Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncoimmunology ; 11(1): 2127508, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36249274

RESUMO

Glioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated tumor-cell specific delivery of an anti-PD-1 immunoadhesin (aPD-1) via a targeted adeno-associated viral vector (AAV) as well as HER2-specific NK-92/5.28.z (anti-HER2.CAR/NK-92) cells as components for a combination immunotherapy. In co-culture experiments, target-activated anti-HER2.CAR/NK-92 cells modified surrounding tumor cells and bystander immune cells by triggering the release of inflammatory cytokines and upregulation of PD-L1. Tumor cell-specific delivery of aPD-1 was achieved by displaying a HER2-specific designed ankyrin repeat protein (DARPin) on the AAV surface. HER2-AAV mediated gene transfer into GB cells correlated with HER2 expression levels, without inducing anti-viral responses in transduced cells. Furthermore, AAV-transduction did not interfere with anti-HER2.CAR/NK-92 cell-mediated tumor cell lysis. After selective transduction of HER2+ cells, aPD-1 expression was detected at the mRNA and protein level. The aPD-1 immunoadhesin was secreted in a time-dependent manner, bound its target on PD-1-expressing cells and was able to re-activate T cells by efficiently disrupting the PD-1/PD-L1 axis. Moreover, high intratumoral and low systemic aPD-1 concentrations were achieved following local injection of HER2-AAV into orthotopic tumor grafts in vivo. aPD-1 was selectively produced in tumor tissue and could be detected up to 10 days after a single HER2-AAV injection. In subcutaneous GL261-HER2 and Tu2449-HER2 immunocompetent mouse models, administration of the combination therapy significantly prolonged survival, including complete tumor control in several animals in the GL261-HER2 model. In summary, local therapy with aPD-1 encoding HER2-AAVs in combination with anti-HER2.CAR/NK-92 cells may be a promising novel strategy for GB immunotherapy with the potential to enhance efficacy and reduce systemic side effects of immune-checkpoint inhibitors.


Assuntos
Glioblastoma , Adenoviridae/genética , Animais , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Citocinas , Glioblastoma/genética , Glioblastoma/terapia , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/transplante , Camundongos , RNA Mensageiro , Receptor ErbB-2/metabolismo , Terapias em Estudo , Microambiente Tumoral
2.
Oncogene ; 29(32): 4517-27, 2010 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-20514015

RESUMO

Aberrant activation of the epidermal growth factor receptor (EGFR) has been found in human cancers of various origins, and has been implicated in cancer pathogenesis. The therapeutic anti-EGFR antibodies cetuximab and matuzumab inhibit both ligand-induced receptor activation and growth of EGFR-expressing tumor cells. The efficacy of such EGFR-targeted therapies may be further enhanced by induction of functionally equivalent endogenous antibody responses. Here we describe novel peptide sequences selected from random peptide libraries for binding to single-chain antibody fragments of cetuximab or matuzumab. Two of these peptides characterized by KTL and YPLG motifs are recognized equally well by cetuximab and matuzumab, although nonoverlapping epitopes were previously reported for these antibodies. Immunization of experimental animals with synthetic KTL- and YPLG-containing peptides led to induction of antibodies that cross-react with human EGFR, and prevent binding of natural EGFR ligands, ligand-induced receptor activation and tumor cell growth in a manner similar to cetuximab and matuzumab. Our findings show that these peptide mimotopes can induce anti-EGFR antibodies with antitumoral activity, which may have implications for EGFR-specific cancer immunotherapy.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Epitopos/imunologia , Receptores ErbB/imunologia , Peptídeos/química , Peptídeos/imunologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Cetuximab , Reações Cruzadas , Epitopos/química , Humanos , Imunoterapia , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Neoplasias/imunologia , Neoplasias/terapia , Biblioteca de Peptídeos , Estrutura Terciária de Proteína
3.
Cell Death Dis ; 1: e68, 2010 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21364672

RESUMO

In an attempt to improve TRAIL's (tumor necrosis factor-related apoptosis-inducing ligand) tumor selective activity a variant was designed, in which the three TRAIL protomers are expressed as a single polypeptide chain (scTRAIL). By genetic fusion with a single-chain antibody fragment (scFv) recognizing the extracellular domain of ErbB2, we further equipped scTRAIL with tumor-targeting properties. We studied tumor targeting and apoptosis induction of scFv-scTRAIL in comparison with non-targeted scTRAIL. Importantly, the tumor antigen-targeted scTRAIL fusion protein showed higher apoptotic activity in vitro, with a predominant action by TRAIL-R2 signaling. Pharmacokinetic studies revealed increased plasma half-life of the targeted scTRAIL fusion protein compared with scTRAIL. In vivo studies in a mouse tumor model with xenotransplanted Colo205 cells confirmed greater response to the ErbB2-specific scTRAIL fusion protein compared with non-targeted scTRAIL both under local and systemic application regimen. Together, in vitro and in vivo data give proof of concept of higher therapeutic activity of tumor-targeted scFv-scTRAIL molecules. Further, we envisage that through targeting of scTRAIL, potential side effects should be minimized. We propose that scFv-mediated tumor targeting of single-chain TRAIL represents a promising strategy to improve TRAIL's antitumoral action and to minimize potential unwanted actions on normal tissues.Cell Death and Disease (2010) 1, e68; doi:10.1038/cddis.2010.45; published online 26 August 2010.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Proteínas Recombinantes de Fusão/uso terapêutico , Anticorpos de Cadeia Única/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Epitopos , Meia-Vida , Humanos , Camundongos , Camundongos Nus , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/farmacocinética , Anticorpos de Cadeia Única/sangue , Anticorpos de Cadeia Única/farmacocinética , Anticorpos de Cadeia Única/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/sangue , Ligante Indutor de Apoptose Relacionado a TNF/farmacocinética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Leukemia ; 23(9): 1634-42, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19357702

RESUMO

The Wilms tumor antigen, WT1, is expressed at high levels in various types of leukemia and solid tumors, including lung, breast, colon cancer and soft tissue sarcomas. The WT1 protein has been found to be highly immunogenic, and spontaneous humoral and cytotoxic T-cell responses have been detected in patients suffering from leukemia. Furthermore, major histocompatibility complexes class I- and II-restricted WT1 peptide epitopes have been shown to elicit immune responses in patients with WT1-expressing tumors. As a consequence, WT1 has become an attractive target for anticancer immunotherapy. In this study, we investigated the feasibility of generating WT1-specific T cells for adoptive immunotherapy after allogeneic stem cell transplantation. We analyzed the incidence of T cells specific for WT1 peptide epitopes in cancer patients and healthy volunteers. It is noted that we could generate WT1-specific responses in nine of ten healthy volunteer donors and established T-cell clones specific for two WT1-derived peptide epitopes. These in vitro expanded WT1-specific T cells effectively lysed WT1-expressing tumor cell lines, indicating the potential clinical impact of ex vivo expanded donor-derived WT1-specific T cells for adoptive immunotherapy after allogeneic stem cell transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Imunoterapia Adotiva , Leucemia/terapia , Sarcoma/terapia , Linfócitos T Citotóxicos/imunologia , Proteínas WT1/imunologia , Linhagem Celular Tumoral , Epitopos , Antígeno HLA-A2/análise , Humanos , Imunofenotipagem , Orthomyxoviridae/imunologia , Transplante Homólogo , Proteínas WT1/genética
5.
Cell Death Differ ; 14(9): 1678-87, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17557115

RESUMO

Fas ligand (FasL) is a type II transmembrane protein belonging to the tumor necrosis factor family. Its binding to the cognate Fas receptor triggers the apoptosis that plays a pivotal role in the maintenance of immune system homeostasis. The cell death-inducing property of FasL has been associated with its extracellular domain, which can be cleaved off by metalloprotease activity to produce soluble FasL. The fate of the remaining membrane-anchored N-terminal part of the FasL molecule has not been determined. Here we show that post-translational processing of overexpressed and endogenous FasL in T-cells by the disintegrin and metalloprotease ADAM10 generates a 17-kDa N-terminal fragment, which lacks the receptor-binding extracellular domain. This FasL remnant is membrane anchored and further processed by SPPL2a, a member of the signal peptide peptidase-like family of intramembrane-cleaving proteases. SPPL2a cleavage liberates a smaller and highly unstable fragment mainly containing the intracellular FasL domain (FasL ICD). We show that this fragment translocates to the nucleus and is capable of inhibiting gene transcription. With ADAM10 and SPPL2a we have identified two proteases implicated in FasL processing and release of the FasL ICD, which has been shown to be important for retrograde FasL signaling.


Assuntos
Proteínas ADAM/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Proteína Ligante Fas/metabolismo , Linfócitos T/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Proteína Ligante Fas/química , Humanos , Microscopia Confocal , Estrutura Terciária de Proteína , RNA Interferente Pequeno
6.
Cell Death Differ ; 13(4): 576-85, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16179940

RESUMO

The serine protease granzyme B (GrB) of cytotoxic lymphocytes efficiently induces apoptosis by direct activation of caspases and cleavage of central caspase substrates. We employed human GrB as an effector function in chimeric fusion proteins that also contain the EGFR ligand TGFalpha or an ErbB2-specific single-chain antibody fragment (scFv) for selective targeting to tumor cells. GrB-TGFalpha (GrB-T) and GrB-scFv(FRP5) (GrB-5) molecules expressed in the yeast Pichia pastoris were bifunctional, cleaving synthetic and natural GrB substrates, and binding specifically to cells expressing EGFR or ErbB2 target receptors. Upon cell binding the chimeric molecules were internalized into intracellular vesicles, but could be released into the cytosol by the endosomolytic reagent chloroquine. Treatment with picomolar to nanomolar concentrations of GrB-5 and GrB-T resulted in selective and rapid tumor cell killing, accompanied by clear signs of apoptosis such as chromatin condensation, membrane blebbing, formation of apoptotic bodies and activation of endogenous initiator and effector caspases.


Assuntos
Antineoplásicos/administração & dosagem , Apoptose , Sistemas de Liberação de Medicamentos , Proteínas Recombinantes de Fusão/administração & dosagem , Serina Endopeptidases/administração & dosagem , Anticorpos/administração & dosagem , Anticorpos/genética , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cloroquina/farmacologia , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/enzimologia , Relação Dose-Resposta a Droga , Endocitose , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Granzimas , Humanos , Concentração Inibidora 50 , Pichia/genética , Pichia/metabolismo , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Serina Endopeptidases/genética , Fator de Crescimento Transformador alfa/administração & dosagem , Fator de Crescimento Transformador alfa/genética
7.
Br J Cancer ; 92(8): 1421-9, 2005 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15812545

RESUMO

Efficient delivery of tumour-associated antigens to appropriate cellular compartments of antigen-presenting cells is of prime importance for the induction of potent, cell-mediated antitumour immune responses. We have designed novel multivalent liposomal constructs that co-deliver the p63-71 cytotoxic T Lymphocyte epitope derived from human ErbB2 (HER2), and HA307-319, a T-helper (Th) epitope derived from influenza haemagglutinin. Both peptides were conjugated to the surface of liposomes via a Pam3CSS anchor, a synthetic lipopeptide with potent adjuvant activity. In a murine model system, vaccination with these constructs completely protected BALB/c mice from subsequent s.c. challenge with ErbB2-expressing, but not ErbB2-negative, murine renal carcinoma (Renca) cells, indicating the induction of potent, antigen-specific immune responses. I.v. re-challenge of tumour-free animals 2 months after the first tumour cell inoculation did not result in the formation of lung tumour nodules, suggesting that long-lasting, systemic immunity had been induced. While still protecting the majority of vaccinated mice, a liposomal construct lacking the Th epitope was less effective than the diepitope construct, also correlating with a lower number of CD8+ IFN-gamma+ T-cells identified upon ex vivo peptide restimulation of splenocytes from vaccinated animals. Importantly, in a therapeutic setting treatment with the liposomal vaccines resulted in cures in the majority of tumour-bearing mice and delayed tumour growth in the remaining ones. Our results demonstrate that liposomal constructs which combine Tc and Th peptide antigens and lipopeptide adjuvants can induce efficient, antigen-specific antitumour immunity, and represent promising synthetic delivery systems for the design of specific antitumour vaccines.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Epitopos de Linfócito T/imunologia , Neoplasias Renais/tratamento farmacológico , Lipossomos/imunologia , Receptor ErbB-2/imunologia , Animais , Carcinoma de Células Renais/imunologia , Modelos Animais de Doenças , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/imunologia , Humanos , Neoplasias Renais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/imunologia , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
8.
J Hematother Stem Cell Res ; 10(4): 523-34, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11522235

RESUMO

T lymphocytes recognize specific antigens through interaction of the T cell receptor (TCR) with short peptides presented by major histocompatibility complex (MHC) class I or II molecules. For initial activation and clonal expansion, naïve T cells are dependent on professional antigen-presenting cells (APCs) that provide additional co-stimulatory signals. TCR activation in the absence of co-stimulation can result in unresponsiveness and clonal anergy. To bypass immunization, different approaches for the derivation of cytotoxic effector cells with grafted recognition specificity have been developed. Chimeric antigen receptors have been constructed that consist of binding domains derived from natural ligands or antibodies specific for cell-surface antigens, genetically fused to effector molecules such as the TCR alpha and beta chains, or components of the TCR-associated CD3 complex. Upon antigen binding, such chimeric receptors link to endogenous signaling pathways in the effector cell and generate activating signals similar to those initiated by the TCR complex. Since the first reports on chimeric antigen receptors, this concept has steadily been refined and the molecular design of chimeric receptors has been optimized. Aided by advances in recombinant antibody technology, chimeric antigen receptors targeted to a wide variety of antigens on the surface of cancer cells and of cells infected by human immunodeficiency virus (HIV) have been generated. In initial clinical studies, infusion of such cells into patients proved to be safe and transient therapeutic effects have been observed.


Assuntos
Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Engenharia Genética , Humanos , Imunoterapia Adotiva/métodos , Receptores de Antígenos/genética , Proteínas Recombinantes de Fusão/genética
9.
Prostate ; 47(1): 21-8, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11304726

RESUMO

BACKGROUND: HER2/neu has been implicated in the oncogenesis of human prostate cancer. Clinical studies have suggested that overexpression of HER2 may be one of the indicators of poor prognosis in prostate cancer patients. METHODS: We used Western blot analysis to examine the expression of HER2 in a panel of established human prostate cancer cell lines and used an MTT assay to evaluate the cytotoxicity on these cells of a recombinant fusion protein consisting of an HER2-specific single-chain antibody and the Pseudomonas exotoxin A, scFv(FRP5)-ETA. RESULTS: LNCaP cells express high levels of HER2 protein. Exposure of LNCaP cells to scFv(FRP5)-ETA caused remarkable cell death. In contrast, PC3M cells, which express an undetectable level of HER2 protein, were resistant to scFv(FRP5)-ETA-induced cytotoxicity. MDA PCa 2a, MDA PCa 2b, and DU145 cells express low-to-medium levels of HER2 protein and showed an HER2 level-dependent response to scFv(FRP5)-ETA-induced cytotoxicity. The scFv(FRP5)-ETA-induced cytotoxicity of LNCaP cells could be inhibited by an anti-HER2 monoclonal antibody (mAb), which downregulated the levels of HER2 protein, indicating the specificity of scFv(FRP5)-ETA in inducing cytotoxicity in LNCaP cells. Using an apoptosis ELISA, we demonstrated that scFv(FRP5)-ETA induced apoptosis in LNCaP cells. The apoptosis was inhibited by the presence of dihydrotestosterone (DHT) in culture medium. Exposure of LNCaP cells to scFv(FRP5)-ETA caused reduction in the level of the prostate-specific antigen (PSA). CONCLUSIONS: Our data suggest that scFv(FRP5)-ETA might be a useful agent for the treatment of human prostate cancer cells with high levels of HER2 expression.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Exotoxinas/toxicidade , Imunotoxinas/farmacologia , Neoplasias da Próstata , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/toxicidade , Fatores de Virulência , Animais , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Di-Hidrotestosterona/farmacologia , Masculino , Antígeno Prostático Específico/biossíntese , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/metabolismo , Exotoxina A de Pseudomonas aeruginosa
10.
Biotechniques ; 30(1): 190-4, 196, 198 passim, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11196310

RESUMO

Vascular targeting agents (VTAs) can be produced by linking antibodies or antibody fragments directed against endothelial cell markers to effector moieties. So far, it has been necessary to produce the components of VTAs (antibody, antibody fragment, linker, and effector) separately and, subsequently, to conjugate them by biochemical reactions. We devised a cloning and expression system to allow rapid generation of recombinant VTAs from hybridoma cell lines. The VTAs consist of a single chain Fv antibody fragment as a targeting moiety and either truncated Pseudomonas exotoxin (resulting in immunotoxins) or truncated human tissue factor (resulting in coaguligands) as effectors. The system was applied to generate recombinant immunotoxins and coaguligands directed against endoglin, vascular endothelial growth factor (VEGF):VEGF receptor (VEGFR) complex and vascular cell adhesion molecule 1 (VCAM-1). The fusion proteins exhibited similar functional activity to analogous biochemical constructs. This is the first report to describe the generation and characterization of recombinant coaguligands.


Assuntos
Fragmentos de Imunoglobulinas/farmacologia , Imunotoxinas/farmacologia , Neovascularização Patológica/prevenção & controle , Tromboplastina/farmacologia , Animais , Antígenos CD , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Clonagem Molecular , Relação Dose-Resposta a Droga , Endoglina , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Fatores de Crescimento Endotelial/imunologia , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fator Xa/efeitos dos fármacos , Fator Xa/metabolismo , Feminino , Humanos , Hibridomas , Fragmentos de Imunoglobulinas/genética , Imuno-Histoquímica , Imunotoxinas/genética , Linfocinas/imunologia , Linfocinas/metabolismo , Camundongos , Plasmídeos/genética , Ligação Proteica , Receptores Proteína Tirosina Quinases/imunologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Superfície Celular , Receptores de Fatores de Crescimento/imunologia , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Tromboplastina/genética , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
11.
Gynecol Oncol ; 80(2): 145-55, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11161852

RESUMO

OBJECTIVES: The HER-2 proto-oncogene is overexpressed in 15-30% of ovarian cancers, providing a target for therapy in a fraction of patients. We previously described the ability of a recombinant single-chain anti-p185HER-2-Pseudomonas exotoxin A fusion protein, scFv(FRP5)-ETA, to inhibit growth of cancer cells in culture and tumor xenografts in vivo. We have also described synergistic interaction between an anti-p185HER-2-ricin A chain immunotoxin and ionizing radiation for inhibiting growth of ovarian and breast cancer cells that overexpress HER-2. Our objective in this report was to evaluate the in vitro and in vivo effects of scFv(FRP5)-ETA against SKOv3 ovarian cancer cells that have been transfected to express >10(6) p185HER-2 receptors per cell (clone-9002-18). METHODS: Inhibition of clonogenic growth was measured in vitro by limiting dilution analysis. Inhibition of tumor growth was measured in vivo using heterografts established with the same ovarian cancer cell lines. RESULTS: Clone-9002-18 cells were substantially more sensitive than parental SKOv3 cells to the cytotoxic effects of scFv(FRP5)-ETA. Exotoxin fusion protein induced apoptosis in clone-9002-18 cells, but did not affect parental SKOv3 cells. Treatment of clone-9002-18 cells with 200- to 2000-cGy external beam irradiation in combination with scFv(FRP5)-ETA produced synergistic elimination of clonogenic tumor cells in culture. In vivo, subcutaneous or intraperitoneal growth of tumor xenografts in nu/nu mice was significantly inhibited (P = 0.004) by treatment for 10 days with scFv(FRP5)-ETA or with 131I-labeled-520C9 anti-p185HER-2 radionuclide conjugate, but additive effects were not observed with combined treatment. CONCLUSION: ScFv(FRP5)-ETA deserves further evaluation for intraperitoneal therapy of ovarian cancers that overexpress p185HER-2.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Exotoxinas/toxicidade , Imunotoxinas/toxicidade , Radioisótopos do Iodo/toxicidade , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/toxicidade , Fatores de Virulência , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/toxicidade , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Terapia Combinada , Sinergismo Farmacológico , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Inibidores do Crescimento/toxicidade , Humanos , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/toxicidade , Imunotoxinas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/radioterapia , Proto-Oncogene Mas , Receptor ErbB-2/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
12.
Clin Cancer Res ; 6(11): 4314-22, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11106249

RESUMO

Efficient T-cell activation requires two signals. The first signal, which confers specificity, is provided by interaction of the T-cell receptor with peptides presented by MHC molecules. One of the second costimulatory signals is induced by binding of B7 proteins on the surface of antigen-presenting cells to CD28 on the T-cell surface. Expression of B7 molecules on tumor cells can result in the activation of tumor specific T lymphocytes and induce protective antitumor immunity. However, at present such gene-therapeutic approaches are limited by the inability to selectively target B7 gene expression to cancer cells. As an alternative approach we exploited recombinant antibody fragments to localize a costimulatory B7 molecule to the surface of tumor cells. We constructed chimeric proteins that contain in a single polypeptide chain a portion of human B7-2 (CD86) genetically fused to single-chain (sc) Fv antibody domains specific for the tumor-associated antigens epidermal growth factor receptor and the closely related ErbB2 receptor tyrosine kinase. A small recombinant fragment of human CD86 was characterized that corresponds to amino acid residues 1-111 (CD86(111)) of the mature protein. CD86(111) produced in the yeast Pichia pastoris and CD86(111) expressed in bacteria was functionally active and displayed specific binding to B7 counter receptors. Bacterially expressed CD86(111)-scFv fusion proteins specifically localized to the respective target antigens on the surface of tumor cells and markedly enhanced the proliferation of primary T cells when bound to immobilized tumor antigen.


Assuntos
Antígenos CD/uso terapêutico , Fragmentos de Imunoglobulinas/uso terapêutico , Glicoproteínas de Membrana/uso terapêutico , Neoplasias/terapia , Fragmentos de Peptídeos/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Antígenos CD/metabolismo , Antígeno B7-2 , Células CHO , Cricetinae , Humanos , Fragmentos de Imunoglobulinas/metabolismo , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
13.
Adv Drug Deliv Rev ; 44(2-3): 153-66, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11072112

RESUMO

The development of vectors for cell-specific gene delivery is a major goal of gene therapeutic strategies. Significant progress has been made in the construction of non-viral vectors that combine different functions required for gene transfer in an artificial complex. To some extent this can be achieved by complexing plasmid DNA with synthetic compounds such as lipids and polycations. Alternative approaches rely on the activities of natural or recombinant DNA-carrier proteins to achieve uptake and intracellular delivery of plasmid DNA. Nuclear proteins such as histones and members of the high mobility group protein family have been shown to condense DNA and transfect cultured cells. Some structural proteins of DNA viruses spontaneously assemble with plasmid DNA and form transfection-competent pseudocapsids. In addition, chimeric fusion proteins have been engineered that incorporate in a single polypeptide chain heterologous protein domains which facilitate binding to plasmid DNA, specific recognition of target cells, induction of receptor-mediated endocytosis, and DNA transport through intracellular compartments.


Assuntos
Capsídeo/metabolismo , Proteínas de Transporte/metabolismo , DNA/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Toxinas Bacterianas/química , Capsídeo/administração & dosagem , DNA/uso terapêutico , Endocitose , Endossomos/metabolismo , Humanos , Transfecção
14.
J Mol Biol ; 301(4): 893-904, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-10966794

RESUMO

Delayed infectivity panning (DIP) is a novel approach for the in vivo isolation of interacting protein pairs. DIP combines phage display and cell surface display of polypeptides as follows: an antigen is displayed in many copies on the surface of F(+) Escherichia coli cells by fusing it to a Lpp-OmpA' hybrid. To prevent premature, non-specific infection by phage, the cells are rendered functionally F(-) by growth at 16 degrees C. The antigen-displaying cells are used to capture antibody-displaying phage by virtue of the antibody-antigen interaction. Following removal of unbound phage, infection of the cells by bound phage is initiated by raising the temperature to 37 degrees C that facilitates F pilus expression. The phage then dissociate from the antigen and infect the bacteria through the F pilus. Using specific scFv antibodies and the human ErbB2 proto-oncogene and IL2-Ralpha chain as model antibody-antigen pairs, we demonstrate enrichment of those phage that display a specific antibody over phage that display an irrelevant antibody of over 1,000,000 in a single DIP cycle. We further show the successful isolation of anti-toxin, anti-receptor, anti-enzyme and anti-peptide antibodies from several immune phage libraries, a shuffled library and a large synthetic human library. The effectiveness of DIP makes it suitable for the isolation of rare clones present in large libraries. Since DIP can be applied for most of the phage libraries already existing, it could be a powerful tool for the rapid isolation and characterization of binders in numerous protein-protein interactions.


Assuntos
Especificidade de Anticorpos/imunologia , Antígenos/imunologia , Clonagem Molecular/métodos , Proteínas de Escherichia coli , Escherichia coli/genética , Lipoproteínas , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/imunologia , Anticorpos/genética , Anticorpos/imunologia , Reações Antígeno-Anticorpo/imunologia , Antígenos/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Bacteriófagos/genética , Bacteriófagos/imunologia , Bacteriófagos/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Escherichia coli/imunologia , Escherichia coli/fisiologia , Escherichia coli/virologia , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/fisiologia , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Ligação Proteica , Proto-Oncogene Mas , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/imunologia , Proteínas Recombinantes de Fusão/genética , Sensibilidade e Especificidade , Temperatura
15.
Int J Cancer ; 86(2): 269-75, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10738256

RESUMO

Overexpression of the ErbB2 and epidermal growth factor receptor (EGFR) tyrosine kinases is frequently observed in squamous cell carcinomas of the head and neck, and has been correlated with shorter overall survival. By immunoblot analysis, we have found EGFR and ErbB2 expression in 6 out of 6 established head and neck cancer cell lines. Elevated EGFR protein levels were noted in 3 and elevated ErbB2 levels in 5 of them. Significant expression of EGFR and ErbB2 was also detected in 17 of 47 and 26 of 45 primary tumor samples. Due to their enhanced expression on the tumor cell surface, these receptors can be regarded as suitable targets for directed cancer therapy. We have analyzed the antitumoral activity of recombinant single-chain antibody toxins specific for ErbB2 and EGFR against head and neck cancer cells in vitro and in vivo. The recombinant toxins consist of the variable domains of the heavy and light chains of monoclonal antibodies (MAbs) genetically fused to a truncated Pseudomonas exotoxin A (ETA). At low concentrations, the ErbB2-specific single-chain antibody (scFv) toxin scFv(FRP5)-ETA and the EGFR-specific toxins scFv(225)-ETA and scFv(14E1)-ETA inhibited the in vitro growth of established head and neck cancer cell lines and primary tumor cells. In a nude mouse tumor model, intratumoral injection of the antibody toxins resulted in the rapid regression of subcutaneously growing CAL 27 tumor xenografts, with scFv(FRP5)-ETA and scFv(14E1)-ETA treatment being most effective and leading to the cure of up to 50% of the animals. Our results suggest that EGFR and ErbB2-specific antibody toxins may become valuable therapeutic reagents for the treatment of squamous cell carcinomas of the head and neck.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Carcinoma de Células Escamosas/patologia , Receptores ErbB/imunologia , Exotoxinas/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Imunotoxinas/farmacologia , Receptor ErbB-2/imunologia , Fatores de Virulência , Animais , Anticorpos Monoclonais , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/tratamento farmacológico , Divisão Celular , Receptores ErbB/análise , Exotoxinas/administração & dosagem , Exotoxinas/uso terapêutico , Neoplasias de Cabeça e Pescoço/química , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Imunotoxinas/imunologia , Imunotoxinas/uso terapêutico , Camundongos , Camundongos Nus , Transplante de Neoplasias , Receptor ErbB-2/análise , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Células Tumorais Cultivadas , Exotoxina A de Pseudomonas aeruginosa
16.
J Virol Methods ; 81(1-2): 71-5, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10488763

RESUMO

Retroviral vectors are used widely as gene transfer vehicles. Vector particles are generated by packaging cell lines, which supply the structural proteins gag, pol and env needed to package the retroviral vector RNA. The most efficient way to introduce the vector genome into the packaging cell line is cross-infection with a retroviral vector. Since the infection of a packaging cell line by the produced virus is blocked due to the down regulation of the retrovirus receptor by the envelope glycoprotein, the vector genome should be introduced by a virus with a host tropism different from the one of the packaging cell line. The murine ecotropic retrovirus receptor was expressed in the human amphotropic packaging cell line FLYA13 to generate a cell line which can be infected by murine ecotropic retroviruses. Vector transfer can now be facilitated by cross-infection with the appropriate ecotropic retroviral vectors and provides a simple and efficient method for the generation of amphotropic packaging lines.


Assuntos
DNA Recombinante/genética , Vetores Genéticos/genética , Vírus da Leucemia Murina/genética , Glicoproteínas de Membrana/genética , Receptores Virais/genética , Retroviridae/genética , Vírion/genética , Montagem de Vírus/genética , Células 3T3 , Animais , Técnicas de Transferência de Genes , Humanos , Glicoproteínas de Membrana/biossíntese , Camundongos , Receptores Virais/biossíntese , Retroviridae/fisiologia , Células Tumorais Cultivadas
17.
Hybridoma ; 18(1): 69-75, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10211791

RESUMO

The ErbB2 receptor tyrosine kinase is often overexpressed in human malignancies and causally involved in transformation. High levels of ErbB2 in tumor cells correlate with an unfavorable prognosis. This makes the ErbB2 receptor an interesting target for tumor therapy, and several strategies have been designed to direct drugs to ErbB2-expressing cells. We established a novel cellular model that allows preclinical evaluation of ErbB2-directed drugs in immunocompetent animals. Renal carcinoma (Renca) cells are an established tumor cell line that originated in Balb/c mice. Upon intravenous transplantation, these cells form pulmonary metastases in Balb/c mice. The transforming genetic lesions in these cells are not fully characterized, but do not seem to involve alterations in ErbB2 gene expression. We transfected Renca cells with the gene encoding the human ErbB2 receptor to provide a target structure for specific drugs and with the bacterial lacZ gene to provide a sensitive means of detection of the tumor cells in the transplanted animals. These genetically modified cells form lung metastasis and can be easily visualized on the surface of lung tissue by staining with an X-gal solution. This allows a quantitative analysis of the number of ErbB2-expressing pulmonary metastasis. We previously used these Renca cells to evaluate the efficacy of an ErbB2-specific tumor toxin on pulmonary metastases in an adjuvant and a palliative treatment setting. In both cases, we achieved a dramatic reduction of disseminated lung lesions. Here we show that even at an advanced stage of metastasis formation, the ErbB2-specific toxin is able to efficiently reduce the number of pulmonary tumors.


Assuntos
Modelos Animais de Doenças , Receptores ErbB/metabolismo , Imunotoxinas/uso terapêutico , Neoplasias Pulmonares/secundário , Metástase Neoplásica/prevenção & controle , Receptor ErbB-2/antagonistas & inibidores , Animais , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/terapia , Receptores ErbB/imunologia , Estudos de Avaliação como Assunto , Humanos , Imunocompetência , Imuno-Histoquímica , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Neoplasias Renais/terapia , Óperon Lac , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Receptor ErbB-2/genética , Transfecção , Células Tumorais Cultivadas
18.
Oncogene ; 18(9): 1711-21, 1999 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-10208432

RESUMO

Cytotoxic strategies which are directed to tumor-associated antigens might be most beneficial for cancer patients with minimal tumor load such as in an adjuvant setting after initial therapy. We have recently described a highly potent single chain antibody-toxin, scFv(14E1)-ETA, which consists of the variable domains of the antibody 14E1 genetically fused to a truncated form of Pseudomonas exotoxin A. ScFv(14E1)-ETA specifically recognizes the human epidermal growth factor receptor (EGFR) and the oncogenically activated receptor variant EGFRvIII, which have been implicated in the development of various human malignancies. Here we have investigated the antimetastatic activity of bacterially expressed scFv(14E1)-ETA and its disulfide-stabilized derivative ds-scFv(14E1)-ETA in a novel model for disseminated disease which is based on murine renal carcinoma cells subsequently transfected with the E. coli beta-galactosidase gene, and human full-length or variant EGFR cDNAs. Intravenous injection of these Renca-lacZ/EGFR and Renca-lacZ/EGFRvIII cells in syngenic Balb/c mice led to the formation of pulmonary metastases which were readily detectable upon excision of the lungs and X-gal staining. Systemic treatment of mice with scFv(14E1)-ETA resulted in the complete suppression of Renca-lacZ/EGFRvIII metastasis formation and drastically reduced the number of pulmonary Renca-lacZ/EGFR tumor nodules. The ds-scFv(14E1)-ETA derivative where the antibody variable regions are connected by an artificial disulfide bond displayed improved thermal stability at physiological temperature but due to reduced cytotoxic activity was less potent than the original scFv(14E1)-ETA in metastasis suppression.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Receptores ErbB/metabolismo , Exotoxinas/metabolismo , Fragmentos de Imunoglobulinas/metabolismo , Imunotoxinas/metabolismo , Fatores de Virulência , Animais , Receptores ErbB/genética , Receptores ErbB/imunologia , Escherichia coli , Exotoxinas/genética , Feminino , Expressão Gênica , Humanos , Fragmentos de Imunoglobulinas/genética , Imunotoxinas/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Testes de Neutralização , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas , Exotoxina A de Pseudomonas aeruginosa
19.
Int J Mol Med ; 1(1): 249-56, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9852227

RESUMO

The data of a closed phase I/II trial in patients with resistant Hodgkin's lymphoma indicate promising results using a chemically linked anti-CD25 ricin-A immunotoxin (IT) (RFT5-SMPT-dgA). This IT is based on the high-affinity moab RFT5. Since recombinant DNA technology permits the readier production of large amounts of ITs, we constructed a new RFT5-based fusion toxin [RFT5(scFv)-ETA']. We isolated mRNA from the hybridoma cell line RFT5, synthesized first strand cDNA and performed RT-PCR. Amplified coding regions of the light and heavy chain variable domains were joined together with a synthetic (Gly4-Ser)3 linker. The resulting single chain variable fragment (scFv) was fused to a modified Pseudomonas aeruginosa exotoxin A (ETA') lacking its cell-binding domain I. After IPTG-induced expression in Escherichia coli, the 70 kDa His-tagged fusion protein [RFT5(scFv)-ETA'] was isolated by osmotic shock and sonication under denaturing conditions. The recombinant toxin was purified on a Ni2+-NTA chelating sepharose and eluted with 250 mM imidazole. Pooled protein was renatured, dialyzed and concentrated by precipitation. Binding properties of RFT5(scFv)-ETA' were assessed on the CD25-expressing cell line L540cy by ELISA, immunohistochemistry and FACS analysis. CD25-specific binding was confirmed by immunoprecipitation experiments with recombinant human IL-2 receptor alpha. The in vitro toxicity of the chimeric protein was tested on the Hodgkin-derived cell lines L540cy, L428, L1236, a monocyte cell line U937 and a Burkitt lymphoma cell line BL38. RFT5(scFv)-ETA' inhibited protein biosynthesis of L540cy and L428 cells by 50% at concentrations (IC50) of 18 and 12 ng/ml, respectively. CD25-specific toxicity was confirmed by competitive toxicity assays. These data confirm for the first time binding specificity and toxicity of a recombinant anti-CD25 immunotoxin, against Hodgkin-derived cell lines; its applicability on Hodgkin's lymphoma needs yet to be evaluated in vivo.


Assuntos
ADP Ribose Transferases , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/farmacocinética , Toxinas Bacterianas , Exotoxinas/farmacologia , Região Variável de Imunoglobulina/farmacologia , Imunotoxinas/farmacologia , Fatores de Virulência , Sequência de Aminoácidos , Anticorpos Monoclonais/genética , Sequência de Bases , Estudos de Avaliação como Assunto , Exotoxinas/genética , Expressão Gênica , Humanos , Região Variável de Imunoglobulina/genética , Imunotoxinas/genética , Dados de Sequência Molecular , Anticorpos de Cadeia Única , Células Tumorais Cultivadas , Células U937 , Exotoxina A de Pseudomonas aeruginosa
20.
Cancer Immunol Immunother ; 47(3): 167-75, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9829842

RESUMO

Tumor necrosis factor (TNF)-alpha has a broad range of biological activities, which depend heavily on cell type and physiological condition. In a panel of human tumor cell lines we analyzed expression of the receptor tyrosine kinases EGFR, ErbB2 and ErbB3, and the response to TNF-alpha. Among the cell lines tested those resistant to TNF-alpha were found to express high levels of either EGFR, or ErbB2 and ErbB3. In TNF-sensitive breast and cervical carcinoma cells activation of EGFR or ErbB2 by the exogenous growth factors EGF and heregulin beta1 resulted in a significant increase in the number of cells surviving TNF-alpha treatment. In contrast, inhibition of EGFR activation in TNF-resistant breast carcinoma cells by the novel antagonistic anti-EGFR antibody 14E1 sensitized the cells to the cytotoxic effects of TNF-alpha. A bacterially expressed fusion protein consisting of a 14E1 single-chain (sc) Fv antibody fragment linked to human TNF-alpha retained TNF-alpha activity. This scFv(14E1)-TNF-alpha molecule localized specifically to EGFR on the surface of tumor cells and activated the NF-kappaB pathway in co-cultured T cells, as demonstrated by electrophoretic mobility shift assays.


Assuntos
Receptores ErbB/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor ErbB-2/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Animais , Neoplasias da Mama , Carcinoma , Técnicas de Cocultura , Resistência a Medicamentos , Feminino , Humanos , Imunoconjugados/metabolismo , Fragmentos de Imunoglobulinas/metabolismo , Camundongos , NF-kappa B/metabolismo , Ligação Proteica , Receptor ErbB-3 , Linfócitos T/citologia , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...