Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
RMD Open ; 10(1)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38316489

ABSTRACT

OBJECTIVE: To evaluate the efficacy, immunogenicity and safety of the proposed biosimilar MSB11456 versus European Union (EU)-approved tocilizumab reference product in patients with rheumatoid arthritis (RA) in a multicentre, randomised, double-blind, multinational, parallel-group study (NCT04512001). METHODS: Adult patients with moderate-to-severe active RA and inadequate clinical response to ≥1 disease-modifying antirheumatic drug (synthetic or biologic) receiving methotrexate were randomised to receive 24 weekly subcutaneous 162 mg injections of either MSB11456 or EU-approved tocilizumab. Equivalence between treatments was considered if the 95% CI (European Medicines Agency)/90% CI (US Food and Drug Administration) for the difference in mean change from baseline to week 24 in Disease Activity Score-28 Joint Count with erythrocyte sedimentation rate (DAS28-ESR) between treatments was entirely within prespecified equivalence intervals (-0.6 to 0.6 and -0.6 to 0.5, respectively). At week 24, patients were rerandomised to continued treatment or MSB11456. Secondary efficacy endpoints to week 52, and safety and immunogenicity to week 55 were also evaluated. RESULTS: At week 24, the least squares mean difference in the change from baseline in DAS28-ESR between treatments was 0.01 (95% CI -0.19 to 0.22) in the 604 randomised patients. Similarity between treatments was shown for all other efficacy, safety and immunogenicity endpoints, including in patients who switched from EU-approved tocilizumab to MSB114466. CONCLUSIONS: Therapeutic equivalence was demonstrated for efficacy endpoints, and safety and immunogenicity analyses support the similarity of the two treatments. The results of this study strengthen the evidence that the proposed biosimilar MSB11456 and EU-approved tocilizumab exert similar clinical effects.


Subject(s)
Antibodies, Monoclonal, Humanized , Antirheumatic Agents , Arthritis, Rheumatoid , Biosimilar Pharmaceuticals , United States , Adult , Humans , Biosimilar Pharmaceuticals/adverse effects , Double-Blind Method , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Antirheumatic Agents/adverse effects
2.
Expert Rev Clin Immunol ; 19(4): 447-455, 2023 04.
Article in English | MEDLINE | ID: mdl-36789991

ABSTRACT

BACKGROUND: Tocilizumab is a monoclonal immunoglobulin G interleukin-6 receptor antagonist. MSB11456 is a proposed tocilizumab biosimilar. OBJECTIVE: To determine the pharmacokinetic equivalence of a single subcutaneous injection of MSB11456, when delivered via autoinjector (AI) and prefilled syringe (PFS), in healthy adult subjects. RESEARCH DESIGN AND METHODS: In this randomized, open-label, single fixed-dose, crossover study, 91 subjects received subcutaneous administration of tocilizumab 162 mg via AI and PFS presentations. The primary endpoint pharmacokinetic parameters were analyzed using analysis of variance. Safety data were summarized descriptively. RESULTS: There were no differences in pharmacokinetic parameters between presentations, and safety parameters were comparable. The 90% confidence intervals for the geometric least squares mean ratios of all primary pharmacokinetic parameters were contained within the predefined 80.00% to 125.00% bioequivalence limits, indicating pharmacokinetic equivalence between the AI and PFS. CONCLUSIONS: MSB11456 administration via AI was bioequivalent to administration via PFS. MSB11456 can be administered by AI or PFS, increasing the available range of self-injection devices. TRIAL REGISTRATION: The trial is registered at EudraCT, number 2020-003419-86.


Tocilizumab is a biologic drug that is used to treat autoimmune diseases, including rheumatoid arthritis. MSB11456 has been shown to be equivalent to the US-licensed and EU-approved tocilizumab when administered by subcutaneous injection. There are different devices available to administer subcutaneous injections, and depending on the device, the patient's experience can be enhanced, convenience and compliance increased, and cost-effectiveness ensured for patients taking this medicine. This randomized, single fixed-dose, crossover study tested the pharmacokinetic similarity of MSB11456 when given subcutaneously via an auto-injector device versus a pre-filled syringe device in 100 healthy subjects. A total of 91 healthy volunteers received MSB11456 via both auto-injector and pre-filled syringe using a crossover design. Blood was collected before the first dose and at regular intervals during the study to determine the pharmacokinetics of tocilizumab and ensure safety. This study found that the pharmacokinetics of tocilizumab following administration using the autoinjector and the prefilled syringe were equivalent, and the safety profiles were similar. These findings indicate that the auto-injector can be considered another option that can be used to subcutaneously inject MSB11456.


Subject(s)
Antibodies, Monoclonal, Humanized , Syringes , Adult , Humans , Cross-Over Studies , Antibodies, Monoclonal, Humanized/pharmacokinetics , Therapeutic Equivalency , Injections, Subcutaneous
3.
Expert Rev Clin Immunol ; 19(4): 439-446, 2023 04.
Article in English | MEDLINE | ID: mdl-36715299

ABSTRACT

BACKGROUND: Tocilizumab, a recombinant monoclonal immunoglobulin G, targets the interleukin-6 receptor. MSB11456 is a proposed tocilizumab biosimilar. OBJECTIVES: To assess pharmacokinetic equivalence of intravenous MSB11456 to US-licensed tocilizumab. RESEARCH DESIGN AND METHODS: In this double-blind, parallel-group, single-dose study, 128 healthy adults were randomized to a single one-hour 8 mg/kg IV infusion of either MSB11456 or US-licensed tocilizumab. Blood samples were collected pre-dose and at regular intervals up to day 48 post-dose. The primary endpoint pharmacokinetic parameter was analyzed using analysis of variance (ANOVA) model on the natural logarithm of the endpoint (AUC0-last), with treatment as a fixed effect. Immunogenicity and safety data were summarized descriptively. RESULTS: Subjects received either MSB11456 (N = 62) or US-licensed tocilizumab (N = 66). Pharmacokinetic bioequivalence, defined as 90% confidence intervals for the geometric least squares mean ratio entirely contained within the 80.00% to 125.00% equivalence limits, was demonstrated between MSB11456 and US-licensed tocilizumab for the primary and secondary pharmacokinetic endpoints. Anti-drug antibody responses, frequency of neutralizing antibodies against tocilizumab, and safety profiles showed no notable between-treatment differences. Safety was comparable between treatments. CONCLUSIONS: Pharmacokinetic similarity of MSB11456 and US-licensed tocilizumab was demonstrated, with comparable immunogenicity and safety profiles, supporting MSB11455 as a biosimilar to US-licensed tocilizumab. The trial is registered at EudraCT, number 2019-003484-22.


Tocilizumab is a biologic drug that is prescribed for autoimmune conditions such as rheumatoid arthritis in adults and arthritis in children where the cause is unknown. Because of the high cost of biologic drugs, alternate similar drugs are being designed and tested to ensure that they are as effective and as safe as drugs that are currently available. These new drugs are called biosimilars. MSB11456 is a proposed tocilizumab biosimilar. Our study tested how the pharmacokinetics, immunogenicity, and safety of intravenously administered MSB11456 compared to that of the already approved tocilizumab drug marketed in the US (US-licensed tocilizumab). One hundred and twenty-eight healthy adult volunteers received a one-hour 8 mg/kg intravenous infusion of either MSB11456 or US-licensed tocilizumab in this randomized, double-blind, parallel-group, single-dose study. Blood samples were taken before and at scheduled times during the study, up to 48 days after the first dose for analysis. In this study, we showed that the pharmacokinetics of MSB11456 were equivalent to the US-licensed tocilizumab. The safety and immune response to the drugs were also similar. These findings indicate that MSB11456 can be considered a biosimilar to tocilizumab. Biosimilars can reduce the cost of drugs by increasing competition and improve access to these, generally expensive, treatment options.


Subject(s)
Biosimilar Pharmaceuticals , Adult , Humans , Area Under Curve , Healthy Volunteers , Antibodies, Monoclonal, Humanized , Therapeutic Equivalency , Double-Blind Method
4.
Expert Rev Clin Immunol ; 18(5): 533-543, 2022 05.
Article in English | MEDLINE | ID: mdl-35354411

ABSTRACT

BACKGROUND: Tocilizumab is a recombinant humanized monoclonal immunoglobulin G1 antibody against the interleukin-6 receptor (IL-6 R). MSB11456 is a proposed tocilizumab biosimilar. OBJECTIVES: To assess the pharmacokinetic and pharmacodynamic similarity of MSB11456 to both US-licensed and EU-approved tocilizumab. METHODS: Healthy adult volunteers (N = 685) received a single 162 mg subcutaneous injection of MSB11456, US-licensed tocilizumab, or EU-approved tocilizumab in this randomized, double-blind, parallel-group study. Blood samples were taken pre-dose and for up to 48 days post-dose. Primary endpoint pharmacokinetic parameters were analyzed using analysis of covariance. Secondary pharmacodynamic measures included serum-soluble IL-6 R and serum C-reactive protein. Safety data were analyzed descriptively. RESULTS: Pharmacokinetic equivalence (with all corresponding 90% confidence intervals for the geometric least squares mean ratios within the predefined 80.00% to 125.00% equivalence margin) was demonstrated between MSB11456 and both US-licensed and EU-approved tocilizumab, as well as between the reference products. Pharmacodynamic analyses demonstrated similarity of MSB11456 and both US-licensed and EU-approved tocilizumab, as well as between the reference products. Safety, tolerability, and immunogenicity were comparable between treatments. CONCLUSION: Pharmacokinetic and pharmacodynamic similarity of MSB11456, US-licensed tocilizumab, and EU-approved tocilizumab were demonstrated, and the three products had comparable immunogenicity and safety, supporting MSB11456 as a biosimilar to tocilizumab.


Tocilizumab is a biologic drug that is used to treat autoimmune diseases, including rheumatoid arthritis. Biologic drugs are very important for the treatment of autoimmune diseases, but their costs limit accessibility. Therefore, the availability of biosimilars, which are biologics that are very similar in structure and function to an existing biologic drug, may provide a significant cost advantage for national healthcare programs and consumers. MSB11456 is a proposed tocilizumab biosimilar. Our study tested the pharmacokinetic and pharmacodynamic similarity of MSB11456 to the approved formulations of tocilizumab in the US and EU (US-licensed and EU-approved tocilizumab) in a large group of healthy adults. Volunteers received a single 162 mg subcutaneous injection of MSB11456, US-licensed tocilizumab, or EU-approved tocilizumab in this randomized, double-blind, parallel-group study. Blood samples were taken before and regularly after the injection, and safety was monitored. We showed that the pharmacokinetics and pharmacodynamics of MSB11456, US-licensed and EU-approved tocilizumab were sufficiently similar to claim equivalence between the three products. Safety and immunogenicity were also comparable between the three treatments. These findings suggest that MSB11456 can be considered as a biosimilar to tocilizumab. Biosimilars have improved price competition and led to a reduction in the net costs of biologics, so tocilizumab biosimilars can be expected to contribute to this and potentially improve access to the best available care.


Subject(s)
Biosimilar Pharmaceuticals , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Area Under Curve , Biosimilar Pharmaceuticals/pharmacokinetics , Biosimilar Pharmaceuticals/therapeutic use , Double-Blind Method , Healthy Volunteers , Humans , Interleukin-6 , Therapeutic Equivalency
5.
Bioanalysis ; 11(9): 875-886, 2019 May.
Article in English | MEDLINE | ID: mdl-31070047

ABSTRACT

Aim: Novel bifunctional VEGF-A neutralizing therapies are being developed for the treatment of retinal vascular diseases such as age-related macular degeneration and diabetic retinopathy. In developing new therapeutic drugs, only small aqueous humor sample volumes are available for analyzing several parameters. Highly sensitive detection methods must be applied in analyzing VEGF-A levels in ocular fluids in order to demonstrate VEGF-A suppression following drug administration. Experimental: A highly sensitive immunoassay for VEGF-A was developed on the single molecule array (Simoa) platform, and validated before being used for the analysis of clinical aqueous humor samples from patients treated with anti-VEGF-A therapeutics. Results: This highly sensitive immunoassay allows the detection of baseline VEGF-A levels and suppression effects after drug administration, even in sample volumes as low as 12 µl. Conclusion: The Simoa VEGF-A assay is a valuable tool for the reliable monitoring of VEGF-A suppression after intravitreal administration of anti-VEGF-A drugs.


Subject(s)
Aqueous Humor/chemistry , Immunoassay/methods , Limit of Detection , Vascular Endothelial Growth Factor A/analysis , Calibration , Diabetes Complications/drug therapy , Humans , Macular Edema/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors
6.
Bioanalysis ; 6(24): 3355-68, 2014.
Article in English | MEDLINE | ID: mdl-25534792

ABSTRACT

The 2014 8th Workshop on Recent Issues in Bioanalysis (8th WRIB), a 5-day full immersion in the evolving field of bioanalysis, took place in Universal City, California, USA. Close to 500 professionals from pharmaceutical and biopharmaceutical companies, contract research organizations and regulatory agencies worldwide convened to share, review, discuss and agree on approaches to address current issues of interest in bioanalysis. The topics covered included both small and large molecules, and involved LCMS, hybrid LBA/LCMS, LBA approaches and immunogenicity. From the prolific discussions held during the workshop, specific recommendations are presented in this 2014 White Paper. As with the previous years' editions, this paper acts as a practical tool to help the bioanalytical community continue advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2014 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations for Large molecules bioanalysis using LBA and Immunogenicity. Part 1 (Small molecules bioanalysis using LCMS) and Part 2 (Hybrid LBA/LCMS, Electronic Laboratory Notebook and Regulatory Agencies' Input) were published in the Bioanalysis issues 6(22) and 6(23), respectively.


Subject(s)
Chemistry Techniques, Analytical , Immunity , Antibodies, Neutralizing/immunology , Biotransformation , Humans , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Polyethylene/chemistry , Practice Guidelines as Topic , United States , United States Food and Drug Administration
7.
J Immunol Methods ; 314(1-2): 67-73, 2006 Jul 31.
Article in English | MEDLINE | ID: mdl-16824538

ABSTRACT

Endotoxins are frequent contaminants of recombinant proteins produced in Escherichia coli. Due to their adverse effects, endotoxins have to be removed from recombinant proteins prior their use in cell-based assays or parenteral application. Reduction of endotoxin to less than 10 EU mg(-1) is, however, one of the most problematic steps during protein purification from E. coli and often associated with substantial loss of biological materials. The present paper describes the use of a single step procedure enabling metal chelate affinity purification and endotoxin clearance from antibody fragments produced in E. coli using a non-ionic detergent. Endotoxin content was as low as 5 to 9 EU mg(-1) with a recovery of antibody fragments of over 90%. Non-ionic detergent treatment did not compromise integrity and functionality of these multimeric molecules. Furthermore, recombinant antibody fragments did not stimulate endotoxin-sensitive cell lines confirming the low endotoxin content. In conclusion, this one-step protocol is a rapid, cost effective and automation-compatible procedure suitable for recombinant antibody fragments.


Subject(s)
Chromatography, Affinity/methods , Endotoxins/chemistry , Immunoglobulin Fragments/chemistry , Metals , Antibodies, Monoclonal/chemistry , Binding Sites, Antibody , Cell Line, Tumor , Detergents/chemistry , Dimerization , Endothelial Cells/physiology , Endotoxins/isolation & purification , Escherichia coli , Humans , Intercellular Adhesion Molecule-1/immunology , Models, Molecular , Octoxynol , Polyethylene Glycols/chemistry
8.
Eur J Immunol ; 32(5): 1300-8, 2002 05.
Article in English | MEDLINE | ID: mdl-11981817

ABSTRACT

Ataxia-Telangiectasia (A-T) and Nijmegen breakage syndrome (NBS) are recessive genetic diseases with similar cellular phenotypes that are caused by mutations in the recently described ATM (encoding ATM) and NBS1 (encoding p95) genes, respectively. Both disorders are accompanied by immunodeficiency in a majority of patients, but the mechanism involved has as yet not been established. We demonstrate that in cells from A-T patients, the switch (S) recombination junctions are aberrant and characterized by a strong dependence on short sequence homologies and devoid of normally occurring mutations around the breakpoint. A low number of S fragments were generated in cells from NBS patients and showed only limited dependence on sequence identity and mutation frequencies were similar to those observed in normal controls. We propose that ATM and p95 are both involved in the final step(s) in class switch recombination with related, but disparate, functional roles. Thus, the general pathway involved in DNA repair also has a major influence on the immunoglobulin isotype switching process.


Subject(s)
Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/immunology , Immunoglobulin Switch Region/genetics , Nuclear Proteins , Recombination, Genetic , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/immunology , Case-Control Studies , Cell Cycle Proteins/genetics , DNA-Binding Proteins , Humans , IgA Deficiency/genetics , IgA Deficiency/immunology , Immunoglobulin A/blood , Immunoglobulin A/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins
9.
Proteomics ; 2(3): 241-6, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11921439

ABSTRACT

Proteins are the molecules that fulfil most cellular functions and represent over 90% of drug targets in the market. Chromophore-assisted laser inactivation (CALI) provides a timely and locally restricted protein inactivation and has proven to specifically destroy protein function using dye-coupled ligands and laser irradiation. CALI involves the generation of short-lived radicals thus limiting the radius of covalent modifications to spatially restricted sites on the target molecule. A transient functional inactivation occurs if the radicals modify amino acids of the target protein that are responsible for function. Here we show specific inactivation of several protein targets, that are members of relevant signal transduction pathways. For each of these targets, simple and high throughput screening-scaleable assays have been developed, making it possible to quantify the observed inactivation. Activities of target proteins have been addressed in cell-free as well as cell-based assays employing human primary and tumor-derived cell lines. In all cases, at least 50% inactivation was achieved. The data presented here demonstrate that CALI is a highly versatile tool for validating disease relevant targets at the protein level. This approach also takes into account post-translational modifications like phosphorylation, glycosylation or acylation, thereby enlarging its applicability for many different types of targets.


Subject(s)
Lasers , Proteins/chemistry , Proteome , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Cell Survival , Cell-Free System , Cells, Cultured , Humans , Indicators and Reagents , Ligands , Mitogen-Activated Protein Kinases/metabolism , Proteins/metabolism , Signal Transduction/physiology , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...