Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Adv Ther ; 38(5): 2418-2434, 2021 05.
Article in English | MEDLINE | ID: mdl-33778929

ABSTRACT

INTRODUCTION: Etrolizumab is a novel, dual-action anti-ß7 integrin antibody studied in phase 3 trials in patients with inflammatory bowel disease. An autoinjector (AI) is being developed in parallel to complement the prefilled syringe with needle safety device (PFS-NSD) for subcutaneous (SC) administration in these trials. Here we demonstrate the comparable pharmacokinetics, tolerability, and safety of both devices. METHODS: This randomized, open-label, two-part study in healthy participants evaluated the comparability of etrolizumab exposure between the AI and the PFS-NSD. Part 1 (pilot) involved a small number of participants, and initial results were used to finalize the design of the larger part 2 (pivotal) study. In both parts, participants were randomly assigned to receive a single SC dose of etrolizumab 105 mg by AI or PFS-NSD. Randomization was stratified by body weight. Primary pharmacokinetic outcomes were Cmax, AUClast, and AUC0-inf. RESULTS: One hundred and eighty healthy participants (part 1, n = 30; part 2, n = 150) received a single SC dose of etrolizumab by AI or PFS-NSD. Primary pharmacokinetic results from part 1 supported modification of the part 2 study design. Results from part 2 demonstrated that etrolizumab exposure was equivalent between devices, with geometric mean ratios (GMRs) between AI and PFS-NSD of 102% (90% confidence interval [CI] 94.2-111) for Cmax, 98.0% (90% CI 89.3-107) for AUClast, and 97.6% (90% CI 88.6-107) for AUC0-inf. Median tmax and mean terminal t1/2 were also similar between devices. GMRs and 90% CIs of all primary pharmacokinetic parameters were fully contained within the predefined equivalence limits (80-125%). CONCLUSION: This pharmacokinetic study demonstrated that single SC injections of etrolizumab 105 mg using an AI or a PFS-NSD resulted in equivalent etrolizumab exposure and similar safety and tolerability in healthy participants. Taken together, these results support the use of an AI for etrolizumab administration. TRIAL REGISTRATION: NCT02996019.


Subject(s)
Antibodies, Monoclonal, Humanized , Syringes , Healthy Volunteers , Humans , Injections, Subcutaneous
2.
Sci Transl Med ; 13(593)2021 05 12.
Article in English | MEDLINE | ID: mdl-33980574

ABSTRACT

Tau has become an attractive alternative target for passive immunotherapy efforts for Alzheimer's disease (AD). The anatomical distribution and extent of tau pathology correlate with disease course and severity better than other disease markers to date. We describe here the generation, preclinical characterization, and phase 1 clinical characterization of semorinemab, a humanized anti-tau monoclonal antibody with an immunoglobulin G4 (igG4) isotype backbone. Semorinemab binds all six human tau isoforms and protects neurons against tau oligomer neurotoxicity in cocultures of neurons and microglia. In addition, when administered intraperitoneally once weekly for 13 weeks, murine versions of semorinemab reduced the accumulation of tau pathology in a transgenic mouse model of tauopathy, independent of antibody effector function status. Semorinemab also showed clear evidence of target engagement in vivo, with increases in systemic tau concentrations observed in tau transgenic mice, nonhuman primates, and humans. Higher concentrations of systemic tau were observed after dosing in AD participants compared to healthy control participants. No concerning safety signals were observed in the phase 1 clinical trial at single doses up to 16,800 mg and multiple doses totaling 33,600 mg in a month.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/drug therapy , Animals , Brain/metabolism , Disease Models, Animal , Humans , Immunization, Passive , Mice , Mice, Transgenic , Tauopathies/drug therapy , tau Proteins/metabolism
3.
J Pathol ; 218(3): 380-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19334050

ABSTRACT

Despite the availability of new targeted therapies, ductal pancreatic adenocarcinoma continues to carry a poor prognosis. Carcinoembryonic antigen-related cell adhesion molecule (CEACAM)6 has been reported as a potential biomarker and therapy target for this malignancy. We have evaluated CEACAM6 as a potential therapy target, using an antibody-drug conjugate (ADC). Expression of CEACAM6 in pancreatic adenocarcinomas was determined using immunohistochemistry on tissue microarrays. The expression pattern in granulocytes and granulocytic precursors was measured by flow cytometry. Murine xenograft and non-human primate models served to evaluate efficacy and safety, respectively. Robust expression of CEACAM6 was found in > 90% of invasive pancreatic adenocarcinomas as well as in intraepithelial neoplastic lesions. In the granulocytic lineage, CEACAM6 was expressed at all stages of granulocytic maturation except for the early lineage-committed precursor cell. The anti-CEACAM6 ADC showed efficacy against established CEACAM6-expressing tumours. In non-human primates, antigen-dependent toxicity of the ADC consisted of dose-dependent and reversible depletion of granulocytes and their precursors. This was associated with preferential and rapid localization of the antibody in bone marrow, as determined by sequential in vivo PET imaging of the radiolabelled anti-CEACAM6. Localization of the radiolabelled tracer could be attenuated by predosing with unlabelled antibody confirming specific accumulation in this compartment. Based on the expression pattern in normal and malignant pancreatic tissues, efficacy against established tumours and limited and reversible bone marrow toxicity, we propose that CEACAM6 should be considered for an ADC-based therapy approach against pancreatic adenocarcinomas and possibly other CEACAM6-positive neoplasms.


Subject(s)
Adenocarcinoma/therapy , Cell Adhesion Molecules/antagonists & inhibitors , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/toxicity , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Drug Evaluation, Preclinical/methods , Feasibility Studies , Female , GPI-Linked Proteins , Hematopoietic Stem Cells/metabolism , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Macaca fascicularis , Male , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neutrophil Activation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
4.
Clin Pharmacol Ther ; 105(1): 177-189, 2019 01.
Article in English | MEDLINE | ID: mdl-29952004

ABSTRACT

Most treatments for epithelial injury target hematopoietic mechanisms, possibly causing immunosuppression. Interleukin (IL)-22 promotes tissue regeneration, acting directly on epithelial cells. UTTR1147A, a human IL-22Fc (immunoglobulin G (IgG)4) fusion protein, activates IL-22 signaling. This phase I placebo-controlled trial of single, ascending, i.v. (1-120 µg/kg) and s.c (3-120 µg/kg) doses of UTTR1147A analyzed its effects on safety, tolerability, pharmacokinetics, and pharmacodynamic biomarkers in healthy volunteers. Most adverse events (AEs) were mild or moderate. The maximum tolerated i.v. dose in healthy volunteers was 90 µg/kg. Predominant AEs were dose-dependent reversible skin effects consistent with IL-22 pharmacology. UTTR1147A exposure increased approximately dose-proportionally, with a half-life of ~1 week. IL-22 biomarkers (regenerating islet protein 3A (REG3A), serum amyloid A (SAA), and C-reactive protein (CRP)) increased dose-dependently. Neither inflammatory symptoms and signs nor cytokines increased with CRP elevations. UTTR1147A demonstrated acceptable safety, pharmacokinetics, and IL-22R engagement, supporting further clinical development.


Subject(s)
Epithelial Cells/drug effects , Immunoglobulin G/administration & dosage , Interleukins/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Adolescent , Adult , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Healthy Volunteers , Humans , Immunoglobulin G/metabolism , Interleukins/metabolism , Male , Middle Aged , Recombinant Fusion Proteins/metabolism , Single-Blind Method , Young Adult , Interleukin-22
5.
Front Pharmacol ; 10: 39, 2019.
Article in English | MEDLINE | ID: mdl-30774593

ABSTRACT

Background: Anti-integrin therapy is a new frontline strategy in the treatment of inflammatory bowel diseases (IBD). The anti-ß7 integrin antibody etrolizumab is currently being investigated for safety and efficacy in Crohn's disease (CD) and ulcerative colitis (UC) in several phase III trials. Mechanistically, etrolizumab is known to block ß7 integrin ligand binding and reduces intestinal trafficking of ß7-expressing cells. Etrolizumab blocks ß7 integrin ligand binding and reduces ß7-positive lymphocyte migration and retention in the inflamed gut mucosa, but the exact mechanisms by which this inhibition occurs are not fully understood. Methods: Cellular effects of etrolizumab or etrolizumab surrogate antibody (etrolizumab-s) were investigated in cell culture models and analyzed by flow cytometry, fluorescence microscopy, ImageStream®, stimulated emission depletion (STED) microscopy and functional dynamic in vitro adhesion assays. Moreover, effects on α4ß7 integrin were compared with the pharmacodynamically similar antibody vedolizumab. Results: As demonstrated by several different approaches, etrolizumab and etrolizumab-s treatment led to internalization of ß7 integrin. This resulted in impaired dynamic adhesion to MAdCAM-1. Internalized ß7 integrin localized in endosomes and re-expression of ß7 was dependent on de novo protein synthesis. In vitro etrolizumab treatment did not lead to cellular activation or cytokine secretion and did not induce cytotoxicity. Internalization of α4ß7 integrin was increased with etrolizumab compared with vedolizumab. Discussion: Our data suggest that etrolizumab does not elicit secondary effector functions on the single cell level. Integrin internalization may be an important mechanism of action of etrolizumab, which might explain some but not all immunological effects observed with etrolizumab.

6.
J Clin Pharmacol ; 58(3): 386-398, 2018 03.
Article in English | MEDLINE | ID: mdl-29178491

ABSTRACT

Etrolizumab, a humanized monoclonal antibody, specifically binds to the ß7 subunit of the heterodimeric integrins α4ß7 and αEß7. Pharmacokinetic (PK) and pharmacodynamic (PD) data were collected from an etrolizumab phase 1 trial in patients with moderate to severe ulcerative colitis (UC). We developed a mechanism-based model to simultaneously describe the kinetics of serum etrolizumab concentration and free ß7 receptors on circulating intestinal-homing CD4+ T lymphocytes. Included in the analysis were 38 phase 1 UC patients who received single or 3 monthly doses of etrolizumab intravenously or subcutaneously across a dose range of 0.3 to 10 mg/kg. A quasi-steady-state target-mediated drug disposition model was developed to describe the dynamic interaction between serum etrolizumab concentration and free ß7 receptors on intestinal-homing CD4+ T lymphocytes in UC patients. The time profiles of serum etrolizumab and absolute counts of ß7+ lymphocytes (expressed as percentage of baseline level) were well described by the quasi-steady-state target-mediated drug disposition model. The model was able to characterize the maximum drug occupancy of ß7 receptors on intestinal-homing CD4+ T lymphocytes and the concentration-dependent duration of occupancy. The 90% effective concentration for etrolizumab to saturate the ß7 receptors on intestinal homing CD4+ T cells was 1.3 µg/mL. PK and PD profiles predicted by the model were consistent with observations from a subsequent phase 2 study. In conclusion, an integrated PK/PD model developed in this analysis reasonably described serum etrolizumab PK profiles and the relationship between PK and PD (free ß7 receptors on circulating intestinal-homing CD4+ T lymphocytes) in UC patients.


Subject(s)
Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacology , Colitis, Ulcerative/blood , Colitis, Ulcerative/drug therapy , Integrins/blood , Models, Biological , Antibodies, Monoclonal, Humanized/pharmacokinetics , Cohort Studies , Colitis, Ulcerative/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Doublecortin Domain Proteins , Female , Gastrointestinal Agents/blood , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/pharmacology , Humans , Immunologic Factors/blood , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Male , Microtubule-Associated Proteins , Neuropeptides
7.
AAPS J ; 21(1): 9, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30547287

ABSTRACT

The interleukin (IL)-17 pathway has been implicated in the pathophysiology of many autoimmune diseases. MCAF5352A is a humanized monoclonal antibody which targets both IL-17A and IL-17F, thereby inhibiting the activity of IL-17 dimers (IL-17AA, IL-17AF, and IL-17FF). The pharmacokinetic profile of MCAF5352A has been characterized in both a Phase Ia single ascending dose study and a Phase Ib multiple ascending dose study. Two qualified enzyme-linked immunosorbent assays were used to measure total IL-17AA and IL-17FF levels in serum as pharmacodynamic biomarkers in the Phase I studies. The two assays demonstrated specificity for IL-17AA or IL-17FF with sensitivity at low picogram/milliliter levels. The assay precision and accuracy also met acceptance criteria. Although total serum IL-17AA and IL-17FF levels were below the assay detection limits prior to administration of MCAF5352A, post-treatment levels in both the single and multiple dose cohorts became detectable and increased in a dose-dependent manner. These data are consistent with target engagement by MCAF5352A. Our work highlights bioanalytical challenges encountered while developing biomarker assays requiring high sensitivity and specificity. Data generated using these assays enabled the confirmation of target engagement during early clinical drug development.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Biomarkers, Pharmacological/blood , Interleukin-17/blood , Adult , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Arthritis, Rheumatoid/drug therapy , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Development , Enzyme-Linked Immunosorbent Assay/methods , Female , Healthy Volunteers , Humans , Interleukin-17/antagonists & inhibitors , Limit of Detection , Male , Mice , Middle Aged , Sensitivity and Specificity , Young Adult
8.
Bioanalysis ; 9(16): 1207-1216, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28766364

ABSTRACT

AIM: Etrolizumab, a humanized monoclonal antibody, has demonstrated clinical remission in a Phase II study of ulcerative colitis patients. In the Phase III program, a second indication, Crohn's disease was added. The pharmacokinetic ELISA used in the Phase I/II studies in normal human and ulcerative colitis sera exhibited matrix interference in the Crohn's disease population, necessitating implementation of a new technology. Methodology & results: Optimization of the original ELISA and assay redevelopment using different antibody pairs did not result in substantive improvements, necessitating implementation of an alternative technology for assay development. CONCLUSION: We highlight the challenges encountered with optimization/redevelopment of the original ELISA and discuss results of the new assay on the Gyros platform.


Subject(s)
Crohn Disease/metabolism , Enzyme-Linked Immunosorbent Assay/instrumentation , Immunoassay/instrumentation , Lab-On-A-Chip Devices , Animals , Artifacts , Crohn Disease/blood , Humans , Tissue Distribution
9.
MAbs ; 6(6): 1500-8, 2014.
Article in English | MEDLINE | ID: mdl-25484037

ABSTRACT

RG7652 is a human immunoglobulin 1 (IgG1) monoclonal antibody (mAb) targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) and is designed for the treatment of hypercholesterolemia. A target-binding enzyme-linked immunosorbent assay (ELISA) was developed to measure RG7652 levels in human serum in a Phase I study. Although target-binding assay formats are generally used to quantify free therapeutic, the actual therapeutic species being measured are affected by assay conditions, such as sample dilution and incubation time, and levels of soluble target in the samples. Therefore, in the presence of high concentrations of circulating target, the choice of reagents and assay conditions can have a significant effect on the observed pharmacokinetic (PK) profiles. Phase I RG7652 PK analysis using the ELISA data resulted in a nonlinear dose normalized exposure. An investigation was conducted to characterize the ELISA to determine whether the assay format and reagents may have contributed to the PK observation. In addition, to confirm the ELISA results, a second orthogonal method, liquid chromatography tandem mass spectrometry (LC-MS/MS) using a signature peptide as surrogate, was developed and implemented. A subset of PK samples, randomly selected from half of the subjects in the 6 single ascending dose (SAD) cohorts in the Phase I clinical study, was analyzed with the LC-MS/MS assay, and the data were found to be comparable to the ELISA data. This paper illustrates the importance of reagent characterization, as well as the benefits of using an orthogonal approach to eliminate bioanalytical contributions when encountering unexpected observations.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Immunoglobulin G/immunology , Proprotein Convertases/immunology , Serine Endopeptidases/immunology , Antibodies, Monoclonal/blood , Antibodies, Monoclonal, Humanized , Chromatography, Liquid , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Proprotein Convertase 9 , Tandem Mass Spectrometry
10.
Bioanalysis ; 4(17): 2127-39, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23013395

ABSTRACT

BACKGROUND: Design of experiments (DOE) is a systematic approach to assess the effects of many factors on a response of an assay. This paper provides a case study whereby DOE was successfully utilized to evaluate robustness parameters for a ligand-binding assay (LBA). METHODOLOGY: A 24-run Plackett-Burman design was developed to investigate factors that may have caused a lack of robustness in this particular LBA. We modeled five main effects and their ten two-way interactions, using the standard curve signal as the response. RESULTS: By utilizing DOE, we were able to quickly identify the factors that affected our assay's performance. The lack of robustness was attributed to the handling of the coat reagent. Factors that had an adverse effect on the coat material were vortexing and freeze-thaw cycles. CONCLUSION: We recommend that a robustness DOE be conducted prior to the validation of an assay for early identification of critical factors that may impact assay performance.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , Research Design/standards , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Humans
SELECTION OF CITATIONS
SEARCH DETAIL