Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Immunol Methods ; 498: 113147, 2021 11.
Article in English | MEDLINE | ID: mdl-34508774

ABSTRACT

Immunogenicity is one major challenge to the successful development of biotherapeutics because it could adversely affect PK/PD, safety, and efficacy. Preclinical immunogenicity risk assessment strategies and assays have been developed and implemented to screen and optimize discovery molecules. Internalization by antigen presenting cells (APC) and innate immune activation are initial prerequisite steps in eliciting immune responses to biotherapeutics. Dendritic cells (DC)- and monocyte-based assays are employed to interrogate such risks, and their value has been well documented in the literature. However, these assays have limited throughput, exhibit higher variability, and entail lengthy and complex procedures as they are based on primary cells such as peripheral blood mononuclear cells (PBMC) from individual donors. Herein, we investigated THP1 cells as surrogate cells to study APC internalization and innate immune activation. Comparability studies showed that THP1 cells could resemble innate immune responses of monocyte-derived DC and primary CD14+ monocytes using a panel of therapeutic antibodies. In addition, an automated high throughput THP1 internalization assay was qualified to enable risk assessment at pre­lead stages. The results demonstrated that THP1 cells can be utilized to assess immunogenicity risk in a high throughput manner.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Monocytes/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Antigen Presentation , Automation, Laboratory , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis , Flow Cytometry , High-Throughput Screening Assays , Humans , Infliximab/pharmacology , Monocytes/immunology , Monocytes/metabolism , THP-1 Cells
2.
MAbs ; 12(1): 1764829, 2020.
Article in English | MEDLINE | ID: mdl-32370596

ABSTRACT

Biologics have the potential to induce an immune response when used therapeutically. A number of in vitro assays are currently used preclinically to predict the risk of immunogenicity, but the validation of these preclinical tools suffers from the relatively small number of accessible immunogenic molecules and the limited understanding of the mechanisms underlying the immunogenicity of biologics. Here, we present the post-hoc analysis of three monoclonal antibodies with high immunogenicity in the clinic. Two of the three antibodies elicited a CD4 T cell proliferative response in multiple donors in a peripheral blood mononuclear cell assay, but required different experimental conditions to induce these responses. The third antibody did not trigger any T cell response in this assay. These distinct capacities to promote CD4 T cell responses in vitro were mirrored by different capacities to stimulate innate immune cells. Only one of the three antibodies was capable of inducing human dendritic cell (DC) maturation; the second antibody promoted monocyte activation while the third one did not induce any innate cell activation in vitro. All three antibodies exhibited a moderate to high internalization by human DCs and MHC-associated peptide proteomics analysis revealed the presence of potential T cell epitopes that were confirmed by a T-cell proliferation assay. Collectively, these findings highlight the existence of distinct immune stimulatory mechanisms for immunogenic antibodies. These findings have implications for the preclinical immunogenicity risk assessment of biologics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Formation/immunology , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Antibodies, Monoclonal/pharmacology , Antibody Formation/drug effects , Antigen Presentation/drug effects , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects
3.
AAPS J ; 22(3): 68, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32300899

ABSTRACT

Treatment-emergent antidrug antibodies (TE-ADA) pose a major challenge to the development of biotherapeutics. The antidrug antibody responses are highly orchestrated and involve many types of immune cells and biological processes. Biological drug internalization and processing by antigen-presenting cells (APCs) are two initial and critical steps in the cascade of events leading to T cell-dependent ADA production. The assays thus far described in literature to evaluate immunogenicity potential/risk as a function of APC activity mainly focus on internalization of labeled drug candidates in vitro. Herein, we describe a high-throughput Förster Resonance Energy Transfer (FRET)-based assay for assessing both internalization and processing using CD14+ monocyte-derived dendritic cells (DCs) as APCs. Antigen-binding fragment F(ab')2 against IgG fragment crystallizable gamma (Fcγ) was labeled with the activatable FRET pair TAMRA-QSY7 as a universal probe for antibodies and proteins with a fragment crystallizable (Fc) domain. The assay was qualified using six mAbs of known clinical immunogenicity and one IgG1 isotype antibody using Design of Experiment (DoE). Correlation analysis of internalization and clinical immunogenicity data showed that this FRET-based internalization assay was able to detect clinically immunogenic antibodies. This method provides a tool for analyzing/screening the immunogenicity risk of biological candidates by assessing one of the critical components of the ADA formation process within the broader context of an immunogenicity risk assessment strategy.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Formation , Dendritic Cells/physiology , Immunogenetic Phenomena , Molecular Probes , Fluorescence Resonance Energy Transfer , Lysosomes/metabolism , Risk Assessment
4.
J Clin Pharmacol ; 58(3): 340-346, 2018 03.
Article in English | MEDLINE | ID: mdl-29106714

ABSTRACT

Etanercept has been recently approved in the United States for the treatment of moderate to severe plaque psoriasis in patients aged 4-17 years. The objective of this study was to characterize etanercept pharmacokinetics, immunogenicity, and efficacy in pediatric patients. Data from a phase 3 study and open-label extension study were analyzed. Etanercept serum concentrations in pediatric patients receiving etanercept 0.8 mg/kg (maximum, 50 mg) weekly were compared with adult psoriasis patients and pediatric patients with juvenile idiopathic arthritis (JIA) who received etanercept 0.4 mg/kg twice weekly. The developments of antietanercept antibodies and efficacy based on the Psoriasis Area and Severity Index were evaluated. Steady-state trough etanercept concentrations were similar across visits from weeks 12-48, between patients aged 4-11 and 12-17 years, between pediatric and adult psoriasis patients, and between pediatric patients with psoriasis or JIA. Etanercept serum concentrations and safety profiles were similar in patients with (15.9%) and without antietanercept antibodies. Dosing used in the study provided similar exposures and efficacy across ranges of weight and body mass index. Pharmacokinetic, immunogenicity, and efficacy data support 0.8 mg/kg (maximum, 50 mg) weekly dosing of etanercept in patients aged 4-17 years.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arthritis, Juvenile/drug therapy , Arthritis, Juvenile/immunology , Etanercept/immunology , Etanercept/pharmacokinetics , Psoriasis/drug therapy , Psoriasis/immunology , Adolescent , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies/immunology , Child , Child, Preschool , Double-Blind Method , Drug Administration Schedule , Etanercept/therapeutic use , Female , Humans , Male , Severity of Illness Index , Treatment Outcome
5.
Rheumatol Ther ; 4(2): 391-404, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28900875

ABSTRACT

INTRODUCTION: To evaluate the efficacy and safety of etanercept treatment in adult patients with moderate to severe rheumatoid arthritis (RA) who failed to respond (primary failure) or lost a satisfactory response (secondary failure) to adalimumab. METHODS: All patients discontinued prior adalimumab treatment and continued methotrexate with etanercept 50 mg once weekly for 24 weeks. The primary study endpoint was American College of Rheumatology 20% improvement criteria (ACR20) at week 12. RESULTS: Eighty-five patients (mean age 56.6 years; female 80.0%) were evaluated for safety and 84 for efficacy. Thirty (35.7%) patients achieved ACR20 at week 12; the lower bound of the 95% confidence interval (CI; 25.6, 46.9) was greater than the prespecified goal of 24% based on previous research. Improvements from baseline in clinical outcomes and patient-reported outcomes were observed at each study visit. In planned subgroup analyses, patients with anti-adalimumab antibodies and secondary adalimumab failure had the highest ACR20 response to etanercept at week 12 (11/17 patients; 64.7%). Among the patients with secondary adalimumab failure, those with anti-adalimumab antibodies were fivefold more likely to have an ACR20 response to etanercept than those without anti-adalimumab antibodies (odds ratio 5.2; 95% CI 2.0, 13.5; P < 0.001). Adverse events were reported for 62 (72.9%) patients and were consistent with previous studies of etanercept. Most adverse events were mild or moderate in severity. CONCLUSION: Switching to etanercept is a therapeutic option in patients with RA who fail adalimumab treatment. The presence of anti-adalimumab antibodies may provide additional support for switching to etanercept, particularly in patients with secondary adalimumab failure. TRIAL REGISTRATION: ClinicalTrials.gov identifier, NCT01927757.

6.
AAPS J ; 19(3): 599-602, 2017 05.
Article in English | MEDLINE | ID: mdl-28247192

ABSTRACT

With more than 100 therapeutic proteins (TP) approved since the first EMA guidance on immunogenicity in 2007, a vast amount of clinical experience with a variety of therapeutic proteins has been gained. This has provided data on anti-drug antibodies (ADA) and their observed clinical impact, or lack thereof. It has become evident that not all ADA responses are clinically relevant. The current "standard practice" is to test for ADA in all patients on every study. It is essential that we acknowledge the immunogenicity data gained from marketed TPs and that options for immunogenicity testing reflect this information. Improvements in bioanalytical support throughout the drug development process will eliminate extraneous, non-impactful practices. We propose that low-risk therapeutic proteins could be supported with an event-driven ("collect-and-hold") immunogenicity testing strategy throughout early phases of the clinical program. In the absence of an event, only pivotal studies (where ADA incidence and impact can be decisively assessed) would include default ADA testing. In keeping with the "standard practice," immunogenicity risk assessment must be an on-going and real-time evaluation. This approach has the potential to deliver meaningful, clinically relevant immunogenicity results while maintaining an emphasis on patient safety.


Subject(s)
Drug Evaluation/methods , Immunity, Active , Proteins/therapeutic use , Clinical Trials as Topic , Humans , Proteins/immunology
7.
Ann Rheum Dis ; 76(3): 526-533, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27466231

ABSTRACT

OBJECTIVE: To demonstrate pharmacokinetic (PK) similarity of biosimilar candidate ABP 501 relative to adalimumab reference product from the USA and European Union (EU) and evaluate safety, tolerability and immunogenicity of ABP 501. METHODS: Randomised, single-blind, single-dose, three-arm, parallel-group study; healthy subjects were randomised to receive ABP 501 (n=67), adalimumab (USA) (n=69) or adalimumab (EU) (n=67) 40 mg subcutaneously. Primary end points were area under the serum concentration-time curve from time 0 extrapolated to infinity (AUCinf) and the maximum observed concentration (Cmax). Secondary end points included safety and immunogenicity. RESULTS: AUCinf and Cmax were similar across the three groups. Geometrical mean ratio (GMR) of AUCinf was 1.11 between ABP 501 and adalimumab (USA), and 1.04 between ABP 501 and adalimumab (EU). GMR of Cmax was 1.04 between ABP 501 and adalimumab (USA) and 0.96 between ABP 501 and adalimumab (EU). The 90% CIs for the GMRs of AUCinf and Cmax were within the prespecified standard PK equivalence criteria of 0.80 to 1.25. Treatment-related adverse events were mild to moderate and were reported for 35.8%, 24.6% and 41.8% of subjects in the ABP 501, adalimumab (USA) and adalimumab (EU) groups; incidence of antidrug antibodies (ADAbs) was similar among the study groups. CONCLUSIONS: Results of this study demonstrated PK similarity of ABP 501 with adalimumab (USA) and adalimumab (EU) after a single 40-mg subcutaneous injection. No new safety signals with ABP 501 were identified. The safety and tolerability of ABP 501 was similar to the reference products, and similar ADAb rates were observed across the three groups. TRIAL REGISTRATION NUMBER: EudraCT number 2012-000785-37; Results.


Subject(s)
Adalimumab/pharmacology , Antirheumatic Agents/pharmacokinetics , Biosimilar Pharmaceuticals/pharmacokinetics , Adalimumab/adverse effects , Adalimumab/blood , Adalimumab/immunology , Adolescent , Adult , Antibodies/blood , Antirheumatic Agents/adverse effects , Antirheumatic Agents/blood , Antirheumatic Agents/immunology , Area Under Curve , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/blood , Europe , Female , Healthy Volunteers , Humans , Male , Middle Aged , Single-Blind Method , Therapeutic Equivalency , United States , Young Adult
8.
Bioanalysis ; 8(17): 1845-57, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27523191

ABSTRACT

We recommend the application of a strategically designed step-wise approach to transfer cell-based assays that includes assessing analytical performance (through a fit for purpose validation and/or design of experiment robustness characterization), clinical performance (i.e., concordance) and performance or proficiency testing for long-term method monitoring. Here we focus on the application of this strategy to cell-based assays for the measurement of neutralizing anti-drug antibodies. This application is unique in that it requires a custom cell-based assay to be used over a long period of time (potentially phase 1a through the life of a marketed product) with the confidence of consistent method performance and result reporting. But, the process is adaptable to a variety of assay types and applications. We present lessons learned from two cell-based assay transfers that met relevant challenges while implementing alternative permutations of the recommended method transfer process.


Subject(s)
Antibodies, Neutralizing/analysis , Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods , Animals , Cytokines/analysis , Evaluation Studies as Topic , Humans
9.
Arthritis Res Ther ; 17: 215, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26290435

ABSTRACT

INTRODUCTION: Blisibimod is a potent B cell-activating factor (BAFF) antagonist that binds to both cell membrane-expressed and soluble BAFF. The goal of these first-in-human studies was to characterize the safety, tolerability, and pharmacokinetic and pharmacodynamic profiles of blisibimod in subjects with systemic lupus erythematosus (SLE). METHODS: SLE subjects with mild disease that was stable/inactive at baseline received either a single dose of blisibimod (0.1, 0.3, 1, or 3 mg/kg subcutaneous [SC] or 1, 3, or 6 mg/kg intravenous [IV]) or placebo (phase 1a; N = 54), or four weekly doses of blisibimod (0.3, 1, or 3 mg/kg SC or 6 mg/kg IV) or placebo (phase 1b; N = 63). Safety and tolerability measures were collected, and B cell subset measurements and pharmacokinetic analyses were performed. RESULTS: All subjects (93 % female; mean age 43.7 years) carried the diagnosis of SLE for ≥ 1 year. Single- and multiple-dose treatment with blisibimod produced a decrease in the number of naïve B cells (24-76 %) and a transient relative increase in the memory B cell compartment, with the greatest effect on IgD(-)CD27+; there were no notable changes in T cells or natural killer cells. With time, memory B cells reverted to baseline, leading to a calculated 30 % reduction in total B cells by approximately 160 days after the first dose. In both the single- and multiple-dosing SC cohorts, the pharmacokinetic profile indicated slow absorption, dose-proportional exposure from 0.3 through 3.0 mg/kg SC and 1 through 6 mg/kg IV, linear pharmacokinetics across the dose range of 1.0-6.0 mg/kg, and accumulation ratios ranging from 2.21 to 2.76. The relative increase in memory B cells was not associated with safety signals, and the incidence of adverse events, anti-blisibimod antibodies, and clinical laboratory abnormalities were comparable between blisibimod- and placebo-treated subjects. CONCLUSIONS: Blisibimod changed the constituency of the B cell pool and single and multiple doses of blisibimod exhibited approximate dose-proportional pharmacokinetics across the dose range 1.0-6.0 mg/kg. The safety and tolerability profile of blisibimod in SLE was comparable with that of placebo. These findings support further studies of blisibimod in SLE and other B cell-mediated diseases. TRIAL REGISTRATION: Clinicaltrials.gov NCT02443506 . Registered 11 May 2015. NCT02411136 Registered 7 April 2015.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocyte Subsets/metabolism , Lupus Erythematosus, Systemic/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Adult , Area Under Curve , B-Cell Activating Factor/antagonists & inhibitors , B-Lymphocyte Subsets/drug effects , Dizziness/chemically induced , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Headache/chemically induced , Humans , Injections, Intravenous , Injections, Subcutaneous , Lupus Erythematosus, Systemic/drug therapy , Lymphocyte Count , Male , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Treatment Outcome , Young Adult
10.
J Immunol Methods ; 419: 1-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25795420

ABSTRACT

Anti-drug neutralizing antibodies (NAbs) formed due to unwanted immunogenicity of a therapeutic protein point towards a mature immune response. NAb detection is important in interpreting the therapeutic's efficacy and safety in vivo. In vitro cell-based NAb assays provide a physiological system for NAb detection, however are complex assays. Non-cell-based competitive ligand binding (CLB) approaches are also employed for NAb detection. Instead of cells, CLB assays use soluble receptor and conjugated reagents and are easier to perform, however have reduced physiological relevance. The aim of this study was to compare the performance of CLB assays to established cell-based assays to determine the former's ability to detect clinically relevant NAbs towards therapeutics that (i) acted as an agonist or (ii) acted as antagonists by binding to a target receptor. We performed a head-to-head comparison of the performance of cell-based and CLB NAb assays for erythropoietin (EPO) and two anti-receptor monoclonal antibodies (AMG-X and AMG 317). Clinically relevant NAb-positive samples identified previously by a cell-based assay were assessed in the corresponding CLB format(s). A panel of 12 engineered fully human anti-EPO monoclonal antibodies (MAbs) was tested in both EPO NAb assay formats. Our results showed that the CLB format was (i) capable of detecting human anti-EPO MAbs of differing neutralizing capabilities and affinities and (ii) provided similar results as the cell-based assay for detecting NAbs in patient samples. The cell-based and CLB assays also behaved comparably in detecting NAbs in clinical samples for AMG-X. In the case of anti-AMG 317 NAbs, the CLB format failed to detect NAbs in more than 50% of the tested samples. We conclude that assay sensitivity, drug tolerance and the selected assay matrix played an important role in the inability of AMG 317 CLB assays to detect clinically relevant NAbs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Biological Assay/methods , Erythropoietin/immunology , Immunologic Techniques/methods , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/immunology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Erythropoietin/metabolism , Erythropoietin/pharmacology , Humans , Interleukin-4/metabolism , Interleukin-4/pharmacology , Ligands , Protein Binding/drug effects , Protein Binding/immunology , Receptors, Erythropoietin/metabolism , Receptors, Interleukin-4/metabolism , Reproducibility of Results
11.
Pharmacol Res Perspect ; 3(6): e00199, 2015 Dec.
Article in English | MEDLINE | ID: mdl-27022472

ABSTRACT

Hu714MuXHu is a recombinant chimeric murine-human monoclonal antibody directed against interleukin-15 (IL-15), a proinflammatory cytokine associated with memory CD8+ and natural killer (NK) T-cell activation and implicated in the pathogenesis of inflammatory diseases. A pharmacokinetic-pharmacodynamic (PK/PD) model was developed to describe the NK cell count reduction in cynomolgus monkeys after treatment with Hu714MuXHu. Cynomolgus monkeys were dosed with Hu714MuXHu in three studies: as a single dose at 0.1 or 1 mg·kg(-1) i.v.; weekly for 5 weeks at 0, 30, 60, or 150 mg·kg(-1) i.v. or 150 mg·kg(-1) s.c.; weekly for 13 weeks at 0, 5, 30, or 150 mg·kg(-1) s.c. Serum Hu714MuXHu concentration-time data were analyzed using noncompartmental analysis and the PK/NK cell count relationship was assessed via simultaneous PK/PD modeling. Hu714MuXHu PK was approximately dose-proportional between 0.1-150 mg·kg(-1) for i.v. and 5-150 mg·kg(-1) for s.c. administration with an elimination half-life of 12.7-18 days. Hu714MuXHu administration resulted in rapid and marked reductions in NK cell counts after the first dose which recovered fully after the serum Hu714MuXHu concentrations approached 0.1 µg·mL(-1) (assay limit of quantification). PK/PD modeled Hu714MuXHu effects on NK cells had an EC 50 of 0.09 µg·mL(-1). In summary, weekly i.v. or s.c. doses with Hu714MuXHu for up to 3 months in cynomolgus monkeys demonstrated linear PK and significant NK cell count reduction, which was described using PK/PD modeling. This approach may be used to guide investigative product dose selections for inflammatory diseases where NK cell count alterations are quantifiable.

12.
J Pharm Biomed Anal ; 102: 176-83, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25279937

ABSTRACT

A robust, quantitative method for assessing the neutralizing capacity of anti-therapeutic protein antibodies was developed and tested using 4 analytical assay platforms typically used for detection of anti-drug neutralizing antibodies. The method described here utilized titration of increasing concentrations of therapeutic protein into serum containing anti-therapeutic protein antibodies, either positive control antibodies or clinical samples. Neutralizing capacities were calculated by determining the EC50 from the titration curves. The neutralizing capacity of purified anti therapeutic protein antibodies was expressed in terms of "µg of drug neutralized per µg of anti-TP antibody" present. In the case of serum originating from clinical study subjects, the neutralizing capacity of the samples was expressed as "µg of drug neutralized per mL of serum". A relative shift in EC50 values was observed as the amount of serum or antibody was changed resulting in a proportional shift of the calculated neutralizing capacity. Application of this approach using different assay platforms was consistent providing evidence for its potential to be a useful approach to characterize, qualify and compare neutralizing positive control antibody preparations or clinical samples. Using this methodology, we were able to draw a clear correlation between the neutralizing capacity and the effect of these antibodies on a clinical pharmacodynamic (PD) marker. Determination of the neutralizing capacity of antibody positive samples from subjects in a clinical study indicated direct correlation of pharmacodynamic results with non-response, partial response and response to high, mid and low neutralizing capacities respectively.


Subject(s)
Antibodies, Anti-Idiotypic/analysis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/analysis , Animals , Antibodies, Monoclonal/therapeutic use , Cells, Cultured , Humans , Rabbits
13.
J Pharm Sci ; 102(10): 3545-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23925953

ABSTRACT

We describe a novel human immunoglobulin G2 (IgG2 )-tolerant and immune-competent heterozygous mouse model (Xeno-het) developed by crossbreeding a human Ig-tolerized XenoMouse® with a C57BL/6J wild-type mouse. The Xeno-het mouse expresses both mouse and human immunoglobulin G (IgG) genes, resulting in B-cells expressing human and mouse IgG, and secretion of human and mouse Ig into serum. This model was utilized to evaluate the immunogenicity risk of aggregated and chemically modified human antibodies. The mice were tested for their ability to break tolerance to self-tolerant monomeric antibodies. Aggregates made by mechanical stirring elicited an anti-drug antibody (ADA) response, but did not induce a robust and long-term memory B and T-cell response. Chemically modified antibodies made by oxidation were only weak and transient inducers of an immune response, as measured by a lack of both an ADA response and a B-cell antigen-specific response. Aggregate size was an important characteristic, as specific-sized protein-coated beads were able to elicit an immune response. We propose the use of this model to identify risk factors such as aggregation during manufacturing at early development for an increased potential immunogenicity risk.


Subject(s)
Antibodies/immunology , Antibody Formation/immunology , Biological Factors/immunology , Immune Tolerance/immunology , Animals , B-Lymphocytes/immunology , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
14.
Br J Clin Pharmacol ; 76(2): 188-202, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23731388

ABSTRACT

The aim of the study was to characterize performance of a complementary set of assays to measure antigen-specific immune responses in subjects immunized with a neoantigen. Healthy volunteers (HV) (n = 8) and patients with systemic lupus erythematosus (SLE) (n = 6) were immunized with keyhole limpet haemocyanin (KLH) on days 1 and 29. Serum antibodies were detected using a flow cytometric bead array (CBA) that multiplexed the KLH response alongside pre-existing anti-tetanus antibodies. Peripheral blood mononuclear cells were studied by B cell ELISPOT. These assays were built upon precedent assay development in cynomolgus monkeys, which pointed towards their utility in humans. Primary anti-KLH IgG responses rose to a mean of 65-93-fold above baseline for HV and SLE patients, respectively, and secondary responses rose to a mean of 260-170-fold above baseline. High levels of anti-tetanus IgG were detected in pre-immunization samples and their levels did not change over the course of study. Anti-KLH IgG1-4 subclasses were characterized by a predominant IgG1 response, with no significant differences in subclass magnitude or distribution between HV and SLE subjects. Anti-KLH IgM levels were detectable, although the overall response was lower. IgM was not detected in two SLE subjects whodid generate an IgG response. All subjects responded to KLH by B cell ELISPOT, with no significant differences observed between HV and SLE subjects. The CBA and B cell ELISPOT assays reliably measured anti-KLH B cell responses, supporting use of this approach and these assays to assess the pharmacodynamic and potential safety impact of marketed/investigational immune-therapeutics.


Subject(s)
Adjuvants, Immunologic/administration & dosage , B-Lymphocytes/immunology , Hemocyanins/administration & dosage , Lupus Erythematosus, Systemic/prevention & control , Vaccination , B-Lymphocytes/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hemocyanins/immunology , Humans , Immunoglobulin G/blood , Lupus Erythematosus, Systemic/immunology
15.
Bioanalysis ; 5(9): 1041-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23641695

ABSTRACT

BACKGROUND: Immunogenicity testing is an important component of clinical development for large-molecule biotherapeutics. New complex types of large molecules, such as antibody-drug conjugates (ADCs), require careful evaluation of the testing strategy and bioanalytical assays used to monitor the development of antitherapeutic antibodies. RESULTS: An electrochemiluminescence-based immunoassay for the detection and epitope characterization of anti-ADC antibodies was validated. Using this assay format, antibodies directed against the monoclonal antibody and linker-drug components of the ADC were successfully detected in a multiple-dose rat toxicity study. CONCLUSION: Immunogenicity assays incorporating epitope determination may provide additional information about the characteristics of induced antitherapeutic antibodies, including the magnitude and timing of the various types of antibody responses.


Subject(s)
Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Immunoconjugates/blood , Immunoconjugates/immunology , Immunogenetic Phenomena/immunology , Pharmaceutical Preparations/blood , Animals , Antibodies, Monoclonal/therapeutic use , Antigen-Antibody Reactions , Humans , Immunoconjugates/therapeutic use
16.
Bioanalysis ; 4(22): 2725-35, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23210655

ABSTRACT

The development of an unwanted immune response to a protein therapeutic is a constant concern and necessitates careful monitoring of this class of drugs during clinical development. Neutralizing antibodies can have a significant impact on bioavailability, efficacy and safety of a protein therapeutic. Consequently, immunogenicity testing is required prior to obtaining regulatory approval and in some cases even after a product is marketed. Given the importance of this testing, it is critical that sensitive and robust assays are developed for detection of clinically relevant neutralizing antibodies. This review will describe considerations and current best practices for developing assays to detect neutralizing antibodies to protein therapeutics.


Subject(s)
Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Immunoassay/methods , Proteins/immunology , Animals , Drug Discovery , Humans , Immunity , Proteins/therapeutic use
17.
BMC Clin Pharmacol ; 11: 17, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22070868

ABSTRACT

BACKGROUND: Panitumumab is a fully human antibody against the epidermal growth factor receptor that is indicated for the treatment of metastatic colorectal cancer (mCRC) after disease progression on standard chemotherapy. The purpose of this analysis was to examine the immunogenicity of panitumumab and to evaluate the effect of anti-panitumumab antibodies on pharmacokinetic and safety profiles in patients with mCRC receiving panitumumab in combination with oxaliplatin- or irinotecan-based chemotherapies. METHODS: Three validated assays (two screening immunoassays and a neutralizing antibody bioassay) were used to detect the presence of anti-panitumumab antibodies in serum samples collected from patients enrolled in four panitumumab combination chemotherapy clinical trials. The impact of anti-panitumumab antibodies on pharmacokinetic and safety profiles was analyzed using population pharmacokinetic analysis and descriptive statistics, respectively. RESULTS: Of 1124 patients treated with panitumumab in combination with oxaliplatin- or irinotecan-based chemotherapy with postbaseline samples available for testing, 20 (1.8%) patients developed binding antibodies and 2 (0.2%) developed neutralizing antibodies. The incidence of anti-panitumumab antibodies was similar in patients with tumors expressing wild-type or mutant KRAS and in patients receiving oxaliplatin- or irinotecan-based chemotherapies. No evidence of an altered pharmacokinetic or safety profile was found in patients who tested positive for anti-panitumumab antibodies. CONCLUSIONS: The immunogenicity of panitumumab in the combination chemotherapy setting was infrequent and similar to the immunogenicity observed in the monotherapy setting. Panitumumab immunogenicity did not appear to alter pharmacokinetic or safety profiles. This low rate of immunogenicity may be attributed to the fully human nature of panitumumab.


Subject(s)
Antibodies, Anti-Idiotypic/analysis , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Drug Hypersensitivity/immunology , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/analysis , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/complications , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Drug Hypersensitivity/blood , Drug Hypersensitivity/complications , Drug Hypersensitivity/epidemiology , Humans , Incidence , Irinotecan , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Panitumumab , Severity of Illness Index
18.
J Biomol Screen ; 15(6): 644-52, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20508254

ABSTRACT

The method described here employs a high-content cell-based assay format for the detection of neutralizing antibodies (NAbs) to panitumumab, a fully human monoclonal antagonistic antibody to the human epidermal growth factor (EGF) receptor in human serum (screening assay). A specificity assay was also developed and qualified to confirm that the neutralizing activity was attributable to the presence of NAbs and not due to serum interference (serum interference assay). The ArrayScan IV HCS reader was used for the measurement of tyrosine phosphorylation of the epidermal growth factor receptor (EGFR) and STAT-1 redistribution between the cytoplasm and nucleus in the human epidermoid carcinoma cell line A431. Assay conditions were developed by (1) optimizing the response of the A431 cells to recombinant human EGF in pooled human serum, (2) evaluating the ability of panitumumab to inhibit the EGF response, and (3) assessing the assay's sensitivity for detecting a positive control affinity purified rabbit polyclonal anti-panitumumab antibody. Panitumumab dose-dependently inhibited 4 ng/mL EGF, and the positive control antibody showed a dose-dependent neutralization of 50 ng/mL panitumumab. The experiments indicated that in comparison to STAT-1 translocation, EGFR phosphorylation was the optimal endpoint for the screening and serum interference assays. Assay cut points were derived for the screening and serum interference assays by obtaining normalized ratios of mean fluorescence intensity values obtained with EGFR phosphorylation by testing sera from healthy human donor sera. The assay sensitivity was determined to be 0.125 microg/mL for the positive control antibody.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antineoplastic Agents/immunology , Automation/methods , Biological Assay/methods , Microscopy, Fluorescence/methods , Cell Line, Tumor , Drug Evaluation, Preclinical , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , Neutralization Tests , Panitumumab , Phosphorylation/drug effects , Protein Transport/drug effects , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
19.
Curr Pharm Biotechnol ; 10(4): 352-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19519410

ABSTRACT

Assessment of immunogenicity is a major aspect in evaluating the safety of biological therapeutic proteins. It is important to evaluate the immunogenic potential of the biologics in an appropriate fashion using clearly defined strategy and clinical trials. The studies must include the appropriate risk assessment procedures using validated methods. The immune responses against the therapeutic biologics can be studied using various methodologies. These include enzyme linked immunoassays (ELISA), surface plasmon resonance (SPR), chemiluminescence, and flowcytometry assays for binding antibodies and cell based assays for neutralizing antibodies. The immune responses to the biologics can widely vary in various cross section of the population, thus a combination of techniques are necessary to fully evaluate the immunogenic potential of the biologics. This review outlines various commonly used technology platforms, its merits and shortcomings for the evaluation of the immune responses.


Subject(s)
Biopharmaceutics/methods , Immunoassay/methods , Immunogenetics/methods , Peptides/immunology , Proteins/immunology , Proteins/therapeutic use , Technology, Pharmaceutical/methods , Animals , Humans , Peptides/chemistry , Peptides/therapeutic use , Proteins/chemistry
20.
J Immunol ; 178(11): 7467-72, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17513798

ABSTRACT

Evaluation of the immunogenicity of panitumumab, a fully human anti-epidermal growth factor receptor mAb approved for use in colorectal cancer patients, led to the development of two separate immunoassays for the detection of anti-panitumumab Abs. The first immunoassay used a bridging ELISA capable of detecting 10 ng/ml positive control anti-panitumumab Ab. The ELISA incorporated an acid dissociation step to reduce drug interference and tolerated the presence of approximately 100-fold molar excess of drug. During eight clinical trials, the ELISA detected developing Ab responses in 2 of 612 (0.3%) subjects. In one of the ELISA positive subjects, neutralizing Abs were detected using an epidermal growth factor receptor phosphorylation bioassay. The second immunoassay used a Biacore biosensor immunoassay format capable of detecting 1 mug/ml positive control Ab while tolerating the presence of equal molar amounts of drug. Although less sensitive and less tolerant to competing drug in the assay, the Biacore assay detected developing Ab responses in 25 of the 604 (4.1%) subjects. Additionally, the Biacore assay identified eight subjects who developed neutralizing Abs. Mouse mAbs with affinities ranging from 1.1 x 10(-6) to 8.4 x 10(-10) M were used to characterize both assay types. The ELISA was more sensitive for the detection of higher affinity mAbs and detected high-affinity mAbs in the presence of higher molar ratio of drug to mAb. The Biacore assay was more sensitive for detection of lower affinity mAbs and detected low affinity Abs in the presence of higher molar ratios of drug to mAb.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Enzyme-Linked Immunosorbent Assay/methods , Surface Plasmon Resonance/methods , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Anti-Idiotypic/blood , Antibodies, Monoclonal/metabolism , Antibody Affinity , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay/standards , Epitope Mapping , ErbB Receptors/immunology , Female , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/metabolism , Mice , Mice, Inbred NOD , Panitumumab , Sensitivity and Specificity , Surface Plasmon Resonance/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...