1
|
Zhou C, Zhou Y, Liu L, Jiang H, Wei H, Zhou C, Ji X. Progress and recognition of idiopathic intracranial hypertension: A narrative review. CNS Neurosci Ther 2024; 30:e14895. [PMID: 39097911 PMCID: PMC11298205 DOI: 10.1111/cns.14895] [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: 02/11/2024] [Revised: 06/03/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Idiopathic intracranial hypertension (IIH) mainly affects obese young women, causing elevated intracranial pressure, headaches, and papilledema, risking vision loss and severe headaches. Despite weight loss as the primary treatment, the underlying mechanisms remain unclear. Recent research explores novel therapeutic targets. AIMS This review aimed to provide a comprehensive understanding of IIH's pathophysiology and clinical features to inform pathogenesis and improve treatment strategies. METHODS Recent publications on IIH were searched and summarized using PubMed, Web of Science, and MEDLINE. RESULTS The review highlights potential pathomechanisms and therapeutic advances in IIH. CONCLUSION IIH incidence is rising, with growing evidence linking it to metabolic and hormonal disturbances. Early diagnosis and treatment remain challenging.
Collapse
Affiliation(s)
- Chenxia Zhou
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Neurology and Intracranial Hypertension and Cerebral Venous Disease CenterNational Health Commission of China, Xuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yifan Zhou
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersBeijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision Medicine, Capital Medical UniversityBeijingChina
| | - Lu Liu
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Neurology and Intracranial Hypertension and Cerebral Venous Disease CenterNational Health Commission of China, Xuanwu Hospital, Capital Medical UniversityBeijingChina
- Zhongguancun Xirui Institute of Precision Medicine for Heart and Brain TumorsBeijingChina
| | - Huimin Jiang
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersBeijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision Medicine, Capital Medical UniversityBeijingChina
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data‐Based Precision MedicineSchool of Biological Science and Medical Engineering, Beihang UniversityBeijingChina
| | - Chen Zhou
- Neurology and Intracranial Hypertension and Cerebral Venous Disease CenterNational Health Commission of China, Xuanwu Hospital, Capital Medical UniversityBeijingChina
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersBeijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision Medicine, Capital Medical UniversityBeijingChina
| | - Xunming Ji
- Neurology and Intracranial Hypertension and Cerebral Venous Disease CenterNational Health Commission of China, Xuanwu Hospital, Capital Medical UniversityBeijingChina
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersBeijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision Medicine, Capital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
2
|
Wei W, Xu Q, Wu L, Gong G, Tian Y, Huang H, Li Z. Drug development and potential targets for Cushing's syndrome. Eur J Med Chem 2024; 270:116333. [PMID: 38569434 DOI: 10.1016/j.ejmech.2024.116333] [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: 12/15/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
Cushing's syndrome (CS) is a complex disorder characterized by the excessive secretion of cortisol, with Cushing's disease (CD), particularly associated with pituitary tumors, exhibiting heightened morbidity and mortality. Although transsphenoidal pituitary surgery (TSS) stands as the primary treatment for CD, there is a crucial need to optimize patient prognosis. Current medical therapy serves as an adjunctive measure due to its unsatisfactory efficacy and unpredictable side effects. In this comprehensive review, we delve into recent advances in understanding the pathogenesis of CS and explore therapeutic options by conducting a critical analysis of potential drug targets and candidates. Additionally, we provide an overview of the design strategy employed in previously reported candidates, along with a summary of structure-activity relationship (SAR) analyses and their biological efficacy. This review aims to contribute valuable insights to the evolving landscape of CS research, shedding light on potential avenues for therapeutic development.
Collapse
Affiliation(s)
- Wei Wei
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Qianqian Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Liuyi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Guangyue Gong
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yucheng Tian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Huidan Huang
- Center of Drug Screening & Evaluation, Wannan Medical College, Wuhu, Anhui, 241000, PR China.
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
3
|
Schiffer L, Oestlund I, Snoep JL, Gilligan LC, Taylor AE, Sinclair AJ, Singhal R, Freeman A, Ajjan R, Tiganescu A, Arlt W, Storbeck KH. Inhibition of the glucocorticoid-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 drives concurrent 11-oxygenated androgen excess. FASEB J 2024; 38:e23574. [PMID: 38551804 DOI: 10.1096/fj.202302131r] [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: 10/19/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 04/02/2024]
Abstract
Aldo-keto reductase 1C3 (AKR1C3) is a key enzyme in the activation of both classic and 11-oxygenated androgens. In adipose tissue, AKR1C3 is co-expressed with 11β-hydroxysteroid dehydrogenase type 1 (HSD11B1), which catalyzes not only the local activation of glucocorticoids but also the inactivation of 11-oxygenated androgens, and thus has the potential to counteract AKR1C3. Using a combination of in vitro assays and in silico modeling we show that HSD11B1 attenuates the biosynthesis of the potent 11-oxygenated androgen, 11-ketotestosterone (11KT), by AKR1C3. Employing ex vivo incubations of human female adipose tissue samples we show that inhibition of HSD11B1 results in the increased peripheral biosynthesis of 11KT. Moreover, circulating 11KT increased 2-3 fold in individuals with type 2 diabetes after receiving the selective oral HSD11B1 inhibitor AZD4017 for 35 days, thus confirming that HSD11B1 inhibition results in systemic increases in 11KT concentrations. Our findings show that HSD11B1 protects against excess 11KT production by adipose tissue, a finding of particular significance when considering the evidence for adverse metabolic effects of androgens in women. Therefore, when targeting glucocorticoid activation by HSD11B1 inhibitor treatment in women, the consequently increased generation of 11KT may offset beneficial effects of decreased glucocorticoid activation.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Imken Oestlund
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Jacky L Snoep
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
- Molecular Cell Biology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Rishi Singhal
- Upper GI Unit and Minimally Invasive Unit, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Adrian Freeman
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ramzi Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - Ana Tiganescu
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
- Medical Research Council Laboratory of Medical Sciences, London, UK
| | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
4
|
Hall L, Hart R. Role of corticosteroids in skin physiology and therapeutic potential of an 11β-HSD1 inhibitor: A review. Int J Dermatol 2024; 63:443-454. [PMID: 38146184 DOI: 10.1111/ijd.16967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 12/27/2023]
Abstract
Skin is a major site of cortisol bioconversion by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzymes which catalyze intracellular inactive cortisone into physiologically active cortisol. 11β-HSD1 is highly expressed in skin, especially in dermal fibroblasts, epidermal keratinocytes, melanocytes, and hair follicles, and plays important roles in regulating keratinocytes, fibroblast proliferation, and has roles in skin aging. Inhibition of 11β-HSD1 may reverse decreased collagen levels observed in extrinsically and intrinsically aged skin. Inhibitors of 11β-HSD1 may also have the potential to reverse decreased collagen observed in skin atrophy induced by glucocorticoid treatment. This systematic review aimed to summarize the current knowledge of roles for 11β-HSD1 inhibitor in skin physiology and potential for future use in medications. Studies have demonstrated that immediately following experimental insult in an animal model, there is increased expression of 11β-HSD1, and that topical application of an 11β-HSD1 inhibitor increases the rate of healing, increases skin collagen content, increases dermal fibroblasts, and increases dermal thickness. Furthermore, in patients with type 2 diabetes mellitus, 11β-HSD1 inhibitors reduce wound diameter after injury. Further development of 11β-HSD1 inhibitors appears to be a promising area for treating aging skin, aiding wound healing, and mitigating effects of systemic glucocorticoid use. Both topically and orally administered 11β-HSD1 inhibitors appear to be viable avenues for future research.
Collapse
Affiliation(s)
- Larissa Hall
- Faculty of Science and Agriculture, Business and Law, School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Robert Hart
- Faculty of Science and Agriculture, Business and Law, School of Science and Technology, University of New England, Armidale, NSW, Australia
| |
Collapse
|
5
|
Gómez C, Alimajstorovic Z, Othonos N, Winter DV, White S, Lavery GG, Tomlinson JW, Sinclair AJ, Odermatt A. Identification of a human blood biomarker of pharmacological 11β-hydroxysteroid dehydrogenase 1 inhibition. Br J Pharmacol 2024; 181:698-711. [PMID: 37740611 DOI: 10.1111/bph.16251] [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: 04/20/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND AND PURPOSE 11β-Hydroxysteroid dehydrogenase-1 (11β-HSD1) catalyses the oxoreduction of cortisone to cortisol, amplifying levels of active glucocorticoids. It is a pharmaceutical target in metabolic disease and cognitive impairments. 11β-HSD1 also converts some 7oxo-steroids to their 7β-hydroxy forms. A recent study in mice described the ratio of tauroursodeoxycholic acid (TUDCA)/tauro-7oxolithocholic acid (T7oxoLCA) as a biomarker for decreased 11β-HSD1 activity. The present study evaluates the equivalent bile acid ratio of glycoursodeoxycholic acid (GUDCA)/glyco-7oxolithocholic acid (G7oxoLCA) as a biomarker for pharmacological 11β-HSD1 inhibition in humans and compares it with the currently applied urinary (5α-tetrahydrocortisol + tetrahydrocortisol)/tetrahydrocortisone ((5αTHF + THF)/THE) ratio. EXPERIMENTAL APPROACH Bile acid profiles were analysed by ultra-HPLC tandem-MS in blood samples from two independent, double-blind placebo-controlled clinical studies of the orally administered selective 11β-HSD1 inhibitor AZD4017. The blood GUDCA/G7oxoLCA ratio was compared with the urinary tetrahydro-glucocorticoid ratio for ability to detect 11β-HSD1 inhibition. KEY RESULTS No significant alterations were observed in bile acid profiles following 11β-HSD1 inhibition by AZD4017, except for an increase of the secondary bile acid G7oxoLCA. The enzyme product/substrate ratio GUDCA/G7oxoLCA was found to be more reliable to detect 11β-HSD1 inhibition than the absolute G7oxoLCA concentration in both cohorts. Comparison of the blood GUDCA/G7oxoLCA ratio with the urinary (5αTHF + THF)/THE ratio revealed that both successfully detect 11β-HSD1 inhibition. CONCLUSIONS AND IMPLICATIONS 11β-HSD1 inhibition does not cause major alterations in bile acid homeostasis. The GUDCA/G7oxoLCA ratio represents the first blood biomarker of pharmacological 11β-HSD1 inhibition and may replace or complement the urinary (5αTHF + THF)/THE ratio biomarker.
Collapse
Affiliation(s)
- Cristina Gómez
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Zerin Alimajstorovic
- Metabolic Neurology, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Nantia Othonos
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Denise V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Sarah White
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Gareth G Lavery
- Department for Biosciences, Nottingham Trent University, Nottingham, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Alexandra J Sinclair
- Metabolic Neurology, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham, Birmingham, UK
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Oestlund I, Snoep J, Schiffer L, Wabitsch M, Arlt W, Storbeck KH. The glucocorticoid-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 catalyzes the activation of testosterone. J Steroid Biochem Mol Biol 2024; 236:106436. [PMID: 38035948 DOI: 10.1016/j.jsbmb.2023.106436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 12/02/2023]
Abstract
Testosterone biosynthesis from its precursor androstenedione is thought to be exclusively catalysed by the 17β-hydroxysteroid dehydrogenases-HSD17B3 in testes, and AKR1C3 in the ovary, adrenal and peripheral tissues. Here we show for the first time that the glucocorticoid activating enzyme 11β-hydroxysteroid dehydrogenase type 1 (HSD11B1) can also catalyse the 17β-reduction of androstenedione to testosterone, using a combination of in vitro enzyme kinetic assays, mathematical modelling, and molecular docking analysis. Furthermore, we show that co-expression of HSD11B1 and AKR1C3 increases testosterone production several-fold compared to the rate observed with AKR1C3 only, and that HSD11B1 is likely to contribute significantly to testosterone production in peripheral tissues.
Collapse
Affiliation(s)
- Imken Oestlund
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Jacky Snoep
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa; Molecular Cell Biology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, University Hospital of Ulm, Ulm, Germany
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK; Medical Research Council Laboratory of Medical Sciences, London, UK
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa; Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK.
| |
Collapse
|
7
|
Chatterjee R, Bhukta S, Angajala KK, Dandela R. Copper catalysed oxidative cascade deamination/cyclization of vinyl azide and benzylamine for the synthesis of 2,4,6-triarylpyridines. Org Biomol Chem 2023. [PMID: 37334911 DOI: 10.1039/d3ob00625e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
A highly efficient one-pot method for the synthesis of 2,4,6-triaryl pyridines has been developed via cascade deamination and annulation. Copper triflate and molecular iodine easily promoted the oxidative cyclization reaction of vinyl azide and benzylamine to access a wide variety of substituted pyridine substrates under an oxygen atmosphere. The presence of benzyl amine enables the cyclization process by providing the aryl functionality and the nitrogen source. Moreover, a broad range of substrates with good functional group tolerance, avoidance of external oxidants, excellent product yields, operational simplicity and mild conditions are the notable advantages of the present protocol.
Collapse
Affiliation(s)
- Rana Chatterjee
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| | - Swadhapriya Bhukta
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| | - Kishore Kumar Angajala
- Department of Humanities and Sciences, Vardhaman College of Engineering, Shamshabad, Hyderabad-501218, Telangana, India
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| |
Collapse
|
8
|
Donnelly LJ, Berthet J, Cantat T. Selective Reduction of Secondary Amides to Imines Catalysed by Schwartz's Reagent**. Angew Chem Int Ed Engl 2022; 61:e202206170. [DOI: 10.1002/anie.202206170] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Liam J. Donnelly
- Université Paris-Saclay CEA CNRS NIMBE 91191 Gif-sur-Yvette France
| | | | - Thibault Cantat
- Université Paris-Saclay CEA CNRS NIMBE 91191 Gif-sur-Yvette France
| |
Collapse
|
9
|
Abbas A, Schini M, Ainsworth G, Brown SR, Oughton J, Crowley RK, Cooper MS, Fairclough RJ, Eastell R, Stewart PM. Effect of AZD4017, a Selective 11β-HSD1 Inhibitor, on Bone Turnover Markers in Postmenopausal Osteopenia. J Clin Endocrinol Metab 2022; 107:2026-2035. [PMID: 35275196 PMCID: PMC9202729 DOI: 10.1210/clinem/dgac100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT The causative link between circulating glucocorticoid excess and osteoporosis is well-established. The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which increases local cortisol production, is expressed in human osteoblasts and its activity increases with age. OBJECTIVE We hypothesized that local 11β-HSD1 might mediate an age-related decrease in bone formation and that selective 11β-HSD1 inhibition may enhance bone formation. METHODS A dual-center, phase II, randomized, double-blind, placebo-controlled trial of 90 days' treatment with AZD4017 (a selective 11β-HSD1 inhibitor) was conducted in 55 postmenopausal women with osteopenia. Participants received 400 mg oral AZD4017 twice daily vs matched placebo over 90 days. The primary outcome measure was the impact on the bone formation marker osteocalcin. Secondary objectives included correlation with 11β-HSD1 activity. RESULTS At 90 days, osteocalcin levels did not differ between treatment groups: active (mean 22.3 [SD 8.6] ng/mL, n = 22) and placebo (21.7 [SD 9.2] ng/mL, n = 24), with a baseline-adjusted treatment effect of 0.95 (95% CI: -2.69, 4.60). The results from the urinary [THF + alloTHF]/THE ratio (index of 11β-HSD1 activity) and the urinary cortisol/cortisone ratio (index of 11β-HSD2 activity) confirmed a > 90% inhibition of 11β-HSD1 but no change in activity of 11β-HSD2. CONCLUSION This trial demonstrates that AZD4017 selectively inhibits 11β-HSD1 activity in vivo in a safe and reversible manner. Following 90 days of treatment, there is no effect on bone formation, indicating that the relative impairment of bone mineral density in postmenopausal women is not mediated by local intracellular production of cortisol under normal physiological concentrations.
Collapse
Affiliation(s)
- Afroze Abbas
- Faculty of Medicine and Health, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| | - Marian Schini
- Academic Unit of Bone Metabolism, University of Sheffield, Sheffield S5 7AU, UK
| | - Gemma Ainsworth
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9JT, UK
| | - Sarah R Brown
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9JT, UK
| | - Jamie Oughton
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9JT, UK
| | - Rachel K Crowley
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Mark S Cooper
- Concord Clinical School, Faculty of Medicine & Health, University of Sydney, NSW 2139, Australia
| | - Rebecca J Fairclough
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Richard Eastell
- Academic Unit of Bone Metabolism, University of Sheffield, Sheffield S5 7AU, UK
| | - Paul M Stewart
- Faculty of Medicine and Health, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| |
Collapse
|
10
|
Donnelly LJ, Berthet J, Cantat T. Selective Reduction of Secondary Amides to Imines Catalysed by Schwartz's Reagent**. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202206170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Liam J. Donnelly
- Université Paris-Saclay CEA CNRS NIMBE 91191 Gif-sur-Yvette France
| | | | - Thibault Cantat
- Université Paris-Saclay CEA CNRS NIMBE 91191 Gif-sur-Yvette France
| |
Collapse
|
11
|
A Combination of Nicotinamide and D-Ribose (RiaGev) Is Safe and Effective to Increase NAD + Metabolome in Healthy Middle-Aged Adults: A Randomized, Triple-Blind, Placebo-Controlled, Cross-Over Pilot Clinical Trial. Nutrients 2022; 14:nu14112219. [PMID: 35684021 PMCID: PMC9183138 DOI: 10.3390/nu14112219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 12/26/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor required for proper functioning of all cells and its decline is correlated with advancing age and disease. This randomized, triple-blind, placebo-controlled, crossover pilot study assessed the efficacy and safety of a combination of nicotinamide with D-ribose (RiaGev) for NAD metabolome enhancement and related benefits in healthy middle-aged adults. Supplementing with 1520 mg RiaGev twice daily for 7 days significantly increased the NAD+ metabolome in blood, especially NADP+ by 27% compared to the placebo group (p = 0.033) and over the baseline (p = 0.007). Increases in glutathione and high energy phosphates were also observed in the blood. Seven-day supplementation with RiaGev significantly (p = 0.013) reduced overall blood glucose without significant changes in insulin secretion (p = 0.796), suggesting an improved insulin sensitivity and glucose tolerance. The waking salivary cortisol of the subjects steadily and significantly decreased (p = 0.026) in the RiaGev group in contrast to the placebo. Subjects in the RiaGev group showed less fatigue, improved mental concentration and motivation over the baseline (p = 0.015, 0.018, and 0.012, respectively) as observed through the Checklist Individual Strength (CIS) questionnaire. There were no clinically relevant adverse events, or alterations in hematology, electrolytes, liver, and kidney markers pre- and post-supplementation. RiaGev appears to be safe and efficacious in increasing NAD+ metabolome in healthy middle-aged adults, as shown by this study.
Collapse
|
12
|
Equisetin is an anti-obesity candidate through targeting 11 β-HSD1. Acta Pharm Sin B 2022; 12:2358-2373. [PMID: 35646525 PMCID: PMC9136616 DOI: 10.1016/j.apsb.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is increasingly prevalent globally, searching for therapeutic agents acting on adipose tissue is of great importance. Equisetin (EQST), a meroterpenoid isolated from a marine sponge-derived fungus, has been reported to display antibacterial and antiviral activities. Here, we revealed that EQST displayed anti-obesity effects acting on adipose tissue through inhibiting adipogenesis in vitro and attenuating HFD-induced obesity in mice, doing so without affecting food intake, blood pressure or heart rate. We demonstrated that EQST inhibited the enzyme activity of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), a therapeutic target of obesity in adipose tissue. Anti-obesity properties of EQST were all offset by applying excessive 11β-HSD1's substrates and 11β-HSD1 inhibition through knockdown in vitro or 11β-HSD1 knockout in vivo. In the 11β-HSD1 bypass model constructed by adding excess 11β-HSD1 products, EQST's anti-obesity effects disappeared. Furthermore, EQST directly bond to 11β-HSD1 protein and presented remarkable better intensity on 11β-HSD1 inhibition and better efficacy on anti-obesity than known 11β-HSD1 inhibitor. Therefore, EQST can be developed into anti-obesity candidate compound, and this study may provide more clues for developing higher effective 11β-HSD1 inhibitors.
Collapse
|
13
|
Gastaldelli A, Stefan N, Häring HU. Liver-targeting drugs and their effect on blood glucose and hepatic lipids. Diabetologia 2021; 64:1461-1479. [PMID: 33877366 PMCID: PMC8187191 DOI: 10.1007/s00125-021-05442-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022]
Abstract
The global epidemic of non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) and the high prevalence among individuals with type 2 diabetes has attracted the attention of clinicians specialising in liver disorders. Many drugs are in the pipeline for the treatment of NAFLD/NASH, and several glucose-lowering drugs are now being tested specifically for the treatment of liver disease. Among these are nuclear hormone receptor agonists (e.g. peroxisome proliferator-activated receptor agonists, farnesoid X receptor agonists and liver X receptor agonists), fibroblast growth factor-19 and -21, single, dual or triple incretins, sodium-glucose cotransporter inhibitors, drugs that modulate lipid or other metabolic pathways (e.g. inhibitors of fatty acid synthase, diacylglycerol acyltransferase-1, acetyl-CoA carboxylase and 11β-hydroxysteroid dehydrogenase type-1) or drugs that target the mitochondrial pyruvate carrier. We have reviewed the metabolic effects of these drugs in relation to improvement of diabetic hyperglycaemia and fatty liver disease, as well as peripheral metabolism and insulin resistance.
Collapse
Affiliation(s)
- Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy.
| | - Norbert Stefan
- Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany.
- Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, Tübingen, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
14
|
Targeting Energy Expenditure-Drugs for Obesity Treatment. Pharmaceuticals (Basel) 2021; 14:ph14050435. [PMID: 34066399 PMCID: PMC8148206 DOI: 10.3390/ph14050435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/18/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity and overweight are associated with lethal diseases. In this context, obese and overweight individuals infected by COVID-19 are at greater risk of dying. Obesity is treated by three main pharmaceutical approaches, namely suppressing appetite, reducing energy intake by impairing absorption, and increasing energy expenditure. Most compounds used for the latter were first envisaged for other medical uses. However, several candidates are now being developed explicitly for targeting obesity by increasing energy expenditure. This review analyzes the compounds that show anti-obesity activity exerted through the energy expenditure pathway. They are classified on the basis of their development status: FDA-approved, Withdrawn, Clinical Trials, and Under Development. The chemical nature, target, mechanisms of action, and description of the current stage of development are described for each one.
Collapse
|
15
|
Grimm F, Rehman J, Stoldt S, Khan TA, Schlötel JG, Nizamov S, John M, Belov VN, Hell SW. Rhodamines with a Chloronicotinic Acid Fragment for Live Cell Superresolution STED Microscopy*. Chemistry 2021; 27:6070-6076. [PMID: 33496998 PMCID: PMC8048976 DOI: 10.1002/chem.202005134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 12/22/2022]
Abstract
Formylation of 2,6-dichloro-5-R-nicotinic acids at C-4 followed by condensation with 3-hydroxy-N,N-dimethylaniline gave analogs of the popular TAMRA fluorescent dye with a 2,6-dichloro-5-R-nicotinic acid residues (R=H, F). The following reaction with thioglycolic acid is selective, involves only one chlorine atom at the carbon between pyridine nitrogen and the carboxylic acid group and affords new rhodamine dyes absorbing at 564/ 573 nm and emitting at 584/ 597 nm (R=H/ F, in aq. PBS). Conjugates of the dyes with "small molecules" provided specific labeling (covalent and non-covalent) of organelles as well as of components of the cytoskeleton in living cells and were combined with fluorescent probes prepared from 610CP and SiR dyes and applied in two-color STED microscopy with a 775 nm STED laser.
Collapse
Affiliation(s)
- Florian Grimm
- Abberior GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Jasmin Rehman
- Abberior GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Stefan Stoldt
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| | - Taukeer A. Khan
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| | - Jan Gero Schlötel
- Abberior-Instruments GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Shamil Nizamov
- Abberior GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Michael John
- Institute of Organic and Biomolecular ChemistryGeorg-August UniversityTammannstr. 237077GöttingenGermany
| | - Vladimir N. Belov
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| | - Stefan W. Hell
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| |
Collapse
|
16
|
Hardy RS, Botfield H, Markey K, Mitchell JL, Alimajstorovic Z, Westgate CSJ, Sagmeister M, Fairclough RJ, Ottridge RS, Yiangou A, Storbeck KHH, Taylor AE, Gilligan LC, Arlt W, Stewart PM, Tomlinson JW, Mollan SP, Lavery GG, Sinclair AJ. 11βHSD1 Inhibition with AZD4017 Improves Lipid Profiles and Lean Muscle Mass in Idiopathic Intracranial Hypertension. J Clin Endocrinol Metab 2021; 106:174-187. [PMID: 33098644 PMCID: PMC7765633 DOI: 10.1210/clinem/dgaa766] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) determines prereceptor metabolism and activation of glucocorticoids within peripheral tissues. Its dysregulation has been implicated in a wide array of metabolic diseases, leading to the development of selective 11β-HSD1 inhibitors. We examined the impact of the reversible competitive 11β-HSD1 inhibitor, AZD4017, on the metabolic profile in an overweight female cohort with idiopathic intracranial hypertension (IIH). METHODS We conducted a UK multicenter phase II randomized, double-blind, placebo-controlled trial of 12-week treatment with AZD4017. Serum markers of glucose homeostasis, lipid metabolism, renal and hepatic function, inflammation and androgen profiles were determined and examined in relation to changes in fat and lean mass by dual-energy X-ray absorptiometry. RESULTS Patients receiving AZD4017 showed significant improvements in lipid profiles (decreased cholesterol, increased high-density lipoprotein [HDL] and cholesterol/HDL ratio), markers of hepatic function (decreased alkaline phosphatase and gamma-glutamyl transferase), and increased lean muscle mass (1.8%, P < .001). No changes in body mass index, fat mass, and markers of glucose metabolism or inflammation were observed. Patients receiving AZD4017 demonstrated increased levels of circulating androgens, positively correlated with changes in total lean muscle mass. CONCLUSIONS These beneficial metabolic changes represent a reduction in risk factors associated with raised intracranial pressure and represent further beneficial therapeutic outcomes of 11β-HSD1 inhibition by AZD4017 in this overweight IIH cohort. In particular, beneficial changes in lean muscle mass associated with AZD4017 may reflect new applications for this nature of inhibitor in the management of conditions such as sarcopenia.
Collapse
Affiliation(s)
- Rowan S Hardy
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Hannah Botfield
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Keira Markey
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - James L Mitchell
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Zerin Alimajstorovic
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Connar S J Westgate
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Michael Sagmeister
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rebecca J Fairclough
- Emerging Innovations Unit, Discovery Sciences. BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Ottridge
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andreas Yiangou
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Karl-Heinz H Storbeck
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology & Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Susan P Mollan
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
- Correspondence and Reprint Requests: Alexandra Sinclair, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK. E-mail:
| |
Collapse
|
17
|
Chuanxin Z, Shengzheng W, Lei D, Duoli X, Jin L, Fuzeng R, Aiping L, Ge Z. Progress in 11β-HSD1 inhibitors for the treatment of metabolic diseases: A comprehensive guide to their chemical structure diversity in drug development. Eur J Med Chem 2020; 191:112134. [PMID: 32088493 DOI: 10.1016/j.ejmech.2020.112134] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/24/2020] [Accepted: 02/06/2020] [Indexed: 12/19/2022]
Abstract
11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) is a key metabolic enzyme that catalyzing the intracellular conversion of inactive glucocorticoids to physiologically active ones. Work over the past decade has demonstrated the aberrant overexpression of 11β-HSD1 contributed to the pathophysiological process of metabolic diseases like obesity, type 2 diabetes mellitus, and metabolic syndromes. The inhibition of 11β-HSD1 represented an attractive therapeutic strategy for the treatment of metabolic diseases. Therefore, great efforts have been devoted to developing 11β-HSD1 inhibitors based on the diverse molecular scaffolds. This review focused on the structural features of the most important 11β-HSD1 inhibitors and categorized them into natural products derivatives and synthetic compounds. We also briefly discussed the optimization process, binding modes, structure-activity relationships (SAR) and biological evaluations of each inhibitor. Moreover, the challenges and directions for 11β-HSD1 inhibitors were discussed, which might provide some useful clues to guide the future discovery of novel 11β-HSD1 inhibitors.
Collapse
Affiliation(s)
- Zhong Chuanxin
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Wang Shengzheng
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Dang Lei
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xie Duoli
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Liu Jin
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute for Research and Continuing Education (IRACE), Hong Kong Baptist University, Shenzhen, China
| | - Ren Fuzeng
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| | - Lu Aiping
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Zhang Ge
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
18
|
Practical application of ligand efficiency metrics in lead optimisation. Bioorg Med Chem 2018; 26:3006-3015. [PMID: 29655612 DOI: 10.1016/j.bmc.2018.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 11/21/2022]
Abstract
The use of composite metrics that normalise biological potency values in relation to markers of physicochemical properties, such as size or lipophilicity, has gained a significant amount of traction with many medicinal chemists in recent years. However, there is no consensus on best practice in the area and their application has attracted some criticism. Here we present our approach to their application in lead optimisation projects, provide an objective discussion of the principles we consider important and illustrate how our use of lipophilic ligand efficiency enabled the progression of a number of our successful drug discovery projects. We derive, from this and some recent literature highlights, a set of heuristic guidelines for lipophilicity based optimisation that we believe are generally applicable across chemical series and protein targets.
Collapse
|
19
|
Daga PR, Bolger MB, Haworth IS, Clark RD, Martin EJ. Physiologically Based Pharmacokinetic Modeling in Lead Optimization. 1. Evaluation and Adaptation of GastroPlus To Predict Bioavailability of Medchem Series. Mol Pharm 2018; 15:821-830. [PMID: 29337578 DOI: 10.1021/acs.molpharmaceut.7b00972] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
When medicinal chemists need to improve bioavailability (%F) within a chemical series during lead optimization, they synthesize new series members with systematically modified properties mainly by following experience and general rules of thumb. More quantitative models that predict %F of proposed compounds from chemical structure alone have proven elusive. Global empirical %F quantitative structure-property (QSPR) models perform poorly, and projects have too little data to train local %F QSPR models. Mechanistic oral absorption and physiologically based pharmacokinetic (PBPK) models simulate the dissolution, absorption, systemic distribution, and clearance of a drug in preclinical species and humans. Attempts to build global PBPK models based purely on calculated inputs have not achieved the <2-fold average error needed to guide lead optimization. In this work, local GastroPlus PBPK models are instead customized for individual medchem series. The key innovation was building a local QSPR for a numerically fitted effective intrinsic clearance (CLloc). All inputs are subsequently computed from structure alone, so the models can be applied in advance of synthesis. Training CLloc on the first 15-18 rat %F measurements gave adequate predictions, with clear improvements up to about 30 measurements, and incremental improvements beyond that.
Collapse
Affiliation(s)
- Pankaj R Daga
- Novartis Institute of Biomedical Research , Emeryville , California 94608 , United States
| | - Michael B Bolger
- Simulations Plus, Inc. , 42505 10th Street West , Lancaster , California 93534 , United States
| | - Ian S Haworth
- Department of Pharmacology and Pharmaceutical Sciences , University of Southern California , Los Angeles , California 90089 , United States
| | - Robert D Clark
- Simulations Plus, Inc. , 42505 10th Street West , Lancaster , California 93534 , United States
| | - Eric J Martin
- Novartis Institute of Biomedical Research , Emeryville , California 94608 , United States
| |
Collapse
|
20
|
Synthesis of 2,3,6-trisubstituted pyridines by transition-metal free cyclization of 1,3-diynes with amino acids. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.06.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
21
|
Schwab D, Sturm C, Portron A, Fuerst-Recktenwald S, Hainzl D, Jordan P, Stewart WC, Tepedino ME, DuBiner H. Oral administration of the 11β-hydroxysteroid-dehydrogenase type 1 inhibitor RO5093151 to patients with glaucoma: an adaptive, randomised, placebo-controlled clinical study. BMJ Open Ophthalmol 2017; 1:e000063. [PMID: 29354707 PMCID: PMC5721631 DOI: 10.1136/bmjophth-2016-000063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/16/2017] [Accepted: 04/08/2017] [Indexed: 11/05/2022] Open
Abstract
Background/aims Cortisol is involved in the regulation of intraocular pressure (IOP). This study aimed to assess the effect of 11β-hydroxysteroid-dehydrogenase type 1 (11βHSD1) inhibition by oral administration of RO5093151 on IOP. Methods The exposure of key ocular compartments following oral administration was assessed in rabbits. An adaptive, randomised, placebo-controlled study gated by a Bayesian decision criterion was performed in 35 patients with primary open angle glaucoma (POAG) or ocular hypertension (OHT). Following a 7-day placebo-controlled run-in period, 200 mg twice daily RO5093151 or placebo (4:1) were administered for 7 days. The extent of 11βHSD1 inhibition was assessed by the ratio of urinary tetrahydrocortisol (5α and 5β)/tetrahydrocortisone (THF/THE). Time-matched IOP assessments were performed. Results A high distribution of RO5093151 into the rabbit eye was observed. In humans, a high and sustained inhibition of 11βHSD1 was shown by the decrease of THF/THE from 0.9 at baseline to 0.18 on day 7. There was no statistically significant difference in change of IOP from baseline. In the ‘worse eye’, the adjusted least square mean change from baseline was −2.7 mm Hg (95% CI −4.2 to –1.2) and −2.9(95% CI −5.9 to 0.1) in the RO5093151 and placebo group, respectively. Conclusions Despite high inhibition of 11βHSD1 and expected moderate to high tissue distribution in ocular tissues, a 7-day treatment with a high oral dose of RO5093151 did not result in a clinically meaningful effect on IOP in patients with POAG or OHT.
Collapse
Affiliation(s)
- Dietmar Schwab
- Roche Innovation Center Basel, Clinical Pharmacology, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Carolina Sturm
- Roche Innovation Center Basel, Clinical Pharmacology, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Agnès Portron
- Roche Innovation Center Basel, Clinical Pharmacology, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Sabine Fuerst-Recktenwald
- Roche Innivation Center Basel, Translational Medicine, Cardiovascular and Metabolism, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Dominik Hainzl
- Roche Innovation Center Basel, Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Paul Jordan
- Department of Biometrics, Product Development, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | | | - Michael E Tepedino
- Cornerstone Eye Care, Division of Health Care, High Point, North Carolina, USA
| | - Harvey DuBiner
- Eye Care Centers Management, Inc., Clayton Eye Center, Morrow, Georgia, USA
| |
Collapse
|
22
|
Ye XY, Chen SY, Wu S, Yoon DS, Wang H, Hong Z, O'Connor SP, Li J, Li JJ, Kennedy LJ, Walker SJ, Nayeem A, Sheriff S, Camac DM, Ramamurthy V, Morin PE, Zebo R, Taylor JR, Morgan NN, Ponticiello RP, Harrity T, Apedo A, Golla R, Seethala R, Wang M, Harper TW, Sleczka BG, He B, Kirby M, Leahy DK, Li J, Hanson RL, Guo Z, Li YX, DiMarco JD, Scaringe R, Maxwell B, Moulin F, Barrish JC, Gordon DA, Robl JA. Discovery of Clinical Candidate 2-((2S,6S)-2-Phenyl-6-hydroxyadamantan-2-yl)-1-(3'-hydroxyazetidin-1-yl)ethanone [BMS-816336], an Orally Active Novel Selective 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor. J Med Chem 2017; 60:4932-4948. [PMID: 28537398 DOI: 10.1021/acs.jmedchem.7b00211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BMS-816336 (6n-2), a hydroxy-substituted adamantyl acetamide, has been identified as a novel, potent inhibitor against human 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzyme (IC50 3.0 nM) with >10000-fold selectivity over human 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2). 6n-2 exhibits a robust acute pharmacodynamic effect in cynomolgus monkeys (ED50 0.12 mg/kg) and in DIO mice. It is orally bioavailable (%F ranges from 20 to 72% in preclinical species) and has a predicted pharmacokinetic profile of a high peak to trough ratio and short half-life in humans. This ADME profile met our selection criteria for once daily administration, targeting robust inhibition of 11β-HSD1 enzyme for the first 12 h period after dosing followed by an "inhibition holiday" so that the potential for hypothalamic-pituitary-adrenal (HPA) axis activation might be mitigated. 6n-2 was found to be well-tolerated in phase 1 clinical studies and represents a potential new treatment for type 2 diabetes, metabolic syndrome, and other human diseases modulated by glucocorticoid control.
Collapse
Affiliation(s)
- Xiang-Yang Ye
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Stephanie Y Chen
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Shung Wu
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David S Yoon
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Haixia Wang
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Zhenqiu Hong
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Stephen P O'Connor
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Jun Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - James J Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Lawrence J Kennedy
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Steven J Walker
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Akbar Nayeem
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Steven Sheriff
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Daniel M Camac
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Vidyhashankar Ramamurthy
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Paul E Morin
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Rachel Zebo
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Joseph R Taylor
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Nathan N Morgan
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Randolph P Ponticiello
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Thomas Harrity
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Atsu Apedo
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Rajasree Golla
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Ramakrishna Seethala
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Mengmeng Wang
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Timothy W Harper
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Bogdan G Sleczka
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Bin He
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Mark Kirby
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David K Leahy
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Jianqing Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Ronald L Hanson
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Zhiwei Guo
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Yi-Xin Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - John D DiMarco
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Raymond Scaringe
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Brad Maxwell
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Frederick Moulin
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Joel C Barrish
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David A Gordon
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Jeffrey A Robl
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
23
|
Jones S, Ahmet J, Ayton K, Ball M, Cockerill M, Fairweather E, Hamilton N, Harper P, Hitchin J, Jordan A, Levy C, Lopez R, McKenzie E, Packer M, Plant D, Simpson I, Simpson P, Sinclair I, Somervaille TCP, Small H, Spencer GJ, Thomson G, Tonge M, Waddell I, Walsh J, Waszkowycz B, Wigglesworth M, Wiseman DH, Ogilvie D. Discovery and Optimization of Allosteric Inhibitors of Mutant Isocitrate Dehydrogenase 1 (R132H IDH1) Displaying Activity in Human Acute Myeloid Leukemia Cells. J Med Chem 2016; 59:11120-11137. [PMID: 28002956 DOI: 10.1021/acs.jmedchem.6b01320] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A collaborative high throughput screen of 1.35 million compounds against mutant (R132H) isocitrate dehydrogenase IDH1 led to the identification of a novel series of inhibitors. Elucidation of the bound ligand crystal structure showed that the inhibitors exhibited a novel binding mode in a previously identified allosteric site of IDH1 (R132H). This information guided the optimization of the series yielding submicromolar enzyme inhibitors with promising cellular activity. Encouragingly, one compound from this series was found to induce myeloid differentiation in primary human IDH1 R132H AML cells in vitro.
Collapse
Affiliation(s)
| | | | | | - Matthew Ball
- Manchester Institute of Biotechnology, University of Manchester , Princess Street, Manchester, M1 7DN, U.K
| | | | | | | | | | | | | | - Colin Levy
- Manchester Institute of Biotechnology, University of Manchester , Princess Street, Manchester, M1 7DN, U.K
| | - Ruth Lopez
- Manchester Institute of Biotechnology, University of Manchester , Princess Street, Manchester, M1 7DN, U.K
| | - Eddie McKenzie
- Manchester Institute of Biotechnology, University of Manchester , Princess Street, Manchester, M1 7DN, U.K
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ryu JH, Lee JA, Kim S, Shin YA, Yang J, Han HY, Son HJ, Kim YH, Sa JH, Kim JS, Lee J, Lee J, Park HG. Discovery of 2-((R)-4-(2-Fluoro-4-(methylsulfonyl)phenyl)-2-methylpiperazin-1-yl)-N-((1R,2s,3S,5S,7S)-5-hydroxyadamantan-2-yl)pyrimidine-4-carboxamide (SKI2852): A Highly Potent, Selective, and Orally Bioavailable Inhibitor of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1). J Med Chem 2016; 59:10176-10189. [DOI: 10.1021/acs.jmedchem.6b01122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Je Ho Ryu
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Jung A Lee
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Shinae Kim
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Young Ah Shin
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Jewon Yang
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Hye Young Han
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Hyun Joo Son
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Yong Hyuk Kim
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Joon Ho Sa
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Jae-Sun Kim
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Jungeun Lee
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Jeeyeon Lee
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Hyeung-geun Park
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| |
Collapse
|
25
|
Abstract
The first mineralocorticoid receptor (MR) antagonist, spironolactone, was developed almost 60 years ago to treat primary aldosteronism and pathological edema. Its use waned in part because of its lack of selectivity. Subsequently, knowledge of the scope of MR function was expanded along with clinical evidence of the therapeutic importance of MR antagonists to prevent the ravages of inappropriate MR activation. Forty-two years elapsed between the first and MR-selective second generation of MR antagonists. Fifteen years later, despite serious shortcomings of the existing antagonists, a third-generation antagonist has yet to be marketed. Progress has been slowed by the lack of appreciation of the large variety of cell types that express the MR and its diverse cell-type-specific actions, and also its unique complex interaction actions at the molecular level. New MR antagonists should preferentially target the inflammatory and fibrotic effects of MR and perhaps its excitatory effects on sympathetic nervous system, but not the renal tubular epithelium or neurons of the cortex and hippocampus. This review briefly describes efforts to develop a third-generation MR antagonist and why fourth generation antagonists and selective agonists based on structural determinants of tissue and ligand-specific MR activation should be contemplated.
Collapse
|
26
|
Murumkar PR, Shinde AC, Sharma MK, Yamaguchi H, Miniyar PB, Yadav MR. Development of a credible 3D-QSAR CoMSIA model and docking studies for a series of triazoles and tetrazoles containing 11β-HSD1 inhibitors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2016; 27:265-292. [PMID: 27094303 DOI: 10.1080/1062936x.2016.1167774] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Type 2 diabetes mellitus is described by insulin resistance and high fasting blood glucose. Increased levels of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzyme result in insulin resistance and metabolic syndrome. Inhibition of 11β-HSD1 decreases glucose production and increases hepatic insulin sensitivity. Use of selective 11β-HSD1 inhibitors could prove to be an effective strategy for the treatment of the disease. It was decided to identify the essential structural features required by any compound to possess 11β-HSD1 inhibitory activity. A dataset of 139 triazoles and tetrazoles having 11β-HSD1 inhibitory activity was used for the development of a 3D-QSAR model. The best comparative molecular field analysis (CoMFA) model was generated with databased alignment, which was further used for comparative molecular similarity indices analysis (CoMSIA). The optimal CoMSIA model showed [Formula: see text] = 0.809 with five components, [Formula: see text] = 0.931, SEE = 0.323 and F-value = 249.126. The CoMSIA model offered better prediction than the CoMFA model with [Formula: see text] = 0.522 and 0.439, respectively, indicating that the CoMSIA model appeared to be a better one for the prediction of activity for the newly designed 11β-HSD1 inhibitors. The selectivity aspect of 11β-HSD1 over 11β-HSD2 was studied with the help of docking studies.
Collapse
Affiliation(s)
- P R Murumkar
- a Faculty of Pharmacy , The Maharaja Sayajirao University of Baroda , Vadodara , India
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - A C Shinde
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - M K Sharma
- a Faculty of Pharmacy , The Maharaja Sayajirao University of Baroda , Vadodara , India
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - H Yamaguchi
- c Department of Pharmacy , Meijo University , Nagoya , Japan
| | - P B Miniyar
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - M R Yadav
- a Faculty of Pharmacy , The Maharaja Sayajirao University of Baroda , Vadodara , India
| |
Collapse
|
27
|
Ryu JH, Kim S, Lee JA, Han HY, Son HJ, Lee HJ, Kim YH, Kim JS, Park HG. Synthesis and optimization of picolinamide derivatives as a novel class of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors. Bioorg Med Chem Lett 2015; 25:1679-1683. [DOI: 10.1016/j.bmcl.2015.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/23/2015] [Accepted: 03/03/2015] [Indexed: 11/15/2022]
|
28
|
Chen XQ, Shao LD, Pal M, Shen Y, Cheng X, Xu G, Peng LY, Wang K, Pan ZH, Li MM, Leng Y, He J, Zhao QS. Hupehenols A-E, selective 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors from Viburnum hupehense. JOURNAL OF NATURAL PRODUCTS 2015; 78:330-334. [PMID: 25590374 DOI: 10.1021/np500896n] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Five selective 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) competitive inhibitors, hupehenols A-E (1-5), were isolated from Viburnum hupehense. The structure elucidation indicated that compounds 1-5 are new 20,21,22,23,24,25,26,27-octanordammarane triterpenoids. Their structures were established on the basis of NMR spectroscopic and mass spectrometric analysis. Hupehenols A-E (1-5) showed inhibition against human 11β-HSD1, with hupehenols B (2) and E (5) having IC50 values of 15.3 and 34.0 nM, respectively. Moreover, hupehenols C (3) and D (4) are highly selective inhibitors of human 11β-HSD1 when compared to murine 11β-HSD1.
Collapse
Affiliation(s)
- Xuan-Qin Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650204, Yunnan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Byun SY, Shin YJ, Nam KY, Hong SP, Ahn SK. A novel highly potent and selective 11β-hydroxysteroid dehydrogenase type 1 inhibitor, UI-1499. Life Sci 2015; 120:1-7. [DOI: 10.1016/j.lfs.2014.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/15/2014] [Accepted: 11/01/2014] [Indexed: 01/27/2023]
|
30
|
Robb GR, Boyd S, Davies CD, Dossetter AG, Goldberg FW, Kemmitt PD, Scott JS, Swales JG. Design of pyrazolo-pyrimidines as 11β-HSD1 inhibitors through optimisation of molecular electrostatic potential. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00043b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Rapid and efficient lead optimisation through quantification of the molecular electrostatic potential using quantum mechanics.
Collapse
|
31
|
Ryu JH, Kim S, Han HY, Son HJ, Lee HJ, Shin YA, Kim JS, Park HG. Synthesis and biological evaluation of picolinamides as potent inhibitors of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Bioorg Med Chem Lett 2014; 25:695-700. [PMID: 25529735 DOI: 10.1016/j.bmcl.2014.11.074] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/25/2014] [Accepted: 11/27/2014] [Indexed: 10/24/2022]
Abstract
Synthesis of a series of 6-substituted picolinamide derivatives and their inhibitory activities against 11β-hydroxysteroid dehydrogenase type 1 are described. Optimization of the initial hit compound, N-cyclohexyl-6-(piperidin-1-yl)picolinamide (1) from high throughput screening of in-house library resulted in the discovery of the highly potent and metabolically stable compound 25, which was efficacious in a mouse ex vivo pharmacodynamic model and reduced the fasting blood glucose and insulin levels in a HF/STZ mouse model after oral dosing.
Collapse
Affiliation(s)
- Je Ho Ryu
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea; Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Shinae Kim
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea
| | - Hye Young Han
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea
| | - Hyun Joo Son
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea
| | - Hyun Jung Lee
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea
| | - Young Ah Shin
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea
| | - Jae-Sun Kim
- Life Science R&D Center, SK Chemicals, 686 Sampyeong-dong, Bundang-gu, Seongnam-si 463-400, Republic of Korea
| | - Hyeung-Geun Park
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
32
|
Gomez-Sanchez EP. Brain mineralocorticoid receptors in cognition and cardiovascular homeostasis. Steroids 2014; 91:20-31. [PMID: 25173821 PMCID: PMC4302001 DOI: 10.1016/j.steroids.2014.08.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/10/2014] [Accepted: 08/04/2014] [Indexed: 12/20/2022]
Abstract
Mineralocorticoid receptors (MR) mediate diverse functions supporting osmotic and hemodynamic homeostasis, response to injury and inflammation, and neuronal changes required for learning and memory. Inappropriate MR activation in kidneys, heart, vessels, and brain hemodynamic control centers results in cardiovascular and renal pathology and hypertension. MR binds aldosterone, cortisol and corticosterone with similar affinity, while the glucocorticoid receptor (GR) has less affinity for cortisol and corticosterone. As glucocorticoids are more abundant than aldosterone, aldosterone activates MR in cells co-expressing enzymes with 11β-hydroxydehydrogenase activity to inactivate them. MR and GR co-expressed in the same cell interact at the molecular and functional level and these functions may be complementary or opposing depending on the cell type. Thus the balance between MR and GR expression and activation is crucial for normal function. Where 11β-hydroxydehydrogenase 2 (11β-HSD2) that inactivates cortisol and corticosterone in aldosterone target cells of the kidney and nucleus tractus solitarius (NTS) is not expressed, as in most neurons, MR are activated at basal glucocorticoid concentrations, GR at stress concentrations. An exception may be pre-autonomic neurons of the PVN which express MR and 11β-HSD1 in the absence of hexose-6-phosphate dehydrogenase required to generate the requisite cofactor for reductase activity, thus it acts as a dehydrogenase. MR antagonists, valuable adjuncts to the treatment of cardiovascular disease, also inhibit MR in the brain that are crucial for memory formation and exacerbate detrimental effects of excessive GR activation on cognition and mood. 11β-HSD1 inhibitors combat metabolic and cognitive diseases related to glucocorticoid excess, but may exacerbate MR action where 11β-HSD1 acts as a dehydrogenase, while non-selective 11β-HSD1&2 inhibitors cause injurious disruption of MR hemodynamic control. MR functions in the brain are multifaceted and optimal MR:GR activity is crucial. Therefore selectively targeting down-stream effectors of MR specific actions may be a better therapeutic goal.
Collapse
Affiliation(s)
- Elise P Gomez-Sanchez
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
33
|
Colin O, Greck C, Prim D, Thomassigny C. Toward Pyridine-Heterocycle Patterns through Prins and Aza-Prins Cyclisations: Application to a Short Synthesis of (±)-Anabasine. European J Org Chem 2014. [DOI: 10.1002/ejoc.201402971] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
34
|
Goldberg FW, Dossetter AG, Scott JS, Robb GR, Boyd S, Groombridge SD, Kemmitt PD, Sjögren T, Gutierrez PM, deSchoolmeester J, Swales JG, Turnbull AV, Wild MJ. Optimization of Brain Penetrant 11β-Hydroxysteroid Dehydrogenase Type I Inhibitors and in Vivo Testing in Diet-Induced Obese Mice. J Med Chem 2014; 57:970-86. [DOI: 10.1021/jm4016729] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | - James S. Scott
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Graeme R. Robb
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Scott Boyd
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Sam D. Groombridge
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Paul D. Kemmitt
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Tove Sjögren
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | | | | | - John G. Swales
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Andrew V. Turnbull
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Martin J. Wild
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| |
Collapse
|
35
|
McCoull W, Augustin M, Blake C, Ertan A, Kilgour E, Krapp S, Moore JE, Newcombe NJ, Packer MJ, Rees A, Revill J, Scott JS, Selmi N, Gerhardt S, Ogg DJ, Steinbacher S, Whittamore PRO. Identification and optimisation of 3,3-dimethyl-azetidin-2-ones as potent and selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). MEDCHEMCOMM 2014. [DOI: 10.1039/c3md00234a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
Scott JS, Goldberg FW, Turnbull AV. Medicinal Chemistry of Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1). J Med Chem 2013; 57:4466-86. [DOI: 10.1021/jm4014746] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- James S. Scott
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| | - Frederick W. Goldberg
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| | - Andrew V. Turnbull
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| |
Collapse
|
37
|
Ling T, Griffith E, Mitachi K, Rivas F. Scalable and Divergent Total Synthesis of (+)-Colletoic Acid, a Selective 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor. Org Lett 2013; 15:5790-3. [DOI: 10.1021/ol402842u] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Taotao Ling
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105-3678, United States
| | - Elizabeth Griffith
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105-3678, United States
| | - Katsuhiko Mitachi
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105-3678, United States
| | - Fatima Rivas
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105-3678, United States
| |
Collapse
|
38
|
Just-Baringo X, Albericio F, Álvarez M. From 2,6-Dichloronicotinic Acid to Thiopeptide Cores. European J Org Chem 2013. [DOI: 10.1002/ejoc.201300877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Böhme T, Engel CK, Farjot G, Güssregen S, Haack T, Tschank G, Ritter K. 1,1-Dioxo-5,6-dihydro-[4,1,2]oxathiazines, a novel class of 11ß-HSD1 inhibitors for the treatment of diabetes. Bioorg Med Chem Lett 2013; 23:4685-91. [PMID: 23845218 DOI: 10.1016/j.bmcl.2013.05.102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 05/29/2013] [Accepted: 05/31/2013] [Indexed: 11/19/2022]
Abstract
Racemic cis-1,1-dioxo-5,6-dihydro-[4,1,2]oxathiazine derivative 4a was isolated as an impurity in a sample of a hit from a HTS campaign on 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). After separation by chiral chromatography the 4a-S, 8a-R enantiomer of compound 4a was identified as the true, potent enzyme inhibitor. The cocrystal structure of 4a with human and murine 11ß-HSD1 revealed the unique binding mode of the oxathiazine series. SAR elucidation and optimization in regard to metabolic stability led to monocyclic tetramethyloxathiazines as exemplified by compound 21g.
Collapse
Affiliation(s)
- Thomas Böhme
- Sanofi Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Nair SK, Matthews JJ, Cripps SJ, Cheng H, Hoffman JE, Smith C, Kupchinsky S, Siu M, Taylor WD, Wang Y, Johnson TO, Dress KR, Edwards MP, Zhou S, Hosea NA, LaPaglia A, Kang P, Castro A, Ermolieff J, Fanjul A, Vogel JE, Rejto P, Dalvie D. N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: Strategies to eliminate reactive metabolites. Bioorg Med Chem Lett 2013; 23:2344-8. [DOI: 10.1016/j.bmcl.2013.02.066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 02/07/2013] [Accepted: 02/13/2013] [Indexed: 01/21/2023]
|
41
|
Discovery of SAR184841, a potent and long-lasting inhibitor of 11β-hydroxysteroid dehydrogenase type 1, active in a physiopathological animal model of T2D. Bioorg Med Chem Lett 2013; 23:2414-21. [DOI: 10.1016/j.bmcl.2013.02.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/31/2013] [Accepted: 02/03/2013] [Indexed: 01/16/2023]
|
42
|
Xia G, You X, Liu L, Liu H, Wang J, Shi Y, Li P, Xiong B, Liu X, Shen J. Design, synthesis and SAR of piperidyl-oxadiazoles as 11β-hydroxysteroid dehydrogenase 1 inhibitors. Eur J Med Chem 2013; 62:1-10. [DOI: 10.1016/j.ejmech.2012.12.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 12/17/2012] [Accepted: 12/23/2012] [Indexed: 01/19/2023]
|
43
|
Udagawa S, Sakami S, Takemura T, Sato M, Arai T, Nitta A, Aoki T, Kawai K, Iwamura T, Okazaki S, Takahashi T, Kaino M. Discovery of novel 7-membered cyclic amide derivatives that inhibit 11beta-hydroxysteroid dehydrogenase type 1. Bioorg Med Chem Lett 2013; 23:1617-21. [DOI: 10.1016/j.bmcl.2013.01.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 01/18/2013] [Accepted: 01/22/2013] [Indexed: 11/13/2022]
|
44
|
Wan ZK, Chenail E, Li HQ, Ipek M, Xiang J, Suri V, Hahm S, Bard J, Svenson K, Xu X, Tian X, Wang M, Li X, Johnson CE, Qadri A, Panza D, Perreault M, Mansour TS, Tobin JF, Saiah E. Discovery of HSD-621 as a Potential Agent for the Treatment of Type 2 Diabetes. ACS Med Chem Lett 2013; 4:118-23. [PMID: 24900572 DOI: 10.1021/ml300352x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 11/23/2012] [Indexed: 12/20/2022] Open
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) catalyzes the conversion of inactive glucocorticoid cortisone to its active form, cortisol. The glucocorticoid receptor (GR) signaling pathway has been linked to the pathophysiology of diabetes and metabolic syndrome. Herein, the structure-activity relationship of a series of piperazine sulfonamide-based 11β-HSD1 inhibitors is described. (R)-3,3,3-Trifluoro-2-(5-(((R)-4-(4-fluoro-2-(trifluoromethyl)phenyl)-2-methylpiperazin-1-yl)sulfonyl)thiophen-2-yl)-2-hydroxypropanamide 18a (HSD-621) was identified as a potent and selective 11β-HSD1 inhibitor and was ultimately selected as a clinical development candidate. HSD-621 has an attractive overall pharmaceutical profile and demonstrates good oral bioavailability in mouse, rat, and dog. When orally dosed in C57/BL6 diet-induced obesity (DIO) mice, HSD-621 was efficacious and showed a significant reduction in both fed and fasting glucose and insulin levels. Furthermore, HSD-621 was well tolerated in drug safety assessment studies.
Collapse
Affiliation(s)
- Zhao-Kui Wan
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Eva Chenail
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Huan-Qiu Li
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Manus Ipek
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Jason Xiang
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Vipin Suri
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Seung Hahm
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Joel Bard
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Kristine Svenson
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Xin Xu
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Xianbin Tian
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Mengmeng Wang
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Xiangping Li
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Christian E. Johnson
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Ariful Qadri
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Darrell Panza
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Mylene Perreault
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Tarek S. Mansour
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - James F. Tobin
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Eddine Saiah
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| |
Collapse
|
45
|
Scott JS, Birch AM, Brocklehurst KJ, Brown HS, Goldberg K, Groombridge SD, Hudson JA, Leach AG, MacFaul PA, McKerrecher D, Poultney R, Schofield P, Svensson PH. Optimisation of aqueous solubility in a series of G protein coupled receptor 119 (GPR119) agonists. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20130e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Solubility improvements in a series of GPR119 agonists are achieved through reduction of lipophilicity together with hydrogen bond acceptor modulation.
Collapse
|
46
|
Goldberg FW, Leach AG, Scott JS, Snelson WL, Groombridge SD, Donald CS, Bennett SNL, Bodin C, Gutierrez PM, Gyte AC. Free-Wilson and Structural Approaches to Co-optimizing Human and Rodent Isoform Potency for 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1) Inhibitors. J Med Chem 2012; 55:10652-61. [DOI: 10.1021/jm3013163] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Andrew G. Leach
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| | - James S. Scott
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| | - Wendy L. Snelson
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| | - Sam D. Groombridge
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| | - Craig S. Donald
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| | | | - Cristian Bodin
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| | | | - Amy C. Gyte
- AstraZeneca, Mereside, Alderley
Park, Macclesfield, SK10 4TG, United Kingdom
| |
Collapse
|
47
|
Scott JS, deSchoolmeester J, Kilgour E, Mayers RM, Packer MJ, Hargreaves D, Gerhardt S, Ogg DJ, Rees A, Selmi N, Stocker A, Swales JG, Whittamore PRO. Novel Acidic 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1) Inhibitor with Reduced Acyl Glucuronide Liability: The Discovery of 4-[4-(2-Adamantylcarbamoyl)-5-tert-butyl-pyrazol-1-yl]benzoic Acid (AZD8329). J Med Chem 2012; 55:10136-47. [DOI: 10.1021/jm301252n] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- James S. Scott
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Joanne deSchoolmeester
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Elaine Kilgour
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Rachel M. Mayers
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Martin J. Packer
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - David Hargreaves
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Stefan Gerhardt
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Derek J. Ogg
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Amanda Rees
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Nidhal Selmi
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Andrew Stocker
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - John G. Swales
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| | - Paul R. O. Whittamore
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom
| |
Collapse
|
48
|
Scott JS, Gill AL, Godfrey L, Groombridge SD, Rees A, Revill J, Schofield P, Sörme P, Stocker A, Swales JG, Whittamore PRO. Optimisation of pharmacokinetic properties in a neutral series of 11β-HSD1 inhibitors. Bioorg Med Chem Lett 2012; 22:6756-61. [PMID: 23013933 DOI: 10.1016/j.bmcl.2012.08.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 08/14/2012] [Accepted: 08/16/2012] [Indexed: 11/25/2022]
Abstract
11β-HSD1 is increasingly seen as an attractive target for the treatment of type II diabetes and other elements of the metabolic syndrome. In this program of work we describe how a series of neutral 2-thioalkyl-pyridine 11β-HSD1 inhibitors were optimized in terms of their pharmacokinetic properties to give compounds with excellent bioavailability in both rat and dog through a core change to pyrimidine. A potential reactive metabolite issue with 4-thioalkyl-pyrimidines was circumvented by a switch from sulfur to carbon substitution.
Collapse
Affiliation(s)
- James S Scott
- Cardiovascular & Gastrointestinal Innovative Medicines Unit, AstraZeneca Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Scott JS, Barton P, Bennett SNL, deSchoolmeester J, Godfrey L, Kilgour E, Mayers RM, Packer MJ, Rees A, Schofield P, Selmi N, Swales JG, Whittamore PRO. Reduction of acyl glucuronidation in a series of acidic 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors: the discovery of AZD6925. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20154b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|