Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 138(14): 1258-1268, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34077951

RESUMO

Hemophilia A is a bleeding disorder resulting from deficient factor VIII (FVIII), which normally functions as a cofactor to activated factor IX (FIXa) that facilitates activation of factor X (FX). To mimic this property in a bispecific antibody format, a screening was conducted to identify functional pairs of anti-FIXa and anti-FX antibodies, followed by optimization of functional and biophysical properties. The resulting bispecific antibody (Mim8) assembled efficiently with FIXa and FX on membranes, and supported activation with an apparent equilibrium dissociation constant of 16 nM. Binding affinity with FIXa and FX in solution was much lower, with equilibrium dissociation constant values for FIXa and FX of 2.3 and 1.5 µM, respectively. In addition, the activity of Mim8 was dependent on stimulatory activity contributed by the anti-FIXa arm, which enhanced the proteolytic activity of FIXa by 4 orders of magnitude. In hemophilia A plasma and whole blood, Mim8 normalized thrombin generation and clot formation, with potencies 13 and 18 times higher than a sequence-identical analogue of emicizumab. A similar potency difference was observed in a tail vein transection model in hemophilia A mice, whereas reduction of bleeding in a severe tail-clip model was observed only for Mim8. Furthermore, the pharmacokinetic parameters of Mim8 were investigated and a half-life of 14 days shown in cynomolgus monkeys. In conclusion, Mim8 is an activated FVIII mimetic with a potent and efficacious hemostatic effect based on preclinical data.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemorragia/tratamento farmacológico , Animais , Fator IXa/antagonistas & inibidores , Fator VIIIa/uso terapêutico , Fator X/antagonistas & inibidores , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL
2.
Glycoconj J ; 30(3): 227-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22878593

RESUMO

Protein glycosylation often changes during cancer development, resulting in the expression of cancer-associated carbohydrate antigens. In particular mucins such as MUC1 are subject to these changes. We previously identified an immunodominant Tn-MUC1 (GalNAc-α-MUC1) cancer-specific epitope not covered by immunological tolerance in MUC1 humanized mice and man. The objective of this study was to determine if mouse antibodies to this Tn-MUC1 epitope induce antibody-dependent cellular cytotoxicity (ADCC) pivotal for their potential use in cancer immunotherapy. Binding affinity of mAb 5E5 directed to Tn-MUC1 was investigated using BiaCore. The availability of Tn-MUC1 on the surface of breast cancer cells was evaluated by immunohistochemistry, confocal microscopy, and flow cytometry, followed by in vitro assessment of antibody-dependent cellular cytotoxicity by mAb 5E5. Biacore analysis demonstrated high affinity binding (KD = 1.7 nM) of mAb 5E5 to its target, Tn-MUC1. Immunolabelling with mAb 5E5 revealed surface expression of the Tn-MUC1 epitope in breast cancer tissue and cell lines, and mAb 5E5 induced ADCC in two human breast cancer cell lines, MCF7 and T47D. Aberrantly glycosylated MUC1 is expressed on the surface of breast cancer cells and a target for antibody-dependent cell-mediated cytotoxicity suggesting that antibodies targeting glycopeptide epitopes on mucins are strong candidates for cancer-specific immunotherapies.


Assuntos
Adenocarcinoma/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Neoplasias da Mama/imunologia , Mucina-1/imunologia , Acetilgalactosamina/química , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Feminino , Glicosilação , Humanos , Células MCF-7 , Mucina-1/química , Mucina-1/metabolismo
3.
Front Pharmacol ; 3: 122, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22754528

RESUMO

Identification of targets for cancer therapy requires the understanding of the in vivo roles of proteins, which can be derived from studies using gene-targeted mice. An alternative strategy is the administration of inhibitory monoclonal antibodies (mAbs), causing acute disruption of the target protein function(s). This approach has the advantage of being a model for therapeutic targeting. mAbs for use in mouse models can be obtained through immunization of gene-deficient mice with the autologous protein. Such mAbs react with both species-specific epitopes and epitopes conserved between species. mAbs against proteins involved in extracellular proteolysis, including plasminogen activators urokinase plasminogen activator (uPA), tissue-type plasminogen activator (tPA), their inhibitor PAI-1, the uPA receptor (uPAR), two matrix metalloproteinases (MMP9 and MMP14), as well as the collagen internalization receptor uPARAP, have been developed. The inhibitory mAbs against uPA and uPAR block plasminogen activation and thereby hepatic fibrinolysis in vivo. Wound healing, another plasmin-dependent process, is delayed by an inhibitory mAb against uPA in the adult mouse. Thromboembolism can be inhibited by anti-PAI-1 mAbs in vivo. In conclusion, function-blocking mAbs are well-suited for targeted therapy in mouse models of different diseases, including cancer.

4.
Cancer Cell ; 21(4): 488-503, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22516258

RESUMO

Little is known about the dynamics of cancer cell death in response to therapy in the tumor microenvironment. Intravital microscopy of chemotherapy-treated mouse mammary carcinomas allowed us to follow drug distribution, cell death, and tumor-stroma interactions. We observed associations between vascular leakage and response to doxorubicin, including improved response in matrix metalloproteinase-9 null mice that had increased vascular leakage. Furthermore, we observed CCR2-dependent infiltration of myeloid cells after treatment and that Ccr2 null host mice responded better to treatment with doxorubicin or cisplatin. These data show that the microenvironment contributes critically to drug response via regulation of vascular permeability and innate immune cell infiltration. Thus, live imaging can be used to gain insights into drug responses in situ.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Cisplatino/farmacocinética , Cisplatino/uso terapêutico , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/efeitos dos fármacos , Células Mieloides/patologia , Receptores CCR2/genética , Receptores CCR2/fisiologia , Células Tumorais Cultivadas
5.
Curr Opin Cell Biol ; 22(5): 697-706, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20822891

RESUMO

The extracellular matrix (ECM) is a key regulator of cell and tissue function. Traditionally, the ECM has been thought of primarily as a physical scaffold that binds cells and tissues together. However, the ECM also elicits biochemical and biophysical signaling. Controlled proteolysis and remodeling of the ECM network regulate tissue tension, generate pathways for migration, and release ECM protein fragments to direct normal developmental processes such as branching morphogenesis. Collagens are major components of the ECM of which basement membrane type IV and interstitial matrix type I are the most prevalent. Here we discuss how abnormal expression, proteolysis and structure of these collagens influence cellular functions to elicit multiple effects on tumors, including proliferation, initiation, invasion, metastasis, and therapy response.


Assuntos
Colágeno/metabolismo , Neoplasias/fisiopatologia , Movimento Celular , Humanos , Neoplasias/metabolismo , Alicerces Teciduais
6.
Clin Chem ; 56(10): 1636-40, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20802096

RESUMO

BACKGROUND: The liberated domain I of the urokinase plasminogen activator receptor [uPAR(I)] is a significant prognostic marker in lung and ovarian cancer, although the uPAR(I) concentration is below the limit of quantification (LOQ) in a substantial proportion of patient samples (Lung Cancer 2005;48:349-55; Clin Cancer Res 2008;14:5785-93; APMIS 2009;117:755-61). This study was undertaken to design an immunoassay with improved functional sensitivity for measuring uPAR(I) and to evaluate the prognostic value of uPAR(I) for colorectal cancer (CRC) patients. METHODS: Surface plasmon resonance analysis identified 2 monoclonal antibodies, R3 and R20, that simultaneously bind to the liberated uPAR(I) but not to intact uPAR. We used R3 for capture and Eu-labeled R20 for detection in designing a 2-site sandwich time-resolved fluorescence immunoassay (TR-FIA 4) for measuring liberated uPAR(I). TR-FIA 4 was validated for use with citrated plasma. The prognostic value of the uPAR(I) concentration was evaluated in 298 CRC patients. The Cox proportional hazards model was used for the uni- and multivariate survival analyses. RESULTS: The LOQ was 0.65 pmol/L. Liberated uPAR(I) was measurable in all patient samples with TR-FIA 4. In the multivariate analysis that included sex, age, tumor stage, tumor localization, and adjuvant treatment, the uPAR(I) concentration measured with TR-FIA 4 (hazard ratio, 1.72; 95% CI, 1.15-2.57; P = 0.009), as well as the concentration of intact soluble uPAR plus the cleaved uPAR fragment containing domains II and III, tumor stage, and age were independent predictors of prognosis. CONCLUSIONS: TR-FIA 4 has a functional sensitivity improved 4-fold over that of the previous uPAR(I) assay. The uPAR(I) concentration measured with TR-FIA 4 is an independent predictor of prognosis in CRC patients.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Colorretais/diagnóstico , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/sangue , Neoplasias Colorretais/mortalidade , Feminino , Humanos , Imunoensaio , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Estrutura Terciária de Proteína , Sensibilidade e Especificidade
7.
Protein Expr Purif ; 72(1): 87-94, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20214994

RESUMO

Matrix metalloproteinase-9 (MMP-9) is a 92-kDa soluble pro-enzyme implicated in pathological events including cancer invasion. It is therefore an attractive target for therapeutic intervention studies in mouse models. Development of inhibitors requires sufficient amounts of correctly folded murine MMP-9. Constructs encoding zymogens of full-length murine MMP-9 and a version lacking the O-glycosylated linker region and hemopexin domains were therefore generated and expressed in stably transfected Drosophila S2 insect cells. After 7 days of induction the expression levels of the full-length and truncated versions were 5 mg/l and 2 mg/l, respectively. The products were >95% pure after gelatin Sepharose chromatography and possessed proteolytic activity when analyzed by gelatin zymography. Using the purified full-length murine MMP-9 we raised polyclonal antibodies by immunizations of rabbits. These antibodies specifically identified pro-MMP-9 in incisional skin wound extracts from mice when used for Western blotting. Immunohistochemical analysis of paraffin embedded skin wounds from mice showed that MMP-9 protein was localized at the leading-edge keratinocytes in front of the migrating epidermal layer. No immunoreactivity was observed when the antibody was probed against skin wound material from MMP-9 deficient mice. In conclusion, we have generated and purified two proteolytically active recombinant murine MMP-9 protein constructs, which are critical reagents for future cancer drug discovery studies.


Assuntos
Drosophila/genética , Expressão Gênica , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/isolamento & purificação , Murinae/genética , Animais , Anticorpos/imunologia , Linhagem Celular , Cromatografia de Afinidade , Drosophila/citologia , Metaloproteinase 9 da Matriz/análise , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Pele/metabolismo
8.
J Immunol Methods ; 339(1): 55-65, 2008 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18761343

RESUMO

The urokinase plasminogen activator receptor (uPAR) is a versatile three-domain GPI-anchored protein, which binds urokinase plasminogen activator (uPA) and thereby focalises plasminogen activation on the cell surface. Generation of a proteolytic potential is essential in both normal physiological and pathological extracellular tissue remodelling processes. uPA can also cleave uPAR, resulting in liberation of the amino-terminal domain I, which encompasses binding sites for both uPA and the adhesion molecule, vitronectin. In order to localise the different uPAR forms on the plasma membrane of murine monocyte macrophage-like P388D.1 cells, we have now generated and characterised two high-affinity murine mAbs, mR3 and mR4, raised against murine uPAR. mR3 was found to recognise an epitope located in domain I of uPAR. Surface plasmon resonance analyses and cell binding studies revealed that this mAb was able to bind preformed complexes of murine pro-uPA and murine uPAR. In contrast, mR4 recognises domains II-III in uPAR and does not bind preformed pro-uPA-uPAR complexes in similar analyses. Immunofluorescence microscopy of P388D.1 cells revealed that mR3 stained the cells equally well in the presence or absence of saturation with the amino-terminal fragment of uPA, ATF. However, the signal intensity obtained using another uPAR domain I specific mAb, mR1, was significantly reduced upon ATF saturation. Furthermore, when adding ATF, mR4 selectively stained the cleaved receptor. Applying these newly generated mAbs, we additionally demonstrated that cleaved and intact uPAR was evenly distributed on the surface of these cells.


Assuntos
Anticorpos Monoclonais/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Animais , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Sítios de Ligação de Anticorpos/imunologia , Linhagem Celular , Membrana Celular/química , Membrana Celular/imunologia , Humanos , Camundongos , Estrutura Terciária de Proteína , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Ressonância de Plasmônio de Superfície/métodos , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/imunologia
9.
J Biol Chem ; 283(47): 32506-15, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18799467

RESUMO

Urokinase-type plasminogen activator (uPA) plays a central role in tissue remodeling processes. Most of our understanding of the role of uPA in vivo is derived from studies using gene-targeted uPA-deficient mice. To enable in vivo studies on the specific interference with uPA functionality in mouse models, we have now developed murine monoclonal antibodies (mAbs) directed against murine uPA by immunization of uPA-deficient mice with the recombinant protein. Guided by enzyme-linked immunosorbent assay, Western blotting, surface plasmon resonance, and enzyme kinetic analyses, we have selected two highly potent and inhibitory anti-uPA mAbs (mU1 and mU3). Both mAbs recognize epitopes located on the B-chain of uPA that encompasses the catalytic site. In enzyme activity assays in vitro, mU1 blocked uPA-catalyzed plasminogen activation as well as plasmin-mediated pro-uPA activation, whereas mU3 only was directed against the first of these reactions. We additionally provide evidence that mU1, but not mU3, successfully targets uPA-dependent processes in vivo. Hence, systemic administration of mU1 (i) rescued mice treated with a uPA-activable anthrax protoxin and (ii) impaired uPA-mediated hepatic fibrinolysis in tissue-type plasminogen activator (tPA)-deficient mice, resulting in a phenotype mimicking that of uPA;tPA double deficient mice. Importantly, this is the first report demonstrating specific antagonist-directed targeting of mouse uPA at the enzyme activity level in a normal physiological process in vivo.


Assuntos
Fibrinólise , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Anticorpos/química , Anticorpos Monoclonais/química , Ensaio de Imunoadsorção Enzimática , Feminino , Cinética , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ligação Proteica , Proteínas Recombinantes/química , Ressonância de Plasmônio de Superfície
10.
Front Biosci ; 13: 6752-62, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508692

RESUMO

The cellular receptor for urokinase, uPAR, localizes its ligand, uPA, and thereby the plasminogen activation, to the cell surface. uPA also cleaves uPAR, liberating the ligand-binding domain I, and thereby inactivates the binding potential of uPAR for both uPA and vitronectin. The uPA-catalyzed cleavage of uPAR is fast on the cell surface, when uPA is bound to a neighboring uPAR molecule. uPAR can be shed from the cell surface. However, the soluble form cannot be cleaved by uPA. Glycolipid-anchored and soluble forms of intact, uPAR(I-III), and cleaved receptor, uPAR(II-III) and uPAR(I), have been identified in tissue and body fluids. It is well-established, that the total amount of all uPAR forms is a strong prognostic marker in different types of cancer. Using immunoassays, measuring the individual uPAR forms, has revealed that the cleaved uPAR forms are even stronger prognostic markers and have diagnostic utility. This review will focus on the mechanism of uPAR cleavage and the functional consequences, as well as the clinical applicability of cleaved uPAR forms.


Assuntos
Neoplasias/diagnóstico , Receptores de Superfície Celular/análise , Ativação Enzimática , Fibrinólise , Humanos , Neoplasias/fisiopatologia , Fragmentos de Peptídeos/análise , Plasminogênio/metabolismo , Prognóstico , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase
11.
Thromb Haemost ; 97(6): 1013-22, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17549305

RESUMO

Binding of urokinase plasminogen activator (uPA) to its cellular receptor, uPAR, potentiates plasminogen activation and localizes it to the cell surface. Focal plasminogen activation is involved in both normal and pathological tissue remodeling processes including cancer invasion. The interaction between uPA and uPAR therefore represents a potential target for anti-invasive cancer therapy. Inhibitors of the human uPA-uPAR interaction have no effect in the murine system. To enable in-vivo studies in murine cancer models we have now generated murine monoclonal antibodies (mAbs) against murine uPAR (muPAR) by immunizing uPAR-deficient mice with recombinant muPAR and screened for antibodies, which inhibit the muPA-muPAR interaction. Two of the twelve mAbs obtained, mR1 and mR2, interfered with the interaction between muPAR and the amino-terminal fragment of muPA (mATF) when analyzed by surface plasmon resonance. The epitope for mR1 is located on domain I of muPAR, while that of mR2 is on domains (II-III). In cell binding experiments using radiolabelled mATF, the maximal inhibition obtained with mR1 was 85% while that obtained with mR2 was 50%. The IC(50) value for mR1 was 0.67 nM compared to 0.14 nM for mATF. In an assay based on modified anthrax toxins, requiring cell-bound muPA activity for its cytotoxity, an approximately 50% rescue of the cells could be obtained by addition of mR1. Importantly, in-vivo efficacy of mR1 was demonstrated by the ability of mR1 to rescue mice treated with a lethal dose of uPA-activatable anthrax toxins.


Assuntos
Anticorpos Monoclonais/farmacologia , Macrófagos/efeitos dos fármacos , Plasminogênio/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Reações Antígeno-Anticorpo , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/toxicidade , Sítios de Ligação de Anticorpos , Ligação Competitiva , Linhagem Celular , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Mapeamento de Epitopos , Feminino , Humanos , Hibridomas/metabolismo , Imunização , Radioisótopos do Iodo/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...