1
|
Pitsiu M, Yalkinoglu Ö, Farrell C, Girard P, Vazquez‐Mateo C, Papasouliotis O. Population pharmacokinetics of atacicept in systemic lupus erythematosus: An analysis of three clinical trials. CPT Pharmacometrics Syst Pharmacol 2023; 12:1157-1169. [PMID: 37332136 PMCID: PMC10431037 DOI: 10.1002/psp4.12982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
B cell stimulating factor (BLyS) and a proliferation-inducing ligand (APRIL) are targets for novel treatments in patients with systemic lupus erythematosus (SLE). Atacicept is a recombinant, soluble fusion protein that blocks BLyS and APRIL activity. This study characterized the pharmacokinetic (PK) profile of atacicept using a population PK model and identified covariates explaining the PK variability. Total atacicept concentrations from a phase I study in healthy volunteers and two phase II studies in patients with SLE, using subcutaneous administration, were modeled using a quasi-steady-state approximation of the target-mediated drug disposition model with first-order absorption. The model included 3640 serum atacicept concentration records from 37 healthy volunteers and 503 patients with SLE and described total atacicept concentrations of the three trials, providing precise estimates of all parameters. Body weight and baseline BLyS concentration were the only statistically significant covariates, whereas no differences were found between patients and healthy volunteers. Apparent clearance and volume of the central compartment increased with body weight and initial target concentration increased with baseline BLyS. The change on atacicept exposure was moderate, with a difference in area under the curve compared with the median of 20%-32% for body weight, and 7%-18% for BLyS. Therefore, the effects of these covariates on atacicept exposure are not expected to be clinically relevant. The model described the complete total atacicept concentration-time profiles without finding any differences between healthy subjects and patients with SLE and supports the 150 mg once weekly dose for further trials.
Collapse
Affiliation(s)
- Maria Pitsiu
- Quantitative Pharmacology and PharmacometricsICON Clinical Research UK LtdMarlowUK
| | - Özkan Yalkinoglu
- Translational Medicinethe healthcare business of Merck KGaADarmstadtGermany
| | - Colm Farrell
- Quantitative Pharmacology and PharmacometricsICON Clinical Research UK LtdMarlowUK
| | - Pascal Girard
- Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, GermanyAres Trading S.A.LausanneSwitzerland
| | | | - Orestis Papasouliotis
- Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, GermanyAres Trading S.A.LausanneSwitzerland
| |
Collapse
|
2
|
Eslami M, Willen D, Papasouliotis O, Schuepbach-Mallpell S, Willen L, Donzé O, Yalkinoglu Ö, Schneider P. Kinetics of free and ligand-bound atacicept in human serum. Front Immunol 2022; 13:1035556. [PMID: 36532058 PMCID: PMC9756848 DOI: 10.3389/fimmu.2022.1035556] [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: 09/02/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
BAFF (B cell activation factor of the TNF family/B lymphocyte stimulator, BLyS) and APRIL (a proliferation-inducing ligand) are targeted by atacicept, a decoy receptor consisting of the extracellular domain of TACI (transmembrane activator and calcium-modulator and cyclophilin (CAML) interactor) fused to the Fc portion of human IgG1. The purpose of the study was to characterize free and ligand-bound atacicept in humans. Total and active atacicept in serum of healthy volunteers receiving a single dose of subcutaneous atacicept or in patients treated weekly for one year were measured by ELISA, Western blot, or cell-based assays. Pharmacokinetics of free and bound atacicept were predicted based on total atacicept ELISA results. Persistence of complexes of purified atacicept bound to recombinant ligands was also monitored in mice. Results show that unbound or active atacicept in human serum exceeded 0.1 µg/ml for one week post administration, or throughout a 1-year treatment with weekly administrations. After a single administration of atacicept, endogenous BAFF bound to atacicept was detected after 8 h then increased about 100-fold within 2 to 4 weeks. Endogenous heteromers of BAFF and APRIL bound to atacicept also accumulated, but atacicept-APRIL complexes were not detected. In mice receiving intravenous injections of purified complexes pre-formed in vitro, atacicept-BAFF persisted longer (more than a week) than atacicept-APRIL (less than a day). Thus, only biologically inactive BAFF and BAFF-APRIL heteromers accumulate on atacicept in vivo. The measure of active atacicept provides further support for the once-weekly dosing regimen implemented in the clinical development of atacicept.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Daniela Willen
- Clinical Pharmacology, Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Orestis Papasouliotis
- Translational Medicine, Merck Institute for Pharmacometrics (an affiliate of Merck KGaA), Lausanne, Switzerland
| | | | - Laure Willen
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Özkan Yalkinoglu
- Clinical Pharmacology, Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland,*Correspondence: Pascal Schneider,
| |
Collapse
|
3
|
Willen D, Uhl W, Wolna P, Papasouliotis O, Yalkinoglu Ö. Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Atacicept in a Randomized Trial in Healthy Caucasian and Japanese Subjects. Eur J Drug Metab Pharmacokinet 2020; 45:27-40. [PMID: 31529406 PMCID: PMC6994531 DOI: 10.1007/s13318-019-00575-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Background and Objective Atacicept is an inhibitor of the B lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL), and is being studied in relation to immunological disease. Currently, limited data on atacicept are available in non-Caucasian subjects. Pharmacokinetic data from earlier studies of atacicept were derived using an enzyme-linked immunosorbent assay (ELISA), which was subsequently found to have inadequacies. Hence, a new bioanalytical ELISA for total atacicept was developed and validated. We conducted this randomized, double-blind, placebo-controlled phase I study to compare the safety, tolerability, pharmacokinetics, and pharmacodynamics of atacicept in healthy Japanese and Caucasian subjects while generating pharmacokinetic data using the new ELISA. Methods Japanese subjects aged ≥ 18 to ≤ 55 years (n = 24) were randomized (1:1:1:1) to a single subcutaneous dose of atacicept 25, 75, or 150 mg or placebo. Caucasian subjects were then enrolled to match the Japanese subjects’ gender, body weight (± 20%), and height (± 15%). Results Atacicept was well tolerated and there were no clinically significant differences in treatment-emergent adverse events (TEAEs), vital signs, or laboratory parameters between the Japanese and Caucasian subjects. Most (90%) TEAEs were mild; no severe or serious TEAEs or deaths occurred. Weight-adjusted atacicept exposure was comparable between ethnicities and across doses: the Japanese/Caucasian ratio of the area under the serum concentration–time curve from time zero to the last sampling point (AUC0–t) was 107.21% (90% CI 93.42–123.02%) and the Japanese/Caucasian ratio of maximum serum concentration (Cmax) was 95.74% (90% CI 74.26–123.43%; ANCOVA). Median time to reach Cmax (tmax) was 20–60 h across all subjects. Dose–exposure relationships were comparable for the two ethnicities, with dose-normalized AUC0–t decreasing with increasing dose, indicating nonlinear pharmacokinetics for the doses examined. There were no statistically significant differences between ethnicities in the pharmacokinetics–dose relationship. Some transient dose-related decreases in mean serum immunoglobulin (Ig)A and IgM, but not IgG, were observed after atacicept administration. There were small transient increases in peripheral B cell numbers in the first 4 days after dosing that were larger with atacicept than with placebo, with no apparent dose relationship. No anti-atacicept antibodies were detected. Conclusion The safety, pharmacokinetic, and pharmacodynamic profiles of atacicept in healthy Japanese subjects were comparable to those in healthy Caucasian subjects. EudraCT-ID: 2013-002703-34. Electronic supplementary material The online version of this article (10.1007/s13318-019-00575-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniela Willen
- Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Wolfgang Uhl
- Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Peter Wolna
- Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Orestis Papasouliotis
- Merck Institute for Pharmacometrics (An Affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | |
Collapse
|
4
|
Robinson MJ, Webster RH, Tarlinton DM. How intrinsic and extrinsic regulators of plasma cell survival might intersect for durable humoral immunity. Immunol Rev 2020; 296:87-103. [DOI: 10.1111/imr.12895] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Marcus J. Robinson
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| | - Rosela H. Webster
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| | - David M. Tarlinton
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| |
Collapse
|
5
|
Schrezenmeier E, Jayne D, Dörner T. Targeting B Cells and Plasma Cells in Glomerular Diseases: Translational Perspectives. J Am Soc Nephrol 2018; 29:741-758. [PMID: 29326157 DOI: 10.1681/asn.2017040367] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The unique contributions of memory B cells and plasma cells in kidney diseases remain unclear. In this review, we evaluate the clinical experience with treatments directed at B cells, such as rituximab, and at plasma cells, such as proteasome inhibition, to shed light on the role of these two B lineage compartments in glomerular diseases. Specifically, analysis of these targeted interventions in diseases such as ANCA-associated vasculitis, SLE, and antibody-mediated transplant rejection permits insight into the pathogenetic effect of these cells. Notwithstanding the limitations of preclinical models and clinical studies (heterogeneous populations, among others), the data suggest that memory B and plasma cells represent two engines of autoimmunity, with variable involvement in these diseases. Whereas memory B cells and plasma cells appear to be key in ANCA-associated vasculitis and antibody-mediated transplant rejection, respectively, SLE seems likely to be driven by both autoimmune compartments. These conclusions have implications for the future development of targeted therapeutics in immune-mediated renal disease.
Collapse
Affiliation(s)
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Thomas Dörner
- Rheumatology and Clinical Immunology, Department of Medicine, Charité Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany; and
| |
Collapse
|
6
|
Yu T, Enioutina EY, Brunner HI, Vinks AA, Sherwin CM. Clinical Pharmacokinetics and Pharmacodynamics of Biologic Therapeutics for Treatment of Systemic Lupus Erythematosus. Clin Pharmacokinet 2017; 56:107-125. [PMID: 27384528 PMCID: PMC5575762 DOI: 10.1007/s40262-016-0426-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease with potentially severe clinical manifestation that mainly affects women of child-bearing age. Patients who do not respond to standard-of-care therapies, such as corticosteroids and immunosuppressants, require biologic therapeutics that specifically target a single or multiple SLE pathogenesis pathways. This review summarizes the clinical pharmacokinetic and pharmacodynamic characteristics of biologic agents that are approved, used off-label, or in the active pipeline of drug development for SLE patients. Depending on the type of target, the interacting biologics may exhibit linear (non-specific) or non-linear (target-mediated) disposition profiles, with terminal half-lives varying from approximately 1 week to 1 month. Biologics given by subcutaneous administration, which offers dosing flexibility over intravenous administration, demonstrated a relatively slow absorption with a time to maximum concentration of approximately 1 day to 2 weeks and a variable bioavailability of 30-82 %. The population pharmacokinetics of monoclonal antibodies were best described by a two-compartment model with central clearance and steady-state volume of distribution ranging from 0.176 to 0.215 L/day and 3.60-5.29 L, respectively. The between-subject variability in pharmacokinetic parameters were moderate (20-79 %) and could be partially explained by body size. The development of linked pharmacokinetic-pharmacodynamic models incorporating SLE disease biomarkers are an attractive strategy for use in dosing regimen simulation and optimization. The relationship between efficacy/adverse events and biologic concentration should be evaluated to improve clinical trial outcomes, especially for biologics in the advanced phase of drug development. New strategies, such as model-based precision dosing dashboards, could be utilized to incorporate information collected from therapeutic drug monitoring into pharmacokinetic/pharmacodynamic models to enable individualized dosing in real time.
Collapse
Affiliation(s)
- Tian Yu
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, SLC, Utah 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Elena Y Enioutina
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, SLC, Utah 295 Chipeta Way, Salt Lake City, UT, 84108, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hermine I Brunner
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Catherine M Sherwin
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, SLC, Utah 295 Chipeta Way, Salt Lake City, UT, 84108, USA.
- Department of Pediatrics, Clinical Trials Office, Salt Lake City, UT, USA.
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
7
|
Zhao Q, Chen X, Li J, Jiang J, Li M, Zhong W, Li Z, Leung SO, Zhang F, Hu P. Pharmacokinetics, Pharmacodynamics and Preliminary Observations for Clinical Activity and Safety of Multiple Doses of Human Mouse Chimeric Anti-CD22 Monoclonal Antibody (SM03) in Chinese Patients with Systemic Lupus Erythematosus. Clin Drug Investig 2017; 36:889-902. [PMID: 27424629 DOI: 10.1007/s40261-016-0426-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVES SM03 is a novel recombinant, human/mouse chimeric immunoglobulin G1 monoclonal antibody directed against the CD22 antigen on human B lymphocytes. This was the first study to investigate the pharmacokinetics, pharmacodynamics, immunogenicity, safety and clinical activity of SM03 in patients with systemic lupus erythematosus (SLE). METHODS This study was an open, multiple-centre, parallel-group, multiple-ascending-dose, phase I study in 29 SLE patients. Pharmacokinetic assessment was conducted in 22 of these patients. Eligible patients received multiple intravenous infusions of SM03 for 4 weeks (240 mg/m2, 600 or 900 mg, once weekly) and were monitored over an 84-day observation period for pharmacokinetics, pharmacodynamics, immunogenicity, safety and clinical response. RESULTS After multiple-dose SM03, the maximal serum concentration of SM03 was reached within 3-7 h. The mean elimination half-life was 15 days. The average accumulation ratios of the area under the time-concentration curve and the maximum concentration after the fourth administration of SM03 were 2.0 and 1.5. CD19+ B-lymphocyte counts were decreased. Infections were the most common adverse events. No drug-related serious adverse events were reported. The therapeutic benefit of SM03 was observed mainly in patients with moderate-to-severe disease activity. CONCLUSION Pharmacokinetic exposure increased in a lower-than-dose-proportional manner up to 900 mg. SM03 was well tolerated at doses ranging from 240 mg/m2 to 900 mg, with no new safety signals identified. SM03 has potential efficacy in Chinese patients with SLE.
Collapse
Affiliation(s)
- Qian Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 41 Damucang, Xicheng District, Beijing, 100032, China
| | - Xia Chen
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 41 Damucang, Xicheng District, Beijing, 100032, China
| | - Jing Li
- Department of Rheumatology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Ji Jiang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 41 Damucang, Xicheng District, Beijing, 100032, China
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Wen Zhong
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 41 Damucang, Xicheng District, Beijing, 100032, China
| | - Zhengdong Li
- SinoMab BioScience, Ltd, BioIncubator No. 1-301, Science and Technology Park, Nanshan District, Shenzhen, 518057, Guangdong, China
| | - Shui-On Leung
- SinoMab BioScience, Ltd, BioIncubator No. 1-301, Science and Technology Park, Nanshan District, Shenzhen, 518057, Guangdong, China.
| | - Fengchun Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 41 Damucang, Xicheng District, Beijing, 100032, China.
| |
Collapse
|
8
|
Zhao Q, Chen X, Hou Y, Jiang J, Zhong W, Yao X, Wang W, Li L, Fang J, Zhang F, Hu P. Pharmacokinetics, Pharmacodynamics, Safety, and Clinical Activity of Multiple Doses of RCT-18 in Chinese Patients With Systemic Lupus Erythematosus. J Clin Pharmacol 2016; 56:948-59. [PMID: 26634642 DOI: 10.1002/jcph.686] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/23/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Qian Zhao
- Clinical Pharmacology Research Center & Translational Medicine Center, Peking Union Medical College Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Xia Chen
- Clinical Pharmacology Research Center & Translational Medicine Center, Peking Union Medical College Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Yong Hou
- Department of Rheumatology; Peking Union Medical College Hospital; Beijing China
| | - Ji Jiang
- Clinical Pharmacology Research Center & Translational Medicine Center, Peking Union Medical College Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Wen Zhong
- Clinical Pharmacology Research Center & Translational Medicine Center, Peking Union Medical College Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Xuejing Yao
- School of Life Science and Technology; Tongji University; Shanghai China
| | - Wenxiang Wang
- RC Biotechnologies Ltd; Economic and Technological Development Zone; Yantai China
| | - Lin Li
- RC Biotechnologies Ltd; Economic and Technological Development Zone; Yantai China
| | - Jianmin Fang
- School of Life Science and Technology; Tongji University; Shanghai China
| | - Fengchun Zhang
- Department of Rheumatology; Peking Union Medical College Hospital; Beijing China
| | - Pei Hu
- Clinical Pharmacology Research Center & Translational Medicine Center, Peking Union Medical College Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| |
Collapse
|
9
|
Cogollo E, Cogollo E, Silva MA, Isenberg D. Profile of atacicept and its potential in the treatment of systemic lupus erythematosus. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1331-9. [PMID: 25834391 PMCID: PMC4357613 DOI: 10.2147/dddt.s71276] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The importance of B cell activating factors in the generation of autoantibodies in patients with systemic lupus erythematosus (SLE) is now recognized. The two key factors, known as BAFF and APRIL, produced by a variety of cells including monocytes, dendritic cells and T cells, also help to regulate B cell maturation, function and survival. Biologic agents that block these factors have now been developed and tried out in large scale clinical trials in SLE patients. Benlysta which blocks BAFF has met some of its end points in clinical trials and is approved for use in patients with skin and joint disease who have failed conventional drugs. In contrast, clinical trials using atacicept which blocks both BAFF and APRIL have been more challenging to interpret. An early study in lupus nephritis was, mistakenly, abandoned due to serious infections thought to be linked to the biologic when in fact the dramatic fall in the immunoglobulin levels took place when the patients were given mycophenolate, prior to the introduction of the atacicept. Likewise the higher dose arm (150 mgm) of a flare prevention study was terminated prematurely when 2 deaths occurred. However, the mortality rate in this study was identical to that seen in the Benlysta studies and a post hoc analysis found a highly significant benefit for the 150mgm arm compared to the lower dose (75 mgm) and placebo arms. Other trials with both Benlysta and atacicept are on-going.
Collapse
Affiliation(s)
| | - Estafania Cogollo
- Department of Internal Medicine, Hospital Principe de Asturias, Alcala de Henares, Madrid, Spain
| | - Marta Amaral Silva
- Department of Internal Medicine, Hospital Distrital da Figueira da Foz, Coimbra, Portugal
| | - David Isenberg
- Centre for Rheumatology, Department of Medicine, University College London, London, UK
| |
Collapse
|
10
|
Pharmacokinetics, Pharmacodynamics, and Tolerability of Single Ascending Doses of RCT-18 in Chinese Patients with Rheumatoid Arthritis. Clin Pharmacokinet 2014; 53:1033-44. [DOI: 10.1007/s40262-014-0175-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
11
|
Nys J, Smulski CR, Tardivel A, Willen L, Kowalczyk C, Donzé O, Huard B, Hess H, Schneider P. No evidence that soluble TACI induces signalling via membrane-expressed BAFF and APRIL in myeloid cells. PLoS One 2013; 8:e61350. [PMID: 23620746 PMCID: PMC3631189 DOI: 10.1371/journal.pone.0061350] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/07/2013] [Indexed: 11/18/2022] Open
Abstract
Myeloid cells express the TNF family ligands BAFF/BLyS and APRIL, which exert their effects on B cells at different stages of differentiation via the receptors BAFFR, TACI (Transmembrane Activator and CAML-Interactor) and/or BCMA (B Cell Maturation Antigen). BAFF and APRIL are proteins expressed at the cell membrane, with both extracellular and intracellular domains. Therefore, receptor/ligand engagement may also result in signals in ligand-expressing cells via so-called “reverse signalling”. In order to understand how TACI-Fc (atacicept) technically may mediate immune stimulation instead of suppression, we investigated its potential to activate reverse signalling through BAFF and APRIL. BAFFR-Fc and TACI-Fc, but not Fn14-Fc, reproducibly stimulated the ERK and other signalling pathways in bone marrow-derived mouse macrophages. However, these effects were independent of BAFF or APRIL since the same activation profile was observed with BAFF- or APRIL-deficient cells. Instead, cell activation correlated with the presence of high molecular mass forms of BAFFR-Fc and TACI-Fc and was strongly impaired in macrophages deficient for Fc receptor gamma chain. Moreover, a TACI-Fc defective for Fc receptor binding elicited no detectable signal. Although these results do not formally rule out the existence of BAFF or APRIL reverse signalling (via pathways not tested in this study), they provide no evidence in support of reverse signalling and point to the importance of using appropriate specificity controls when working with Fc receptor-expressing myeloid cells.
Collapse
Affiliation(s)
- Josquin Nys
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Aubry Tardivel
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Laure Willen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | | - Bertrand Huard
- Department of Patho-Immunology, Medical University Centre, Geneva, Switzerland
- Division of Hematology, Geneva University Hospital, Geneva, Switzerland
| | | | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
- * E-mail:
| |
Collapse
|
12
|
Blüml S, McKeever K, Ettinger R, Smolen J, Herbst R. B-cell targeted therapeutics in clinical development. Arthritis Res Ther 2013; 15 Suppl 1:S4. [PMID: 23566679 PMCID: PMC3624127 DOI: 10.1186/ar3906] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
B lymphocytes are the source of humoral immunity and are thus a critical component of the adaptive immune system. However, B cells can also be pathogenic and the origin of disease. Deregulated B-cell function has been implicated in several autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis. B cells contribute to pathological immune responses through the secretion of cytokines, costimulation of T cells, antigen presentation, and the production of autoantibodies. DNA-and RNA-containing immune complexes can also induce the production of type I interferons, which further promotes the inflammatory response. B-cell depletion with the CD20 antibody rituximab has provided clinical proof of concept that targeting B cells and the humoral response can result in significant benefit to patients. Consequently, the interest in B-cell targeted therapies has greatly increased in recent years and a number of new biologics exploiting various mechanisms are now in clinical development. This review provides an overview on current developments in the area of B-cell targeted therapies by describing molecules and subpopulations that currently offer themselves as therapeutic targets, the different strategies to target B cells currently under investigation as well as an update on the status of novel therapeutics in clinical development. Emerging data from clinical trials are providing critical insight regarding the role of B cells and autoantibodies in various autoimmune conditions and will guide the development of more efficacious therapeutics and better patient selection.
Collapse
Affiliation(s)
- Stephan Blüml
- MedImmune, LLC, Department of Research, One MedImmune Way, Gaithersburg, MD 20854, USA
| | | | | | | | | |
Collapse
|
13
|
Lutalo PMK, D'Cruz DP. Belimumab for the management of systemic lupus erythematosus. Expert Opin Biol Ther 2012; 12:957-63. [DOI: 10.1517/14712598.2012.682980] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
14
|
Chugh PK. Lupus: novel therapies in clinical development. Eur J Intern Med 2012; 23:212-8. [PMID: 22385876 DOI: 10.1016/j.ejim.2011.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/30/2011] [Accepted: 11/02/2011] [Indexed: 01/09/2023]
Abstract
There have been significant advancements in understanding the immunopathogenesis of systemic lupus erythematosus. However, the developments in therapeutics have been rather slow. Belimumab, a B lymphocyte stimulator (BLyS) inhibitor has been approved for the treatment of this disease after more than 50 years. Numerous biological agents are being developed which target the B cells, T cells, and various cytokines. Among anti-B cell therapy, drugs target CD20+ cells (ocrelizumab, SBI-087), CD22+ cells (epratuzumab) \or the receptors of tumor necrosis factor (TNF) superfamily (atacicept, LY2127399, A-623). Monoclonal antibodies targeting interferon alpha (IFN-α) and gamma (IFN-γ) and interleukins (IL-6, 10) are being investigated for SLE. Novel targets include toll like receptors, phosphodiesterases, CD40 ligand and retinoid receptors. This review discusses various drugs which are in different phases of clinical trials and hold promise for patients suffering from this chronic debilitating disease.
Collapse
Affiliation(s)
- Preeta Kaur Chugh
- Department of Pharmacology, Maulana Azad Medical College and Associated Hospitals, Bahadur Shah Zafar Marg, New Delhi 110002, India.
| |
Collapse
|
15
|
Genovese MC, Kinnman N, de La Bourdonnaye G, Pena Rossi C, Tak PP. Atacicept in patients with rheumatoid arthritis and an inadequate response to tumor necrosis factor antagonist therapy: results of a phase II, randomized, placebo-controlled, dose-finding trial. ACTA ACUST UNITED AC 2011; 63:1793-803. [PMID: 21452293 DOI: 10.1002/art.30373] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To assess the efficacy, safety, and biologic activity of atacicept in patients with rheumatoid arthritis (RA) in whom the response to treatment with tumor necrosis factor antagonists was inadequate. METHODS The Atacicept for Reduction of Signs and Symptoms in Rheumatoid Arthritis Trial (AUGUST I) was a multicenter, phase II, double-blind, placebo-controlled dose-finding study involving 256 patients randomized 1:1:1:1 to receive atacicept (25 mg, 75 mg, or 150 mg) or placebo twice weekly for 4 weeks, then weekly for 21 weeks, with a 13-week treatment-free followup period (week 38). The primary end point was a response at week 26 according to the American College of Rheumatology criteria for 20% improvement in disease severity, using the C-reactive protein level. RESULTS No statistically significant differences were observed in the efficacy end points at week 26 (P = 0.410 for overall treatment effect). However, atacicept significantly reduced immunoglobulin and rheumatoid factor (RF) levels, but not anti-citrullinated protein antibody levels, in a dose-dependent manner, with levels returning toward baseline values during followup. The effects of treatment on IgG-RF and IgA-RF were more pronounced than the effects on total IgG and IgA. Adverse events (AEs), including serious AEs, leading to withdrawal were more common among patients treated with atacicept compared with placebo. AEs were variable in nature, and no dose-dependent trends were observed. The frequency of infection-related AEs was similar across treatments. No notable effect of treatment on immunization status (protective versus nonprotective titer) was observed after initiation of treatment. CONCLUSION This study did not meet the primary efficacy end point. However, clear biologic activity consistent with the proposed mechanism of action was observed. The results suggest that decreasing the expression of RF may not be sufficient to induce clinical improvement in RA. The safety of atacicept was considered acceptable in this patient population.
Collapse
Affiliation(s)
- M C Genovese
- Stanford University, Palo Alto, California, USA.
| | | | | | | | | |
Collapse
|
16
|
Abstract
Multiple sclerosis (MS) has traditionally been considered to be a T-cell-mediated disease. However, there is an increasing body of evidence for the involvement of B cells and autoantibodies in the pathology of this disease, providing a rationale for treatments directed against B cells. In this paper we summarize evidence for the key role of B cells in the immunopathology of MS and review data supporting the use of a novel B-cell targeted therapy, atacicept, in this condition. Atacicept is a human recombinant fusion protein that comprises the binding portion of a receptor for both BLyS (B-Lymphocyte Stimulator) and APRIL (A PRoliferation-Inducing Ligand), two cytokines that have been identified as important regulators of B-cell maturation, function and survival. Atacicept has shown selective effects on cells of the B-cell lineage, acting on mature B cells and blocking plasma cells and late stages of B-cell development while sparing B-cell progenitors and memory cells. The efficacy of atacicept in animal models of autoimmune disease and the biological activity of atacicept in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) has been demonstrated. Clinical studies were initiated to investigate the safety, tolerability and efficacy of atacicept in patients with MS. An unexpected increase in inflammatory activity in one of the trials, however, led to suspension of all atacicept trials in MS.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | | |
Collapse
|
17
|
van Vollenhoven RF, Kinnman N, Vincent E, Wax S, Bathon J. Atacicept in patients with rheumatoid arthritis and an inadequate response to methotrexate: Results of a phase II, randomized, placebo-controlled trial. ACTA ACUST UNITED AC 2011; 63:1782-92. [DOI: 10.1002/art.30372] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
18
|
Development and characterization of APRIL antagonistic monoclonal antibodies for treatment of B-cell lymphomas. Blood 2011; 117:6856-65. [PMID: 21543761 DOI: 10.1182/blood-2011-01-330852] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
APRIL (A proliferation-inducing ligand) is a TNF family member that binds two TNF receptor family members, TACI and BCMA. It shares these receptors with the closely related TNF family member, B-cell activating factor (BAFF). Contrary to BAFF, APRIL binds heparan sulfate proteoglycans (HSPGs), which regulates cross-linking of APRIL and efficient signaling. APRIL was originally identified as a growth promoter of solid tumors, and more recent evidence defines APRIL also as an important survival factor in several human B-cell malignancies, such as chronic lymphocytic leukemia (CLL). To target APRIL therapeutically, we developed two anti-human APRIL antibodies (hAPRIL.01A and hAPRIL.03A) that block APRIL binding to BCMA and TACI. Their antagonistic properties are unique when compared with a series of commercially available monoclonal anti-human APRIL antibodies as they prevent in vitro proliferation and IgA production of APRIL-reactive B cells. In addition, they effectively impair the CLL-like phenotype of aging APRIL transgenic mice and, more importantly, block APRIL binding to human B-cell lymphomas and prevent the survival effect induced by APRIL. We therefore conclude that these antibodies have potential for further development as therapeutics to target APRIL-dependent survival in B-cell malignancies.
Collapse
|
19
|
Engel P, Gómez-Puerta JA, Ramos-Casals M, Lozano F, Bosch X. Therapeutic targeting of B cells for rheumatic autoimmune diseases. Pharmacol Rev 2011; 63:127-56. [PMID: 21245206 DOI: 10.1124/pr.109.002006] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Autoreactive B cells are characterized by their ability to secrete autoantibodies directed against self-peptides. During the last decade, it has become increasingly apparent that B lymphocytes not only produce autoantibodies but also exert important regulatory roles independent of their function as antibody-producing cells. This is especially relevant in the context of autoimmunity, because autoreactive B cells have been shown to possess the ability to activate pathogenic T cells, to produce pro-inflammatory cytokines, and to promote the formation of tertiary lymphoid tissue in target organs. The production of monoclonal antibodies against B-cell-surface molecules has facilitated the characterization of several distinct B lymphocyte subsets. These cell-surface molecules have not only served as useful cell differentiation markers but have also helped to unravel the important biological functions of these cells. Some of these molecules, all of which are expressed on the cell surface, have proven to be effective therapeutic targets. In both animal models and in clinical assays, the efficient elimination of B lymphocytes has been shown to be useful in the treatment of rheumatoid arthritis and other autoimmune diseases. The treatment of most rheumatic autoimmune diseases relies mainly on the use of cytotoxic immunosuppressants and corticosteroids. Although this has resulted in improved disease survival, patients may nonetheless suffer severe adverse events and, in some cases, their relapse rate remains high. The increasing need for safer and more effective drugs along with burgeoning new insights into the pathogenesis of these disorders has fueled interest in biological agents; clinical trials involving the B-cell depletion agent rituximab have been especially promising. This article reviews the current knowledge of B-cell biology and pathogenesis as well as the modern therapeutic approaches for rheumatic autoimmune diseases focusing in particular on the targeting of B-cell-specific surface molecules and on the blocking of B-cell activation and survival.
Collapse
Affiliation(s)
- Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
20
|
Abstract
This overview of 25 monoclonal antibody (mAb) and 5 Fc fusion protein therapeutics provides brief descriptions of the candidates, recently published clinical study results and on-going Phase 3 studies. In alphanumeric order, the 2011 therapeutic antibodies to watch list comprises AIN-457, bapineuzumab, brentuximab vedotin, briakinumab, dalotuzumab, epratuzumab, farletuzumab, girentuximab (WX-G250), naptumomab estafenatox, necitumumab, obinutuzumab, otelixizumab, pagibaximab, pertuzumab, ramucirumab, REGN88, reslizumab, solanezumab, T1h , teplizumab, trastuzumab emtansine, tremelimumab, vedolizumab, zalutumumab and zanolimumab. In alphanumeric order, the 2011 Fc fusion protein therapeutics to watch list comprises aflibercept, AMG-386, atacicept, Factor VIII and Factor IX-Fc. Commercially-sponsored mAb and Fc fusion therapeutics that have progressed only as far as Phase 2/3 or 3 were included. Candidates undergoing regulatory review or products that have been approved may also be in Phase 3 studies, but these were excluded. Due to the large body of primary literature about the candidates, only selected references are given and results from recent publications and articles that were relevant to Phase 3 studies are emphasized. Current as of September 2010, the information presented here will serve as a baseline against which future progress in the development of antibody-based therapeutics can be measured.
Collapse
|
21
|
Leishman AJ, Sims GP, Sleeman M, Braddock M. Emerging small molecule and biological therapeutic approaches for the treatment of autoimmunity. Expert Opin Investig Drugs 2010; 20:23-39. [PMID: 21118058 DOI: 10.1517/13543784.2011.540569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
IMPORTANCE OF THE FIELD Biological therapeutics targeting TNF-α, IL-6, CD20 and CD80/86 is proving to be an important weapon in the clinicians' armory to fight autoimmunity alongside long-standing small molecule therapeutics such as methotrexate and glucocorticoids. However, there still remains a high unmet clinical need in the field of autoimmunity and many researchers are continuing to discover and develop new therapeutics to address this. AREAS COVERED IN THIS REVIEW A new wave of small molecule and biological therapeutics targeting different pathways is being developed which could generate exciting new options for clinicians. This review aims to highlight those emerging therapies that are most advanced in clinical development. WHAT THE READER WILL GAIN The reader will gain an appreciation of new approaches being developed to address the high unmet clinical need in the field of autoimmunity. TAKE HOME MESSAGE Despite recent success in the development of therapeutics to treat autoimmunity, new therapeutic strategies are being developed to address the remaining areas of a high unmet clinical need.
Collapse
Affiliation(s)
- Andrew J Leishman
- AstraZeneca R&D Charnwood, Bioscience Department, Loughborough, LE11 5RH, UK.
| | | | | | | |
Collapse
|
22
|
Hall J, Dennler P, Haller S, Pratsinis A, Säuberli K, Towbin H, Walther K, Woytschak J. Genomics drugs in clinical trials. Nat Rev Drug Discov 2010; 9:988. [DOI: 10.1038/nrd1552-c1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|