1
|
Huang X, Hong X, Yang S, Ye L, Yang X, Cui C, Wu Q, Wang W, Huang J, Yang G. Tolerability, safety, and pharmacokinetics of GR1603 injection in healthy subjects: a randomized, double-blind, placebo-controlled single-dose escalation clinical trial. Expert Opin Investig Drugs 2024:1-7. [PMID: 39681526 DOI: 10.1080/13543784.2024.2443756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/02/2024] [Accepted: 12/14/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND GR1603 is a monoclonal antibody targeting the type I interferon receptor. The aim of this study was to evaluate the safety, tolerability, pharmacokinetics, immunogenicity and pharmacodynamics of GR1603 in healthy volunteers. METHODS Healthy adults (≥18 years old) were enrolled in a placebo control, dose-escalation Phase I clinical trial receiving a single injectable dose of GR1603. Follow-up was 12 weeks. Adverse event (AE) profiles, vital signs, and blood samples were collected for assessment of safety, PK, and expression of type I interferon inducible genes. RESULTS Of the 46 subjects, 44 completed treatment. In the experimental group of 34 subjects (mean age 26.6 years), 30 experienced treatment-emergent adverse events (TEAEs), with a total of 102 occurrences, resulting in an incidence rate of 88.2%. The most commonly reported drug-related AEs were upper respiratory tract infection (17.6%), all of which were ≤ grade 2. GR1603 exhibits non-linear PK in the dose range of 0.1 mg/kg to 9 mg/kg. All samples were negative for anti-drug antibodies before and after dosing. The degrees of IFN gene signature were significantly inhibited in the higher dose groups. CONCLUSION The safety/tolerability, PK and exploratory metrics observed in this study support further clinical development of GR1603. CLINICAL TRIAL REGISTRATION www.chictr.org.cn/searchproj.html identifier is ChiCTR2100045628.
Collapse
Affiliation(s)
- Xin Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Hong
- Center for Experimental Medicine, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuang Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Ye
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoyan Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chang Cui
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Wu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- BioPharmaceuticals R&D, Chongqing Genrix Biopharmaceutical Co. Ltd, Chongqing, China
| | - Jie Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
2
|
Martín-Torregrosa D, Mansilla-Polo M, Morgado-Carrasco D. [Translated article] Use of Anifrolumab in Systemic Lupus Erythematosus, Cutaneous Lupus Erythematosus, and Other Autoimmune Dermatoses. ACTAS DERMO-SIFILIOGRAFICAS 2024:S0001-7310(24)00765-8. [PMID: 39389344 DOI: 10.1016/j.ad.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 10/12/2024] Open
Abstract
Anifrolumab is an inhibitor of the type I interferon receptor subunit 1 (IFNAR1) recently approved for the management of moderate-to-severe systemic lupus erythematosus (SLE). In 2 clinical trials, it has proven effective to treat cutaneous signs. Although anifrolumab has not been indicated for cutaneous lupus erythematosus (CLE), multiple cases and case series (20 publications with a total of 78 patients) have shown good and rapid responses with this drug, both in subacute CLE and discoid lupus erythematosus, as well as in lupus panniculitis and perniosis. Two case reports of dermatomyositis have also experienced clinical improvement with anifrolumab. Clinical trials of this drug are ongoing for subacute CLE and discoid lupus erythematosus, systemic sclerosis, and progressive vitiligo. Its most common adverse effects are respiratory infections and herpes zoster. Anifrolumab may be a well-tolerated alternative in the management of CLE.
Collapse
Affiliation(s)
- D Martín-Torregrosa
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, Spain
| | - M Mansilla-Polo
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, Spain
| | - D Morgado-Carrasco
- Servicio de Dermatología, Hospital de Figueres, Fundació Salut Empordà, Girona, Spain; Servicio de Dermatología, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Sharma P, Boulton DW, Bertagnolli LN, Tang W. Physiology-based pharmacokinetic model with relative transcriptomics to evaluate tissue distribution and receptor occupancy of anifrolumab. CPT Pharmacometrics Syst Pharmacol 2024. [PMID: 39360565 DOI: 10.1002/psp4.13245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/16/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Type I interferons contribute to the pathogenesis of several autoimmune disorders, including systemic lupus erythematosus (SLE), systemic sclerosis, cutaneous lupus erythematosus, and myositis. Anifrolumab is a monoclonal antibody that binds to subunit 1 of the type I interferon receptor (IFNAR1). Results of phase IIb and phase III trials led to the approval of intravenous anifrolumab 300 mg every 4 weeks (Q4W) alongside standard therapy in patients with moderate-to-severe SLE. Here, we built a population physiology-based pharmacokinetic (PBPK) model of anifrolumab by utilizing the physiochemical properties of anifrolumab, binding kinetics to the Fc gamma neonatal receptor, and target-mediated drug disposition properties. A novel relative transcriptomics approach was employed to determine IFNAR1 expression in tissues (blood, skin, gastrointestinal tract, lungs, and muscle) using mRNA abundances from bioinformatic databases. The IFNAR1 expression and PBPK model were validated by testing their ability to predict clinical pharmacokinetics over a large dose range from different clinical scenarios after subcutaneous and intravenous anifrolumab dosing. The validated PBPK model predicted high unbound local concentrations of anifrolumab in blood, skin, gastrointestinal tract, lungs, and muscle, which exceeded its IFNAR1 dissociation equilibrium constant values. The model also predicted high IFNAR1 occupancy with subcutaneous and intravenous anifrolumab dosing. The model predicted more sustained IFNAR1 occupancy ≥90% with subcutaneous anifrolumab 120 mg once-weekly dosing vs. intravenous 300 mg Q4W dosing. The results informed the dosing of phase III studies of anifrolumab in new indications and present a novel approach to PBPK modeling coupled with relative transcriptomics in simulating pharmacokinetics of therapeutic monoclonal antibodies.
Collapse
Affiliation(s)
- Pradeep Sharma
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - David W Boulton
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Lynn N Bertagnolli
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Weifeng Tang
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| |
Collapse
|
4
|
Martín-Torregrosa D, Mansilla-Polo M, Morgado-Carrasco D. Use of Anifrolumab in Systemic Lupus Erythematosus, Cutaneous Lupus Erythematosus, and Other Autoimmune Dermatoses. ACTAS DERMO-SIFILIOGRAFICAS 2024:S0001-7310(24)00533-7. [PMID: 38972582 DOI: 10.1016/j.ad.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 07/09/2024] Open
Abstract
Anifrolumab is an inhibitor of the type I interferon receptor subunit 1 (IFNAR1) recently approved for the management of moderate-to-severe systemic lupus erythematosus (SLE). In 2 clinical trials, it has proven effective to treat cutaneous signs. Although anifrolumab has not been indicated for cutaneous lupus erythematosus (CLE), multiple cases and case series (20 publications with a total of 78 patients) have shown good and rapid responses with this drug, both in subacute CLE and discoid lupus erythematosus, as well as in lupus panniculitis and perniosis. Two case reports of dermatomyositis have also experienced clinical improvement with anifrolumab. Clinical trials of this drug are ongoing for subacute CLE and discoid lupus erythematosus, systemic sclerosis, and progressive vitiligo. Its most common adverse effects are respiratory infections and herpes zoster. Anifrolumab may be a well-tolerated alternative in the management of CLE.
Collapse
Affiliation(s)
- D Martín-Torregrosa
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, España
| | - M Mansilla-Polo
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, España
| | - D Morgado-Carrasco
- Servicio de Dermatología, Hospital de Figueres, Fundació Salut Empordà, Girona, España; Servicio de Dermatología, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, España.
| |
Collapse
|
5
|
Li X, Li B, Wang M, Fang M, Lou J, Liu J, Chen H, Ding Y. Safety, Tolerability, Pharmacokinetics, and Immunogenicity of the Anti-IFNAR1 Monoclonal Antibody QX006N: A First-in-Human Single Ascending Dose Study in Healthy Chinese Volunteers. BioDrugs 2024; 38:313-321. [PMID: 38148466 DOI: 10.1007/s40259-023-00637-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND AND OBJECTIVE QX006N is a novel, humanized, IgG4κ monoclonal antibody targeting IFNAR1, developed for the treatment of systemic lupus erythematosus. This study aims to investigate the pharmacokinetics, safety, tolerability, and immunogenicity of QX006N when administered intravenously to healthy Chinese individuals. METHODS A double-blind, randomized, placebo-controlled, single-ascending-dose, phase I clinical trial was conducted comprising five cohorts (n = 10 per cohort, except n = 5 for the first cohort). Subjects in each cohort were randomly assigned in a 4:1 ratio to receive a single intravenous infusion of QX006N (0.3 mg/kg, 1.0 mg/kg, 3.0 mg/kg, 6.0 mg/kg, or 10.0 mg/kg) or placebo for 30 minutes. Tolerability assessments included adverse events, vital signs, 12-lead electrocardiogram, physical examination, and clinical laboratory tests. The serum concentration of QX006N was measured using the enzyme-linked immunosorbent assay method, and the anti-drug antibodies were detected using the electrochemiluminescence assay method. RESULTS QX006N demonstrated a favorable safety and tolerability profile throughout the study. All treatment-emergent adverse events were of Grade 1-2 (CTCAE Version 5.0), and no serious adverse events, deaths, or drug discontinuations because of treatment-emergent adverse events were observed. All drug-related treatment-emergent adverse events showed no clear dose-related trends. Following an intravenous infusion of QX006N at doses that ranged from 0.3 mg/kg to 10 mg/kg, the half-life increased from 24.7 to 208 hours in a dose-dependent manner, while clearance decreased from 0.0828 to 0.0065 L/h. The maximum concentration exhibited nearly dose-proportional increases, and the area under the curve displayed a more than dose-proportional increment with non-linear pharmacokinetic characteristics. The incidence of anti-drug antibodies was observed to increase over time for doses that ranged from 1.0 mg/kg to 10.0 mg/kg of QX006N, reaching its peak at day 57 (range 62.50-87.50%). Conversely, the incidence of anti-drug antibodies in the QX006N 0.3-mg/kg and placebo cohorts remained low. CONCLUSIONS QX006N demonstrated acceptable safety, tolerability, and pharmacokinetic characteristics in healthy subjects when administered as a single intravenous infusion at doses that ranged from 0.3 mg/kg to 10.0 mg/kg. Based on the pharmacokinetic and safety outcomes, a recommended effective dose of 300 mg is proposed for future phase Ib studies. CLINICAL TRIAL REGISTRATION This study has been registered at http://www.chinadrugtrials.org.cn/ under identifier CTR20212834.
Collapse
Affiliation(s)
- Xiaojiao Li
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Bing Li
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Meng Wang
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Min Fang
- Qyuns Therapeutics Co., Ltd., Jiangsu, China
| | - Jinfeng Lou
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Jingrui Liu
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Hong Chen
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Yanhua Ding
- Phase I Clinical Trial Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China.
| |
Collapse
|
6
|
Gensous N, Lazaro E, Blanco P, Richez C. Anifrolumab: first biologic approved in the EU not restricted to patients with a high degree of disease activity for the treatment of moderate to severe systemic lupus erythematosus. Expert Rev Clin Immunol 2024; 20:21-30. [PMID: 37800604 DOI: 10.1080/1744666x.2023.2268284] [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: 06/08/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Type 1 interferons (IFNs) play a crucial role in the pathogenesis of systemic lupus erythematosus (SLE) and various type I IFNs targeting therapeutic approaches have been developed. Anifrolumab, a monoclonal antibody that binds to the subunit 1 of the type I IFN receptor, has acquired considerable interest and has entered different clinical human trials willing to evaluate its efficacy and safety. AREAS COVERED This review summarizes the data obtained in phases 1, 2, and 3 clinical trials of anifrolumab for SLE patients. A focus is made on data of clinical efficacy and safety obtained in MUSE, TULIP-1 and TULIP-2 trials. EXPERT OPINION/COMMENTARY Anifrolumab is a promising therapeutic option for patients with SLE, currently authorized for moderate-to-severe SLE. Extensive real-world use is now going to generate data required to gain experience on the type of patients who benefit the most from the drug, and the exact positioning of anifrolumab in the therapeutic plan.
Collapse
Affiliation(s)
- Noémie Gensous
- Department of Internal Medicine and Clinical Immunology, CHU Bordeaux, Hôpital Saint-André, Bordeaux, France
- UMR/CNRS 5164, ImmunoConcEpT, CNRS, University of Bordeaux, Bordeaux, France
| | - Estibaliz Lazaro
- UMR/CNRS 5164, ImmunoConcEpT, CNRS, University of Bordeaux, Bordeaux, France
- Department of Internal Medicine and Infectious Diseases, Centre National de Référence des Maladies Auto-immunes Systémiques Rares RESO, CHU Bordeaux, Hôpital Haut Leveque, Pessac, France
| | - Patrick Blanco
- UMR/CNRS 5164, ImmunoConcEpT, CNRS, University of Bordeaux, Bordeaux, France
- Department of Immunology and Immunogenetics, CHU Bordeaux, Hôpital Pellegrin, Bordeaux, France
| | - Christophe Richez
- UMR/CNRS 5164, ImmunoConcEpT, CNRS, University of Bordeaux, Bordeaux, France
- Department of Rheumatology, Centre National de Référence des Maladies Auto-immunes Systémiques Rares RESO, CHU de Bordeaux, Hôpital Pellegrin, Bordeaux, France
| |
Collapse
|
7
|
Tang W, Tummala R, Almquist J, Hwang M, White WI, Boulton DW, MacDonald A. Clinical Pharmacokinetics, Pharmacodynamics, and Immunogenicity of Anifrolumab. Clin Pharmacokinet 2023; 62:655-671. [PMID: 37148484 PMCID: PMC10182164 DOI: 10.1007/s40262-023-01238-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2023] [Indexed: 05/08/2023]
Abstract
The type I interferon (IFN) signaling pathway is implicated in the pathogenesis of systemic lupus erythematosus (SLE). Anifrolumab is a monoclonal antibody that targets the type I IFN receptor subunit 1. Anifrolumab is approved in several countries for patients with moderate to severe SLE receiving standard therapy. The approved dosing regimen of anifrolumab is a 300-mg dose administered intravenously every 4 weeks; this was initially based on the results of the Phase 2b MUSE and further confirmed in the Phase 3 TULIP-1 and TULIP-2 trials, in which anifrolumab 300-mg treatment was associated with clinically meaningful improvements in disease activity with an acceptable safety profile. There have been several published analyses of the pharmacokinetic and pharmacodynamic profile of anifrolumab, including a population-pharmacokinetic analysis of 5 clinical studies of healthy volunteers and patients with SLE, in which body weight and type I IFN gene expression were significant covariates identified for anifrolumab exposure and clearance. Additionally, the pooled Phase 3 SLE population has been used to evaluate how serum exposure may be related to clinical responses, safety risks, and pharmacodynamic effects of the 21-gene type I IFN gene signature (21-IFNGS). The relevance of 21-IFNGS with regard to clinical efficacy outcomes has also been analyzed. Herein, the clinical pharmacokinetics, pharmacodynamics, and immunogenicity of anifrolumab as well as results of population-pharmacokinetics and exposure-response analyses are reviewed.
Collapse
Affiliation(s)
- Weifeng Tang
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Raj Tummala
- Clinical Development, Late Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Joachim Almquist
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Hwang
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Wendy I White
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - David W Boulton
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Alexander MacDonald
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
8
|
Ahmed AA, Osman N, Furie R. An evaluation of anifrolumab for use in adults with systemic lupus erythematosus. Expert Rev Clin Immunol 2022; 18:1095-1106. [PMID: 36083692 DOI: 10.1080/1744666x.2022.2123793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Type 1 interferons play a key role in the pathogenesis of systemic lupus erythematosus (SLE). An important clinical question is whether inhibiting the type 1 interferon pathway reduce the disease activity in SLE patients. This review evaluates the safety and efficacy of the monoclonal antibody against the type 1 interferon alpha receptor, anifrolumab, in patients with SLE. AREAS COVERED Key terms (SLE, type 1 interferon, anifrolumab) were used to query the PubMed database for phase 1, 2 and 3 clinical trials of anifrolumab for SLE patients. Phase 1 studies showed anifrolumab has non-linear pharmacokinetics and the optimal safe dose is 300 mg given intravenously every four weeks. The MUSE (phase 2) and the TULIP-2 (phase 3) trials showed that anifrolumab when added to standard therapy significantly reduced disease activity in SLE patients. Common adverse events associated with anifrolumab were upper respiratory and urinary infections as well as shingles. EXPERT OPINION Anifrolumab is an exciting new therapeutic for SLE patients. Additional analyses of the combined TULIP-1 and TULIP-2 datasets as well as future studies with anifrolumab will generate yet more data in SLE. No doubt anifrolumab will be studied in other diseases where type I interferons play an important role.
Collapse
Affiliation(s)
- Abdullah Ali Ahmed
- Rheumatology, Stony Brook University The State University of New York101 Nicolls Road, Stony Brook, New York 11794-0701, United States
| | - Naureen Osman
- Rheumatology, Northwell Health865 Northern Boulevard, Great Neck, New York 11021, United States
| | - Richard Furie
- Rheumatology, Northwell Health865 Northern Boulevard, Great Neck, New York 11021, United States
| |
Collapse
|
9
|
Almquist J, Kuruvilla D, Mai T, Tummala R, White WI, Tang W, Roskos L, Chia YL. Nonlinear Population Pharmacokinetics of Anifrolumab in Healthy Volunteers and Patients With Systemic Lupus Erythematosus. J Clin Pharmacol 2022; 62:1106-1120. [PMID: 35383948 PMCID: PMC9540432 DOI: 10.1002/jcph.2055] [Citation(s) in RCA: 4] [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: 12/07/2021] [Accepted: 03/27/2022] [Indexed: 12/03/2022]
Abstract
We characterized the population pharmacokinetics of anifrolumab, a type I interferon receptor-blocking antibody. Pharmacokinetic data were analyzed from the anifrolumab (intravenous [IV], every 4 weeks) arms from 5 clinical trials in patients with systemic lupus erythematosus (SLE) (n = 664) and healthy volunteers (n = 6). Population pharmacokinetic modeling was performed using a 2-compartment model with parallel linear and nonlinear elimination pathways. The impact of covariates (demographics, interferon gene signature [IFNGS, high/low], disease characteristics, renal/hepatic function, SLE medications, and antidrug antibodies) on pharmacokinetics was evaluated. Time-varying clearance (CL) was characterized using an empirical sigmoidal time-dependent function. Anifrolumab exposure increased more than dose-proportionally from 100 to 1000 mg IV every 4 weeks. Based on population pharmacokinetics modeling, the baseline median linear CL was 0.193 L/day in IFNGS-high patients and 0.153 L/day in IFNGS-low/healthy volunteers. After a year, median anifrolumab linear CL decreased by 8.4% from baseline. Body weight and IFNGS were significant pharmacokinetic covariates, whereas age, sex, race, disease activity, SLE medications, and presence of antidrug antibodies had no significant effect on anifrolumab pharmacokinetics. Anifrolumab at a concentration of 300 mg IV every 4 weeks was predicted to be below the lower limit of quantitation in 95% of patients ≈10 weeks after a single dose and ≈16 weeks after stopping dosing at steady state. To conclude, anifrolumab exhibited nonlinear pharmacokinetics and time-varying linear CL; doses ≥300 mg IV every 4 weeks provided sustained anifrolumab concentrations. This study provides further evidence to support the use of anifrolumab 300 mg IV every 4 weeks in patients with moderate to severe SLE.
Collapse
Affiliation(s)
- Joachim Almquist
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&DAstraZenecaGothenburgSweden
| | - Denison Kuruvilla
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
GenentechSouth San FranciscoCaliforniaUSA
| | - Tu Mai
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
GenentechSouth San FranciscoCaliforniaUSA
| | - Raj Tummala
- Clinical Development, Late Respiratory & Immunology, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Wendy I. White
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety Sciences, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Weifeng Tang
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety Sciences, R&DAstraZenecaGaithersburgMarylandUSA
| | - Lorin Roskos
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
ExelixisAlamedaCaliforniaUSA
| | - Yen Lin Chia
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
SeagenSouth San FranciscoCaliforniaUSA
| |
Collapse
|
10
|
Sim TM, Ong SJ, Mak A, Tay SH. Type I Interferons in Systemic Lupus Erythematosus: A Journey from Bench to Bedside. Int J Mol Sci 2022; 23:2505. [PMID: 35269647 PMCID: PMC8910773 DOI: 10.3390/ijms23052505] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 01/15/2023] Open
Abstract
Dysregulation of type I interferons (IFNs) has been implicated in the pathogenesis of systemic lupus erythematosus (SLE) since the late 1970s. The majority of SLE patients demonstrate evidence of type I IFN pathway activation; however, studies attempting to address the relationship between type I IFN signature and SLE disease activity have yielded conflicting results. In addition to type I IFNs, type II and III IFNs may overlap and also contribute to the IFN signature. Different genetic backgrounds lead to overproduction of type I IFNs in SLE and contribute to the breakdown of peripheral tolerance by activation of antigen-presenting myeloid dendritic cells, thus triggering the expansion and differentiation of autoreactive lymphocytes. The consequence of the continuous stimulation of the immune system is manifested in different organ systems typical of SLE (e.g., mucocutaneous and cardiovascular involvement). After the discovery of the type I IFN signature, a number of different strategies have been developed to downregulate the IFN system in SLE patients, finally leading to the successful trial of anifrolumab, the second biologic to be approved for the treatment of SLE in 10 years. In this review, we will discuss the bench to bedside translation of the type I IFN pathway and put forward some issues that remain unresolved when selecting SLE patients for treatment with biologics targeting type I IFNs.
Collapse
Affiliation(s)
- Tao Ming Sim
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (T.M.S.); (A.M.)
| | - Siying Jane Ong
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (T.M.S.); (A.M.)
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Sen Hee Tay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (T.M.S.); (A.M.)
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| |
Collapse
|
11
|
Mucke J, Schneider M. Innovationen in der Arzneimitteltherapie des systemischen Lupus erythematodes. Internist (Berl) 2022; 63:566-572. [DOI: 10.1007/s00108-021-01250-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 11/30/2022]
|
12
|
Abstract
Anifrolumab (anifrolumab-fnia; Saphnelo™) is a monoclonal antibody antagonist of the type 1 interferon receptor (IFNAR). It is being developed by AstraZeneca (under license from Medarex, now Bristol-Myers Squibb) for the treatment of autoimmune disorders, including systemic lupus erythematosus (SLE) and lupus nephritis, the underlying pathogenesis of which involves type 1 interferon. In July 2021, intravenous anifrolumab was approved in the USA for the treatment of adult patients with moderate to severe SLE who are receiving standard therapy. Anifrolumab (intravenous or subcutaneous) continues to be assessed in clinical studies in SLE in various countries, and the intravenous formulation is under regulatory review in the EU and Japan. This article summarizes the milestones in the development of anifrolumab leading to this first approval for the treatment of moderate to severe SLE.
Collapse
Affiliation(s)
- Emma D Deeks
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
13
|
Zheng F, Hou P, Corpstein CD, Park K, Li T. Multiscale pharmacokinetic modeling of systemic exposure of subcutaneously injected biotherapeutics. J Control Release 2021; 337:407-416. [PMID: 34324897 DOI: 10.1016/j.jconrel.2021.07.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 07/24/2021] [Indexed: 12/19/2022]
Abstract
Subcutaneously injected formulations have been developed for many biological products including monoclonal antibodies (mAbs). A knowledge gap nonetheless remains regarding the absorption and catabolism mechanisms and kinetics of a large molecule at the administration site. A multiscale pharmacokinetic (PK) model was thus developed by coupling multiphysics simulations of subcutaneous (SC) absorption kinetics with whole-body pharmacokinetic (PK) modeling, bridged by consideration of the presystemic clearance by the initial lymph. Our local absorption simulation of SC-injected albumin enabled the estimation of its presystemic clearance and led to the whole-body PK modeling of systemic exposure. The local absorption rate of albumin was found to be influential on the PK profile. Additionally, nineteen mAbs were explored via this multiscale simulation and modeling framework. The computational results suggest that stability propensities of the mAbs are correlated with the presystemic clearance, and electrostatic charges in the complementarity-determining region influence the local absorption rate. Still, this study underscores a critical need to experimentally determine various biophysical characteristics of a large molecule and the biomechanical properties of human skin tissues.
Collapse
Affiliation(s)
- Fudan Zheng
- Industrial & Physical Pharmacy, Purdue University West Lafayette, Indiana, USA
| | - Peng Hou
- Industrial & Physical Pharmacy, Purdue University West Lafayette, Indiana, USA
| | | | - Kinam Park
- Industrial & Physical Pharmacy, Purdue University West Lafayette, Indiana, USA
| | - Tonglei Li
- Industrial & Physical Pharmacy, Purdue University West Lafayette, Indiana, USA.
| |
Collapse
|
14
|
Bruce IN, Nami A, Schwetje E, Pierson ME, Rouse T, Chia YL, Kuruvilla D, Abreu G, Tummala R, Lindholm C. Pharmacokinetics, pharmacodynamics, and safety of subcutaneous anifrolumab in patients with systemic lupus erythematosus, active skin disease, and high type I interferon gene signature: a multicentre, randomised, double-blind, placebo-controlled, phase 2 study. THE LANCET. RHEUMATOLOGY 2021; 3:e101-e110. [PMID: 38279367 DOI: 10.1016/s2665-9913(20)30342-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND 300 mg of intravenous anifrolumab every 4 weeks added to standard-of-care treatment for patients with systemic lupus erythematosus (SLE) reduced disease activity and glucocorticoid requirement in a previous phase 3 trial. Because patients might find subcutaneous administration more convenient than intravenous delivery, we aimed to evaluate the pharmacokinetics, pharmacodynamics, safety, and efficacy of subcutaneous anifrolumab in patients with SLE, active skin disease, and a high type I interferon gene signature. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 2 study was done at 12 hospitals and outpatient clinics in Hungary, South Korea, Poland, and the USA. Eligible patients were aged 18-70 years, and had SLE with high type I interferon gene signature and an activity score on the Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) of at least 10. Enrolled participants were randomly assigned (3:1:3:1) by use of a voice-web response system to receive either 150 mg of subcutaneous anifrolumab or corresponding placebo, or 300 mg of subcutaneous anifrolumab or corresponding placebo in addition to stable standard-of-care treatment. The study was double-blinded with respect to intervention but not dose, until 12 weeks. Doses of oral glucocorticoids were tapered after week 12. The primary pharmacokinetic endpoint was the serum concentration of anifrolumab based on the maximum concentration after the first dose and the minimum (trough) concentration before subsequent doses and was measured in all patients who received anifrolumab and had at least one quantifiable serum pharmacokinetics observation following the first dose. The primary pharmacodynamic endpoint was neutralisation of the type I interferon pharmacodynamic signature at week 12 and was assessed in all patients with a high type I interferon pharmacodynamics signature at baseline based on a 21-gene test. Safety was evaluated in the full analysis set, which included all patients who received at least one dose of anifrolumab. This trial is completed and is registered at ClinicalTrials.gov, NCT02962960. FINDINGS Between March 14, 2017, and Oct 26, 2017, 36 patients were randomly assigned to receive 150 mg of anifrolumab (n=14), 300 mg of anifrolumab (n=13), or placebo (n=9). Two patients in the anifrolumab 150 mg group were excluded from the pharmacodynamic analysis set (n=34). Ten (71%) of 14 patients in the anifrolumab 150 mg group, ten (77%) of 13 patients in the anifrolumab 300 mg group, and nine (100%) of the nine patients in the placebo group completed 52 weeks of treatment. At week 12, pre-dose mean trough serum concentrations of anifrolumab were more than dose proportional between the anifrolumab 150 mg group (19·82 μg/mL [SD 15·01]) and the anifrolumab 300 mg group (60·28 μg/mL [43·66]), and the pharmacokinetics were non-linear. At week 12, the median percentage neutralisation of the type I interferon gene signature was higher with 150 mg (88·0% [median absolute deviation 7·4]) and 300 mg (90·7% [3·3]) of anifrolumab than with placebo (18·5% [8·1]), and more patients in the anifrolumab 150 mg group and the anifrolumab 300 mg group than in the placebo group had neutralisation of 75% or more (eight [67%] of 12 vs ten [77%] of 13 vs one [11%] of nine). At least one adverse event was reported by 23 (85%) of 27 patients in the anifrolumab groups and by seven (78%) of nine patients in the placebo group; most adverse events were of mild-to-moderate severity. Serious adverse events were reported in six (22%) of 27 patients in the anifrolumab groups (four patients in the 150 mg group and two in the 300 mg group). No serious adverse events were reported in the placebo group. Herpes zoster infection was reported by three (11%) of 27 patients in the anifrolumab groups and by one (11%) of nine patients in the placebo group. There were no treatment-related deaths. INTERPRETATION Anifrolumab, administered subcutaneously every 2 weeks to patients with SLE and moderate-to-severe skin manifestations, had non-linear pharmacokinetics that were more than dose proportional, and neutralised the type I interferon gene signature in a dose-dependent manner. The safety profile was consistent with previous studies of intravenous anifrolumab, supporting the continued development of anifrolumab as a subcutaneously administered therapy for patients with SLE. FUNDING AstraZeneca.
Collapse
Affiliation(s)
- Ian N Bruce
- National Institute for Health Research Manchester Biomedical Research Centre, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tanaka Y, Tummala R. Anifrolumab, a monoclonal antibody to the type I interferon receptor subunit 1, for the treatment of systemic lupus erythematosus: an overview from clinical trials. Mod Rheumatol 2020; 31:1-12. [PMID: 32814461 DOI: 10.1080/14397595.2020.1812201] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic activation of the type I interferon (IFN) pathway plays a critical role in systemic lupus erythematosus (SLE) pathogenesis. Anifrolumab is a human monoclonal antibody to the type I IFN receptor subunit 1, which blocks the action of type I IFNs. Two phase 3 studies (TULIP-1 and TULIP-2) and a phase 2b study (MUSE) provide substantial evidence for the efficacy and safety of anifrolumab for moderately to severely active SLE. In all three studies, monthly intravenous anifrolumab 300 mg was associated with treatment differences >16% compared with placebo at Week 52 in British Isles Lupus Assessment Group-based Composite Lupus Assessment response rates. The combined data across a range of other clinically significant endpoints (e.g. oral corticosteroid reduction, improved skin disease, flare reduction) further support the efficacy of anifrolumab for SLE treatment. The safety profile of anifrolumab was generally similar across all studies; serious adverse events occurred in 8-16% and 16-19% of patients receiving anifrolumab and placebo, respectively. Herpes zoster incidence was greater with anifrolumab (≤7%) vs placebo (≤2%). Evidence from these clinical trials suggests that in patients with active SLE, anifrolumab is superior to placebo in achieving composite endpoints of disease activity response and oral corticosteroid reduction.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Raj Tummala
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
16
|
Paredes JL, Niewold TB. Type I interferon antagonists in clinical development for lupus. Expert Opin Investig Drugs 2020; 29:1025-1041. [PMID: 32700979 PMCID: PMC7924012 DOI: 10.1080/13543784.2020.1797677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a severe chronic and incurable autoimmune disease. Treatment includes glucocorticoids and immunosuppressants which typically result in partial responses, and hence there is a great need for new therapies. The type I interferon (IFN) pathway is activated in more than 50% of SLE patients, and it is strongly implicated as a pathogenic factor in SLE. AREAS COVERED We searched the literature using 'SLE and interferon antagonists' as search terms. This identified a number of therapeutics that have entered clinical development targeting type I IFN in SLE. These include monoclonal antibodies against type I IFN cytokines and a kinoid vaccination strategy to induce anti-IFN antibodies. EXPERT OPINION Type I IFN antagonists have had some success, but many molecules have not progressed to phase III. These varied results are likely attributed to the multiple concurrent cytokine abnormalities present in SLE, the imprecise nature of the IFN signature as a readout for type I IFN and difficulties with clinical trials such as background medication use and diffuse composite disease activity measures. Despite these challenges, it seems likely that a type I IFN antagonist will come to clinical utility for SLE given the large unmet need and the recent phase III success with anifrolumab.
Collapse
Affiliation(s)
- Jacqueline L Paredes
- Colton Center for Autoimmunity, New York University School of Medicine , New York, NY, USA
| | - Timothy B Niewold
- Colton Center for Autoimmunity, New York University School of Medicine , New York, NY, USA
| |
Collapse
|
17
|
Anderson E, Furie R. Anifrolumab in systemic lupus erythematosus: current knowledge and future considerations. Immunotherapy 2020; 12:275-286. [PMID: 32237942 DOI: 10.2217/imt-2020-0017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that is potentially life-threatening and can affect any organ. The complex pathogenesis and heterogeneity of the disease, among other factors, present significant challenges in developing new therapies. Knowledge gained over many years has implicated type I interferon (IFN) in the pathogenesis of SLE and anti-IFN therapies hold promise as a much-needed future treatment for SLE. Anifrolumab, a human monoclonal antibody against the type I IFN receptor, has recently been evaluated in two Phase III clinical trials for the treatment of moderate-to-severe SLE. Here, we review the clinical efficacy and safety of anifrolumab and discuss the potential challenges in determining the optimal SLE patient subgroup for treatment.
Collapse
Affiliation(s)
- Erik Anderson
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Richard Furie
- Division of Rheumatology, Zucker School of Medicine at Hofstra/Northwell, 865 Northern Boulevard, Great Neck, NY 11021, USA
| |
Collapse
|
18
|
Pathogenic Role of Type I Interferons in HIV-Induced Immune Impairments in Humanized Mice. Curr HIV/AIDS Rep 2020; 16:224-229. [PMID: 31055732 DOI: 10.1007/s11904-019-00444-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW Recent findings on the critical pathogenic role of type 1 interferons (IFN-I) in HIV-1 persistence in humanized mice suggest that inhibiting IFN-I signaling transiently will reverse HIV-induced inflammatory diseases and rescue anti-HIV immunity to control HIV-1 reservoirs. RECENT FINDINGS In both humanized mice and in monkeys, IFN-I signaling is functionally defined to play an important role in suppressing early HIV-1 and SIV infection. During persistent infection in humanized mice, however, IFN-I signaling is revealed to induce T cell depletion and impairment. Interestingly, in HIV-infected mice with effective combination antiretroviral therapy (cART), blocking IFN-I signaling reverses HIV-induced inflammation, rescues anti-HIV T cells, and reduces HIV-1 reservoirs. These findings functionally define the role of IFN-I in HIV-1 reservoir persistence and suggest that blocking IFN-I signaling will provide a novel therapeutic strategy to (i) reverse inflammation-associated diseases in HIV patients under cART, (ii) rescue host anti-HIV immunity, and (iii) reduce or control HIV-1 reservoirs.
Collapse
|
19
|
Varkhede N, Forrest L. Understanding the Monoclonal Antibody Disposition after Subcutaneous Administration using a Minimal Physiologically based Pharmacokinetic Model. JOURNAL OF PHARMACY AND PHARMACEUTICAL SCIENCES 2019; 21:130s-148s. [PMID: 30011390 DOI: 10.18433/jpps30028] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Monoclonal antibodies (mAbs) are commonly administered by subcutaneous (SC) route. However, bioavailability is often reduced after SC administration. In addition, the sequential transfer of mAbs through the SC tissue and lymphatic system is not completely understood. Therefore, major objectives of this study were a) To understand absorption of mAbs via the lymphatic system after SC administration using physiologically based pharmacokinetic (PBPK) modeling, and b) to demonstrate application of the model for prediction of SC pharmacokinetics (PK) of mAbs. METHODS A minimal PBPK model was constructed using various physiological parameters related to the SC injection site and lymphatic system. The remainder of the body organs were represented using a 2-compartment model (central and peripheral compartments), with parameters derived from available intravenous (IV) PK data. The IV and SC clinical PK data of a total of 10 mAbs were obtained from literature. The SC PK data were used to estimate the lymphatic trunk-lymph node (LN) clearance. RESULTS The mean estimated lymphatic trunk-LN clearance obtained from 37 SC PK profiles of mAbs was 0.00213 L/h (0.001332 to 0.002928, 95% confidence intervals). The estimated lymphatic trunk-LN clearance was greater for the mAbs with higher isoelectric point (pI). In addition, the estimated clearance increased with decrease in the bioavailability. CONCLUSION The minimal PBPK model identified SC injection site lymph flow, afferent and efferent lymph flows, and volumes associated with the SC injection site, lymphatic capillaries and lymphatic trunk-LN as important physiological parameters governing the absorption of mAbs after SC administration. The model may be used to predict PK of mAbs using the relationship of lymphatic trunk-LN clearance and the pI. In addition, the model can be used as a bottom platform to incorporate SC and lymphatic in vitro clearance data for mAb PK prediction in the future.
Collapse
Affiliation(s)
- Ninad Varkhede
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, USA
| | - Laird Forrest
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, USA
| |
Collapse
|
20
|
Felten R, Scher F, Sagez F, Chasset F, Arnaud L. Spotlight on anifrolumab and its potential for the treatment of moderate-to-severe systemic lupus erythematosus: evidence to date. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1535-1543. [PMID: 31190735 PMCID: PMC6514126 DOI: 10.2147/dddt.s170969] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022]
Abstract
Previous reports have described the appearance of systemic lupus erythematosus (SLE) cases following interferon-α (IFN-α) therapy, IFN-regulated gene expression is significantly increased in SLE, and an association between SLE and gene variants belonging to IFN downstream pathways has been shown. Based on this, targeting of IFN and of their signaling pathways has appeared to be interesting developments within the field of SLE therapy. Different specific type I IFN antagonists have been studied in clinical trials and some of those have already reached Phase III. A potential approach would be to target IFN receptors rather than IFN themselves. Anifrolumab (previously MEDI-546) is a fully human monoclonal antibody (Ab) that binds to subunit 1 of the type I IFN receptor (IFNAR1), blocking the action of different type I IFNs (IFN-α, IFN-β and IFN-ω). This drug has been assessed in 11 clinical studies: 9 in SLE, 1 in systemic sclerosis and 1 in rheumatoid arthritis. In SLE, clinical development reached Phase I for 1 study and Phases II and III for 5 and 3 trials, respectively. The Phase IIb, randomized control trial (RCT), double-blind, placebo-controlled study of adults with moderate-to-severe SLE (MUSE trial) showed positive results on the composite primary endpoint SRI-4. Greater efficacy was seen in patients with high baseline IFN gene signature compared with those with low baseline IFN gene signature. Anifrolumab also demonstrated promising results on cutaneous and arthritic manifestations, especially among patients with a high IFN gene signature. The pivotal Treatment of Uncontrolled Lupus via the Interferon IFN Pathway (TULIP 1 and 2 studies are now completed. In August 2018, the promoter announced that the TULIP 1 Phase III trial did not reach its primary endpoint. The release of the completed but not yet published Phase II studies and of the TULIP pivotal trials results will further inform us on the actual therapeutic potential of anifrolumab.
Collapse
Affiliation(s)
- Renaud Felten
- Rheumatology Department, University Hospital of Strasbourg, Université de Strasbourg, Strasbourg, F-67000, France.,National Reference Centre for Rare Systemic and Autoimmune Diseases East South-West (RESO), Strasbourg, France.,Immunology Laboratory, "Immunopathologie et Chimie Thérapeutique", Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, Strasbourg, F-67000, France
| | - Florence Scher
- Pharmacy-Sterilisation Department, University Hospital of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Flora Sagez
- Rheumatology Department, University Hospital of Strasbourg, Université de Strasbourg, Strasbourg, F-67000, France.,National Reference Centre for Rare Systemic and Autoimmune Diseases East South-West (RESO), Strasbourg, France
| | - François Chasset
- Faculty of Medicine at Sorbonne University, AP-HP, Dermatology and Allergology Department, Tenon Hospital, Sorbonne University, Paris, F-75020, France
| | - Laurent Arnaud
- Rheumatology Department, University Hospital of Strasbourg, Université de Strasbourg, Strasbourg, F-67000, France.,National Reference Centre for Rare Systemic and Autoimmune Diseases East South-West (RESO), Strasbourg, France.,Immuno-Rheumatology Laboratory, "Laboratoire d'ImmunoRhumatologie Moléculaire", INSERM UMR_S1109, Strasbourg, F-67000, France
| |
Collapse
|
21
|
Tanaka Y, Takeuchi T, Okada M, Ishii T, Nakajima H, Kawai S, Nagashima T, Hayashi N, Wang L, Tummala R. Safety and tolerability of anifrolumab, a monoclonal antibody targeting type I interferon receptor, in Japanese patients with systemic lupus erythematosus: A multicenter, phase 2, open-label study. Mod Rheumatol 2019; 30:101-108. [PMID: 30793642 DOI: 10.1080/14397595.2019.1583833] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objectives: This study evaluated the safety and tolerability of anifrolumab, a monoclonal antibody targeting the type I interferon (IFN) receptor, in Japanese patients with moderate-to-severe systemic lupus erythematosus (SLE).Methods: In this open-label, phase 2, dose-escalation study, patients received intravenous (IV) anifrolumab 100, 300, or 1000 mg every 4 weeks from days 29 to 337 (Stage 1). Patients who completed Stage 1 continued anifrolumab 300 mg every 4 weeks for 156 weeks (Stage 2). The primary objective was to evaluate the safety of anifrolumab for 48 weeks (Stage 1) and 156 weeks (Stage 2). The pharmacokinetics and pharmacodynamics of anifrolumab were also assessed.Results: Of 20 patients enrolled in Stage 1, 17 received IV anifrolumab 100 mg (n = 6), 300 mg (n = 5), or 1000 mg (n = 6). Adverse events (AE) and serious AE (SAE) incidences were similar between dose cohorts. SAEs occurred in 41% (Stage 1) and 33% (Stage 2) of patients; AEs leading to discontinuation occurred in 24% (Stage 1) and 22% (Stage 2) of patients. Anifrolumab had non-linear pharmacokinetics after the first and last dose and dose-dependently suppressed the IFN gene signature.Conclusion: Anifrolumab was well tolerated among Japanese patients with moderate-to-severe SLE.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masato Okada
- Division of Allergy & Rheumatology, St. Luke's International Hospital, Tokyo, Japan
| | - Tomonori Ishii
- Department of Rheumatology and Hematology, Tohoku University Hospital, Miyagi, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Chiba University Hospital, Chiba, Japan
| | - Shinichi Kawai
- Department of Inflammation & Pain Control Research, Toho University School of Medicine, Tokyo, Japan
| | - Takao Nagashima
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Nobuya Hayashi
- Research and Development, AstraZeneca K.K., Osaka, Japan
| | - Liangwei Wang
- Global Medicine Division, AstraZeneca Pharmaceuticals, Gaithersburg, MD, USA
| | - Raj Tummala
- Global Medicine Division, AstraZeneca Pharmaceuticals, Gaithersburg, MD, USA
| |
Collapse
|