Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharm ; 16(1): 86-95, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30444371

RESUMO

The collection of aqueous humor (phase 1 b/2 Mahalo study) from patients dosed intravitreally with anti-factor D (AFD; FCFD4514S, lampalizumab), a humanized antibody fragment previously under investigation to treat geographic atrophy (GA) secondary to age-related macular degeneration, presented a unique opportunity to examine AFD properties in clinical samples. We investigated AFD stability and target-binding characteristics to set up strategies for engineering and evaluating optimized molecules that enable less frequent dosing. Two variants, AFD.v8 and AFD.v14, were evaluated as alternatives to AFD for longer-acting treatments. Mass spectrometry, surface plasmon resonance, and immunoassay were used to assess AFD stability and binding activity in aqueous humor samples from Mahalo patients. In vitro stability and binding activity of AFD, AFD.v8, and AFD.v14 were assessed in human vitreous humor versus buffer at 37 °C over 16 weeks and in vivo in rabbits over 28 days along with pharmacokinetic determinations. In human aqueous humor, AFD specific binding was >85% through 30 days, and deamidation was <3% through 60 days, consistent with the AFD stability and binding activity in vitreous humor from humans in vitro and rabbits in vivo. Target binding, stability, and rabbit pharmacokinetic parameters of AFD.v8 and AFD.v14 were similar to those of AFD. Physiological stability and activity of AFD translated across in vitro and in vivo studies in humans and rabbits. The two variants AFD.v8 and AFD.v14 demonstrated comparable potency and pharmacokinetics. These findings, along with previously demonstrated improved solubility of AFD.v8 and AFD.v14, provide proof-of-concept for developing other similar long-acting therapeutic variants.


Assuntos
Humor Aquoso/metabolismo , Fator D do Complemento/antagonistas & inibidores , Fragmentos Fab das Imunoglobulinas/metabolismo , Animais , Atrofia Geográfica/metabolismo , Humanos , Imunoensaio , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Degeneração Macular/metabolismo , Masculino , Espectrometria de Massas , Coelhos , Ressonância de Plasmônio de Superfície , Corpo Vítreo/metabolismo
2.
Eur J Pharm Biopharm ; 101: 53-61, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26820920

RESUMO

Treatment of diseases of the central nervous system by monoclonal antibodies may be limited by the restricted uptake of antibodies across the blood-brain barrier (BBB). An antibody targeting transferrin receptor (TfR) has been shown to take advantage of the receptor-mediated transcytosis properties of TfR in order to cross the BBB in mice, with the uptake in the brain being dependent on the affinity to TfR. In the bispecific format with arms targeting both TfR and ß-secretase 1 (BACE1), altering the affinity to TfR has been shown to impact systemic exposure and safety profiles. In this work, a mathematical model incorporating pharmacokinetic/pharmacodynamic (PKPD) and safety profiles is developed for bispecific TfR/BACE1 antibodies with a range of affinities to TfR in order to guide candidate selection. The model captures the dependence of both systemic and brain exposure on TfR affinity and the subsequent impact on brain Aß40 lowering and circulating reticulocyte levels. Model simulations identify the optimal affinity for the TfR arm of the bispecific to maximize Aß reduction while maintaining reticulocyte levels. The model serves as a useful tool to prioritize and optimize preclinical studies and has been used to support the selection of additional candidates for further development.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/farmacocinética , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/metabolismo , Receptores da Transferrina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Transporte Biológico/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Camundongos , Modelos Teóricos , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , Transcitose/efeitos dos fármacos , Transferrina/metabolismo
3.
Nat Biotechnol ; 30(2): 184-9, 2012 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-22267010

RESUMO

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


Assuntos
Anticorpos/sangue , Anticorpos/imunologia , Sítios de Ligação de Anticorpos/imunologia , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoglobulina G/química , Engenharia de Proteínas , Aminobenzoatos/química , Aminobenzoatos/imunologia , Animais , Anticorpos/química , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular , Cisteína/química , Humanos , Imunoconjugados/administração & dosagem , Imunoglobulina G/imunologia , Macaca fascicularis , Maleimidas/química , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/imunologia , Maitansina/química , Maitansina/imunologia , Camundongos , Camundongos Nus , Modelos Moleculares , Oligopeptídeos/química , Oligopeptídeos/imunologia , Conformação Proteica , Ratos , Relação Estrutura-Atividade , Compostos de Sulfidrila/química , Trastuzumab
4.
Sci Transl Med ; 3(84): 84ra44, 2011 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-21613623

RESUMO

Monoclonal antibodies have therapeutic potential for treating diseases of the central nervous system, but their accumulation in the brain is limited by the blood-brain barrier (BBB). Here, we show that reducing the affinity of an antibody for the transferrin receptor (TfR) enhances receptor-mediated transcytosis of the anti-TfR antibody across the BBB into the mouse brain where it reaches therapeutically relevant concentrations. Anti-TfR antibodies that bind with high affinity to TfR remain associated with the BBB, whereas lower-affinity anti-TfR antibody variants are released from the BBB into the brain and show a broad distribution 24 hours after dosing. We designed a bispecific antibody that binds with low affinity to TfR and with high affinity to the enzyme ß-secretase (BACE1), which processes amyloid precursor protein into amyloid-ß (Aß) peptides including those associated with Alzheimer's disease. Compared to monospecific anti-BACE1 antibody, the bispecific antibody accumulated in the mouse brain and led to a greater reduction in brain Aß after a single systemic dose. TfR-facilitated transcytosis of this bispecific antibody across the BBB may enhance its potency as an anti-BACE1 therapy for treating Alzheimer's disease.


Assuntos
Anticorpos/metabolismo , Anticorpos/uso terapêutico , Afinidade de Anticorpos/imunologia , Encéfalo/metabolismo , Receptores da Transferrina/imunologia , Transcitose/imunologia , Peptídeos beta-Amiloides/biossíntese , Animais , Anticorpos/administração & dosagem , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/farmacocinética , Anticorpos Biespecíficos/uso terapêutico , Vasos Sanguíneos/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Células HEK293 , Humanos , Camundongos , Modelos Biológicos , Transporte Proteico
5.
Ther Deliv ; 2(9): 1121-3, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22833909

RESUMO

Leading off the event, specialized workshops with panels of experts were conducted that brough together students and scientists to the current advances in their specialty. Delivery of therapeutics across the blood-brain barrier for the treatment of neurological conditions such as pain, Alzheimer's and Parkinson's disease is a field of great development with an expanding aging population of patients. Manoj Rajadhyaksha, Pfizer, Inc. and Saileta Prabhu, Genentech, Inc., brought together a panel describing the anatomy of the CNS and improvements in drug delivery. The conference that followed expanded upon the current approaches.


Assuntos
Biotecnologia/métodos , Doenças do Sistema Nervoso Central/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Animais , Barreira Hematoencefálica/metabolismo , Sistema Nervoso Central/fisiologia , Sistema Nervoso Central/fisiopatologia , Doenças do Sistema Nervoso Central/fisiopatologia , Humanos
6.
Drug Metab Dispos ; 38(12): 2309-19, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20823292

RESUMO

3A5 is a novel antibody that binds repeated epitopes within CA125, an ovarian tumor antigen that is shed into the circulation. Binding to shed antigen may limit the effectiveness of therapeutic antibodies because of unproductive immune complex (IC) formation and/or altered antibody distribution. To evaluate this possibility, we characterized the impact of shed CA125 on the in vivo distribution of 3A5. In vitro, 3A5 and CA125 were found to form ICs in a concentration-dependent manner. This phenomenon was then evaluated in vivo using quantitative whole-body autoradiography to assess the tissue distribution of (125)I-3A5 in an orthotopic OVCAR-3 tumor mouse model at different stages of tumor burden. Low doses of 3A5 (75 µg/kg) and pathophysiological levels of shed CA125 led to the formation of ICs in vivo that were rapidly distributed to the liver. Under these conditions, increased clearance of 3A5 from normal tissues was observed in mice bearing CA125-expressing tumors. Of importance, despite IC formation, 3A5 uptake by tumors was sustained over time. At a therapeutically relevant dose of 3A5 (3.5 mg/kg), IC formation was undetectable and distribution to normal tissues followed that of blood. In contrast, increased levels of radioactivity were observed in the tumors. These data demonstrate that CA125 and 3A5 do form ICs in vivo and that the liver is involved in their uptake. However, at therapeutic doses of 3A5 and clinically relevant CA125 levels, IC formation consumes only a minor fraction of 3A5, and tumor targeting seems to be unaffected.


Assuntos
Anticorpos/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Antígeno Ca-125/imunologia , Neoplasias Ovarianas/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Distribuição Tecidual
7.
J Med Chem ; 50(6): 1146-57, 2007 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-17315988

RESUMO

By the screening of a combinatorial library for inhibitors of nitric oxide (NO) formation by the inducible isoform of nitric oxide synthase (iNOS) using a whole-cell assay, 2-(imidazol-1-yl)pyrimidines were identified. Compounds were found to inhibit the dimerization of iNOS monomers, thus preventing the formation of the dimeric, active form of the enzyme. Optimization led to the selection of the potent, selective, and orally available iNOS dimerization inhibitor, 21b, which significantly ameliorated adjuvant-induced arthritis in a rat model. Analysis of the crystal structure of the 21b--iNOS monomer complex provided a rationalization for both the SAR and the mechanism by which 21b blocks the formation of the protein--protein interaction present in the dimeric form of iNOS.


Assuntos
Anti-Inflamatórios não Esteroides/síntese química , Benzodioxóis/síntese química , Imidazóis/síntese química , Óxido Nítrico Sintase Tipo II/metabolismo , Pirimidinas/síntese química , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Artrite Experimental/terapia , Benzodioxóis/química , Benzodioxóis/farmacologia , Linhagem Celular , Chlorocebus aethiops , Cristalografia por Raios X , Dimerização , Imidazóis/química , Imidazóis/farmacologia , Masculino , Modelos Moleculares , Pirimidinas/química , Pirimidinas/farmacologia , Ratos , Ratos Endogâmicos Lew
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA