An evaluation of MSDC-0160, a prototype mTOT modulating insulin sensitizer, in patients with mild Alzheimer's disease

Raj C Shah, Dawn C Matthews, Randolph D Andrews, Ana W Capuano, Debra A Fleischman, James T VanderLugt, Jerry R Colca, Raj C Shah, Dawn C Matthews, Randolph D Andrews, Ana W Capuano, Debra A Fleischman, James T VanderLugt, Jerry R Colca

Abstract

Alzheimer's disease (AD) is associated with insulin resistance and specific regional declines in cerebral metabolism. The effects of a novel mTOT modulating insulin sensitizer (MSDC-0160) were explored in non-diabetic patients with mild AD to determine whether treatment would impact glucose metabolism measured by FDG-PET in regions that decline in AD. MSDC-0160 (150 mg once daily; N=16) compared to placebo (N=13) for 12 weeks did not result in a significant difference in glucose metabolism in pre-defined regions when referenced to the pons or whole brain. However, glucose metabolism referenced to cerebellum was maintained in MSDC-0160 treated participants while it significantly declined for placebo patients in anterior and posterior cingulate, and parietal, lateral temporal, medial temporal cortices. Voxel-based analyses showed additional differences in FDG-PET related to MSDC-0160 treatment. These exploratory results suggest central effects of MSDC-0160 and provide a basis for further investigation of mTOT modulating insulin sensitizers in AD patients.

Figures

Fig. (1)
Fig. (1)
Effect on change in FDG-PET, representative views from sagittal, coronal, and transaxial aspects, referenced to the cerebellum. Significant clusters from with-in group and subtraction analyses relative to cerebellar gray reference region are superimposed on selected transaxial MRI template slices. Blue indicates a decrease in glucose metabolism over the treatment period relative to the reference region, and red a relative increase (or preservation). The placebo group showed an AD-like pattern of decline that was absent in the MSDC-0160 group. The lower panel is a subtraction to show the relative effect of the MSDC-0160 treatment.
Fig. (2)
Fig. (2)
Overview of voxel based analyses using three reference regions. Significant clusters fromwithin-group analyses were superimposed on selected transaxial MRI template slices.Blue indicates a decrease in glucose metabolism over the treatment period relative to the reference region, and red a relative increase (or preservation). While the Placebo group exhibited an AD-like pattern of decline (and relative preservation), the MSDC-0160 group showed relative increases in regions including parahippocampus when referenced to whole brain and cerebellum, and decreases most evident in frontal and cerebellar gray regions when referenced to pons.

References

    1. Alzheimer’s Association.2013 Alzheimer s disease facts and figures. Alzheimers Dement. 2013;9:208–245.
    1. Hyman BT, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Carrillo MC , et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimer Dement. 2013:1–13.
    1. Pellerin L. Food for thought: the importance of glucose and other energy substrates for sustaining brain function under varying levels of activity. Diabetes Metab. 2010;36:S59–63.
    1. Chen X, Yan SD. Mitochondrial Abeta: a potential cause of metabolic dysfunction in Alzheimer’s disease. IUBMB Life. 2006;58:686–694.
    1. Blass JP. Brain metabolism and brain disease: is metabolic deficiency the proximate cause of Alzheimer dementia?. J Neurosci Res. 2001;66:851–856.
    1. Cunnane S, Nugent S, Roy M, Courchesne-Loyer A, Croteau E, Tremblay S , et al. Brain fuel metabolism. aing.and Alzheimer s disease. . Nutrition . 2011;27:3–20.
    1. Li Y, Rinne JO, Mosconi L, Pirraglia L, Rusinek H, DeSanti S, Nina Kemppainen N , et al. Regional analysis of FDG and PIB-PET images in normal aging. mild cognitive impairent.and Alzheimer's disease. Eur J Nucl Med Mol Imaging. 2008; 3512 :2169–2181.
    1. Landau SM, Harvey D, Madison CM, Koeppe RA, Reiman EM, Foster NL , et al. Associations between cognitive. functinal.and FDG-PET measures of decline in AD and MCI. Neurobiol Aging. 2011; 327:1207–1218.
    1. Chen K, Langbaum JB, Fleisher AS, Ayutyanont N, Reschke C, Lee W , et al. Twelve-month metabolic declines in probable Alzheimer's disease and amnestic mild cognitive impairment assessed using an empirically pre-defined statistical region-of-interest: findings from the Alzheimer's Disease Neuroimaging Initiative. Neuroimage. 2010;51:654–664.
    1. Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A , et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance. IRS-1 dysregulaion.and cognitive decline. J Clin Invest. 2012; 1224 :1316–1338.
    1. Parihar M, Brewer GJ. Mitoenergetic failure in Alzheimer disease. Am J Physiol Cell Physiol. 2007;292:C8–C23.
    1. Swerdlow RH. Brain aging. Alzheimer s disase.and mitochondria. Biochim Biophys Acta. 2011; 1812:1630–1639.
    1. Silva DF, Selfridge JE, Lu J, Lezi E, Cardoso SM, Swerdlow RH. Mitochondrial abnormalities in Alzheimer’s disease: possible targets for therapeutic intervention. Adv Pharmacol. 2012;64:83–126.
    1. de la Monte SM. Brain insulin resistance and deficiency as therapeutic targets in Alzheimer’s disease. CurrAlz. 2012;Res 9:35–66.
    1. Colca JR, Feinstein DL. Altering mitochondrial dysfunction as an approach to treating Alzheimer's disease. Adv Pharmacol. 2012;64:155–176.
    1. Hanyu H, Sato T, Kiuchi A, Sakurai H, Iwamoto T. Pioglitazone improved cognition in a pilot study on patients with Alzheimer’s disease and mile cognitive impairment with diabetes mellitus. J Am Geriatr Soc. 2009;571:177–179.
    1. Hanyu H, Sato T, Sakuri H, Iwamoto T. The role of tumor necrosis factor alpha in the cognitive improvement after peroxisome proliferator activator receptor gamma agonist pioglitazone treatment in Alzheimer’s disease. J Am Geriatr Soc. 2010;585:1000–1001.
    1. Miller BW, Willett KC, Desilets AR. Rosiglitazone and pioglitazone for the treatment of Alzheimer's disease. Ann Pharmacother. 2011;45:1416–1424.
    1. Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, Iwamoto T. Efficacy of PPAR-? agonist pioglitazone in mild Alzheimer disease. Neurobiol Aging. 2011;32:1626–1633.
    1. Tzimopoulou S, Cunningham VJ, Nichols TE, Searle G, Bird NP, Mistry P , et al. A multi-center randomized proof-of-concept clinical trial applying [18F]FDG-PET for evaluation of metabolic therapy with rosiglitazone XR in mild to moderate Alzheimer’s disease. J. Alzheimers Dis. 2010;22:1241–1256.
    1. Harrington C, Sawchak S, Chiang C, Davies J, Donovan C, Saunders AM , et al. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimer's disease: two phase 3 studies. curr Alzheimer Res. 2011;85:592–606.
    1. Colca JR, McDonald WG, Cavey GS, Cole SL, Holewa DD, Brightwell-Conrad AS , et al. Identification of a mitochondrial target of thiazolidinedione insulin sensitizers mTOT)-relationship to newly identified mitochondrial pyruvate carrier proteins. PLOS One. 2013;8 e61551:1–10.
    1. Divakaruni AS, Wiley SE, Rogers GW, Andreyev AY, Petrosyan S, Loviscach M , et al. Thiazolidinediones are acute. specific inhibitors of the mitochondrial pyruvate carrier. PNAS. 2013;110:5422–5427.
    1. Colca JR, VanderLugt JT, Adams WJ, Shashlo A, McDonald WG, Liang J , et al. Clinical proof of concept with MSDC-0160. a prototype mTOT modulating insulin sensitizer. Clin Pharm Ther. 2013;93:352–359.
    1. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s disease. Neurology. 1984;34:939–944.
    1. Folstein MF, Folstein SE, McHugh PR. “Mini-Mental State”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189–198.
    1. Bauer CM, Cabral HJ, Greve DN, Killiany RJ. Differentiating between normal aging. Mild Cognitive Impairent.and Alzheimer s disease with FDG-PET Effects of normalization region and partial volume correcting method. . J Alzheimers Dis Parkinsonism. 2013;31
    1. Galea E, Feinstein DL, Lacombe P. Pioglitazone does not increase cerebral glucose utilization in a murine model of Alzheimer’s disease and decreases it in wild-type mice. Diabetologia. 2006;49:2153–2161.
    1. Volkow NDFowler JS, Wang GJ, Telang F, Logan J, Wong C , et al. Methylphenidate decreased the amount of glucose needed by the brain to perform a cognitive task. PLOS One. 2008;3:e2017.
    1. Edison P, Archer HA, Hinz R, Hammers A, Pavese N, Tai YF , et al. Amyloid. hypometaboism.and cognition in Alzheimer disease An [11C]PIB and [18F]FDG PET study. Neurology . 2007;8: 501–508.
    1. Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R , et al. Microglial activation and amyloid deposition in mild cognitive impairment: A PET study. Neurology. 2009;72:56–62.
    1. Mosconi L, Tsui WH, Rusinek H, De Santi S, Li Y, Wang GJ , et al. Quantitation. regional vulnerabiity.and kinetic modeling of brain glucose metabolism in mild Alzheimer s disease. Eur J Nucl Med Mol Imaging . 2007; 34:1467–1479.
    1. Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, Iwamoto T. Efficacy of PPAR agonist pioglitazone in mild Alzheimer’s disease. Neurobiol Aging. 2011;32:16261633.
    1. Colca JR, Tanis SP, McDonald WG, Kletzein RF. Insulin sensitizers in 2013 New insights for the development of novel therapeutic agents to treat metabolic disease. Exp Opin Invest Drugs. 2013;23:1–7.
    1. Colca JR, McDonald WG, Kletzien RF. The Mitochondrial Target of Thiazolidinediones Diabetes. Obesity and Metabolism in press. 2014
    1. Schneider LS, Lahiri DK. The perils of Alzheimer's drug development. Curr Alzheimer Res. 2009;61:77–8.

Source: PubMed

3
Sottoscrivi