Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 70(12): 3451-3460, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33880648

RESUMEN

Cancer immunotherapies have generated remarkable clinical responses for some patients with advanced/metastatic disease, prompting exploration of rational combination therapies to bolster anti-tumor immunity in patients with limited response or those who experience tumor progression following an initial response to immunotherapy. In contrast to other tumor indications, objective response rates to single-agent PD-1/PD-L1 blockade in ovarian cancer are limited, suggesting a need to identify combinatorial approaches that lead to tumor regression in a setting where checkpoint blockade alone is ineffective. Using a pre-clinical model of aggressive intraperitoneal ovarian cancer, we have previously reported on a heterologous prime/boost cancer vaccine that elicits robust anti-tumor immunity, prolongs survival of tumor-bearing mice, and which is further improved when combined with checkpoint blockade. As tumor control in this model is CD8 + T cell dependent, we reasoned that the prime/boost vaccine platform could be used to explore additional treatment combinations intended to bolster the effects of CD8 + T cells. Using whole tumor transcriptomic data, we identified candidate therapeutic targets anticipated to rationally combine with prime/boost vaccination. In the context of a highly effective cancer vaccine, CD27 agonism or antibody-mediated depletion of granulocytic cells each modestly increased tumor control following vaccination, with anti-PD-1 therapy further improving treatment efficacy. These findings support the use of immunotherapies with well-defined mechanisms(s) of action as a valuable platform for identifying candidate combination approaches for further therapeutic testing in ovarian cancer.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Terapia Combinada/métodos , Femenino , Inmunoterapia/métodos , Ratones , Receptor de Muerte Celular Programada 1/inmunología
2.
Bone Marrow Transplant ; 50(7): 924-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25915810

RESUMEN

Fms-like tyrosine kinase-3 ligand (Flt3L) uniquely binds the Flt3 (CD135) receptor expressed on hematopoietic stem cells (HSCs), early progenitor cells, immature thymocytes and steady-state dendritic cells (DCs) and induces their proliferation, differentiation, development and mobilization in the bone marrow, peripheral blood and lymphoid organs. CDX-301 has an identical amino-acid sequence and comparable biological activity to the previously tested rhuFlt3L, which ceased clinical development over a decade ago. This Phase 1 trial assessed the safety, pharmacokinetic, pharmacodynamic and immunologic profile of CDX-301, explored alternate dosing regimens and examined the impact of rhuFlt3L on key immune cell subsets. Thirty healthy volunteers received CDX-301 (1-75 µg/kg/day) over 5-10 days. One event of Grade 3 community-acquired pneumonia occurred. There were no other infections, dose-limiting toxicities or serious adverse events. CDX-301 resulted in effective peripheral expansion of monocytes, hematopoietic stem and progenitor cells and key subsets of myeloid DCs and plasmacytoid DCs, with no clear effect on regulatory T cells. These data from healthy volunteers support the potential for CDX-301, as monotherapy or in combination with other agents, in various indications including allogeneic HSC transplantation and immunotherapy, but the effects of CDX-301 will need to be investigated in each of these patient populations.


Asunto(s)
Células Dendríticas/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Acondicionamiento Pretrasplante/métodos , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Adolescente , Adulto , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
J Intern Med ; 271(2): 183-92, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22126373

RESUMEN

Current vaccines primarily work by inducing protective antibodies. However, in many infections like HIV, malaria and tuberculosis as well as cancers, there remains a need for durable and protective T-cell immunity. Here, we summarize our efforts to develop a safe T-cell-based protein vaccine that exploits the pivotal role of dendritic cells (DC) in initiating adaptive immunity. Focusing on HIV, gag-p24 protein antigen is introduced into a monoclonal antibody (mAb) that efficiently and specifically targets the DEC-205 antigen uptake receptor on DC. When administered together with synthetic double-stranded RNA, polyriboinosinic:polyribocytidylic acid (poly IC) or its analogue poly IC stabilized with carboxymethylcellulose and poly-L-lysine (poly ICLC), as adjuvant, HIV gag-p24 within anti-DEC-205 mAb is highly immunogenic in mice, rhesus macaques, and in ongoing research, healthy human volunteers. Human subjects form both T- and B-cell responses to DC-targeted protein. Thus, DC-targeted protein vaccines are a potential new vaccine platform, either alone or in combination with highly attenuated viral vectors, to induce integrated immune responses against microbial or cancer antigens, with improved ease of manufacturing and clinical use.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Celular/inmunología , Linfocitos T/inmunología , Vacunas/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Carboximetilcelulosa de Sodio/análogos & derivados , Carboximetilcelulosa de Sodio/farmacología , Productos del Gen gag/inmunología , Humanos , Inductores de Interferón/farmacología , Lectinas Tipo C/inmunología , Ratones , Antígenos de Histocompatibilidad Menor , Poli I-C/farmacología , Polilisina/análogos & derivados , Polilisina/farmacología , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología
4.
Oncogene ; 26(25): 3758-67, 2007 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-17530028

RESUMEN

The specificity and high affinity binding of antibodies provides these molecules with ideal properties for delivering a payload to target cells. This concept has been commercialized for cancer therapies using toxin- or radionucleotide-conjugated antibodies that are designed to selectively deliver cytotoxic molecules to cancer cells. Exploiting the same effective characteristics of antibodies, antibody-targeted vaccines (ATV) are designed to deliver disease-specific antigens to professional antigen-presenting cells (APCs), thus enabling the host's immune system to recognize and eliminate malignant or infected cells through adaptive immunity. The concept of ATVs has been in development for many years, and recently has entered clinical trials. Early studies with ATVs focused on the ability to induce humoral immunity in the absence of adjuvants. More recently, ATVs targeted to C-type lectin receptors have been exploited for induction of potent helper and cytolytic T-cell responses. To maximize their stimulatory capacity, the ATVs are being evaluated with a variety of adjuvants or other immunostimulatory agents. In the absence of co-administered immunostimulatory signals, APC-targeting can induce antigen-specific tolerance and, thus, may also be exploited in developing specific treatments for autoimmune and allergic diseases, or for preventing transplant rejection. The successful clinical application of this new class of antibody-based products will clearly depend on using appropriate combinations with other strategies that influence the immune system.


Asunto(s)
Anticuerpos/inmunología , Vacunas contra el Cáncer/inmunología , Animales , Antígenos/inmunología , Autoinmunidad/inmunología , Humanos , Hipersensibilidad/inmunología , Lectinas Tipo C/inmunología
5.
Eur J Haematol ; 70(2): 67-74, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12581186

RESUMEN

The study was undertaken to delineate mechanisms of platelet destruction by phagocytosis during fetal/neonatal alloimmune thrombocytopenia (FAIT/NAIT) because of maternal antibodies against human platelet antigen 1a (HPA-1a). By employing a platelet phagocytosis assay based on the ORPEGEN flow cytometric bacterial phagocytosis test, we measured monocyte ingestion of platelets mediated by anti-HPA-1a antibodies. Moreover, we tested, as potential therapeutic agents, FcgammaR reactive reagents, for their inhibition of this process. Four of six anti-HPA-1a sera tested mediated phagocytosis of HPA-1a-positive platelets in a concentration-dependent manner. Monocyte ingestion of platelets was almost completely inhibited by cytochalasin D. No anti-HPA-1a-mediated phagocytosis was observed with anti-HPA-1a-negative platelets. The humanised anti-FcgammaRI monoclonal antibody H22 at concentrations 1-100 microg/ml, completely inhibited anti-HPA-1a-mediated phagocytosis as did similar concentrations of ivIg. By contrast, a mouse monoclonal anti-FcgammaRII (IV.3, Fab) at 10 microg/ml caused little or no suppression of platelet phagocytosis mediated by two anti-HPA-1 sera. Furthermore, the addition of anti-FcgammaRII (10 microg/ml) to sub-optimal concentrations of H22 did not significantly increase the inhibitory effect of the latter compound. Monomeric IgG (0.1-10 microg/ml) failed to suppress anti-HPA-1 mediated platelet ingestion by the phagocytes, as did anti-FcgammaRIII. To our knowledge this is a rare example of an assay that measures platelet phagocytosis in vitro. The results suggest that FcgammaRI plays a major role in anti-HPA-1a-mediated platelet phagocytosis by monocytes while FcgammaRIIa, is of little or minor importance only. Moreover, the findings indicate the use of H22 as an alternative to interavenous Ig (ivIg) in the management of FAIT/NAIT.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos de Plaqueta Humana/inmunología , Plaquetas/inmunología , Monocitos/inmunología , Fagocitosis/efectos de los fármacos , Receptores de IgG/inmunología , Adulto , Pruebas Inmunológicas de Citotoxicidad , Femenino , Citometría de Flujo , Humanos , Indicadores y Reactivos/farmacología , Recién Nacido , Enfermedades del Recién Nacido/inmunología , Integrina beta3 , Intercambio Materno-Fetal/inmunología , Embarazo , Trombocitopenia/inmunología
6.
J Immunol ; 167(11): 6303-11, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11714794

RESUMEN

The high-affinity IgG receptor, FcgammaRI (CD64), is constitutively expressed exclusively on professional APCs. Human FcgammaRI binds monomeric IgG with high affinity and is, therefore, saturated in vivo. The binding of IgG to FcgammaRI causes receptor recycling, while Abs that cross-link FcgammaRI cause rapid down-modulation of surface FcgammaRI. Because studies performed in the absence of ligand may not be representative of FcgammaRI modulation in vivo, we investigated the ability of FcgammaRI-cross-linking Abs and non-cross-linking derivatives to modulate FcgammaRI in the presence and absence of ligand. In the absence of ligand mAb H22 and wH22xeGFP, an enhanced green fluorescent protein (eGFP)-labeled fusion protein of H22, cross-linked and rapidly down-modulated surface FcgammaRI on the human myeloid cell line, U937, and its high FcgammaRI-expressing subclone, 10.6. This effect was dependent on the concentration of fusion protein and the level of FcgammaRI expression and correlated with internalization of both wH22xeGFP and FcgammaRI, itself, as assessed by confocal microscopy. A single-chain Fv version, sFv22xeGFP, which does not cross-link FcgammaRI, was unable to modulate FcgammaRI in the absence of IgG. However, if ligand was present, treatment with either monovalent or cross-linking fusion protein led to intracellular receptor accumulation. These findings suggest at least two alternate mechanisms of internalization that are influenced by ligand and demonstrate the physiologic potential of FcgammaRI to transport a large antigenic load into APCs for processing. These studies may lead to the development of better FcgammaRI-targeted vaccines, as well as therapies to down-modulate FcR involved in autoimmune diseases.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Inmunoglobulina G/farmacología , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Adyuvantes Inmunológicos/biosíntesis , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Sitios de Unión de Anticuerpos/genética , Células Clonales , Relación Dosis-Respuesta Inmunológica , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Vectores Genéticos/metabolismo , Vectores Genéticos/farmacología , Proteínas Fluorescentes Verdes , Humanos , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Ligandos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Modelos Inmunológicos , Receptores de IgG/biosíntesis , Receptores de IgG/inmunología , Proteínas Recombinantes de Fusión/farmacología , Células U937
7.
J Immunol Methods ; 248(1-2): 113-23, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11223073

RESUMEN

CD30 is a molecule that is overexpressed on the surface of Hodgkin's lymphoma cells. Therefore, CD30 represents a potential candidate for immunotherapy. In this study, we report the in vitro results of two bispecific molecules (BSMs) that target CD30 to trigger molecules expressed on myeloid effector cells. The first BSM is composed of the Fab' fragment of a CD30-specific antibody, Ki-4, chemically linked to the Fab' fragment of the humanized CD64 (FcgammaRI)-specific antibody, H22 (H22xKi-4). In the second BSM, the H22 Fab' is replaced with the Fab' fragment of the CD89 (FcalphaR)-specific, antibody, A77 (A77xKi-4). Both BSMs were able to bind specifically to lymphoma cell lines expressing CD30. In addition, the H22xKi-4 and A77xKi-4 BSMs were shown to bind cells expressing CD64 and CD89, respectively. Both BSMs mediated potent, dose-dependent antibody dependent cell-mediated cytotoxicity (ADCC) of CD30-expressing tumor cell lines when human monocytes were used as effector cells. In addition, freshly prepared polymorphonuclear leukocytes (PMNs) and effector cells in whole blood were able to mediate the ADCC of targets in conjunction with the A77xKi-4 BSM in some, but not all, experiments. Furthermore, we examined the ability of monocyte-derived macrophages (MDMs) to phagocytose CD30-expressing tumor cell lines in conjunction with the BSM. MDM-mediated phagocytosis was significantly enhanced in the presence of both BSMs. These results demonstrate that targeting lymphoma cells via CD30 to the myeloid high affinity Fc receptor for IgG and to the Fc receptor for IgA results in potent in vitro anti-tumor activity.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD/inmunología , Enfermedad de Hodgkin/terapia , Antígeno Ki-1/inmunología , Receptores Fc/inmunología , Receptores de IgG/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Humanos , Ratones , Fagocitosis , Células Tumorales Cultivadas
8.
J Immunol Methods ; 248(1-2): 167-82, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11223077

RESUMEN

Studies from our laboratory and others have established that both mononuclear phagocytes and neutrophils mediate very efficient cytotoxicity when targeted through Fc receptors using a suitable monoclonal or bispecific antibody (BsAb). Cross-linking an Fc receptor for IgG (FcgammaR) triggers multiple anti-tumor activities including superoxide generation, cytokine and enzyme release, phagocytosis and antibody-dependent cellular cytotoxicity (ADCC). In this report, using unfractionated leukocytes and two color flow cytometric analysis, we describe the phagocytic capacity of peripheral blood polymorphonuclear cells (PMN) and monocytes isolated from patients enrolled in a phase I clinical trial of MDX-H210 given in combination with IFNgamma. MDX-H210 is a BsAb targeting the myeloid trigger molecule FcgammaRI and the HER-2/neu proto-oncogene product overexpressed on a variety of adenocarcinomas. In this trial, cohorts of patients received escalating doses of MDX-H210 3 times per week for 3 weeks. Interferon-gamma (IFNgamma) was given 24 h prior to each BsAb infusion. Our results demonstrate that monocytes from these patients were inherently capable of phagocytosing the HER-2/neu positive SK-BR-3 cell line and that addition of MDX-H210 into the assay significantly enhanced the number of targets phagocytosed. Two days after administration of an immunologically active dose of MDX-H210 (10 mg/m2), monocytes from these patients were able to phagocytose greater amounts of target cell material, indicating that these cells remained armed with functionally sufficient BsAb for at least 48 h. PMN from these patients very efficiently mediated phagocytosis through FcgammaRI after being treated with IFNgamma, but not before. We conclude that phagocytosis is not only an efficient mechanism of myeloid cell-mediated cytotoxicity, but may also be a mechanism by which antigens from phagocytosed cells can enter a professional antigen presenting cell for processing and presentation.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Monocitos/inmunología , Neoplasias/terapia , Neutrófilos/inmunología , Fagocitosis , Receptor ErbB-2/análisis , Animales , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Citometría de Flujo , Humanos , Interferón gamma/farmacología , Ratones , Neoplasias/inmunología , Proto-Oncogenes Mas , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Células Tumorales Cultivadas
9.
J Immunol ; 165(12): 6738-42, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11120792

RESUMEN

Previous studies have documented that targeting foreign Ags to IgG FcgammaR leads to enhanced Ag-specific responses in vitro and in vivo. However, the ability to overcome immunologic nonresponsiveness by targeting poorly immunogenic Ags to FcgammaR has not been investigated. To address this question in a simple model, we immunized transgenic mice expressing human CD64 (FcgammaRI) and their nontransgenic littermates with Fab' derived from the murine anti-human CD64 mAb m22. The m22 Fab' served as both the targeting molecule and the Ag. We found that only CD64-expressing mice developed anti-Id titers to m22. Furthermore, chemically linked multimers of m22 Fab', which mediated efficient internalization of the human CD64, were significantly more potent than monomeric m22 F(ab')(2) at inducing anti-Id responses. In all cases, the humoral responses were specific for m22 Id and did not react with other murine IgG1 Fab' fragments. Chemical addition of a second murine Fab' (520C9 anti-human HER2/neu) to m22 Fab' multimers demonstrated that IgG1 and IgG2a anti-Id titers could be generated to 520C9 only in the CD64-expressing mice. These results show that targeting to CD64 can overcome immunological nonresponsiveness to a weak immunogen. Therefore, targeting to CD64 may be an effective method to enhance the activity of nonimmunogenic tumor vaccines.


Asunto(s)
Anticuerpos Antiidiotipos/biosíntesis , Antígenos/inmunología , Ratones Transgénicos/inmunología , Receptor ErbB-2/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Animales , Anticuerpos Antiidiotipos/metabolismo , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/genética , Antígenos/administración & dosificación , Sitios de Unión de Anticuerpos/genética , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Isotipos de Inmunoglobulinas/biosíntesis , Ratones , Ratones Endogámicos , Modelos Inmunológicos , Receptor ErbB-2/administración & dosificación , Receptores de IgG/metabolismo , Factores de Tiempo
10.
J Immunol ; 164(11): 5746-52, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10820252

RESUMEN

Macrophages represent an important effector cell for Ab-mediated tumor therapy. Previous studies have documented that cytokines can influence Fc receptor (FcR) expression and function. In this study we examined the tumoricidal activities of the type I receptors for IgG (Fc gamma RI) and the IgA FcR (Fc alpha RI) on monocyte-derived macrophages (MDM) cultured in the presence of IFN-gamma, M-CSF, or GM-CSF. Bispecific Abs were used to target a Her2/neu breast carcinoma cell line, SKBR-3, to Fc alpha RI or Fc gamma RI on MDM. Although Fc alpha RI and Fc gamma RI share a common signaling pathway contingent on association with the gamma-chain (FcR gamma subunit), a marked difference in their efficiency in mediating tumoricidal functions was seen in response to specific cytokines. M-CSF- and GM-CSF-treated MDM mediated efficient phagocytosis of SKBR-3 cells by Fc alpha RI and Fc gamma RI; however, IFN-gamma-treated MDM phagocytosed tumor cells only with the Fc gamma RI-directed bispecific Abs. Similarly, IFN-gamma-cultured MDM lysed tumor cells more efficiently via Fc gamma RI then by Fc alpha RI as measured in Ab-dependent cellular cytotoxicity assays. Conversely, GM-CSF-treated MDM mediated more efficient lysis of tumor cells via Fc alpha RI than Fc gamma RI, while M-CSF-cultured MDM were relatively less efficient in mediating Ab-dependent cellular cytotoxicity through either receptor. With the exception of IFN-gamma-mediated enhancement of Fc gamma RI expression and Fc gamma RI gamma-chain complexes, the regulation of Fc gamma RI- or Fc alpha RI-mediated activity occurred without significant change in either receptor expression or total complexes with gamma subunit. These data suggest that the efficiency of Ab-mediated tumor therapy, which depends on FcR effector cell functions, may be modified by the use of specific cytokines.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD/fisiología , Citocinas/farmacología , Inmunoglobulina A/metabolismo , Macrófagos/inmunología , Monocitos/inmunología , Receptores Fc/fisiología , Receptores de IgG/fisiología , Adyuvantes Inmunológicos/farmacología , Adulto , Anticuerpos Biespecíficos/farmacología , Diferenciación Celular/inmunología , Supervivencia Celular/inmunología , Pruebas Inmunológicas de Citotoxicidad/métodos , Citometría de Flujo , Humanos , Macrófagos/citología , Monocitos/citología , Fagocitosis/inmunología , Células Tumorales Cultivadas/inmunología
11.
Eur J Haematol ; 65(6): 399-406, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11168497

RESUMEN

To gain insight into mechanisms of platelet destruction and its possible inhibition during fetal/neonatal alloimmune thrombocytopenia (FAIT/NAIT) the binding to monocytes (Mo) of anti-HPA-1a-sensitised platelets, the initial step in IgG-mediated destruction by effector cells, was evaluated in an in vitro assay. Neonatal Mo were compared with adult Mo as effectors in the assay. Moreover, the potential involvement of Fcgamma receptor (FcgammaR) classes during platelet destruction in the disease was tested by using FcgammaR class-specific reagents as inhibitors of the binding reaction. Neonatal Mo were 37% less active than adult Mo in their interaction with anti-HPA-1a-sensitised platelets (p < 0.05). The FcgammaRI-specific reagents human monomeric IgG and humanised anti-FcgammaRI monoclonal H22 caused virtually complete inhibition of platelet binding to Mo. When compared to an intravenous immunoglobulin preparation the inhibitory activity of H22 was 10-100 x greater than that of the latter compound. Monoclonal anti-FcgammaRII IV.3 and anti-FcgammaRIII 3G8 decreased platelet binding by 70% and 64%, respectively, but only the anti-FcgammaRII inhibition was statistically significant (p < 0.001). Finally, anti-HPA-1a-sensitised platelets bound to 131H- but not to 131R- FcgammaRIIa transfected 3T6 mouse fibroblasts (p < 0.01), in an anti-HPA-1a-concentration-dependent manner. The results suggest that FcgammaRI and FcgammaRIIa may be involved in anti-HPA-1a-mediated platelet destruction by mononuclear phagocytes during FAIT/NAIT. Moreover, the much greater potency ofmonoclonal H22 than of intravenous immunoglobulin as an inhibitor of anti-HPA-1a-mediated Mo-platelet interaction, might render it superior to the latter agent in the maternal therapy of the disorder.


Asunto(s)
Antígenos de Plaqueta Humana/farmacología , Plaquetas/efectos de los fármacos , Monocitos/citología , Receptores de IgG/metabolismo , Adulto , Animales , Anticuerpos Monoclonales/farmacología , Antígenos de Plaqueta Humana/sangre , Antígenos de Plaqueta Humana/inmunología , Plaquetas/citología , Comunicación Celular/inmunología , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Inmunoglobulinas Intravenosas/farmacología , Recién Nacido , Integrina beta3 , Isoanticuerpos/sangre , Isoanticuerpos/inmunología , Ratones , Monocitos/metabolismo , Madres , Receptores de IgG/inmunología , Trombocitopenia/etiología , Trombocitopenia/inmunología , Transfección
12.
Blood ; 94(2): 808-17, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10397749

RESUMEN

Within multi-subunit Ig receptors, the FcR gamma-chain immunoreceptor tyrosine-based activation motif (ITAM) plays a crucial role in enabling antigen presentation. This process involves antigen-capture and targeting to specific degradation and major histocompatibility complex (MHC) class II loading compartments. Antigenic epitopes are then presented by MHC class II molecules to specific T cells. The high-affinity receptor for IgG, hFcgammaRIa, is exclusively expressed on myeloid lineage cells and depends on the FcR gamma-chain for surface expression, efficient ligand binding, and most phagocytic effector functions. However, we show in this report, using the IIA1.6 cell model, that hFcgammaRIa can potentiate MHC class II antigen presentation, independently of a functional FcR gamma-chain ITAM. Immunoelectron microscopic analyses documented hFcgammaRIa alpha-chain/rabbit IgG-Ovalbumin complexes to be internalized and to migrate via sorting endosomes to MHC class II-containing late endosomes. Radical deletion of the hFcgammaRIa alpha-chain cytoplasmic tail did not affect internalization of rabbit IgG-Ovalbumin complexes. Importantly, however, this resulted in diversion of receptor-ligand complexes to the recycling pathway and decreased antigen presentation. These results show the hFcgammaRIa cytoplasmic tail to contain autonomous targeting information for intracellular trafficking of receptor-antigen complexes, although deficient in canonical tyrosine- or dileucine-targeting motifs. This is the first documentation of autonomous targeting by a member of the multichain FcR family that may critically impact the immunoregulatory role proposed for hFcgammaRIa (CD64).


Asunto(s)
Presentación de Antígeno , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunoglobulina G/inmunología , Receptores de IgG/inmunología , Secuencia de Aminoácidos , Animales , Línea Celular , Endocitosis , Peroxidasa de Rábano Silvestre , Humanos , Ratones , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Ovalbúmina/inmunología , Conejos , Receptores de IgG/química , Receptores de IgG/genética , Proteínas Recombinantes de Fusión/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Fracciones Subcelulares/química , Transfección
13.
Breast Cancer Res Treat ; 53(3): 199-207, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10369066

RESUMEN

BACKGROUND: MDX-210 is a bispecific antibody (BsAb) with specificity for both the proto-oncogene product of HER-2/neu (c-erbB-2) and FcgammaRI (CD64). HER-2/neu is overexpressed in malignant tissue of approximately 30% of patients with breast cancer, and FcgammaRI is expressed on human monocytes, macrophages, and IFN-gamma activated granulocytes. We investigated phagocytosis and cytolysis of cultured human breast cancer cells by human monocyte-derived macrophages (MDM) mediated by BsAb MDX-210, its partially humanized derivative (MDX-H210), and its parent MoAb 520C9 (anti-HER-2/neu) under various conditions. MATERIALS AND METHODS: Purified monocytes were cultured with GM-CSF, M-CSF, or no cytokine for five or six days. Antibody dependent cellular phagocytosis (ADCP) and cytolysis (ADCC) assays were performed with the MDM and HER-2/neu positive target cells (SK-BR-3). ADCP was measured by two-color fluorescence flow cytometry using PKH2 (green fluorescent dye) and phycoerythrin-conjugated (red) monoclonal antibodies (MoAb) against human CD14 and CD11b. ADCC was measured with a non-radioactive LDH detection kit. RESULTS: Both BsAb MDX-210 (via FcgammaRI) and MoAb 520C9 (mouse IgG1, via FcgammaRII) mediated similar levels of ADCP and ADCC. ADCP mediated by BsAb MDX-H210 was identical to that mediated by BsAb MDX-210. Confocal microscopy demonstrated that dual-labeled cells represented true phagocytosis. Both ADCP and ADCC were higher when MDM were pre-incubated with GM-CSF than when incubated with M-CSF. CONCLUSIONS: BsAb MDX-210 is as active in vitro as the parent MoAb 520C9 in inducing both phagocytosis and cytolysis of MDM. MDX-210 and its partially humanized derivative, MDX-H210, mediated similar levels of ADCP. GM-CSF appears to superior to M-CSF in inducing MDM-mediated ADCC and ADCP. These studies support the ongoing clinical investigations of BsAb MDX-210 and its partially humanized derivative.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Neoplasias de la Mama/inmunología , Carcinoma/inmunología , Fagocitosis/efectos de los fármacos , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma/metabolismo , Carcinoma/patología , Citocinas/farmacología , Pruebas Inmunológicas de Citotoxicidad , Relación Dosis-Respuesta a Droga , Epítopos , Femenino , Citometría de Flujo , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Microscopía Confocal , Fagocitosis/inmunología , Proto-Oncogenes Mas , Células Tumorales Cultivadas , Células U937
14.
Hum Antibodies ; 9(1): 47-54, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10331185

RESUMEN

A trispecific F(ab')3 antibody conjugate (TAC) with specificities for the Fc gamma receptor I (Fc gamma RI/CD64), the epidermal growth factor receptor (EGFR) and the HER2/neu antigen has been developed to redirect effector cell-mediated cytotoxicity against cancer cells expressing both or either of the tumor-associated antigens. The TAC was constructed in two steps using the sulfhydryl-specific cross-linker o-phenylenedimaleimide (o-PDM). In step one, a bispecific antibody was prepared by linking the Fab' fragments of mAb m22 (a murine IgG1 specific for Fc gamma RI) and mAb H425 (a humanized IgG1 antibody recognizing EGFR). The conjugation efficiency was about 60%. In the second step, the Fab' fragment of mAb 520C9, a murine IgG1 specific for HER2/neu, was coupled to the bispecific antibody made in step one. About 40% of the bispecific conjugate was derivatized to form the trispecific antibody. The purity of the TAC was more than 90% after gel filtration purification. The TAC was characterized in vitro for its ability to bind specifically to all the three antigens and to kill target cells expressing the tumor antigens. In contrast to bispecific conjugates that can only target cells expressing either of the tumor antigens, the TAC was able to bind both the antigens more efficiently in cell-binding assays and to kill tumor cells expressing EGFR and HER2/neu antigens.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Monoclonales/biosíntesis , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos de Neoplasias/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/inmunología , Receptores ErbB/inmunología , Humanos , Hibridomas/inmunología , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Ratones , Receptor ErbB-2/inmunología , Células Tumorales Cultivadas
15.
Clin Cancer Res ; 4(9): 2237-43, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9748144

RESUMEN

A bispecific antibody was made by chemical conjugation of Fab' fragments from humanized antibodies specific for tumor-associated glycoprotein-72 (TAG-72) and high-affinity immunoglobulin receptor, FcgammaA receptor type I (FcgammaRI). The purified anti-TAG-72 x anti-FcgammaRI (HCC49xH22) bispecific antibody had an approximate Mr of 111,000, consistent with a F(ab')2, and bound specifically to KLEB and LS174T tumor cell lines, which express the TAG-72 tumor antigen. Furthermore, HCC49x H22 was shown to simultaneously bind to KLEB cells and a soluble FcgammaRI fusion protein, demonstrating the bifunctional nature of the molecule. Using IFN-gamma-treated monocytes as effector cells, concentrations of the bispecific antibody in the range of 1-10,000 ng/ml mediated specific lysis of TAG-72-positive tumor cells. In contrast, the bispecific antibody did not promote antibody-dependent cellular cytotoxicity of a cell line that was negative for TAG-72 antigen. Importantly, the antibody-dependent cellular cytotoxicity activity of the bispecific antibody was significantly greater than that of the monoclonal antibody HCC49. These in vitro data indicate that the humanized bispecific antibody HCC49xH22 has the appropriate specificity and functional activity for further evaluation as potential immunotherapy for TAG-72-positive malignancies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos de Neoplasias/inmunología , Glicoproteínas/inmunología , Neoplasias/metabolismo , Receptores de IgG/inmunología , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/metabolismo , Citometría de Flujo , Glicoproteínas/metabolismo , Humanos , Ratones , Ratones SCID , Neoplasias/inmunología , Células Tumorales Cultivadas
16.
J Immunol ; 160(4): 1677-86, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9469424

RESUMEN

The FcR for IgA (Fc alpha RI, CD89) is primarily expressed on cytotoxic immune effector cells. By chemically cross-linking F(ab') fragments of the FcR for IgA (Fc alpha RI)-specific mAb (A77) with tumor Ag-specific mAb (anti-HER2/neu and anti-epidermal growth factor receptor), we have developed bispecific molecules (BSM) that simultaneously bind to respective tumor Ags and Fc alpha RI-expressing effector cells in whole blood. These BSM mediated up to 55% of specific lysis of appropriate tumor Ag-expressing target cells (from a variety of tumors) with purified polymorphonuclear leukocytes, monocytes, or whole blood effector cells without preactivation with exogenous cytokines. To our knowledge, this is the first demonstration of Ab-dependent cell-mediated cytotoxic activity via Fc alpha RI in whole blood. Also, monocyte-derived macrophages mediated phagocytosis of HER2/neu-expressing tumor cells (>95% tumor cell loss). These BSM-mediated cytotoxic activities were completely inhibited by F(ab')2 of A77, demonstrating the specific role of Fc alpha RI as a trigger molecule. Furthermore, the binding of these BSM to monocytes or polymorphonuclear leukocytes in whole blood did not induce modulation of Fc alpha RI in the absence of the target Ag. Therefore, immune effector cells may be "armed" with Fc alpha RI-directed BSM in whole blood. These Fc alpha RI-directed BSM may offer new treatment options for various malignancies and other disease conditions.


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Inmunoglobulina A/metabolismo , Receptores Fc/metabolismo , Animales , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/farmacología , Antígenos CD/sangre , Antígenos CD/inmunología , Antígenos de Neoplasias/sangre , Sitios de Unión de Anticuerpos , Humanos , Inmunidad Celular , Inmunoglobulina A/sangre , Ratones , Monocitos/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Plasmacitoma , Unión Proteica/inmunología , Receptores Fc/sangre , Receptores Fc/inmunología , Células Tumorales Cultivadas
17.
Cancer Res ; 57(18): 4008-14, 1997 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9307286

RESUMEN

A bispecific antibody, MDX-H210, was developed to target cytotoxic effector cells expressing Fc gamma receptor type I (Fc gammaRI, CD64) to HER2/neu-overexpressing tumor cells. HER2/neu is an appropriate target for immunotherapy due to the high level of expression of this proto-oncogene in a variety of malignancies. The expression of Fc gammaRI is limited primarily to cytotoxic immune cells, including monocytes, macrophages, and cytokine-activated polymorphonuclear (PMN) cells. Therefore, tumor cells bound with MDX-H210 can be selectively recognized by effector cells with cytotoxic potential. MDX-H210 was prepared by chemical conjugation of Fab' fragments derived from the HER2/neu-specific monoclonal antibody, 520C9, and the Fc gammaRI-specific monoclonal antibody, H22. This bispecific molecule demonstrated specific, dose-dependent, and saturable binding to both HER2/neu- and Fc gammaRI-expressing cells. A solid-phase immunoassay that demonstrated simultaneous and specific binding to both antigens was used to confirm the bispecific nature of MDX-H210. Monocytes and PMN cells mediated MDX-H210-dependent lysis of HER2/neu-overexpressing cell lines derived from breast, ovarian, and lung carcinomas. IFN-gamma treatment of monocytes enhanced antibody-dependent cellular cytotoxicity, whereas IFN-gamma and granulocyte colony-stimulating factor were required for PMN cell-mediated tumor cell lysis. In addition, MDX-H210 elicited tumor necrosis factor-alpha secretion from monocytes when cultured in the presence of HER2/neu-positive target cells. These in vitro data suggest that targeting tumor cells to Fc gammaRI with MDX-H210 may be an effective treatment for malignancies that overexpress HER2/neu. The in vivo cytotoxic potential of MDX-H210 may be enhanced by combination therapy with the cytokines granulocyte colony-stimulating factor and IFN-gamma, which up-regulate Fc gammaRI expression on cytotoxic effector cells.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Citotoxicidad Inmunológica , Humanos , Inmunoterapia/métodos , Proto-Oncogenes Mas , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Leukoc Biol ; 62(4): 469-79, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9335317

RESUMEN

About 10-15% of patients with immune thrombocytopenic purpura (ITP) cannot be controlled by corticosteroid therapy and splenectomy. For these patients treatment with high-dose IVIgG induces partial or complete responses. The clinical benefits of IVIgG could be due to blockade of Fc receptors for IgG (FcgammaR), because several model systems clearly show that functional FcgammaR are essential for establishment of ITP and related diseases. However, the specific contributions of the three individual classes of FcgammaR remain to be more completely defined. Recently monoclonal antibody (mAb) H22, which recognizes an epitope on FcgammaRI (CD64) outside the ligand binding domain, was humanized by grafting its complementarity determining regions onto human IgG1 constant domains. Because FcgammaRI has a high affinity for human IgG1 antibodies, we predicted mAb H22 would also bind to FcgammaRI through its Fc domain and block FcgammaRI-mediated phagocytosis. These studies demonstrate that mAb H22 blocked phagocytosis of opsonized red blood cells 1000 times more effectively than an irrelevant IgG. Moreover, cross-linking FcgammaRI with mAb H22 rapidly down-modulated FcgammaRI expression on monocytes without affecting other surface antigens. We conclude that because mAb H22 is a humanized mAb that blocks the FcgammaRI ligand binding domain and down-modulates FcgammaRI expression, it is a particularly good candidate for evaluating the role of FcgammaRI in patients with ITP.


Asunto(s)
Anticuerpos Monoclonales , Monocitos/inmunología , Fagocitosis , Receptores de IgG/fisiología , Animales , Antígenos CD/biosíntesis , Células Cultivadas , Epítopos/análisis , Eritrocitos/inmunología , Citometría de Flujo , Humanos , Regiones Constantes de Inmunoglobulina , Inmunoglobulina G , Cinética , Ratones , Modelos Inmunológicos , Receptores de IgG/biosíntesis , Receptores de IgG/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...