A pilot randomized, placebo controlled, double blind phase I trial of the novel SIRT1 activator SRT2104 in elderly volunteers

Vincenzo Libri, Andrew P Brown, Giulio Gambarota, Jonathan Haddad, Gregory S Shields, Helen Dawes, David J Pinato, Ethan Hoffman, Peter J Elliot, George P Vlasuk, Eric Jacobson, Martin R Wilkins, Paul M Matthews, Vincenzo Libri, Andrew P Brown, Giulio Gambarota, Jonathan Haddad, Gregory S Shields, Helen Dawes, David J Pinato, Ethan Hoffman, Peter J Elliot, George P Vlasuk, Eric Jacobson, Martin R Wilkins, Paul M Matthews

Abstract

Background: SRT2104 has been developed as a selective small molecule activator of SIRT1, a NAD(+)-dependent deacetylase involved in the regulation of energy homeostasis and the modulation of various metabolic pathways, including glucose metabolism, oxidative stress and lipid metabolism. SIRT1 has been suggested as putative therapeutic target in multiple age-related diseases including type 2 diabetes and dyslipidemias. We report the first clinical trial of SRT2104 in elderly volunteers.

Methods: Oral doses of 0.5 or 2.0 g SRT2104 or matching placebo were administered once daily for 28 days. Pharmacokinetic samples were collected through 24 hours post-dose on days 1 and 28. Multiple pharmacodynamic endpoints were explored with oral glucose tolerance tests (OGTT), serum lipid profiles, magnetic resonance imaging (MRI) for assessment of whole body visceral and subcutaneous fat, maximal aerobic capacity test and muscle 31P magnetic resonance spectroscopy (MRS) for estimation of mitochondrial oxidative capacity.

Results: SRT2104 was generally safe and well tolerated. Pharmacokinetic exposure increased less than dose-proportionally. Mean Tmax was 2-4 hours with elimination half-life of 15-20 hours. Serum cholesterol, LDL levels and triglycerides decreased with treatment. No significant changes in OGTT responses were observed. 31P MRS showed trends for more rapid calculated adenosine diphosphate (ADP) and phosphocreatine (PCr) recoveries after exercise, consistent with increased mitochondrial oxidative phosphorylation.

Conclusions: SRT2104 can be safely administered in elderly individuals and has biological effects in humans that are consistent with SIRT1 activation. The results of this study support further development of SRT2104 and may be useful in dose selection for future clinical trials in patients.

Trial registration: ClinicalTrials.gov NCT00964340.

Conflict of interest statement

Competing Interests: At the time this publication was written, JH, EH, PEGPV and EJ were employed by Sirtris Pharmaceuticals, a GlaxoSmithKline company developing SR2104 and related molecules. APB, GG, and PMM were employed by GlaxoSmithKline Research and Development (GSK R&D), Ltd. VL was employed by GSK R&D until April 2009. APB is currently employed by Imanova, Centre of Imaging Sciences, London, UK. The other authors declared no conflicts of interest. The above competing interests do not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. CONSORT 2010 Flow Diagram.
Figure 1. CONSORT 2010 Flow Diagram.
The schema graphically outlines the design and conduct of the clinical study.
Figure 2. Mean plasma concentration versus time…
Figure 2. Mean plasma concentration versus time plots for SRT2104 on day 1 and 28, following multiple dose administration of 0.5 g/day (top panel) and 2.0 g/day (lower panel) to elderly male and female volunteers.
Figure 3. Lipid profile at baseline and…
Figure 3. Lipid profile at baseline and after 28 days treatment with placebo and SRT2104 (0.5 g/day and 2.0 g/day).
[Black dots indicate points that are two interquartile ranges outside the means; * P

Figure 4. Oral glucose tolerance tests (OGTTs)…

Figure 4. Oral glucose tolerance tests (OGTTs) at baseline and after 28 days treatment placebo…

Figure 4. Oral glucose tolerance tests (OGTTs) at baseline and after 28 days treatment placebo (upper) SRT2104 0.5 g/day (middle) or SRT2104 2.0 g/day (lower panel).

Figure 5. Insulin (left) and C-Peptide (right)…

Figure 5. Insulin (left) and C-Peptide (right) concentration–time curves at baseline (screening) and after 28…

Figure 5. Insulin (left) and C-Peptide (right) concentration–time curves at baseline (screening) and after 28 days treatment with placebo (upper) SRT2104 0.5 g/day (middle) or SRT2104 2.0 g/day (lower panel).

Figure 6. Representative example of magnetic resonance…

Figure 6. Representative example of magnetic resonance images and 31 P Magnetic resonance spectroscopy results.

Top…

Figure 6. Representative example of magnetic resonance images and 31P Magnetic resonance spectroscopy results.
Top left panel: intra-abdominal (red) and subcutaneous (yellow) adipose tissue maps overlaid on coronal and sagittal body MRI images from a single subject. Bottom left panel: dynamic series of 31P MRS spectra acquired serially during exercise and recovery from a single subject as described in Methods. Right panel: box plot of PCr recovery time change from baseline (day −1) to day 27 for the placebo, SRT2104 0.5 g/day and 2.0 g/day groups.
Figure 4. Oral glucose tolerance tests (OGTTs)…
Figure 4. Oral glucose tolerance tests (OGTTs) at baseline and after 28 days treatment placebo (upper) SRT2104 0.5 g/day (middle) or SRT2104 2.0 g/day (lower panel).
Figure 5. Insulin (left) and C-Peptide (right)…
Figure 5. Insulin (left) and C-Peptide (right) concentration–time curves at baseline (screening) and after 28 days treatment with placebo (upper) SRT2104 0.5 g/day (middle) or SRT2104 2.0 g/day (lower panel).
Figure 6. Representative example of magnetic resonance…
Figure 6. Representative example of magnetic resonance images and 31P Magnetic resonance spectroscopy results.
Top left panel: intra-abdominal (red) and subcutaneous (yellow) adipose tissue maps overlaid on coronal and sagittal body MRI images from a single subject. Bottom left panel: dynamic series of 31P MRS spectra acquired serially during exercise and recovery from a single subject as described in Methods. Right panel: box plot of PCr recovery time change from baseline (day −1) to day 27 for the placebo, SRT2104 0.5 g/day and 2.0 g/day groups.

References

    1. Haigis MC, Sinclair DA (2010) Mammalian sirtuins: biological insights and disease relevance. Annu Rev Pathol 5: 253–295.
    1. Leibiger IB, Berggren P-O (2006) Sirt1: a metabolic master switch that modulates lifespan. Nat Med 12: 34–36.
    1. Camins A, Sureda FX, Junyent F, Verdaguer E, Folch J, et al. (2010) Sirtuin activators: designing molecules to extend life span. Biochim Biophys Acta 1799: 740–749.
    1. Minor RK, Baur JA, Gomes AP, Ward TM, Csiszar A, et al. (2011) SRT1720 improves survival and healthspan of obese mice. Sci Rep (Nature) 1: 1–10.
    1. Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ, et al. (2007) Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450: 712–716.
    1. Feige JN, Lagouge M, Canto C, Strehle A, Houten SM, et al. (2008) Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab 8: 347–358.
    1. Heilbronn LK, de Jonge L, Frisard MI, DeLany JP, Larson-Meyer DE, et al. (2006) Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: a randomized controlled trial. JAMA 295: 1539–1548.
    1. Hou X, Xu S, Maitland-Toolan KA, Sato K, Jiang B, et al. (2008) SIRT1 regulates hepatocyte lipid metabolism through activating AMP-activated protein kinase. J Biol Chem 283: 20015–20026.
    1. Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, et al. (2004) Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature 429: 771–776.
    1. Bordone L, Motta MC, Picard F, Robinson A, Jhala US, et al. (2006) Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol 4.
    1. Imai S, Armstrong CM, Kaeberlein M, Guarente L (2000) Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403: 795–800.
    1. Baur JA (2010) Resveratrol, sirtuins, and the promise of a DR mimetic. Mech Ageing Dev 131: 261–269.
    1. Elliott PJ, Walpole S, Morelli L, Lambert PD, Lunsmann W, et al. (2009) Resveratrol/SRT-501. Drugs Fut 34 4: 291.
    1. Nayagam VM, Wang X, Tan YC, Poulsen A, Goh KC, et al. (2006) SIRT1 modulating compounds from high-throughput screening as anti-inflammatory and insulin-sensitizing agents. J Biomol Screen 11: 959–967.
    1. Yang H, Baur JA, Chen A, Miller C, Adams JK, et al. (2007) Design and synthesis of compounds that extend yeast replicative lifespan. Aging Cell 6: 35–43.
    1. Bemis JE, Vu CB, Xie R, Nunes JJ, Ng PY, et al. (2009) Discovery of oxazolo[4,5-b]pyridines and related heterocyclic analogs as novel SIRT1 activators. Bioorg Med Chem Lett 19: 2350–2353.
    1. Qi Y DM, Hirsch ML, Cote AM, Lainez EO, Johnson MO, et al... (2010) Activation of Sirtuin1 (SIRT1) by the novel small molecule SRT2104 promotes body weight loss, increases exercise capacity and improves insulin sensitivity in diet-induced obese mice. ADA, 70th Scientific Sessions 390.
    1. Suri V DM, Qi Y, Lainez EO, Cote AM, Johnson MO, Gagne DJ, et al... (2011) The Metabolomic Profile of SRT2104, a Selective SIRT1 Activator in High Fat Diet Fed Mice Reveals Significant Improvements in Metabolic Function in Liver, Heart and Skeletal Muscle. ADA, 71th Scientific Sessions 1706.
    1. Hoffmann E, Wald J, Lavu S, Roberts J, Beaumont C, et al. (2012) Pharmacokinetics and Tolerability of SRT2104, a First-In-Class Small Molecule Activator of SIRT1, after Single and Repeated Oral Administration in Man. Br J Clin Pharmacol doi:.
    1. DECODE Study Group on behalf of the European Diabetes Epidemiology Study Group (1998) Will new diagnostic criteria for diabetes mellitus change phenotype of patients with diabetes? Reanalysis of European epidemiological data. BMJ 8: 371–375.
    1. He Y-L, Wang Y, Bullock JM, Deacon CF, Holst JJ, et al. (2007) Pharmacodynamics of vildagliptin in patients with type 2 diabetes during OGTT. J Clin Pharmacol 47: 633–641.
    1. Matthews PM, Allaire C, Shoubridge EA, Karpati G, Carpenter S, et al. (1991) In vivo muscle magnetic resonance spectroscopy in the clinical investigation of mitochondrial disease. Neurology 41: 114–120.
    1. Larson-Meyer DE, Newcomer BR, Hunter GR, Hetherington HP, Weinsier RL (2000) 31P MRS measurement of mitochondrial function in skeletal muscle: reliability, force-level sensitivity and relation to whole body maximal oxygen uptake. NMR Biomed 13: 14–27.
    1. Schocke MFH, Esterhammer R, Kammerlander C, Rass A, Kremser C, et al. (2004) High-energy phosphate metabolism during incremental calf exercise in humans measured by 31 phosphorus magnetic resonance spectroscopy (31P MRS). Magn Reson Imaging 22: 109–115.
    1. Kemp GJ, Radda GK (1994) Quantitative interpretation of bioenergetic data from 31P and 1H magnetic resonance spectroscopic studies of skeletal muscle: an analytical review. Magn Reson Q 10: 43–63.
    1. Larson-Meyer DE, Newcomer BR, Hunter GR, Joanisse DR, Weinsier RL, et al. (2001) Relation between in vivo and in vitro measurements of skeletal muscle oxidative metabolism. Muscle Nerve 24: 1665–1676.
    1. Hume R (1966) Prediction of lean body mass from height and weight. J Clin Pathol 19: 389–391.
    1. Naressi A, Couturier C, Devos JM, Janssen M, Mangeat C, et al. (2001) Java-based graphical user interface for the MRUI quantitation package. MAGMA 12: 141–152.
    1. Vanderthommen M, Duteil S, Wary C, Raynaud JS, Leroy-Willig A, et al. (2003) A comparison of voluntary and electrically induced contractions by interleaved 1H- and 31P-NMRS in humans. J Appl Physiol 94: 1012–1024.
    1. Rofsky NM, Lee VS, Laub G, Pollack MA, Krinsky GA, et al. (1999) Abdominal MR imaging with a volumetric interpolated breath-hold examination. Radiology 212: 876–884.
    1. Thomas EL, Saeed N, Hajnal JV, Brynes A, Goldstone AP, et al. (1998) Magnetic resonance imaging of total body fat. J Appl Physiol 85: 1778–1785.
    1. Medicine ACoS (2009) ACSM's Guidelines for exercise testing and prescription. Philadelphia, PA: Lippincott, Williams and Wilkins.
    1. Rostami-Hodjegan A, Tucker GT (2007) Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 6: 140–148.
    1. Zern TL, West KL, Fernandez ML (2003) Grape polyphenols decrease plasma triglycerides and cholesterol accumulation in the aorta of ovariectomized guinea pigs. J Nutr 133: 2268–2272.
    1. Do G-M, Kwon E-Y, Kim H-J, Jeon S-M, Ha T-Y, et al. (2008) Long-term effects of resveratrol supplementation on suppression of atherogenic lesion formation and cholesterol synthesis in apo E-deficient mice. Biochem Biophys Res Commun 374: 55–59.
    1. Li X, Zhang S, Blander G, Tse JG, Krieger M, et al. (2007) SIRT1 deacetylates and positively regulates the nuclear receptor LXR. Mol Cell 28: 91–9106.
    1. Jacobson EL, Kim H, Kim M, Jacobson MK (2012) Niacin: vitamin and antidyslipidemic drug. Subcell Biochem 56: 37–47.
    1. Fabbrini E, Mohammed BS, Korenblat KM, Magkos F, McCrea J, et al. (2010) Effect of Fenofibrate and Niacin on Intrahepatic Triglyceride Content, Very Low-Density Lipoprotein Kinetics, and Insulin Action in Obese Subjects with Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 95: 2727–35.
    1. Ponugoti B, Kim D-H, Xiao Z, Smith Z, Miao J, et al. (2010) SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism. J Biol Chem 285: 33959–33970.
    1. Mooy JM, Grootenhuis PA, de Vries H, Kostense PJ, Popp-Snijders C, et al. (1996) Intra-individual variation of glucose, specific insulin and proinsulin concentrations measured by two oral glucose tolerance tests in a general Caucasian population: the Hoorn Study. Diabetologia 39: 298–305.
    1. Utzschneider KM, Prigeon RL, Tong J, Gerchman F, Carr DB, et al. (2007) Within-subject variability of measures of beta cell function derived from a 2 h OGTT: implications for research studies. Diabetologia 50: 2516–2525.
    1. Gurd BJ, Yoshida Y, McFarlan JT, Holloway GP, Moyes CD, et al. (2011) Nuclear SIRT1 activity, but not protein content, regulates mitochondrial biogenesis in rat and human skeletal muscle. Am J Physiol Regul Integr Comp Physiol 301: 67–75.
    1. Menzies KJ, Hood DA (2012) The role of SirT1 in muscle mitochondrial turnover. Mitochondrion 12: 5–13.
    1. Tomlin DL, Wenger HA (2001) The relationship between aerobic fitness and recovery from high intensity intermittent exercise. Sports Med 31: 1–11.
    1. Bassett DR, Howley ET (2000) Limiting factors for maximum oxygen uptake and determinants of endurance performance. Med Sci Sports Exerc 32: 70–84.
    1. Dandurand RJ, Matthews PM, Arnold DL, Eidelman DH (1995) Mitochondrial disease. Pulmonary function, exercise performance, and blood lactate levels. Chest 108: 182–9.
    1. Layec G, Haseler LJ, Richardson RS (2012) Reduced muscle oxidative capacity is independent of O2 availability in elderly people. Age doi:.
    1. Thomas EL, Makwana A, Newbould R, Rao AW, Gambarota G, et al. (2011) Pragmatic study of orlistat 60 mg on abdominal obesity. Eur J Clin Nutr 65: 1256–1262.
    1. Kelley DE, Kuller LH, McKolanis TM, Harper P, Mancino J, et al. (2004) Effects of moderate weight loss and orlistat on insulin resistance, regional adiposity, and fatty acids in type 2 diabetes. Diabetes Care 27: 33–40.
    1. Berggren JR, Boyle KE, Chapman WH, Houmard JA (2008) Skeletal muscle lipid oxidation and obesity: influence of weight loss and exercise. Am J Physiol Endocrinol Metab 294: 726–32.

Source: PubMed

3
Se inscrever