1
|
Santiso A, Heinemann A, Kargl J. Prostaglandin E2 in the Tumor Microenvironment, a Convoluted Affair Mediated by EP Receptors 2 and 4. Pharmacol Rev 2024; 76:388-413. [PMID: 38697857 DOI: 10.1124/pharmrev.123.000901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 05/05/2024] Open
Abstract
The involvement of the prostaglandin E2 (PGE2) system in cancer progression has long been recognized. PGE2 functions as an autocrine and paracrine signaling molecule with pleiotropic effects in the human body. High levels of intratumoral PGE2 and overexpression of the key metabolic enzymes of PGE2 have been observed and suggested to contribute to tumor progression. This has been claimed for different types of solid tumors, including, but not limited to, lung, breast, and colon cancer. PGE2 has direct effects on tumor cells and angiogenesis that are known to promote tumor development. However, one of the main mechanisms behind PGE2 driving cancerogenesis is currently thought to be anchored in suppressed antitumor immunity, thus providing possible therapeutic targets to be used in cancer immunotherapies. EP2 and EP4, two receptors for PGE2, are emerging as being the most relevant for this purpose. This review aims to summarize the known roles of PGE2 in the immune system and its functions within the tumor microenvironment. SIGNIFICANCE STATEMENT: Prostaglandin E2 (PGE2) has long been known to be a signaling molecule in cancer. Its presence in tumors has been repeatedly associated with disease progression. Elucidation of its effects on immunological components of the tumor microenvironment has highlighted the potential of PGE2 receptor antagonists in cancer treatment, particularly in combination with immune checkpoint inhibitor therapeutics. Adjuvant treatment could increase the response rates and the efficacy of immune-based therapies.
Collapse
Affiliation(s)
- Ana Santiso
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
2
|
Corminboeuf O, Diethelm S, Zumbrunn C, Lyothier I, Niggli N, Gnerre C, Jeay S, Lehembre F, Boss C. Design of Dual EP2/EP4 Antagonists through Scaffold Merging of Selective Inhibitors. ChemMedChem 2024; 19:e202300606. [PMID: 37983645 DOI: 10.1002/cmdc.202300606] [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: 11/01/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 11/22/2023]
Abstract
Prostaglandin E2 (PGE2) plays a key role in various stages of cancer. PGE2 signals through the EP2 and the EP4 receptors, promoting tumorigenesis, metastasis, and/or immune suppression. Dual inhibition of both the EP2 and the EP4 receptors has the potential to counteract the effect of PGE2 and to result in antitumor efficacy. We herein disclose for the first time the structure of dual EP2/EP4 antagonists. By merging the scaffolds of EP2 selective and EP4 selective inhibitors, we generated a new chemical series of compounds blocking both receptors with comparable potency. In vitro and in vivo profiling suggests that the newly identified compounds are promising lead structures for further development into dual EP2/EP4 antagonists for use in cancer therapy.
Collapse
Affiliation(s)
- Olivier Corminboeuf
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Stefan Diethelm
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Cornelia Zumbrunn
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Isabelle Lyothier
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Nadja Niggli
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Carmela Gnerre
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Sébastien Jeay
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - François Lehembre
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Christoph Boss
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| |
Collapse
|
3
|
Iwasa S, Koyama T, Nishino M, Kondo S, Sudo K, Yonemori K, Yoshida T, Tamura K, Shimizu T, Fujiwara Y, Kitano S, Shimomura A, Sato J, Yokoyama F, Iida H, Kondo M, Yamamoto N. First-in-human study of ONO-4578, an antagonist of prostaglandin E 2 receptor 4, alone and with nivolumab in solid tumors. Cancer Sci 2022; 114:211-220. [PMID: 36082616 PMCID: PMC9807514 DOI: 10.1111/cas.15574] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 01/07/2023] Open
Abstract
EP4, a prostaglandin E2 receptor, has shown an immunosuppressive activity on cancer cells. This first-in-human study evaluated ONO-4578, a highly selective EP4 antagonist, as monotherapy and in combination with nivolumab in patients with advanced or metastatic solid tumors. A daily dose ranging from 30 mg to 100 mg of ONO-4578 monotherapy and that ranging from 2 mg to 60 mg of ONO-4578 with biweekly nivolumab 240 mg were administered. A total of 31 patients were enrolled, 10 receiving monotherapy and 21 receiving combination therapy. Overall, 26 patients experienced treatment-related adverse events. Dose-limiting toxicities were observed in three patients; one of six patients receiving 100 mg monotherapy developed grade 3 duodenal ulcer and two of six patients receiving 60 mg combination therapy developed either grade 3 erythema multiforme or grade 3 increased amylase and grade 4 increased lipase. One patient with small-cell lung cancer who received 40 mg combination therapy had a partial response, and three patients with monotherapy and six patients with combination therapy had stable disease. Pharmacodynamics analyses showed that ONO-4578 had EP4 antagonistic activity at doses as low as 2 mg. In conclusion, the maximum tolerated dose of ONO-4578 alone or in combination with nivolumab was not reached. ONO-4578 was well tolerated at the tested doses and showed signs of antitumor activity. Considering safety, efficacy, and pharmacokinetics/pharmacodynamics results, ONO-4578 40 mg daily with nivolumab 240 mg biweekly was selected as the recommended dose for future clinical trials. (Registration: JapicCTI-173,496 and NCT03155061).
Collapse
Affiliation(s)
- Satoru Iwasa
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Takafumi Koyama
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Makoto Nishino
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Shunsuke Kondo
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Kazuki Sudo
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Kan Yonemori
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan,Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Tatsuya Yoshida
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan,Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Kenji Tamura
- Department of Breast and Medical OncologyNational Cancer Center HospitalTokyoJapan,Present address:
Department of Medical OncologyShimane University HospitalShimaneJapan
| | - Toshio Shimizu
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan,Present address:
Department of Medical Oncology/Cancer CenterWakayama Medical University HospitalWakayamaJapan
| | - Yutaka Fujiwara
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan,Present address:
Department of Thoracic OncologyAichi Cancer Center HospitalAichiJapan
| | - Shigehisa Kitano
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan,Present address:
Advanced Medical Development CenterCancer Institute Hospital of Japanese Foundation for Cancer ResearchTokyoJapan
| | - Akihiko Shimomura
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan,Present address:
Department of Breast and Medical OncologyCenter Hospital of the National Center for Global Health and MedicineTokyoJapan
| | - Jun Sato
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Fumiharu Yokoyama
- Translational Research LaboratoriesOno Pharmaceutical Co., Ltd.OsakaJapan
| | - Hiroyuki Iida
- Clinical PharmacologyOno Pharmaceutical Co., Ltd.OsakaJapan
| | - Maki Kondo
- Oncology Clinical DevelopmentOno Pharmaceutical Co., Ltd.OsakaJapan
| | - Noboru Yamamoto
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
4
|
Hoffmann SL, Seminoff K, McKemie DS, Kass PH, Knych HK. Pharmacokinetics of grapiprant and effects on TNF-alpha concentrations following oral administration to horses. J Vet Pharmacol Ther 2022; 45:467-472. [PMID: 35652132 PMCID: PMC10919302 DOI: 10.1111/jvp.13076] [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: 03/25/2022] [Revised: 04/27/2022] [Accepted: 05/19/2022] [Indexed: 12/01/2022]
Abstract
Grapiprant is a prostaglandin E2 receptor antagonist that has been found to be an effective anti-inflammatory in dogs and that is devoid of some of the adverse effects associated with traditional NSAIDs that elicit their effects through inhibition of PGE2 production. Previously published reports have described the pharmacokinetics of this drug in horses when administered at 2 mg/kg; however, pharmacodynamic effects in this species have yet to be described. The objective of the current study was to describe the pharmacokinetics and pharmacodynamics of grapiprant at a higher dose. Eight horses received a single oral administration of 15 mg/kg. Plasma concentrations were determined for 96 h using liquid chromatography-tandem mass spectrometry. Non-compartmental analysis was used to determine pharmacokinetic parameters. Pharmacodynamic effects were assessed ex vivo by stimulating blood samples with PGE2 and determining TNF-ɑ concentrations. Maximum concentration, time to maximum concentration and area under the curve were 327.5 (188.4-663.0) ng/ml, 1 (0.75-2.0) hour and 831.8 (512.6-1421.6) h*ng/ml, respectively. The terminal half-life was 11.1 (8.27-21.2) hr. Significant stimulation of TNF alpha was noted for 2-4 h post-drug administration. Results of this study suggest a short duration of EP4 receptor engagement when administered at a dose of 15 mg/kg.
Collapse
Affiliation(s)
- Silke L. Hoffmann
- K.L Maddy Equine Analytical Pharmacology Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Kelsey Seminoff
- K.L Maddy Equine Analytical Pharmacology Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Daniel S. McKemie
- K.L Maddy Equine Analytical Pharmacology Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Philip H. Kass
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Heather K. Knych
- K.L Maddy Equine Analytical Pharmacology Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
5
|
Wang W, He J, Yang J, Zhang C, Cheng Z, Zhang Y, Zhang Q, Wang P, Tang S, Wang X, Liu M, Lu W, Zhang HK. Scaffold Hopping Strategy to Identify Prostanoid EP4 Receptor Antagonists for Cancer Immunotherapy. J Med Chem 2022; 65:7896-7917. [PMID: 35640059 DOI: 10.1021/acs.jmedchem.2c00448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer cells can effectively suppress the natural immune response in humans, and prostaglandin E2 (PGE2) is a key mediator in the development of tumor cell resistance to immunotherapy. As a major contributor to PGE2-elicited immunosuppressive activity, the EP4 receptor promotes tumor development and progression in the tumor microenvironment, and the development of selective and potent EP4 receptor antagonists should have promising potential for tumor immunotherapy. Aiming at improving the drug-like properties, a series of 4,7-dihydro-5H-thieno[2,3-c]pyran derivatives were designed and synthesized through a scaffold hopping strategy. The most promising compound 47 exhibited good EP4 antagonistic activity and excellent subtype selectivity, as well as favorable drug-like properties. It effectively suppressed the expression of multiple immunosuppression-related genes in macrophages. Meanwhile, oral administration of compound 47, alone or in combination with anti-PD-1 antibody, significantly enhanced the antitumor immune response and inhibited tumor growth in the mouse CT26 colon carcinoma model.
Collapse
Affiliation(s)
- Wei Wang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Jiacheng He
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Junjie Yang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Chan Zhang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Zhiyuan Cheng
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Yao Zhang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qiansen Zhang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Peili Wang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Shuowen Tang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Xin Wang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Mingyao Liu
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Weiqiang Lu
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Han-Kun Zhang
- Drug Discovery Unit, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| |
Collapse
|
6
|
Wang H, Qin Z, Yan A. Classification models and SAR analysis on CysLT1 receptor antagonists using machine learning algorithms. Mol Divers 2021; 25:1597-1616. [PMID: 33534023 DOI: 10.1007/s11030-020-10165-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022]
Abstract
Cysteinyl leukotrienes 1 (CysLT1) receptor is a promising drug target for rhinitis or other allergic diseases. In our study, we built classification models to predict bioactivities of CysLT1 receptor antagonists. We built a dataset with 503 CysLT1 receptor antagonists which were divided into two groups: highly active molecules (IC50 < 1000 nM) and weakly active molecules (IC50 ≥ 1000 nM). The molecules were characterized by several descriptors including CORINA descriptors, MACCS fingerprints, Morgan fingerprint and molecular SMILES. For CORINA descriptors and two types of fingerprints, we used the random forests (RF) and deep neural networks (DNN) to build models. For molecular SMILES, we used recurrent neural networks (RNN) with the self-attention to build models. The accuracies of test sets for all models reached 85%, and the accuracy of the best model (Model 2C) was 93%. In addition, we made structure-activity relationship (SAR) analyses on CysLT1 receptor antagonists, which were based on the output from the random forest models and RNN model. It was found that highly active antagonists usually contained the common substructures such as tetrazoles, indoles and quinolines. These substructures may improve the bioactivity of the CysLT1 receptor antagonists.
Collapse
Affiliation(s)
- Hongzhao Wang
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, University of Chemical Technology, Beijing, People's Republic of China
| | - Zijian Qin
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, University of Chemical Technology, Beijing, People's Republic of China
| | - Aixia Yan
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, University of Chemical Technology, Beijing, People's Republic of China.
| |
Collapse
|
7
|
Heeney A, Rogers AC, Mohan H, Mc Dermott F, Baird AW, Winter DC. Prostaglandin E 2 receptors and their role in gastrointestinal motility - Potential therapeutic targets. Prostaglandins Other Lipid Mediat 2021; 152:106499. [PMID: 33035691 DOI: 10.1016/j.prostaglandins.2020.106499] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/20/2020] [Accepted: 10/01/2020] [Indexed: 12/19/2022]
Abstract
Prostaglandin E2 (PGE2) is found throughout the gastrointestinal tract in a diverse variety of functions and roles. The recent discovery of four PGE2 receptor subtypes in intestinal muscle layers as well as in the enteric plexus has led to much interest in the study of their roles in gut motility. Gut dysmotility has been implicated in functional disease processes including irritable bowel syndrome (IBS) and slow transit constipation, and lubiprostone, a PGE2 derivative, has recently been licensed to treat both conditions. The diversity of actions of PGE2 in the intestinal tract is attributed to its differing effects on its downstream receptor types, as well as their varied distribution in the gut, in both health and disease. This review aims to identify the role and distribution of PGE2 receptors in the intestinal tract, and aims to elucidate their distinct role in gut motor function, with a specific focus on functional intestinal pathologies.
Collapse
Affiliation(s)
- A Heeney
- Department of Physiology, College of Life Sciences, University College Dublin, Dublin, Ireland; Department of Surgery, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.
| | - A C Rogers
- Department of Physiology, College of Life Sciences, University College Dublin, Dublin, Ireland; Department of Surgery, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - H Mohan
- Department of Physiology, College of Life Sciences, University College Dublin, Dublin, Ireland; Department of Surgery, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - F Mc Dermott
- Department of Physiology, College of Life Sciences, University College Dublin, Dublin, Ireland
| | - A W Baird
- Department of Physiology, College of Life Sciences, University College Dublin, Dublin, Ireland
| | - D C Winter
- Institute for Clinical Outcomes, Research and Education (ICORE), St Vincent's University Hospital, Elm Park, Dublin 4, Ireland; Department of Surgery, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| |
Collapse
|
8
|
Robertson-Plouch C, Stille JR, Liu P, Smith C, Brown D, Warner M, Hu L, Fisher MJ. A randomized clinical efficacy study targeting mPGES1 or EP4 in dogs with spontaneous osteoarthritis. Sci Transl Med 2020; 11:11/516/eaaw9993. [PMID: 31666405 DOI: 10.1126/scitranslmed.aaw9993] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
Abstract
Canine studies of spontaneous osteoarthritis (OA) pain add valuable data supporting drug treatment mechanisms that may translate to humans. A multicenter, randomized, double-blind, placebo- and active-controlled study was conducted in client-owned dogs with moderate OA pain to evaluate efficacy of LYA, an inhibitor of microsomal prostaglandin E synthase-1 (mPGES1), an EP4 antagonist (LYB), and carprofen, versus placebo. Of 255 dogs screened, 163 were randomized (placebo/LYA/LYB/carprofen: n = 43/39/42/39) and 158 completed treatment. Efficacy versus placebo was assessed using Bayesian mixed-effect model for repeated measure analyses of the Canine Brief Pain Inventory (CBPI) pain interference score (PIS; primary endpoint), pain severity score, and overall impression, as well as the Liverpool Osteoarthritis in Dogs (LOAD) mobility score. The posterior probability that the difference to placebo was <0 at week 2 was 80% for LYA and 54% for LYB for CBPI PIS (both <95% predefined threshold). For secondary endpoints, the posterior probability that the difference to placebo was <0 at week 2 ranged from 89 to 96% for LYA and from 56 to 89% for LYB. The posterior probabilities comparing carprofen to placebo groups were ≥90% for all efficacy endpoints. The proportion of dogs with one or more adverse event was not significantly different from placebo (32.6%) for LYA (35.9%) or carprofen (25.6%), but the rate for LYB (59.5%) was higher versus placebo (P = 0.017). LYA treatment demonstrated consistent improvement in all efficacy measures, suggesting that inhibition of mPGES1 may be an effective treatment for chronic pain associated with OA.
Collapse
Affiliation(s)
| | - John R Stille
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| | - Peng Liu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Claire Smith
- Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - Dorothy Brown
- Elanco, Eli Lilly and Company, Indianapolis, IN 46140, USA.,School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Margaret Warner
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Leijun Hu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Matthew J Fisher
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| |
Collapse
|
9
|
Abstract
BACKGROUND Patent ductus arteriosus (PDA) is common in premature infants. Cyclooxygenase inhibitors such as indomethacin, which inhibit prostaglandin E2(PGE2) synthesis, are currently the sole treatments for patients with PDA. Their efficacy are, however, frequently limited, and adverse effects are problematic. Because the PGE2-specific receptor EP4 selectively expresses in rat ductus arteriosus (DA), it is hypothesized that EP4 inhibition would promote DA closure with fewer side-effects.Methods and Results:A new chemical compound EP4 antagonist, RQ-15986 (renamed from CJ-042794), was used. Whether RQ-15986 selectively contracted the DA was examined by measuring the isometric tension of rat DA ex vivo at embryonic day 19 (e19) and e21. RQ-15986 at a dose of 10-6mol/L increased the isometric tension of the DA up to 44.8±6.2% and 69.1±12.9% to the maximal KCl-induced tension at e19 and e21 respectively. The effect of RQ-15986 on rat DA in vivo was also tested by using a rapid whole-body freezing method. RQ-15986 inhibited PGE1-induced DA dilatation in neonatal rats. Furthermore, RQ-15986 contracted the DA in a dose-dependent manner, and the constriction was greater at e21 than at e19. Moreover, RQ-15986 did not contract the aorta or the marginal artery of the colon. CONCLUSIONS EP4 inhibition contracts rat DA with fewer side-effects. EP4 inhibition is a promising alternative strategy to treat patients with PDA.
Collapse
Affiliation(s)
- Toshiki Sakuma
- Department of Cell Physiology, The Jikei University School of Medicine
| | - Toru Akaike
- Department of Cell Physiology, The Jikei University School of Medicine
| | - Susumu Minamisawa
- Department of Cell Physiology, The Jikei University School of Medicine
| |
Collapse
|
10
|
Mamun A, Yokoyama U, Saito J, Ito S, Hiromi T, Umemura M, Fujita T, Yasuda S, Minami T, Goda M, Uchida K, Suzuki S, Masuda M, Ishikawa Y. A selective antagonist of prostaglandin E receptor subtype 4 attenuates abdominal aortic aneurysm. Physiol Rep 2018; 6:e13878. [PMID: 30230255 PMCID: PMC6144453 DOI: 10.14814/phy2.13878] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 12/15/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive disease that has an increasing prevalence with aging, but no effective pharmacological therapy to attenuate AAA progression is currently available. We reported that the prostaglandin E receptor EP4 plays roles in AAA progression. Here, we show the effect of CJ-42794, a selective EP4 antagonist, on AAA using two mouse models (angiotensin II- and CaCl2 -induced AAAs) and human aortic smooth muscle cells isolated from AAA tissue. Oral administration of CJ-42794 (0.2 mg/kg per day) for 4 weeks significantly decreased AAA formation in ApoE-/- mice infused with angiotensin II (1 μg/kg per min), in which elastic fiber degradation and activations of matrix metalloproteinase (MMP)-2 and MMP-9 were attenuated. Interleukin-6 (IL-6) proteins were highly expressed in the medial layer of angiotensin II-induced mouse AAA tissues, whereas this expression was significantly decreased in mice treated with CJ-42794. AAA formation induced by periaortic CaCl2 application in wild-type mice was also reduced by oral administration of CJ-42794 for 4 weeks. After oral administration of CJ-42794 beginning 2 weeks after periaortic CaCl2 application and continuing for an additional 4 weeks, the aortic diameter and elastic fiber degradation grade were significantly smaller in CJ-42794-treated mice than in untreated mice. Additionally, in smooth muscle cells isolated from human AAA tissues, stimulation of CJ-42794 inhibited PGE2 -induced IL-6 secretion in a dose-dependent manner and decreased PGE2 -induced MMP-2 activity. These data suggest that inhibition of EP4 has the potential to be a pharmacological strategy for attenuation of AAA progression.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Apolipoproteins E/deficiency
- Cells, Cultured
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Sulfonylurea Compounds/pharmacology
- Sulfonylurea Compounds/therapeutic use
Collapse
Affiliation(s)
- Al Mamun
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Utako Yokoyama
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Junichi Saito
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Satoko Ito
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Taro Hiromi
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
- Department of Emergency medicineGraduate School of MedicineYokohama City UniversityYokohamaJapan
| | - Masanari Umemura
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Takayuki Fujita
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Shota Yasuda
- Department of SurgeryYokohama City UniversityYokohamaJapan
| | - Tomoyuki Minami
- Cardiovascular CenterYokohama City University Medical CenterYokohamaJapan
| | - Motohiko Goda
- Department of SurgeryYokohama City UniversityYokohamaJapan
| | - Keiji Uchida
- Cardiovascular CenterYokohama City University Medical CenterYokohamaJapan
| | | | - Munetaka Masuda
- Department of SurgeryYokohama City UniversityYokohamaJapan
- Cardiovascular CenterYokohama City University Medical CenterYokohamaJapan
| | | |
Collapse
|
11
|
Selectively targeting prostanoid E (EP) receptor-mediated cell signalling pathways: Implications for lung health and disease. Pulm Pharmacol Ther 2018; 49:75-87. [DOI: 10.1016/j.pupt.2018.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/05/2018] [Accepted: 01/25/2018] [Indexed: 12/18/2022]
|
12
|
Jin Y, Smith C, Hu L, Coutant DE, Whitehurst K, Phipps K, McNearney TA, Yang X, Ackermann B, Pottanat T, Landschulz W. LY3127760, a Selective Prostaglandin E4 (EP4) Receptor Antagonist, and Celecoxib: A Comparison of Pharmacological Profiles. Clin Transl Sci 2017; 11:46-53. [PMID: 28857461 PMCID: PMC5759725 DOI: 10.1111/cts.12497] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022] Open
Abstract
Safety, tolerability, and pharmacology profiles of LY3127760, an EP4 antagonist, were explored in healthy subjects in a subject/investigator‐blind, parallel‐group, multiple‐ascending dose study. Cohorts consisted of 13 patients randomized to LY3127760, celecoxib (400 mg), or placebo (9:2:2 ratio) for 28 days. LY3127760 was well tolerated; the most commonly observed adverse events were gastrointestinal, similar to celecoxib. LY3127760 increased release of ex vivo tumor necrosis factor alpha after lipopolysaccharide/prostaglandin E2 stimulation when compared with placebo, suggesting a dose‐dependent blockade of the EP4 receptor. Compared with placebo, 24‐h urinary excretion of prostaglandin E metabolite was modestly increased; prostacyclin metabolite was inhibited; and thromboxane A2 metabolite was unchanged. Effects on sodium and potassium excretion were similar to those of celecoxib. We conclude that LY3127760 demonstrated similar effects on prostacyclin synthesis and renal sodium retention as celecoxib. These data support exploration of LY3127760 at daily doses of 60 mg to 600 mg in phase II trials. This trial's registration number: NCT01968070.
Collapse
Affiliation(s)
- Yan Jin
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Claire Smith
- Eli Lilly and Company, Lilly UK, Windlesham, Surrey, UK
| | - Leijun Hu
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | | | | | - Xiao Yang
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | | |
Collapse
|
13
|
Chandrasekhar S, Yu X, Harvey AK, Oskins JL, Lin C, Wang X, Blanco M, Fisher MJ, Kuklish SL, Schiffler MA, Vetman T, Warshawsky AM, York JS, Bendele AM, Chambers MG. Analgesic and anti-inflammatory properties of novel, selective, and potent EP4 receptor antagonists. Pharmacol Res Perspect 2017; 5:e00316. [PMID: 28603634 PMCID: PMC5464344 DOI: 10.1002/prp2.316] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 11/07/2022] Open
Abstract
Prostaglandin (PG) E2 is the key driver of inflammation associated with arthritic conditions. Inhibitors of PGE 2 production (NSAIDs and Coxibs) are used to treat these conditions, but carry significant side effect risks due to the inhibition of all prostanoids that play important physiological function. The activities of PGE 2 are transduced through various receptor sub-types. Prostaglandin E2 type 4 receptor (EP4) is associated with the development of inflammation and autoimmunity. We therefore are interested in identifying novel EP4 antagonists to treat the signs and symptoms of arthritis without the potential side effects of PGE 2 modulators such as NSAIDs and Coxibs. Novel EP4 antagonists representing distinct chemical scaffolds were identified using a variety of in vitro functional assays and were shown to be selective and potent. The compounds were shown to be efficacious in animal models of analgesia, inflammation, and arthritis.
Collapse
Affiliation(s)
| | - Xiao‐Peng Yu
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Anita K. Harvey
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Jennifer L. Oskins
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Chaohua Lin
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Xushan Wang
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Maria‐Jesus Blanco
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Matthew J. Fisher
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Steven L. Kuklish
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | | | - Tatiana Vetman
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Alan M. Warshawsky
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | - Jeremy S. York
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| | | | - Mark G. Chambers
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndiana46285
| |
Collapse
|
14
|
Synthesis and evaluation of 18 F-labeled CJ-042794 for imaging prostanoid EP4 receptor expression in cancer with positron emission tomography. Bioorg Med Chem Lett 2017; 27:2094-2098. [DOI: 10.1016/j.bmcl.2017.03.078] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 03/24/2017] [Accepted: 03/25/2017] [Indexed: 12/25/2022]
|
15
|
Bhooshan N, Staats PN, Fulton AM, Feliciano JL, Edelman MJ. Prostaglandin E Receptor EP4 expression, survival and pattern of recurrence in locally advanced NSCLC. Lung Cancer 2016; 101:88-91. [PMID: 27794413 DOI: 10.1016/j.lungcan.2016.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/01/2016] [Accepted: 09/14/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Elevated COX-2 expression has been correlated with inferior outcome in NSCLC. COX-2 catalyzes the transformation of arachidonate to PGE2. We and others have demonstrated that PGE2 induces proliferation and metastatic spread and immunosuppression through the G protein-coupled EP4 receptor. We hypothesized that EP4 expression on malignant cells would correlate with outcome and patterns of relapse after treatment of LANSCLC stage IIIA (7th edition, N2+). METHODS Tissue specimens from 41 pts treated for LANSC at UMGCC were obtained. A tissue microarray was prepared and examined for EP4 expression. Intensity of staining was scored semi-quantitatively as 0-4 in both the nuclear and cytoplasmic compartments by a pathologist blinded to the clinical data. RESULTS EP4 nuclear staining 0-1 vs. 2+ was associated with overall survival, (OS) (44.3 vs. 18 mo; HR=0.41, p=0.024) and numerically superior progression free survival (PFS) (16.4 vs. 10.2 mo, p=0.16). EP4 cytoplasmic staining did not correlate with OS (0-1 vs. 2+, 23.8 vs. 28.8 mo; HR=1.2, p=0.81). Relapse pattern (no relapse or local vs. systemic) did not correlate with EP nuclear staining (p=1.0, X2). CONCLUSIONS This is the first clinical study of EP4 expression in lung cancer. There was a significant correlation between OS and nuclear EP4 expression, indicating that this is a potential therapeutic target. Studies with AT-007, a specific inhibitor of EP4, are planned to commence this year.
Collapse
Affiliation(s)
- Neha Bhooshan
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA
| | - Paul N Staats
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA
| | - Amy M Fulton
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA; Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Josephine L Feliciano
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA
| | - Martin J Edelman
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
16
|
Gill SK, Yao Y, Kay LJ, Bewley MA, Marriott HM, Peachell PT. The anti-inflammatory effects of PGE 2 on human lung macrophages are mediated by the EP 4 receptor. Br J Pharmacol 2016; 173:3099-3109. [PMID: 27460634 DOI: 10.1111/bph.13565] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE PGE2 inhibits cytokine generation from human lung macrophages. However, the EP receptor that mediates this beneficial anti-inflammatory effect of PGE2 has not been defined. The aim of this study was to identify the EP receptor by which PGE2 inhibits cytokine generation from human lung macrophages. This was determined by using recently developed EP receptor ligands. EXPERIMENTAL APPROACH The effects of PGE2 and EP-selective agonists on LPS-induced generation of TNF-α and IL-6 from macrophages were evaluated. The effects of EP2 -selective (PF-04852946, PF-04418948) and EP4 -selective (L-161,982, CJ-042794) receptor antagonists on PGE2 responses were studied. The expression of EP receptor subtypes by human lung macrophages was determined by RT-PCR. KEY RESULTS PGE2 inhibited LPS-induced and Streptococcus pneumoniae-induced cytokine generation from human lung macrophages. Analysis of mRNA levels indicated that macrophages expressed EP2 and EP4 receptors. L-902,688 (EP4 receptor-selective agonist) was considerably more potent than butaprost (EP2 receptor-selective agonist) as an inhibitor of TNF-α generation from macrophages. EP2 receptor-selective antagonists had marginal effects on the PGE2 inhibition of TNF-α generation, whereas EP4 receptor-selective antagonists caused rightward shifts in the PGE2 concentration-response curves. CONCLUSIONS AND IMPLICATIONS These studies demonstrate that the EP4 receptor is the principal receptor that mediates the anti-inflammatory effects of PGE2 on human lung macrophages. This suggests that EP4 receptor agonists could be effective anti-inflammatory agents in human lung disease.
Collapse
Affiliation(s)
- Sharonjit K Gill
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Yiwen Yao
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Linda J Kay
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Martin A Bewley
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Helen M Marriott
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Peter T Peachell
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK.
| |
Collapse
|
17
|
Mazaleuskaya LL, Lawson JA, Li X, Grant G, Mesaros C, Grosser T, Blair IA, Ricciotti E, FitzGerald GA. A broad-spectrum lipidomics screen of antiinflammatory drug combinations in human blood. JCI Insight 2016; 1. [PMID: 27547824 DOI: 10.1172/jci.insight.87031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Current methods of drug screening in human blood focus on the immediate products of the affected pathway and mostly rely on approaches that lack sensitivity and the capacity for multiplex analysis. We have developed a sensitive and selective method based on ultra-performance liquid chromatography-tandem mass spectrometry to scan the effect of drugs on the bioactive eicosanoid lipidome in vitro and ex vivo. Using small sample sizes, we can reproducibly measure a broad spectrum of eicosanoids in human blood and capture drug-induced substrate rediversion and unexpected shifts in product formation. Microsomal prostaglandin E synthase-1 (mPGES-1) is an antiinflammatory drug target alternative to COX-1/-2. Contrasting effects of targeting mPGES-1 versus COX-1/-2, due to differential substrate shifts across the lipidome, were observed and can be used to rationalize and evaluate drug combinations. Finally, the in vitro results were extrapolated to ex vivo studies by administration of the COX-2 inhibitor, celecoxib, to volunteers, illustrating how this approach can be used to integrate preclinical and clinical studies during drug development.
Collapse
Affiliation(s)
- Liudmila L Mazaleuskaya
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John A Lawson
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xuanwen Li
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory Grant
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Clementina Mesaros
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tilo Grosser
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Schiffler MA, Chandrasekhar S, Fisher MJ, Harvey A, Kuklish SL, Wang XS, Warshawsky AM, York JS, Yu XP. Discovery and characterization of a potent and selective EP4 receptor antagonist. Bioorg Med Chem Lett 2015; 25:3176-8. [DOI: 10.1016/j.bmcl.2015.05.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 12/18/2022]
|
19
|
Ma X, Holt D, Kundu N, Reader J, Goloubeva O, Take Y, Fulton AM. A prostaglandin E (PGE) receptor EP4 antagonist protects natural killer cells from PGE 2-mediated immunosuppression and inhibits breast cancer metastasis. Oncoimmunology 2014; 2:e22647. [PMID: 23482441 PMCID: PMC3583931 DOI: 10.4161/onci.22647] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cyclooxygenase-2 is frequently upregulated in epithelial tumors and contributes to poor outcomes in multiple malignancies. The COX-2 product prostaglandin E2 (PGE2) promotes tumor growth and metastasis by acting on a family of four G protein-coupled receptors (EP1-4). Using a novel small molecule EP4 antagonist (RQ-15986) and a syngeneic murine model of metastatic breast cancer, we determined the effect of EP4 blockade on innate immunity and tumor biology. Natural killer (NK)-cell functions are markedly depressed in mice bearing murine mammary tumor 66.1 or 410.4 cells owing to the actions of PGE2 on NK cell EP4 receptors. The EP4 agonist PGE1-OH inhibits NK functions in vitro, and this negative regulation is blocked by RQ-15986. Likewise, the treatment of tumor-bearing mice with RQ-15986 completely protected NK cells from the immunosuppressive effects of the tumor microenvironment in vivo. RQ-15986 also has direct effects on EP4 expressed by tumor cells, inhibiting the PGE2-mediated activation of adenylate cyclase and blocking PGE2-induced tumor cell migration. The pretreatment of tumor cells with a non-cytotoxic concentration of RQ-15986 inhibited lung colonization, a beneficial effect that was lost in mice depleted of NK cells. The oral administration of RQ-15986 inhibited the growth of tumor cells implanted into mammary glands and their spontaneous metastatic colonization to the lungs, resulting in improved survival. Our findings reveal that EP4 antagonism prevents tumor-mediated NK-cell immunosuppression and demonstrates the anti-metastatic activity of a novel EP4 antagonist. These observations support the investigation of EP4 antagonists in clinical trials.
Collapse
Affiliation(s)
- Xinrong Ma
- University of Maryland Greenebaum Cancer Center; Baltimore, MD USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Majumder M, Xin X, Liu L, Girish GV, Lala PK. Prostaglandin E2 receptor EP4 as the common target on cancer cells and macrophages to abolish angiogenesis, lymphangiogenesis, metastasis, and stem-like cell functions. Cancer Sci 2014; 105:1142-51. [PMID: 24981602 PMCID: PMC4462388 DOI: 10.1111/cas.12475] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 06/21/2014] [Accepted: 06/26/2014] [Indexed: 01/09/2023] Open
Abstract
We previously established that COX-2 overexpression promotes breast cancer progression and metastasis. As long-term use of COX-2 inhibitors (COX-2i) can promote thrombo-embolic events, we tested an alternative target, prostaglandin E2 receptor EP4 subtype (EP4), downstream of COX-2. Here we used the highly metastatic syngeneic murine C3L5 breast cancer model to test the role of EP4-expressing macrophages in vascular endothelial growth factor (VEGF)-C/D production, angiogenesis, and lymphangiogenesis in situ, the role of EP4 in stem-like cell (SLC) functions of tumor cells, and therapeutic effects of an EP4 antagonist RQ-15986 (EP4A). C3L5 cells expressed all EP receptors, produced VEGF-C/D, and showed high clonogenic tumorsphere forming ability in vitro, functions inhibited with COX-2i or EP4A. Treating murine macrophage RAW 264.7 cell line with COX-2i celecoxib and EP4A significantly reduced VEGF-A/C/D production in vitro, measured with quantitative PCR and Western blots. Orthotopic implants of C3L5 cells in C3H/HeJ mice showed rapid tumor growth, angiogenesis, lymphangiogenesis (CD31/LYVE-1 and CD31/PROX1 immunostaining), and metastasis to lymph nodes and lungs. Tumors revealed high incidence of EP4-expressing, VEGF-C/D producing macrophages identified with dual immunostaining of F4/80 and EP4 or VEGF-C/D. Celecoxib or EP4A therapy at non-toxic doses abrogated tumor growth, lymphangiogenesis, and metastasis to lymph nodes and lungs. Residual tumors in treated mice revealed markedly reduced VEGF-A/C/D and phosphorylated Akt/ERK proteins, VEGF-C/D positive macrophage infiltration, and proliferative/apoptotic cell ratios. Knocking down COX-2 or EP4 in C3L5 cells or treating cells in vitro with celecoxib or EP4A and treating tumor-bearing mice in vivo with the same drug reduced SLC properties of tumor cells including preferential co-expression of COX-2 and SLC markers ALDH1A, CD44, OCT-3/4, β-catenin, and SOX-2. Thus, EP4 is an excellent therapeutic target to block stem-like properties, angiogenesis, and lymphangiogenesis induced by VEGF-A/C/D secreted by cancer cells and tumor infiltrating macrophages.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/drug therapy
- Adenocarcinoma/secondary
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Celecoxib
- Cell Line, Tumor
- Cell Proliferation
- Cyclooxygenase 2/metabolism
- Drug Screening Assays, Antitumor
- Female
- Lung Neoplasms/blood supply
- Lung Neoplasms/drug therapy
- Lung Neoplasms/secondary
- Lymphangiogenesis
- Lymphatic Metastasis
- Macrophages/metabolism
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred C3H
- Molecular Targeted Therapy
- Neoplasm Transplantation
- Neoplastic Stem Cells
- Neovascularization, Pathologic/drug therapy
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Tumor Burden
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Mousumi Majumder
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, Ontario, Canada
| | - Xiping Xin
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, Ontario, Canada
| | - Ling Liu
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, Ontario, Canada
| | - Gannareddy V Girish
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, Ontario, Canada
| | - Peeyush K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, Ontario, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, Ontario, Canada
| |
Collapse
|
21
|
Kay LJ, Gilbert M, Pullen N, Skerratt S, Farrington J, Seward EP, Peachell PT. Characterization of the EP receptor subtype that mediates the inhibitory effects of prostaglandin E2 on IgE-dependent secretion from human lung mast cells. Clin Exp Allergy 2014; 43:741-51. [PMID: 23786281 DOI: 10.1111/cea.12142] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/16/2013] [Accepted: 04/18/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Prostaglandin E2 (PGE2 ) has been shown to inhibit IgE-dependent histamine release from human lung mast cells. This effect of PGE2 is believed to be mediated by EP2 receptors. However, definitive evidence that this is the case has been lacking in the absence of EP2 -selective antagonists. Moreover, recent evidence has suggested that PGE2 activates EP4 receptors to inhibit respiratory cell function. OBJECTIVE The aim of this study was to determine the receptor by which PGE2 inhibits human lung mast cell responses by using recently developed potent and selective EP2 and EP4 receptor antagonists alongside other established EP receptor ligands. METHODS The effects of non-selective (PGE2 , misoprostol), EP2 -selective (ONO-AE1-259, AH13205, butaprost-free acid) and EP4 -selective (L-902,688, TCS251) agonists on IgE-dependent histamine release and cyclic-AMP generation in mast cells were determined. The effects of EP2 -selective (PF-04418948, PF-04852946) and EP4 -selective (CJ-042794, L-161,982) antagonists on PGE2 responses of mast cells were studied. The expression of EP receptor subtypes was determined by RT-PCR. RESULTS Prostaglandin E2 , EP2 agonists and EP4 agonists inhibited IgE-dependent histamine release from mast cells. PGE2 and EP2 agonists, but not EP4 agonists, increased cyclic-AMP levels in mast cells. EP4 -selective antagonists did not affect the PGE2 inhibition of histamine release, whereas EP2 -selective antagonists caused rightward shifts in the PGE2 concentration-response curves. RT-PCR studies indicated that mast cells expressed EP2 and EP4 receptors. CONCLUSIONS AND CLINICAL RELEVANCE Although human lung mast cells may express both EP2 and EP4 receptors, the principal mechanism by which PGE2 inhibits mediator release in mast cells is by activating EP2 receptors.
Collapse
Affiliation(s)
- L J Kay
- Academic Unit of Respiratory Medicine, The Medical School, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | |
Collapse
|
22
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 505] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
23
|
Prostaglandin E receptor EP4 is a therapeutic target in breast cancer cells with stem-like properties. Breast Cancer Res Treat 2013; 143:19-31. [PMID: 24281828 PMCID: PMC3889836 DOI: 10.1007/s10549-013-2779-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 11/15/2013] [Indexed: 01/15/2023]
Abstract
The cyclooxygenase pathway is strongly implicated in breast cancer progression but the role of this pathway in the biology of breast cancer stem/progenitor cells has not been defined. Recent attention has focused on targeting the cyclooxygenase 2 (COX-2) pathway downstream of the COX-2 enzyme by blocking the activities of individual prostaglandin E (EP) receptors. Prostaglandin E receptor 4 (EP4) is widely expressed in primary invasive ductal carcinomas of the breast and antagonizing this receptor with small molecule inhibitors or shRNA directed to EP4 inhibits metastatic potential in both syngeneic and xenograft models. Breast cancer stem/progenitor cells are defined as a subpopulation of cells that drive tumor growth, metastasis, treatment resistance, and relapse. Mammosphere-forming breast cancer cells of human (MDA-MB-231, SKBR3) or murine (66.1, 410.4) origin of basal-type, Her-2 phenotype and/or with heightened metastatic capacity upregulate expression of both EP4 and COX-2 and are more tumorigenic compared to the bulk population. In contrast, luminal-type or non-metastatic counterparts (MCF7, 410, 67) do not increase COX-2 and EP4 expression in mammosphere culture. Treatment of mammosphere-forming cells with EP4 inhibitors (RQ-15986, AH23848, Frondoside A) or EP4 gene silencing, but not with a COX inhibitor (Indomethacin) reduces both mammosphere-forming capacity and the expression of phenotypic markers (CD44hi/CD24low, aldehyde dehydrogenase) of breast cancer stem cells. Finally, an orally delivered EP4 antagonist (RQ-08) reduces the tumor-initiating capacity and markedly inhibits both the size of tumors arising from transplantation of mammosphere-forming cells and phenotypic markers of stem cells in vivo. These studies support the continued investigation of EP4 as a potential therapeutic target and provide new insight regarding the role of EP4 in supporting a breast cancer stem cell/tumor-initiating phenotype.
Collapse
|
24
|
Sekiguchi F, Aoki Y, Nakagawa M, Kanaoka D, Nishimoto Y, Tsubota-Matsunami M, Yamanaka R, Yoshida S, Kawabata A. AKAP-dependent sensitization of Ca(v) 3.2 channels via the EP(4) receptor/cAMP pathway mediates PGE(2) -induced mechanical hyperalgesia. Br J Pharmacol 2013; 168:734-45. [PMID: 22924591 DOI: 10.1111/j.1476-5381.2012.02174.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 07/30/2012] [Accepted: 08/15/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE The Ca(v) 3.2 isoform of T-type Ca(2+) channels (T channels) is sensitized by hydrogen sulfide, a pro-nociceptive gasotransmitter, and also by PKA that mediates PGE(2) -induced hyperalgesia. Here we examined and analysed Ca(v) 3.2 sensitization via the PGE(2) /cAMP pathway in NG108-15 cells that express Ca(v) 3.2 and produce cAMP in response to PGE(2) , and its impact on mechanical nociceptive processing in rats. EXPERIMENTAL APPROACH In NG108-15 cells and rat dorsal root ganglion (DRG) neurons, T-channel-dependent currents (T currents) were measured with the whole-cell patch-clamp technique. The molecular interaction of Ca(v) 3.2 with A-kinase anchoring protein 150 (AKAP150) and its phosphorylation were analysed by immunoprecipitation/immunoblotting in NG108-15 cells. Mechanical nociceptive threshold was determined by the paw pressure test in rats. KEY RESULTS In NG108-15 cells and/or rat DRG neurons, dibutyryl cAMP (db-cAMP) or PGE(2) increased T currents, an effect blocked by AKAP St-Ht31 inhibitor peptide (AKAPI) or KT5720, a PKA inhibitor. The effect of PGE(2) was abolished by RQ-00015986-00, an EP(4) receptor antagonist. AKAP150 was co-immunoprecipitated with Ca(v) 3.2, regardless of stimulation with db-cAMP, and Ca(v) 3.2 was phosphorylated by db-cAMP or PGE(2) . In rats, intraplantar (i.pl.) administration of db-cAMP or PGE(2) caused mechanical hyperalgesia, an effect suppressed by AKAPI, two distinct T-channel blockers, NNC 55-0396 and ethosuximide, or ZnCl(2) , known to inhibit Ca(v) 3.2 among T channels. Oral administration of RQ-00015986-00 suppressed the PGE(2) -induced mechanical hyperalgesia. CONCLUSION AND IMPLICATIONS Our findings suggest that PGE(2) causes AKAP-dependent phosphorylation and sensitization of Ca(v) 3.2 through the EP(4) receptor/cAMP/PKA pathway, leading to mechanical hyperalgesia in rats.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Division of Pharmacology & Pathophysiology, Kinki University School of Pharmacy, Higashi-Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Konya V, Marsche G, Schuligoi R, Heinemann A. E-type prostanoid receptor 4 (EP4) in disease and therapy. Pharmacol Ther 2013; 138:485-502. [PMID: 23523686 PMCID: PMC3661976 DOI: 10.1016/j.pharmthera.2013.03.006] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/07/2013] [Indexed: 01/06/2023]
Abstract
The large variety of biological functions governed by prostaglandin (PG) E2 is mediated by signaling through four distinct E-type prostanoid (EP) receptors. The availability of mouse strains with genetic ablation of each EP receptor subtype and the development of selective EP agonists and antagonists have tremendously advanced our understanding of PGE2 as a physiologically and clinically relevant mediator. Moreover, studies using disease models revealed numerous conditions in which distinct EP receptors might be exploited therapeutically. In this context, the EP4 receptor is currently emerging as most versatile and promising among PGE2 receptors. Anti-inflammatory, anti-thrombotic and vasoprotective effects have been proposed for the EP4 receptor, along with its recently described unfavorable tumor-promoting and pro-angiogenic roles. A possible explanation for the diverse biological functions of EP4 might be the multiple signaling pathways switched on upon EP4 activation. The present review attempts to summarize the EP4 receptor-triggered signaling modules and the possible therapeutic applications of EP4-selective agonists and antagonists.
Collapse
Key Words
- ampk, amp-activated protein kinase
- camp, cyclic adenylyl monophosphate
- cftr, cystic fibrosis transmembrane conductance regulator
- clc, chloride channel
- cox, cyclooxygenase
- creb, camp-response element-binding protein
- dp, d-type prostanoid receptor
- dss, dextran sodium sulfate
- egfr, epidermal growth factor receptor
- enos, endothelial nitric oxide synthase
- ep, e-type prostanoid receptor
- epac, exchange protein activated by camp
- eprap, ep4 receptor-associated protein
- erk, extracellular signal-regulated kinase
- fem1a, feminization 1 homolog a
- fp, f-type prostanoid receptor
- grk, g protein-coupled receptor kinase
- 5-hete, 5-hydroxyeicosatetraenoic acid
- icer, inducible camp early repressor
- icam-1, intercellular adhesion molecule-1
- ig, immunoglobulin
- il, interleukin
- ifn, interferon
- ip, i-type prostanoid receptor
- lps, lipopolysaccharide
- map, mitogen-activated protein kinase
- mcp, monocyte chemoattractant protein
- mek, map kinase kinase
- nf-κb, nuclear factor kappa-light-chain-enhancer of activated b cells
- nsaid, non-steroidal anti-inflammatory drug
- pg, prostaglandin
- pi3k, phosphatidyl insositol 3-kinase
- pk, protein kinase
- tp, t-type prostanoid receptor
- tx, thromboxane receptor
- prostaglandins
- inflammation
- vascular disease
- cancerogenesis
- renal function
- osteoporosis
Collapse
Affiliation(s)
| | | | | | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| |
Collapse
|
26
|
Abstract
Prostaglandin antagonists, with their pharmacological effects, are well-known drugs capable of treating widely diffused illnesses, including pain and inflammation disorders. In recent years, a major research focus has been devoted to the identification of agents able to selectively antagonize each receptor with which prostaglandins interact. This review attempts to give a broad overview of molecules capable of selectively blocking the prostaglandin PGE2 EP4 receptor. Further therapeutic applications and uses have also been disccussed, including the first drug candidate to have reached clinical trials within the last few years.
Collapse
|
27
|
Elberg D, Turman MA, Pullen N, Elberg G. Prostaglandin E2 stimulates cystogenesis through EP4 receptor in IMCD-3 cells. Prostaglandins Other Lipid Mediat 2012; 98:11-6. [PMID: 22503965 DOI: 10.1016/j.prostaglandins.2012.03.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 03/21/2012] [Accepted: 03/30/2012] [Indexed: 10/28/2022]
Abstract
Previously, we demonstrated that prostaglandin E(2) (PGE(2)) induced cAMP and cyst formation through PGE(2) receptor-2 (EP2) activity in human autosomal-dominant polycystic kidney disease (ADPKD) epithelial cells. In this study, we determined the role of EP2 and EP4 receptors in mediating PGE(2) stimulation of cAMP signaling and cystogenesis in mouse renal epithelial cells using the inner medullary collecting duct-3 (IMCD-3) cell line. In contrast to human ADPKD cells, using novel EP2 and EP4 antagonists, we found that IMCD-3 cells expressed functional EP4 but not EP2, which stimulated cAMP formation and led to cyst formation in 3D culture system. The involvement of EP4 receptors in IMCD-3 cells was further supported by the specific effect of EP4 siRNA that inhibited PGE(2)-induced cystogenesis. We also observed different cellular localization of EP2 or EP4 receptors in IMCD-3 transfected cells. Collectively, our results suggest an important role of different expression of EP2 or EP4 receptors in the regulation of cystogenesis.
Collapse
Affiliation(s)
- Dorit Elberg
- Department of Pediatrics, Section of Nephrology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | | | | | | |
Collapse
|
28
|
af Forselles KJ, Root J, Clarke T, Davey D, Aughton K, Dack K, Pullen N. In vitro and in vivo characterization of PF-04418948, a novel, potent and selective prostaglandin EP₂ receptor antagonist. Br J Pharmacol 2012; 164:1847-56. [PMID: 21595651 DOI: 10.1111/j.1476-5381.2011.01495.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Studies of the role of the prostaglandin EP(2) receptor) have been limited by the availability of potent and selective antagonist tools. Here we describe the in vitro/in vivo pharmacological characterization of a novel EP(2) receptor antagonist, PF-04418948 (1-(4-fluorobenzoyl)-3-{[(6-methoxy-2-naphthyl)oxy]methyl} azetidine-3-carboxylic acid). EXPERIMENTAL APPROACH Functional antagonist potency was assessed in cell-based systems expressing human EP(2) receptors and native tissue preparations from human, dog and mouse. The selectivity of PF-04418948 was assessed against related receptors and a panel of GPCRs, ion channels and enzymes. The ability of PF-04418948 to pharmacologically block EP(2) receptor function in vivo was tested in rats. KEY RESULTS PF-04418948 inhibited prostaglandin E(2)(PGE(2))-induced increase in cAMP in cells expressing EP(2) receptors with a functional K(B) value of 1.8 nM. In human myometrium, PF-04418948 produced a parallel, rightward shift of the butaprost-induced inhibition of the contractions induced by electrical field stimulation with an apparent K(B) of 5.4 nM. In dog bronchiole and mouse trachea, PF-04418948 produced parallel rightward shifts of the PGE(2)-induced relaxation curve with a K(B) of 2.5 nM and an apparent K(B) of 1.3 nM respectively. Reversal of the PGE(2)-induced relaxation in the mouse trachea by PF-04418948 produced an IC(50) value of 2.7 nM. Given orally, PF-04418948 attenuated the butaprost-induced cutaneous blood flow response in rats. PF-04418948 was selective for EP(2) receptors over homologous and unrelated receptors, enzymes and channels. CONCLUSIONS AND IMPLICATIONS PF-04418948 is an orally active, potent and selective surmountable EP(2) receptor antagonist that should aid further elaboration of EP(2) receptor function.
Collapse
|
29
|
Stasi LP, Bhimani K, Borriello M, Canciani L, Caselli G, Colace F, Ferioli C, Kaswala M, Mennuni L, Piepoli T, Pucci S, Salvi M, Shirsath V, Zanelli T, Zerbi S. Synthesis, pharmacophore modeling and in vitro activity of 10,11-dihydrodibenzo[b,f]oxepine-4-carboxamide derivatives as novel and potent antagonists of the prostaglandin EP4 receptor. Bioorg Med Chem Lett 2011; 21:6336-40. [PMID: 21930381 DOI: 10.1016/j.bmcl.2011.08.102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 08/24/2011] [Accepted: 08/25/2011] [Indexed: 10/17/2022]
Abstract
The construction of a EP(4) antagonists pharmacophore model and the discovery of a highly potent oxepinic series of EP(4) antagonists is discussed. Compound 1a exhibits an excellent selectivity profile toward EP(2) receptor subtype and low cytochrome P450 inhibition potential.
Collapse
Affiliation(s)
- Luigi Piero Stasi
- Rottapharm Madaus, Medicinal Chemistry Department, via Valosa di Sopra 9, Monza 20900, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Holt D, Ma X, Kundu N, Fulton A. Prostaglandin E(2) (PGE (2)) suppresses natural killer cell function primarily through the PGE(2) receptor EP4. Cancer Immunol Immunother 2011; 60:1577-86. [PMID: 21681369 DOI: 10.1007/s00262-011-1064-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 06/07/2011] [Indexed: 12/29/2022]
Abstract
The COX-2 product prostaglandin E(2) (PGE(2)) contributes to the high metastatic capacity of breast tumors. Our published data indicate that inhibiting either PGE(2) production or PGE(2)-mediated signaling through the PGE(2) receptor EP4 reduces metastasis by a mechanism that requires natural killer (NK) cells. It is known that NK cell function is compromised by PGE(2), but very little is known about the mechanism by which PGE(2) affects NK effector activity. We now report the direct effects of PGE(2) on the NK cell. Endogenous murine splenic NK cells express all four PGE(2) receptors (EP1-4). We examined the role of EP receptors in three NK cell functions: migration, cytotoxicity, and cytokine release. Like PGE(2), the EP4 agonist PGE(1)-OH blocked NK cell migration to FBS and to four chemokines (ITAC, MIP-1α, SDF-1α, and CCL21). The EP2 agonist, Butaprost, inhibited migration to specific chemokines but not in response to FBS. In contrast to the inhibitory actions of PGE(2), the EP1/EP3 agonist Sulprostone increased migration. Unlike the opposing effects of EP4 vs. EP1/EP3 on migration, agonists of each EP receptor were uniformly inhibiting to NK-mediated cytotoxicity. The EP4 agonist, PGE(1)-OH, inhibited IFNγ production from NK cells. Agonists for EP1, EP2, and EP3 were not as effective at inhibiting IFNγ. Agonists of EP1, EP2, and EP4 all inhibited TNFα; EP4 agonists were the most potent. Thus, the EP4 receptor consistently contributed to loss of function. These results, taken together, support a mechanism whereby inhibiting PGE(2) production or preventing signaling through the EP4 receptor may prevent suppression of NK functions that are critical to the control of breast cancer metastasis.
Collapse
Affiliation(s)
- Dawn Holt
- Department of Pathology, School of Medicine, University of Maryland, Baltimore, USA
| | | | | | | |
Collapse
|
31
|
Colucci J, Boyd M, Berthelette C, Chiasson JF, Wang Z, Ducharme Y, Friesen R, Wrona M, Levesque JF, Denis D, Mathieu MC, Stocco R, Therien AG, Clarke P, Rowland S, Xu D, Han Y. Discovery of 4-[1-[([1-[4-(trifluoromethyl)benzyl]-1H-indol-7-yl]carbonyl)amino]cyclopropyl]benzoic acid (MF-766), a highly potent and selective EP4 antagonist for treating inflammatory pain. Bioorg Med Chem Lett 2010; 20:3760-3. [PMID: 20471829 DOI: 10.1016/j.bmcl.2010.04.065] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 04/14/2010] [Accepted: 04/16/2010] [Indexed: 11/18/2022]
Abstract
The discovery of a highly potent and selective EP(4) antagonist MF-766 is discussed. This N-benzyl indole derivative exhibits good pharmacokinetic profile and unprecedented in vivo potency in the rat AIA model.
Collapse
Affiliation(s)
- John Colucci
- Department of Medicinal Chemistry, Merck Frosst Canada Ltd, Kirkland, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Blouin M, Han Y, Burch J, Farand J, Mellon C, Gaudreault M, Wrona M, Lévesque JF, Denis D, Mathieu MC, Stocco R, Vigneault E, Therien A, Clark P, Rowland S, Xu D, O’Neill G, Ducharme Y, Friesen R. The Discovery of 4-{1-[({2,5-Dimethyl-4-[4-(trifluoromethyl)benzyl]-3-thienyl}carbonyl)amino]cyclopropyl}benzoic Acid (MK-2894), A Potent and Selective Prostaglandin E2 Subtype 4 Receptor Antagonist. J Med Chem 2010; 53:2227-38. [DOI: 10.1021/jm901771h] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
33
|
Novel strategies for the treatment of inflammatory hyperalgesia. Eur J Clin Pharmacol 2010; 66:429-44. [DOI: 10.1007/s00228-010-0784-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Accepted: 01/11/2010] [Indexed: 12/24/2022]
|
34
|
|
35
|
|
36
|
Jones RL, Giembycz MA, Woodward DF. Prostanoid receptor antagonists: development strategies and therapeutic applications. Br J Pharmacol 2009; 158:104-45. [PMID: 19624532 PMCID: PMC2795261 DOI: 10.1111/j.1476-5381.2009.00317.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2009] [Accepted: 04/07/2009] [Indexed: 01/17/2023] Open
Abstract
Identification of the primary products of cyclo-oxygenase (COX)/prostaglandin synthase(s), which occurred between 1958 and 1976, was followed by a classification system for prostanoid receptors (DP, EP(1), EP(2) ...) based mainly on the pharmacological actions of natural and synthetic agonists and a few antagonists. The design of potent selective antagonists was rapid for certain prostanoid receptors (EP(1), TP), slow for others (FP, IP) and has yet to be achieved in certain cases (EP(2)). While some antagonists are structurally related to the natural agonist, most recent compounds are 'non-prostanoid' (often acyl-sulphonamides) and have emerged from high-throughput screening of compound libraries, made possible by the development of (functional) assays involving single recombinant prostanoid receptors. Selective antagonists have been crucial to defining the roles of PGD(2) (acting on DP(1) and DP(2) receptors) and PGE(2) (on EP(1) and EP(4) receptors) in various inflammatory conditions; there are clear opportunities for therapeutic intervention. The vast endeavour on TP (thromboxane) antagonists is considered in relation to their limited pharmaceutical success in the cardiovascular area. Correspondingly, the clinical utility of IP (prostacyclin) antagonists is assessed in relation to the cloud hanging over the long-term safety of selective COX-2 inhibitors. Aspirin apart, COX inhibitors broadly suppress all prostanoid pathways, while high selectivity has been a major goal in receptor antagonist development; more targeted therapy may require an intermediate position with defined antagonist selectivity profiles. This review is intended to provide overviews of each antagonist class (including prostamide antagonists), covering major development strategies and current and potential clinical usage.
Collapse
Affiliation(s)
- R L Jones
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | | | | |
Collapse
|