Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Biochem Soc Trans ; 30(4): 487-90, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12196120

RESUMEN

The binding sites on human IgG1 for human Fc gamma receptor (Fc gamma R) I, Fc gamma RIIa, Fc gamma RIIb, Fc gamma RIIIa and neonatal FcR have been mapped. A common set of IgG1 residues is involved in binding to all Fc gamma Rs, while Fc gamma RII and Fc gamma RIII utilize distinct sites outside this common set. In addition to residues which abrogated binding to the Fc gamma R, several positions were found which improved binding only to specific Fc gamma Rs or simultaneously improved binding to one type of Fc gamma R and reduced binding to another type. Selected IgG1 variants with improved binding to Fc gamma RIIIa were then tested in an in vitro antibody-dependent cellular cytotoxicity (ADCC) assay and showed an enhancement in ADCC when either peripheral blood mononuclear cells or natural killer cells were used.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Ingeniería Genética/métodos , Receptores de IgG/inmunología , Anticuerpos Monoclonales/genética , Citotoxicidad Celular Dependiente de Anticuerpos , Linfocitos B/inmunología , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Células Asesinas Naturales/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
2.
Surg Endosc ; 16(10): 1494-5, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12098030

RESUMEN

Imaging of the gallbladder and biliary tract has changed dramatically in the past 20 years. Magnetic resonancecholangiopancreatography provides a noninvasive alternative to endoscopic retrograde cholangiopancreatography and percutaneous transhepatic cholangiography in the diagnosis of Mirizzi syndrome. In this laparoscopic era, when diagnosis is certain, surgeons must choose between a laparoscopic and a traditional open approach. The authors review their cases of hepatobiliary surgery during the period 1993-2000. Three cases of Mirizzi syndrome (0.4%) were observed among 712 surgical hepatobiliary patients (two type 1 cases and one type 2 case). The authors suggest that with Mirizzi syndrome type 1, laparoscopy together with peroperative cholangiography should be used to resolve anatomic doubts. If clipping of the cystic duct is possible and certain, then laparoscopy may be continued and finished. In the case of cholecystocholedochal fistula (Mirizzi syndrome type 2), when the diagnosis is determined before surgery, the authors believe that laparoscopy is dangerous. Adhesions, inflammation, and anatomy changes may cause injuries to the main bile duct, so an open traditional approach is suggested.


Asunto(s)
Colangiopancreatografia Retrógrada Endoscópica/métodos , Colelitiasis/complicaciones , Colelitiasis/cirugía , Colestasis Extrahepática/diagnóstico , Colestasis Extrahepática/cirugía , Laparoscopía/métodos , Imagen por Resonancia Magnética/métodos , Colestasis Extrahepática/etiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndrome
3.
Science ; 295(5557): 1086-9, 2002 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11834839

RESUMEN

The protein-protein interaction between leukocyte functional antigen-1 (LFA-1) and intercellular adhesion molecule-1 (ICAM-1) is critical to lymphocyte and immune system function. Here, we report on the transfer of the contiguous, nonlinear epitope of ICAM-1, responsible for its association with LFA-1, to a small-molecule framework. These LFA-1 antagonists bound LFA-1, blocked binding of ICAM-1, and inhibited a mixed lymphocyte reaction (MLR) with potency significantly greater than that of cyclosporine A. Furthermore, in comparison to an antibody to LFA-1, they exhibited significant anti-inflammatory effects in vivo. These results demonstrate the utility of small-molecule mimics of nonlinear protein epitopes and the protein epitopes themselves as leads in the identification of novel pharmaceutical agents.


Asunto(s)
Inmunosupresores/farmacología , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Tiofenos/síntesis química , Tiofenos/farmacología , beta-Alanina/síntesis química , beta-Alanina/farmacología , Secuencia de Aminoácidos , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Ciclosporina/farmacología , Dermatitis Irritante/tratamiento farmacológico , Dinitrofluorobenceno , Diseño de Fármacos , Ensayo de Inmunoadsorción Enzimática , Epítopos , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/farmacología , Inmunosupresores/síntesis química , Inmunosupresores/química , Inmunosupresores/metabolismo , Molécula 1 de Adhesión Intercelular/química , Prueba de Cultivo Mixto de Linfocitos , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Ratones Endogámicos BALB C , Imitación Molecular , Mutagénesis , Estructura Secundaria de Proteína , Relación Estructura-Actividad , Tiofenos/química , Tiofenos/metabolismo , beta-Alanina/análogos & derivados , beta-Alanina/química , beta-Alanina/metabolismo
4.
G Chir ; 23(11-12): 405-12, 2002.
Artículo en Italiano | MEDLINE | ID: mdl-12652913

RESUMEN

Pancreaticoduodenectomy represents the only therapeutic option for cefalo-pancreatic and periampullary cancers. Surgical and anaesthesiological techniques development over the last twenty years has granted an operative mortality decrease. However, surgical morbidity is still high, with an incidence of 30-50%. A 20 year experience of a single Centre is examined retrospectively: 121 patients underwent pancreatic resection with radical intent. Type of operation or re-operation, operative mortality within 30 days, general and surgical morbidity, postoperative hospital stay were analysed. Average recovery time was 24 days (range 12-65); operative mortality was 5.8% (7/121); general morbidity, including medical and surgical complications, was observed in 47 patients (38.8%). Pancreatic fistula occurred in 16 patients (13.2%); ten of these underwent a second operation. Patients who underwent pancreaticoduodenctomy were divided as follows: 76 pts. received a pylours-preserving pancreaticoduodenectomy and 45 a Whipple's resection. Neither surgical complications incidence nor mortality rate were significantly different between the two groups. Postoperative complications following pancreaticoduodenectomy are still frequent and severe. In particular, pancreatic fistula represents the most relevant complication following pancreaticoduodenectomy. The Authors suggest that standard and meticulous surgical procedures together with continued efforts to improve postoperative follow-up, support early detection of complications and improvement of results in most patients.


Asunto(s)
Pancreaticoduodenectomía/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Estudios Retrospectivos
5.
Cytokine ; 16(3): 106-19, 2001 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-11741351

RESUMEN

A neutralizing anti-interleukin-(IL-)8 monoclonal antibody was humanized by grafting the complementary determining regions onto the human IgG framework. Subsequent alanine scanning mutagenesis and phage display enabled the production of an affinity matured antibody with a >100-fold improvement in IL-8 binding. Antibody fragments can be efficiently produced in Escherichia coli but have the limitation of rapid clearance rates in vivo. The Fab' fragment of the antibody was therefore modified with polyethylene glycol (PEG) in order to obtain a more desirable pharmacokinetic profile. PEG (5-40 kDa) was site-specifically conjugated to the Fab' via the single free cysteine residue in the hinge region. In vitro binding and bioassays showed little or no loss of activity. The pharmacokinetic profiles of the 20 kDa, 30 kDa, 40 kDa, and 40 kDa branched PEG-Fab' molecules were evaluated in rabbits. Relative to the native Fab', the clearance rates of the PEGylated molecules were decreased by 44-175-fold. In a rabbit ear model of ischemia/reperfusion injury, all PEGylated Fab' molecules were as efficacious in reducing oedema as the original monoclonal antibody. These studies demonstrate that it is possible to customize the pharmacokinetic properties of a Fab' while retaining its antigen binding activity.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Inmunoglobulina G/inmunología , Interleucina-8/inmunología , Interleucina-8/farmacocinética , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Alanina/metabolismo , Animales , Anticuerpos Monoclonales/química , Reacciones Antígeno-Anticuerpo , ADN Complementario/metabolismo , Edema/terapia , Electroforesis en Gel de Poliacrilamida , Humanos , Concentración 50 Inhibidora , Interleucina-8/metabolismo , Cinética , Ratones , Mutagénesis , Biblioteca de Péptidos , Unión Proteica , Conejos , Proteínas Recombinantes de Fusión/metabolismo , Daño por Reperfusión , Factores de Tiempo , Tripsina/farmacología
6.
J Mol Biol ; 313(1): 83-97, 2001 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-11601848

RESUMEN

The outstanding importance of the antigen-antibody recognition process for the survival and defence strategy of higher organisms is in sharp contrast to the limited high resolution structural data available on antibody-antigen pairs with antigenic proteins. The limitation is the most severe for structural data not restricted to the antigen-antibody complex but extending to the uncomplexed antigen and antibody. We report the crystal structure of the complex between tissue factor (TF) and the humanized Fab fragment D3h44 at a resolution of 1.85 A together with the structure of uncomplexed D3h44 at the same resolution. In conjunction with the previously reported 1.7 A crystal structure of uncomplexed TF, a unique opportunity is generated to explore details of the recognition process. The TF.D3h44 interface is characterised by a high number of polar interactions, including as may as 46 solvent molecules. Conformational changes upon complex formation are very small and almost exclusively limited to the reorientation of side-chains. The binding epitope is in complete agreement with earlier mutagenesis experiments. A revaluation of two other antibody-antigen pairs reported at similar resolutions, shows that all these complexes are very similar with respect to the solvation of the interface, the number of solvent positions conserved in the uncomplexed and complexed proteins and the number of water molecules expelled from the surface and replaced by hydrophilic atoms from the binding partner upon complex formation. A strategy is proposed on how to exploit this high resolution structural data to guide the affinity maturation of humanised antibodies.


Asunto(s)
Sitios de Unión de Anticuerpos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Tromboplastina/química , Tromboplastina/inmunología , Agua/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Solventes , Tromboplastina/metabolismo , Agua/química
7.
Cytokine ; 15(5): 250-60, 2001 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-11594789

RESUMEN

We have developed a panel of murine monoclonal antibodies that recognize human interferon alpha. One of these mononclonal antibodies binds and neutralizes, with high affinity, all of seven tested recombinant human interferon alphas. This mononclonal antibody also neutralizes the interferon activity present in two independent pools of interferon alphas prepared following stimulation of human peripheral blood leukocytes. The complementary determining regions from this murine mononclonal antibody were transferred to a human IgG2 heavy chain and to a human kappa1 light chain. In addition, six (heavy chain) and two (light chain) amino acids were transferred from the framework regions. This generated a humanized mononclonal antibody that retained the specificity of the mouse parent. The humanized anti-interferon alpha antibody is a candidate therapeutic for those diseases, such as insulin-dependent diabetes, systemic lupus erythematosis, psoriasis and Crohn's disease, which are all characterized by pathological expression of interferon alpha.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Interferón-alfa/química , Lupus Eritematoso Sistémico/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Técnicas Biosensibles , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Células HeLa , Humanos , Inmunoglobulina G/química , Interferón-alfa/inmunología , Interferón beta/química , Interferón beta/inmunología , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo
8.
Thromb Haemost ; 85(3): 379-89, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11307801

RESUMEN

Blocking the cofactor function of human tissue factor may be beneficial in various coagulation-mediated diseases. The murine antibody D3 binds to the membrane proximal substrate interaction region of human tissue factor and blocks tissue factor function even in the presence of bound factor VIIa. The cloned murine D3 antibody was humanized and affinity matured by exchanging amino acids in the complementarity determining regions as well as in the antibody framework. The humanized antibody, D3H44, bound to tissue factor with a 100-fold increased affinity (KD 0.1 nM) as compared to the original murine and chimeric versions. Depending on the particular disease, different pharmacokinetic properties of the antibody may be required and, therefore, several antibody variants-- F(ab), F(ab')2, IgG2, IgG4 and IgG4b-were generated. In vitro, the humanized D3 antibodies displayed potent inhibition of plasma clotting and tissue factor: factor VIIa-mediated activation of factors IX and X (e.g. D3H44-F(ab')2, IC50(F.X) 47 pM). In addition, D3H44-F(ab')2 completely prevented fibrin deposition in a human ex vivo thrombosis model under venous blood flow conditions (IC50 37 nM). The humanized D3 antibodies may be utilized for treatment of cardiovascular diseases which involve tissue factor activity, e.g. acute coronary syndrome and venous thrombosis.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Afinidad de Anticuerpos/inmunología , Tromboplastina/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Pruebas de Coagulación Sanguínea , Clonación Molecular , Factor IX/efectos de los fármacos , Factor IX/metabolismo , Factor X/efectos de los fármacos , Factor X/metabolismo , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Humanos , Fragmentos de Inmunoglobulinas/genética , Ratones , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Alineación de Secuencia
9.
J Immunol ; 166(4): 2571-5, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11160318

RESUMEN

This manuscript describes two sites in a human IgG1 that, when mutated individually or in combination, result in a dramatic increase in C1q binding and complement-dependent cytotoxicity activity. These two residues, K326 and E333, are located at the extreme ends of the C1q binding epicenter in the C(H)2 domain of a human IgG. A mutation to tryptophan at K326 debilitates Ab-dependent cell-mediated cytotoxicity activity. In addition, substitutions of the residues E333 with serine and of K326 with tryptophan in a human IgG2 confer biological activity in the complement-dependent cytotoxicity assay in which the wild-type IgG2 is inactive. This study reveals that the residues K326 and E333 play a significant role in the control of the biological activity of an IgG molecule and can rescue the activity of an inactive IgG isotype.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Activación de Complemento/genética , Complemento C1q/inmunología , Complemento C1q/metabolismo , Mutagénesis Sitio-Dirigida , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales de Origen Murino , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Antígenos CD20/inmunología , Antígenos CD20/metabolismo , Sitios de Unión de Anticuerpos/genética , Carbohidratos/análisis , Línea Celular Transformada , Ácido Glutámico/genética , Glicosilación , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Isotipos de Inmunoglobulinas/genética , Isotipos de Inmunoglobulinas/metabolismo , Isotipos de Inmunoglobulinas/farmacología , Lisina/genética , Rituximab
10.
J Biol Chem ; 276(9): 6591-604, 2001 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-11096108

RESUMEN

Immunoglobulin G (IgG) Fc receptors play a critical role in linking IgG antibody-mediated immune responses with cellular effector functions. A high resolution map of the binding site on human IgG1 for human Fc gamma RI, Fc gamma RIIA, Fc gamma RIIB, Fc gamma RIIIA, and FcRn receptors has been determined. A common set of IgG1 residues is involved in binding to all Fc gamma R; Fc gamma RII and Fc gamma RIII also utilize residues outside this common set. In addition to residues which, when altered, abrogated binding to one or more of the receptors, several residues were found that improved binding only to specific receptors or simultaneously improved binding to one type of receptor and reduced binding to another type. Select IgG1 variants with improved binding to Fc gamma RIIIA exhibited up to 100% enhancement in antibody-dependent cell cytotoxicity using human effector cells; these variants included changes at residues not found at the binding interface in the IgG/Fc gamma RIIIA co-crystal structure (Sondermann, P., Huber, R., Oosthuizen, V., and Jacob, U. (2000) Nature 406, 267-273). These engineered antibodies may have important implications for improving antibody therapeutic efficacy.


Asunto(s)
Inmunoglobulina G/química , Receptores de IgG/química , Citotoxicidad Celular Dependiente de Anticuerpos , Sitios de Unión , Cristalización , Glicosilación , Humanos , Inmunoglobulina G/clasificación , Inmunoglobulina G/metabolismo , Ingeniería de Proteínas , Receptores de IgG/metabolismo , Relación Estructura-Actividad
11.
Transplantation ; 69(10): 2005-13, 2000 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-10852588

RESUMEN

INTRODUCTION: Leukocyte function-associated antigen-1 (LFA-1, CD11a) monoclonal antibody (mAb) affects many leukocyte functions without cell depletion. We hypothesized that the use of a humanized, anti-rhesus modified LFA-1 mAb (H2C12) in rhesus monkeys would cause: (1) prolonged heart allograft survival, (2) inhibition of primary but not secondary antibody responses, and (3) minimal drug toxicity. METHODS AND RESULTS: Control (n=5) and H2C12-treated (n=7) (8-20 mg/kg i.v. on day -1 followed by 10 mg/kg/day) adult male rhesus recipients were inoculated with GP120 protein antigen on day -28 and -1 and grafted with heterotopic abdominal hearts (day 0). Donor-recipient pairs were equally MLR mismatched (4329.8+/-1124.1 CPM controls vs. 7289.0+/-1926.5 treated, P=NS). Mean heart allograft survival as evaluated by daily abdominal palpation was significantly prolonged in high dose recipients (23.0+/-2.6, n=4) vesus controls (8.2+/-1.3, n=5, P<0.02, Mann-Whitney U test). H2C12 treatment did not produce signs of cytokine release or toxicity, was nondepleting, but down-modulated PBL CD11a expression to 43.4+/-3.6% (n=4) of control levels (n=5) at day 7 as demonstrated by flow cytometry. It had no effect on postoperative Con A or MLR and did not prevent mAb clearance due to the rhesus-antihuman antibody response. The addition of mycophenolate mofitil prevented rhesus-antihuman antibody response with therapeutic H2C12 levels seen for >35 days. CONCLUSIONS: The use of this mAb to block CD11a had the benefit of being a well tolerated, highly targeted therapy. These are the first results showing that monotherapy with anti-leukocyte function-associated antigen-1 mAb prolonged survival of MLR mismatched allogenic cardiac grafts in primates.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Animales , Anticuerpos Monoclonales/farmacocinética , Prueba de Histocompatibilidad , Humanos , Terapia de Inmunosupresión/métodos , Inmunosupresores/uso terapéutico , Recuento de Linfocitos , Macaca mulatta , Masculino , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Daño por Reperfusión Miocárdica , Factores de Tiempo , Trasplante Homólogo
12.
J Biol Chem ; 275(11): 7870-7, 2000 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-10713102

RESUMEN

Neurotrophins are important for the development and maintenance of the vertebrate nervous system, mediating their signal into the cell by specific interaction with tyrosine kinase receptors of the Trk family. The extracellular portion of the Trk receptors has been previously proposed to consist of a cysteine-rich motif, a leucine-rich motif, a second cysteine-rich motif followed by two immunoglobulin-like domains. Earlier studies have shown that a major neurotrophin-binding site in the Trk receptors resides in the second immunoglobulin-like domain. Although the individual amino acids in TrkA involved in binding to nerve growth factor (NGF) and those in TrkC involved in binding to neurotrophin-3 have been mapped in this domain, the Trk amino acids that provide specificity remained unclear. In this study, a minimum set of residues in the human TrkC second immunoglobulin-like domain, which does not bind nerve growth factor (NGF), were substituted with those from human TrkA. The resulting Trk variant recruited binding of NGF equivalent to TrkA, maintained neurotrophin-3 binding equivalent to TrkC, and also bound brain-derived neurotrophin, although with lower affinity compared with TrkB. This implies that the amino acids in the second immunoglobulin-like domain that determine Trk specificity are distinct for each Trk.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Receptor trkC/metabolismo , Secuencia de Aminoácidos , Aminoácidos , Sitios de Unión/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Neurotrofina 3/metabolismo , Unión Proteica , Ingeniería de Proteínas , Receptor trkA/genética , Receptor trkC/genética , Homología de Secuencia de Aminoácido
13.
Nat Med ; 6(4): 443-6, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10742152

RESUMEN

Inhibitory receptors have been proposed to modulate the in vivo cytotoxic response against tumor targets for both spontaneous and antibody-dependent pathways. Using a variety of syngenic and xenograft models, we demonstrate here that the inhibitory FcgammaRIIB molecule is a potent regulator of antibody-dependent cell-mediated cytotoxicity in vivo, modulating the activity of FcgammaRIII on effector cells. Although many mechanisms have been proposed to account for the anti-tumor activities of therapeutic antibodies, including extended half-life, blockade of signaling pathways, activation of apoptosis and effector-cell-mediated cytotoxicity, we show here that engagement of Fcgamma receptors on effector cells is a dominant component of the in vivo activity of antibodies against tumors. Mouse monoclonal antibodies, as well as the humanized, clinically effective therapeutic agents trastuzumab (Herceptin(R)) and rituximab (Rituxan(R)), engaged both activation (FcgammaRIII) and inhibitory (FcgammaRIIB) antibody receptors on myeloid cells, thus modulating their cytotoxic potential. Mice deficient in FcgammaRIIB showed much more antibody-dependent cell-mediated cytotoxicity; in contrast, mice deficient in activating Fc receptors as well as antibodies engineered to disrupt Fc binding to those receptors were unable to arrest tumor growth in vivo. These results demonstrate that Fc-receptor-dependent mechanisms contribute substantially to the action of cytotoxic antibodies against tumors and indicate that an optimal antibody against tumors would bind preferentially to activation Fc receptors and minimally to the inhibitory partner FcgammaRIIB.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD/inmunología , Linfoma de Células B/inmunología , Melanoma Experimental/inmunología , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Antígenos CD/genética , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Femenino , Humanos , Inmunización Pasiva , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptores de IgG/genética , Rituximab , Trastuzumab
14.
J Immunol ; 164(8): 4178-84, 2000 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10754313

RESUMEN

Rituxan (Rituximab) is a chimeric mAb with human IgG1 constant domains used in the therapy of non-Hodgkin's B cell lymphomas. This Ab targets B cells by binding to the cell-surface receptor, CD20. In our investigation of the mechanism of B cell depletion mediated by Rituximab, we first constructed mutants of Rituximab defective in complement activation but with all other effector functions intact. Our results demonstrate that the previously described C1q binding motif in murine IgG2b constituting residues E318, K320, and K322 is not applicable to a human IgG1 when challenged with either human, rabbit, or guinea pig complement. Alanine substitution at positions E318 and K320 in Rituximab had little or no effect on C1q binding and complement activation, whereas alanine substitution at positions D270, K322, P329, and P331 significantly reduced the ability of Rituximab to bind C1q and activate complement. We have also observed that concentrations of complement approaching physiological levels are able to rescue >60% of the activity of these mutant Abs with low affinity for C1q. These data localize the C1q binding epicenter on human IgG1 and suggest that there are species-specific differences in the C1q binding site of Igs.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Sitios de Unión de Anticuerpos , Complemento C1q/inmunología , Complemento C1q/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Alanina/genética , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino , Afinidad de Anticuerpos/genética , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Ácido Aspártico/genética , Sitios de Unión de Anticuerpos/genética , Complemento C1q/química , Cobayas , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/química , Inmunoglobulina G/genética , Lisina/genética , Ratones , Mapeo Peptídico/métodos , Fenilalanina/genética , Conejos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Rituximab
15.
J Biol Chem ; 273(44): 28937-44, 1998 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-9786897

RESUMEN

By extensive mutagenic analysis of the inserted domain (I-domain) of the alpha-chain (CD11a) of the leukocyte function-associated antigen-1 (LFA-1), we have defined a putative binding surface for intercellular adhesion molecules 1 and 2 (ICAM-1 and -2). This analysis showed that individually mutating Leu-205 or Glu-241 to alanine completely abolished LFA-1 binding to ICAM-1 or -2 without affecting I-domain structure, as assayed by antibody binding. Mutating Thr-243 to alanine also had a profound effect on LFA-1 binding to ICAM-1 and -2, as seen by complete loss of binding to ICAM-1 and a significant reduction (70% decrease) in binding to ICAM-2. Mutating Glu-146 to alanine reduced LFA-1 binding to ICAM-1 or -2 by 70%, and mutating His-264 or Glu-293 to alanine reduced binding to ICAM-1 or -2 by about 30-40%. Mutating Thr-175 to alanine reduced binding to ICAM-1 by about 30% and binding to ICAM-2 by about 70%. Interestingly, mutating Lys-263 to alanine preferentially abolished LFA-1 binding to ICAM-2. Using these data, we have generated a model of the interface between the LFA-1 I-domain and residues in the first domain of ICAM-1 that have been shown to be critical for this interaction. In addition, this model, together with the ICAM-2 crystal structure, has been used to map residues that are likely to mediate LFA-1 I-domain binding to ICAM-2.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Secuencia de Aminoácidos , Antígenos CD/química , Antígenos CD/genética , Sitios de Unión , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Epítopos/metabolismo , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/genética , Antígeno-1 Asociado a Función de Linfocito/química , Antígeno-1 Asociado a Función de Linfocito/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis
16.
Protein Eng ; 11(4): 321-8, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9680194

RESUMEN

A murine monoclonal antibody to the zymogen form of human protein C that blocks protein C activation by thrombin-thrombomodulin both in vitro and in vivo has been humanized using the consensus and resurfacing methods. While the binding of the parent murine antibody to protein C is calcium-dependent (1.5-2-fold decrease in binding without calcium), the humanized antibody exhibited a significant increase in its calcium-dependence (5-fold decrease in binding without calcium). Two exposed human framework residues in the variable light domain of the humanized antibody, aspartic acid L1 and glutamine L3, are responsible for the increase in calcium-dependence.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Calcio/metabolismo , Proteína C/inmunología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/química , Clonación Molecular , Simulación por Computador , ADN Complementario , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido
17.
Nat Biotechnol ; 16(7): 677-81, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9661204

RESUMEN

Production of bispecific IgG (BsIgG) by coexpressing two different antibodies is inefficient due to unwanted pairings of the component heavy and light chains. To overcome this problem, heavy chains were remodeled for heterodimerization using engineered disulfide bonds in combination with previously identified "knobs-into-holes" mutations. One of the variants, S354C:T366W/Y349'C:T366'S:L368'A:Y407++ +'V, gave near quantitative (approximately 95%) heterodimerization. Light chain mispairing was circumvented by using an identical light chain for each arm of the BsIgG. Antibodies with identical light chains that bind to different antigens were identified from an scFv phage library with a very restricted light chain repertoire for the majority (50/55) of antigen pairs tested. A BsIgG capable of simultaneously binding to the human receptors HER3 and cMpI was prepared by coexpressing the common light chain and corresponding remodeled heavy chains followed by protein A chromatography. The engineered heavy chains retain their ability to support antibody-dependent cell-mediated cytotoxicity as demonstrated with an anti-HER2 antibody.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Inmunoglobulina G/biosíntesis , Proteínas de Neoplasias , Receptores Inmunológicos/metabolismo , Complejo Antígeno-Anticuerpo , Citotoxicidad Inmunológica , Dimerización , Disulfuros , Receptores ErbB/metabolismo , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Modelos Moleculares , Mutación/genética , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-3 , Receptores de Citocinas/metabolismo , Receptores de Trombopoyetina
18.
J Biol Chem ; 273(10): 5829-40, 1998 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-9488719

RESUMEN

Neurotrophic factors are important for survival and maintenance of neurons during developmental and adult stages of the vertebrate nervous system. The neurotrophins mediate their signal into the cell by specific interaction with tyrosine kinase receptors of the Trk family. The extracellular immunoglobulin-like domain of the Trk receptors adjacent to the membrane has previously been shown to be the dominant element for specific neurotrophin binding. Using computer graphics models of the human TrkA and TrkC immunoglobulin-like domains as a guide, the residues involved in binding to their respective neurotrophins were mapped by mutational analysis. TrkC primarily utilizes loop EF, between beta-strands E and F, for binding. In contrast, TrkA utilizes the EF loop as well as additional residues, the latter being prime candidates for determining the specificity of TrkA versus TrkC. When selected TrkC and TrkA mutants with reduced binding were expressed on NIH3T3 cells, neurotrophin-induced autophosphorylation was strongly reduced or absent.


Asunto(s)
Amina Oxidasa (conteniendo Cobre) , Factores de Crecimiento Nervioso/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Tirosina Quinasas Receptoras/química , Receptores de Factor de Crecimiento Nervioso/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Sitios de Unión/fisiología , Moléculas de Adhesión Celular/química , Línea Celular , Mapeo Epitopo , Expresión Génica/genética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida/genética , Neurotrofina 3 , Fosforilación , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor trkA , Receptor trkC , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Alineación de Secuencia
19.
J Biol Chem ; 273(6): 3216-22, 1998 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-9452434

RESUMEN

Flt-1 is one of two receptor tyrosine kinases through which the angiogenic factor vascular endothelial growth factor (VEGF) functions. Placenta growth factor (PlGF) is an additional ligand for Flt-1. The second immunoglobulin-like domain in the extracellular domain of Flt-1 has previously been identified as the region containing the critical ligand-binding determinants. We analyzed the contribution of charged residues within the first three domains of Flt-1 to ligand binding by alanine-scanning mutagenesis. Domain 2 residues Arg159, Glu208 and His223-Arg224 (together) affect both VEGF and PlGF binding, while Glu137, Lys171, His223, and Arg224 affect PlGF but not VEGF. Several charged residues, especially Asp187, are important in maintaining the structural integrity of domain 2. In addition, some residues in domain 3 contribute to binding (Asp231) or provide for additional discrimination between ligands (Arg280-Asp283).


Asunto(s)
Factores de Crecimiento Endotelial/química , Inmunoglobulinas/química , Linfocinas/química , Proteínas Proto-Oncogénicas/química , Proteínas Tirosina Quinasas Receptoras/química , Alanina/química , Alanina/metabolismo , Secuencia de Aminoácidos , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Humanos , Inmunoglobulinas/metabolismo , Linfocinas/genética , Linfocinas/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Homología de Secuencia de Aminoácido , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Cancer Res ; 57(20): 4593-9, 1997 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9377574

RESUMEN

Vascular endothelial growth factor (VEGF) is a major mediator of angiogenesis associated with tumors and other pathological conditions, including proliferative diabetic retinopathy and age-related macular degeneration. The murine anti-human VEGF monoclonal antibody (muMAb VEGF) A.4.6.1 has been shown to potently suppress angiogenesis and growth in a variety of human tumor cells lines transplanted in nude mice and also to inhibit neovascularization in a primate model of ischemic retinal disease. In this report, we describe the humanization of muMAb VEGF A.4.6.1. by site-directed mutagenesis of a human framework. Not only the residues involved in the six complementarity-determining regions but also several framework residues were changed from human to murine. Humanized anti-VEGF F(ab) and IgG1 variants bind VEGF with affinity very similar to that of the original murine antibody. Furthermore, recombinant humanized MAb VEGF inhibits VEGF-induced proliferation of endothelial cells in vitro and tumor growth in vivo with potency and efficacy very similar to those of muMAb VEGF A.4.6.1. Therefore, recombinant humanized MAb VEGF is suitable to test the hypothesis that inhibition of VEGF-induced angiogenesis is a valid strategy for the treatment of solid tumors and other disorders in humans.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/uso terapéutico , Factores de Crecimiento Endotelial/inmunología , Endotelio Vascular/fisiología , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Linfocinas/inmunología , Neovascularización Patológica/prevención & control , Rabdomiosarcoma/terapia , Corteza Suprarrenal/irrigación sanguínea , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Capilares , Bovinos , Células Cultivadas , Clonación Molecular , Secuencia de Consenso , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/química , Inmunoglobulina G/uso terapéutico , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/química , Región Variable de Inmunoglobulina/química , Ratones , Ratones Desnudos , Modelos Estructurales , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/uso terapéutico , Rabdomiosarcoma/irrigación sanguínea , Alineación de Secuencia , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...