Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 32(28): 9677-89, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22787053

ABSTRACT

Passive immunization against ß-amyloid (Aß) has become an increasingly desirable strategy as a therapeutic treatment for Alzheimer's disease (AD). However, traditional passive immunization approaches carry the risk of Fcγ receptor-mediated overactivation of microglial cells, which may contribute to an inappropriate proinflammatory response leading to vasogenic edema and cerebral microhemorrhage. Here, we describe the generation of a humanized anti-Aß monoclonal antibody of an IgG4 isotype, known as MABT5102A (MABT). An IgG4 subclass was selected to reduce the risk of Fcγ receptor-mediated overactivation of microglia. MABT bound with high affinity to multiple forms of Aß, protected against Aß1-42 oligomer-induced cytotoxicity, and increased uptake of neurotoxic Aß oligomers by microglia. Furthermore, MABT-mediated amyloid plaque removal was demonstrated using in vivo live imaging in hAPP((V717I))/PS1 transgenic mice. When compared with a human IgG1 wild-type subclass, containing the same antigen-binding variable domains and with equal binding to Aß, MABT showed reduced activation of stress-activated p38MAPK (p38 mitogen-activated protein kinase) in microglia and induced less release of the proinflammatory cytokine TNFα. We propose that a humanized IgG4 anti-Aß antibody that takes advantage of a unique Aß binding profile, while also possessing reduced effector function, may provide a safer therapeutic alternative for passive immunotherapy for AD. Data from a phase I clinical trial testing MABT is consistent with this hypothesis, showing no signs of vasogenic edema, even in ApoE4 carriers.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Immunoglobulin G/pharmacology , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , CX3C Chemokine Receptor 1 , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Humans , Immunoglobulin G/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Middle Aged , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/metabolism , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/genetics , Statistics, Nonparametric , Time Factors , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
MAbs ; 4(6): 724-31, 2012.
Article in English | MEDLINE | ID: mdl-23778267

ABSTRACT

Modulating the binding affinities to IgE or changing the FcγR binding properties of anti-IgE antibodies offers an opportunity to enhance the therapeutic potential of anti-IgE antibodies, but the influence of increased affinity to IgE or reduced Fc effector function on the pharmacological properties of anti-IgE therapies remains unclear. Our studies were designed to characterize the pharmacokinetics, pharmacodynamics and immune-complex distribution of two high-affinity anti-IgE monoclonal antibodies, high-affinity anti-IgE antibody (HAE) 1 and 2, in mice and monkeys. HAE1, also known as PRO98498, is structurally similar to omalizumab (Xolair®), a humanized anti-IgE IgG1 marketed for the treatment of asthma, but differs by 9 amino acid changes in the complementarity-determining region resulting in a 23-fold improvement in affinity. HAE2 is similar to HAE1, but its Fc region was altered to reduce binding to Fcγ receptors. As expected given the decreased binding to Fcγ receptors, systemic exposure to pre-formed HAE2:IgE complexes in mice was greater (six-fold) and distribution to the liver lower (four-fold) compared with HAE1:IgE complexes. In monkeys, systemic exposure to HAE1 was similar to that previously observed for omalizumab in this species, but required comparatively lower serum drug concentrations to suppress free IgE levels. HAE2 treatment resulted in greater exposure and greater increase of total IgE, relative to HAE1, because of decreased clearance of HAE2:IgE complexes. Overall, these data suggest that increased binding affinity to IgE may provide a more effective therapeutic for asthma patients, and that retaining FcγR binding of the anti-IgE antibody is important for elimination of anti-IgE:IgE complexes.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibody Affinity , Asthma/therapy , Receptors, Fc/metabolism , Animals , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibody Affinity/genetics , Antibody Affinity/immunology , Antigen-Antibody Complex/metabolism , Asthma/immunology , Haplorhini , Humans , Immunoglobulin E/immunology , Metabolic Clearance Rate , Mice , Omalizumab , Protein Binding/genetics , Receptors, Fc/genetics
3.
Sci Transl Med ; 3(84): 84ra43, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21613622

ABSTRACT

Reducing production of amyloid-ß (Aß) peptide by direct inhibition of the enzymes that process amyloid precursor protein (APP) is a central therapeutic strategy for treating Alzheimer's disease. However, small-molecule inhibitors of the ß-secretase (BACE1) and γ-secretase APP processing enzymes have shown a lack of target selectivity and poor penetrance of the blood-brain barrier (BBB). Here, we have developed a high-affinity, phage-derived human antibody that targets BACE1 (anti-BACE1) and is anti-amyloidogenic. Anti-BACE1 reduces endogenous BACE1 activity and Aß production in human cell lines expressing APP and in cultured primary neurons. Anti-BACE1 is highly selective and does not inhibit the related enzymes BACE2 or cathepsin D. Competitive binding assays and x-ray crystallography indicate that anti-BACE1 binds noncompetitively to an exosite on BACE1 and not to the catalytic site. Systemic dosing of mice and nonhuman primates with anti-BACE1 resulted in sustained reductions in peripheral Aß peptide concentrations. Anti-BACE1 also reduces central nervous system Aß concentrations in mouse and monkey, consistent with a measurable uptake of antibody across the BBB. Thus, BACE1 can be targeted in a highly selective manner through passive immunization with anti-BACE1, providing a potential approach for treating Alzheimer's disease. Nevertheless, therapeutic success with anti-BACE1 will depend on improving antibody uptake into the brain.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Antibodies/pharmacology , Antibodies/therapeutic use , Aspartic Acid Endopeptidases/antagonists & inhibitors , Amino Acid Sequence , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Antibodies/chemistry , Antibodies/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/metabolism , Biological Assay , Brain/drug effects , Brain/metabolism , Brain/pathology , Crystallography, X-Ray , Endocytosis/drug effects , Humans , Macaca fascicularis , Mice , Models, Molecular , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Peptide Library , Protein Binding/drug effects , Treatment Outcome
4.
MAbs ; 3(1): 61-6, 2011.
Article in English | MEDLINE | ID: mdl-20962582

ABSTRACT

The pharmacokinetics (PK) of therapeutic antibodies is determined by target and non-target mediated mechanisms. These antibody-specific factors need to be considered during prediction of human PK based upon preclinical information. Principles of allometric scaling established for small molecules using data from multiple animal species cannot be directly applied to antibodies. Here, different methods for projecting human clearance (CL) from animal PK data for 13 therapeutic monoclonal antibodies (mAbs) exhibiting linear PK over the tested dose ranges were examined: simple allometric scaling (CL versus body weight), allometric scaling with correction factors, allometric scaling based on rule of exponent and scaling from only cynomolgus monkey PK data. A better correlation was obtained between the observed human CL and the estimated human CL based on cynomolgus monkey PK data and an allometric scaling exponent of 0.85 for CL than other scaling approaches. Human concentration-time profiles were also reasonably predicted from the cynomolgus monkey data using species-invariant time method with a fixed exponent of 0.85 for CL and 1.0 for volume of distribution. In conclusion, we expanded our previous work and others and further confirmed that PK from cynomolgus monkey alone can be successfully scaled to project human PK profiles within linear range using simplify allometry and Dedrick plots with fixed exponent.


Subject(s)
Algorithms , Antibodies, Monoclonal/pharmacokinetics , Models, Biological , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Drug Evaluation, Preclinical , Humans , Macaca fascicularis , Metabolic Clearance Rate , Mice , Rats , Species Specificity
5.
J Clin Pharmacol ; 45(3): 286-98, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15703364

ABSTRACT

Efalizumab pharmacokinetics, pharmacodynamics, and efficacy were assessed after subcutaneous administration of 1.0 or 2.0 mg/kg/wk for 12 weeks with 12 weeks of follow-up in subjects with psoriasis. Steady-state serum concentrations were achieved by 4 and 8 weeks, respectively. C(max) was 12 and 31 microg/mL, occurring approximately 2 days after a SC dose. Serum trough levels were 9 and 24 microg/mL, and CL/F(ss) was 24 and 16 mL/kg/d. At both doses, CD11a expression on T lymphocytes was maximally down-modulated to approximately 20% of baseline, and CD11a binding sites were >95% saturated. The extent of this PD effect was less for other leukocytes. Leukocyte counts increased by approximately 40%, with the majority of this increase related to a significant but reversible increase in the lymphocyte population. Maximal pharmacodynamic effects were sustained at both dose levels through the course of treatment and were commensurate with improvements in psoriasis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD11a Antigen/immunology , Psoriasis/therapy , Adolescent , Adult , Aged , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antigen-Antibody Reactions , Down-Regulation , Female , Humans , Leukocyte Count , Male , Middle Aged , Psoriasis/blood , Psoriasis/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...