1
|
Jakob F, Hennen S, Gautrois M, Khalil F, Lockhart A. Novel selective glucocorticoid receptor modulator GRM-01 demonstrates dissociation of anti-inflammatory effects from adverse effects on glucose and bone metabolism. Front Pharmacol 2025; 16:1542351. [PMID: 40110125 PMCID: PMC11920646 DOI: 10.3389/fphar.2025.1542351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction The development of selective GR agonist and modulators (SEGRAMs) aimed to minimize the adverse effects of chronic glucocorticoid treatment (e.g., hyperglycemia and osteoporosis) by separating the transactivation and transrepression activities of the glucocorticoid receptor (GR). Herein we report the pharmacologic profile of clinical candidate GRM-01, a novel, orally available, non-steroidal SEGRAM. Methods In vitro GR, progesterone receptor (PR), and mineralocorticoid receptor (MR) binding and reporter gene assays were conducted to determine GRM-01 potency and selectivity. Anti-inflammatory effects were investigated in vitro using functional assays in rat and human whole blood, human lung cells, and primary fibroblast-like synoviocytes from human donors with rheumatoid arthritis. In vitro assays measured tyrosine aminotransferase [TAT] activity in human hepatocytes and osteoprotegerin release from human osteoblasts as markers of glucose and bone metabolism, respectively. In vivo studies examined the effect of GRM-01 on biomarkers in a rat model of inflammation and on cortisol levels in Cynomolgus monkeys. Animal pharmacokinetics (PK) for GRM-01 were determined and used to predict its human PK. Results GRM-01 is a potent and selective ligand of human GR versus human PR and MR (inhibition constant = 12 vs. 3,700 and >10,000 nM, respectively). GRM-01 displayed partial induction (transactivation) at the GR (half-maximal effective concentration [EC50] = 60.2 nM, efficacy 31.8%) versus prednisolone (EC50 = 24.3 nM, efficacy 80.5%). GRM-01 demonstrated anti-inflammatory efficacy, inhibiting tumor necrosis factor-α and interferon-γ release in whole blood assays, and interleukin-6 release in cellular assays. GRM-01 weakly increased TAT activity in HepG2 cells (efficacy 14.0% vs. 92.4% with prednisolone) and partially inhibited osteoprotegerin release in MG-63 cells (by 58% vs. 100%). In vivo, GRM-01 dose-dependently reduced rat ankle swelling, had anti-nociceptive effects, and did not increase blood glucose. In Cynomolgus monkeys, GRM-01 dose-dependently reduced plasma cortisol. Animal PK found that GRM-01 had high oral bioavailability, generally low clearance, and good tissue partitioning. The predicted human total plasma clearance of GRM-01 was 0.25 mL/min/kg, volume of distribution 2.124 L/kg, and half-life ∼98 h. Conclusion GRM-01 displays a favorable preclinical pharmacologic profile consistent with a SEGRAM, and based on this is currently in Phase 1 development.
Collapse
Affiliation(s)
- Florian Jakob
- Research and Development, Grünenthal GmbH, Aachen, Germany
| | | | | | - Feras Khalil
- Research and Development, Grünenthal GmbH, Aachen, Germany
| | | |
Collapse
|
2
|
Ambery P, Zajac G, Almquist J, Prothon S, Astbury C, Brown MN, Nemes S, Nsabimana J, Edman K, Öberg L, Lepistö M, Edenro G, Dillmann I, Mitra S, Belfield G, Keen C, Heise T. The effect of AZD9567 vs. prednisolone on glycaemic control in patients with type 2 diabetes mellitus: Results from a phase 2a clinical trial. Br J Clin Pharmacol 2024; 90:1921-1931. [PMID: 38690606 DOI: 10.1111/bcp.16082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
AIMS Corticosteroids are the treatment of choice for many inflammatory diseases but often lead to adverse effects, including hyperglycaemia. This study investigated the mechanisms driving differential effects on glucose control for AZD9567, an oral nonsteroidal selective glucocorticoid receptor modulator vs. prednisolone in 46 patients with type 2 diabetes mellitus. METHODS In this randomized, double-blind, 2-way cross-over study (NCT04556760), participants received either AZD9567 72 mg and prednisolone 40 mg daily (cohort 1); AZD9567 40 mg and prednisolone 20 mg daily (cohort 2); or placebo and prednisolone 5 mg daily (cohort 3). Treatment duration was 3 days with a 3-week washout between treatment periods. Glycaemic control was assessed after a standardized meal and with continuous glucose monitoring. RESULTS A significant difference between AZD9567 and prednisolone in favour of AZD9567 was observed for the change from baseline to Day 4 glucose excursions postmeal in cohort 1 (glucose area under the curve from 0 to 4 h -4.54%; 95% confidence interval [CI]: -8.88, -0.01; P = .049), but not in cohort 2 (-5.77%; 95% CI: -20.92, 12.29; P = .435). In cohort 1, significant differences between AZD9567 and prednisolone were also seen for the change from baseline to day 4 in insulin and glucagon secretion postmeal (P < .001 and P = .005, respectively) and change from baseline to Day 4 in GLP-1 response (P = .022). Significant differences between AZD9567 and prednisolone for 24-h glucose control were observed for both cohort 1 (-1.507 mmol/L; 95% CI: -2.0820, -0.9314; P < .001) and cohort 2 (-1.110 mmol/L; 95% CI -1.7257, -0.4941; P < .001). CONCLUSION AZD9567 significantly reduced treatment-induced hyperglycaemia compared with prednisolone.
Collapse
Affiliation(s)
- Philip Ambery
- Clinical Development, Research and Late Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Grzegorz Zajac
- Clinical Development, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Warsaw, Poland
| | - Joachim Almquist
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Susanne Prothon
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Carol Astbury
- Projects Department, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Mary N Brown
- Clinical Development, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, USA
| | - Szilard Nemes
- Early Biometrics and Statistical Innovation, Statistics, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Joselyne Nsabimana
- Early Biometrics and Statistical Innovation, Statistical Programming, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karl Edman
- Mechanistic and Structural Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lisa Öberg
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Matti Lepistö
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Goran Edenro
- Respiratory, Inflammation and Autoimmunity BioScience, AstraZeneca, Gothenburg, Sweden
| | - Inken Dillmann
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Suman Mitra
- Canther, UMR9020 -CNRS-1277 -INSERM, F-59045, University de Lille, CHU de Lille, Lille, France
- IMED Respiratory, Inflammation and Autoimmunity, AstraZeneca, Gothenburg, Sweden
| | - Graham Belfield
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christina Keen
- Clinical Development, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
3
|
Chiang YTT, Kassotis CD. Molecular Assessment of Proadipogenic Effects for Common-Use Contraceptives and Their Mixtures. Endocrinology 2024; 165:bqae050. [PMID: 38648498 PMCID: PMC11081078 DOI: 10.1210/endocr/bqae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/12/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Hormonal contraceptives are widely prescribed due to their effectiveness and convenience and have become an integral part of family planning strategies worldwide. In the United States, approximately 65% of reproductive-aged women are estimated to be using contraceptive options, with approximately 33% using one or a combination of hormonal contraceptives. While these methods have undeniably contributed to improved reproductive health, recent studies have raised concerns regarding their potential effect on metabolic health. Despite widespread anecdotal reports, epidemiological research has been mixed as to whether hormonal contraceptives contribute to metabolic health effects. As such, the goals of this study were to assess the adipogenic activity of common hormonal contraceptive chemicals and their mixtures. Five different models of adipogenesis were used to provide a rigorous assessment of metabolism-disrupting effects. Interestingly, every individual contraceptive (both estrogens and progestins) and each mixture promoted significant adipogenesis (eg, triglyceride accumulation and/or preadipocyte proliferation). These effects appeared to be mediated in part through estrogen receptor signaling, particularly for the contraceptive mixtures, as cotreatment with fulvestrant acted to inhibit contraceptive-mediated proadipogenic effects on triglyceride accumulation. In conclusion, this research provides valuable insights into the complex interactions between hormonal contraceptives and adipocyte development. The results suggest that both progestins and estrogens within these contraceptives can influence adipogenesis, and the specific effects may vary based on the receptor disruption profiles. Further research is warranted to establish translation of these findings to in vivo models and to further assess causal mechanisms underlying these effects.
Collapse
Affiliation(s)
- Yu-Ting Tiffany Chiang
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
4
|
van Laar JM, Lei A, Safy‐Khan M, Almquist J, Belfield G, Edman K, Öberg L, Angermann BR, Dillmann I, Berntsson P, Etal D, Dainty I, Astbury C, Belvisi MG, Nemes S, Platt A, Prothon S, Samuelsson S, Svanberg P, Keen C. AZD9567 versus prednisolone in patients with active rheumatoid arthritis: A phase IIa, randomized, double-blind, efficacy, and safety study. Clin Transl Sci 2023; 16:2494-2506. [PMID: 37873558 PMCID: PMC10719483 DOI: 10.1111/cts.13624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 10/25/2023] Open
Abstract
Oral corticosteroid use is limited by side effects, some caused by off-target actions on the mineralocorticoid receptor that disrupt electrolyte balance. AZD9567 is a selective, nonsteroidal glucocorticoid receptor modulator. The efficacy, safety, and tolerability of AZD9567 and prednisolone were assessed in a phase IIa study. Anti-inflammatory mechanism of action was also evaluated in vitro in monocytes from healthy donors. In this randomized, double-blind, parallel-group, multicenter study, patients with active rheumatoid arthritis were randomized 1:1 to AZD9567 40 mg or prednisolone 20 mg once daily orally for 14 days. The primary end point was change from baseline in DAS28-CRP at day 15. Secondary end points included components of DAS28-CRP, American College of Rheumatology (ACR) response criteria (ACR20, ACR50, and ACR70), and safety end points, including serum electrolytes. Overall, 21 patients were randomized to AZD9567 (n = 11) or prednisolone (n = 10), and all completed the study. As anticipated, AZD9567 had a similar efficacy profile to prednisolone, with no clinically meaningful (i.e., >1.0) difference in change from baseline to day 15 in DAS28-CRP between AZD9567 and prednisolone (least-squares mean difference: 0.47, 95% confidence interval: -0.49 to 1.43). Similar results were observed for the secondary efficacy end points. In vitro transcriptomic analysis showed that anti-inflammatory responses were similar for AZD9567, prednisolone, and dexamethasone. Unlike prednisolone, AZD9567 had no effect on the serum sodium:potassium ratio. The safety profile was not different from that of prednisolone. Larger studies of longer duration are required to determine whether AZD9567 40 mg may in the future be an alternative to prednisolone in patients with inflammatory disease.
Collapse
Affiliation(s)
- Jacob M. van Laar
- Division of Internal Medicine and Dermatology, Department of Rheumatology & Clinical ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Alejhandra Lei
- Patient Safety BioPharmaceuticalsChief Medical Office, R&D, AstraZenecaBarcelonaSpain
| | - Mary Safy‐Khan
- Division of Internal Medicine and Dermatology, Department of Rheumatology & Clinical ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Joachim Almquist
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Graham Belfield
- Translational Genomics, Discovery Biology SE, Discovery SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Karl Edman
- Mechanistic and Structural Biology, Discovery SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Lisa Öberg
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Bastian R. Angermann
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Inken Dillmann
- Translational Genomics, Discovery Biology SE, Discovery SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Pia Berntsson
- Bioscience COPD/IPF, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Damla Etal
- Translational Genomics, Discovery Biology SE, Discovery SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Ian Dainty
- Bioscience COPD/IPF, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Carol Astbury
- Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Maria G. Belvisi
- Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
- Respiratory Pharmacology, National Heart and Lung InstituteImperial College LondonLondonUK
| | - Szilárd Nemes
- Early Biometrics and Statistical Innovation, Data Science & AIBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Adam Platt
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Susanne Prothon
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Sara Samuelsson
- Clinical Development, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Petter Svanberg
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Christina Keen
- Clinical Development, Research and Early Development, Respiratory & ImmunologyBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| |
Collapse
|
5
|
Pofi R, Caratti G, Ray DW, Tomlinson JW. Treating the Side Effects of Exogenous Glucocorticoids; Can We Separate the Good From the Bad? Endocr Rev 2023; 44:975-1011. [PMID: 37253115 PMCID: PMC10638606 DOI: 10.1210/endrev/bnad016] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/01/2023]
Abstract
It is estimated that 2% to 3% of the population are currently prescribed systemic or topical glucocorticoid treatment. The potent anti-inflammatory action of glucocorticoids to deliver therapeutic benefit is not in doubt. However, the side effects associated with their use, including central weight gain, hypertension, insulin resistance, type 2 diabetes (T2D), and osteoporosis, often collectively termed iatrogenic Cushing's syndrome, are associated with a significant health and economic burden. The precise cellular mechanisms underpinning the differential action of glucocorticoids to drive the desirable and undesirable effects are still not completely understood. Faced with the unmet clinical need to limit glucocorticoid-induced adverse effects alongside ensuring the preservation of anti-inflammatory actions, several strategies have been pursued. The coprescription of existing licensed drugs to treat incident adverse effects can be effective, but data examining the prevention of adverse effects are limited. Novel selective glucocorticoid receptor agonists and selective glucocorticoid receptor modulators have been designed that aim to specifically and selectively activate anti-inflammatory responses based upon their interaction with the glucocorticoid receptor. Several of these compounds are currently in clinical trials to evaluate their efficacy. More recently, strategies exploiting tissue-specific glucocorticoid metabolism through the isoforms of 11β-hydroxysteroid dehydrogenase has shown early potential, although data from clinical trials are limited. The aim of any treatment is to maximize benefit while minimizing risk, and within this review we define the adverse effect profile associated with glucocorticoid use and evaluate current and developing strategies that aim to limit side effects but preserve desirable therapeutic efficacy.
Collapse
Affiliation(s)
- Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Giorgio Caratti
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford OX37LE, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
6
|
Postel S, Wissler L, Johansson CA, Gunnarsson A, Gordon E, Collins B, Castaldo M, Köhler C, Öling D, Johansson P, Fröderberg Roth L, Beinsteiner B, Dainty I, Delaney S, Klaholz BP, Billas IML, Edman K. Quaternary glucocorticoid receptor structure highlights allosteric interdomain communication. Nat Struct Mol Biol 2023; 30:286-295. [PMID: 36747092 DOI: 10.1038/s41594-022-00914-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2023]
Abstract
The glucocorticoid receptor (GR) is a ligand-activated transcription factor that binds DNA and assembles co-regulator complexes to regulate gene transcription. GR agonists are widely prescribed to people with inflammatory and autoimmune diseases. Here we present high-resolution, multidomain structures of GR in complex with ligand, DNA and co-regulator peptide. The structures reveal how the receptor forms an asymmetric dimer on the DNA and provide a detailed view of the domain interactions within and across the two monomers. Hydrogen-deuterium exchange and DNA-binding experiments demonstrate that ligand-dependent structural changes are communicated across the different domains in the full-length receptor. This study demonstrates how GR forms a distinct architecture on DNA and how signal transmission can be modulated by the ligand pharmacophore, provides a platform to build a new level of understanding of how receptor modifications can drive disease progression and offers key insight for future drug design.
Collapse
Affiliation(s)
- Sandra Postel
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Lisa Wissler
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Carina A Johansson
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Gunnarsson
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Euan Gordon
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Barry Collins
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marie Castaldo
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian Köhler
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - David Öling
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Patrik Johansson
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Brice Beinsteiner
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, Institute of Genetics and of Molecular and Cellular Biology (IGBMC), Illkirch, France
- Université de Strasbourg, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France
- Centre National de la Recherche Scientifique (CNRS) UMR 7104, Illkirch, France
| | - Ian Dainty
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stephen Delaney
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bruno P Klaholz
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, Institute of Genetics and of Molecular and Cellular Biology (IGBMC), Illkirch, France
- Université de Strasbourg, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France
- Centre National de la Recherche Scientifique (CNRS) UMR 7104, Illkirch, France
| | - Isabelle M L Billas
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, Institute of Genetics and of Molecular and Cellular Biology (IGBMC), Illkirch, France
- Université de Strasbourg, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France
- Centre National de la Recherche Scientifique (CNRS) UMR 7104, Illkirch, France
| | - Karl Edman
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
7
|
Pang JP, Hu XP, Wang YX, Liao JN, Chai X, Wang XW, Shen C, Wang JJ, Zhang LL, Wang XY, Zhu F, Weng QJ, Xu L, Hou TJ, Li D. Discovery of a novel nonsteroidal selective glucocorticoid receptor modulator by virtual screening and bioassays. Acta Pharmacol Sin 2022; 43:2429-2438. [PMID: 35110698 PMCID: PMC8809242 DOI: 10.1038/s41401-021-00855-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/27/2021] [Indexed: 12/31/2022]
Abstract
Synthetic glucocorticoids (GCs) have been widely used in the treatment of a broad range of inflammatory diseases, but their clinic use is limited by undesired side effects such as metabolic disorders, osteoporosis, skin and muscle atrophies, mood disorders and hypothalamic-pituitary-adrenal (HPA) axis suppression. Selective glucocorticoid receptor modulators (SGRMs) are expected to have promising anti-inflammatory efficacy but with fewer side effects caused by GCs. Here, we reported HT-15, a prospective SGRM discovered by structure-based virtual screening (VS) and bioassays. HT-15 can selectively act on the NF-κB/AP1-mediated transrepression function of glucocorticoid receptor (GR) and repress the expression of pro-inflammation cytokines (i.e., IL-1β, IL-6, COX-2, and CCL-2) as effectively as dexamethasone (Dex). Compared with Dex, HT-15 shows less transactivation potency that is associated with the main adverse effects of synthetic GCs, and no cross activities with other nuclear receptors. Furthermore, HT-15 exhibits very weak inhibition on the ratio of OPG/RANKL. Therefore, it may reduce the side effects induced by normal GCs. The bioactive compound HT-15 can serve as a starting point for the development of novel therapeutics for high dose or long-term anti-inflammatory treatment.
Collapse
Affiliation(s)
- Jin-Ping Pang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xue-Ping Hu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yun-Xia Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia-Ning Liao
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xin Chai
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xu-Wen Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chao Shen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia-Jia Wang
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, 310058, China
| | - Lu-Lu Zhang
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, 310058, China
| | - Xin-Yue Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng Zhu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qin-Jie Weng
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, 310058, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China
| | - Ting-Jun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dan Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Li JX, Cummins CL. Fresh insights into glucocorticoid-induced diabetes mellitus and new therapeutic directions. Nat Rev Endocrinol 2022; 18:540-557. [PMID: 35585199 PMCID: PMC9116713 DOI: 10.1038/s41574-022-00683-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 02/08/2023]
Abstract
Glucocorticoid hormones were discovered to have use as potent anti-inflammatory and immunosuppressive therapeutics in the 1940s and their continued use and development have successfully revolutionized the management of acute and chronic inflammatory diseases. However, long-term use of glucocorticoids is severely hampered by undesirable metabolic complications, including the development of type 2 diabetes mellitus. These effects occur due to glucocorticoid receptor activation within multiple tissues, which results in inter-organ crosstalk that increases hepatic glucose production and inhibits peripheral glucose uptake. Despite the high prevalence of glucocorticoid-induced hyperglycaemia associated with their routine clinical use, treatment protocols for optimal management of the metabolic adverse effects are lacking or underutilized. The type, dose and potency of the glucocorticoid administered dictates the choice of hypoglycaemic intervention (non-insulin or insulin therapy) that should be provided to patients. The longstanding quest to identify dissociated glucocorticoid receptor agonists to separate the hyperglycaemic complications of glucocorticoids from their therapeutically beneficial anti-inflammatory effects is ongoing, with selective glucocorticoid receptor modulators in clinical testing. Promising areas of preclinical research include new mechanisms to disrupt glucocorticoid signalling in a tissue-selective manner and the identification of novel targets that can selectively dissociate the effects of glucocorticoids. These research arms share the ultimate goal of achieving the anti-inflammatory actions of glucocorticoids without the metabolic consequences.
Collapse
Affiliation(s)
- Jia-Xu Li
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Hu X, Pang J, Zhang J, Shen C, Chai X, Wang E, Chen H, Wang X, Duan M, Fu W, Xu L, Kang Y, Li D, Xia H, Hou T. Discovery of Novel GR Ligands toward Druggable GR Antagonist Conformations Identified by MD Simulations and Markov State Model Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102435. [PMID: 34825505 PMCID: PMC8787434 DOI: 10.1002/advs.202102435] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/30/2021] [Indexed: 06/13/2023]
Abstract
Binding of different ligands to glucocorticoid receptor (GR) may induce different conformational changes and even trigger completely opposite biological functions. To understand the allosteric communication within the GR ligand binding domain, the folding pathway of helix 12 (H12) induced by the binding of the agonist dexamethasone (DEX), antagonist RU486, and modulator AZD9567 are explored by molecular dynamics simulations and Markov state model analysis. The ligands can regulate the volume of the activation function-2 through the residues Phe737 and Gln738. Without ligand or with agonist binding, H12 swings from inward to outward to visit different folding positions. However, the binding of RU486 or AZD9567 perturbs the structural state, and the passive antagonist state appears more stable. Structure-based virtual screening and in vitro bioassays are used to discover novel GR ligands that bias the conformation equilibria toward the passive antagonist state. HP-19 exhibits the best anti-inflammatory activity (IC50 = 0.041 ± 0.011 µm) in nuclear factor-kappa B signaling pathway, which is comparable to that of DEX. HP-19 also does not induce adverse effect-related transactivation functions of GR. The novel ligands discovered here may serve as promising starting points for the development of GR modulators.
Collapse
Affiliation(s)
- Xueping Hu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
- State Key Lab of CAD&CGZhejiang UniversityHangzhouZhejiang310058China
| | - Jinping Pang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Jintu Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Chao Shen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xin Chai
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Ercheng Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Haiyi Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xuwen Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Mojie Duan
- Key Laboratory of magnetic Resonance in Biological SystemsState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsNational Center for Magnetic Resonance in WuhanWuhan Institute of Physics and MathematicsChinese Academy of SciencesWuhanHubei430071China
| | - Weitao Fu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Lei Xu
- Institute of Bioinformatics and Medical EngineeringSchool of Electrical and Information EngineeringJiangsu University of TechnologyChangzhou213001China
| | - Yu Kang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Dan Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Hongguang Xia
- Department of Biochemistry and Research Center of Clinical Pharmacy of The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
- State Key Lab of CAD&CGZhejiang UniversityHangzhouZhejiang310058China
| |
Collapse
|
10
|
La Sala G, Gunnarsson A, Edman K, Tyrchan C, Hogner A, Frolov AI. Unraveling the Allosteric Cross-Talk between the Coactivator Peptide and the Ligand-Binding Site in the Glucocorticoid Receptor. J Chem Inf Model 2021; 61:3667-3680. [PMID: 34156843 DOI: 10.1021/acs.jcim.1c00323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The glucocorticoid receptor (GR) is a nuclear receptor that controls critical biological processes by regulating the transcription of specific genes. There is a known allosteric cross-talk between the ligand and coregulator binding sites within the GR ligand-binding domain that is crucial for the control of the functional response. However, the molecular mechanisms underlying such an allosteric control remain elusive. Here, molecular dynamics (MD) simulations, bioinformatic analysis, and biophysical measurements are integrated to capture the structural and dynamic features of the allosteric cross-talk within the GR. We identified a network of evolutionarily conserved residues that enables the allosteric signal transduction, in agreement with experimental data. MD simulations clarify how such a network is dynamically interconnected and offer a mechanistic explanation of how different peptides affect the intensity of the allosteric signal. This study provides useful insights to elucidate the GR allosteric regulation, ultimately providing a foundation for designing novel drugs.
Collapse
Affiliation(s)
- Giuseppina La Sala
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Gunnarsson
- Discovery Science, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karl Edman
- Discovery Science, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian Tyrchan
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Hogner
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrey I Frolov
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
11
|
Kurimoto T, Tamai I, Nakagawa T, Miyai A, Yamamoto Y, Kosugi Y, Deai K, Hata T, Ohta T, Matsushita M, Yamada T. JTP-117968, a novel selective glucocorticoid receptor modulator, exhibits significant anti-inflammatory effect while maintaining bone mineral density in mice. Eur J Pharmacol 2021; 895:173880. [PMID: 33476654 DOI: 10.1016/j.ejphar.2021.173880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/25/2020] [Accepted: 01/12/2021] [Indexed: 01/10/2023]
Abstract
Classic glucocorticoids have been prescribed for various inflammatory diseases, such as rheumatoid arthritis, due to their outstanding anti-inflammatory effects. However, glucocorticoids cause numerous unwanted side effects, including osteoporosis and diabetes. Hence, selective glucocorticoid receptor modulators (SGRMs), which retain anti-inflammatory effects with minimized side effects, are among the most anticipated drugs in the clinical field. The assumption is that there are two major mechanisms of action via glucocorticoid receptors, transrepression (TR) and transactivation (TA). In general, anti-inflammatory effects of glucocorticoids are largely due to TR, while the side effects associated with glucocorticoids are mostly mediated through TA. We previously reported that JTP-117968, a novel SGRM, maintained partial TR activity while remarkably reducing the TA activity. In this study, we investigated the anti-inflammatory effect of JTP-117968 on a lipopolysaccharide (LPS) challenge model and collagen-induced arthritis (CIA) model in mice. Meanwhile, we tested the effect of JTP-117968 on the bone mineral density (BMD) in mouse femur to evaluate the side effect. Based on the evaluation, JTP-117968 reduced the plasma levels of tumor necrosis factor α induced by LPS challenge in mice significantly. Remarkably, CIA development was suppressed by JTP-117968 comparably with prednisolone and PF-802, an active form of fosdagrocorat that has been developed clinically as an orally available SGRM. Strikingly, the side effect of JTP-117968 on mouse femoral BMD was much lower than those of PF-802 and prednisolone. Therefore, JTP-117968 has attractive potential as a new therapeutic option against inflammatory diseases with minimized side effects compared to classic glucocorticoids.
Collapse
Affiliation(s)
- Takafumi Kurimoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan; Faculty of Agriculture, Department of Agrobiology, Niigata University, Niigata, Japan.
| | - Isao Tamai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Takashi Nakagawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Atsuko Miyai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Yasuo Yamamoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Yoshinori Kosugi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Katsuya Deai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Takahiro Hata
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | | | - Takahisa Yamada
- Faculty of Agriculture, Department of Agrobiology, Niigata University, Niigata, Japan
| |
Collapse
|
12
|
Almquist J, Sadiq MW, Eriksson UG, Hegelund Myrbäck T, Prothon S, Leander J. Estimation of Equipotent Doses for Anti-Inflammatory Effects of Prednisolone and AZD9567, an Oral Selective Nonsteroidal Glucocorticoid Receptor Modulator. CPT Pharmacometrics Syst Pharmacol 2020; 9:444-455. [PMID: 32501650 PMCID: PMC7438818 DOI: 10.1002/psp4.12536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/05/2020] [Indexed: 01/08/2023] Open
Abstract
AZD9567 is a potent and selective nonsteroidal oral glucocorticoid receptor modulator. It is developed as an anti-inflammatory drug with improved safety profile compared with steroids like prednisolone. Throughout the clinical development of AZD9567, dose selection and data interpretation require a method for determining doses with the same anti-inflammatory effect as prednisolone. Equipotent doses of AZD9567 and prednisolone were defined by the same average inhibition of TNFα release, a biomarker of anti-inflammatory effect, measured in a lipopolysaccharide-stimulated whole blood ex vivo assay. Based on pharmacokinetic-pharmacodynamic models, TNFα dose-response relationships for AZD9567 and prednisolone were established. A comparison of the dose-response curves enabled estimation of an equipotency relationship. Specifically, 20 mg prednisolone was estimated to be equipotent to 40 mg AZD9567 (95% confidence interval: 29-54 mg). Static concentration-response analyses showed that the relative potencies for inhibition of TNFα release of AZD9567 and prednisolone were well aligned with several other pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Joachim Almquist
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Muhammad Waqas Sadiq
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Ulf G. Eriksson
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Tove Hegelund Myrbäck
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Susanne Prothon
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Jacob Leander
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| |
Collapse
|
13
|
Köhler C, Carlström G, Gunnarsson A, Weininger U, Tångefjord S, Ullah V, Lepistö M, Karlsson U, Papavoine T, Edman K, Akke M. Dynamic allosteric communication pathway directing differential activation of the glucocorticoid receptor. SCIENCE ADVANCES 2020; 6:eabb5277. [PMID: 32832645 PMCID: PMC7439413 DOI: 10.1126/sciadv.abb5277] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/05/2020] [Indexed: 05/07/2023]
Abstract
Allosteric communication within proteins is a hallmark of biochemical signaling, but the dynamic transmission pathways remain poorly characterized. We combined NMR spectroscopy and surface plasmon resonance to reveal these pathways and quantify their energetics in the glucocorticoid receptor, a transcriptional regulator controlling development, metabolism, and immune response. Our results delineate a dynamic communication network of residues linking the ligand-binding pocket to the activation function-2 interface, where helix 12, a switch for transcriptional activation, exhibits ligand- and coregulator-dependent dynamics coupled to graded activation. The allosteric free energy responds to variations in ligand structure: subtle changes gradually tune allostery while preserving the transmission pathway, whereas substitution of the entire pharmacophore leads to divergent allosteric control by apparently rewiring the communication network. Our results provide key insights that should aid in the design of mechanistically differentiated ligands.
Collapse
Affiliation(s)
- C. Köhler
- Respiratory, Inflammation and Autoimmunity, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - G. Carlström
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P.O. Box 124, 221 00 Lund, Sweden
| | - A. Gunnarsson
- Discovery Sciences, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - U. Weininger
- Division of Biophysical Chemistry, Center for Molecular Protein Science, Department of Chemistry, Lund University, P. O. Box 124, 221 00 Lund, Sweden
| | - S. Tångefjord
- Respiratory, Inflammation and Autoimmunity, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Discovery Sciences, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - V. Ullah
- Respiratory, Inflammation and Autoimmunity, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - M. Lepistö
- Respiratory, Inflammation and Autoimmunity, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - U. Karlsson
- Discovery Sciences, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - T. Papavoine
- Respiratory, Inflammation and Autoimmunity, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - K. Edman
- Discovery Sciences, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - M. Akke
- Division of Biophysical Chemistry, Center for Molecular Protein Science, Department of Chemistry, Lund University, P. O. Box 124, 221 00 Lund, Sweden
| |
Collapse
|