1
|
Meguro M, Miyauchi S, Kanao-Arisumi Y, Naito S, Suzuki K, Inoue S, Yamada K, Homma T, Chiba K, Nara F, Furuzono S. Identification of sulfonylpyrimidines as novel selective aldosterone synthase (CYP11B2) inhibitors. Bioorg Med Chem 2024; 108:117775. [PMID: 38851000 DOI: 10.1016/j.bmc.2024.117775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
4-[(5-[2-Methyl-5-(methylsulfonyl)pentan-2-yl]sulfonylpyrimidin-4-yl)amino]benzonitrile 2 was identified as a novel potent aldosterone synthase inhibitor. Compound 2 was found to inhibit human CYP11B2 in the nanomolar range, and showed an aldosterone-lowering effect in a furosemide-treated cynomolgus monkey model. Although human CYP11B2 has the high homology sequence with human CYP11B1, compound 2 showed more than 80 times higher selectivity over human CYP11B1 in vitro.
Collapse
Affiliation(s)
- Masaki Meguro
- Shinagawa R&D Center, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | - Satoru Miyauchi
- Technology Division, Technology Business Management Group, Daiichi Sankyo Co., Ltd., 1-12-1, Shinomiya, Hiratsuka-shi, Kanagawa 254-0014, Japan
| | - Yukiko Kanao-Arisumi
- Pharmaunion Co., Ltd., 1-23-39 Hiikawa, Jounan-ku, Fukuoka-shi, Fukuoka 814-0153, Japan
| | - Satoru Naito
- Site Operations Department, Shinagawa Site Operation Group, Daiichi Sankyo Business Associe Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kanae Suzuki
- Shinagawa R&D Center, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shinichi Inoue
- Daiichi Sankyo Inc., 211 Mt. Airy Road, Basking Ridge, NJ 07920, USA
| | - Keisuke Yamada
- Medical Affairs Division, Medical Affairs Planning Department, Daiichi Sankyo Co., Ltd., 3-5-1, Nihonbashi-Honcho, Chuo-ku, Tokyo 103-8426, Japan
| | - Tsuyoshi Homma
- Shinagawa R&D Center, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kiyoshi Chiba
- Shinagawa R&D Center, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Futoshi Nara
- Shin Nippon Biomedical Laboratories, Ltd., 8-1-28, Akashicho, Chuo-ku, Tokyo 104-0044, Japan
| | - Shinji Furuzono
- Shinagawa R&D Center, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
2
|
Tinivella A, Banchi M, Gambacorta G, Borghi F, Orlandi P, Baxendale IR, Di Paolo A, Bocci G, Pinzi L, Rastelli G. Discovery of a Potent Dual Inhibitor of Aromatase and Aldosterone Synthase. ACS Pharmacol Transl Sci 2023; 6:1870-1883. [PMID: 38093846 PMCID: PMC10714424 DOI: 10.1021/acsptsci.3c00183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2024]
Abstract
Estrogen deficiency derived from inhibition of estrogen biosynthesis is a typical condition of postmenopausal women and breast cancer (BCs) patients undergoing antihormone therapy. The ensuing increase in aldosterone levels is considered to be the major cause for cardiovascular diseases (CVDs) affecting these patients. Since estrogen biosynthesis is regulated by aromatase (CYP19A1), and aldosterone biosynthesis is modulated by aldosterone synthase (CYP11B2), a dual inhibitor would allow the treatment of BC while reducing the cardiovascular risks typical of these patients. Moreover, this strategy would help overcome some of the disadvantages often observed in single-target or combination therapies. Following an in-depth analysis of a library of synthesized benzylimidazole derivatives, compound X21 was found to be a potent and selective dual inhibitor of aromatase and aldosterone synthase, with IC50 values of 2.3 and 29 nM, respectively. Remarkably, the compound showed high selectivity with respect to 11β-hydroxylase (CYP11B1), as well as CYP3A4 and CYP1A2. When tested in cells, X21 showed potent antiproliferative activity against BC cell lines, particularly against the ER+ MCF-7 cells (IC50 of 0.26 ± 0.03 μM at 72 h), and a remarkable pro-apoptotic effect. In addition, the compound significantly inhibited mTOR phosphorylation at its IC50 concentration, thereby negatively modulating the PI3K/Akt/mTOR axis, which represents an escape for the dependency from ER signaling in BC cells. The compound was further investigated for cytotoxicity on normal cells and potential cardiotoxicity against hERG and Nav1.5 ion channels, demonstrating a safe biological profile. Overall, these assays demonstrated that the compound is potent and safe, thus constituting an excellent candidate for further evaluation.
Collapse
Affiliation(s)
- Annachiara Tinivella
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via G. Campi, Modena 41125, Italy
| | - Marta Banchi
- Department
of Clinical and Experimental Medicine, University
of Pisa, Via Roma 55, Pisa 56126, Italy
| | - Guido Gambacorta
- Department
of Chemistry, University of Durham, Lower Mount Joy, South Rd, Durham DH1 3LE, U.K.
| | - Federica Borghi
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via G. Campi, Modena 41125, Italy
| | - Paola Orlandi
- Department
of Clinical and Experimental Medicine, University
of Pisa, Via Roma 55, Pisa 56126, Italy
| | - Ian R. Baxendale
- Department
of Chemistry, University of Durham, Lower Mount Joy, South Rd, Durham DH1 3LE, U.K.
| | - Antonello Di Paolo
- Department
of Clinical and Experimental Medicine, University
of Pisa, Via Roma 55, Pisa 56126, Italy
| | - Guido Bocci
- Department
of Clinical and Experimental Medicine, University
of Pisa, Via Roma 55, Pisa 56126, Italy
| | - Luca Pinzi
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via G. Campi, Modena 41125, Italy
| | - Giulio Rastelli
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via G. Campi, Modena 41125, Italy
| |
Collapse
|
3
|
Yu Y, Liu A, Dhawan G, Mei H, Zhang W, Izawa K, Soloshonok VA, Han J. Fluorine-containing pharmaceuticals approved by the FDA in 2020: Synthesis and biological activity. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.05.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
4
|
Kolkhof P, Joseph A, Kintscher U. Nonsteroidal mineralocorticoid receptor antagonism for cardiovascular and renal disorders - New perspectives for combination therapy. Pharmacol Res 2021; 172:105859. [PMID: 34461222 DOI: 10.1016/j.phrs.2021.105859] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023]
Abstract
During the recent 30 years, there has been a dramatic increase in knowledge about the role of aldosterone and the mineralocorticoid receptor (MR) in the pathophysiology of cardiovascular (CV) and kidney diseases. The scientific perspective on the aldosterone/MR ensemble extended from a previously renal epithelial-centered focus on sodium-potassium exchange to a broader view as systemic modulators of extracellular matrix, inflammation and fibrosis. Spironolactone was launched as the first antagonist of aldosterone 27 years before the MR was cloned. It was classified as a potassium-sparing diuretic, based on its initial clinical characterization as a diuretic and its preferred activity to compensate for the potassium loss induced by loop diuretics when used in combination. The second steroidal MR antagonist was eplerenone which was discovered at a time when the role of aldosterone and MR in cardiac fibrosis was rediscovered. The constraint of developing potentially life-threatening hyperkalaemia when used in combination with other inhibitors of the renin-angiotensin-system (RAS) in patients with reduced kidney function initiated extensive research and development activities with the goal to identify novel nonsteroidal MR antagonists with an improved benefit-risk ratio. Here we summarize major current clinical trials with MRAs in different CV and renal diseases. Addition of the nonsteroidal MRA finerenone to optimal RAS blockade recently reduced CV and kidney outcomes in two large phase III trials in patients with chronic kidney disease (CKD) and type 2 diabetes (T2D). We provide an outlook on further opportunities for combination therapy of nonsteroidal MRA finerenone with RAS inhibitors and sodium-glucose cotransporter-2 inhibitors (SGLT2i).
Collapse
Affiliation(s)
- Peter Kolkhof
- Cardiovascular Research, Research and Early Development, R&D Pharmaceuticals, Bayer AG, Wuppertal, Germany.
| | - Amer Joseph
- Cardiology and Nephrology, Clinical Development, R&D Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Ulrich Kintscher
- Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal Research Center, 10115 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
5
|
Wu J, Ding X, Tan X. A patent review of aldosterone synthase inhibitors (2014-present). Expert Opin Ther Pat 2021; 32:13-28. [PMID: 34365871 DOI: 10.1080/13543776.2021.1965991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Aldosterone synthase (AS) is a key enzyme involved in the final three rate-limiting steps of the biosynthesis pathway of aldosterone, and its inhibition has been considered as an effective strategy to treat hypertension, heart failure, and related cardio-metabolic diseases. AREA COVERED This review provides an update on the discovery and development of aldosterone synthase inhibitors by means of patents published between January 2014 and March 2021. The molecules are classified by pharmaceutical company with progress that has been made in clinical trials being highlighted. EXPERT OPINION Mineralocorticoid receptor antagonists (MRAs) and aldosterone synthase inhibitors (ASI) represent two of the main approaches for the blockade of aldosterone. Clinical success, as well as foreseen side effects of steroidal MRAs, prompted the discovery and development of ASI. Since the observation of decreased cortisol levels in clinical trials for LCI699, subsequent efforts have been largely focused on improving its selectivity over hCYP11B1. Candidates with improved potency and selectivity are under investigation across a wide range of indications. Whether ASI will provide an additional therapeutic advantage over current safe and selective non-steroidal MRAs is highly anticipated.
Collapse
Affiliation(s)
- Jun Wu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai, China
| | - Xiao Ding
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai, China
| | - Xuefei Tan
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai, China
| |
Collapse
|
6
|
New drug targets for hypertension: A literature review. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166037. [PMID: 33309796 DOI: 10.1016/j.bbadis.2020.166037] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Hypertension is one of the most prevalent cardiovascular diseases worldwide. However, in the population of resistant hypertension, blood pressure is difficult to control effectively. Moreover, antihypertensive drugs may have adverse effect currently. Hence, new therapeutic targets and treatments are needed to uncovered and exploited to control hypertension and its comorbidities. In the past, classical drug targets, such as the aldosterone receptor, aldosterone synthase, and ACE2/angiotensin 1-7/Mas receptor axis, have been investigated. Recently, vaccines and drugs targeting the gastrointestinal microbiome, which represent drug classes, have also been investigated for the management of blood pressure. In this review, we summarized current knowledge on classical and new drug targets and discussed the potential utility of new drugs in the treatment of hypertension.
Collapse
|
7
|
Abstract
The mineralocorticoid aldosterone is an important regulator of blood pressure and electrolyte balance. However, excess aldosterone can be deleterious as a driver of inflammation, vascular remodeling and tissue fibrosis associated with cardiometabolic diseases. Mineralocorticoid receptor antagonists (MRA) and renin-angiotensin-aldosterone system (RAAS) antagonists are current clinical therapies used to antagonize deleterious effects of aldosterone in patients. MRAs compete with aldosterone for binding at its cognate receptor thereby limiting its effect while RAS antagonists reduce aldosterone levels indirectly by blocking the stimulatory effect of angiotensin. Both MRAs and RAS antagonists can result in incomplete inhibition of the harmful effects of excess aldosterone. Aldosterone synthase (AS) inhibitors (ASI) attenuate the production of aldosterone directly and have been proposed as an alternative to MRAs and RAS blockers. Cortisol synthase (CS) is an enzyme closely related to AS and responsible for generating the important glucocorticoid cortisol, required for maintaining critical metabolic and immune responses. The importance of selectivity against CS is shown by early examples of ASIs that were only modestly selective and as such, attenuated cortisol responses when evaluated in patients. Recently, next-generation, highly selective ASIs have been described and are presently being evaluated in the clinic as an alternative to angiotensin and MR antagonists for cardiometabolic disease. Herein we provide a brief review of the challenges associated with discovery of selective ASIs and the transition from the early compounds that paved the way toward the next-generation of highly selective ASIs currently under development.
Collapse
Affiliation(s)
- Steven M Weldon
- Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States.
| | - Nicholas F Brown
- Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| |
Collapse
|
8
|
Meguro M, Miyauchi S, Kanao Y, Naito S, Suzuki K, Inoue S, Yamada K, Homma T, Chiba K, Nara F, Furuzono S. 4-Anilino-pyrimidine, novel aldosterone synthase (CYP11B2) inhibitors bearing pyrimidine structures. Bioorg Med Chem Lett 2017; 27:1902-1906. [DOI: 10.1016/j.bmcl.2017.03.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 01/20/2023]
|
9
|
Weldon SM, Cerny MA, Gueneva-Boucheva K, Cogan D, Guo X, Moss N, Parmentier JH, Richman JR, Reinhart GA, Brown NF. Selectivity of BI 689648, a Novel, Highly Selective Aldosterone Synthase Inhibitor: Comparison with FAD286 and LCI699 in Nonhuman Primates. J Pharmacol Exp Ther 2016; 359:142-50. [PMID: 27481775 DOI: 10.1124/jpet.116.236463] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/29/2016] [Indexed: 03/08/2025] Open
Abstract
The mineralocorticoid aldosterone is an important regulator of blood pressure, volume, and electrolyte balance. However, excess aldosterone can be deleterious as a driver of vascular remodeling and tissue fibrosis associated with cardiometabolic diseases. Aldosterone synthase (AS) inhibitors (ASI) attenuate the production of aldosterone directly and have been proposed as an alternative to mineralocorticoid receptor antagonists for blocking the pathologic effects of excess aldosterone. Discovery of selective ASIs has been challenging because of the high sequence identity (93%) AS shares with cortisol synthase (CS), and the low identity of rodent AS compared with human (63%). Using cynomolgus (cyno) monkey-based models, we identified BI 689648 [6-(5-methoxymethyl-pyridin-3-yl)-3,4-dihydro-2H-[1,8]naphthyridine-1-carboxylic acid amide], a novel, highly selective ASI that exhibits an in vitro IC50 of 2 nM against AS and 300 nm against CS (150-fold selectivity) compared with the recently described ASIs FAD286 [4-(5,6,7,8-tetrahydroimidazo[1,5-a]pyridin-5-yl)benzonitrile] (3 nM AS; 90 nM CS; 40-fold) and LCI699 (4-[(5R)-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl]-3-fluorobenzonitrile) (10 nM AS; 80 nM CS; 8-fold). After oral administration in cyno monkeys, BI 689648 (5 mg/kg) exhibits a peak plasma concentration of ∼500 nM. For in vivo profiling we used an adrenocorticotropin-challenge model in which BI 689648 was >20-fold more selective compared with FAD286 and LCI699. Because both FAD286 and LCI699 failed to provide adequate selectivity for CS when tested in patients, the desire for more selective molecules to test the ASI hypothesis remains high. Therefore, highly selective aldosterone synthase inhibitors such as BI 689648 represent an important step forward toward developing ASIs with greater potential for clinical success in cardiometabolic diseases.
Collapse
Affiliation(s)
- Steven M Weldon
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Matthew A Cerny
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Kristina Gueneva-Boucheva
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Derek Cogan
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Xin Guo
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Neil Moss
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Jean-Hugues Parmentier
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Jeremy R Richman
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Glenn A Reinhart
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Nicholas F Brown
- CardioMetabolic Diseases Research (S.M.W., J-H. P., J.R.R., G.A.R., N.F.B.), and Medicinal Chemistry (M.A.C., K.G.B., D.C., X.G., N.M.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
10
|
Dinh Cat AN, Friederich-Persson M, White A, Touyz RM. Adipocytes, aldosterone and obesity-related hypertension. J Mol Endocrinol 2016; 57:F7-F21. [PMID: 27357931 DOI: 10.1530/jme-16-0025] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 12/15/2022]
Abstract
Understanding the mechanisms linking obesity with hypertension is important in the current obesity epidemic as it may improve therapeutic interventions. Plasma aldosterone levels are positively correlated with body mass index and weight loss in obese patients is reported to be accompanied by decreased aldosterone levels. This suggests a relationship between adipose tissue and the production/secretion of aldosterone. Aldosterone is synthesized principally by the adrenal glands, but its production may be regulated by many factors, including factors secreted by adipocytes. In addition, studies have reported local synthesis of aldosterone in extra-adrenal tissues, including adipose tissue. Experimental studies have highlighted a role for adipocyte-secreted aldosterone in the pathogenesis of obesity-related cardiovascular complications via the mineralocorticoid receptor. This review focuses on how aldosterone secretion may be influenced by adipose tissue and the importance of these mechanisms in the context of obesity-related hypertension.
Collapse
Affiliation(s)
- Aurelie Nguyen Dinh Cat
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Malou Friederich-Persson
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Anna White
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| |
Collapse
|
11
|
Affiliation(s)
- Rita Bernhardt
- Lehrstuhl für Biochemie, Universität des Saarlandes, Saarbrücken, Germany
| |
Collapse
|
12
|
Papillon JPN, Lou C, Singh AK, Adams CM, Ksander GM, Beil ME, Chen W, Leung-Chu J, Fu F, Gan L, Hu CW, Jeng AY, LaSala D, Liang G, Rigel DF, Russell KS, Vest JA, Watson C. 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; 58:9382-94. [PMID: 26540564 DOI: 10.1021/acs.jmedchem.5b01545] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human clinical studies conducted with LCI699 established aldosterone synthase (CYP11B2) inhibition as a promising novel mechanism to lower arterial blood pressure. However, LCI699's low CYP11B1/CYP11B2 selectivity resulted in blunting of adrenocorticotropic hormone-stimulated cortisol secretion. This property of LCI699 prompted its development in Cushing's disease, but limited more extensive clinical studies in hypertensive populations, and provided an impetus for the search for cortisol-sparing CYP11B2 inhibitors. This paper summarizes the discovery, pharmacokinetics, and pharmacodynamic data in preclinical species and human subjects of the selective CYP11B2 inhibitor 8.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael E Beil
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Wei Chen
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Jennifer Leung-Chu
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Fumin Fu
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | | | - Chii-Whei Hu
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Arco Y Jeng
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Daniel LaSala
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | | | - Dean F Rigel
- Cardiovascular and Metabolism, Novartis Institutes for BioMedical Research , One Health Plaza, East Hanover, New Jersey 07936, United States
| | | | | | | |
Collapse
|
13
|
Papillon JPN, Adams CM, Hu QY, Lou C, Singh AK, Zhang C, Carvalho J, Rajan S, Amaral A, Beil ME, Fu F, Gangl E, Hu CW, Jeng AY, LaSala D, Liang G, Logman M, Maniara WM, Rigel DF, Smith SA, Ksander GM. Structure–Activity Relationships, Pharmacokinetics, and in Vivo Activity of CYP11B2 and CYP11B1 Inhibitors. J Med Chem 2015; 58:4749-70. [DOI: 10.1021/acs.jmedchem.5b00407] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Julien P. N. Papillon
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Christopher M. Adams
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Qi-Ying Hu
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Changgang Lou
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Alok K. Singh
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Chun Zhang
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jose Carvalho
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Srinivan Rajan
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Adam Amaral
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Michael E. Beil
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Fumin Fu
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Eric Gangl
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Chii-Whei Hu
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Arco Y. Jeng
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Daniel LaSala
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Guiqing Liang
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Michael Logman
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Wieslawa M. Maniara
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Dean F. Rigel
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Sherri A. Smith
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Gary M. Ksander
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
14
|
Abstract
Hypertension is the most common modifiable risk factor for cardiovascular disease and death, and lowering blood pressure with antihypertensive drugs reduces target organ damage and prevents cardiovascular disease outcomes. Despite a plethora of available treatment options, a substantial portion of the hypertensive population has uncontrolled blood pressure. The unmet need of controlling blood pressure in this population may be addressed, in part, by developing new drugs and devices/procedures to treat hypertension and its comorbidities. In this Compendium Review, we discuss new drugs and interventional treatments that are undergoing preclinical or clinical testing for hypertension treatment. New drug classes, eg, inhibitors of vasopeptidases, aldosterone synthase and soluble epoxide hydrolase, agonists of natriuretic peptide A and vasoactive intestinal peptide receptor 2, and a novel mineralocorticoid receptor antagonist are in phase II/III of development, while inhibitors of aminopeptidase A, dopamine β-hydroxylase, and the intestinal Na
+
/H
+
exchanger 3, agonists of components of the angiotensin-converting enzyme 2/angiotensin(1–7)/Mas receptor axis and vaccines directed toward angiotensin II and its type 1 receptor are in phase I or preclinical development. The two main interventional approaches, transcatheter renal denervation and baroreflex activation therapy, are used in clinical practice for severe treatment resistant hypertension in some countries. Renal denervation is also being evaluated for treatment of various comorbidities, eg, chronic heart failure, cardiac arrhythmias and chronic renal failure. Novel interventional approaches in early development include carotid body ablation and arteriovenous fistula placement. Importantly, none of these novel drug or device treatments has been shown to prevent cardiovascular disease outcomes or death in hypertensive patients.
Collapse
Affiliation(s)
- Suzanne Oparil
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama (S.O.); and Department of Nephrology and Hypertension, University Hospital of the University Erlangen-Nürnberg, Germany (R.E.S.)
| | - Roland E. Schmieder
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama (S.O.); and Department of Nephrology and Hypertension, University Hospital of the University Erlangen-Nürnberg, Germany (R.E.S.)
| |
Collapse
|
15
|
Namsolleck P, Unger T. Aldosterone synthase inhibitors in cardiovascular and renal diseases. Nephrol Dial Transplant 2014; 29 Suppl 1:i62-i68. [PMID: 24493871 DOI: 10.1093/ndt/gft402] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aldosterone is involved in various cardiovascular pathologies, including hypertension, heart failure, atherosclerosis and fibrosis. Mineralocorticoid receptor (MR)-dependent and -independent, genomic and non-genomic processes mediate its complex effects. Spironolactone and eplerenone, both MR antagonists, are the only commercially available compounds targeting directly the actions of aldosterone. However, due to the poor selectivity (spironolactone), low potency (eplerenone) and the fact that only MR-dependent effects of aldosterone can be inhibited, these drugs have limited clinical use. An attractive approach to abolish potentially all of aldosterone-mediated pathologies is the inhibition of aldosterone synthase. This review summarizes current knowledge on the complex effects mediated by aldosterone, potential advantages and disadvantages of aldosterone inhibition and novel directions in the development of aldosterone synthase inhibitors.
Collapse
Affiliation(s)
- Pawel Namsolleck
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
16
|
Aldosterone synthase inhibition for the treatment of hypertension and the derived mechanistic requirements for a new therapeutic strategy. J Hypertens 2014; 31:2085-93. [PMID: 24107737 PMCID: PMC3771574 DOI: 10.1097/hjh.0b013e328363570c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Context: We describe the clinical investigation of the first generation aldosterone synthase inhibitor, LCI699, in patients with essential, uncontrolled, resistant, or secondary hypertension. LCI699 competitively reduced blood pressure at lower doses yet counterintuitive effects were observed at higher doses. Objective and method: An extensive endocrine biomarker analysis was performed to better understand the pharmacological mechanism of the drug. Results: The interference of LCI699 in the renin–angiotensin–aldosterone system occurred with limited target selectivity, as a dose-dependent compensatory stimulation of the hypothalamic-pituitary-adrenal feedback axis was discovered. Thus, LCI699 affected two endocrine feedback loops that converged at a single point, inhibiting the 11β-hydroxylase reaction in the adrenal gland, leading to supraphysiological levels of 11-deoxycortiscosterone. The accumulation of this potent mineralocorticoid may explain the blunted blood pressure response to LCI699. Conclusion: Future aldosterone synthase inhibitors may improve their target selectivity by sparing the 11β-hydroxylase reaction and preferentially inhibiting one of the two other enzymatic reactions mediated by aldosterone synthase.
Collapse
|
17
|
Meredith EL, Ksander G, Monovich L, Papillon JPN, Liu Q, Miranda K, Morris P, Rao C, Burgis R, Capparelli M, Hu QY, Singh A, Rigel DF, Jeng AY, Beil M, Fu F, Hu CW, LaSala D. Discovery and in Vivo Evaluation of Potent Dual CYP11B2 (Aldosterone Synthase) and CYP11B1 Inhibitors. ACS Med Chem Lett 2013; 4:1203-7. [PMID: 24900631 PMCID: PMC4027133 DOI: 10.1021/ml400324c] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/10/2013] [Indexed: 12/16/2022] Open
Abstract
Aldosterone is a key signaling component of the renin-angiotensin-aldosterone system and as such has been shown to contribute to cardiovascular pathology such as hypertension and heart failure. Aldosterone synthase (CYP11B2) is responsible for the final three steps of aldosterone synthesis and thus is a viable therapeutic target. A series of imidazole derived inhibitors, including clinical candidate 7n, have been identified through design and structure-activity relationship studies both in vitro and in vivo. Compound 7n was also found to be a potent inhibitor of 11β-hydroxylase (CYP11B1), which is responsible for cortisol production. Inhibition of CYP11B1 is being evaluated in the clinic for potential treatment of hypercortisol diseases such as Cushing's syndrome.
Collapse
Affiliation(s)
- Erik L. Meredith
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Gary Ksander
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Lauren
G. Monovich
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Julien P. N. Papillon
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Qian Liu
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Karl Miranda
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Patrick Morris
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Chang Rao
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Robin Burgis
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Michael Capparelli
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Qi-Ying Hu
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Alok Singh
- Novartis
Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Dean F. Rigel
- Novartis
Pharmaceuticals Corporation, East
Hanover, New Jersey 07936, United States
| | - Arco Y. Jeng
- Novartis
Pharmaceuticals Corporation, East
Hanover, New Jersey 07936, United States
| | - Michael Beil
- Novartis
Pharmaceuticals Corporation, East
Hanover, New Jersey 07936, United States
| | - Fumin Fu
- Novartis
Pharmaceuticals Corporation, East
Hanover, New Jersey 07936, United States
| | - Chii-Whei Hu
- Novartis
Pharmaceuticals Corporation, East
Hanover, New Jersey 07936, United States
| | - Daniel LaSala
- Novartis
Pharmaceuticals Corporation, East
Hanover, New Jersey 07936, United States
| |
Collapse
|
18
|
Strushkevich N, Gilep AA, Shen L, Arrowsmith CH, Edwards AM, Usanov SA, Park HW. Structural insights into aldosterone synthase substrate specificity and targeted inhibition. Mol Endocrinol 2013; 27:315-24. [PMID: 23322723 PMCID: PMC5417327 DOI: 10.1210/me.2012-1287] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aldosterone is a major mineralocorticoid hormone that plays a key role in the regulation of electrolyte balance and blood pressure. Excess aldosterone levels can arise from dysregulation of the renin-angiotensin-aldosterone system and are implicated in the pathogenesis of hypertension and heart failure. Aldosterone synthase (cytochrome P450 11B2, CYP11B2) is the sole enzyme responsible for the production of aldosterone in humans. Blocking of aldosterone synthesis by mediating aldosterone synthase activity is thus a recently emerging pharmacological therapy for hypertension, yet a lack of structural information has limited this approach. Here, we present the crystal structures of human aldosterone synthase in complex with a substrate deoxycorticosterone and an inhibitor fadrozole. The structures reveal a hydrophobic cavity with specific features associated with corticosteroid recognition. The substrate binding mode, along with biochemical data, explains the high 11β-hydroxylase activity of aldosterone synthase toward both gluco- and mineralocorticoid formation. The low processivity of aldosterone synthase with a high extent of intermediates release might be one of the mechanisms of controlled aldosterone production from deoxycorticosterone. Although the active site pocket is lined by identical residues between CYP11B isoforms, most of the divergent residues that confer additional 18-oxidase activity of aldosterone synthase are located in the I-helix (vicinity of the O(2) activation path) and loops around the H-helix (affecting an egress channel closure required for retaining intermediates in the active site). This intrinsic flexibility is also reflected in isoform-selective inhibitor binding. Fadrozole binds to aldosterone synthase in the R-configuration, using part of the active site cavity pointing toward the egress channel. The structural organization of aldosterone synthase provides critical insights into the molecular mechanism of catalysis and enables rational design of more specific antihypertensive agents.
Collapse
Affiliation(s)
- Natallia Strushkevich
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada M5G 1L7.
| | | | | | | | | | | | | |
Collapse
|
19
|
Azizi M, Amar L, Menard J. Aldosterone synthase inhibition in humans. Nephrol Dial Transplant 2012; 28:36-43. [PMID: 23045428 DOI: 10.1093/ndt/gfs388] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Aldosterone synthase (CYP11B2) inhibition has emerged as a new option for the treatment of hypertension, heart failure and renal disorders, in addition to mineralocorticoid receptor (MR) blockade. The aim is to decrease aldosterone concentrations in both plasma and tissues, thereby decreasing MR-dependent and MR-independent effects in the cardiac, vascular and renal target organs. LCI699 was the first orally active aldosterone-synthase inhibitor to be developed for human use. Its structure is similar to that of FAD286, the dextroenantiomer of the aromatase inhibitor, fadrozole. It dose-dependently decreases plasma and urine aldosterone concentrations by up to 70 or 80% and increases plasma renin activity in healthy male subjects on a low-sodium diet. LCI699 does not decrease basal plasma cortisol concentrations at doses of 0.5-3 mg q.d., but it blocks the cortisol response to adrenocorticotropic hormone (ACTH) at doses ≥ 3 mg q.d. In a proof-of-concept study in patients with primary aldosteronism (PA), LCI699 (0.5-1 mg b.i.d.) induced a dose-dependent and reversible 70-80% decrease in plasma and urinary aldosterone concentration accompanied by a massive dose-dependent accumulation of deoxycorticosterone (>+700%), the aldosterone precursor, in the plasma, thereby confirming the inhibition of the CYP11B2 gene product. This effect was associated with a rapid correction of hypokalaemia, a modest decrease in blood pressure (BP) and a mild increase in plasma renin concentration in patients with PA. LCI699 administration induced biological signs of partial inhibition of the glucocorticoid axis, such as dose-dependent increases in both plasma ACTH and 11-deoxycortisol (the precursor of cortisol) concentrations, consistent with the inhibition of the CYP11B1 gene product. An 8-week placebo-controlled dose-response study on patients with Stage 1 and 2 essential hypertension reported an optimal decrease in BP with a dose of 1 mg LCI699 q.d., which had an antihypertensive effect similar to that of 50 mg b.i.d. eplerenone. A blunted cortisol response to ACTH was observed in 20% of patients, but the clinical and biological safety and tolerability of LCI699 were similar to those of placebo and eplerenone. The discovery of this first orally active aldosterone synthase inhibitor, LCI699, has provided new opportunities to assess the feasibility and the haemodynamic, biological and safety consequences as well as the limitations of this new approach to block the aldosterone pathway in hypertensive patients. However, as the effects of LCI699 on the glucocorticoid axis limit the use of higher doses range because of the loss of selectivity for CYP11B2, this aldosterone synthase inhibitor cannot replace the MR blockade in patients with hypertension, other cardiovascular or renal disorders. The development of second-generation aldosterone synthase inhibitors with a higher selectivity index for CYP11B2 than LCI699 should make it possible to test this approach at much higher doses in these patients, after the necessary toxicology and Phase I studies.
Collapse
Affiliation(s)
- Michel Azizi
- Faculté de Médecine, The Université Paris Descartes, Paris F-75006, France.
| | | | | |
Collapse
|
20
|
Rigel DF, Fu F, Beil M, Hu CW, Liang G, Jeng AY. Pharmacodynamic and pharmacokinetic characterization of the aldosterone synthase inhibitor FAD286 in two rodent models of hyperaldosteronism: comparison with the 11beta-hydroxylase inhibitor metyrapone. J Pharmacol Exp Ther 2010; 334:232-43. [PMID: 20354176 DOI: 10.1124/jpet.110.167148] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aldosterone synthase (CYP11B2) inhibitors (ASIs) represent an attractive therapeutic approach for mitigating the untoward effects of aldosterone. We characterized the pharmacokinetic/pharmacodynamic relationships of a prototypical ASI, (+)-(5R)-4-(5,6,7,8-tetrahydroimidazo[1,5-a]pyridin-5-yl]benzonitrile hydrochloride (CGS020286A, FAD286, FAD) and compared these profiles to those of the 11beta-hydroxylase inhibitor metyrapone (MET) in two rodent models of secondary hyperaldosteronism and corticosteronism. In chronically cannulated Sprague-Dawley rats, angiotensin II (ANG II) (300 ng/kg bolus + 100 ng/kg/min infusion) or adrenocorticotropin (100 ng/kg + 30 ng/kg/min) acutely elevated plasma aldosterone concentration (PAC) from approximately 0.26 nM to a sustained level of approximately 2.5 nM for 9 h. Adrenocorticotropin but not ANG II elicited a sustained increase in plasma corticosterone concentration (PCC) from approximately 300 to approximately 1340 nM. After 1 h of Ang II or adrenocorticotropin infusion, FAD (0.01-100 mg/kg p.o.) or MET (0.1-300 mg/kg p.o.) dose- and drug plasma concentration-dependently reduced the elevated PACs over the ensuing 8 h. FAD was approximately 12 times more dose-potent than MET in reducing PAC but of similar or slightly greater potency on a plasma drug concentration basis. Both agents also decreased PCC in the adrenocorticotropin model at relatively higher doses and with similar dose potencies, whereas FAD was 6-fold weaker based on drug exposures. FAD was approximately 50-fold selective for reducing PAC versus PCC, whereas MET was only approximately 3-fold selective. We conclude that FAD is a potent, orally active, and relatively selective ASI in two rat models of hyperaldosteronism. MET is an order of magnitude less selective than FAD but is, nevertheless, more potent as an ASI than as an 11beta-hydroxylase inhibitor.
Collapse
Affiliation(s)
- Dean F Rigel
- Cardiovascular and Metabolism Research, Novartis Institutes for BioMedical Research, Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936-1080, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Adams CM, Hu CW, Jeng AY, Karki R, Ksander G, Lasala D, Leung-Chu J, Liang G, Liu Q, Meredith E, Rao C, Rigel DF, Shi J, Smith S, Springer C, Zhang C. The discovery of potent inhibitors of aldosterone synthase that exhibit selectivity over 11-beta-hydroxylase. Bioorg Med Chem Lett 2010; 20:4324-7. [PMID: 20615692 DOI: 10.1016/j.bmcl.2010.06.086] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2010] [Revised: 06/12/2010] [Accepted: 06/15/2010] [Indexed: 12/25/2022]
Abstract
Aldosterone, the final component of the renin-angiotensin-aldosterone system, plays an important role in the pathophysiology of hypertension and congestive heart failure. Aldosterone synthase (CYP11B2) catalyzes the last three steps of aldosterone biosynthesis, and as such appears to be a target for the treatment of these disorders. A sulfonamide-imidazole scaffold has proven to be a potent inhibitor of CYP11B2. Furthermore, this scaffold can achieve high levels of selectivity for CYP11B2 over CYP11B1, a key enzyme in the biosynthesis of cortisol.
Collapse
Affiliation(s)
- Christopher M Adams
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Roumen L, Peeters JW, Emmen JMA, Beugels IPE, Custers EMG, de Gooyer M, Plate R, Pieterse K, Hilbers PAJ, Smits JFM, Vekemans JAJ, Leysen D, Ottenheijm HCJ, Janssen HM, Hermans JJR. Synthesis, biological evaluation, and molecular modeling of 1-benzyl-1H-imidazoles as selective inhibitors of aldosterone synthase (CYP11B2). J Med Chem 2010; 53:1712-25. [PMID: 20121113 DOI: 10.1021/jm901356d] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Reducing aldosterone action is beneficial in various major diseases such as heart failure. Currently, this is achieved with mineralocorticoid receptor antagonists, however, aldosterone synthase (CYP11B2) inhibitors may offer a promising alternative. In this study, we used three-dimensional modeling of CYP11B2 to model the binding modes of the natural substrate 18-hydroxycorticosterone and the recently published CYP11B2 inhibitor R-fadrozole as a rational guide to design 44 structurally simple and achiral 1-benzyl-1H-imidazoles. Their syntheses, in vitro inhibitor potencies, and in silico docking are described. Some promising CYP11B2 inhibitors were identified, with our novel lead MOERAS115 (4-((5-phenyl-1H-imidazol-1-yl)methyl)benzonitrile) displaying an IC(50) for CYP11B2 of 1.7 nM, and a CYP11B2 (versus CYP11B1) selectivity of 16.5, comparable to R-fadrozole (IC(50) for CYP11B2 6.0 nM, selectivity 19.8). Molecular docking of the inhibitors in the models enabled us to generate posthoc hypotheses on their binding modes, providing a valuable basis for future studies and further design of CYP11B2 inhibitors.
Collapse
Affiliation(s)
- Luc Roumen
- BioModeling and bioInformatics, Eindhoven University of Technology, PO Box 513, Eindhoven 5600 MB, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Minnaard-Huiban M, Emmen JMA, Roumen L, Beugels IPE, Cohuet GMS, van Essen H, Ruijters E, Pieterse K, Hilbers PAJ, Ottenheijm HCJ, Plate R, de Gooyer ME, Smits JFM, Hermans JJR. Fadrozole reverses cardiac fibrosis in spontaneously hypertensive heart failure rats: discordant enantioselectivity versus reduction of plasma aldosterone. Endocrinology 2008; 149:28-31. [PMID: 17884944 DOI: 10.1210/en.2007-0584] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Reversal of cardiac fibrosis is a major determinant of the salutary effects of mineralocorticoid receptor antagonists in heart failure. Recently, R-fadrozole was coined as an aldosterone biosynthesis inhibitor, offering an appealing alternative to mineralocorticoid receptor antagonists to block aldosterone action. The present study aimed to evaluate the effects of R- and S-fadrozole on plasma aldosterone and urinary aldosterone excretion rate and to compare their effectiveness vs. the mineralocorticoid receptor antagonist potassium canrenoate to reverse established cardiac fibrosis. Male lean spontaneously hypertensive heart failure (SHHF) rats (40 wk) were treated for 8 wk by sc infusions of low (0.24 mg/kg.d) or high (1.2 mg/kg.d) doses of R- or S-fadrozole or by potassium canrenoate via drinking water (7.5 mg/kg.d). At the high dose, plasma aldosterone levels were decreased similarly by R- and S-fadrozole, whereas urinary aldosterone excretion rate was reduced only by S-fadrozole. In contrast, whereas at the high dose, R-fadrozole effectively reversed preexistent left ventricular interstitial fibrosis by 50% (vs. 42% for canrenoate), S-fadrozole was devoid of an antifibrotic effect. The low doses of the fadrozole enantiomers did not change cardiac fibrosis or plasma aldosterone but similarly reduced urinary aldosterone excretion rate. In conclusion, R-fadrozole may possess considerable therapeutic merit because of its potent antifibrotic actions in the heart. However, the observed discordance between the aldosterone-lowering and antifibrotic effects of the fadrozole enantiomers raises some doubt about the mechanism by which R-fadrozole diminishes cardiac collagen and about the generality of the concept of lowering aldosterone levels to treat the diseased heart.
Collapse
Affiliation(s)
- Monica Minnaard-Huiban
- Department of Pharmacology and Toxicology, University of Maastricht, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|