Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cytotherapy ; 11(2): 206-17, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19148842

RESUMO

BACKGROUND: Adoptive immunotherapy with cytotoxic T cells has shown promising clinical results in patients with metastatic melanoma and post-transplant-associated viral infections. Cell transfer therapies often require the ex vivo expansion of large numbers of reactive lymphocytes. Therefore interleukin-2 (IL-2), a potent T-cell mitogenic cytokine that critically affects the features and effectiveness of T cells, is frequently added to cell culture media. METHODS: We examined the influence of various IL-2 concentrations on cell growth, cytotoxicity, cytokine release and surface marker expression of tumor-infiltrating lymphocytes (TIL) during a standard 14-day rapid expansion phase. The study was conducted under good manufacturing practice (GMP) conditions, using approved reagents in a class 10000 laboratory. RESULTS: T-cell cultures grown in very high IL-2 concentrations (600-6000 IU/mL) expanded massively and maximally secreted interferon (IFN)-gamma in response to antigenic stimulation, but exhibited only low direct cytotoxicity. On the other hand, TIL cultures grown in low concentrations of IL-2 throughout the rapid expansion phase expanded to a lower extent and barely secreted IFN-gamma but displayed high cytotoxic activity. A combined approach of starting with 10-120 IU/mL IL-2 during the first week, followed by increasing the IL-2 concentration to 6000 IU/mL during the second week, results in T cells that expand well, maximally produce IFN-gamma and are highly cytotoxic against tumor cells. DISCUSSION: Fine tuning of the IL-2 concentration during ex vivo expansion of T cells can yield high numbers of T cells with optimal features for clinical use.


Assuntos
Imunoterapia Adotiva , Interleucina-2/metabolismo , Linfócitos do Interstício Tumoral/patologia , Melanoma/patologia , Linfócitos T Citotóxicos/patologia , Antígenos de Diferenciação/biossíntese , Antígenos de Neoplasias/metabolismo , Proliferação de Células , Técnicas de Cocultura , Meios de Cultura , Citotoxicidade Imunológica , Humanos , Interferon gama/metabolismo , Interleucina-2/imunologia , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Melanoma/imunologia , Melanoma/metabolismo , Metástase Neoplásica , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
2.
Oncogene ; 26(25): 3714-33, 2007 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-17530025

RESUMO

In recent years antibodies, whether generated by traditional hybridoma technology or by recombinant DNA strategies, have evolved from Paul Ehrlich's 'magic bullets' to a modern age 'guided missile'. In the recent years of immunologic research, we are witnessing development in the fields of antigen screening and protein engineering in order to create specific anticancer remedies. The developments in the field of recombinant DNA, protein engineering and cancer biology have let us gain insight into many cancer-related mechanisms. Moreover, novel techniques have facilitated tools allowing unique distinction between malignantly transformed cells, and regular ones. This understanding has paved the way for the rational design of a new age of pharmaceuticals: monoclonal antibodies and their fragments. Antibodies can select antigens on both a specific and a high-affinity account, and further implementation of these qualities is used to target cancer cells by specifically identifying exogenous antigens of cancer cell populations. The structure of the antibody provides plasticity resonating from its functional sites. This review will screen some of the many novel antibodies and antibody-based approaches that are being currently developed for clinical applications as the new generation of anticancer agents.


Assuntos
Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/uso terapêutico , Imunoterapia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Animais , Antígenos de Neoplasias/imunologia , Citocinas/imunologia , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Receptores de Antígenos de Linfócitos T/imunologia
3.
Int J Cancer ; 94(6): 864-72, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11745490

RESUMO

The MDR1 gene product P-glycoprotein (Pgp) plays a key role in multidrug resistance of cancer cells. Pgp is an ATP-driven efflux pump that extrudes a variety of dissimilar hydrophobic cytotoxic compounds. P-glycoprotein overexpression results in multidrug resistance (MDR) of tumor cell lines in vitro as well as in cancer patients. To selectively target and eliminate MDR tumor cells, we have isolated a monoclonal antibody that specifically reacts with the first extracellular loop of the human Pgp. We have cloned the variable domain genes of this antibody and assembled a functional single-chain Fv fragment capable of specifically targeting various Pgp-expressing MDR carcinoma cells lines. Targeting and specific elimination of Pgp-dependent MDR human cancer cells was achieved by constructing a single-chain immunotoxin in which the scFv fragment was fused to a truncated form of Pseudomonas exotoxin (PE38). We conclude that recombinant Fv-immunotoxins or other Fv-based molecules armed with potent cytotoxins represent an effective tool in targeted cancer therapy aimed at specific elimination of MDR tumor cell sub-populations. Recombinant antibody fragments targeting MDR proteins such as Pgp may be also used for intracellular expression and consequent phenotypic knockout of MDR.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Imunotoxinas/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Clonagem Molecular , Cricetinae , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes/farmacologia
4.
Int J Cancer ; 93(6): 848-54, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11519047

RESUMO

Resistance of tumor cells to lysis by complement is generally attributed to several protective mechanisms. The relative impact of these mechanisms in the same tumor cell, however, has not been assessed yet. We have analyzed the interaction of the human erythroleukemia tumor cell line K562 with human complement. K562 cells express the membrane complement regulatory proteins CD59, CD55 and CD46. As shown here for the first time, K562 also spontaneously release the soluble regulators C1 inhibitor, factor H, and soluble CD59. Complement resistance of K562 cells is augmented upon treatment with PMA, TNF or even with sublytic complement. Unlike TNF and sublytic complement, PMA enhanced the expression of membrane-bound CD55 and CD59 and led to increased secretion of soluble CD59. In addition, we show that complement-resistant K562 cells express a membrane-associated proteolytic activity, higher than the complement-sensitive K562/S cells. Treatment of complement-resistant K562 cells with serine protease inhibitors enhance their sensitivity to complement-mediated lysis. Inhibitors of protein kinase C (PKC) also sensitize K562 cells to complement lysis, implicating PKC-mediated signaling in cell resistance to complement. Neutralization of the CD55 and CD59 but not of CD46 regulatory activity with specific antibodies significantly increases complement-mediated K562 cell lysis. Treatment of K562 cells with a mixture of inhibitory reagents results in a significant additive enhancing effect on complement-mediated lysis of K562. In conclusion, K562 cells resist a complement attack by concomitantly using multiple molecular evasion strategies. Future attempts in antibody-based tumor therapy should include strategies to interfere with those resistance mechanisms.


Assuntos
Proteínas do Sistema Complemento/imunologia , Leucemia Eritroblástica Aguda/imunologia , Antígenos CD/imunologia , Antígenos CD55/imunologia , Antígenos CD59/imunologia , Membrana Celular/metabolismo , Ativação do Complemento , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Eritrócitos/metabolismo , Citometria de Fluxo , Humanos , Células K562 , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/imunologia , Naftalenos/farmacologia , Neutrófilos/metabolismo , Proteínas de Plantas/farmacologia , Proteína Quinase C/metabolismo , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia , Inibidores da Tripsina , Fator de Necrose Tumoral alfa/farmacologia , alfa-Amilases/antagonistas & inibidores
5.
Curr Pharm Biotechnol ; 2(1): 19-46, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11482347

RESUMO

Recombinant Fv-immunotoxins are a new class of biologic anticancer agents composed of a recombinant antibody fragment linked to a very potent bacterial toxin. These potent molecules are designed to specifically bind and kill cancer cells that express a specific target antigen on their cell surface. Recombinant Fv-immunotoxins are an excellent example for the concept of rational drug design. They combine the progress in understanding cancer biology, -the recent knowledge on the mechanisms of malignant transformation and the special properties of cancer cells, -with the enormous developments in recombinant DNA technology and antibody engineering. Recombinant Fv immunotoxins were developed for solid tumors and hematological malignancies and have been characterized intensively for their biological activity in vitro and in vivo in animal models. The excellent in vitro and in vivo activities of recombinant Fv-immunotoxins have lead to their pre-clinical development and to the initiation of clinical trial protocols. Recent trials have demonstrated potent clinical efficacy in patients with malignant diseases that are refractory to traditional modalities of cancer treatment. It is thus suggested that this strategy can be developed into a separate modality of cancer treatment with the basic rationale of specifically targeting cancer cells on the basis of their unique surface markers combined with potent effective biological toxic agents that directly kill the cancer cell. Efforts are now being made to improve the current molecules and to develop new agents with better clinical efficacy. In this review, we will describe the rationale in designing Fv-immunotoxins and will review current progress made in using these agents for cancer treatment.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Fragmentos de Imunoglobulinas/administração & dosagem , Região Variável de Imunoglobulina/administração & dosagem , Imunotoxinas/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes/administração & dosagem , Animais , Humanos , Fragmentos de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Imunotoxinas/genética , Proteínas Recombinantes/genética
6.
Adv Cancer Res ; 81: 93-124, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11430597

RESUMO

Targeted cancer therapy in general and immunotherapy in particular combines rational drug design with the progress in understanding cancer biology. This approach takes advantage of our recent knowledge of the mechanisms by which normal cells are transformed into cancer cells, thus using the special properties of cancer cells to device novel therapeutic strategies. Recombinant immunotoxins are excellent examples of such processes, combining the knowledge of antigen expression by cancer cells with the enormous developments in recombinant DNA technology and antibody engineering. Recombinant immunotoxins are composed of a very potent protein toxin fused to a targeting moiety such as a recombinant antibody fragment or growth factor. These molecules bind to surface antigens specific for cancer cells and kill the target cells by catalytic inhibition of protein synthesis. Recombinant immunotoxins are developed for solid tumors and hematological malignancies and have been characterized intensively for their biological activity in vitro on cultured tumor cell lines as well as in vivo in animal models of human tumor xenografts. The excellent in vitro and in vivo activities of recombinant immunotoxins have lead to their preclinical development and to the initiation of clinical trail protocols. Recent trail results have demonstrated potent clinical efficacy in patients with malignant diseases that are refractory to traditional modalities of cancer treatment: surgery, radiation therapy, and chemotherapy. The results demonstrate that such strategies can be developed into a separate modality of cancer treatment with the basic rationale of specifically targeting cancer cells on the basis of their unique surface markers. Efforts are now being made to improve the current molecules and to develop new agents with better clinical efficacy. This can be achieved by development of novel targeting moieties with improved specificity that will reduce toxicity to normal tissues. In this review, the design, construction, characterization, and applications of recombinant immunotoxins are described. Results of recent clinical trails are presented, and future directions for development of recombinant immunotoxins as a new modality for cancer treatment are discussed.


Assuntos
Imunoterapia/métodos , Imunotoxinas/uso terapêutico , Neoplasias/terapia , Proteínas Recombinantes/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Imunotoxinas/toxicidade , Pró-Fármacos/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes/toxicidade
7.
J Immunol ; 167(1): 270-6, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11418659

RESUMO

There are conflicting opinions about the role that the T cell coreceptors CD4 and CD8 play in TCR binding and activation. Recent evidence from transgenic mouse models suggests that CD8 plays a critical role in TCR binding and activation by peptide-MHC complex multimers (tetramers). Here we show with a human CTL clone specific for a tumor-associated MHC-peptide complex that the binding of tetramers to the TCR on these cells is completely blocked by anti-human CD8 Abs. Moreover, the staining of CTLs with specific MHC-peptide tetramers simultaneously with anti-CD8 Abs was completely blocked with three different anti-CD8 Abs. This blockage was mediated by anti-CD8 Abs but not anti-CD3 Abs and was dose dependent. The blocking effect of the anti-CD8 Abs was attributable to directly inhibiting tetramer binding and was not attributable to Ab-mediated TCR-CD8 internalization and down-regulation. Our results have important implications in TCR binding to MHC-peptide tetramers. MHC-peptide tetramers are widely used today in combination with anti-CD8 Abs for the phenotypic analysis of T cell populations and in the study of T cell responses under various pathological conditions such as infectious diseases and cancer. Our results indicate that also in the human system CD8 plays a critical role in the interaction of MHC-peptide multimers with TCR.


Assuntos
Anticorpos Bloqueadores/farmacologia , Antígenos de Neoplasias/metabolismo , Sítios de Ligação de Anticorpos/imunologia , Antígenos CD8/imunologia , Antígeno HLA-A2/metabolismo , Oligopeptídeos/metabolismo , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Microglobulina beta-2/metabolismo , Anticorpos Bloqueadores/metabolismo , Ligação Competitiva/imunologia , Antígenos CD8/fisiologia , Linhagem Celular , Células Clonais , Regulação para Baixo/imunologia , Meia-Vida , Humanos , Oligopeptídeos/imunologia , Ligação Proteica/imunologia , Receptores de Antígenos de Linfócitos T/biossíntese , Receptores de Antígenos de Linfócitos T/metabolismo , Coloração e Rotulagem , Estereoisomerismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas , Microglobulina beta-2/antagonistas & inibidores
8.
Eur J Immunol ; 30(12): 3522-32, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11093172

RESUMO

Soluble recombinant MHC-peptide complexes are valuable tools for molecular characterization of immune responses as well as for other functional and structural studies. In this study, soluble recombinant single-chain human MHC (scMHC)-peptide complexes were generated by in vitro refolding of inclusion bodies from bacterially expressed engineered HLA-A2 in the presence of tumor-associated or viral peptides. The scMHC molecule was composed of beta2-microglobulin connected to the first three domains of the HLA-A2 heavy chain through a 15-amino acid flexible linker. Highly purified scMHC-peptide complexes were obtained in high yield using several peptides derived from the melanoma antigens gp100 and MART-1 or a viral peptide derived from HTLV-1. The scMHC complexes were characterized in detail and were found to be correctly folded and able to specifically bind HLA-A2-restricted peptides. We also generated scMHC-peptide tetramers, which were biologically functional; they induced a peptide-specific CTL clone to be activated and secrete IFN-gamma, and were able to stain specifically CTL lines. Such recombinant soluble scMHC-peptide complexes and tetramers should prove of great value for characterization of immune responses involving CTL, for visualization of antigen-specific immune responses, for in vitro primary CTL induction, and for peptide binding assays and structural studies.


Assuntos
Antígenos de Neoplasias/imunologia , Antígeno HLA-A2/química , Dobramento de Proteína , Linfócitos T Citotóxicos/imunologia , Antígenos de Neoplasias/química , Escherichia coli/genética , Produtos do Gene tax/química , Produtos do Gene tax/imunologia , Antígeno HLA-A2/imunologia , Humanos , Espectrometria de Massas , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação
9.
J Mol Biol ; 290(3): 685-98, 1999 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-10395823

RESUMO

To develop very small antibody-derived recognition units for experimental, medical, and drug design purposes, a heavy chain variable region (VH) single-domain phage-display library was designed and constructed. The scaffold that was used for library construction was a native sequence of a monoclonal antibody with a unique VH/VL interface. There was no need to modify any residues in the VL interface to avoid non-specific binding of VH domain. The library repertoire, consisting of 4x10(8)independent clones, was generated by the randomization of nine amino acid residues in complementary determining region 3. The library was screened by binding to protein antigens, and individual clones were isolated. The VH genes encoding for specific binding clones were rescued and large amounts of soluble and stable single-domain VH protein were made from insoluble inclusion bodies by in vitro refolding and purification. Biochemical and biophysical characterization of the VH protein revealed a highly specific, correctly folded, and stable monomeric molecule. Binding studies demonstrated an affinity of 20 nM. The properties of these molecules make them attractive for clinical, industrial, and research applications, as well as a step toward improvement in the design of small molecules that are based on the hypervariable loops of antibodies.


Assuntos
Bacteriófagos/genética , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , DNA Recombinante , Cadeias Pesadas de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/metabolismo , Dados de Sequência Molecular
10.
Trends Biotechnol ; 16(12): 513-20, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9881483

RESUMO

Recombinant immunotoxins are new agents being developed for cancer therapy. They are composed of Fv fragments of antibodies that bind to cancer cells fused to a truncated form of a very potent bacterial toxin. The antibody moiety directs the toxin to cancer cells, which are killed, while normal cells are not recognized and thus survive. The excellent preclinical results in vitro and in vivo have led to the initiation of several clinical trials.


Assuntos
Região Variável de Imunoglobulina/genética , Imunotoxinas/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/terapia , Animais , Humanos , Imunotoxinas/genética , Leucemia/terapia , Linfoma/terapia , Pró-Fármacos/administração & dosagem , Radioimunoterapia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
11.
Proc Natl Acad Sci U S A ; 94(9): 4631-6, 1997 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-9114042

RESUMO

Specificity in the immune system is dictated and regulated by specific recognition of peptide/major histocompatibility complex (MHC) complexes by the T cell receptor. Such peptide/MHC complexes are a desirable target for novel approaches in immunotherapy because of their highly restricted fine specificity. Recently, phage display was used to isolate an antibody that has T cell receptor-like specificity. It recognizes mouse MHC class I H-2Kk molecules complexed with a H-2Kk-restricted influenza virus-derived hemagglutinin peptide (Ha255-262) but does not bind to class I H-2Kk alone, peptide alone, or H-2Kk complexed with other peptides. We have used this antibody to make a recombinant antibody-toxin fusion protein (immunotoxin) and show herein that it specifically kills antigen-presenting cells in a peptide-dependent manner and with T cell receptor-like specificity. We find a striking correlation between the fine specificity of binding of the antibody and the cytotoxic activity of the recombinant immunotoxin. We also show specific killing of influenza virus-infected target cells. The results suggest that it should be possible to develop novel immunotherapeutic strategies against human cancer by making recombinant antibodies that will recognize cancer-related peptides complexed with MHC class I molecules on the surface of cancer cells and using these to deliver toxins, radioisotopes, or cytotoxic drugs to the cancer cells.


Assuntos
ADP Ribose Transferases , Anticorpos/farmacologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Toxinas Bacterianas/farmacologia , Exotoxinas/farmacologia , Antígenos de Histocompatibilidade Classe I , Imunotoxinas/farmacologia , Poli(ADP-Ribose) Polimerases/farmacologia , Fatores de Virulência , Animais , Anticorpos/metabolismo , Toxinas Bacterianas/metabolismo , Citotoxicidade Imunológica , Exotoxinas/metabolismo , Antígenos H-2 , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Imunoterapia/métodos , Imunotoxinas/metabolismo , Vírus da Influenza A/efeitos dos fármacos , Camundongos , Peptídeos/imunologia , Poli(ADP-Ribose) Polimerases/metabolismo , Ligação Proteica , Pseudomonas aeruginosa , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sensibilidade e Especificidade , Exotoxina A de Pseudomonas aeruginosa
12.
Nat Biotechnol ; 14(10): 1239-45, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9631086

RESUMO

Disulfide-stabilized Fv fragments of antibodies (dsFv) are molecules in which the VH-VL heterodimer is stabilized by an interchain disulfide bond engineered between structurally conserved framework positions distant from complementarity-determining regions (CDRs). This method of stabilization is applicable for the stabilization of many antibody Fvs and has also been applied to a T-cell receptor Fv. A summary of the design strategy, and the construction and production of various dsFvs and dsFv-fusion proteins is presented. Included in the discussion are the biochemical features of dsFvs in comparison with scFvs, the effect of disulfide stabilization on Fv binding and activity, and various applications of dsFvs and dsFv-immunotoxins for tumor imaging and the treatment of solid tumors in animal models.


Assuntos
Fragmentos de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/terapia , Animais , Bacteriófagos/genética , Bacteriófagos/imunologia , Biotecnologia , Dissulfetos/química , Desenho de Fármacos , Estabilidade de Medicamentos , Humanos , Fragmentos de Imunoglobulinas/química , Região Variável de Imunoglobulina/química , Testes Imunológicos , Imunoterapia , Imunotoxinas/química , Imunotoxinas/uso terapêutico , Modelos Moleculares , Pró-Fármacos/uso terapêutico , Conformação Proteica , Engenharia de Proteínas , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico
13.
Int J Cancer ; 67(1): 113-23, 1996 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-8690511

RESUMO

Monoclonal antibody (MAb) 55.1 specifically recognizes an antigen on the surface of human colon adenocarcinoma cells. We constructed recombinant immunotoxins composed of the heavy- and light-chain variable regions of MAb 55.1 fused to a recombinant form of Pseudomonas exotoxin (PE). The heavy- and light-chain variable regions are stabilized by 2 means. One is by a flexible peptide linker to form a single-chain antigen binding protein (scFv) and the second by an interchain disulfide bond engineered between structurally conserved framework regions. These are termed disulfide stabilized Fvs (dsFv). The 2 Fv forms are fused to truncated forms of PE lacking the cell binding domain. The recombinant scFv- and dsFv-immunotoxins were expressed in E. coli and purified to near homogeneity. The scFv- and dsFv-immunotoxins were shown to be specifically cytotoxic to human colon adenocarcinoma cell lines. The scFv-immunotoxin containing PE38KDEL was more active than the immunotoxin containing PE38 with the native carboxyl terminus (REDLK). However, the PE38KDEL immunotoxin is about 2-fold more toxic in mice, and therefore it does not appreciably increase the therapeutic window in mice. Intravenous administration of the scFv- and dsFv- recombinant immunotoxins caused complete regression of a human colon carcinoma (Colo205) growing subcutaneously in immunodeficient mice. The dsFv-immunotoxin has better antitumor activity compared with its scFv-immunotoxin counterpart.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Neoplasias do Colo/terapia , Exotoxinas/uso terapêutico , Fragmentos de Imunoglobulinas/uso terapêutico , Imunotoxinas/uso terapêutico , Fatores de Virulência , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/uso terapêutico , Sequência de Bases , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Proteínas Recombinantes/uso terapêutico , Transplante Heterólogo , Exotoxina A de Pseudomonas aeruginosa
14.
Clin Cancer Res ; 2(2): 245-52, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9816166

RESUMO

Recombinant immunotoxins are chimeric proteins in which a truncated toxin is fused to a recombinant antigen-binding domain such as a recombinant Fv or Fab. Recombinant immunotoxins target cell surface receptors and other antigens on tumor cells. The antigen-binding and -targeting domains in recombinant immunotoxins are usually single-chain Fvs (scFv), which are the antibody variable regions connected by a flexible peptide linker and fused directly to a bacterial toxin. However, Fabs have also been used. Recombinant immunotoxins have very good activity in vitro on cultured human tumor cell lines and have produced complete regressions and cures of established tumor xenografts in nude mouse models. Problems with the stability and binding of some scFv immunotoxins as well as scFvs not linked to toxin led to the development of a new type of recombinant Fv immunotoxin in which the targeting variable domains of the Fv are stabilized by an interchain disulfide bond located in structurally conserved framework positions of the VH and VL domains. These are termed disulfide-stabilized Fvs (dsFv) or dsFv immunotoxins. dsFvs and dsFv immunotoxins have several advantages over scFv immunotoxins. This review summarizes the design, construction, activities in vitro and in vivo, and biochemical characteristics of dsFv immunotoxins and compares them with scFv immunotoxins.


Assuntos
Fragmentos de Imunoglobulinas/farmacologia , Imunotoxinas/farmacologia , Neoplasias/terapia , Engenharia de Proteínas , Animais , Dissulfetos , Desenho de Fármacos , Humanos , Camundongos , Proteínas Recombinantes/farmacologia
15.
Protein Eng ; 8(12): 1323-31, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8869646

RESUMO

Using molecular modeling technology we have recently identified positions in conserved framework regions of Fvs which can be used to stabilize antibody Fvs by an interchain disulfide bond engineered in between the structurally conserved framework positions of the variable domains of heavy (VH) and light (VL) immunoglobulin chains (disulfide-stabilized Fv; dsFv). The computer model indicated the existence of other potential sites in the framework regions that might be suitable for disulfide bond formation between VH and VL. The possibility of obtaining dsFvs using these positions is evaluated here experimentally by constructing dsFv immunotoxins in which the Fv moiety is fused to a truncated form of Pseudomonas exotoxin. We analyzed the extent of dsFv formation and the activity of the resulting dsFv immunotoxins, and compared various dsFv molecules with the scFv immunotoxin. Our results demonstrate that position H44-L105 is the only one which gives high production yields of active dsFv. All other positions gave either low yields and activity or completely failed to produce active dsFv. With one exception, the formation and activities of the dsFvs corresponded to the C alpha-C alpha distance between the VH and VL positions, with an optimal distance of 5.7 A producing the best dsFv. Distances of 6.0-6.9 A resulted in a low yield of protein that was still capable of binding antigen, whereas distances > 7.0 A resulted in molecules in which dsFv formation was not obtained.


Assuntos
Dissulfetos/metabolismo , Fragmentos de Imunoglobulinas/química , Imunotoxinas/química , Proteínas Recombinantes de Fusão/química , Sequência de Aminoácidos , Sequência de Bases , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Primers do DNA/química , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Fluorescência , Fragmentos de Imunoglobulinas/genética , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/genética , Imunotoxinas/genética , Imunotoxinas/isolamento & purificação , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/genética , Desnaturação Proteica , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/toxicidade , Triptofano/metabolismo , Ureia/farmacologia
16.
J Immunol ; 155(4): 2203-10, 1995 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-7636268

RESUMO

The complement membrane attack complex (MAC), the cytolytic granule protein of cytotoxic lymphocytes perforin, the streptococcal exotoxin streptolysin O (SLO), and the bee venom polypeptide melittin utilize a similar mechanism to incorporate into cell membranes, induce a Ca2+ influx and a rise in intracellular Ca2+ concentration, and produce cell lysis. At sublytic concentrations, these proteins trigger several cellular activities, including protein phosphorylation and synthesis. We have recently demonstrated that human leukemic cells treated with sublytic doses of human complement become more resistant to lytic complement doses. The study has now been extended to include three other pore-formers: murine perforin, SLO and melittin. As shown here, sublytic MAC induces in the K562 human erythroleukemic cells protection from lytic perforin, and vice versa, sublytic perforin induces protection from complement. Also, sublytic SLO and melittin increase resistance of K562 cells to lytic complement and perforin doses. The capacity of Ca2+ ionophores to induce resistance to the lytic proteins has been examined. Exposure of K562 cells to sublytic concentrations of ionomycin or A23187 for 1 h at 37 degrees C confers on them resistance to complement- and perforin-mediated lysis. The protective effects of the ionophores can be abrogated by chelation of extracellular Ca2+ and by inhibition of RNA or protein synthesis in the cells. These results indicate the following: 1) nucleated cells exposed to sublytic complement MAC, perforin, SLO, or melittin may become resistant to the four pore-formers. Physiologically, this may be regarded as an immunologic tachyphylaxis. 2) Ca2+ influx induced by these pore-formers is an essential and sufficient factor to produce this tachyphylaxis.


Assuntos
Cálcio/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Citotoxicidade Imunológica , Leucemia Eritroblástica Aguda/imunologia , Meliteno/farmacologia , Glicoproteínas de Membrana/farmacologia , Estreptolisinas/farmacologia , Proteínas de Bactérias , Humanos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Células Tumorais Cultivadas
17.
Int J Cancer ; 62(3): 351-5, 1995 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-7628878

RESUMO

B1 (dsFv)-PE38 and B3(dsFv)-PE38 are recombinant immunotoxins in which the Fv fragments of MAbs B1 and B3, respectively, are stabilized by an engineered interchain disulfide bond and are fused at their C-termini to a modified Pseudomonas exotoxin from which the cell-binding domain has been deleted (PE38). Both immunotoxins have been shown to be specifically cytotoxic toward human cancer cell lines which express Le gamma-related carbohydrates on their surface, and when given i.v., eradicated 30- to 50-mm3 s.c. A431 tumors growing in nude mice. A major advantage of dsFv-immunotoxins is their stability at 37 degrees C compared with the relatively unstable single-chain Fvs. This allows them to be given continuously by osmotic pumps placed in the peritoneal cavity. In an attempt to increase the therapeutic index of the immunotoxins, we have now delivered them continuously for 6 days through mini-osmotic pumps placed in the peritoneal cavity of tumor-bearing nude mice. Using this mode of administration, we were able to maintain a constant level of immunotoxin in the serum which was non-toxic to the mice, but caused complete regressions of large 150- to 200-mm3 tumors which lasted for over a month at 1/11 of the LD50 with B1(dsFv)-PE38 and 1/6 of the LD50 with B3(dsFv)-PE38. Complete regression of tumors of similar size could also be achieved by i.v. bolus injections of these immunotoxins at 1/7 of the LD50 with B1(dsFv)-PE38) and 1/3 of the LD50 with B3(dsFv)-PE38. These results suggest that in patients it may be advantageous to administer dsFv-immunotoxins by continuous infusion, since a larger therapeutic index is achieved.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Dissulfetos/administração & dosagem , Exotoxinas/administração & dosagem , Imunotoxinas/administração & dosagem , Neoplasias Experimentais/terapia , Fatores de Virulência , Animais , Anticorpos Monoclonais/administração & dosagem , Dissulfetos/química , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Exotoxinas/sangue , Exotoxinas/química , Feminino , Humanos , Fragmentos de Imunoglobulinas/administração & dosagem , Fragmentos de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/administração & dosagem , Cadeias Leves de Imunoglobulina/administração & dosagem , Região Variável de Imunoglobulina/administração & dosagem , Imunotoxinas/sangue , Imunotoxinas/química , Bombas de Infusão Implantáveis , Infusões Parenterais , Injeções Intravenosas , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo , Exotoxina A de Pseudomonas aeruginosa
18.
Mol Immunol ; 32(4): 249-58, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7723770

RESUMO

Recombinant DNA techniques now allow the production of "mini-antibodies" called Fv fragments. These have been produced either as the independent variable domains of the heavy and light chains non-covalently associated in one-to-one stoichiometry or as single-chain gene products with the two domains linked by an intervening peptide sequence. Although Fv fragments can have excellent binding properties, they are often difficult to produce in good yield and lack the characteristic stability of whole antibodies. To improve the stability of the Fv molecule, we have introduced a cysteine residue into conserved framework regions of both the heavy and light variable domains from the anti-Tac antibody at positions compatible with the formation of an interdomain disulfide linkage (i.e. VH-44 and VL-99). The mutant subunits form a disulfide-bonded Fv molecule, which binds to the alpha-subunit of the IL2 receptor (IL2R alpha) with an affinity identical to that of humanized anti-Tac IgG. This disulfide-stabilized Fv (dsFv) proved to be substantially more resistant to denaturation by heat or urea treatment than the single-chain Fv (scFv). Furthermore, the yield of dsFv is -four-fold higher than that of the single-chain analog.


Assuntos
Fragmentos de Imunoglobulinas/química , Receptores de Interleucina-2/imunologia , Sequência de Aminoácidos , Afinidade de Anticorpos , Sequência de Bases , Ligação Competitiva , Desenho Assistido por Computador , Primers do DNA/química , Dissulfetos , Escherichia coli , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes , Relação Estrutura-Atividade
19.
Immunity ; 2(3): 281-7, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7697545

RESUMO

Disulfide-stabilized Fvs (dsFv) are recombinant Fv fragments of antibodies in which the inherently unstable VH-VL heterodimer is stabilized by an interchain disulfide bond engineered between structurally conserved framework positions. We now design and produce a disulfide-stabilized Fv of a T cell receptor. It is composed of V alpha and V beta variable domains of the 2B4 TCR stabilized by a disulfide bond between framework residues of the TCR Fv at a site corresponding to that used for disulfide stabilization of antibody Fvs. For ease of production and detection, the TCRdsFv was fused to a truncated form of Pseudomonas exotoxin (PE38). The TCR(dsFv) retains its native conformation and is much more stable than a TCR scFv. Moreover, it is functional in biological assays. Because successful disulfide stabilization of the TCR Fv by the positions used for antibody Fv stabilization would not occur unless the mutated residues in TCR Fv are at positions closely similar to those in antibody Fvs, most likely within less than 1.5 A, these results provide very strong experimental evidence for the structural similarity between immunoglobulin and TCR antigen-binding variable domains.


Assuntos
Região Variável de Imunoglobulina/química , Receptores de Antígenos de Linfócitos T alfa-beta/química , Sequência de Aminoácidos , Anticorpos Monoclonais , Sequência de Bases , Dissulfetos , Dados de Sequência Molecular , Plasmídeos , Conformação Proteica , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia
20.
Int J Cancer ; 58(1): 142-9, 1994 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8014011

RESUMO

Disulfide-stabilized Fv (dsFv)-immunotoxins are recombinant immunotoxins in which the inherently unstable Fv moiety, composed of the VH-VL heterodimer, is stabilized by a disulfide bond engineered between structurally conserved framework positions of VH and VL. Anti-Tac(dsFv)-PE38KDEL is composed of such a dsFv, directed to the alpha subunit of the IL2 receptor (IL2R), and containing a truncated form of Pseudomonas exotoxin (PE38KDEL). We have found this new type of immunotoxin to be indistinguishable in its in vitro activity and specificity from its single-chain immunotoxin counterpart, anti-Tac(Fv)-PE38KDEL. We have now examined the therapeutically relevant factors, including stability, pharmacokinetics, and antitumor activity of this new disulfide-stabilized Fv-immunotoxin. We found that anti-Tac(dsFv)-PE38KDEL was specifically cytotoxic to human activated T-lymphocytes in addition to IL2R bearing cell lines. Anti-Tac(dsFv)-PE38KDEL was considerably more stable at 37 degrees C in human serum and in buffered saline than the single-chain immunotoxin, anti-Tac(Fv)-PE38KDEL. The half-life in blood was similar for both immunotoxins (approx. 20 min). The therapeutic potential of the disulfide-stabilized immunotoxin was evaluated using an animal model of immunodeficient mice bearing subcutaneous tumor xenografts of human IL2R-bearing cells. Anti-Tac(dsFv)-PE38KDEL caused complete regression of tumors with no toxic effects in mice. Because dsFv-immunotoxins are more stable and can be produced with significantly improved yields compared to scFv-immunotoxins, dsFv-immunotoxin may be more useful for therapeutic applications.


Assuntos
ADP Ribose Transferases , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Toxinas Bacterianas , Dissulfetos/farmacologia , Exotoxinas/farmacocinética , Exotoxinas/toxicidade , Imunotoxinas/metabolismo , Imunotoxinas/toxicidade , Receptores de Interleucina-2/imunologia , Fatores de Virulência , Animais , Estabilidade de Medicamentos , Feminino , Humanos , Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacocinética , Fragmentos de Peptídeos/toxicidade , Plasmídeos , Proteínas Recombinantes/toxicidade , Exotoxina A de Pseudomonas aeruginosa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...