1
|
Guo C, Zhang G, Wu C, Lei Y, Wang Y, Yang J. Emerging trends in small molecule inhibitors targeting aldosterone synthase: A new paradigm in cardiovascular disease treatment. Eur J Med Chem 2024; 274:116521. [PMID: 38820853 DOI: 10.1016/j.ejmech.2024.116521] [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: 03/28/2024] [Revised: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Aldosterone synthase (CYP11B2) is the rate-limiting enzyme in aldosterone production. In recent years, CYP11B2 has become an appealing target for treating conditions associated with excess aldosterone, such as hypertension, heart failure, and cardiometabolic diseases. Several small-molecule inhibitors of CYP11B2 have demonstrated efficacy in both preclinical studies and clinical trials. Among them, the tetrahydroisoquinoline derivative Baxdrostat has entered clinical trial phases and demonstrated efficacy in treating patients with hypertension. However, the high homology (>93 %) between CYP11B2 and steroid-11β-hydroxylase (CYP11B1), which catalyzes cortisol production, implies that insufficient drug specificity can lead to severe side effects. Developing selective inhibitors for CYP11B2 remains a considerable challenge that requires ongoing attention. This review summarizes recent research progress on small-molecule inhibitors targeting CYP11B2, focusing on structure-activity relationships (SAR) and structural optimization. It discusses strategies for enhancing the specificity and inhibitory activity of inhibitors, while also exploring potential applications and future prospects for CYP11B2 inhibitors, providing a theoretical foundation for developing the new generation of CYP11B2-targeted medications.
Collapse
Affiliation(s)
- Cuiyu Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guangbing Zhang
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Lei
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, General Practice Research Institute, West China Hospital, Sichuan University, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Launonen H, Luiskari L, Linden J, Siltari A, Salmenkari H, Korpela R, Vapaatalo H. Adverse effects of an aldosterone synthase (CYP11B2) inhibitor, fadrozole (FAD286), on inflamed rat colon. Basic Clin Pharmacol Toxicol 2023; 133:211-225. [PMID: 37345281 DOI: 10.1111/bcpt.13918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023]
Abstract
Recently, we described local aldosterone production in the murine large intestine. Upregulated local aldosterone synthesis in different tissues has been linked with inflammatory conditions, which have been attenuated by the aldosterone synthase (CYP11B2) inhibitor, fadrozole (FAD286). Therefore, we investigated the effect of inhibition of intestinal aldosterone synthesis on the development of intestinal inflammation. Sprague-Dawley rats were administered 5% (v/w) dextran sodium sulphate (DSS) for 7 days with or without daily FAD286 (30 mg/kg/d) subcutaneous injections on 3 days before, during and one day after DSS. Tissue aldosterone concentrations were evaluated by ELISA, CYP11B2 by Western blot and RT-qPCR. FAD286 halved adrenal aldosterone production but, intriguingly, increased the colonic aldosterone concentration. The lack of inhibitory effect of FAD286 in the colon might have been affected by the smaller size of colonic vs. adrenal CYP11B2, as seen in Western blot. When combined with DSS, FAD286 aggravated the macroscopic and histological signs of intestinal inflammation, lowered the animals' body weight gain and increased the incidence of gastrointestinal bleeding and the permeability to iohexol in comparison to DSS-animals. To conclude, FAD286 exerted harmful effects during intestinal inflammation. Local intestinal aldosterone did not seem to play any role in the inflammatory pathogenesis occurring in the intestine.
Collapse
Affiliation(s)
- Hanna Launonen
- Faculty of Medicine, Pharmacology, University of Helsinki, Helsinki, Finland
| | - Lotta Luiskari
- Faculty of Medicine, Pharmacology, University of Helsinki, Helsinki, Finland
| | - Jere Linden
- Faculty of Veterinary Medicine, Department of Veterinary Biosciences and Finnish Centre for Laboratory Animal Pathology (FCLAP), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aino Siltari
- Faculty of Medicine, Pharmacology, University of Helsinki, Helsinki, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hanne Salmenkari
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riitta Korpela
- Faculty of Medicine, Pharmacology, University of Helsinki, Helsinki, Finland
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | - Heikki Vapaatalo
- Faculty of Medicine, Pharmacology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Mehlich A, Bolanowski M, Mehlich D, Witek P. Medical treatment of Cushing's disease with concurrent diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1174119. [PMID: 37139336 PMCID: PMC10150952 DOI: 10.3389/fendo.2023.1174119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Cushing's disease (CD) is a severe endocrine disorder characterized by chronic hypercortisolaemia secondary to an overproduction of adrenocorticotropic hormone (ACTH) by a pituitary adenoma. Cortisol excess impairs normal glucose homeostasis through many pathophysiological mechanisms. The varying degrees of glucose intolerance, including impaired fasting glucose, impaired glucose tolerance, and Diabetes Mellitus (DM) are commonly observed in patients with CD and contribute to significant morbidity and mortality. Although definitive surgical treatment of ACTH-secreting tumors remains the most effective therapy to control both cortisol levels and glucose metabolism, nearly one-third of patients present with persistent or recurrent disease and require additional treatments. In recent years, several medical therapies demonstrated prominent clinical efficacy in the management of patients with CD for whom surgery was non-curative or for those who are ineligible to undergo surgical treatment. Cortisol-lowering medications may have different effects on glucose metabolism, partially independent of their role in normalizing hypercortisolaemia. The expanding therapeutic landscape offers new opportunities for the tailored therapy of patients with CD who present with glucose intolerance or DM, however, additional clinical studies are needed to determine the optimal management strategies. In this article, we discuss the pathophysiology of impaired glucose metabolism caused by cortisol excess and review the clinical efficacy of medical therapies of CD, with particular emphasis on their effects on glucose homeostasis.
Collapse
Affiliation(s)
- Anna Mehlich
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Marek Bolanowski
- Chair and Department of Endocrinology, Diabetes, and Isotope Treatment, Wroclaw Medical University, Wroclaw, Poland
| | - Dawid Mehlich
- Laboratory of Molecular OncoSignalling, International Institute of Molecular Mechanisms and Machines (IMol) Polish Academy of Sciences, Warsaw, Poland
- Doctoral School of Medical University of Warsaw, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Witek
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
- *Correspondence: Przemysław Witek,
| |
Collapse
|
4
|
Yin L, Pan Y, Xue Y, Chen X, You T, Huang J, Xu Q, Hu Q. Design, Synthesis, and Biological Evaluations of Pyridyl 4,5,6,7-Tetrahydro-4,7-Methanobenzo[ d]isoxazoles as Potent and Selective Inhibitors of 11β-Hydroxylase. J Med Chem 2022; 65:11876-11888. [PMID: 35975976 DOI: 10.1021/acs.jmedchem.2c01037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Inhibition of CYP11B1 is a promising therapy for severe diseases caused by excessive cortisol. Enantiomer discrimination provides clues to achieve selectivity that CYP11B1 and homologous CYP11B2 were selectively bound by S- and R-fadrozole, respectively, in distinct binding modes. Pyridyl 4,5,6,7-tetrahydro-4,7-methanobenzo[d]isoxazoles showing a similar binding mode to S-fadrozole in CYP11B1 were designed as potent and selective CYP11B1 inhibitors. Compound 7aa exhibited a highly potent CYP11B1 inhibition similar to that of the drug osilodrostat (IC50's of 9 and 6 nM, respectively) but was 1500-fold more selective over CYP11B2 compared to osilodrostat (selectivity factors of 125 versus 0.08, respectively). Strong reductions of plasma cortisol concentrations by compound 7aa were demonstrated in rats without interference in aldosterone production after oral application. It showed no inhibition against a panel of steroidogenic and hepatic CYP enzymes. Exhibiting a good pharmacokinetic profile, compound 7aa was considered as a drug candidate for further development.
Collapse
Affiliation(s)
- Lina Yin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Youtian Pan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Yuanyuan Xue
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Xiaoli Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Taiyun You
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Jiahui Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Qihao Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| | - Qingzhong Hu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 East Waihuan Road, 510006 Panyu, Guangzhou, P. R. China
| |
Collapse
|
5
|
|
6
|
Brixius-Anderko S, Scott EE. Aldosterone Synthase Structure With Cushing Disease Drug LCI699 Highlights Avenues for Selective CYP11B Drug Design. Hypertension 2021; 78:751-759. [PMID: 34247511 DOI: 10.1161/hypertensionaha.121.17615] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
| | - Emily E Scott
- Departments of Medicinal Chemistry (S.B.-A., E.E.S.), University of Michigan, Ann Arbor.,Pharmacology (E.E.S.), University of Michigan, Ann Arbor
| |
Collapse
|
7
|
Maternal Undernutrition Modulates Neonatal Rat Cerebrovascular Structure, Function, and Vulnerability to Mild Hypoxic-Ischemic Injury via Corticosteroid-Dependent and -Independent Mechanisms. Int J Mol Sci 2021; 22:ijms22020680. [PMID: 33445547 PMCID: PMC7827870 DOI: 10.3390/ijms22020680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/27/2022] Open
Abstract
The present study explored the hypothesis that an adverse intrauterine environment caused by maternal undernutrition (MUN) acted through corticosteroid-dependent and -independent mechanisms to program lasting functional changes in the neonatal cerebrovasculature and vulnerability to mild hypoxic-ischemic (HI) injury. From day 10 of gestation until term, MUN and MUN-metyrapone (MUN-MET) group rats consumed a diet restricted to 50% of calories consumed by a pair-fed control; and on gestational day 11 through term, MUN-MET groups received drinking water containing MET (0.5 mg/mL), a corticosteroid synthesis inhibitor. P9/P10 pups underwent unilateral carotid ligation followed 24 h later by 1.5 h exposure to 8% oxygen (HI treatment). An ELISA quantified MUN-, MET-, and HI-induced changes in circulating levels of corticosterone. In P11/P12 pups, MUN programming promoted contractile differentiation in cerebrovascular smooth muscle as determined by confocal microscopy, modulated calcium-dependent contractility as revealed by cerebral artery myography, enhanced vasogenic edema formation as indicated by T2 MRI, and worsened neurobehavior MUN unmasked HI-induced improvements in open-field locomotion and in edema resolution, alterations in calcium-dependent contractility and promotion of contractile differentiation. Overall, MUN imposed multiple interdependent effects on cerebrovascular smooth muscle differentiation, contractility, edema formation, flow-metabolism coupling and neurobehavior through pathways that both required, and were independent of, gestational corticosteroids. In light of growing global patterns of food insecurity, the present study emphasizes that infants born from undernourished mothers may experience greater risk for developing neonatal cerebral edema and sensorimotor impairments possibly through programmed changes in neonatal cerebrovascular function.
Collapse
|
8
|
Varlamov EV, Han AJ, Fleseriu M. Updates in adrenal steroidogenesis inhibitors for Cushing's syndrome - A practical guide. Best Pract Res Clin Endocrinol Metab 2021; 35:101490. [PMID: 33707082 DOI: 10.1016/j.beem.2021.101490] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Medical therapy is essential in the management of patients with Cushing's syndrome (CS) when curative surgery has failed, surgery is not feasible, when awaiting radiation effect, and in recurrent cases of CS. Steroidogenesis inhibitors have a rapid onset of action and are effective in reducing hypercortisolism, however, adverse effects, including adrenal insufficiency require very close patient monitoring. Osilodrostat is the only steroidogenesis inhibitor to have been assessed in prospective randomized controlled trials and approved for Cushing's disease (CD) by the US Food and Drug Administration and for CS by the European Medical Agency (EMA). Osilodrostat has been shown to be highly effective at maintaining normal urinary free cortisol in patients with CD. Drugs such as metyrapone, ketoconazole (both EMA approved), and etomidate lack prospective evaluation(s). There is, however, considerable clinical experience and retrospective data that show a very wide efficacy range in treating patients with CS. In the absence of head-to-head comparative clinical trials, therapy choice is determined by the specific clinical setting, risk of adverse events, cost, availability, and other factors. In this review practical points to help clinicians who are managing patients with CS being treated with steroidogenesis inhibitors are presented.
Collapse
Affiliation(s)
- Elena V Varlamov
- Departments of Medicine (Endocrinology, Diabetes and Clinical Nutrition) and Neurological Surgery, and Pituitary Center, Oregon Health & Science University, Mail Code CH8N, 3303 South Bond Avenue, Portland, OR, 97239, USA
| | - Ashley J Han
- School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Pk. Rd, Portland, OR, 97239, USA
| | - Maria Fleseriu
- Departments of Medicine (Endocrinology, Diabetes and Clinical Nutrition) and Neurological Surgery, and Pituitary Center, Oregon Health & Science University, Mail Code CH8N, 3303 South Bond Avenue, Portland, OR, 97239, USA.
| |
Collapse
|
9
|
Shaqura M, Li L, Mohamed DM, Li X, Treskatsch S, Buhrmann C, Shakibaei M, Beyer A, Mousa SA, Schäfer M. Neuronal aldosterone elicits a distinct genomic response in pain signaling molecules contributing to inflammatory pain. J Neuroinflammation 2020; 17:183. [PMID: 32532285 PMCID: PMC7291517 DOI: 10.1186/s12974-020-01864-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/02/2020] [Indexed: 11/10/2022] Open
Abstract
Background Recently, mineralocorticoid receptors (MR) were identified in peripheral nociceptive neurons, and their acute antagonism was responsible for immediate and short-lasting (non-genomic) antinociceptive effects. The same neurons were shown to produce the endogenous ligand aldosterone by the enzyme aldosterone synthase. Methods Here, we investigate whether endogenous aldosterone contributes to inflammation-induced hyperalgesia via the distinct genomic regulation of specific pain signaling molecules in an animal model of Freund’s complete adjuvant (FCA)-induced hindpaw inflammation. Results Chronic intrathecal application of MR antagonist canrenoate-K (over 4 days) attenuated nociceptive behavior in rats with FCA hindpaw inflammation suggesting a tonic activation of neuronal MR by endogenous aldosterone. Consistently, double immunofluorescence confocal microscopy showed abundant co-localization of MR with several pain signaling molecules such as TRPV1, CGRP, Nav1.8, and trkA whose enhanced expression of mRNA and proteins during inflammation was downregulated following i.t. canrenoate-K. More importantly, inhibition of endogenous aldosterone production in peripheral sensory neurons by continuous intrathecal delivery of a specific aldosterone synthase inhibitor prevented the inflammation-induced enhanced transcriptional expression of TRPV1, CGRP, Nav1.8, and trkA and subsequently attenuated nociceptive behavior. Evidence for such a genomic effect of endogenous aldosterone was supported by the demonstration of an enhanced nuclear translocation of MR in peripheral sensory dorsal root ganglia (DRG) neurons. Conclusion Taken together, chronic inhibition of local production of aldosterone by its processing enzyme aldosterone synthase within peripheral sensory neurons may contribute to long-lasting downregulation of specific pain signaling molecules and may, thus, persistently reduce inflammation-induced hyperalgesia.
Collapse
Affiliation(s)
- Mohammed Shaqura
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Li Li
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Doaa M Mohamed
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany.,Department of Zoology, Faculty of Science, Aswan University, Tingar, Egypt
| | - Xiongjuan Li
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, No. 250, Hai'zhu District, Guangzhou, 510260, China
| | - Sascha Treskatsch
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Constanze Buhrmann
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mehdi Shakibaei
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Antje Beyer
- Department of Anaesthesiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Shaaban A Mousa
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany.
| | - Michael Schäfer
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
10
|
Franco PN, Durrant LM, Carreon D, Haddad E, Vergara A, Cascavita C, Obenaus A, Pearce WJ. Prenatal metyrapone treatment modulates neonatal cerebrovascular structure, function, and vulnerability to mild hypoxic-ischemic injury. Am J Physiol Regul Integr Comp Physiol 2019; 318:R1-R16. [PMID: 31577477 DOI: 10.1152/ajpregu.00145.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study explored the hypothesis that late gestational reduction of corticosteroids transforms the cerebrovasculature and modulates postnatal vulnerability to mild hypoxic-ischemic (HI) injury. Four groups of Sprague-Dawley neonates were studied: 1) Sham-Control, 2) Sham-MET, 3) HI-Control, and 4) HI-MET. Metyrapone (MET), a corticosteroid synthesis inhibitor, was administered via drinking water from gestational day 11 to term. In Shams, MET administration 1) decreased reactivity of the hypothalamic-pituitary-adrenal (HPA) axis to surgical trauma in postnatal day 9 (P9) pups by 37%, 2) promoted cerebrovascular contractile differentiation in middle cerebral arteries (MCAs), 3) decreased compliance ≤46% and increased depolarization-induced calcium mobilization in MCAs by 28%, 4) mildly increased hemispheric cerebral edema by 5%, decreased neuronal degeneration by 66%, and increased astroglial and microglial activation by 10- and 4-fold, respectively, and 5) increased righting reflex times by 29%. Regarding HI, metyrapone-induced fetal transformation 1) diminished reactivity of the HPA axis to HI-induced stress in P9/P10 pups, 2) enhanced HI-induced contractile dedifferentiation in MCAs, 3) lessened the effects of HI on MCA compliance and calcium mobilization, 4) decreased HI-induced neuronal injury but unmasked regional HI-induced depression of microglial activation, and 5) attenuated the negative effects of HI on open-field exploration but enhanced the detrimental effects of HI on negative geotaxis responses by 79%. Overall, corticosteroids during gestation appear essential for normal cerebrovascular development and glial quiescence but induce persistent changes that in neonates manifest beneficially as preservation of postischemic contractile differentiation but detrimentally as worsened ischemic cerebrovascular compliance, increased ischemic neuronal injury, and compromised neurobehavior.
Collapse
Affiliation(s)
- P Naomi Franco
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Lara M Durrant
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Desirelys Carreon
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Elizabeth Haddad
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, California
| | - Adam Vergara
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Catherine Cascavita
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, California
| | - William J Pearce
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
11
|
Creemers SG, Feelders RA, de Jong FH, Franssen GJH, de Rijke YB, van Koetsveld PM, Hofland LJ. Osilodrostat Is a Potential Novel Steroidogenesis Inhibitor for the Treatment of Cushing Syndrome: An In Vitro Study. J Clin Endocrinol Metab 2019; 104:3437-3449. [PMID: 31127821 DOI: 10.1210/jc.2019-00217] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/17/2019] [Indexed: 01/05/2023]
Abstract
CONTEXT Metyrapone and ketoconazole, frequently used steroidogenesis inhibitors for treatment of Cushing syndrome, can be associated with side effects and limited efficacy. Osilodrostat is a CYP11B1 and CYP11B2 inhibitor, with unknown effects on other steroidogenic enzymes. OBJECTIVE To compare the effects of osilodrostat, metyrapone, and ketoconazole on adrenal steroidogenesis, and pituitary adenoma cells in vitro. METHODS HAC15 cells, 17 primary human adrenocortical cell cultures, and pituitary adenoma cells were incubated with osilodrostat, metyrapone, or ketoconazole (0.01 to 10 µM). Cortisol and ACTH were measured using chemiluminescence immunoassays, and steroid profiles by liquid chromatography-mass spectrometry. RESULTS In HAC15 cells, osilodrostat inhibited cortisol production more potently (IC50: 0.035 µM) than metyrapone (0.068 µM; P < 0.0001), and ketoconazole (0.621 µM; P < 0.0001). IC50 values of osilodrostat and metyrapone for basal cortisol production varied with a 25- and 18-fold difference, respectively, with comparable potency. Aldosterone production was inhibited more potently by osilodrostat vs metyrapone and ketoconazole. Osilodrostat and metyrapone treatment resulted in strong inhibition of corticosterone and cortisol, 11-deoxycortisol accumulation, and modest effects on adrenal androgens. No pituitary-directed effects of osilodrostat were observed. CONCLUSIONS Under our study conditions, osilodrostat is a potent cortisol production inhibitor in human adrenocortical cells, comparable with metyrapone. All steroidogenesis inhibitors showed large variability in sensitivity between primary adrenocortical cultures. Osilodrostat might inhibit CYP11B1 and CYP11B2, in some conditions to a lesser extent CYP17A1 activity, and a proximal step in the steroidogenesis. Osilodrostat is a promising treatment option for Cushing syndrome, and in vivo differences with metyrapone are potentially driven by pharmacokinetic differences.
Collapse
Affiliation(s)
- Sara G Creemers
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| | - Richard A Feelders
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| | - Frank H de Jong
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| | - Gaston J H Franssen
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| | - Peter M van Koetsveld
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| | - Leo J Hofland
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| |
Collapse
|
12
|
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
|
13
|
Brixius-Anderko S, Scott EE. Structure of human cortisol-producing cytochrome P450 11B1 bound to the breast cancer drug fadrozole provides insights for drug design. J Biol Chem 2018; 294:453-460. [PMID: 30425102 DOI: 10.1074/jbc.ra118.006214] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/12/2018] [Indexed: 11/06/2022] Open
Abstract
Human cytochrome P450 11B1 (CYP11B1) is responsible for the final step generating the steroid hormone cortisol, which controls stress and immune responses and glucose homeostasis. CYP11B1 is a promising drug target to manage Cushing's disease, a disorder arising from excessive cortisol production. However, the design of selective inhibitors has been hampered because structural information for CYP11B1 is unavailable and the enzyme has high amino acid sequence identity (93%) to a closely related enzyme, the aldosterone-producing CYP11B2. Here we report the X-ray crystal structure of human CYP11B1 (at 2.1 Å resolution) in complex with fadrozole, a racemic compound normally used to treat breast cancer by inhibiting estrogen-producing CYP19A1. Comparison of fadrozole-bound CYP11B1 with fadrozole-bound CYP11B2 revealed that despite conservation of the active-site residues, the overall structures and active sites had structural rearrangements consistent with distinct protein functions and inhibition. Whereas fadrozole binds to both CYP11B enzymes by coordinating the heme iron, CYP11B2 binds to the R enantiomer of fadrozole, and CYP11B1 binds to the S enantiomer, each with distinct orientations and interactions. These results provide insights into the cross-reactivity of drugs across multiple steroidogenic cytochrome P450 enzymes, provide a structural basis for understanding human steroidogenesis, and pave the way for the design of more selective inhibitors of each human CYP11B enzyme.
Collapse
Affiliation(s)
| | - Emily E Scott
- From the Departments of Medicinal Chemistry and .,Pharmacology and the Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
14
|
Fuertes M, Tkatch J, Rosmino J, Nieto L, Guitelman MA, Arzt E. New Insights in Cushing Disease Treatment With Focus on a Derivative of Vitamin A. Front Endocrinol (Lausanne) 2018; 9:262. [PMID: 29881371 PMCID: PMC5976796 DOI: 10.3389/fendo.2018.00262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/07/2018] [Indexed: 12/19/2022] Open
Abstract
Cushing's disease (CD) is an endocrine disorder originated by a corticotroph tumor. It is linked with high mortality and morbidity due to chronic hypercortisolism. Treatment goals are to control cortisol excess and achieve long-term remission, therefore, reducing both complications and patient's mortality. First-line of treatment for CD is pituitary's surgery. However, 30% of patients who undergo surgery experience recurrence in long-term follow-up. Persistent or recurrent CD demands second-line treatments, such as pituitary radiotherapy, adrenal surgery, and/or pharmacological therapy. The latter plays a key role in cortisol excess control. Its targets are inhibition of adrenocorticotropic hormone (ACTH) production, inhibition of adrenal steroidogenesis, or antagonism of cortisol action at its peripheral receptor. Retinoic acid (RA) is a metabolic product of vitamin A (retinol) and has been studied for its antiproliferative effects on corticotroph tumor cells. It has been shown that this drug regulates the expression of pro-opiomelanocortin (POMC), ACTH secretion, and tumor growth in corticotroph tumor mouse cell lines and in the nude mice experimental model, via inhibition of POMC transcription. It has been shown to result in tumor reduction, normalization of cortisol levels and clinical improvement in dogs treated with RA for 6 months. The orphan nuclear receptor COUP-TFI is expressed in normal corticotroph cells, but not in corticotroph tumoral cells, and inhibits RA pathways. A first clinical human study demonstrated clinical and biochemical effectiveness in 5/7 patients treated with RA for a period of up to 12 months. In a recent second clinical trial, 25% of 16 patients achieved eucortisolemia, and all achieved a cortisol reduction after 6- to 12-month treatment. The goal of this review is to discuss in the context of the available and future pharmacological treatments of CD, RA mechanisms of action on corticotroph tumor cells, and future perspectives, focusing on potential clinical implementation.
Collapse
Affiliation(s)
- Mariana Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Julieta Tkatch
- División Endocrinología, Hospital General de Agudos “Carlos G. Durand”, Buenos Aires, Argentina
| | - Josefina Rosmino
- División Endocrinología, Hospital General de Agudos “Carlos G. Durand”, Buenos Aires, Argentina
| | - Leandro Nieto
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | | | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- *Correspondence: Eduardo Arzt,
| |
Collapse
|
15
|
Simola N, Paci E, Serra M, Costa G, Morelli M. Modulation of Rat 50-kHz Ultrasonic Vocalizations by Glucocorticoid Signaling: Possible Relevance to Reward and Motivation. Int J Neuropsychopharmacol 2017; 21:73-83. [PMID: 29182715 PMCID: PMC5795343 DOI: 10.1093/ijnp/pyx106] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Rats emit 50-kHz ultrasonic vocalizations (USVs) to communicate positive emotional states, and these USVs are increasingly being investigated in preclinical studies on reward and motivation. Although it is the activation of dopamine receptors that initiates the emission of 50-kHz USVs, non-dopaminergic mechanisms may modulate calling in the 50 kHz frequency band. To further elucidate these mechanisms, the present study investigated whether the pharmacological manipulation of glucocorticoid signaling influenced calling. METHODS Rats were administered corticosterone (1-5 mg/kg, s.c.), the glucocorticoid receptor antagonist mifepristone (40 or 100 mg/kg, s.c.), or the corticosterone synthesis inhibitor metyrapone (50 or 100 mg/kg, i.p.). The effects of these drugs on calling initiation and on calling recorded during nonaggressive social contacts or after the administration of amphetamine (0.25 or 1 mg/kg, i.p.) were then evaluated. RESULTS Corticosterone failed to initiate the emission of 50-kHz USVs and did not influence pro-social and amphetamine-stimulated calling. Similarly, mifepristone and metyrapone did not initiate calling. However, metyrapone suppressed pro-social calling and calling stimulated by a moderate dose (1 mg/kg, i.p.) of amphetamine. Conversely, mifepristone attenuated calling stimulated by a low (0.25 mg/kg, i.p.), but not moderate (1 mg/kg, i.p.), dose of amphetamine and had no influence on pro-social calling. CONCLUSIONS The present results demonstrate that glucocorticoid signaling modulates calling in the 50 kHz frequency band only in certain conditions and suggest that mechanisms different from the inhibition of corticosterone synthesis may participate in the suppression of calling by metyrapone.
Collapse
Affiliation(s)
- Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy,National Institute of Neuroscience, University of Cagliari, Cagliari, Italy,Correspondence: Nicola Simola, PhD, Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale, 72, 09124, Cagliari, Italy ()
| | - Elena Paci
- Department of Physiology, University of Bristol, Bristol, United Kingdom,Department of Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy,National Institute of Neuroscience, University of Cagliari, Cagliari, Italy,NCR, National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| |
Collapse
|
16
|
Emmerich J, van Koppen CJ, Burkhart JL, Hu Q, Siebenbürger L, Boerger C, Scheuer C, Laschke MW, Menger MD, Hartmann RW. Lead Optimization Generates CYP11B1 Inhibitors of Pyridylmethyl Isoxazole Type with Improved Pharmacological Profile for the Treatment of Cushing’s Disease. J Med Chem 2017; 60:5086-5098. [DOI: 10.1021/acs.jmedchem.7b00437] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Juliette Emmerich
- Pharmaceutical
and Medicinal Chemistry, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | | | | | - Qingzhong Hu
- Department
of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | | | - Carsten Boerger
- PharmBioTec GmbH, Science Park 1, 66123 Saarbrücken, Germany
| | - Claudia Scheuer
- Institute
for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Matthias W. Laschke
- Institute
for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Michael D. Menger
- Institute
for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Rolf W. Hartmann
- Pharmaceutical
and Medicinal Chemistry, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
17
|
Shi H, Gao T, Liu Z, Sun L, Jiang X, Chen L, Wang D. Blockage of androgen and administration of estrogen induce transdifferentiation of testis into ovary. J Endocrinol 2017; 233:65-80. [PMID: 28148717 DOI: 10.1530/joe-16-0551] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/01/2017] [Indexed: 12/24/2022]
Abstract
Induction of sex reversal of XY fish has been restricted to the sex undifferentiated period. In the present study, differentiated XY tilapia were treated with trilostane (TR), metopirone (MN) and glycyrrhetinic acid (GA) (inhibitor of 3β-HSD, Cyp11b2 and 11β-HSD, respectively) alone or in combination with 17β-estradiol (E2) from 30 to 90 dah (days after hatching). At 180 dah, E2 alone resulted in 8.3%, and TR, MN and GA alone resulted in no secondary sex reversal (SSR), whereas TR + E2, MN + E2 and GA + E2 resulted in 88.3, 60.0 and 46.7% of SSR, respectively. This sex reversal could be rescued by simultaneous administration of 11-ketotestosterone (11-KT). Compared with the control XY fish, decreased serum 11-KT and increased E2 level were detected in SSR fish. Immunohistochemistry analyses revealed that Cyp19a1a, Cyp11b2 and Dmrt1 were expressed in the gonads of GA + E2, MN + E2 and TR + E2 SSR XY fish at 90 dah, but only Cyp19a1a was expressed at 180 dah. When the treatment was applied from 60 to 120 dah, TR + E2 resulted in 3.3% of SSR, MN + E2 and GA + E2 resulted in no SSR. These results demonstrated that once 11-KT was synthesized, it could antagonize E2-induced male-to-female SSR, which could be abolished by simultaneous treatment with the inhibitor of steroidogenic enzymes. The upper the enzyme was located in the steroidogenic pathway, the higher SSR rate was achieved when it was inhibited as some of the precursors, such as androstenedione, testosterone and 5α-dihydrotestosterone, could act as androgens. These results highlight the key role of androgen in male sex maintenance.
Collapse
Affiliation(s)
- Hongjuan Shi
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Tian Gao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Zhilong Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Lina Sun
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Xiaolong Jiang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Lili Chen
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education)Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| |
Collapse
|
18
|
Salt-dependent Blood Pressure in Human Aldosterone Synthase-Transgenic Mice. Sci Rep 2017; 7:492. [PMID: 28352088 PMCID: PMC5412599 DOI: 10.1038/s41598-017-00461-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/28/2017] [Indexed: 01/19/2023] Open
Abstract
Hypertension is one of the most important, preventable causes of premature morbidity and mortality in the developed world. Aldosterone is a major mineralocorticoid hormone that plays a key role in the regulation of blood pressure and is implicated in the pathogenesis of hypertension and heart failure. Aldosterone synthase (AS, cytochrome P450 11B2, cyp11B2) is the sole enzyme responsible for the production of aldosterone in humans. To determine the effects of increased expression of human aldosterone synthase (hAS) on blood pressure (BP), we established transgenic mice carrying the hAS gene (cyp11B2). We showed that hAS overexpression increased levels of aldosterone in hAS+/- mice. On high salt diet (HS), BPs of hAS+/- mice were significantly increased compared with WT mice. Fadrozole (an inhibitor of aldosterone synthase) treatment significantly reduced BPs of hAS+/- mice on HS. This is the first time overexpression of AS in a transgenic mouse line has shown an ability to induce HP. Specifically inhibiting AS activity in these mice is a promising therapy for reducing hypertension. This hAS transgenic mouse model is therefore an ideal animal model for hypertension therapy studies.
Collapse
|
19
|
Abstract
PURPOSE Endogenous Cushing's syndrome (CS) is a rare disease that results from exposure to high levels of cortisol; Cushing's disease (CD) is the most frequent form of CS. Patients with CS suffer from a variety of comorbidities that increase the risk of mortality. Surgical resection of the disease-causing lesion is generally the first-line treatment of CS. However, some patients may not be eligible for surgery due to comorbidities, and approximately 25 % of patients, especially those with CD, have recurrent disease. For these patients, adrenal steroidogenesis inhibitors may control cortisol elevation and subsequent symptomatology. CS is rare overall, and clinical studies of adrenal steroidogenesis inhibitors are often small and, in many cases, data are limited regarding the efficacy and safety of these treatments. Our aim was to better characterize the profiles of efficacy and safety of currently available adrenal steroidogenesis inhibitors, including drugs currently in development. METHODS We performed a systematic review of the literature regarding adrenal steroidogenesis inhibitors, focusing on novel drugs. RESULTS Currently available adrenal steroidogenesis inhibitors, including ketoconazole, metyrapone, etomidate, and mitotane, have variable efficacy and significant side effects, and none are approved by the US Food and Drug Administration for CS. Therefore, there is a clear need for novel, prospectively studied agents that have greater efficacy and a low rate of adverse side effects. Efficacy and safety data of current and emerging adrenal steroidogenesis inhibitors, including osilodrostat (LCI699) and levoketoconazole (COR-003), show promising results that will have to be confirmed in larger-scale phase 3 studies (currently ongoing). CONCLUSIONS The management of CS, and particularly CD, remains challenging. Adrenal steroidogenesis inhibitors can be of major interest to control the hypercortisolism at any time point, either before or after surgery, as discussed in this review.
Collapse
Affiliation(s)
- Maria Fleseriu
- Departments of Medicine and Neurological Surgery, and Northwest Pituitary Center, Oregon Health & Science University, Mail Code: CH8N, 3303 SW Bond Ave, Portland, OR, 97239, USA.
| | - Frederic Castinetti
- Aix Marseille University, CNRS, CRN2M, Department of Endocrinology, Assistance Publique Hopitaux de Marseille, Marseille, France
| |
Collapse
|
20
|
Hofmann A, Brunssen C, Peitzsch M, Martin M, Mittag J, Jannasch A, Engelmann F, Brown NF, Weldon SM, Huber J, Streicher R, Deussen A, Eisenhofer G, Bornstein SR, Morawietz H. Aldosterone Synthase Inhibition Improves Glucose Tolerance in Zucker Diabetic Fatty (ZDF) Rats. Endocrinology 2016; 157:3844-3855. [PMID: 27526033 DOI: 10.1210/en.2016-1358] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Plasma aldosterone is elevated in type 2 diabetes and obesity in experimental and clinical studies and can act to inhibit both glucose-stimulated insulin secretion by the β-cell and insulin signaling. Currently mineralocorticoid receptor antagonism is the best characterized treatment to ameliorate aldosterone-mediated effects. A second alternative is inhibition of aldosterone synthase, an approach with protective effects on end-organ damage in heart or kidney in animal models. The effect of aldosterone synthase inhibition on metabolic parameters in type 2 diabetes is not known. Therefore, male Zucker diabetic fatty (ZDF) rats were treated for 11 weeks with the aldosterone synthase inhibitor FAD286, beginning at 7 weeks of age. Results were compared with the mineralocorticoid receptor antagonist eplerenone. Plasma aldosterone was abolished by FAD286 and elevated more than 9-fold by eplerenone. The area under the curve calculated from an oral glucose tolerance test (OGTT) was lower and overall insulin response during OGTT was increased by FAD286. In contrast, eplerenone elevated blood glucose levels and blunted insulin secretion during the OGTT. Fasting glucose was lowered and fasting insulin was increased by FAD286 in the prediabetic state. Glycated hemoglobin was lowered by FAD286, whereas eplerenone showed no effect. We conclude that aldosterone synthase inhibition, in contrast to mineralocorticoid receptor antagonism, has the potential for beneficial effects on metabolic parameters in type 2 diabetes.
Collapse
Affiliation(s)
- Anja Hofmann
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Mirko Peitzsch
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Melanie Martin
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Jennifer Mittag
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Anett Jannasch
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Felix Engelmann
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Nicholas F Brown
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Steven M Weldon
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Jochen Huber
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Rüdiger Streicher
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Andreas Deussen
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Graeme Eisenhofer
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Stefan R Bornstein
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation (A.H., C.B., J.M., F.E., H.M.) and Division of Clinical Neurochemistry (M.P., G.E.), Institute of Clinical Chemistry and Laboratory Medicine, Department of Medicine III (G.E., S.R.B.), University Hospital Carl Gustav Carus Dresden, and Institute of Physiology (M.M., A.D.) and Department of Cardiac Surgery (A.J.), Herzzentrum Dresden, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cardio Metabolic Diseases (N.F.B., S.M.W.), Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut 06877; Department of Cardio Metabolic Diseases (J.H., R.S.), Boehringer Ingelheim Pharma GmbH and Co KG, 88400 Biberach, Germany; and Department of Endocrinology and Diabetes (S.R.B.), Division of Diabetes and Nutritional Sciences, Rayne Institute, Faculty of Life Sciences and Medicine, Kings College London, London, SE5 9PJ, United Kingdom
| |
Collapse
|
21
|
Affiliation(s)
- Rita Bernhardt
- Lehrstuhl für Biochemie, Universität des Saarlandes, Saarbrücken, Germany
| |
Collapse
|
22
|
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
|
23
|
Cerny MA, Csengery A, Schmenk J, Frederick K. Development of CYP11B1 and CYP11B2 assays utilizing homogenates of adrenal glands: Utility of monkey as a surrogate for human. J Steroid Biochem Mol Biol 2015; 154:197-205. [PMID: 26303746 DOI: 10.1016/j.jsbmb.2015.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 01/04/2023]
Abstract
Elevated levels of aldosterone are associated with arterial hypertension, congestive heart failure, chronic kidney disease, and obesity. Aldosterone is produced predominantly in the zona glomerulosa of the cortex of the adrenal gland by the enzyme aldosterone synthase (CYP11B2). Treatment of the above indications by decreasing production of aldosterone is thought to be of therapeutic benefit by lessening the deleterious effects of aldosterone mediated through both the mineralocorticoid receptor and also through so called non-genomic pathways. However, inhibition of the highly similar enzyme, CYP11B1, which is responsible for the production of cortisol, must be avoided in the development of clinically useful aldosterone synthase inhibitors due to the resulting impairment of the cortisol-induced stress response. In efforts to assess the interactions of compounds with the CYP11B enzymes, a variety of cell-based inhibitor screening assays for both CYP11B1 and CYP11B2 have been reported. Herein we report details of assays employing both cynomolgus monkey adrenal homogenate (CAH) and human adrenal homogenate (HAH) as sources of CYP11B1 and CYP11B2 enzymes. Utilizing both CAH and HAH, we have characterized the kinetics of the CYP11B1-mediated conversion of 11-deoxycortisol to cortisol and the CYP11B2-mediated oxidation of corticosterone to aldosterone. Inhibition assays for both CYP11B1 and CYP11B2 were subsequently developed. Based on a comparison of human and monkey amino acid sequences, kinetics data, and inhibition values derived from the HAH and CAH assays, evidence is provided in support of using cynomolgus monkey tissue-derived cell homogenates as suitable surrogates for the human enzymes.
Collapse
Affiliation(s)
- Matthew A Cerny
- Boehringer Ingelheim Pharmaceuticals, Inc., Department of Medicinal Chemistry, Drug Discovery Support (DMPK), USA.
| | - Alexander Csengery
- Boehringer Ingelheim Pharmaceuticals, Inc., Department of Medicinal Chemistry, Drug Discovery Support (DMPK), USA
| | - Jennifer Schmenk
- Boehringer Ingelheim Pharmaceuticals, Inc., Department of Medicinal Chemistry, Drug Discovery Support (DMPK), USA
| | - Kosea Frederick
- Boehringer Ingelheim Pharmaceuticals, Inc., Department of Medicinal Chemistry, Drug Discovery Support (DMPK), USA
| |
Collapse
|
24
|
Hoyt SB, Petrilli W, London C, Liang GB, Tata J, Hu Q, Yin L, van Koppen CJ, Hartmann RW, Struthers M, Wisniewski T, Ren N, Bopp C, Sok A, Cai TQ, Stribling S, Pai LY, Ma X, Metzger J, Verras A, McMasters D, Chen Q, Tung E, Tang W, Salituro G, Buist N, Clemas J, Zhou G, Gibson J, Maxwell CA, Lassman M, McLaughlin T, Castro-Perez J, Szeto D, Forrest G, Hajdu R, Rosenbach M, Xiong Y. Discovery of Triazole CYP11B2 Inhibitors with in Vivo Activity in Rhesus Monkeys. ACS Med Chem Lett 2015; 6:861-5. [PMID: 26288685 DOI: 10.1021/acsmedchemlett.5b00048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/17/2015] [Indexed: 12/21/2022] Open
Abstract
Hit-to-lead efforts resulted in the discovery of compound 19, a potent CYP11B2 inhibitor that displays high selectivity vs related CYPs, good pharmacokinetic properties in rat and rhesus, and lead-like physical properties. In a rhesus pharmacodynamic model, compound 19 displays robust, dose-dependent aldosterone lowering efficacy, with no apparent effect on cortisol levels.
Collapse
Affiliation(s)
- Scott B. Hoyt
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Whitney Petrilli
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Clare London
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gui-Bai Liang
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jim Tata
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Qingzhong Hu
- Department
of Pharmaceutical and Medicinal Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus
C2-3, D-66123 Saarbrücken, Germany
| | - Lina Yin
- Department
of Pharmaceutical and Medicinal Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus
C2-3, D-66123 Saarbrücken, Germany
- ElexoPharm GmbH, Im Stadtwald, D-66123 Saarbrücken, Germany
| | | | - Rolf W. Hartmann
- Department
of Pharmaceutical and Medicinal Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus
C2-3, D-66123 Saarbrücken, Germany
| | - Mary Struthers
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Tom Wisniewski
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Ning Ren
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Charlene Bopp
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Andrea Sok
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Tian-Quan Cai
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Sloan Stribling
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Lee-Yuh Pai
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Xiuying Ma
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Joe Metzger
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Andreas Verras
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Daniel McMasters
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Qing Chen
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Elaine Tung
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Wei Tang
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gino Salituro
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Nicole Buist
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Joe Clemas
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gaochao Zhou
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jack Gibson
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | | | - Mike Lassman
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | | | - Jose Castro-Perez
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Daphne Szeto
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gail Forrest
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Richard Hajdu
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Mark Rosenbach
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Yusheng Xiong
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| |
Collapse
|
25
|
Daniel E, Newell-Price JDC. Therapy of endocrine disease: steroidogenesis enzyme inhibitors in Cushing's syndrome. Eur J Endocrinol 2015; 172:R263-80. [PMID: 25637072 DOI: 10.1530/eje-14-1014] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/29/2015] [Indexed: 11/08/2022]
Abstract
Steroidogenesis enzyme inhibitors are the mainstay of medical therapy in Cushing's syndrome (CS). Ketoconazole (KTZ) and metyrapone are the most commonly used agents. Although there is considerable experience of their use in individual specialist centres, these drugs have not been rigorously tested in prospective clinical trials. Clinicians face uncertainties and concerns with respect to the safety profile of these agents, and best means to monitor effect. We review steroidogenesis inhibitors in the management of CS, including older agents (KTZ, metyrapone, etomidate and mitotane) and those currently under development (LCI699, non-racemic KTZ), and offer a practical approach for their use in clinical practice.
Collapse
Affiliation(s)
- Eleni Daniel
- Department of Human MetabolismAcademic Unit of EndocrinologyDepartment of Endocrinology, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - John D C Newell-Price
- Department of Human MetabolismAcademic Unit of EndocrinologyDepartment of Endocrinology, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| |
Collapse
|
26
|
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
|
27
|
Hoyt SB, Park MK, London C, Xiong Y, Tata J, Bennett DJ, Cooke A, Cai J, Carswell E, Robinson J, MacLean J, Brown L, Belshaw S, Clarkson TR, Liu K, Liang GB, Struthers M, Cully D, Wisniewski T, Ren N, Bopp C, Sok A, Cai TQ, Stribling S, Pai LY, Ma X, Metzger J, Verras A, McMasters D, Chen Q, Tung E, Tang W, Salituro G, Buist N, Kuethe J, Rivera N, Clemas J, Zhou G, Gibson J, Maxwell CA, Lassman M, McLaughlin T, Castro-Perez J, Szeto D, Forrest G, Hajdu R, Rosenbach M, Ali A. Discovery of Benzimidazole CYP11B2 Inhibitors with in Vivo Activity in Rhesus Monkeys. ACS Med Chem Lett 2015; 6:573-8. [PMID: 26005536 DOI: 10.1021/acsmedchemlett.5b00054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/07/2015] [Indexed: 11/28/2022] Open
Abstract
We report the discovery of a benzimidazole series of CYP11B2 inhibitors. Hit-to-lead and lead optimization studies identified compounds such as 32, which displays potent CYP11B2 inhibition, high selectivity versus related CYP targets, and good pharmacokinetic properties in rat and rhesus. In a rhesus pharmacodynamic model, 32 produces dose-dependent aldosterone lowering efficacy, with no apparent effect on cortisol levels.
Collapse
Affiliation(s)
- Scott B. Hoyt
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Min K. Park
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Clare London
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Yusheng Xiong
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jim Tata
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | | | - Andrew Cooke
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - Jiaqiang Cai
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - Emma Carswell
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - John Robinson
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - John MacLean
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - Lindsay Brown
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - Simone Belshaw
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - Thomas R. Clarkson
- Merck Research Laboratories, Newhouse, Lanarkshire ML1 5SH, United Kingdom
| | - Kun Liu
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gui-Bai Liang
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Mary Struthers
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Doris Cully
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Tom Wisniewski
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Ning Ren
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Charlene Bopp
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Andrea Sok
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Tian-Quan Cai
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Sloan Stribling
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Lee-Yuh Pai
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Xiuying Ma
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Joe Metzger
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Andreas Verras
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Daniel McMasters
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Qing Chen
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Elaine Tung
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Wei Tang
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gino Salituro
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Nicole Buist
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jeff Kuethe
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Nelo Rivera
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Joe Clemas
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gaochao Zhou
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jack Gibson
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | | | - Mike Lassman
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | | | - Jose Castro-Perez
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Daphne Szeto
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gail Forrest
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Richard Hajdu
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Mark Rosenbach
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Amjad Ali
- Merck Research Laboratories, Rahway, New Jersey 07065, United States
| |
Collapse
|
28
|
Ménard J, Rigel DF, Watson C, Jeng AY, Fu F, Beil M, Liu J, Chen W, Hu CW, Leung-Chu J, LaSala D, Liang G, Rebello S, Zhang Y, Dole WP. Aldosterone synthase inhibition: cardiorenal protection in animal disease models and translation of hormonal effects to human subjects. J Transl Med 2014; 12:340. [PMID: 25491597 PMCID: PMC4301837 DOI: 10.1186/s12967-014-0340-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/22/2014] [Indexed: 01/31/2023] Open
Abstract
Background Aldosterone synthase inhibition provides the potential to attenuate both the mineralocorticoid receptor-dependent and independent actions of aldosterone. In vitro studies with recombinant human enzymes showed LCI699 to be a potent, reversible, competitive inhibitor of aldosterone synthase (Ki = 1.4 ± 0.2 nmol/L in humans) with relative selectivity over 11β-hydroxylase. Methods Hormonal effects of orally administered LCI699 were examined in rat and monkey in vivo models of adrenocorticotropic hormone (ACTH) and angiotensin-II-stimulated aldosterone release, and were compared with the mineralocorticoid receptor antagonist eplerenone in a randomized, placebo-controlled study conducted in 99 healthy human subjects. The effects of LCI699 and eplerenone on cardiac and renal sequelae of aldosterone excess were investigated in a double-transgenic rat (dTG rat) model overexpressing human renin and angiotensinogen. Results Rat and monkey in vivo models of stimulated aldosterone release predicted human dose– and exposure–response relationships, but overestimated the selectivity of LCI699 in humans. In the dTG rat model, LCI699 dose-dependently blocked increases in aldosterone, prevented development of cardiac and renal functional abnormalities independent of blood pressure changes, and prolonged survival. Eplerenone prolonged survival to a similar extent, but was less effective in preventing cardiac and renal damage. In healthy human subjects, LCI699 0.5 mg selectively reduced plasma and 24 h urinary aldosterone by 49 ± 3% and 39 ± 6% respectively (Day 1, mean ± SEM; P < 0.001 vs placebo), which was associated with natriuresis and an increase in plasma renin activity. Doses of LCI699 greater than 1 mg inhibited basal and ACTH-stimulated cortisol. Eplerenone 100 mg increased plasma and 24 h urinary aldosterone while stimulating natriuresis and increasing renin activity. In contrast to eplerenone, LCI699 increased the aldosterone precursor 11-deoxycorticosterone and urinary potassium excretion. Conclusions These results provide new insights into the cardiac and renal effects of inhibiting aldosterone synthase in experimental models and translation of the hormonal effects to humans. Selective inhibition of aldosterone synthase appears to be a promising approach to treat diseases associated with aldosterone excess. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0340-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joël Ménard
- Université Paris Descartes, Faculté de Médecine and INSERM/AP-HP Clinical Investigation Center, Georges Pompidou Hospital, Paris, France.
| | - Dean F Rigel
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Catherine Watson
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Arco Y Jeng
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA. .,Current address: Golda Och Academy, 1418 Pleasant Valley Way, West Orange, NJ, 07052, USA.
| | - Fumin Fu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Michael Beil
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Jing Liu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Wei Chen
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Chii-Whei Hu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | | | - Daniel LaSala
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Guiqing Liang
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Sam Rebello
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Yiming Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - William P Dole
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| |
Collapse
|
29
|
Raff H, Gehrand A, Bruder ED, Hoffman MJ, Engeland WC, Moreno C. Renin knockout rat: control of adrenal aldosterone and corticosterone synthesis in vitro and adrenal gene expression. Am J Physiol Regul Integr Comp Physiol 2014; 308:R73-7. [PMID: 25394830 DOI: 10.1152/ajpregu.00440.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The classic renin-angiotensin system is partly responsible for controlling aldosterone secretion from the adrenal cortex via the peptide angiotensin II (ANG II). In addition, there is a local adrenocortical renin-angiotensin system that may be involved in the control of aldosterone synthesis in the zona glomerulosa (ZG). To characterize the long-term control of adrenal steroidogenesis, we utilized adrenal glands from renin knockout (KO) rats and compared steroidogenesis in vitro and steroidogenic enzyme expression to wild-type (WT) controls (Dahl S rat). Adrenal capsules (ZG; aldosterone production) and subcapsules [zona reticularis/fasciculata (ZFR); corticosterone production] were separately dispersed and studied in vitro. Plasma renin activity and ANG II concentrations were extremely low in the KO rats. Basal and cAMP-stimulated aldosterone production was significantly reduced in renin KO ZG cells, whereas corticosterone production was not different between WT and KO ZFR cells. As expected, adrenal renin mRNA expression was lower in the renin KO compared with the WT rat. Real-time PCR and immunohistochemical analysis showed a significant decrease in P450aldo (Cyp11b2) mRNA and protein expression in the ZG from the renin KO rat. The reduction in aldosterone synthesis in the ZG of the renin KO adrenal seems to be accounted for by a specific decrease in P450aldo and may be due to the absence of chronic stimulation of the ZG by circulating ANG II or to a reduction in locally released ANG II within the adrenal gland.
Collapse
Affiliation(s)
- Hershel Raff
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Aurora Research Institute, Milwaukee, Wisconsin; Departments of Medicine, Surgery, and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin;
| | - Ashley Gehrand
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Aurora Research Institute, Milwaukee, Wisconsin
| | - Eric D Bruder
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Aurora Research Institute, Milwaukee, Wisconsin
| | - Matthew J Hoffman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - William C Engeland
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Carol Moreno
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| |
Collapse
|
30
|
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
|
31
|
Rhesus monkey model for concurrent analyses of in vivo selectivity, pharmacokinetics and pharmacodynamics of aldosterone synthase inhibitors. J Pharmacol Toxicol Methods 2014; 71:137-46. [PMID: 25304940 DOI: 10.1016/j.vascn.2014.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/15/2014] [Accepted: 09/30/2014] [Indexed: 11/21/2022]
Abstract
INTRODUCTION In vivo profiles of aldosterone synthase inhibitors (ASIs) have been investigated utilizing various rodent models. Due to lack of CYP17 activity, rodents produce corticosterone rather than cortisol as that of humans, which raised concern to their effectiveness in translational pharmacological characterization of ASI. METHODS A rhesus monkey model that combines a low sodium diet with adrenocorticotropin (ACTH) treatment was developed. Plasma concentrations of steroid metabolites associated with reactions catalyzed by CYP11B2 and CYP11B1 were measured concurrently by a UPLC/MS method. RESULTS Plasma concentration of aldosterone in regular diet fed rhesus monkeys was low at 109pg/mL. Aldosterone concentrations were increased to 252pg/mL when animals were maintained on a low sodium diet for 3weeks, and to 300pg/mL with ACTH treatment at 0.3mg/kg. The combination of low sodium diet with ACTH treatment further increased plasma concentration of aldosterone to 730pg/mL and other steroid metabolites at various levels. Intravenous administration of ASI, fadrozole (0.001-1mg/kg) or LCI699 (0.003-3mg/kg), led to dose-dependent reductions in aldosterone and 18-hydroxycorticosterone, increases in 11-deoxycorticosterone and 11-deoxycortisol, and bell-shaped changes in cortisol and corticosterone. In vivo selectivity of CYP11B2/CYP11B1 for fadrazole was 26-fold and LCI-699 was 27-fold, which was consistent with relative selectivity using in vitro values from recombinant cells transfected with rhesus monkey CYP11B2 and CYP11B1. DISCUSSION This model enables concurrent characterization of pharmacokinetics, pharmacodynamics and selectivity of CYP11B2 over CYP11B1 inhibition in the same animal. It may be used as a translational model for pharmacological characterization of ASI.
Collapse
|
32
|
Hamlyn JM, Linde CI, Gao J, Huang BS, Golovina VA, Blaustein MP, Leenen FHH. Neuroendocrine humoral and vascular components in the pressor pathway for brain angiotensin II: a new axis in long term blood pressure control. PLoS One 2014; 9:e108916. [PMID: 25275393 PMCID: PMC4183521 DOI: 10.1371/journal.pone.0108916] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/26/2014] [Indexed: 11/29/2022] Open
Abstract
Central nervous system (CNS) administration of angiotensin II (Ang II) raises blood pressure (BP). The rise in BP reflects increased sympathetic outflow and a slower neuromodulatory pressor mechanism mediated by CNS mineralocorticoid receptors (MR). We investigated the hypothesis that the sustained phase of hypertension is associated also with elevated circulating levels of endogenous ouabain (EO), and chronic stimulation of arterial calcium transport proteins including the sodium-calcium exchanger (NCX1), the type 6 canonical transient receptor potential protein (TRPC6), and the sarcoplasmic reticulum calcium ATPase (SERCA2). Wistar rats received a chronic intra-cerebroventricular infusion of vehicle (C) or Ang II (A, 2.5 ng/min, for 14 days) alone or combined with the MR blocker, eplerenone (A+E, 5 µg/day), or the aldosterone synthase inhibitor, FAD286 (A+F, 25 µg/day). Conscious mean BP increased (P<0.05) in A (123±4 mm Hg) vs all other groups. Blood, pituitary and adrenal samples were taken for EO radioimmunoassay (RIA), and aortas for NCX1, TRPC6 and SERCA2 immunoblotting. Central infusion of Ang II raised plasma EO (0.58±0.08 vs C 0.34±0.07 nM (P<0.05), but not in A + E and A + F groups as confirmed by off-line liquid chromatography (LC)-RIA and LC-multistage mass spectrometry. Two novel isomers of EO were elevated by Ang II; the second less polar isomer increased >50-fold in the A+F group. Central Ang II increased arterial expression of NCX1, TRPC6 and SERCA2 (2.6, 1.75 and 3.7-fold, respectively; P<0.01)) but not when co-infused with E or F. Adrenal and pituitary EO were unchanged. We conclude that brain Ang II activates a CNS-humoral axis involving plasma EO. The elevated EO reprograms peripheral ion transport pathways known to control arterial Na+ and Ca2+ homeostasis; this increases contractility and augments sympathetic effects. The new axis likely contributes to the chronic pressor effect of brain Ang II.
Collapse
Affiliation(s)
- John M. Hamlyn
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
- * E-mail:
| | - Cristina I. Linde
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Junjie Gao
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Bing S. Huang
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Vera A. Golovina
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Mordecai P. Blaustein
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | | |
Collapse
|
33
|
Hu Q, Yin L, Hartmann RW. Aldosterone Synthase Inhibitors as Promising Treatments for Mineralocorticoid Dependent Cardiovascular and Renal Diseases. J Med Chem 2014; 57:5011-22. [DOI: 10.1021/jm401430e] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Qingzhong Hu
- Pharmaceutical and Medicinal
Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, D-66123 Saarbrücken, Germany
| | - Lina Yin
- Pharmaceutical and Medicinal
Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, D-66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal
Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, D-66123 Saarbrücken, Germany
| |
Collapse
|
34
|
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: 34] [Impact Index Per Article: 2.8] [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
|
35
|
Stehlin E, Malpas SC, Budgett DM, Barrett CJ, McCormick D, Whalley G, Fu F, Beil M, Rigel DF, Guild SJ. Chronic measurement of left ventricular pressure in freely moving rats. J Appl Physiol (1985) 2013; 115:1672-82. [DOI: 10.1152/japplphysiol.00683.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Measurements of left ventricular pressure (LVP) in conscious freely moving animals are uncommon, yet could offer considerable opportunity for understanding cardiovascular disease progression and treatment. The aim of this study was to develop surgical methods and validate the measurements of a new high-fidelity, solid-state pressure-sensor telemetry device for chronically measuring LVP and dP/d t in rats. The pressure-sensor catheter tip (2-Fr) was inserted into the left ventricular chamber through the apex of the heart, and the telemeter body was implanted in the abdomen. Data were measured up to 85 days after implant. The average daytime dP/d t max was 9,444 ± 363 mmHg/s, ranging from 7,870 to 10,558 mmHg/s ( n = 7). A circadian variation in dP/d t max and heart rate (HR) was observed with an average increase during the night phase in dP/d t max of 918 ± 84 mmHg/s, and in HR of 38 ± 3 bpm. The β-adrenergic-agonist isoproterenol, β1-adrenergic agonist dobutamine, Ca2+ channel blocker verapamil, and the calcium sensitizer levosimendan were administered throughout the implant period, inducing dose-dependent time course changes and absolute changes in dP/d t max of −6,000 to +13,000 mmHg/s. The surgical methods and new technologies demonstrated long-term stability, sensitivity to circadian variation, and the ability to measure large drug-induced changes, validating this new solution for chronic measurement of LVP in conscious rats.
Collapse
Affiliation(s)
- Ellyce Stehlin
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Simon C. Malpas
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Physiology, University of Auckland, Auckland, New Zealand,
- Millar Inc, Auckland, New Zealand
| | - David M. Budgett
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Millar Inc, Auckland, New Zealand
| | - Carolyn J. Barrett
- Department of Physiology, University of Auckland, Auckland, New Zealand,
| | - Daniel McCormick
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Millar Inc, Auckland, New Zealand
| | - Gillian Whalley
- Faculty of Social and Health Sciences, Unitec, Auckland, New Zealand; and
| | - Fumin Fu
- Novartis Institutes for BioMedical Research, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Michael Beil
- Novartis Institutes for BioMedical Research, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Dean F. Rigel
- Novartis Institutes for BioMedical Research, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Sarah-Jane Guild
- Department of Physiology, University of Auckland, Auckland, New Zealand,
- Millar Inc, Auckland, New Zealand
| |
Collapse
|
36
|
Ritz E, Pitt B. Mineralocorticoid receptor blockade-a novel approach to fight hyperkalaemia in chronic kidney disease. Clin Kidney J 2013; 6:464-8. [PMID: 26120440 PMCID: PMC4438399 DOI: 10.1093/ckj/sft084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 06/28/2013] [Indexed: 01/06/2023] Open
Abstract
Hyperkalaemia continues to be a major hazard of mineralocorticoid receptor blockade in an effort to retard the progression of chronic kidney disease (CKD). In cardiac patients on mineralocorticoid receptor blockade, RLY-5016 which captures K+ in the colon has been effective in reducing the risk of hyperkalaemia. This compound might be useful in CKD as well.
Collapse
Affiliation(s)
- E Ritz
- Nierenzentrum , University of Heidelberg , Heidelberg , Germany
| | - B Pitt
- Internal Medicine and Cardiovascular Disease , University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
37
|
Besedovsky L, Born J, Lange T. Endogenous glucocorticoid receptor signaling drives rhythmic changes in human T-cell subset numbers and the expression of the chemokine receptor CXCR4. FASEB J 2013; 28:67-75. [PMID: 24051033 DOI: 10.1096/fj.13-237958] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In humans, numbers of circulating naive T cells strongly decline in the morning, which was suggested to be mediated by cortisol, inducing a CXCR4 up-regulation with a subsequent extravasation of the cells. As a systematic evaluation of this assumption is lacking, we investigated in two human placebo-controlled studies the effects of the glucocorticoid receptor (GR) antagonist mifepristone (200 mg orally at 23:00) and of suppressing endogenous cortisol with metyrapone (1 g orally at 04:00) on temporal changes in CXCR4 expression and numbers of different T-cell subsets using flow cytometry. Mifepristone attenuated, and metyrapone completely blocked, the morning increase in CXCR4 expression on naive T cells. In parallel, both substances also hindered the decline in naive T-cell numbers with this effect, however, being less apparent after mifepristone. We identified, and confirmed in additional in vitro studies, a partial agonistic GR effect of mifepristone at night (i.e., between 02:00 and 03:30) that could explain the lower antagonistic efficacy of the substance on CXCR4 expression and naive T-cell counts. CXCR4 expression emerged to be a most sensitive marker of GR signaling. Our studies jointly show that endogenous cortisol, specifically via GR activation, causes the morning increase in CXCR4 expression and the subsequent extravasation of naive T cells, thus revealing an important immunological function of the morning cortisol rise.
Collapse
Affiliation(s)
- Luciana Besedovsky
- 1Department of Neuroendocrinology, University of Lübeck, Ratzeburger Allee 160, Lübeck 23538, Germany.
| | | | | |
Collapse
|
38
|
Huang BS, White RA, Ahmad M, Leenen FHH. Role of brain corticosterone and aldosterone in central angiotensin II-induced hypertension. Hypertension 2013; 62:564-71. [PMID: 23856493 DOI: 10.1161/hypertensionaha.113.01557] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circulating angiotensin II (Ang II) activates a central aldosterone-mineralocorticoid receptor neuromodulatory pathway, which mediates most of the Ang II-induced hypertension. This study examined whether specific central infusion of Ang II also activates this central aldosterone-mineralocorticoid receptor pathway. Intracerebroventricular infusion of Ang II at 1.0, 2.5, and 12.5 ng/min for 2 weeks caused dose-related increases in water intake, Ang II concentration in the cerebrospinal fluid, and blood pressure. Intracerebroventricular Ang II, at 2.5 and 12.5 ng/min, increased hypothalamic aldosterone and corticosterone, as well as plasma aldosterone and corticosterone without affecting plasma Ang II levels. Intracerebroventricular infusion of the aldosterone synthase inhibitor FAD286-but not the mineralocorticoid receptor blocker eplerenone-inhibited by ≈60% the Ang II-induced increase in hypothalamic aldosterone. Both blockers attenuated by ≈50% the increase in plasma aldosterone and corticosterone with only minimal effects on hypothalamic corticosterone. By telemetry, intracerebroventricular infusion of Ang II maximally increased blood pressure within the first day with no further increase over the next 2 weeks. Intracerebroventricular infusion of FAD286 or eplerenone did not affect the initial pressor responses but similarly prevented 60% to 70% of the chronic pressor responses to intracerebroventricular infusion of Ang II. These results indicate distinctly different patterns of blood pressure increase by circulating versus central Ang II and support the involvement of a brain aldosterone-mineralocorticoid receptor-activated neuromodulatory pathway in the chronic hypertension caused by both circulating and central Ang II.
Collapse
Affiliation(s)
- Bing S Huang
- Hypertension Unit, University of Ottawa Heart Institute, H3238-40 Ruskin St, Ottawa, Ontario K1Y 4W7, Canada
| | | | | | | |
Collapse
|
39
|
Scheuer DA. Stimulation of aldosterone synthesis by angiotensin II in the brain: support for positive feedback in hypertension? Hypertension 2013; 62:459-60. [PMID: 23856491 DOI: 10.1161/hypertensionaha.113.01649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Berger ML, Hammerschmidt F, Qian R, Hahner S, Schirbel A, Stichelberger M, Schibli R, Yu J, Arion VB, Woschek A, Öhler E, Zolle IM. [(3)H]metyrapol and 4-[(131)i]iodometomidate label overlapping, but not identical, binding sites on rat adrenal membranes. Mol Pharm 2013; 10:1119-30. [PMID: 23343186 DOI: 10.1021/mp3006227] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metyrapone, metyrapol, and etomidate are competitive inhibitors of 11-deoxycorticosterone hydroxylation by 11β-hydroxylase. [(3)H]Metyrapol and 4-[(131)I]iodometomidate bind with high affinity to membranes prepared from bovine and rat adrenals. Here we report inhibitory potencies of several compounds structurally related to one or both of these adrenostatic drugs, against the binding of both radioligands to rat adrenal membranes. While derivatives of etomidate inhibited the binding of both radioligands with similar potencies, derivatives of metyrapone inhibited the binding of 4-[(131)I]iodometomidate about 10 times weaker than the binding of [(3)H]metyrapol. By X-ray structure analysis the absolute configuration of (+)-1-(2-fluorophenyl)-2-methyl-2-(pyridin-3-yl)-1-propanol [(+)-11, a derivative of metyrapol] was established as (R). We introduce 1-(2-fluorophenyl)-2-methyl-2-(pyridin-3-yl)-1-propanone (9; Ki = 6 nM), 2-(1-imidazolyl)-2-methyl-1-phenyl-1-propanone (13; 2 nM), and (R)-(+)-[1-(4-iodophenyl)ethyl]-1H-imidazole (34; 4 nM) as new high affinity ligands for the metyrapol binding site on 11β-hydroxylase and discuss our results in relation to a proposed active site model of 11β-hydroxylase.
Collapse
Affiliation(s)
- Michael L Berger
- Center for Brain Research, Medical University of Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Management of hyperkalaemia consequent to mineralocorticoid-receptor antagonist therapy. Nat Rev Nephrol 2012; 8:691-9. [DOI: 10.1038/nrneph.2012.217] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
42
|
Rajamohan SB, Raghuraman G, Prabhakar NR, Kumar GK. NADPH oxidase-derived H(2)O(2) contributes to angiotensin II-induced aldosterone synthesis in human and rat adrenal cortical cells. Antioxid Redox Signal 2012; 17:445-59. [PMID: 22214405 PMCID: PMC3365360 DOI: 10.1089/ars.2011.4176] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The Renin-Angiotensin-Aldosterone-System plays a pivotal role in hypertension. Angiotensin II (Ang II) is a major regulator of aldosterone synthesis and secretion, and it is known to facilitate reactive oxygen species (ROS) generation in many cell types. AIMS Here, we assessed the role of ROS signaling in Ang II-induced aldosterone synthesis by focusing on the regulation of aldosterone synthase (CYP11B2), a cytochrome P450 oxidase that catalyzes the final step in aldosterone biosynthetic pathway. RESULTS Ang II increased CYP11B2 activity, mRNA and protein with a concomitant elevation of 6-Carboxy- 2',7'-dichlorodihydrofluorescein diacetate fluorescence, malondialdehyde and protein carbonyl levels (indices of ROS), NADPH oxidase (Nox) activity, and H(2)O(2) levels in human and rat adrenal cortical cells. The expression of nuclear receptor related 1 protein, a transcription factor known to regulate CYP11B2 expression, was also augmented by Ang II. These Ang II-evoked effects were either abolished or attenuated by pretreatment of cells with either Ang II type I receptor (AT(1)R) antagonist, or antioxidants or Nox inhibitor or siRNA silencing of Nox1, 2 and 4, or inhibitors of phospholipase C and protein kinase C. Exogenous H(2)O(2) mimicked the facilitatory effects of Ang II on CYP11B2 activity, mRNA, and protein expression, and these changes were significantly reduced by PEG-catalase. INNOVATION ROS, particularly H(2)O(2), is identified as a key regulator of aldosterone production. CONCLUSION Our results suggest that Ang II facilitates CYP11B2 activity and the ensuing aldosterone production via activation of AT(1)R-Nox-H(2)O(2) signaling pathway.
Collapse
Affiliation(s)
- Senthilkumar B Rajamohan
- Department of Medicine, Institute for Integrative Physiology, University of Chicago, Chicago, IL 60637-1470, USA
| | | | | | | |
Collapse
|
43
|
|
44
|
Deliyanti D, Miller AG, Tan G, Binger KJ, Samson AL, Wilkinson-Berka JL. Neovascularization is attenuated with aldosterone synthase inhibition in rats with retinopathy. Hypertension 2012; 59:607-13. [PMID: 22275532 DOI: 10.1161/hypertensionaha.111.188136] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neovascularization is a hallmark feature of retinopathy of prematurity and diabetic retinopathy. Type 1 angiotensin receptor blockade reduces neovascularization in experimental retinopathy of prematurity, known as oxygen-induced retinopathy (OIR). We investigated in OIR whether inhibiting aldosterone with the aldosterone synthase inhibitor FAD286 reduced neovascularization as effectively as angiotensin receptor blockade (valsartan). OIR was induced in neonatal Sprague-Dawley rats, and they were treated with FAD286 (30 mg/kg per day), valsartan (10 mg/kg per day), or FAD286+valsartan. The cellular sources of aldosterone synthase, the mineralocorticoid receptor, and 11β-hydroxysteroid dehydrogenase 2 were evaluated in retinal cells involved in neovascularization (primary endothelial cells, pericytes, microglia, ganglion cells, and glia). In OIR, FAD286 reduced neovascularization and neovascular tufts by 89% and 67%, respectively, and normalized the increase in vascular endothelial growth factor mRNA (1.74-fold) and protein (4.74-fold) and was as effective as valsartan and FAD286+valsartan. In retina, aldosterone synthase mRNA was reduced with FAD286 but not valsartan. Aldosterone synthase was detected in microglia, ganglion cells, and glia, whereas mineralocorticoid receptor and 11β-hydroxysteroid dehydrogenase 2 were present in all of the cell types studied. Given the location of aldosterone synthase in microglia and their contribution to retinal inflammation and neovascularization in OIR, the effects of FAD286 on microglial density were studied. The increase in microglial density (ionized calcium binding adaptor protein 1 immunolabeling) in OIR was reduced with all of the treatments. In OIR, FAD286 reduced the increase in mRNA for tumor necrosis factor-α, intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and monocyte chemoattractant molecule 1. These findings indicate that aldosterone inhibition may be a potential treatment for retinal neovascularization.
Collapse
Affiliation(s)
- Devy Deliyanti
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
45
|
Bramlage P, Turgonyi E, Montalescot G. Aldosterone blockade: current research and future trends. Eur Heart J Suppl 2011. [DOI: 10.1093/eurheartj/sur005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
46
|
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; 56:831-8. [PMID: 20837883 DOI: 10.1161/hypertensionaha.110.157271] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We report the first administration of an orally active aldosterone synthase inhibitor, LCI699, to 14 patients with primary aldosteronism. After a 2-week placebo run-in, patients received oral LCI699 (0.5 mg BID) for 2 weeks, LCI699 (1.0 mg BID) for 2 weeks, and placebo for 1 week. We assessed changes in hormone concentrations, plasma potassium levels, and 24-hour ambulatory systolic blood pressure and safety. The supine plasma aldosterone concentration decreased from 540 pmol/L (95% CI: 394 to 739 pmol/L) to 171 pmol/L (95% CI: 128 to 230 pmol/L) after 0.5 mg of LCI699 (-68%; P<0.0001) and to 133 pmol/L (95% CI: 100 to 177 pmol/L) after 1.0 mg of LCI699 (-75%; P<0.0001). Plasma 11-deoxycorticosterone concentrations increased by 710% after 0.5 mg of LCI699 (P<0.0001) and by 1427% after 1.0 mg of LCI699 (P<0.0001). The plasma potassium concentration increased from 3.27±0.31 to 4.03±0.33 mmol/L (P<0.0001) after only 1 week on 0.5 mg of LCI699. Twenty-four-hour ambulatory systolic blood pressure decreased by -4.1 mm Hg (95% CI: -8.1 to -0.1 mm Hg) after 4 weeks of treatment (P=0.046). Basal plasma cortisol concentrations remained unchanged, whereas plasma adrenocorticotropic hormone concentrations increased by 35% after 0.5 mg of LCI699 (P=0.08) and by 113% after 1.0 mg of LCI699 (P<0.0001), and the plasma cortisol response to an adrenocorticotropic hormone test was blunted. All of the variables except plasma 11-deoxycorticosterone concentration returned to initial levels after the placebo. LCI699 was well tolerated. In conclusion, the administration of LCI699, up to 1.0 mg BID, effectively and safely inhibits aldosterone synthase in patients with primary aldosteronism. This 4-week treatment corrected the hypokalemia and mildly decreased blood pressure. The effects on the glucocorticoid axis were consistent with a latent inhibition of cortisol synthesis.
Collapse
Affiliation(s)
- Laurence Amar
- Université Paris Descartes, Faculté de Médecine, Paris, France
| | | | | | | | | | | |
Collapse
|