1
|
Saini DK, Devaramani MS, Shanmugavel H, Tabassum SZ, Mudnakudu-Nagaraju KK, Siddesha JM, Shetty R. Development and validation of a recombinant human TNF-α based ELISA to detect and quantify adalimumab. Biochem Biophys Rep 2025; 42:102005. [PMID: 40248135 PMCID: PMC12005340 DOI: 10.1016/j.bbrep.2025.102005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
Adalimumab, a humanized IgG1 monoclonal antibody is currently used to treat inflammatory diseases. However, a sensitive, in-house ELISA for evaluating inter- and intra-individual pharmacokinetic variability of adalimumab remains limited. In this study, an ELISA was developed to measure adalimumab levels, using recombinant human TNF-α (rhTNF-α) as capture antibody. Initially, surface plasma resonance showed acceptable binding kinetics (KD) of 2.38x10-07 nM for adalimumab. Next, a standard curve of adalimumab (1.54 ng/ml to 300 ng/ml), with five quality control points (5.2, 16, 27, 150, and 200 ng/ml) was evaluated for inter and intra-assay accuracy and precision, using serum matrix, by four independent validations. The linear range of the validated assay was 5.2 ng/ml to 200 ng/ml, upper limit of quantification (ULOQ) and lower limit of quantification (LLOQ) were 200 ng/ml and 5.2 ng/ml, respectively. The assay specificity was validated by testing cross-reactivity of rituximab with rhTNF-α, which was found to be non-reactive. Further, the hook effect was over-ruled by diluting the highest concentration of adalimumab tested to assay linear range, and dilution integrity was observed for entire concentrations within linear range (%RE ≤ 20 %), as recommended by European Medicines Agency. Collectively, this rhTNF-α binding-based ELISA method is highly sensitive, reproducible, and useful for monitoring adalimumab.
Collapse
Affiliation(s)
- Dinesh Kumar Saini
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
- Denovo Biolabs Pvt Ltd., Electronic City, Phase I, Bengaluru, 560100, India
| | - Manjunath S. Devaramani
- Denovo Biolabs Pvt Ltd., Electronic City, Phase I, Bengaluru, 560100, India
- Department of Biotechnology & Bioinformatics, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | | | | | - Kiran Kumar Mudnakudu-Nagaraju
- Department of Biotechnology & Bioinformatics, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Jalahalli Mariswamy Siddesha
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Radhakrishna Shetty
- National Food Institute Research Group for Chemical Risk Assessment and GMO Research Group for Microbial Biotechnology and Biorefining, Henrik Dams Allé, 2800, Kgs. Lyngby, Denmark
| |
Collapse
|
2
|
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
|
3
|
Fayyaz S, Lotke R, Haußmann I, Petersen M, Müller E, Schwarzer-Sperber HS, Schwarzer R, Sauter D. Reactivation of latent HIV-1 by the glucocorticoid receptor modulator AZD9567. J Virol 2025; 99:e0188624. [PMID: 39817774 PMCID: PMC11853017 DOI: 10.1128/jvi.01886-24] [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: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
One key determinant of HIV-1 latency reversal is the activation of the viral long terminal repeat (LTR) by cellular transcription factors such as NF-κB and AP-1. Interestingly, the activity of these two transcription factors can be modulated by glucocorticoid receptors (GRs). Furthermore, the HIV-1 genome contains multiple binding sites for GRs. We therefore hypothesized that glucocorticoids and other GR modulators may influence HIV-1 latency and reactivation. To investigate how GR signaling affects latent HIV-1 reservoirs, we assembled a representative panel of GR modulators including natural steroidal agonists, selective and non-selective GR modulators, and clinically approved GR-modulating drugs. The effects of these compounds on HIV-1 reactivation were assessed using latently HIV-1-infected cell lines and primary cells, as well as reporter assays that monitored GR and LTR activities. We found that AZD9567 (Mizacorat), a non-steroidal partial GR agonist, reactivates latent HIV-1 in both lymphoid and myeloid cell lines and primary CD4+ T cells. Conversely, the GR antagonist mifepristone suppresses HIV-1 LTR-driven gene expression. Mechanistic analyses revealed that AZD9567-mediated reactivation partially depends on both GR and AP-1 binding sites in the LTR. In summary, we, here, identify the GR modulator AZD9567 as novel latency-reversing agent that activates LTR-driven gene expression, which may aid in advancing current shock-and-kill approaches in the treatment of HIV-1 infection.IMPORTANCELatently infected cells of people living with HIV are constantly exposed to fluctuating levels of glucocorticoid hormones such as cortisol. In addition, many HIV-infected individuals regularly take corticosteroids as anti-inflammatory drugs. Although corticosteroids are known to affect the activity of the viral long terminal repeat (LTR) promoter and influence ongoing HIV-1 replication, relatively little is known about the effect of corticosteroid hormones and other glucocorticoid receptor (GR) modulators on latent HIV-1. By systematically comparing natural and synthetic GR modulators, we, here, identify a first first-in-class, oral, partial GR agonist that reactivates latent HIV-1 from different cell types. This drug, AZD9567, was previously tested in clinical trials for rheumatoid arthritis. Mutational analyses shed light on the underlying mode of action and revealed transcription factor binding sites in the HIV-1 LTR that determine responsiveness to AZD9567.
Collapse
Affiliation(s)
- Sharmeen Fayyaz
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
- National Institute of Virology, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rishikesh Lotke
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Isabell Haußmann
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Moritz Petersen
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Eva Müller
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, Essen, Germany
| | | | - Roland Schwarzer
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, Essen, Germany
| | - Daniel Sauter
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Thangadurai M, Sethuraman S, Subramanian A. Drug Delivery Approaches for Rheumatoid Arthritis: Recent Advances and Clinical Translation Aspects. Crit Rev Ther Drug Carrier Syst 2025; 42:1-54. [PMID: 40084516 DOI: 10.1615/critrevtherdrugcarriersyst.v42.i3.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease characterized with symmetrical progression of joint deformity that is often diagnosed at a chronic condition with other associated pathological conditions such as pericarditis, keratitis, pulmonary granuloma. Despite the understanding of RA pathophysiology in disease progression, current clinical treatment options such as disease-modifying anti-rheumatic drugs (DMARDs), biologics, steroids, and non-steroidal anti-inflammatory drugs (NSAIDs) provide only palliative therapy while causing adverse side effects such as off-target multi-organ toxicity and risk of infections. Further, available drug delivery strategies to treat RA pathogenicity does not successfully reach the site of action due to various barriers such as phagocytosis and first pass effect in addition to the disease complexity and unknown etiology, thereby leading to the development of irreversible joint dysfunction. Therefore, novel and effective strategies remain an unmet need to control the disease progression and to maintain the balance between pro- and anti-inflammatory cytokines. This review provides a comprehensive outlook on the RA pathophysiology and its corresponding disease progression. Contributions of synoviocytes such as macrophages, fibroblast-like cells in increasing invasiveness to exacerbate joint damage is also outlined in this review, which could be a potential future therapeutic target to complement the existing treatment regimens in controlling RA pathogenesis. Further, various smart drug delivery approaches under research to achieve maximum therapeutic efficacy with minimal adverse side effects have been discussed, which in turn emphasize the unmet challenges and future perspectives in addressing RA complications.
Collapse
Affiliation(s)
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
5
|
Jaumotte JD, El Khoury N, Min CK, Wang J, Madigan C, Jano A, Russo Kobylski RJ, Solt LA, Dhavan RS, Short KL, Lei T, Chandran U, Cole TJ, Monaghan-Nichols AP, Sampath V, Houtman R, Nettles KW, DeFranco DB. Physiologic and structural characterization of desisobutyryl-ciclesonide, a selective glucocorticoid receptor modulator in newborn rats. PNAS NEXUS 2025; 4:pgae573. [PMID: 39781096 PMCID: PMC11707230 DOI: 10.1093/pnasnexus/pgae573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
Bronchopulmonary dysplasia, the most prevalent chronic lung disease of prematurity, is often treated with glucocorticoids (GCs) such as dexamethasone (DEX), but their use is encumbered with several adverse somatic, metabolic, and neurologic effects. We previously reported that systemic delivery of the GC prodrug ciclesonide (CIC) in neonatal rats activated glucocorticoid receptor (GR) transcriptional responses in lung but did not trigger multiple adverse effects caused by DEX. To determine whether limited systemic metabolism of CIC was solely responsible for its enhanced safety profile, we treated neonatal rats with its active metabolite desisobutyryl-ciclesonide (Des-CIC). DEX but not Des-CIC caused a reduction in body weight as well as reduced insulin-like growth factor-1 serum levels and chronic hyperglycemia in neonatal rats. However, Des-CIC was as effective as DEX in reducing the expression of various bleomycin-induced proinflammatory cytokine mRNAs. In vitro studies with various cell types demonstrate the potent GR transactivation and transrepression activity of Des-CIC, although genome-wide transcriptomic analyses reveal differences in DEX vs. Des-CIC responses in neonatal rat lung and liver tissue. Des-CIC is a GR super-agonist as revealed by an in vitro coregulator peptide binding assay. In addition, molecular dynamics simulations revealed unique Des-CIC-dependent allosteric signaling pathways between specific residues in the GR ligand-binding domain and receptor surfaces interacting with coregulator peptides. Thus, Des-CIC is a potential novel selective GR modulator that could impart a favorable therapeutic index for CIC use for even modest durations of GC exposure which could have long-lasting adverse somatic, metabolic, or neurologic effects.
Collapse
Affiliation(s)
- Juliann D Jaumotte
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| | - Nathalie El Khoury
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| | - Charles K Min
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiefei Wang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Caroline Madigan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| | - Antalya Jano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| | - Robin J Russo Kobylski
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Laura A Solt
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rutu S Dhavan
- Department of Biochemistry and Molecular Biology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Kelly L Short
- Department of Biochemistry and Molecular Biology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Tianhua Lei
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Uma Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Timothy J Cole
- Department of Biochemistry and Molecular Biology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Ann Paula Monaghan-Nichols
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Venkatesh Sampath
- Department of Pediatrics/Division of Neonatology, Children's Mercy, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - René Houtman
- Department of Research and Development, Precision Medicine Lab, Oss 5349, The Netherlands
| | - Kendall W Nettles
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Donald B DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| |
Collapse
|
6
|
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
|
7
|
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
|