Preclinical and Early Clinical Profile of a Highly Selective and Potent Oral Inhibitor of Aldosterone Synthase (CYP11B2)

Katrijn Bogman, Dietmar Schwab, Marie-Laure Delporte, Giuseppe Palermo, Kurt Amrein, Susanne Mohr, Maria Cristina De Vera Mudry, Morris J Brown, Philippe Ferber, Katrijn Bogman, Dietmar Schwab, Marie-Laure Delporte, Giuseppe Palermo, Kurt Amrein, Susanne Mohr, Maria Cristina De Vera Mudry, Morris J Brown, Philippe Ferber

Abstract

Primary hyperaldosteronism is a common cause of resistant hypertension. Aldosterone is produced in the adrenal by aldosterone synthase (AS, encoded by the gene CYP11B2). AS shares 93% homology to 11β-hydroxylase (encoded by the gene CYP11B1), responsible for cortisol production. This homology has hitherto impeded the development of a drug, which selectively suppresses aldosterone but not cortisol production, as a new treatment for primary hyperaldosteronism. We now report the development of RO6836191 as a potent (Ki 13 nmol/L) competitive inhibitor of AS, with in vitro selectivity >100-fold over 11β-hydroxylase. In cynomolgus monkeys challenged with synthetic adrenocorticotropic hormone, single doses of RO6836191 inhibited aldosterone synthesis without affecting the adrenocorticotropic hormone-induced rise in cortisol. In repeat-dose toxicity studies in monkeys, RO6836191 reproduced the adrenal changes of the AS-/- mouse: expansion of the zona glomerulosa; increased expression of AS (or disrupted green fluorescent protein gene in the AS-/- mouse); hypertrophy, proliferation, and apoptosis of zona glomerulosa cells. These changes in the monkey were partially reversible and partially preventable by electrolyte supplementation and treatment with an angiotensin-converting enzyme inhibitor. In healthy subjects, single doses of RO6836191, across a 360-fold dose range, reduced plasma and urine aldosterone levels with maximum suppression at a dose of 10 mg, but unchanged cortisol, on adrenocorticotropic hormone challenge, up to 360 mg, and increase in the precursors 11-deoxycorticosterone and 11-deoxycortisol only at or >90 mg. In conclusion, RO6836191 demonstrates that it is possible to suppress aldosterone production completely in humans without affecting cortisol production.

Clinical trial registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01995383.

Keywords: CYP11B2; adrenal cortex; aldosterone; cytochrome P-450; hydrocortisone; hyperaldosteronism; zona glomerulosa.

© 2016 The Authors.

Figures

Figure 1.
Figure 1.
Histopathologic changes in the cynomolgus monkey zona glomerulosa in the high-dose (A) and mechanistic (B) safety studies. A, Histopathology of representative monkeys in the high-dose study. Animals were administered RO6836191 at 0, 1, 7, or 40 mg/kg for 4 weeks and recovery after 40 mg/kg. Staining with hematoxylin-eosin (upper); immunostaining with human anti-CYP11B2 antibody to visualize the zona glomerulosa (lower). Scale bar=100 µm. Dotted lines delineate the demarcation between the zona glomerulosa from the underlying zonal fasciculata. B, Histopathology of representative monkeys in the mechanistic study. Animals were administered RO6836191 at 1 mg/kg for 4 weeks, with or without electrolytes (E) or angiotensin-converting enzyme inhibitor lisinopril 0.5 mg/kg (A), recovery after treatment with 1 mg/kg with electrolytes (E*), control monkeys were administered electrolytes alone. Staining with hematoxylin-eosin (scale bar=100 µm; upper), double-sided arrows delineate width of the zona glomerulosa (ZG), single-sided arrows indicate the transitional zone of the ZG. Immunostaining with human anti-CYP11B2 antibody to visualize the zona glomerulosa (scale bar=50 µm; lower).
Figure 2.
Figure 2.
Dose-dependent effect of RO6836191 on plasma aldosterone, cortisol, and precursors in cynomolgus monkeys. Data represent individual % change from vehicle in plasma concentration (measured at 1 hour post-adrenocorticotropic hormone challenge) for aldosterone and precursors (A) and cortisol and precursors (B) by dose group. Means by dose group are connected by a line (n=2/dose group). 11-DOC indicates 11-deoxycorticosterone.
Figure 3.
Figure 3.
Dose-dependent effect of RO6836191 on plasma aldosterone, cortisol, and precursors in healthy subjects (study part 1). Aldosterone (A) and cortisol (B) plasma concentration-time profiles after single ascending doses of RO6836191 (0–360 mg) administered orally to healthy subjects 1 hour before the adrenocorticotropic hormone (ACTH) challenge. Downward arrow indicates time of ACTH challenge (1 hour postdose) and upward arrow time of postural change (6 hours postdose). Data represent mean plasma concentrations (+SD) by dose group (n=6/dose group and n=12 for placebo). Below limit of quantification (BLQ) values were set to lower limit of quantification (LLOQ; 5 pg/mL), for doses >10 mg, aldosterone concentrations were BLQ from 2.5 hours postdose onward. Effect of RO6836191 on plasma aldosterone (C), cortisol (D), and precursors after a single oral dose (0–360 mg) in healthy subjects administered 1 hour before the ACTH challenge. Data represent mean (and 90% confidence interval) % change from baseline in plasma exposure (AUC0-24) for aldosterone and precursors (left graph) and cortisol and precursors (right graph) by dose group (n=6/dose group and n=12 for placebo). Filled symbols indicate P<0.01 compared with placebo. 11-DOC indicates 11-deoxycorticosterone.
Figure 4.
Figure 4.
Urinary sodium-to-potassium ratio after single-dose administration of 1, 3, 10 mg RO6836191 or placebo to healthy subjects under low-salt diet (study part 2). Box whiskers show interquartile range (box) and min–max (whiskers) for Na/K ratio from 24-hour urine collections, by dose group (n=6/dose group) at baseline (BL), on day 1 and day 2.

References

    1. Calhoun DA. Hyperaldosteronism as a common cause of resistant hypertension. Annu Rev Med. 2013;64:233–247. doi: 10.1146/annurev-med-042711-135929.
    1. Connell JM, Fraser R, MacKenzie S, Davies E. Is altered adrenal steroid biosynthesis a key intermediate phenotype in hypertension? Hypertension. 2003;41:993–999. doi: 10.1161/01.HYP.0000064344.00173.44.
    1. Te Riet L, van Esch JH, Roks AJ, van den Meiracker AH, Danser AH. Hypertension: renin-angiotensin-aldosterone system alterations. Circ Res. 2015;116:960–975. doi: 10.1161/CIRCRESAHA.116.303587.
    1. Hargovan M, Ferro A. Aldosterone synthase inhibitors in hypertension: current status and future possibilities. JRSM Cardiovasc Dis. 2014;3:2048004014522440. doi: 10.1177/2048004014522440.
    1. Falkenstein E, Christ M, Feuring M, Wehling M. Specific nongenomic actions of aldosterone. Kidney Int. 2000;57:1390–1394. doi: 10.1046/j.1523-1755.2000.00980.x.
    1. Brown NJ. Contribution of aldosterone to cardiovascular and renal inflammation and fibrosis. Nat Rev Nephrol. 2013;9:459–469. doi: 10.1038/nrneph.2013.110.
    1. Azizi M, Amar L, Menard J. Aldosterone synthase inhibition in humans. Nephrol Dial Transplant. 2013;28:36–43. doi: 10.1093/ndt/gfs388.
    1. Bassett MH, White PC, Rainey WE. The regulation of aldosterone synthase expression. Mol Cell Endocrinol. 2004;217:67–74. doi: 10.1016/j.mce.2003.10.011.
    1. Mornet E, Dupont J, Vitek A, White PC. Characterization of two genes encoding human steroid 11 beta-hydroxylase (P-450(11) beta). J Biol Chem. 1989;264:20961–20967.
    1. Hattangady NG, Olala LO, Bollag WB, Rainey WE. Acute and chronic regulation of aldosterone production. Mol Cell Endocrinol. 2012;350:151–162. doi: 10.1016/j.mce.2011.07.034.
    1. Gardner DG, Shoback D. Greenspan’s Basic and Clinical Endocrinology. 8th. New York, NY: McGraw Hill Medical; 2007.
    1. Menard J, Watson C, Rebello S, Zhang YM, Dole WP. Hormonal and electrolyte responses to the aldosterone synthase inhibitor LCI699 in sodium depleted healthy subjects. JACC. 2010;55:A61.E583.
    1. Amar L, Azizi M, Menard J, Peyrard S, Watson C, Plouin PF. Aldosterone synthase inhibition with LCI699: a proof-of-concept study in patients with primary aldosteronism. Hypertension. 2010;56:831–838. doi: 10.1161/HYPERTENSIONAHA.110.157271.
    1. Calhoun DA, White WB, Krum H, Guo W, Bermann G, Trapani A, Lefkowitz MP, Ménard J. Effects of a novel aldosterone synthase inhibitor for treatment of primary hypertension: results of a randomized, double-blind, placebo- and active-controlled phase 2 trial. Circulation. 2011;124:1945–1955. doi: 10.1161/CIRCULATIONAHA.111.029892.
    1. Andersen K, Hartman D, Peppard T, Hermann D, Van Ess P, Lefkowitz M, Trapani A. The effects of aldosterone synthase inhibition on aldosterone and cortisol in patients with hypertension: a phase II, randomized, double-blind, placebo-controlled, multicenter study. J Clin Hypertens (Greenwich) 2012;14:580–587. doi: 10.1111/j.1751-7176.2012.00667.x.
    1. Karns AD, Bral JM, Hartman D, Peppard T, Schumacher C. Study of aldosterone synthase inhibition as an add-on therapy in resistant hypertension. J Clin Hypertens (Greenwich) 2013;15:186–192. doi: 10.1111/jch.12051.
    1. Schumacher CD, Steele RE, Brunner HR. Aldosterone synthase inhibition for the treatment of hypertension and the derived mechanistic requirements for a new therapeutic strategy. J Hypertens. 2013;31:2085–2093. doi: 10.1097/HJH.0b013e328363570c.
    1. Bertagna X, Pivonello R, Fleseriu M, Zhang Y, Robinson P, Taylor A, Watson CE, Maldonado M, Hamrahian AH, Boscaro M, Biller BM. LCI699, a potent 11β-hydroxylase inhibitor, normalizes urinary cortisol in patients with Cushing’s disease: results from a multicenter, proof-of-concept study. J Clin Endocrinol Metab. 2014;99:1375–1383. doi: 10.1210/jc.2013-2117.
    1. Heath V. Pharmacotherapy: a new option for Cushing disease? Nat Rev Endocrinol. 2014;10:127. doi: 10.1038/nrendo.2013.266.
    1. Martin RE, Aebi JD, Hornsperger B, et al. Discovery of 4-Aryl-5,6,7,8-tetrahydroisoquinolines as potent, selective, and orally active aldosterone synthase (CYP11B2) inhibitors: in vivo evaluation in rodents and cynomolgus monkeys. J Med Chem. 2015;58:8054–8065. doi: 10.1021/acs.jmedchem.5b00851.
    1. Cerny MA. Progress towards clinically useful aldosterone synthase inhibitors. Curr Top Med Chem. 2013;13:1385–1401.
    1. Wolkersdörfer GW, Bornstein SR. Tissue remodelling in the adrenal gland. Biochem Pharmacol. 1998;56:163–171.
    1. Papillon JP, Lou C, Singh AK, et al. Discovery of N-[5-(6-Chloro-3-cyano-1-methyl-1H-indol-2-yl)-pyridin-3-ylmethyl]-ethanesulfonamide, a cortisol-sparing CYP11B2 inhibitor that lowers aldosterone in human subjects. J Med Chem. 2015;58:9382–9394. doi: 10.1021/acs.jmedchem.5b01545.
    1. Aoyama H, Asaishi K, Abe R, Kajiwara T, Enomoto K, Yoshida M, Ohasi Y, Tominaga T, Abe O. [Clinical evaluation of CGS16949A in advanced or recurrent breast cancer—a multi-institutional late phase II clinical trial]. Gan To Kagaku Ryoho. 1994;21:477–484.
    1. Wada T, Nomura Y, Oohashi Y, Abe O, Koyama H, Takashima S. [Late phase II study of CGS16949A, a new aromatase inhibitor—a multicentral cooperative study (Western Japan Group)]. Gan To Kagaku Ryoho. 1994;21:485–493.
    1. Lee G, Makhanova N, Caron K, Lopez ML, Gomez RA, Smithies O, Kim HS. Homeostatic responses in the adrenal cortex to the absence of aldosterone in mice. Endocrinology. 2005;146:2650–2656. doi: 10.1210/en.2004-1102.
    1. Eudy RJ, Sahasrabudhe V, Sweeney K, Tugnait M, King-Ahmad A, Near K, Loria P, Banker ME, Piotrowski DW, Boustany-Kari CM. The use of plasma aldosterone and urinary sodium to potassium ratio as translatable quantitative biomarkers of mineralocorticoid receptor antagonism. J Transl Med. 2011;9:180. doi: 10.1186/1479-5876-9-180.
    1. Croom KF, Perry CM. Eplerenone: a review of its use in essential hypertension. Am J Cardiovasc Drugs. 2005;5:51–69.
    1. Pitt B, Kober L, Ponikowski P, Gheorghiade M, Filippatos G, Krum H, Nowack C, Kolkhof P, Kim SY, Zannad F. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J. 2013;34:2453–2463. doi: 10.1093/eurheartj/eht187.
    1. Berger S, Bleich M, Schmid W, Cole TJ, Peters J, Watanabe H, Kriz W, Warth R, Greger R, Schütz G. Mineralocorticoid receptor knockout mice: pathophysiology of Na+ metabolism. Proc Natl Acad Sci U S A. 1998;95:9424–9429.

Source: PubMed

3
Prenumerera