Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Mol Ther Methods Clin Dev ; 32(1): 101217, 2024 Mar 14.
Article En | MEDLINE | ID: mdl-38496304

Recombinant adeno-associated virus (AAV) vectors are the leading delivery vehicle used for in vivo gene therapies. Anti-AAV antibodies (AAV Abs) can interact with the viral capsid component of an AAV-based gene therapy (GT). Therefore, patients with preexisting AAV Abs (seropositive patients) are often excluded from GT trials to prevent treatment of patients who are unlikely to benefit1 or may have a higher risk for adverse events outweighing treatment benefits. On the contrary, unnecessary exclusion of patients with high unmet medical need should be avoided. Instead, a risk-benefit assessment that weighs the potential risks due to seropositivity vs. severity of disease and available treatment options, should drive the decision if patient selection is required. Assays for patient selection must be validated according to their intended use following national regulations/standards for diagnostic assays in appropriate laboratories. In this review, we summarize the current process of patient selection, including assay cutoff criteria and related assay validation approaches. We further provide considerations on regulatory requirements for the development of in vitro diagnostic tests supporting market authorization of a corresponding GT.

2.
Neurol Neuroimmunol Neuroinflamm ; 11(3): e200216, 2024 May.
Article En | MEDLINE | ID: mdl-38484217

BACKGROUND AND OBJECTIVES: Autoantibody discovery in complex autoimmune diseases is challenging. Diverse successful antigen identification strategies are available, but, so far, have often been unsuccessful, especially in the discovery of protein antigens in which conformational and post-translational modification are critical. Our study assesses the utility of a human membrane and secreted protein microarray technology to detect autoantibodies in chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). METHODS: A cell microarray consisting of human embryonic kidney-293 cells expressing >5,000 human proteins was used. First, a validation step was performed with 4 serum samples from patients with autoimmune nodopathy (AN) to assess the ability of this technology to detect circulating known autoantibodies. The ability of the cell microarray technology to discover novel IgG autoantibodies was assessed incubating the array with 8 CIDP serum samples. Identified autoantibodies were subsequently validated using cell-based assays (CBAs), ELISA, and/or tissue immunohistochemistry and analyzed in a cohort of CIDP and AN (n = 96) and control (n = 100) samples. RESULTS: Serum anti-contactin-1 and anti-neurofascin-155 were detected by the human cell microarray technology. Nine potentially relevant antigens were found in patients with CIDP without other detectable antibodies; confirmation was possible in six of them: ephrin type-A receptor 7 (EPHA7); potassium-transporting ATPase alpha chain 1 and subunit beta (ATP4A/4B); leukemia-inhibitory factor (LIF); and interferon lambda 1, 2, and 3 (IFNL1, IFNL2, IFNL3). Anti-ATP4A/4B and anti-EPHA7 antibodies were detected in patients and controls and considered unrelated to CIDP. Both anti-LIF and anti-IFNL antibodies were found in the same 2 patients and were not detected in any control. Both patients showed the same staining pattern against myelinating fibers of peripheral nerve tissue and of myelinating neuron-Schwann cell cocultures. Clinically relevant correlations could not be established for anti-LIF and anti-IFNL3 antibodies. DISCUSSION: Our work demonstrates the utility of human cell microarray technology to detect known and discover unknown autoantibodies in human serum samples. Despite potential CIDP-associated autoantibodies (anti-LIF and anti-IFNL3) being identified, their clinical and pathogenic relevance needs to be elucidated in bigger cohorts.


Autoimmune Diseases , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Humans , Autoantibodies , Proteome , Neurons/chemistry
3.
Mult Scler ; 25(3): 392-398, 2019 03.
Article En | MEDLINE | ID: mdl-29336205

BACKGROUND: Anti-drug antibodies (ADA) against natalizumab develop early during treatment. ADA persistency is defined by two consecutive positive results as performed by the current qualitative ELISA assay (positive/negative). Very little is known about the magnitude of the natalizumab ADA response and persistency. DESIGN/METHODS: We developed a highly sensitive natalizumab ADA titration assay on the Meso Scale Discovery (MSD) platform and a pharmacokinetic (PK) assay. We included 43 patients with a positive ELISA-ADA result within 6 months of treatment initiation (baseline) of whom a follow-up serum sample was available 12-30 months after treatment start. MSD-ADA titres and drug levels were measured. RESULTS: Median MSD-ADA titre at baseline was 4881 and 303 at follow-up. A titre of >400 at baseline had a 94% sensitivity and 89% specificity to predict ADA persistency. Reversion to ADA negativity occurred in 10 patients with mean drug levels of 10.8 µg/mL. The median trough drug level in ADA-positive samples was 0 µg/mL. PK levels and ADA titres correlated strongly negatively ( r = -0.67). CONCLUSION: High baseline natalizumab ADA titres accurately predict persistency. Despite continuous treatment, the majority of patients with persistent ADA had no detectable drug levels indicating loss of efficacy in line with phase 3 study results.


Antibodies/immunology , Immunoassay/standards , Immunologic Factors/immunology , Multiple Sclerosis/drug therapy , Natalizumab/immunology , Outcome Assessment, Health Care , Adult , Antibodies/blood , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Immunologic Factors/blood , Male , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Natalizumab/blood , Sensitivity and Specificity , Young Adult
4.
Int Immunopharmacol ; 66: 362-365, 2019 Jan.
Article En | MEDLINE | ID: mdl-30529500

Primary immune thrombocytopenia (ITP) is an autoimmune disease characterized by pathogenic immunoglobulin G (IgG) autoantibodies that bind to platelets, causing their phagocytic removal and leading to reductions in platelet number. The neonatal Fc receptor (FcRn) selectively salvages and recycles IgG, including pathogenic IgG, thereby extending the half-life of IgG in plasma. Two anti-mouse FcRn monoclonal antibodies (mAb) (4470 and 4464) were generated to evaluate the effect of inhibiting IgG recycling. Statistically significant reductions in plasma IgG concentration were observed upon administration of 4470 (10, 30 and 100 mg/kg) in wild-type mice. In a passive mouse model of ITP, 4464 alleviated the reduction in platelet number and/or preserved newly produced platelets when dosed prophylactically as well as in a therapeutic dosing regimen once platelet numbers had already been reduced. These results support the investigation of anti-FcRn therapy as a potential treatment for ITP.


Antibodies, Monoclonal/therapeutic use , Blood Platelets/immunology , Immunoglobulin G/blood , Immunoglobulins, Intravenous/therapeutic use , Immunotherapy/methods , Single-Chain Antibodies/therapeutic use , Thrombocytopenia/therapy , Animals , Antibodies, Monoclonal/genetics , Autoantibodies/metabolism , Cell Line , Disease Models, Animal , Female , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Humoral , Mice , Mice, Inbred C57BL , Platelet Count , Receptors, Fc/immunology , Single-Chain Antibodies/genetics , Thrombocytopenia/immunology
5.
MAbs ; 10(7): 1111-1130, 2018 10.
Article En | MEDLINE | ID: mdl-30130439

Rozanolixizumab (UCB7665), a humanized high-affinity anti-human neonatal Fc receptor (FcRn) monoclonal antibody (IgG4P), has been developed to reduce pathogenic IgG in autoimmune and alloimmune diseases. We document the antibody isolation and compare rozanolixizumab with the same variable region expressed in various mono-, bi- and trivalent formats. We report activity data for rozanolixizumab and the different molecular formats in human cells, FcRn-transgenic mice, and cynomolgus monkeys. Rozanolixizumab, considered the most effective molecular format, dose-dependently and selectively reduced plasma IgG concentrations in an FcRn-transgenic mouse model (no effect on albumin). Intravenous (IV) rozanolixizumab dosing in cynomolgus monkeys demonstrated non-linear pharmacokinetics indicative of target-mediated drug disposition; single IV rozanolixizumab doses (30 mg/kg) in cynomolgus monkeys reduced plasma IgG concentration by 69% by Day 7 post-administration. Daily IV administration of rozanolixizumab (initial 30 mg/kg loading dose; 5 mg/kg daily thereafter) reduced plasma IgG concentrations in all cynomolgus monkeys, with low concentrations maintained throughout the treatment period (42 days). In a 13-week toxicology study in cynomolgus monkeys, supra-pharmacological subcutaneous and IV doses of rozanolixizumab (≤ 150 mg/kg every 3 days) were well tolerated, inducing sustained (but reversible) reductions in IgG concentrations by up to 85%, with no adverse events observed. We have demonstrated accelerated natural catabolism of IgG through inhibition of IgG:FcRn interactions in mice and cynomolgus monkeys. Inhibition of FcRn with rozanolixizumab may provide a novel therapeutic approach to reduce pathogenic IgG in human autoimmune disease. Rozanolixizumab is being investigated in patients with immune thrombocytopenia (NCT02718716) and myasthenia gravis (NCT03052751).


Antibodies, Monoclonal, Humanized/chemistry , Histocompatibility Antigens Class I/immunology , Immunosuppressive Agents/chemistry , Myasthenia Gravis/drug therapy , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Receptors, Fc/immunology , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , Clinical Trials as Topic , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/blood , Immunosuppressive Agents/metabolism , Macaca fascicularis , Mice , Mice, Transgenic , Protein Binding , Receptors, Fc/genetics , Transgenes/genetics
6.
Sci Transl Med ; 9(414)2017 11 01.
Article En | MEDLINE | ID: mdl-29093180

Pathogenic immunoglobulin G (IgG) autoantibodies characterize some human autoimmune diseases; their high concentration and long half-life are dependent on recycling by the neonatal Fc receptor (FcRn). Inhibition of FcRn is an attractive new treatment concept for IgG-mediated autoimmune diseases. Rozanolixizumab (UCB7665; CA170_01519.g57 IgG4P) is an anti-human FcRn monoclonal antibody. In cynomolgus monkeys, rozanolixizumab reduced IgG (maximum 75 to 90% by about day 10), was well tolerated, and did not increase risk of infection. We also report a first-in-human, randomized, double-blind, placebo-controlled, dose-escalating study of intravenous (IV) or subcutaneous (SC) rozanolixizumab in healthy subjects (NCT02220153). The primary objective was to evaluate safety and tolerability. Secondary objectives were assessment of rozanolixizumab pharmacokinetics and pharmacodynamics, including effects on circulating IgG concentrations. Forty-nine subjects were randomized to receive rozanolixizumab (n = 36) or placebo (n = 13) across six cohorts. The first three cohorts received IV doses, and the subsequent three cohorts received SC doses, of rozanolixizumab 1, 4, or 7 mg/kg (n = 6 for each cohort; plus n = 7 or 6 for placebo, respectively). The most frequent treatment-emergent adverse event [TEAE; headache, 14 of 36 (38.9%) subjects] was dose-dependent and more prominent after IV administration. Severe TEAEs occurred in four subjects, all in the highest-dose IV group [headache (n = 3) and back pain (n = 1)]. Rozanolixizumab pharmacokinetics demonstrated nonlinear increases with dose. There were sustained dose-dependent reductions in serum IgG concentrations (IV and SC rozanolixizumab). These data provide clinical evidence for the therapeutic potential of rozanolixizumab.


Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Immunoglobulin G/blood , Receptors, Fc/antagonists & inhibitors , Adult , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Area Under Curve , Demography , Dose-Response Relationship, Drug , Female , Histocompatibility Antigens Class I/metabolism , Humans , Macaca fascicularis , Male , Middle Aged , Receptors, Fc/metabolism , Toxicity Tests
7.
MAbs ; 8(7): 1319-1335, 2016 10.
Article En | MEDLINE | ID: mdl-27532598

An antibody format, termed Fab-dsFv, has been designed for clinical indications that require monovalent target binding in the absence of direct Fc receptor (FcR) binding while retaining substantial serum presence. The variable fragment (Fv) domain of a humanized albumin-binding antibody was fused to the C-termini of Fab constant domains, such that the VL and VH domains were individually connected to the Cκ and CH1 domains by peptide linkers, respectively. The anti-albumin Fv was selected for properties thought to be desirable to ensure a durable serum half-life mediated via FcRn. The Fv domain was further stabilized by an inter-domain disulfide bond. The bispecific format was shown to be thermodynamically and biophysically stable, and retained good affinity and efficacy to both antigens simultaneously. In in vivo studies, the serum half-life of Fab-dsFv, 2.6 d in mice and 7.9 d in cynomolgus monkeys, was equivalent to Fab'-PEG.


Antibodies, Bispecific/blood , Immunoglobulin Fab Fragments , Immunoglobulin Variable Region , Serum Albumin/metabolism , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Half-Life , Humans , Immunoglobulin Fab Fragments/blood , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Variable Region/blood , Immunoglobulin Variable Region/chemistry , Mice , Serum Albumin/immunology
8.
Clin Vaccine Immunol ; 20(3): 377-90, 2013 Mar.
Article En | MEDLINE | ID: mdl-23324518

Clostridium difficile infections are a major cause of antibiotic-associated diarrhea in hospital and care facility patients. In spite of the availability of effective antibiotic treatments, C. difficile infection (CDI) is still a major cause of patient suffering, death, and substantial health care costs. Clostridium difficile exerts its major pathological effects through the actions of two protein exotoxins, TcdA and TcdB, which bind to and disrupt gut tissue. Antibiotics target the infecting bacteria but not the exotoxins. Administering neutralizing antibodies against TcdA and TcdB to patients receiving antibiotic treatment might modulate the effects of the exotoxins directly. We have developed a mixture of three humanized IgG1 monoclonal antibodies (MAbs) which neutralize TcdA and TcdB to address three clinical needs: reduction of the severity and duration of diarrhea, reduction of death rates, and reduction of the rate of recurrence. The UCB MAb mixture showed higher potency in a variety of in vitro binding and neutralization assays (∼10-fold improvements), higher levels of protection in a hamster model of CDI (82% versus 18% at 28 days), and higher valencies of toxin binding (12 versus 2 for TcdA and 3 versus 2 for TcdB) than other agents in clinical development. Comparisons of the MAb properties also offered some insight into the potential relative importance of TcdA and TcdB in the disease process.


Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridium Infections/therapy , Enterotoxins/antagonists & inhibitors , Immunologic Factors/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Cricetinae , Disease Models, Animal , Enterotoxins/immunology , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Immunoglobulin G/therapeutic use , Immunologic Factors/immunology , Immunologic Factors/isolation & purification , Treatment Outcome
...