A randomized controlled study to evaluate the effect of bexarotene on amyloid-β and apolipoprotein E metabolism in healthy subjects

Kaushik Ghosal, Michael Haag, Philip B Verghese, Tim West, Tim Veenstra, Joel B Braunstein, Randall J Bateman, David M Holtzman, Gary E Landreth, Kaushik Ghosal, Michael Haag, Philip B Verghese, Tim West, Tim Veenstra, Joel B Braunstein, Randall J Bateman, David M Holtzman, Gary E Landreth

Abstract

Introduction: We conducted a phase Ib proof of mechanism trial to determine whether bexarotene (Targretin) increases central nervous system (CNS) apolipoprotein E (apoE) levels and alters Aβ metabolism in normal healthy individuals with the APOE ε3/ε3 genotype.

Methods: We used stable isotope labeling kinetics (SILK-ApoE and SILK-Aβ) to measure the effect of bexarotene on the turnover rate of apoE and Aβ peptides and stable isotope spike absolute quantitation (SISAQ) to quantitate their concentrations in the cerebrospinal fluid (CSF). Normal subjects were treated for 3 days with bexarotene (n = 3 women, 3 men, average 32 years old) or placebo (n = 6 women, average 30.2 years old) before administration of C13-leucine and collection of plasma and CSF over the next 48 hours. Bexarotene concentrations in plasma and CSF were also measured.

Results: Oral administration of bexarotene resulted in plasma levels of 1 to 2 μM; however, only low nM levels were found in CSF. Bexarotene increased CSF apoE by 25% but had no effect on metabolism of Aβ peptides.

Discussion: Bexarotene has poor CNS penetration in normal human subjects. Drug treatment resulted in a modest increase in CSF apoE levels. The absence of an effect on Aβ metabolism is likely reflective of the low CNS levels of bexarotene achieved. This study documents the utility of SILK-ApoE technology in measuring apoE kinetics in humans.

Trial registration: This trial is registered at clinicaltrials.gov (NCT02061878).

Keywords: Alzheimer's disease; Apolipoprotein E; Bexarotene; Retinoid X receptor; β amyloid.

Figures

Fig. 1
Fig. 1
Pharmacokinetics of bexarotene. The concentration of bexarotene in plasma and cerebrospinal fluid (CSF) samples was measured hourly in subjects receiving drug. The average maximal concentration of drug in plasma was 1.46 ± 1 μM, and the Tmax was 3.45 ± 11.41 h after dosing. The levels of bexarotene in CSF was below the level of detection (0.021 μM) in >95% of samples. In samples in which bexarotene could reliably be quantified, peak CSF concentrations were approximately 20 nM.
Fig. 2
Fig. 2
Stable isotope labeling kinetics (SILK) and stable isotope spike absolute quantitation of ApoE in cerebrospinal fluid (CSF). The SILK data are plotted as the normalized tracer-to-tracee ratio (TTR), which is the amount of 13C6-Leu labeled apoE divided by the amount of unlabeled apoE. (A) The mean values are plotted ± 95% confidence intervals (CIs) of placebo- (blue) and bexarotene- (red) treated subjects. (B) The fractional synthesis rates (FSRs), measured from 6 to 17 hours or (C) fractional clearance rates (FCRs) determined from 23 to 48 hours were not significantly different between placebo- and drug-treated subjects. The absolute concentration of apoE in the CSF was calculated by adding the concentration values for the unlabeled and 13C6-labeled apoE. (D) ApoE concentrations of the individual subjects treated with placebo (blue) or bexarotene (red) and (E) their average values (±95% CIs). (F) There was a significant 25% increase (P = .0367) in the mean weighted area under the full concentration curves of apoE in the CSF of the bexarotene-treated subjects. Quantitation of the amount of newly synthesized apoE in (G) individual subject and (H) their average values (±95% CIs) revealed (I) a nonsignificant change between placebo- and bexarotene-treated subjects.
Fig. 3
Fig. 3
Stable isotope labeling kinetics (SILK) of total Aβ and of Aβ40 in cerebrospinal fluid (CSF). The synthesis and clearance rates of total Aβ were measured in CSF using an Aβ1-x capture antibody. The SILK data are plotted as the normalized tracer-to-tracee ratio (TTR), which is the amount of 13C6-Leu labeled Aβ divided by the amount of total unlabeled Aβ. (A) The values for the individual subject treated with placebo (blue) or bexarotene (red), and (B) the mean values are plotted ± 95% confidence intervals (CIs). (C) The fractional synthesis rates (FSRs), measured from 6 to 17 hours. (D) Fractional clearance rates (FCRs) determined from 23 to 48 hours were not significantly different between placebo- and drug-treated subjects. The SILK data are plotted as the normalized TTR, which is the amount of 13C6-Leu labeled Aβ40 divided by the amount of unlabeled Aβ40. (E) The values for the individual subject and (F) the mean values are plotted ± 95% CIs. (G) The FSRs, measured from 6 to 17 hours or (H) FCRs determined from 23 to 48 hours were not significantly different between placebo- and drug-treated subjects.
Fig. 4
Fig. 4
Stable isotope spike absolute quantitation of total Aβ and Aβ40 and newly synthesized total Aβ and Aβ40 in cerebrospinal fluid (CSF). The absolute concentration of total Aβ and Aβ40 peptides in the CSF was calculated by adding the concentration values for the unlabeled and 13C6-labeled peptides using antibodies directed at Aβ1-x or at the C-terminal Aβ40 epitope. (A) Total Aβ or (D) Aβ40 concentrations of the individual subjects treated with placebo (blue) or bexarotene (red) and (B, E) their average values (±95% confidence intervals [CIs]). There was no significant difference in the mean weighted area under the full concentration curves of (C) total Aβ or (F) Aβ40 in the CSF of the bexarotene-treated subjects. The absolute concentration of newly synthesized total Aβ and Aβ40 peptides in the CSF was calculated by adding the concentration values for the unlabeled and 13C6-labeled Aβ peptides at each time point. Quantitation of the mean amount of newly synthesized (G) total Aβ and (J) Aβ40 (±95% CIs) in placebo- and bexarotene-treated subjects revealed no difference in the amount of Aβ peptides (H, K) synthesized or (I, L) cleared in the placebo-treated compared with bexarotene-treated subjects. Abbreviation: AUC, area under the curve.

References

    1. Querfurth H.W., LaFerla F.M. Alzheimer's disease. N Engl J Med. 2010;362:329–344.
    1. Mawuenyega K.G., Sigurdson W., Ovod V., Munsell L., Kasten T., Morris J.C. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science. 2010;330:1774–1776.
    1. Patterson B.W., Elbert D.L., Mawuenyega K.G., Kasten T., Ovod V., Ma S. Age and amyloid effects on human central nervous system amyloid-beta kinetics. Ann Neurol. 2015;78:439–453.
    1. Palop J.J., Mucke L. Amyloid-beta-induced neuronal dysfunction in Alzheimer's disease: from synapses toward neural networks. Nat Neurosci. 2010;13:812–818.
    1. Roses A.D., Saunders A.M. APOE is a major susceptibility gene for Alzheimer's disease. Curr Opin Biotechnol. 1994;5:663–667.
    1. Kim J., Basak J.M., Holtzman D.M. The role of apolipoprotein E in Alzheimer's disease. Neuron. 2009;63:287–303.
    1. Castellano J.M., Kim J., Stewart F.R., Jiang H., DeMattos R.B., Patterson B.W. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci Transl Med. 2011;3:89ra57.
    1. Jiang Q., Lee C.Y., Mandrekar S., Wilkinson B., Cramer P., Zelcer N. ApoE promotes the proteolytic degradation of Abeta. Neuron. 2008;58:681–693.
    1. Donkin J.J., Stukas S., Hirsch-Reinshagen V., Namjoshi D., Wilkinson A., May S. ATP-binding cassette transporter A1 mediates the beneficial effects of the liver-X-receptor agonist GW3965 on object recognition memory and amyloid burden in APP/PS1 mice. J Biol Chem. 2010;285:34144–34154.
    1. Fitz N.F., Cronican A., Pham T., Fogg A., Fauq A.H., Chapman R. Liver X receptor agonist treatment ameliorates amyloid pathology and memory deficits caused by high-fat diet in APP23 mice. J Neurosci. 2010;30:6862–6872.
    1. Riddell D.R., Zhou H., Comery T.A., Kouranova E., Lo C.F., Warwick H.K. The LXR agonist TO901317 selectively lowers hippocampal Abeta42 and improves memory in the Tg2576 mouse model of Alzheimer's disease. Mol Cell Neurosci. 2007;34:621–628.
    1. Suon S., Zhao J., Villarreal S.A., Anumula N., Liu M., Carangia L.M. Systemic treatment with liver X receptor agonists raises apolipoprotein E, cholesterol, and amyloid-beta peptides in the cerebral spinal fluid of rats. Mol Neurodegener. 2010;5:44.
    1. Zelcer N., Tontonoz P. Liver X receptors as integrators of metabolic and inflammatory signaling. J Clin Invest. 2006;116:607–614.
    1. Beaven S.W., Tontonoz P. Nuclear receptors in lipid metabolism: targeting the heart of dyslipidemia. Annu Rev Med. 2006;57:313–329.
    1. Skerrett R., Malm T., Landreth G.E. Nuclear receptors in neurodegenerative diseases. Neurobiol Dis. 2014;72 Pt A:104–116.
    1. Lee C.Y., Tse W., Smith J.D., Landreth G.E. Apolipoprotein E promotes beta-amyloid trafficking and degradation by modulating microglial cholesterol levels. J Biol Chem. 2012;287:2032–2044.
    1. Cramer P.E., Cirrito J.R., Wesson D.W., Lee C.Y., Karlo J.C., Zinn A.E. ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models. Science. 2012;335:1503–1506.
    1. Fitz N.F., Cronican A.A., Lefterov I., Koldamova R. Comment on “ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models”. Science. 2013;340:924-c.
    1. Tai L.M., Koster K.P., Luo J., Lee S.H., Wang Y.T., Collins N.C. Amyloid-beta pathology and APOE genotype modulate retinoid X receptor agonist activity in vivo. J Biol Chem. 2014;289:30538–30555.
    1. Mandrekar-Colucci S., Karlo J.C., Landreth G.E. Mechanisms underlying the rapid peroxisome proliferator-activated receptor-gamma-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer's disease. J Neurosci. 2012;32:10117–10128.
    1. Yamanaka M., Ishikawa T., Griep A., Axt D., Kummer M.P., Heneka M.T. PPARgamma/RXRalpha-induced and CD36-mediated microglial amyloid-beta phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci. 2012;32:17321–17331.
    1. Ulrich J.D., Burchett J.M., Restivo J.L., Schuler D.R., Verghese P.B., Mahan T.E. In vivo measurement of apolipoprotein E from the brain interstitial fluid using microdialysis. Mol Neurodegener. 2013;8:13.
    1. Boehm M.F., Zhang L., Badea B.A., White S.K., Mais D.E., Berger E. Synthesis and structure-activity relationships of novel retinoid X receptor-selective retinoids. J Med Chem. 1994;37:2930–2941.
    1. Tesseur I., Lo A.C., Roberfroid A., Dietvorst S., Van Broeck B., Borgers M. Comment on “ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models”. Science. 2013;340:924-e.
    1. FDA. Bexarotene, new drug application 21-055. 1999. Available at: . Accessed June 20, 2016.
    1. Farol L.T., Hymes K.B. Bexarotene: a clinical review. Expert Rev Anticancer Ther. 2004;4:180–188.
    1. Scarisbrick J.J., Morris S., Azurdia R., Illidge T., Parry E., Graham-Brown R. UK consensus statement on safe clinical prescribing of bexarotene for patients with cutaneous T-cell lymphoma. Br J Dermatol. 2013;168:192–200.
    1. Wildsmith K.R., Han B., Bateman R.J. Method for the simultaneous quantitation of apolipoprotein E isoforms using tandem mass spectrometry. Anal Biochem. 2009;395:116–118.
    1. Bateman R.J., Munsell L.Y., Morris J.C., Swarm R., Yarasheski K.E., Holtzman D.M. Human amyloid-beta synthesis and clearance rates as measured in cerebrospinal fluid in vivo. Nat Med. 2006;12:856–861.
    1. Wildsmith K.R., Basak J.M., Patterson B.W., Pyatkivskyy Y., Kim J., Yarasheski K.E. In vivo human apolipoprotein E isoform fractional turnover rates in the CNS. PLoS One. 2012;7:e38013.
    1. Howell S.R., Shirley M.A., Grese T.A., Neel D.A., Wells K.E., Ulm E.H. Bexarotene metabolism in rat, dog, and human, synthesis of oxidative metabolites, and in vitro activity at retinoid receptors. Drug Metab Dispos. 2001;29:990–998.
    1. Yarasheski K.E., Smith S.R., Powderly W.G. Reducing plasma HIV RNA improves muscle amino acid metabolism. Am J Physiol Endocrinol Metab. 2005;288:E278–E284.
    1. Mandrekar-Colucci S., Landreth G.E. Nuclear receptors as therapeutic targets for Alzheimer's disease. Expert Opin Ther Targets. 2011;15:1085–1097.
    1. Aagaard M.M., Siersbaek R., Mandrup S. Molecular basis for gene-specific transactivation by nuclear receptors. Biochim Biophys Acta. 2011;1812:824–835.
    1. Rotstein B.H., Hooker J.M., Woo J., Collier T.L., Brady T.J., Liang S.H. Synthesis of [(11)C]Bexarotene by Cu-mediated [(11)C] carbon dioxide fixation and preliminary PET imaging. ACS Med Chem Lett. 2014;5:668–672.
    1. Landreth G.E., Cramer P.E., Lakner M.M., Cirrito J.R., Wesson D.W., Brunden K.R. “Response to comments on ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models”. Science. 2013;340:924-g.
    1. Kobayashi T., Furusawa Y., Yamada S., Akehi M., Takenaka F., Sasaki T. Positron emission tomography to elucidate pharmacokinetic differences of regioisomeric retinoid x receptor agonists. ACS Med Chem Lett. 2015;6:334–338.
    1. Sheline Y.I., West T., Yarasheski K., Swarm R., Jasielec M.S., Fisher J.R. An antidepressant decreases CSF Abeta production in healthy individuals and in transgenic AD mice. Sci Transl Med. 2014;6:236re4.
    1. Montagne A., Barnes S.R., Sweeney M.D., Halliday M.R., Sagare A.P., Zhao Z. Blood-brain barrier breakdown in the aging human hippocampus. Neuron. 2015;85:296–302.
    1. Cummings J.L., Zhong K., Kinney J.W., Heaney C., Moll-Tudla J., Joshi A. Double-blind, placebo-controlled, proof-of-concept trial of bexarotene Xin moderate Alzheimer's disease. Alzheimers Res Ther. 2016;8:4.
    1. Pierrot N., Lhommel R., Quenon L., Hanseeuw B., Dricot L., Sindic C. Targretin improves cognitive and biological markers in a patient with Alzheimer's disease. J Alzheimers Dis. 2015;49:271–276.
    1. Lerner V., Miodownik C., Gibel A., Sirota P., Bush I., Elliot H. The retinoid X receptor agonist bexarotene relieves positive symptoms of schizophrenia: a 6-week, randomized, double-blind, placebo-controlled multicenter trial. J Clin Psychiatry. 2013;74:1224–1232.
    1. Heneka M.T., Fink A., Doblhammer G. Effect of pioglitazone medication on the incidence of dementia. Ann Neurol. 2015;78:284–294.
    1. McFarland K., Spalding T.A., Hubbard D., Ma J.N., Olsson R., Burstein E.S. Low dose bexarotene treatment rescues dopamine neurons and restores behavioral function in models of Parkinson's disease. ACS Chem Neurosci. 2013;4:1430–1438.
    1. Roy A., Jana M., Kundu M., Corbett G.T., Rangaswamy S.B., Mishra R.K. HMG-CoA reductase inhibitors bind to PPARalpha to upregulate neurotrophin expression in the brain and improve memory in mice. Cell Metab. 2015;22:253–265.
    1. Mounier A., Georgiev D., Nam K.N., Fitz N.F., Castranio E.L., Wolfe C.M. Bexarotene-activated retinoid X receptors regulate neuronal differentiation and dendritic complexity. J Neurosci. 2015;35:11862–11876.
    1. Tachibana M., Shinohara M., Yamazaki Y., Liu C.C., Rogers J., Bu G. Rescuing effects of RXR agonist bexarotene on aging-related synapse loss depend on neuronal LRP1. Exp Neurol. 2016;277:1–9.
    1. Nam K.N., Mounier A., Fitz N.F., Wolfe C., Schug J., Lefterov I. RXR controlled regulatory networks identified in mouse brain counteract deleterious effects of Abeta oligomers. Sci Rep. 2016;6:24048.
    1. Riancho J., Ruiz-Soto M., Berciano M.T., Berciano J., Lafarga M. Neuroprotective effect of bexarotene in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Front Cell Neurosci. 2015;9:250.

Source: PubMed

3
Předplatit