Selection and early clinical evaluation of the brain-penetrant 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitor UE2343 (Xanamem™)

Scott P Webster, Andrew McBride, Margaret Binnie, Karen Sooy, Jonathan R Seckl, Ruth Andrew, T David Pallin, Hazel J Hunt, Trevor R Perrior, Vincent S Ruffles, J William Ketelbey, Alan Boyd, Brian R Walker, Scott P Webster, Andrew McBride, Margaret Binnie, Karen Sooy, Jonathan R Seckl, Ruth Andrew, T David Pallin, Hazel J Hunt, Trevor R Perrior, Vincent S Ruffles, J William Ketelbey, Alan Boyd, Brian R Walker

Abstract

Background and purpose: Reducing glucocorticoid exposure in the brain via intracellular inhibition of the cortisol-regenerating enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) has emerged as a therapeutic strategy to treat cognitive impairment in early Alzheimer's disease (AD). We sought to discover novel, brain-penetrant 11β-HSD1 inhibitors as potential medicines for the treatment of AD.

Experimental approach: Medicinal chemistry optimization of a series of amido-thiophene analogues was performed to identify potent and selective 11β-HSD1 inhibitors with optimized oral pharmacokinetics able to access the brain. Single and multiple ascending dose studies were conducted in healthy human subjects to determine the safety, pharmacokinetic and pharmacodynamic characteristics of the candidate compound.

Results: UE2343 was identified as a potent, orally bioavailable, brain-penetrant 11β-HSD1 inhibitor and selected for clinical studies. No major safety issues occurred in human subjects. Plasma adrenocorticotropic hormone was elevated (a marker of systemic enzyme inhibition) at doses of 10 mg and above, but plasma cortisol levels were unchanged. Following multiple doses of UE2343, plasma levels were approximately dose proportional and the terminal t1/2 ranged from 10 to 14 h. The urinary tetrahydrocortisols/tetrahydrocortisone ratio was reduced at doses of 10 mg and above, indicating maximal 11β-HSD1 inhibition in the liver. Concentrations of UE2343 in the CSF were 33% of free plasma levels, and the peak concentration in CSF was ninefold greater than the UE2343 IC50 .

Conclusions and implications: UE2343 is safe, well tolerated and reaches the brain at concentrations predicted to inhibit 11β-HSD1. UE2343 is therefore a suitable candidate to test the hypothesis that 11β-HSD1 inhibition in brain improves memory in patients with AD.

© 2016 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

Figure 1
Figure 1
Lead structure and properties. Pharmacokinetics in rats was conducted by i.v. (n = 3 rats) and p.o. administration (n = 5 rats). The number of individual experiments [n] for measurement of in vitro human 11β‐HSD1 inhibition, plasma protein binding and aqueous solubility are shown. Data are reported as mean ± SD.
Figure 2
Figure 2
UE2343 concentrations following a single oral dose of UE2343. Six healthy male and two healthy female subjects per dose level.
Figure 3
Figure 3
Plasma ACTH following a single oral dose of UE2343. Six healthy male and two healthy female subjects per dose level. (A) ACTH levels up to 125 h post‐dose. (B) ACTH at 23 h post‐dose; ANOVA *P < 0.05.
Figure 4
Figure 4
Urinary THFs/THE ratios. (A) Effects on Day 1 following a single dose of UE2343; ANOVA *P < 0.05. Six healthy male and two healthy female subjects per dose level. (B) Effects up to Day 12 following multiple doses of UE2343; ANOVA, 10 (P < 0.05), 20 (P < 0.05) and 35 mg (P < 0.05). Eight healthy male subjects per dose level.
Figure 5
Figure 5
UE2343 levels in plasma and CSF following 4 days of administration of 35 mg b.i.d in four healthy male subjects.

References

    1. Alexander SPH, Catterall WA, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015a). The Concise Guide to PHARMACOLOGY 2015/16: Voltage‐gated ion channels. Br J Pharmacol 172: 5904–5941.
    1. Alexander SPH, Cidlowski JA, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015b). The Concise Guide to PHARMACOLOGY 2015/16: Nuclear hormone receptors. Br J Pharmacol 172: 5956–5978.
    1. Alexander SPH, Fabbro D, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015c). The Concise Guide to PHARMACOLOGY 2015/16: Enzymes. Br J Pharmacol 172: 6024–6109.
    1. Anagnostis P, Athyros VG, Tziomalos K, Karagiannis A, Mikhailidis DP (2009). Clinical review: the pathogenetic role of cortisol in the metabolic syndrome: a hypothesis. J Clin Endocrinol Metab 94: 2692–2701.
    1. Best R, Walker BR (1997). Additional value of measurement of urinary cortisone and unconjugated cortisol metabolites in assessing the activity of 11β‐hydroxysteroid dehydrogenase in vivo . Clin Endocrinol (Oxf) 47: 231–236.
    1. Boonen E, Vervenne H, Meersseman P, Andrew R, Mortier L, Declercq PE et al. (2013). Reduced cortisol metabolism during critical illness. N Engl J Med 368: 1477–1488.
    1. Cernansky JG, Dong H, Fagan AM, Wang L, Xiong C, Holtzman DM et al. (2006). Plasma cortisol and progression of dementia in subjects with Alzheimer‐type dementia. Am J Psychiatry 163: 2164–2169.
    1. Curtis MJ, Bond RA, Spina D, hluwalia A A, Alexander SPA, Giembycz MA et al. (2015). Experimental design and analysis and their reporting: new guidance for publication in BJP. Br J Pharmacol 172: 3461–3471.
    1. Feig PU, Shah S, Hermanowski‐Vosatka A, Plotkin D, Springer MS, Donahue S et al. (2011). Effects of an 11β‐hydroxysteroid dehydrogenase type 1 inhibitor, MK‐0916, in patients with type 2 diabetes mellitus and metabolic syndrome. Diabetes Obes Metab 13: 498–504.
    1. Freude S, Heise T, Woerle HJ, Jungnik A, Rauch T, Hamilton B et al. (2016). Safety, pharmacokinetics and pharmacodynamics of BI 135585, a selective 11β‐hydroxysteroid dehydrogenase‐1 (HSD1) inhibitor in humans: liver and adipose tissue 11β‐HSD1 inhibition after acute and multiple administrations over 2 weeks. Diabetes Obes Metab 18: 486–490.
    1. Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM (2006). Glucocorticoids increase amyloid‐β and tau pathology in a mouse model of Alzheimer's disease. J Neurosci 26: 9047–9056.
    1. Heise T, Morrow L, Hompesch M, Häring HU, Kapitza C, Abt M et al. (2014). Safety, efficacy and weight effect of two 11β‐HSD1 inhibitors in metformin‐treated patients with type 2 diabetes. Diabetes Obes Metab 16: 1070–1077.
    1. Katz DA, Liu W, Locke C, Jacobson P, Barnes DM, Basu R et al. (2013). Peripheral and central nervous system inhibition of 11β‐hydroxysteroid dehydrogenase type 1 in man by the novel inhibitor ABT‐384. Transl Psychiatry 3: e295.
    1. Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG (2010). Animal research: reporting in vivo experiments: the ARRIVE guidelines. Br J Pharmacol 160: 1577–1579.
    1. Kornhuber J, Jessen F (2015). Cerebrospinal fluid cortisol and clinical disease progression in MCI and dementia of Alzheimer's type. Neurobiol Aging 36: 601–607.
    1. Lui W, Katz DA, Locke C, Daszkowski D, Wang Y, Rieser MJ et al. (2013). Clinical safety, pharmacokinetics, and pharmacodynamics of the 11β‐hydroxysteroid dehydrogenase type 1 Inhibitor ABT‐384 in healthy volunteers and elderly adults. Clin Pharmacol Drug Dev 2: 133–151.
    1. Lupien SJ, Leon M, Santi S, Convit A, Tarshish C, Nair NP et al. (1998). Cortisol levels during human aging predict hippocampal atrophy and memory deficits. Nat Neurosci 1: 69–73.
    1. Lupien SJ, McEwen BS, Gunnar MR, Heim C (2009). Effects of stress throughout the lifespan on the brain, behavior and cognition. Nat Rev Neurosci 10: 434–445.
    1. MacLullich AM, Deary IJ, Starr JM, Ferguson KJ, Wardlaw JM, Seckl JR (2005). Plasma cortisol levels, brain volumes and cognition in healthy elderly men. Psychoneuroendocrinology 30: 505–515.
    1. Marek GJ, Katz DA, Meier A, Greco N, Zhang W, Liu W et al. (2014). Efficacy and safety evaluation of HSD‐1 inhibitor ABT‐384 in Alzheimer's disease. Alzheimers Dement 10: S364–S373.
    1. McGrath JC, Lilley E (2015). Implementing guidelines on reporting research using animals (ARRIVE etc.): new requirements for publication in BJP. Br J Pharmacol 172: 3189–3193.
    1. Mohler EG, Browman KE, Roderwald VA, Cronin EA, Markosyn S, Bitner RS et al. (2011). Acute inhibition of 11β‐hydroxysteroid dehydrogenase type‐1 improves memory in rodent models of cognition. J Neurosci 31: 5406–5413.
    1. Peskind ER, Wilkinson CW, Petrie EC, Schellenberg GD, Raskind MA (2001). Increased CSF cortisol in AD is a function of APOE genotype. Neurology 56: 1094–1098.
    1. Popp J, Wolfsgruber S, Heuser I, Peters O, Hüll M, Schröder J et al. (2015). Cerebrospinal fluid cortisol and clinical disease progression in MCI and dementia of Alzheimer's type. Neurobiol Aging 36: 601–607.
    1. Reynolds RM, Strachan MW, Labad J, Lee AJ, Frier BM, Fowkes FG et al. (2010). Morning cortisol levels and cognitive abilities in people with type 2 diabetes: the Edinburgh type 2 diabetes study. Diabetes Care 33: 714–720.
    1. Reynolds RM, Webster SP (2015). Translational research in stress neuroendocrinology In: Russell JA, Shipston MJ. (eds). Neuroendocrinology of Stress, First edn. John Wiley & Sons: Singapore.
    1. Rosenstock J, Banarer S, Fonseca VA, Inzucchi SE, Sun W, Yao W et al. (2010). The 11β‐hydroxysteroid dehydrogenase type 1 inhibitor INCB13739 improves hyperglycemia in patients with type 2 diabetes inadequately controlled by metformin monotherapy. Diabetes Care 33: 1516–1522.
    1. Sandeep TC, Yau JLW, MacLullich AM, Noble J, Deary IJ, Walker BR et al. (2004). 11β‐hydroxysteroid dehydrogenase inhibition improves cognitive function in healthy elderly men and type 2 diabetics. Proc Natl Acad Sci U S A 101: 6734–6739.
    1. Shah S, Hermanowski‐Vosatka A, Gibson K, Ruck RA, Jia G, Zhang J et al. (2011). Efficacy and safety of the selective 11β‐HSD‐1 inhibitors MK‐0736 and MK‐0916 in overweight and obese patients with hypertension. J Am Soc Hypertens 5: 166–176.
    1. Sooy K, Webster SP, Noble J, Binnie M, Walker BR, Seckl JR et al. (2010). Partial deficiency or short term inhibition of 11β‐hydroxysteroid dehydrogenase type 1 improves cognitive function in aging mice. J Neurosci 30: 13867–13872.
    1. Sooy K, Noble J, McBride A, Binnie M, Yau JL, Seckl JR et al. (2015). Cognitive and disease‐modifying effects of 11ß‐hydroxysteroid dehydrogenase type 1 inhibition in male Tg2576 mice, a model of Alzheimer's disease. Endocrinology 156: 4592–4603.
    1. Southan C, Sharman JL, Benson HE, Faccenda E, Pawson AJ, Alexander SP et al. (2016). The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucl. Acids Res. 44: D1054–D1068.
    1. Starkman MN, Giordani B, Gebarski SS, Berent S, Schork MA, Schteingart DE (1999). Decrease in cortisol reverses human hippocampal atrophy following treatment of Cushing's disease. Biol Psychiatry 46: 1595–1602.
    1. Wager TT, Hou X, Verhoest PR, Villalobos A (2010). Moving beyond rules: the development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chem Nerosci 1: 435–449.
    1. Webster SP, Seckl JR, Walker BR, Ward P, Pallin TD, Dyke HJ et al. (2011a). (4‐Phenyl‐piperidin‐1‐yl)‐[5‐(1H‐pyrazol‐4‐yl)‐thiophen‐3‐yl]‐methanone Compounds and Their Use. Int PCT Application WO2011033255.
    1. Webster SP, Seckl JR, Walker BR, Ward P, Pallin TD, Dyke HJ et al. (2011b). 3,3‐Disubstituted‐(8‐aza‐bicyclo[3.2.1]oct‐8‐yl)‐[5‐(1 h‐pyrazol‐4‐yl)‐thiophen‐3‐yl]‐methanone and related compounds and their use. Int PCT Application WO2011135276.
    1. Yau JL, Noble J, Kenyon CJ, Hibberd C, Kotelevtsev Y, Mullins JJ et al. (2001). Lack of tissue glucocorticoid reactivation in 11beta‐hydroxysteroid dehydrogenase type 1 knockout mice ameliorates age‐related learning impairments. Proc Natl Acad Sci U S A 98: 4716–4721.
    1. Yau JLW, Wheelan N, Noble J, Walker BR, Webster SP, Kenyon CJ et al. (2014). Intrahippocampal glucocorticoids generated by 11β‐HSD1 affect memory in aged mice. Neurobiol Aging 36: 334–343.

Source: PubMed

3
Abonner