1
|
Biendl S, Häberli C, Chen G, Wang W, Zhong L, Saunders J, Pham T, Wang X, Wu J, Charman SA, Vennerstrom JL, Keiser J. In Vitro and In Vivo Antischistosomal Activity Profiling and Pharmacokinetics of Ozonide Carboxylic Acids. ACS Infect Dis 2023; 9:643-652. [PMID: 36794836 PMCID: PMC10858445 DOI: 10.1021/acsinfecdis.2c00581] [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] [Academic Contribution Register] [Indexed: 02/17/2023]
Abstract
Praziquantel, the only drug in clinical use for the treatment and control of schistosomiasis, is inactive against developing infections. Ozonides are synthetic peroxide derivatives inspired by the naturally occurring artemisinin and show particularly promising activity against juvenile schistosomes. We conducted an in-depth characterization of the in vitro and in vivo antischistosomal activity and pharmacokinetics of lead ozonide carboxylic acid OZ418 and four of its active analogs. In vitro, the ozonides featured rapid and consistent activity against schistosomula and adult schistosomes at double-digit micromolar EC50 values. Potency did not vary considerably between Schistosoma spp. The zwitterionic OZ740 and OZ772 were more active in vivo compared to their non-amphoteric carboxylic acids OZ418 and OZ748, despite their much lower systemic plasma exposure (AUC). The most active compound in vivo was ethyl ester OZ780, which was rapidly transformed to its parent zwitterion OZ740 and achieved ED50 values of 35 ± 2.4 and 29 ± 2.4 mg/kg against adult and juvenile Schistosoma mansoni, respectively. Ozonide carboxylic acids represent promising candidates for further optimization and development due to their good efficacy against both life stages together with their broad activity range against all relevant parasite species.
Collapse
Affiliation(s)
- Stefan Biendl
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Cécile Häberli
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Wen Wang
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Longjin Zhong
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jessica Saunders
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Thao Pham
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Xiaofang Wang
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States of America
| | - Jianbo Wu
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States of America
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jonathan L Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States of America
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| |
Collapse
|
2
|
Han S, Li H, Chen W, Yang L, Tong X, Zuo J, Hu Y. Discovery of potent ebola entry inhibitors with (3S,4aS,8aS)-2-(3-amino-2-hydroxypropyl) decahydroisoquinoline-3-carboxamide scaffold. Eur J Med Chem 2022; 240:114608. [PMID: 35872393 DOI: 10.1016/j.ejmech.2022.114608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/07/2022] [Revised: 07/03/2022] [Accepted: 07/10/2022] [Indexed: 11/15/2022]
Abstract
Ebola virus (EBOV), one member of the family Filoviridae, can causes hemorrhagic fever and other severe diseases in humans with a high mortality rate (25-90%). Until recently, there were no approved drugs and very limited treatment method for Ebola virus disease. In this study, we discovered a series of potent Ebola entry inhibitors with the (3S,4aS,8aS)-2-(3-amino-2-hydroxypropyl)decahydroisoquinoline-3-carboxamide scaffold from high-throughput screening in reported pseudotyped virus system. Further optimization resulted a most potent compound 28 (IC50= 0.05 μM, SI = 98), which displayed 3-fold potency compared to the known inhibitor Toremifene (IC50= 0.17 μM, SI = 55). Moreover, compound 28 exhibited the remarkable selectivity between EBOV-GP and VSV-G (Spec. Index = 58), thus could exclude nonspecific effects. Structure-activity relationship and molecular docking analysis of the new chemical scaffold provided more information on the binding modes and the spare volume at the binding cavity, thus can guide the design of the further potent compounds.
Collapse
Affiliation(s)
- Sheng Han
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Heng Li
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weixiong Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Yang
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiankun Tong
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jianping Zuo
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Youhong Hu
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, 1 Xiangshanzhi Road, Hangzhou, 310024, China.
| |
Collapse
|
3
|
Riccardi D, Ward JPT, Yarova PL, Janssen LJ, Lee TH, Ying S, Corrigan CJ. Topical therapy with negative allosteric modulators of the calcium-sensing receptor (calcilytics) for the management of asthma: the beginning of a new era? Eur Respir J 2022; 60:13993003.02103-2021. [DOI: 10.1183/13993003.02103-2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/19/2021] [Accepted: 12/16/2021] [Indexed: 11/05/2022]
Abstract
In this review article we present the evidence to date supporting the role of the calcium-sensing receptor (CaSR) as a key, pluripotential molecular trigger for asthma and speculate on the likely benefits of topical therapy of asthma with negative allosteric modulators of the CaSR: calcilytics.
Collapse
|
4
|
Diao J, DeBono A, Josephs TM, Bourke JE, Capuano B, Gregory KJ, Leach K. Therapeutic Opportunities of Targeting Allosteric Binding Sites on the Calcium-Sensing Receptor. ACS Pharmacol Transl Sci 2021; 4:666-679. [PMID: 33860192 DOI: 10.1021/acsptsci.1c00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/04/2021] [Indexed: 01/24/2023]
Abstract
The CaSR is a class C G protein-coupled receptor (GPCR) that acts as a multimodal chemosensor to maintain diverse homeostatic functions. The CaSR is a clinical therapeutic target in hyperparathyroidism and has emerged as a putative target in several other diseases. These include hyper- and hypocalcaemia caused either by mutations in the CASR gene or in genes that regulate CaSR signaling and expression, and more recently in asthma. The development of CaSR-targeting drugs is complicated by the fact that the CaSR possesses many different binding sites for endogenous and exogenous agonists and allosteric modulators. Binding sites for endogenous and exogenous ligands are located throughout the large CaSR protein and are interconnected in ways that we do not yet fully understand. This review summarizes our current understanding of CaSR physiology, signaling, and structure and how the many different binding sites of the CaSR may be targeted to treat disease.
Collapse
Affiliation(s)
- Jiayin Diao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Aaron DeBono
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| | - Ben Capuano
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| |
Collapse
|
5
|
Tuffour A, Kosiba AA, Zhang Y, Peprah FA, Gu J, Shi H. Role of the calcium-sensing receptor (CaSR) in cancer metastasis to bone: Identifying a potential therapeutic target. Biochim Biophys Acta Rev Cancer 2021; 1875:188528. [PMID: 33640382 DOI: 10.1016/j.bbcan.2021.188528] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/18/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/20/2022]
Abstract
Cancer is a major cause of morbidity and mortality worldwide due to its ability to evade immune surveillance and metastasize from its origin to a secondary point of contact. Though several treatment techniques have been developed to suppress or manage cancer spread, a strategy for total control over the disease continues to evade researchers. In considering ways to control or prevent cancer from metastasizing to the bone, we analyze the impact of the calcium-sensing receptor (CaSR), whose primary role is to maintain calcium (Ca2+) homeostasis in cellular and systemic physiological processes. CaSR is a pleiotropic receptor capable of enhancing the proliferation of some cancers such as breast, lung, prostate and kidney cancers at its primary site(s) and stimulating bone metastasis, while exerting a suppressive effect in others such as colon cancer. The activity of CaSR not only increases cancer cell proliferation, migration and suppression of apoptosis in the organs indicated, but also increases the secretion of parathyroid hormone-related protein (PTHrP) and epiregulin, which induce osteolytic activity and osteoblastic suppression. In addition, released cytokines and Ca2+ from bone resorption are critical factors that further promote cancer proliferation. In this review, we seek to highlight previous viewpoints on CaSR, discuss its role in a new context, and consider its potential clinical application in cancer treatment.
Collapse
Affiliation(s)
- Alex Tuffour
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | | | - Yao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Frank Addai Peprah
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
6
|
Zhou S, Huang G, Chen G. Synthesis and biological activities of drugs for the treatment of osteoporosis. Eur J Med Chem 2020; 197:112313. [PMID: 32335412 DOI: 10.1016/j.ejmech.2020.112313] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/26/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022]
Abstract
Osteoporosis is an asymptomatic progressive disease. With the improvement of people's living standard and the aging of population, osteoporosis and its fracture have become one of the main diseases threatening the aging society. The serious medical and social burden caused by this has aroused wide public concern. Osteoporosis is listed as one of the three major diseases of the elderly. At present, the drugs for osteoporosis include bone resorption inhibitors and bone formation promoters. The purpose of these anti-osteoporosis drugs is to balance osteoblast bone formation and osteoclast bone resorption. With the development of anti-osteoporosis drugs, new anti osteoporosis drugs have been designed and synthesized. There are many kinds of new compounds with anti osteoporosis activity, but most of them are concentrated on the original drugs with anti osteoporosis activity, or the natural products with anti-osteoporosis activity are extracted from the natural products for structural modification to obtain the corresponding derivatives or analogues. These target compounds showed good ALP activity in vitro and in vivo, promoted osteoblast differentiation and mineralization, or had anti TRAP activity, inhibited osteoclast absorption. This work attempts to systematically review the studies on the synthesis and bioactivity of anti-osteoporosis drugs in the past 10 years. The structure-activity relationship was discussed, which provided a reasonable idea for the design and development of new anti-osteoporosis drugs.
Collapse
Affiliation(s)
- Shiyang Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Gangliang Huang
- Active Carbohydrate Research Institute, Chongqing Key Laboratory of Green Synthesis and Application, College of Chemistry, Chongqing Normal University, Chongqing, 401331, China.
| | - Guangying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China.
| |
Collapse
|
7
|
Nemeth EF, Van Wagenen BC, Balandrin MF. Discovery and Development of Calcimimetic and Calcilytic Compounds. PROGRESS IN MEDICINAL CHEMISTRY 2018; 57:1-86. [PMID: 29680147 DOI: 10.1016/bs.pmch.2017.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/08/2023]
Abstract
The extracellular calcium receptor (CaR) is a G protein-coupled receptor (GPCR) and the pivotal molecule regulating systemic Ca2+ homeostasis. The CaR was a challenging target for drug discovery because its physiological ligand is an inorganic ion (Ca2+) rather than a molecule so there was no structural template to guide medicinal chemistry. Nonetheless, small molecules targeting this receptor were discovered. Calcimimetics are agonists or positive allosteric modulators of the CaR, while calcilytics are antagonists and all to date are negative allosteric modulators. The calcimimetic cinacalcet was the first allosteric modulator of a GPCR to achieve regulatory approval and is a first-in-class treatment for secondary hyperparathyroidism in patients on dialysis, and for hypercalcemia in some forms of primary hyperparathyroidism. It is also useful in treating some rare genetic diseases that cause hypercalcemia. Two other calcimimetics are now on the market (etelcalcetide) or under regulatory review (evocalcet). Calcilytics stimulate the secretion of parathyroid hormone and were initially developed as treatments for osteoporosis. Three different calcilytics of two different chemotypes failed in clinical trials due to lack of efficacy. Calcilytics are now being repurposed and might be useful in treating hypoparathyroidism and several rare genetic diseases causing hypocalcemia. The challenges ahead for medicinal chemists are to design compounds that select conformations of the CaR that preferentially target a particular signalling pathway and/or that affect the CaR in a tissue-selective manner.
Collapse
|
8
|
Frees S, Breuksch I, Haber T, Bauer HK, Chavez-Munoz C, Raven P, Moskalev I, D Costa N, Tan Z, Daugaard M, Thüroff JW, Haferkamp A, Prawitt D, So A, Brenner W. Calcium-sensing receptor (CaSR) promotes development of bone metastasis in renal cell carcinoma. Oncotarget 2018; 9:15766-15779. [PMID: 29644008 PMCID: PMC5884663 DOI: 10.18632/oncotarget.24607] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/21/2017] [Accepted: 02/25/2018] [Indexed: 12/26/2022] Open
Abstract
Bone metastasis is an important prognostic factor in renal cell carcinoma (RCC). The calcium-sensing receptor (CaSR) has been associated with bone metastasis in several different malignancies. We analyzed the impact of CaSR in bone metastasis in RCC in vitro and in vivo. The RCC cell line 786-O was stably transfected with the CaSR gene and treated with calcium alone or in combination with the CaSR antagonist NPS2143. Afterwards migration, adhesion, proliferation and prominent signaling molecules were analyzed. Calcium treated CaSR-transfected 768-O cells showed an increased adhesion to endothelial cells and the extracellular matrix components fibronectin and collagen I, but not to collagen IV. The chemotactic cell migration and proliferation was also induced by calcium. The activity of SHC, AKT, ERK, P90RSK and JNK were enhanced after calcium treatment of CaSR-transfected cells. These effects were abolished by NPS2143. Development of bone metastasis was evaluated in vivo in a mouse model. Intracardiac injection of CaSR-transfected 768-O cells showed an increased rate of bone metastasis. The results indicate CaSR as an important component in the mechanism of bone metastasis in RCC. Therefore, targeting CaSR might be beneficial in patients with bone metastatic RCC with a high CaSR expression.
Collapse
Affiliation(s)
- Sebastian Frees
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada.,Department of Urology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ines Breuksch
- Department of Gynecology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Tobias Haber
- Department of Urology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Heide-Katharina Bauer
- Department of Gynecology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Claudia Chavez-Munoz
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Peter Raven
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Igor Moskalev
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Ninadh D Costa
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Zheng Tan
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Mads Daugaard
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Joachim W Thüroff
- Department of Urology, Johannes Gutenberg University Medical Center, Mainz, Germany.,Current address: Department of Urology, University Clinic Mannheim, Mannheim, Germany
| | - Axel Haferkamp
- Department of Urology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Dirk Prawitt
- Department of Pediatrics, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Alan So
- Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, British Columbia, Canada
| | - Walburgis Brenner
- Department of Urology, Johannes Gutenberg University Medical Center, Mainz, Germany.,Department of Gynecology, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
9
|
Seo B, Kim H, Kim YG, Baek Y, Um K, Lee PH. Synthesis of Bicyclic Isothiazoles through an Intramolecular Rhodium-Catalyzed Transannulation of Cyanothiadiazoles. J Org Chem 2017; 82:10574-10582. [PMID: 28889744 DOI: 10.1021/acs.joc.7b02077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
An intramolecular rhodium-catalyzed transannulation of readily available cyanothiadiazoles containing an ester, amide, or ether as a linker is described. It provides a wide range of bicyclic isothiazoles in good to excellent yields together with the release of molecular nitrogen. These results indicate that the carbon atom in the α-thiavinyl carbene is nucleophilic and that the sulfur atom is electrophilic.
Collapse
Affiliation(s)
- Boram Seo
- Department of Chemistry, Kangwon National University , Chuncheon 24341, Republic of Korea
| | - Hyunseok Kim
- Department of Chemistry, Kangwon National University , Chuncheon 24341, Republic of Korea
| | - Ya Gob Kim
- Department of Chemistry, Kangwon National University , Chuncheon 24341, Republic of Korea
| | - Yonghyeon Baek
- Department of Chemistry, Kangwon National University , Chuncheon 24341, Republic of Korea
| | - Kyusik Um
- Department of Chemistry, Kangwon National University , Chuncheon 24341, Republic of Korea
| | - Phil Ho Lee
- Department of Chemistry, Kangwon National University , Chuncheon 24341, Republic of Korea
| |
Collapse
|
10
|
Lin TY, Wu HH, Feng JJ, Zhang J. Transfer of Chirality in the Rhodium-Catalyzed Chemoselective and Regioselective Allylic Alkylation of Hydroxyarenes with Vinyl Aziridines. Org Lett 2017; 19:2897-2900. [DOI: 10.1021/acs.orglett.7b01136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/02/2023]
Affiliation(s)
- Tao-Yan Lin
- Shanghai Key Laboratory of
Green Chemistry and Chemical Processes, School of Chemistry and Molecular
Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Hai-Hong Wu
- Shanghai Key Laboratory of
Green Chemistry and Chemical Processes, School of Chemistry and Molecular
Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Jian-Jun Feng
- Shanghai Key Laboratory of
Green Chemistry and Chemical Processes, School of Chemistry and Molecular
Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Junliang Zhang
- Shanghai Key Laboratory of
Green Chemistry and Chemical Processes, School of Chemistry and Molecular
Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| |
Collapse
|
11
|
Shen C, Guo X, Yu J, Zeng XG, Peng L, Zhao CM, Zhang FL. Synthesis of 2-(phenoxymethyl)oxirane derivatives through unexpected rearrangement of oxiran-2-ylmethyl benzenesulfonates. SYNTHETIC COMMUN 2017. [DOI: 10.1080/00397911.2016.1258579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/20/2022]
Affiliation(s)
- Chuang Shen
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Xiang Guo
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Jun Yu
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Xian-Guo Zeng
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Li Peng
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Chuan-meng Zhao
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Fu-Li Zhang
- China State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, China
| |
Collapse
|
12
|
Liang GB, Zhou C, Huo X, Wang H, Yang X, Huang S, Wang H, Wilkinson H, Luo L, Tang W, Sutton D, Li H, Zaller D, Meinke PT. Discovery of novel dihydrobenzofuran cyclopropane carboxylic acid based calcium sensing receptor antagonists for the treatment of osteoporosis. Bioorg Med Chem Lett 2016; 26:4077-80. [DOI: 10.1016/j.bmcl.2016.06.073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/07/2016] [Revised: 06/06/2016] [Accepted: 06/25/2016] [Indexed: 01/12/2023]
|
13
|
Schepelmann M, Yarova PL, Lopez-Fernandez I, Davies TS, Brennan SC, Edwards PJ, Aggarwal A, Graça J, Rietdorf K, Matchkov V, Fenton RA, Chang W, Krssak M, Stewart A, Broadley KJ, Ward DT, Price SA, Edwards DH, Kemp PJ, Riccardi D. The vascular Ca2+-sensing receptor regulates blood vessel tone and blood pressure. Am J Physiol Cell Physiol 2015; 310:C193-204. [PMID: 26538090 DOI: 10.1152/ajpcell.00248.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/31/2015] [Accepted: 10/29/2015] [Indexed: 12/28/2022]
Abstract
The extracellular calcium-sensing receptor CaSR is expressed in blood vessels where its role is not completely understood. In this study, we tested the hypothesis that the CaSR expressed in vascular smooth muscle cells (VSMC) is directly involved in regulation of blood pressure and blood vessel tone. Mice with targeted CaSR gene ablation from vascular smooth muscle cells (VSMC) were generated by breeding exon 7 LoxP-CaSR mice with animals in which Cre recombinase is driven by a SM22α promoter (SM22α-Cre). Wire myography performed on Cre-negative [wild-type (WT)] and Cre-positive (SM22α)CaSR(Δflox/Δflox) [knockout (KO)] mice showed an endothelium-independent reduction in aorta and mesenteric artery contractility of KO compared with WT mice in response to KCl and to phenylephrine. Increasing extracellular calcium ion (Ca(2+)) concentrations (1-5 mM) evoked contraction in WT but only relaxation in KO aortas. Accordingly, diastolic and mean arterial blood pressures of KO animals were significantly reduced compared with WT, as measured by both tail cuff and radiotelemetry. This hypotension was mostly pronounced during the animals' active phase and was not rescued by either nitric oxide-synthase inhibition with nitro-l-arginine methyl ester or by a high-salt-supplemented diet. KO animals also exhibited cardiac remodeling, bradycardia, and reduced spontaneous activity in isolated hearts and cardiomyocyte-like cells. Our findings demonstrate a role for CaSR in the cardiovascular system and suggest that physiologically relevant changes in extracellular Ca(2+) concentrations could contribute to setting blood vessel tone levels and heart rate by directly acting on the cardiovascular CaSR.
Collapse
Affiliation(s)
- M Schepelmann
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - P L Yarova
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - I Lopez-Fernandez
- School of Biosciences, Cardiff University, Cardiff, United Kingdom; Faculty of Pharmacy, Université de Picardie Jules Verne, Amiens, France
| | - T S Davies
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - S C Brennan
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - P J Edwards
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - A Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - J Graça
- School of Biosciences, Cardiff University, Cardiff, United Kingdom; Pathology Sciences, AstraZeneca, Macclesfield, Cheshire, United Kingdom
| | - K Rietdorf
- Faculty of Science, Department for Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - V Matchkov
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - R A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - W Chang
- Endocrine Research Unit, Department of Veteran Affairs Medical Center, Department of Medicine, University of California, San Francisco, Califonia
| | - M Krssak
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - A Stewart
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - K J Broadley
- School of Pharmacy and Pharmaceutical Sciences, Division of Pharmacology, Cardiff University, Cardiff, United Kingdom
| | - D T Ward
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom; and
| | - S A Price
- Pathology Sciences, AstraZeneca, Macclesfield, Cheshire, United Kingdom
| | - D H Edwards
- Cardiff University, Wales Heart Research Institute, Cardiff, United Kingdom
| | - P J Kemp
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - D Riccardi
- School of Biosciences, Cardiff University, Cardiff, United Kingdom;
| |
Collapse
|
14
|
Nikolov NG, Dybdahl M, Jónsdóttir SÓ, Wedebye EB. hERG blocking potential of acids and zwitterions characterized by three thresholds for acidity, size and reactivity. Bioorg Med Chem 2014; 22:6004-13. [DOI: 10.1016/j.bmc.2014.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/08/2014] [Revised: 08/26/2014] [Accepted: 09/05/2014] [Indexed: 02/01/2023]
|
15
|
Zhou ZL, Liu HL, Wu JW, Tsao CW, Chen WH, Liu KT, Ho Y. Combining Structure-Based Pharmacophore andIn SilicoApproaches to Discover Novel Selective Serotonin Reuptake Inhibitors. Chem Biol Drug Des 2013; 82:705-17. [DOI: 10.1111/cbdd.12192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2013] [Revised: 06/20/2013] [Accepted: 07/09/2013] [Indexed: 01/04/2023]
Affiliation(s)
- Zheng-Li Zhou
- Institute of Biochemical and Biomedical Engineering; National Taipei University of Technology; 1 Sec. 3 ZhongXiao E. Road Taipei 10608 Taiwan
| | - Hsuan-Liang Liu
- Institute of Biochemical and Biomedical Engineering; National Taipei University of Technology; 1 Sec. 3 ZhongXiao E. Road Taipei 10608 Taiwan
- Department of Chemical Engineering and Biotechnology; National Taipei University of Technology; 1 Sec. 3 ZhongXiao E. Road Taipei 10608 Taiwan
| | - Josephine W. Wu
- Department of Optometry; Central Taiwan University of Science and Technology; 666 Buzih Road Taichung 40601 Taiwan
| | - Cheng-Wen Tsao
- Department of Applied Cosmetology; Taoyuan Innovation Institute of Technology; 414 Sec. 3, Jhongshan E. Road Jhongli City Taoyuan County 32091 Taiwan
| | - Wei-Hsi Chen
- Chemistry Division; Institute of Nuclear Energy Research; 1000 Wunhua Road Longtan Township Taoyuan County 32546 Taiwan
| | - Kung-Tien Liu
- Everlight Chemical Industrial Corporation; 6th Fl, 77, Tun Hua South Road, Sec.2 Taipei 106 Taiwan
| | - Yih Ho
- School of Pharmacy; Taipei Medical University; 250 Wu-Hsing Street Taipei 110 Taiwan
| |
Collapse
|
16
|
Maag H. Overcoming poor permeability – the role of prodrugs for oral drug delivery. DRUG DISCOVERY TODAY: TECHNOLOGIES 2012; 9:e71-e174. [PMID: 0 DOI: 10.1016/j.ddtec.2012.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/26/2023]
|
17
|
Melancon BJ, Hopkins CR, Wood MR, Emmitte KA, Niswender CM, Christopoulos A, Conn PJ, Lindsley CW. Allosteric modulation of seven transmembrane spanning receptors: theory, practice, and opportunities for central nervous system drug discovery. J Med Chem 2012; 55:1445-64. [PMID: 22148748 DOI: 10.1021/jm201139r] [Citation(s) in RCA: 186] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/27/2022]
Affiliation(s)
- Bruce J Melancon
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
The calcium-sensing receptor (CaR) is the key controller of extracellular calcium (Ca(2+)(o)) homeostasis via its regulation of parathyroid hormone (PTH) secretion and renal Ca(2+) reabsorption. The CaR-selective calcimimetic drug Cinacalcet stimulates the CaR to suppress PTH secretion in chronic kidney disease and represents the world's first clinically available receptor positive allosteric modulator (PAM). Negative CaR allosteric modulators (NAMs), known as calcilytics, can increase PTH secretion and are being investigated as possible bone anabolic treatments against age-related osteoporosis. Here we address the current state of development and clinical use of a series of positive and negative CaR modulators. In addition, clinical CaR mutations and transgenic mice carrying tissue-specific CaR deletions have provided a novel understanding of the relative functional importance of CaR in both calciotropic tissues and those elsewhere in the body. The development of CaR-selective modulators and signalling reagents have provided us with a more detailed appreciation of how the CaR signals in vivo. Thus, both of these areas of CaR research will be reviewed.
Collapse
Affiliation(s)
- Donald T Ward
- Faculty of Life Sciences, The University of ManchesterManchester, UK
| | | |
Collapse
|
19
|
Calcilytics: antagonists of the calcium-sensing receptor for the treatment of osteoporosis. Future Med Chem 2011; 3:535-47. [PMID: 21526895 DOI: 10.4155/fmc.11.17] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022] Open
Abstract
The only bone anabolic agents currently available on the market are based on the parathyroid hormone (PTH). Secretion of endogenous PTH is controlled by a calcium-sensing receptor at the surface of the parathyroid glands. Antagonists of this receptor (calcilytics) induce the release of the hormone. Provided the effect of the calcilytic is of short duration, a bone anabolic effect should also result. Although the first calcilytic series became known approximately 10 years ago, the number of different structural types is still small today. This article outlines the quest from hits to potent development candidates of all relevant calcilytic series currently known. Even after the front-runners unexpectedly failed in the clinic, the approach for an oral alternative to parenteral PTH remains highly attractive.
Collapse
|
20
|
Abraham AK, Maurer TS, Kalgutkar AS, Gao X, Li M, Healy DR, Petersen DN, Griffith DA, Mager DE. Pharmacodynamic model of parathyroid hormone modulation by a negative allosteric modulator of the calcium-sensing receptor. AAPS JOURNAL 2011; 13:265-73. [PMID: 21437757 DOI: 10.1208/s12248-011-9266-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/03/2010] [Accepted: 02/22/2011] [Indexed: 11/30/2022]
Abstract
In this study, a pharmacodynamic model is developed, based on calcium-parathyroid hormone (PTH) homeostasis, which describes the concentration-effect relationship of a negative allosteric modulator of the calcium-sensing receptor (CaR) in rats. Plasma concentrations of drug and PTH were determined from plasma samples obtained via serial jugular vein sampling following single subcutaneous doses of 1, 5, 45, and 150 mg/kg to male Sprague-Dawley rats (n = 5/dose). Drug pharmacokinetics was described by a one-compartment model with first-order absorption and linear elimination. Concentration-time profiles of PTH were characterized using a model in which the compound allosterically modulates Ca(+2) binding to the CaR that, in turn, modulates PTH through a precursor-pool indirect response model. Additionally, negative feedback was incorporated to account for tolerance observed at higher dose levels. Model fitting and parameter estimation were conducted using the maximum likelihood algorithm. The proposed model well characterized the data and provided compound specific estimates of the K(i) and cooperativity constant (α) of 1.47 ng/mL and 0.406, respectively. In addition, the estimated model parameters for PTH turnover were comparable to that previously reported. The final generalized model is capable of characterizing both PTH-Ca(+2) homeostasis and the pharmacokinetics and pharmacodynamics associated with the negative allosteric CaR modulator. As such, the model provides a simple platform for analysis of drugs targeting the PTH-Ca(+2) system.
Collapse
Affiliation(s)
- Anson K Abraham
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Amherst, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Shinagawa Y, Inoue T, Katsushima T, Kiguchi T, Ikenogami T, Ogawa N, Fukuda K, Hirata K, Harada K, Takagi M, Nakagawa T, Kimura S, Matsuo Y, Maekawa M, Hayashi M, Soejima Y, Takahashi M, Shindo M, Hashimoto H. Discovery of a potent and short-acting oral calcilytic with a pulsatile secretion of parathyroid hormone. ACS Med Chem Lett 2011; 2:238-42. [PMID: 24900301 PMCID: PMC4018071 DOI: 10.1021/ml100268k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2010] [Accepted: 12/21/2010] [Indexed: 12/24/2022] Open
Abstract
Short-acting oral calcilytics, calcium-sensing receptor (CaSR) antagonists, have been considered as alternatives for parathyroid hormone (PTH), an injectable bone anabolic drug used in the treatment of osteoporosis. Previously, we identified aminopropandiol 1, which transiently stimulated endogenous PTH secretion in rats. However, the inhibition of cytochrome P450 (CYP) 2D6 and the low bioavailability of 1 remain to be solved. Attempts to change the physicochemical properties of the highly lipophilic amine 1 by introduction of a carboxylic acid group as well as further structural modifications led to the discovery of the highly potent biphenylcarboxylic acid 15, with a markedly reduced CYP2D6 inhibition and a significantly improved bioavailability. Compound 15 evoked a rapid and transient elevation of endogenous PTH levels in rats after oral administration in a dose-dependent manner at a dose as low as 1 mg/kg. The PTH secretion pattern correlated with the pharmacokinetic profile and agreed well with that of the exogenous PTH injection which exerts a bone anabolic effect.
Collapse
Affiliation(s)
| | | | | | - Toshihiro Kiguchi
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Taku Ikenogami
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Naoki Ogawa
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Kenji Fukuda
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Kazuyuki Hirata
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Kazuhito Harada
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Masaki Takagi
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Takashi Nakagawa
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Shuichi Kimura
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yushi Matsuo
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Mariko Maekawa
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Mikio Hayashi
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yuki Soejima
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Mitsuru Takahashi
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Masanori Shindo
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Hiromasa Hashimoto
- Central Pharmaceutical Research Institute, Japan Tobacco
Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| |
Collapse
|
22
|
Urwyler S. Allosteric modulation of family C G-protein-coupled receptors: from molecular insights to therapeutic perspectives. Pharmacol Rev 2011; 63:59-126. [PMID: 21228259 DOI: 10.1124/pr.109.002501] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/27/2023] Open
Abstract
Allosteric receptor modulation is an attractive concept in drug targeting because it offers important potential advantages over conventional orthosteric agonism or antagonism. Allosteric ligands modulate receptor function by binding to a site distinct from the recognition site for the endogenous agonist. They often have no effect on their own and therefore act only in conjunction with physiological receptor activation. This article reviews the current status of allosteric modulation at family C G-protein coupled receptors in the light of their specific structural features on the one hand and current concepts in receptor theory on the other hand. Family C G-protein-coupled receptors are characterized by a large extracellular domain containing the orthosteric agonist binding site known as the "venus flytrap module" because of its bilobal structure and the dynamics of its activation mechanism. Mutational analysis and chimeric constructs have revealed that allosteric modulators of the calcium-sensing, metabotropic glutamate and GABA(B) receptors bind to the seven transmembrane domain, through which they modify signal transduction after receptor activation. This is in contrast to taste-enhancing molecules, which bind to different parts of sweet and umami receptors. The complexity of interactions between orthosteric and allosteric ligands is revealed by a number of adequate biochemical and electrophysiological assay systems. Many allosteric family C GPCR modulators show in vivo efficacy in behavioral models for a variety of clinical indications. The positive allosteric calcium sensing receptor modulator cinacalcet is the first drug of this type to enter the market and therefore provides proof of principle in humans.
Collapse
Affiliation(s)
- Stephan Urwyler
- Department of Chemistry and Biochemistry, University of Berne, P/A Weissensteinweg 3, CH-3303 Jegenstorf, Berne, Switzerland.
| |
Collapse
|
23
|
Southers JA, Bauman JN, Price DA, Humphries PS, Balan G, Sagal JF, Maurer TS, Zhang Y, Oliver R, Herr M, Healy DR, Li M, Kapinos B, Fate GD, Riccardi KA, Paralkar VM, Brown TA, Kalgutkar AS. Metabolism-guided design of short-acting calcium-sensing receptor antagonists. ACS Med Chem Lett 2010; 1:219-23. [PMID: 24900198 DOI: 10.1021/ml100058w] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2010] [Accepted: 05/10/2010] [Indexed: 12/22/2022] Open
Abstract
As part of a strategy to deliver short-acting calcium-sensing receptor (CaSR) antagonists, the metabolically labile thiomethyl functionality was incorporated into the zwitterionic amino alcohol derivative 3 with the hope of increasing human clearance through oxidative metabolism, while delivering a pharmacologically inactive sulfoxide metabolite. The effort led to the identification of thioanisoles 22 and 23 as potent and orally active CaSR antagonists with a rapid onset of action and short pharmacokinetic half-lives, which led to a rapid and transient stimulation of parathyroid hormone in a dose-dependent fashion following oral administration to rats. On the basis of the balance between target pharmacology, safety, and human disposition profiles, 22 and 23 were advanced as clinical candidates for the treatment of osteoporosis.
Collapse
Affiliation(s)
| | | | - David A. Price
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | | | - Gayatri Balan
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | - John F. Sagal
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | | | - Yan Zhang
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Robert Oliver
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Michael Herr
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | - David R. Healy
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Mei Li
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Brendon Kapinos
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | | | | | | | - Thomas A. Brown
- Pfizer Global Research and Development, Groton, Connecticut 06340
| | | |
Collapse
|
24
|
Kalgutkar AS, Griffith DA, Ryder T, Sun H, Miao Z, Bauman JN, Didiuk MT, Frederick KS, Zhao SX, Prakash C, Soglia JR, Bagley SW, Bechle BM, Kelley RM, Dirico K, Zawistoski M, Li J, Oliver R, Guzman-Perez A, Liu KKC, Walker DP, Benbow JW, Morris J. Discovery Tactics To Mitigate Toxicity Risks Due to Reactive Metabolite Formation with 2-(2-Hydroxyaryl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one Derivatives, Potent Calcium-Sensing Receptor Antagonists and Clinical Candidate(s) for the Treatment of Osteoporosis. Chem Res Toxicol 2010; 23:1115-26. [DOI: 10.1021/tx100137n] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
Affiliation(s)
- Amit S. Kalgutkar
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - David A. Griffith
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Tim Ryder
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Hao Sun
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Zhuang Miao
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Jonathan N. Bauman
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Mary T. Didiuk
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Kosea S. Frederick
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Sabrina X. Zhao
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Chandra Prakash
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - John R. Soglia
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Scott W. Bagley
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Bruce M. Bechle
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Ryan M. Kelley
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Kenneth Dirico
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Michael Zawistoski
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Jianke Li
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Robert Oliver
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Angel Guzman-Perez
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Kevin K. C. Liu
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Daniel P. Walker
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - John W. Benbow
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Joel Morris
- Pharmacokinetics, Dynamics and Metabolism Department and Department of Medicinal Chemistry, Pfizer Global Research and Development, Groton, Connecticut 06340
| |
Collapse
|