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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Allergy ; 73(2): 328-340, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28921585

RESUMO

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.


Assuntos
Hipersensibilidade/imunologia , Hipersensibilidade/terapia , Tolerância Imunológica/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Dessensibilização Imunológica/métodos , Humanos
2.
Allergy ; 72(6): 866-887, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28032353

RESUMO

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.


Assuntos
Hipersensibilidade/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Anticorpos , Humanos , Imunoglobulina E/imunologia , Vigilância Imunológica , Imunoterapia/tendências , Neoplasias/terapia , Células Th2/imunologia
3.
Allergy ; 63(10): 1255-66, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18671772

RESUMO

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.


Assuntos
Hipersensibilidade/imunologia , Imunoglobulina E/fisiologia , Neoplasias/imunologia , Animais , Basófilos/imunologia , Eosinófilos/imunologia , Humanos , Imunoglobulina E/uso terapêutico , Mastócitos/imunologia , Neoplasias/epidemiologia , Neoplasias/terapia
4.
J Interferon Cytokine Res ; 18(8): 597-607, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9726441

RESUMO

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.


Assuntos
Anticorpos Anti-Idiotípicos/uso terapêutico , Antineoplásicos/imunologia , Imunoglobulina G/uso terapêutico , Interleucina-2/uso terapêutico , Linfoma de Células B/terapia , Proteínas Recombinantes de Fusão/imunologia , Animais , Reações Antígeno-Anticorpo , Antígenos de Neoplasias/imunologia , Relação Dose-Resposta Imunológica , Feminino , Câmaras gama , Meia-Vida , Linfoma de Células B/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Cintilografia
5.
J Interferon Cytokine Res ; 21(9): 709-20, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11576465

RESUMO

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.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Interleucina-12/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/imunologia , Complexo CD3/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Genes erbB-2/imunologia , Humanos , Imunoglobulina G/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Células Matadoras Naturais/imunologia , Fígado/imunologia , Neoplasias Pulmonares/secundário , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Proteínas Nucleares , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Células Th1/imunologia , Distribuição Tecidual
6.
J Immunol Methods ; 248(1-2): 91-101, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11223071

RESUMO

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.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Imunoglobulina G/uso terapêutico , Interleucina-12/uso terapêutico , Interleucina-2/uso terapêutico , Neoplasias/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Humanos , Camundongos
7.
Clin Exp Med ; 4(2): 57-64, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15672942

RESUMO

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.


Assuntos
Anticorpos/uso terapêutico , Citocinas/uso terapêutico , Neoplasias/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Interleucina-12/uso terapêutico , Interleucina-2/uso terapêutico , Fator de Necrose Tumoral alfa/uso terapêutico
8.
Hum Antibodies ; 8(3): 106-18, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9322080

RESUMO

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.


Assuntos
Anticorpos/imunologia , Interleucina-2/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos/genética , Humanos , Imunoterapia , Interleucina-2/genética , Neoplasias/terapia , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico
9.
Hum Antibodies ; 10(1): 43-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11455061

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Imunoglobulina G/uso terapêutico , Interleucina-2/uso terapêutico , Neoplasias Experimentais/terapia , Receptor ErbB-2/biossíntese , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/uso terapêutico , Linhagem Celular , Feminino , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Interleucina-2/genética , Camundongos , Camundongos Endogâmicos BALB C , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Trastuzumab
10.
Vet Parasitol ; 57(4): 339-49, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7660571

RESUMO

Current methods for the control of cattle tick Boophilus microplus infestations are not effective and the parasite remains a serious problem for the cattle industry in tropical and subtropical areas. Recently, we developed a vaccine against B. microplus employing a recombinant Bm86 (rBm86) antigen preparation (Gavac, Heber Biotec) and it was shown to induce a protective response in vaccinated animals under controlled conditions. Here we show that, under field conditions in grazing cattle, the vaccine is able to control B. microplus populations. Two parasite-free farms were employed for the study. In the first farm, animals were vaccinated with the recombinant vaccine, while, in the second, animals received a saline injection in adjuvant. After immunization, animals were artificially infected and the infestation rate was recorded. Over the 33 weeks of the experiment, the infestation rate was lower in the vaccinated group compared with the control group. At the end of the experiment it was necessary to use chemicals in the control farm after serious losses in production and animals.


Assuntos
Doenças dos Bovinos/prevenção & controle , Infestações por Carrapato/veterinária , Vacinação/veterinária , Animais , Antígenos/isolamento & purificação , Bovinos , Doenças dos Bovinos/parasitologia , Feminino , Infestações por Carrapato/parasitologia , Infestações por Carrapato/prevenção & controle , Carrapatos/imunologia , Fatores de Tempo , Vacinas Sintéticas/isolamento & purificação
11.
Leukemia ; 24(9): 1555-65, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20574452

RESUMO

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.


Assuntos
Morte Celular/genética , Genes Homeobox , Ferro/metabolismo , Linfoma de Células B/patologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Linfoma de Células B/metabolismo , Reação em Cadeia da Polimerase
12.
Leukemia ; 23(1): 59-70, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18946492

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Hematológicas/tratamento farmacológico , Imunoglobulina G/farmacologia , Receptores da Transferrina/imunologia , Xantonas/farmacologia , Avidina , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Humanos , Imunoglobulina G/uso terapêutico , Ferro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Xantonas/uso terapêutico
13.
Hum Gene Ther ; 20(5): 453-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19215191

RESUMO

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.


Assuntos
Vacinas Anticâncer/imunologia , Interleucina-12/imunologia , Síndromes Neoplásicas Hereditárias/prevenção & controle , Receptor ErbB-2/genética , Proteína Supressora de Tumor p53/genética , Vacinas de DNA/imunologia , Animais , Citocinas/biossíntese , Citocinas/imunologia , Citotoxicidade Imunológica , Feminino , Terapia Genética , Imunoglobulina G/sangue , Imunoterapia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-12/metabolismo , Complexo Principal de Histocompatibilidade/imunologia , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/patologia , Camundongos , Síndromes Neoplásicas Hereditárias/terapia , Ratos , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Glândulas Salivares/imunologia , Glândulas Salivares/patologia , Transfecção , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
14.
J Immunol ; 163(1): 250-8, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10384123

RESUMO

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.


Assuntos
Especificidade de Anticorpos/genética , Antineoplásicos/química , Imunoglobulina G/genética , Interleucina-12/genética , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos de Neoplasias/metabolismo , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Sítios de Ligação de Anticorpos/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Citotoxicidade Imunológica/imunologia , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Injeções Subcutâneas , Interleucina-12/administração & dosagem , Interleucina-12/metabolismo , Células K562 , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/genética , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12 , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Células Tumorais Cultivadas
15.
J Immunol ; 165(9): 5112-21, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11046042

RESUMO

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.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/genética , Antineoplásicos/síntese química , Citocinas/fisiologia , Epitopos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunoglobulina G/genética , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adenocarcinoma/prevenção & controle , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/química , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Divisão Celular/genética , Divisão Celular/imunologia , Linhagem Celular , Membrana Celular/genética , Membrana Celular/imunologia , Membrana Celular/metabolismo , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Citotoxicidade Imunológica/genética , Epitopos/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Meia-Vida , Humanos , Imunoglobulina G/administração & dosagem , Isotipos de Imunoglobulinas/biossíntese , Isotipos de Imunoglobulinas/classificação , Injeções Intravenosas , Injeções Subcutâneas , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/farmacocinética , Células Tumorais Cultivadas
16.
J Immunol ; 163(8): 4421-6, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10510383

RESUMO

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.


Assuntos
Especificidade de Anticorpos , Avidina/genética , Encéfalo/metabolismo , Imunoglobulina G/genética , Oligonucleotídeos Antissenso/metabolismo , Receptores da Transferrina/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Animais , Fármacos Anti-HIV/metabolismo , Especificidade de Anticorpos/genética , Avidina/metabolismo , Biotina/sangue , Biotina/farmacocinética , Encéfalo/imunologia , Marcação de Genes , Humanos , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/metabolismo , Imunoglobulina G/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/metabolismo , Radioisótopos do Iodo/farmacocinética , Masculino , Camundongos , Ácidos Nucleicos Peptídicos/farmacocinética , Ratos , Ratos Sprague-Dawley , Receptores da Transferrina/genética , Proteínas Recombinantes de Fusão/genética , Trítio
17.
Lab Anim Sci ; 49(2): 179-88, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10331548

RESUMO

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.


Assuntos
Expressão Gênica , Receptor ErbB-2/genética , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Citometria de Fluxo , Humanos , Imunoglobulina G/análise , Linfoma/imunologia , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Proto-Oncogene Mas , Receptor ErbB-2/análise , Linfócitos T/imunologia , Transfecção , Células Tumorais Cultivadas
18.
Cancer Immunol Immunother ; 49(12): 649-62, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11258791

RESUMO

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.


Assuntos
Anticorpos Antineoplásicos/imunologia , Idiótipos de Imunoglobulinas/imunologia , Linfoma de Células B/imunologia , Regressão Neoplásica Espontânea/patologia , Receptor ErbB-2/metabolismo , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Anticorpos Monoclonais/uso terapêutico , Divisão Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Humanos , Imunidade , Imunização Passiva , Imunoglobulina G/imunologia , Injeções Intravenosas , Injeções Subcutâneas , Linfoma de Células B/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Transplante de Neoplasias , Retroviridae/genética , Linfócitos T/imunologia , Transfecção , Células Tumorais Cultivadas
19.
J Immunol ; 160(7): 3419-26, 1998 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9531302

RESUMO

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.


Assuntos
Especificidade de Anticorpos/genética , Antígeno B7-1/imunologia , Imunoglobulina G/genética , Ativação Linfocitária/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Sequência de Bases , Sítios de Ligação de Anticorpos/genética , Células CHO , Membrana Celular/genética , Membrana Celular/imunologia , Cricetinae , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/fisiologia , Ativação Linfocitária/genética , Dados de Sequência Molecular , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/fisiologia
20.
J Immunol ; 161(7): 3729-36, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9759898

RESUMO

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.


Assuntos
Antígenos de Neoplasias/genética , Quimiocina CCL5/imunologia , Quimiocinas/fisiologia , Epitopos/genética , Imunoglobulina G/genética , Proteínas Recombinantes de Fusão/química , Sequência de Aminoácidos , Antígenos de Neoplasias/fisiologia , Sequência de Bases , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiocinas/genética , Quimiotaxia , Endotélio Vascular/imunologia , Endotélio Vascular/fisiologia , Epitopos/fisiologia , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/metabolismo , Dados de Sequência Molecular , Ligação Proteica/genética , Ligação Proteica/imunologia , Engenharia de Proteínas , Receptor ErbB-2/genética , Receptores CCR5/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/imunologia , Veias Umbilicais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa