Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Allergy ; 73(2): 328-340, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28921585

RESUMEN

While desired for the cure of allergy, regulatory immune cell subsets and nonclassical Th2-biased inflammatory mediators in the tumour microenvironment can contribute to immune suppression and escape of tumours from immunological detection and clearance. A key aim in the cancer field is therefore to design interventions that can break immunological tolerance and halt cancer progression, whereas on the contrary allergen immunotherapy exactly aims to induce tolerance. In this position paper, we review insights on immune tolerance derived from allergy and from cancer inflammation, focusing on what is known about the roles of key immune cells and mediators. We propose that research in the field of AllergoOncology that aims to delineate these immunological mechanisms with juxtaposed clinical consequences in allergy and cancer may point to novel avenues for therapeutic interventions that stand to benefit both disciplines.


Asunto(s)
Hipersensibilidad/inmunología , Hipersensibilidad/terapia , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Desensibilización Inmunológica/métodos , Humanos
2.
Allergy ; 72(6): 866-887, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28032353

RESUMEN

Th2 immunity and allergic immune surveillance play critical roles in host responses to pathogens, parasites and allergens. Numerous studies have reported significant links between Th2 responses and cancer, including insights into the functions of IgE antibodies and associated effector cells in both antitumour immune surveillance and therapy. The interdisciplinary field of AllergoOncology was given Task Force status by the European Academy of Allergy and Clinical Immunology in 2014. Affiliated expert groups focus on the interface between allergic responses and cancer, applied to immune surveillance, immunomodulation and the functions of IgE-mediated immune responses against cancer, to derive novel insights into more effective treatments. Coincident with rapid expansion in clinical application of cancer immunotherapies, here we review the current state-of-the-art and future translational opportunities, as well as challenges in this relatively new field. Recent developments include improved understanding of Th2 antibodies, intratumoral innate allergy effector cells and mediators, IgE-mediated tumour antigen cross-presentation by dendritic cells, as well as immunotherapeutic strategies such as vaccines and recombinant antibodies, and finally, the management of allergy in daily clinical oncology. Shedding light on the crosstalk between allergic response and cancer is paving the way for new avenues of treatment.


Asunto(s)
Hipersensibilidad/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Anticuerpos , Humanos , Inmunoglobulina E/inmunología , Vigilancia Inmunológica , Inmunoterapia/tendencias , Neoplasias/terapia , Células Th2/inmunología
3.
Leukemia ; 24(9): 1555-65, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20574452

RESUMEN

The HOX genes encode a family of transcription factors that are dysregulated in several malignancies and have been implicated in oncogenesis and cancer cell survival. Disruption of HOX protein function using the peptide HXR9 has shown anti-tumor effects against melanoma, lung cancer and renal cancer. In this report, we evaluated the expression of all 39 HOX genes in a panel of six malignant B-cell lines, including multiple myeloma cells and found different levels of expression of HOX family members suggesting that they also have a role in malignant B-cell survival. We show that disrupting HOX function using the peptide HXR9 induces significant cytotoxicity in the entire panel of cell lines. Importantly, we found that the cytotoxic effects of HXR9 can be enhanced by combining it with ch128.1Av, an antibody-avidin fusion protein specific for the human transferrin receptor 1 (CD71). Iron starvation induced by the fusion protein contributes to the enhanced effect and involves, at least in part, the induction of a caspase-independent pathway. These results show the relevance of HOX proteins in malignant B-cell survival and suggest that our therapeutic strategy may be effective in the treatment of incurable B-cell malignancies such as multiple myeloma.


Asunto(s)
Muerte Celular/genética , Genes Homeobox , Hierro/metabolismo , Linfoma de Células B/patología , Línea Celular Tumoral , Proliferación Celular , Humanos , Linfoma de Células B/metabolismo , Reacción en Cadena de la Polimerasa
4.
Hum Gene Ther ; 20(5): 453-64, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19215191

RESUMEN

The highly aggressive cancer syndrome of female mice carrying a p53 knockout allele and a rat HER-2/neu (Neu) transgene (BALB-p53Neu) can be prevented by a cell vaccine presenting three components: Neu, interleukin (IL)-12 production, and allogeneic major histocompatibility complex (MHC) alleles (Triplex cell vaccine). Here we tested a second-generation Triplex DNA-based vaccine (Tri-DNA), consisting of the combination of three gene components (a transmembrane-extracellular domain fragment of the Neu gene, IL-12 genes, and the H-2D(q) allogeneic MHC gene), carried by separate plasmids. The Tri-DNA vaccine was at least as effective as the Triplex cell vaccine for cancer immunoprevention, giving a similar delay in the onset of mammary cancer and complete protection from salivary cancer. Both vaccines induced anti-Neu antibodies of the murine IgG2a isotype at similar levels. The Tri-DNA vaccine gave more restricted immunostimulation, consisting of a fully helper T cell type 1 (Th1)-polarized response, with effective production of interferon (IFN)-gamma in response to the vaccine but no spontaneous production, and no induction of anti-Neu IgG3 antibodies. On the other hand, the Triplex cell vaccine induced both Th1 and Th2 cytokines, a strong increase in spontaneous IFN-gamma production, and high levels of IgG3 antibodies recognizing Neu-positive syngeneic cells. In conclusion, the Tri-DNA vaccine is as effective as Triplex cell vaccine, exploiting a more restricted immune stimulation.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Interleucina-12/inmunología , Síndromes Neoplásicos Hereditarios/prevención & control , Receptor ErbB-2/genética , Proteína p53 Supresora de Tumor/genética , Vacunas de ADN/inmunología , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Citotoxicidad Inmunológica , Femenino , Terapia Genética , Inmunoglobulina G/sangre , Inmunoterapia , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/metabolismo , Complejo Mayor de Histocompatibilidad/inmunología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/patología , Ratones , Síndromes Neoplásicos Hereditarios/terapia , Ratas , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Glándulas Salivales/inmunología , Glándulas Salivales/patología , Transfección , Proteína p53 Supresora de Tumor/inmunología , Proteína p53 Supresora de Tumor/metabolismo
5.
Leukemia ; 23(1): 59-70, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18946492

RESUMEN

The human transferrin receptor (hTfR) is a target for cancer immunotherapy due to its overexpression on the surface of cancer cells. We previously developed an antibody-avidin fusion protein that targets hTfR (anti-hTfR IgG3-Av) and exhibits intrinsic cytotoxicity against certain malignant cells. Gambogic acid (GA), a drug that also binds hTfR, induces cytotoxicity in several malignant cell lines. We now report that anti-hTfR IgG3-Av and GA induce cytotoxicity in a new broader panel of hematopoietic malignant cell lines. Our results show that the effect of anti-hTfR IgG3-Av is iron-dependent whereas that of GA is iron-independent in all cells tested. In addition, we observed that GA exerts a TfR-independent cytotoxicity. We also found that GA increases the generation of reactive oxygen species that may play a role in the cytotoxicity induced by this drug. Additive cytotoxicity was observed by simultaneous combination treatment with these drugs and synergy by using anti-hTfR IgG3-Av as a chemosensitizing agent. In addition, we found a concentration of GA that is toxic to malignant hematopoietic cells but not to human hematopoietic progenitor cells. Our results suggest that these two compounds may be effective, alone or in combination, for the treatment of human hematopoietic malignancies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Inmunoglobulina G/farmacología , Receptores de Transferrina/inmunología , Xantonas/farmacología , Avidina , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Inmunoglobulina G/uso terapéutico , Hierro/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Xantonas/uso terapéutico
6.
Allergy ; 63(10): 1255-66, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18671772

RESUMEN

Epidemiological studies have suggested inverse associations between allergic diseases and malignancies. As a proof of concept for the capability of immunoglobulin E (IgE) to destruct tumor cells, several experimental strategies have evolved to specifically target this antibody class towards relevant tumor antigens. It could be demonstrated that IgE antibodies specific to overexpressed tumor antigens have been superior to any other immunoglobulin class with respect to antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) reactions. In an alternative approach, IgE nonspecifically attached to tumor cells proved to be a powerful adjuvant establishing tumor-specific immune memory. Active Th2 immunity could also be achieved by applying an oral immunization regimen using mimotopes, i.e. epitope mimics of tumor antigens. The induced IgE antibodies could be cross-linked by live tumor cells leading to tumoricidic mediator release. Thus, IgE antibodies may not only act in natural tumor surveillance, but could possibly also be exploited for tumor control in active and passive immunotherapy settings. Thereby, eosinophils, mast cells and macrophages can be armed with the cytophilic IgE and become potent anti-tumor effectors, able to trace viable tumor cells in the tissues. It is strongly suggested that the evolving new field AllergoOncology will give new insights into the role of IgE-mediated allergy in malignancies, possibly opening new avenues for tumor therapy.


Asunto(s)
Hipersensibilidad/inmunología , Inmunoglobulina E/fisiología , Neoplasias/inmunología , Animales , Basófilos/inmunología , Eosinófilos/inmunología , Humanos , Inmunoglobulina E/uso terapéutico , Mastocitos/inmunología , Neoplasias/epidemiología , Neoplasias/terapia
7.
Clin Exp Med ; 4(2): 57-64, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15672942

RESUMEN

Cancer patients who are administered therapeutic doses of cytokines (e.g., interleukin-2, granulocyte macrophage colony stimulating factor, interleukin-12, and tumor necrosis factor-alpha) frequently develop devastating toxic side effects that can lead to discontinuation of therapy. This problem has compelled numerous investigators to design innovative strategies that will reduce prolonged systemic cytokine exposure and promote cytokine accumulation at the site of the tumor. One such strategy involves the use of antibody-cytokine fusion proteins consisting of immunoenhancing cytokines genetically fused to antibodies that are able to target specific antigens exclusively expressed or overexpressed on the surface of tumor cells. Preclinical studies examining their therapeutic efficacy demonstrate that they posses potent tumoricidal activity, suggesting that they may be clinically useful as novel cancer therapeutic agents.


Asunto(s)
Anticuerpos/uso terapéutico , Citocinas/uso terapéutico , Neoplasias/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Interleucina-12/uso terapéutico , Interleucina-2/uso terapéutico , Factor de Necrosis Tumoral alfa/uso terapéutico
9.
J Interferon Cytokine Res ; 21(9): 709-20, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11576465

RESUMEN

We have constructed an antibody interleukin-12 (IL-12) fusion protein (mscIL-12.her2.IgG3) that demonstrates significant antitumor activity against the murine carcinoma CT26-expressing human HER2/neu. We now report that this antitumor activity is dose dependent and comparable to or better than recombinant murine IL-12 (rMuIL-12) using subcutaneous and metastatic models of disease. The antitumor activity of mscIL-12.her2.IgG3 is reduced in Rag2 knockout mice, suggesting that T cells play a role in tumor rejection. In SCID-beige mice, the antitumor activity is further reduced, suggesting that natural killer (NK) cells or macrophages or both are also important. The isotype of the antibody response to HER2/neu is consistent with a switch from a Th2 to a Th1 immune response and the infiltration of mononuclear cell in tumors from mice treated with mscIL-12.her2.IgG3. Immunohistochemistry reveals that mscIL-12.her2.IgG3 is antiangiogenic. Thus, the mechanism of the antitumor activity exhibited by mscIL-12.her2.IgG3 is highly complex and involves a combination of T and NK cell activity, a switch to a Th1 immune response, and antiantiogenic activity. This is the first study comparing the in vivo antitumor activity of an antibody-IL-12 fusion protein and free IL-12. Our results suggest that antibody-IL-12 fusion proteins may be useful for the treatment of human cancer.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Interleucina-12/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Anticuerpos Antineoplásicos/genética , Anticuerpos Antineoplásicos/inmunología , Complejo CD3/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Genes erbB-2/inmunología , Humanos , Inmunoglobulina G/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Neoplasias Pulmonares/secundario , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Proteínas Nucleares , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología , Células TH1/inmunología , Distribución Tisular
10.
Hum Antibodies ; 10(1): 43-9, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11455061

RESUMEN

Anti-HER2/neu therapy of human HER2/neu expressing malignancies such as breast cancer has shown only partial success in clinical trials. To expand the clinical potential of this approach, we have genetically engineered an anti-HER2/neu human IgG3 fusion protein containing interleukin-2 (IL-2) fused at its carboxyl terminus. Anti-Her2/neu IgG3-(IL-2) retained antibody and cytokine related activity. Treatment of immunocompentent mice with this antibody fusion protein resulted in significant retardation in the subcutaneous (s.c.) growth of CT26-HER2/neu tumors suggesting that anti-HER2/neu IgG3-(IL-2) fusion protein will be useful in the treatment of HER2/neu expressing tumors. We also found that fusing IL-2 to human IgG3 results in a significant enhancement of the murine anti-human antibody (MAHA) response.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoglobulina G/uso terapéutico , Interleucina-2/uso terapéutico , Neoplasias Experimentales/terapia , Receptor ErbB-2/biosíntesis , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Anticuerpos Antineoplásicos/genética , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/uso terapéutico , Línea Celular , Femenino , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Interleucina-2/genética , Ratones , Ratones Endogámicos BALB C , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Trastuzumab
11.
Cancer Immunol Immunother ; 49(12): 649-62, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11258791

RESUMEN

In the present study we describe a novel murine tumor model in which the highly malignant murine B cell lymphoma 38C13 has been transduced with the cDNA encoding human tumor-associated antigen HER2/neu. This new cell line (38C13-HER2/neu) showed stable surfiace expression but not secretion of human HER2/neu. It also maintained expression of the idiotype (Id) of the surface immunoglobulin of 38C13, which serves as another tumor-associated antigen. Surprisingly, spontaneous tumor regression was observed following s.c. but not i.v. injection of 38C13-HER2/neu cells in immunocompetent syngeneic mice. Regression was more frequently observed with larger tumor cell challenges and was mediated through immunological mechanisms because it was not observed in syngeneic immunodeficient mice. Mice that showed complete tumor regression were immune to challenge with the parental cell line 38C13 and V1, a variant of 38C13 that does not express the Id. Immunity could be transferred with sera, suggesting that an antibody response mediated rejection and immunity. Continuously growing s.c. tumors as well as metastatic tumors obtained after the i.v. injection of 38C13-HER2/neu maintained expression of human HER2/neu, which can serve as a target for active immunotherapy. As spontaneous tumor regression has not been observed in other human murine models expressing human HER2/neu, our results illustrate the enormous differences that can exist among different murine tumors expressing the same antigen. The present model provides a useful tool for the study of the mechanisms of protective immunity to B cell lymphoma and for the evaluation of different therapeutic approaches based on the stimulation or suppression of the immune response.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Idiotipos de Inmunoglobulinas/inmunología , Linfoma de Células B/inmunología , Regresión Neoplásica Espontánea/patología , Receptor ErbB-2/metabolismo , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Anticuerpos Monoclonales/uso terapéutico , División Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Expresión Génica , Humanos , Inmunidad , Inmunización Pasiva , Inmunoglobulina G/inmunología , Inyecciones Intravenosas , Inyecciones Subcutáneas , Linfoma de Células B/patología , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Trasplante de Neoplasias , Retroviridae/genética , Linfocitos T/inmunología , Transfección , Células Tumorales Cultivadas
12.
J Immunol Methods ; 248(1-2): 91-101, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11223071

RESUMEN

Advances in genetic engineering and expression systems have led to rapid progress in the development of antibodies fused to other proteins. These 'antibody fusion proteins' have novel properties and include antibodies with specificity for tumor associated antigens fused to cytokines such as interleukin-2 (IL2), granulocyte/macrophage colony-stimulating factor (GM-CSF), and interleukin-12 (IL12). The goal of this approach to cancer therapy is to concentrate the cytokine in the tumor microenvironment and in so doing directly enhance the tumoricidal effect of the antibody and/or enhance the host immune response (T-cell, B-cell or NK) against the tumor. In the past decade, multiple antibody-cytokine fusion proteins have been developed with different specificities targeting a broad variety of tumors. These novel molecules retain both antibody and cytokine associated functions. In addition, in animals bearing tumors, antibody-cytokine fusion proteins are able to target the tumor and to elicit a significant anti-tumor response that in some cases results in a complete elimination of the tumor. These results suggest that antibody-cytokine fusion proteins have potential for use in the treatment of human cancer. In the present review, we describe strategies for construction of antibody-cytokine fusion proteins and discuss the properties of several antibody-cytokine fusion proteins with IgG genetically fused to the cytokines IL2, GM-CSF or IL12.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Inmunoglobulina G/uso terapéutico , Interleucina-12/uso terapéutico , Interleucina-2/uso terapéutico , Neoplasias/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Humanos , Ratones
13.
J Immunol ; 165(9): 5112-21, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11046042

RESUMEN

Anti-HER2/neu therapy of human HER2/neu-expressing malignancies such as breast cancer has shown only partial success in clinical trials. To expand the clinical potential of this approach, we have genetically engineered an anti-HER2/neu IgG3 fusion protein containing GM-CSF. Anti-HER2/neu IgG3-(GM-CSF) expressed in myeloma cells was correctly assembled and secreted. It was able to target HER2/neu-expressing cells and to support growth of a GM-CSF-dependent murine myeloid cell line, FDC-P1. The Ab fusion protein activated J774.2 macrophage cells so that they exhibit an enhanced cytotoxic activity and was comparable to the parental Ab in its ability to effect Ab-dependent cellular cytotoxicity-mediated tumor cell lysis. Pharmacokinetic studies showed that anti-HER2/neu IgG3-(GM-CSF) is stable in the blood. Interestingly, the half-life of anti-HER2/neu IgG3-(GM-CSF) depended on the injected dose with longer in vivo persistence observed at higher doses. Biodistribution studies showed that anti-HER2/neu IgG3-(GM-CSF) is mainly localized in the spleen. In addition, anti-HER2/neu IgG3-(GM-CSF) was able to target the HER2/neu-expressing murine tumor CT26-HER2/neu and enhance the immune response against the targeted Ag HER2/neu. Anti-HER2/neu IgG3-(GM-CSF) is able to enhance both Th1- and Th2-mediated immune responses and treatment with this Ab fusion protein resulted in significant retardation in the growth of s.c. CT26-HER2/neu tumors. Our results suggest that anti-HER2/neu IgG3-(GM-CSF) fusion protein is useful in the treatment of HER2/neu-expressing tumors.


Asunto(s)
Anticuerpos Antineoplásicos/administración & dosificación , Anticuerpos Antineoplásicos/genética , Antineoplásicos/síntesis química , Citocinas/fisiología , Epítopos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Inmunoglobulina G/genética , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/prevención & control , Animales , Anticuerpos Antiidiotipos/biosíntesis , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , División Celular/genética , División Celular/inmunología , Línea Celular , Membrana Celular/genética , Membrana Celular/inmunología , Membrana Celular/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Citotoxicidad Inmunológica/genética , Epítopos/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Semivida , Humanos , Inmunoglobulina G/administración & dosificación , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/clasificación , Inyecciones Intravenosas , Inyecciones Subcutáneas , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Unión Proteica/genética , Unión Proteica/inmunología , Receptor ErbB-2/administración & dosificación , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/farmacocinética , Células Tumorales Cultivadas
14.
J Immunol ; 163(8): 4421-6, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10510383

RESUMEN

In the present study a novel Ab-avidin fusion protein has been constructed to deliver biotinylated compounds across the blood brain barrier. This fusion molecule consists of an Ab specific for the transferrin receptor genetically fused to avidin. The Ab-avidin fusion protein (anti-TfR IgG3-CH3-Av) expressed in murine myeloma cells was correctly assembled and secreted and showed both Ab- and avidin-related activities. In animal models, it showed much longer serum half-life than the chemical conjugate between OX-26 and avidin. Most importantly, this fusion protein demonstrated superior [3H]biotin uptake into brain parenchyma in comparison with the chemical conjugate. We also delivered a biotinylated 18-mer antisense peptide-nucleic acid specific for the rev gene of HIV-1 to the brain. Brain uptake of the HIV antisense drug was increased at least 15-fold when it was bound to the anti-TfR IgG3-CH3-Av, suggesting its potential use in neurologic AIDS. This novel Ab fusion protein should have general utility as a universal vehicle to effectively deliver biotinylated compounds across the blood-brain barrier for diagnosis and/or therapy of a broad range of CNS disorders such as infectious diseases, brain tumors as well as Parkinson's and Huntington's diseases.


Asunto(s)
Especificidad de Anticuerpos , Avidina/genética , Encéfalo/metabolismo , Inmunoglobulina G/genética , Oligonucleótidos Antisentido/metabolismo , Receptores de Transferrina/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Animales , Fármacos Anti-VIH/metabolismo , Especificidad de Anticuerpos/genética , Avidina/metabolismo , Biotina/sangre , Biotina/farmacocinética , Encéfalo/inmunología , Marcación de Gen , Humanos , Regiones Constantes de Inmunoglobulina/genética , Regiones Constantes de Inmunoglobulina/metabolismo , Inmunoglobulina G/metabolismo , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Radioisótopos de Yodo/farmacocinética , Masculino , Ratones , Ácidos Nucleicos de Péptidos/farmacocinética , Ratas , Ratas Sprague-Dawley , Receptores de Transferrina/genética , Proteínas Recombinantes de Fusión/genética , Tritio
15.
J Immunol ; 163(1): 250-8, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10384123

RESUMEN

IL-12 is a heterodimeric cytokine with many actions on innate and cellular immunity that may have antitumor and antimetastatic effects. However, systemic administration of IL-12 can be toxic. Tumor-specific Abs provide a means to selectively target a metastatic/residual nodule and deliver therapeutic quantities of an immunostimulatory molecule like IL-12 with lower systemic levels and ideally, toxicity. We report the construction and characterization of an Ab fusion protein in which single-chain murine IL-12 is fused to an anti-Her2/neu Ab at the amino terminus (mscIL-12.her2.IgG3). The use of single-chain IL-12 in the fusion protein simplifies vector construction, ensures equimolar concentrations of the two IL-12 subunits, and may confer greater stability to the fusion protein. SDS-PAGE analysis shows this 320-kDa protein is secreted and correctly assembled. FACS analysis demonstrates that this fusion protein binds to cells transfected with the Her2/neu Ag, thus retaining Ab specificity; this fusion protein also binds to a cell line and to PHA-activated PBMC that express the IL-12R, thus demonstrating cytokine receptor specificity. T cell proliferation assays and NK cytotoxicity assays demonstrate that this fusion protein exhibits IL-12 bioactivity comparable to recombinant murine IL-12. In vivo studies demonstrate that this fusion protein has antitumor activity. These results are significant and suggest that this IL-12 Ab fusion protein can effectively combine the therapeutic potential of IL-12 with the tumor-targeting ability of the Ab and may provide a viable alternative to systemic administration of IL-12.


Asunto(s)
Especificidad de Anticuerpos/genética , Antineoplásicos/química , Inmunoglobulina G/genética , Interleucina-12/genética , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/terapia , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos de Neoplasias/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Sitios de Unión de Anticuerpos/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Citotoxicidad Inmunológica/inmunología , Femenino , Humanos , Inmunoglobulina G/administración & dosificación , Inyecciones Subcutáneas , Interleucina-12/administración & dosificación , Interleucina-12/metabolismo , Células K562 , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Unión Proteica/genética , Unión Proteica/inmunología , Receptor ErbB-2/administración & dosificación , Receptor ErbB-2/genética , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12 , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/biosíntesis , Células Tumorales Cultivadas
16.
Lab Anim Sci ; 49(2): 179-88, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10331548

RESUMEN

BACKGROUND AND PURPOSE: Expression of the HER2/neu proto-oncogene, a receptor-like transmembrane protein expressed at low levels on some normal cells, is markedly increased in a subset of human breast, colon, lung, and ovarian cancers. A humanized HER2/neu antibody has been tested as a therapeutic agent in several clinical trials, with promising results. We have developed a family of anti-HER2/neu fusion proteins. To evaluate the immunologic efficacy of these proteins, it is critical that tumors expressing the target antigen can grow in immunologically intact mice. METHOD: To produce murine tumors expressing human HER2/neu on the surface, CT26, MC38, and EL4 murine cell lines were transduced by use of a retroviral construct containing the cDNA encoding the human HER2/neu gene. RESULTS: Histologic features and kinetics of tumor growth in subcutaneous space of the human HER2/neu-expressing cells were similar to those of the respective parental cell lines. Intravenous inoculation with these cells induced disseminated malignant disease. Flow cytometric and immmunohistochemical analyses of freshly isolated tumors revealed in vivo expression of human HER2/neu. Secretion of antigen was not detected by use of an ELISA. CONCLUSION: Although an antibody response against the human HER2/neu antigen was observed, this response does not affect the growth rate of the HER2/neu-expressing cells. These murine models may be useful tools for evaluation of anti-cancer therapeutic approaches that target human HER2/neu.


Asunto(s)
Expresión Génica , Receptor ErbB-2/genética , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citometría de Flujo , Humanos , Inmunoglobulina G/análisis , Linfoma/inmunología , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Proto-Oncogenes Mas , Receptor ErbB-2/análisis , Linfocitos T/inmunología , Transfección , Células Tumorales Cultivadas
17.
J Immunol ; 161(7): 3729-36, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9759898

RESUMEN

The successful eradication of cancer cells in the setting of minimal residual disease may require targeting of metastatic tumor deposits that evade the immune system. We combined the targeting flexibility and specificity of mAbs with the immune effector function of the chemokine RANTES to target established tumor deposits. We describe the construction of an Ab fusion molecule with variable domains directed against the tumor-associated Ag HER2/neu, linked to sequences encoding the chemokine RANTES (RANTES.her2.IgG3). RANTES is a potent chemoattractant of T cells, NK cells, monocytes, and dendritic cells, and expression of RANTES has been shown to enhance immune responses against tumors in murine models. RANTES.her2.IgG3 fusion protein bound specifically to HER2/neu Ag expressed on EL4 cells and on SKBR3 breast cancer cells as assayed by flow cytometry. RANTES.her2.IgG3 could elicit actin polymerization of THP-1 cells and transendothelial migration of primary T lymphocytes. RANTES.her2.IgG3 prebound to SKBR3 cells also facilitated migration of T cells. RANTES.her2.IgG3 bound specifically to the CCR5 chemokine receptor, as demonstrated by flow cytometry, and inhibited HIV-1 infection via the CCR5 coreceptor. RANTES.her2.IgG3, alone or in combination with other chemokine or cytokine fusion Abs, may be a suitable reagent for recruitment and activation of an expanded repertoire of effector cells to tumor deposits.


Asunto(s)
Antígenos de Neoplasias/genética , Quimiocina CCL5/inmunología , Quimiocinas/fisiología , Epítopos/genética , Inmunoglobulina G/genética , Proteínas Recombinantes de Fusión/química , Secuencia de Aminoácidos , Antígenos de Neoplasias/fisiología , Secuencia de Bases , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiocinas/genética , Quimiotaxis , Endotelio Vascular/inmunología , Endotelio Vascular/fisiología , Epítopos/fisiología , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/metabolismo , Datos de Secuencia Molecular , Unión Proteica/genética , Unión Proteica/inmunología , Ingeniería de Proteínas , Receptor ErbB-2/genética , Receptores CCR5/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T/inmunología , Venas Umbilicales
18.
J Interferon Cytokine Res ; 18(8): 597-607, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9726441

RESUMEN

Antibody (Ab)-based tumor therapeutics use the tumor-binding specificity of the Ab to target Fc functions or associated molecules to the site of the tumor. We have used an Ab-interleukin-2 (IL-2) fusion protein to deliver IL-2 to a murine B cell lymphoma (38C13). This anti-Id IgG3-CH3-IL-2, which recognizes the idiotype present on the surface of the lymphoma has a half-life in mice approximately 17-fold longer than the half-life reported for IL-2. Gamma camera studies showed that anti-Id IgG3-CH3-IL-2 localizes at the site of a subcutaneous tumor in mice. The anti-Id IgG3-CH3-IL-2 also shows enhanced antitumor activity compared with the combination of Ab and IL-2 administered together. However, the mechanism of antitumor activity appears to depend on the dose and the treatment schedule used. A single dose of fusion protein prevented tumor in only 50% of the animals, although all the survivors showed some evidence of immunologic memory. Although multiple doses are more effective in preventing tumor growth (87% survivors), they are ineffective in generating protective immunologic memory. Our results suggest that Ab-IL-2 fusion proteins will be useful in the diagnosis and treatment of human B cell lymphomas and other related malignancies.


Asunto(s)
Anticuerpos Antiidiotipos/uso terapéutico , Antineoplásicos/inmunología , Inmunoglobulina G/uso terapéutico , Interleucina-2/uso terapéutico , Linfoma de Células B/terapia , Proteínas Recombinantes de Fusión/inmunología , Animales , Reacciones Antígeno-Anticuerpo , Antígenos de Neoplasias/inmunología , Relación Dosis-Respuesta Inmunológica , Femenino , Cámaras gamma , Semivida , Linfoma de Células B/diagnóstico por imagen , Ratones , Ratones Endogámicos C3H , Ratones Desnudos , Cintigrafía
19.
J Immunol ; 160(7): 3419-26, 1998 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9531302

RESUMEN

We describe the construction and characterization of an Ab fusion protein specific for the tumor-associated Ag HER2/neu linked to sequences encoding the extracellular domain of the B7.1 T cell costimulatory ligand. The Ab domain of the fusion molecule will specifically target HER2/neu-expressing tumor cells, while the B7.1 domain is designed to activate a specific immune response. We show that the B7.1 fusion Ab retained ability to selectively bind to the HER2/neu Ag and to the CTLA4/CD28 counter-receptors for B7.1. Specific T cell activation was observed when the B7.1 Ab fusion protein was bound to HER2/neu-expressing cells. The use of the B7.1 Ab fusion protein may overcome limitations of gene transfer and/or standard Ab therapy and represents a novel approach to the eradication of minimal residual disease.


Asunto(s)
Especificidad de Anticuerpos/genética , Antígeno B7-1/inmunología , Inmunoglobulina G/genética , Activación de Linfocitos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Secuencia de Bases , Sitios de Unión de Anticuerpos/genética , Células CHO , Membrana Celular/genética , Membrana Celular/inmunología , Cricetinae , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/fisiología , Activación de Linfocitos/genética , Datos de Secuencia Molecular , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/fisiología
20.
Hum Antibodies ; 8(3): 106-18, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9322080

RESUMEN

Advances in genetic engineering and expression systems have led to a rapid progress in the development of immunoglobulins fused to other proteins. These 'antibody fusion proteins' have novel properties and include antibodies fused to the cytokine interleukin-2. In the present review we describe strategies for construction of these antibody-interleukin-2 fusion proteins and discuss their in vitro and in vivo properties. Antibody-interleukin-2 fusion proteins retain both antibody associated functions such as antigen binding, complement activation and Fc gamma receptor binding as well as interleukin-2 associated functions such as the stimulation of proliferation of CTLL2 cells. In vivo, they produce strong potentiation of the host immune response against any associated antigen. In addition, these novel molecules are able to target tumor cells and produce a specific and effective T cell response capable of eliminating the tumor. These properties suggest that antibody-interleukin-2 fusion proteins will be useful in the diagnosis and/or treatment of human cancer as well as in the potentiation of human response against any associated antigen.


Asunto(s)
Anticuerpos/inmunología , Interleucina-2/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos/genética , Humanos , Inmunoterapia , Interleucina-2/genética , Neoplasias/terapia , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA