Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 53(6): 497-509, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14648071

ABSTRACT

PURPOSE: In the field of cancer immunotherapy research, the targeting of effector cells with specific antibodies is a very promising approach. Recent advances in genetic engineering have made it possible to prepare immunoglobulin fragments consisting of variable domains using bacterial expression systems. METHODS: We have produced an anti-epidermal growth-factor receptor (EGFR) x anti-CD3 bispecific diabody (Ex3 diabody) in an Escherichia coli (E. coli) expression system with refolding method. The Ex3 diabody targets lymphokine-activated killer cells with a T-cell phenotype (T-LAK cells) to EGFR positive bile duct carcinoma cells with dramatic enhancement of cytotoxicity in vitro. This specific killing of EGFR-positive cells was completely inhibited by parental mAb IgGs directed to EGFR and the CD3 antigen. RESULTS: When T-LAK cells were cultured with EGFR-positive tumor cells in the presence of Ex3 diabody, they produced much higher levels of IFN-gamma, GM-CSF, and TNF-alpha than in its absence, this being a possible mechanism underlying specific antitumor activity. The Ex3 diabody showed good stability when tested at 37 degrees C for 48 h, and also markedly inhibited tumor growth of bile duct carcinoma xenografts in severe combined immunodeficient (SCID) mice. When Ex3 diabody (20 microg/mouse) was administrated intravenously, together with T-LAK cells and interleukin-2 (IL-2), complete cure of tumors were observed in three of six mice, and the other three showed marked retardation of tumor growth. CONCLUSION: The Ex3 diabody can be considered a highly promising reagent for study of specific targeting immunotherapy against bile duct and other EGFR-positive carcinomas.


Subject(s)
Antibodies, Bispecific/immunology , Bile Duct Neoplasms/therapy , CD3 Complex/immunology , ErbB Receptors/immunology , Immunotherapy , Killer Cells, Lymphokine-Activated/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/therapeutic use , Bile Duct Neoplasms/immunology , Cells, Cultured , Escherichia coli/metabolism , Female , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Hybridomas/immunology , Immunoglobulin G/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Mice , Mice, Inbred ICR , Mice, SCID , Phenotype , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
2.
Cancer Immunol Immunother ; 52(2): 97-106, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12594573

ABSTRACT

4-1BB ligand (4-1BBL), a member of the tumor necrosis factor (TNF) superfamily, interacts with 4-1BB (CDw137) expressed on activated T cells and delivers a costimulatory signal for T cell activation and growth. Various studies have demonstrated a role for murine 4-1BB in immune function, but relatively few investigations of human 4-1BB have been conducted. Here we report on the construction of a recombinant E1/E3-deleted adenovirus encoding human 4-1BBL (Ad4-1BBL) and its stimulation of antitumor immunity. Ad4-1BBL was able to efficiently infect several human adenocarcinoma cell lines and induce 4-1BBL expression on the cell surface within 24 h, this enhancing the antitumor activity not only of lymphokine-activated killer cells with a T cell phenotype (T-LAK) but also naive peripheral blood mononuclear cells (PBMC). This antitumor activity with T-LAK cells was further enhanced by addition of bispecific antibody (BsAb; anti-MUC1xanti-CD3). Cocultivation of Ad4-1BBL-infected tumor cells with either T-LAK cells or PBMC resulted in significant elevation of interferon-gamma (IFN-gamma), interleukin-2 (IL-2), and granulocyte-macrophage colony-stimulating factor (GM-CSF) production. Furthermore, remarkable tumor growth inhibition was observed in cholangiocarcinoma-grafted severe combined immunodeficient (SCID) mice to which Ad4-1BBL and T-LAK cells were administered when tumor size exceeded 5 mm in diameter. These results provide strong evidence in support of the efficacy of adenovirally delivered 4-1BBL for genetic immunotherapy of cancer.


Subject(s)
Adenoviridae/genetics , Bile Ducts, Intrahepatic , Cholangiocarcinoma/therapy , Genetic Therapy , Killer Cells, Lymphokine-Activated/immunology , Tumor Necrosis Factor-alpha/genetics , 4-1BB Ligand , Animals , Antibodies, Bispecific/therapeutic use , Antigens, CD , Bile Duct Neoplasms/therapy , CD3 Complex/immunology , Cholangiocarcinoma/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, SCID , Mucin-1/immunology , Neoplasm Transplantation , Neoplasms, Experimental/therapy , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Tumor Necrosis Factor-alpha/physiology
3.
Immunol Lett ; 81(2): 99-106, 2002 Apr 22.
Article in English | MEDLINE | ID: mdl-11852114

ABSTRACT

For the purpose of establishing a new adoptive immunotherapy for bile duct carcinoma (BDC), we previously constructed two kinds of bispecific antibodies (bsAbs), anti-MUC1 x anti-CD3 (M x 3) and anti-MUC1 x anti-CD28 (M x 28), which activate T cells and form bridges between them and MUC1-expressing tumor cells. In our previous studies [Cancer Res. 56 (1996) 4205] specific targeting therapy (STT) consisting of i.v. administration of lymphokine activated killer cells with a T cell phenotype (T-LAK) sensitized with two kinds of bsAbs to human BDC-grafted severe combined immunodeficient (SCID) mice demonstrated remarkable inhibition of tumor growth. However, complete cures could not be obtained. In order to improve antitumor efficacy, we have paid attention to anti-CD2 monoclonal antibodies (mAbs), thought to play an important roles in signal transduction in T cell activation or control of T cell receptor (TCR)-driven activation. Therefore, we developed another bsAb, anti-MUC1 x anti-CD2 (M x 2), in order to examine if this would show synergism with the two previously described bsAbs. The combination of the three bsAbs (M x 3, M x 28 and M x 2 bsAbs) showed highest cytotoxicity against MUC1-expressing BDC cells when given simultaneously with peripheral blood mononuclear cells (PBMCs) or T-LAK cells in vitro. When 2 x 10(7) T-LAK cells sensitized with different combinations of bsAbs were administered four times i.v. to BDC-grafted SCID mice, the best therapeutic result was obtained with a combination of all three bsAbs. These results indicate usefulness of combination of three bsAbs for targeting cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bile Duct Neoplasms/therapy , CD2 Antigens/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive/methods , Mucin-1/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibody Specificity , Female , Humans , Interferon-gamma/biosynthesis , Killer Cells, Lymphokine-Activated/immunology , Leukocytes/cytology , Leukocytes/immunology , Lymphocyte Subsets/immunology , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/therapy , Tumor Cells, Cultured
4.
Cancer Immunol Immunother ; 51(1): 33-44, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11845258

ABSTRACT

In cancer immunotherapy research, many bispecific antibodies (BsAbs) have been developed for directing T cells toward tumor cells. Recent advances in genetic engineering have made it possible to prepare immunoglobulin fragments consisting of variable domains using bacterial expression systems. Therefore, recombinant BsAbs, termed diabodies, have attracted particular attention. We have previously produced an anti-MUC1 x anti-CD3 diabody (Mx3 diabody) in an Escherichia coli ( E. coli) expression system. In order to reinforce the antitumor effects of the Mx3 diabody, mutated superantigen staphylococcal enterotoxin A (SEA) D227A was genetically fused to the Mx3 diabody. The SEA D227A fusion Mx3 diabody (SEA D227A-Mx3 diabody) thus constructed showed remarkable MUC1-specific antitumor effects when used with effector cells (lymphokine-activated killer cells with T-cell phenotype [T-LAK] and peripheral blood mononuclear cells [PBMCs]). In the bile duct carcinoma (BDC)-xenografted severe combined immunodeficient (SCID) mouse model, it also demonstrated strong antitumor activity when administered i.v. together with T-LAK cells and interleukin-2 (IL-2). In this experiment, the complete disappearance of tumors was observed in 3 out of 6 mice, and the other 3 showed marked retardation of tumor growth. Therefore, the SEA D227A-Mx3 diabody is considered to be a promising reagent in specific targeted immunotherapy for BDC and other MUC1-positive carcinomas. This is the first report on a diabody that is effective in treating human solid cancers in the xenografted SCID mouse experimental model.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , Bile Duct Neoplasms/therapy , CD3 Complex/immunology , Carcinoma/therapy , Immunotherapy , Mucin-1/immunology , Neoplasm Proteins/immunology , Superantigens/immunology , Animals , Antibodies, Bispecific/therapeutic use , Bile Duct Neoplasms/immunology , Carcinoma/immunology , Enterotoxins/genetics , Enterotoxins/immunology , Escherichia coli , Flow Cytometry , Genetic Vectors/genetics , Humans , Hybridomas/immunology , Killer Cells, Lymphokine-Activated/immunology , Mice , Mice, SCID , Molecular Weight , Protein Folding , Protein Structure, Tertiary , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Superantigens/therapeutic use , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...