Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Immunol ; 186(6): 3735-44, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21300821

ABSTRACT

1E10 is a murine anti-idiotypic mAb specific for an idiotypic mAb that reacts with NeuGc-containing gangliosides, sulfatides, and Ags expressed in some human tumors. In melanoma, breast, and lung cancer patients, this anti-idiotypic Ab was able to induce a specific Ab response against N-glycosylated gangliosides, attractive targets for cancer immunotherapy as these glycolipids are not naturally expressed in humans. A clinical study with nonsmall cell lung cancer patients showed encouraging clinical benefits. Immunological studies performed in 20 of these patients suggested a correlation between the induction of Abs against NeuGcGM3 and longer survival times. The induced anti-NeuGcGM3 Abs recognized and directly killed tumor cells expressing the Ag, by a mechanism independent of complement activation. In the present work, we show that this cytotoxicity differs from apoptosis because it is temperature independent, no chromatin condensation or caspase 3 induction are detected, and the DNA fragmentation induced has a different pattern than the one characteristic for apoptosis. It is a very quick process and involves cytosqeleton reorganization. The Abs induce cellular swelling and the formation of big membrane lesions that allow the leakage of cytoplasm and the loss of the cell membrane integrity. All of these characteristics resemble a process of oncotic necrosis. To our knowledge, this is the first report of the active induction in cancer patients of NeuGcGM3-specific Abs able to induce complement independent oncotic necrosis to tumor cells. These results contribute to reinforcing the therapeutic potential of anti-idiotypic vaccines and the importance of NeuGcGM3 ganglioside as antitumor target.


Subject(s)
Antibodies, Neoplasm/physiology , Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/immunology , Immunoglobulin Idiotypes/physiology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Animals , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/biosynthesis , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Carcinoma, Lewis Lung/ultrastructure , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Death/immunology , Cell Line, Tumor , Dogs , Horses , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin Idiotypes/administration & dosage , Immunoglobulin M/biosynthesis , Leukemia L1210/immunology , Leukemia L1210/pathology , Leukemia L1210/therapy , Lung Neoplasms/ultrastructure , Mice , Mice, Inbred BALB C , Plasmacytoma/immunology , Plasmacytoma/pathology , Plasmacytoma/therapy
2.
J Immunol ; 184(8): 4170-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20220087

ABSTRACT

By studying BALB/c mice deficient in immune components, we show that the protective immunity to rat ErbB2(+) tumors rests on the Ab response elicited by the electroporation of a DNA vaccine encoding the extracellular and transmembrane domains of rat ErbB2. In vivo, the adoptive transfer of vaccine-elicited anti-rat ErbB2 Abs protected against a challenge of rat ErbB2(+) carcinoma cells (TUBO cells). In vitro, such Abs inhibited TUBO cell growth by impairing cell cycle progression and inducing apoptosis. To correlate intrinsic mechanisms of Ab action with their tumor-inhibitory potential, first we showed that TUBO cells constitutively express phosphorylated transgenic ErbB2/autochthonous ErbB3 heterodimers and exhibit a basal level of Akt phosphorylation, suggesting a constitutive activation of the PI3K/Akt pathway. Treatment with anti-ErbB2 Abs caused a drastic reduction in the basal level of Akt phosphorylation in the absence of an impairment of PI3K enzymatic activity. Notably, the same Ab treatment induced an increase in PTEN phosphatase activity that correlated with a reduced PTEN phosphorylation. In conclusion, vaccine-induced anti-ErbB2 Abs directly affected the transformed phenotype of rat ErbB2(+) tumors by impairing ErbB2-mediated PI3K/Akt signaling.


Subject(s)
Antibodies, Neoplasm/physiology , Mammary Neoplasms, Experimental/prevention & control , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Receptor, ErbB-2/immunology , Signal Transduction/immunology , Vaccines, DNA/immunology , 3T3 Cells , Animals , Antibodies, Neoplasm/biosynthesis , Cell Line, Tumor , Electroporation , Extracellular Space/genetics , Extracellular Space/immunology , Female , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , PTEN Phosphohydrolase/physiology , Phosphorylation/immunology , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, ErbB-2/administration & dosage , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
3.
J Immunol ; 182(3): 1242-6, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155468

ABSTRACT

We studied the growth of transgenic adenocarcinoma of mouse prostate (TRAMP)-C1 tumor cells expressing human prostate-specific Ag (PSA) in HLA-DRB1*1501 (DR2b) transgenic mice. TRAMP-PSA tumors were frequently rejected by HLA-DR2b(-) mice but had increased incidence in HLA-DR2b(+) littermates. The levels of PSA-specific CD8 T cell responses were significantly higher in the HLA-DR2b(-) mice that rejected TRAMP-PSA tumors compared with HLA-DR2b(+) tumor-bearing littermates. In contrast, Ab responses to PSA were strong in HLA-DR2b(+) mice bearing TRAMP-PSA tumors and were virtually undetectable in HLA-DR2b(-) littermates. The analysis of CD4 T cell responses to PSA revealed the presence of several CD4 T cell epitopes in HLA-DR2b(+) mice but failed to identify strong I-A(b)-restricted epitopes in HLA-DR2b(-) mice. Our data demonstrate that the expression of a permissive HLA class II allele can change the pattern of the immune response to a tumor Ag, resulting in the failure of tumor rejection.


Subject(s)
Adenocarcinoma/immunology , Alleles , Graft Survival/immunology , HLA-DR Antigens/genetics , HLA-DR2 Antigen/genetics , Prostatic Neoplasms/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8 Antigens/biosynthesis , CD8 Antigens/immunology , Cell Line, Tumor , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/genetics , HLA-DR Antigens/immunology , HLA-DR2 Antigen/immunology , HLA-DRB1 Chains , Histocompatibility Antigens Class II/immunology , Humans , Incidence , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostate-Specific Antigen/biosynthesis , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
4.
J Clin Invest ; 111(8): 1161-70, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12697735

ABSTRACT

Since the mechanisms by which specific immunity destroys Her-2/neu carcinoma cells are highly undetermined, these were assessed in BALB/c mice vaccinated with plasmids encoding extracellular and transmembrane domains of the protein product (p185(neu)) of the rat Her-2/neu oncogene shot into the skin by gene gun. Vaccinated mice rejected a lethal challenge of TUBO carcinoma cells expressing p185(neu). Depletion of CD4 T cells during immunization abolished the protection, while depletion of CD8 cells during the effector phase halved it, and depletion of polymorphonuclear granulocytes abolished all protection. By contrast, Ig mu-chain gene KO mice, as well as Fcgamma receptor I/III, beta-2 microglobulin, CD1, monocyte chemoattractant protein 1 (MCP1), IFN-gamma, and perforin gene KO mice were protected. Only mice with both IFN-gamma and perforin gene KOs were not protected. Although immunization also cured all BALB/c mice bearing established TUBO carcinomas, it did not cure any of the perforin KO or perforin and IFN-gamma KO mice. Few mice were cured that had knockouts of the gene for Ig mu-chain, Fcgamma receptor I/III, IFN-gamma, or beta-2 microglobulin. Moreover, vaccination cured half of the CD1 and the majority of the MCP1 KO mice. The eradication of established p185(neu) carcinomas involves distinct mechanisms, each endowed with a different curative potential.


Subject(s)
Antibodies, Neoplasm/physiology , Cancer Vaccines/immunology , Cytokines/physiology , Membrane Glycoproteins/physiology , Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Vaccines, DNA/immunology , Animals , Biolistics , Female , Graft Rejection , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , Vaccination
5.
J Clin Invest ; 78(2): 349-54, 1986 Aug.
Article in English | MEDLINE | ID: mdl-3734096

ABSTRACT

Hodgkin's disease-derived giant cell lines (HD-cells) express high levels of ectosialyltransferase activity presumed to be a galactose-specific lectin recognizing the desialylated 3-fucosyl-N-acetyllactosamine structure (X-hapten). Both the anti-X-hapten monoclonal antibody VIM-D5 and a polyclonal antiserum to another galactose-lectin, the hepatic asialoglycoprotein receptor (HBP), recognize a 55,000-mol wt HD-cell protein (Paietta, E., R. J. Stockert, A. G. Morell, V. Diehl, and P. H. Weirnik. 1986. Proc. Natl. Acad. Sci. USA. 83:3451-3455.) That the expression of the 55,000-mol wt protein is restricted to HD-cells among X-hapten positive cells lines is confirmed in this study. The 55,000-mol wt protein is shown to be present on the cell surface and intracellularly, where an additional immunocrossreactive 150,000-mol wt protein is recognized. Extraction of the 55,000 mol wt protein from HD-cell lysates by affinity chromatography results in the loss of sialyltransferase activity. While evidence for a single protein possessing both the antigenic and the enzymatic activity is not direct, these results suggest that the ectosialyltransferase unique to HD-cells is a 55,000-mol wt membrane glycoprotein possessing the X-hapten oligosaccharide.


Subject(s)
Antigens, Neoplasm/analysis , Asialoglycoprotein Receptor , Hodgkin Disease/enzymology , Sialyltransferases/immunology , Transferases/immunology , Antibodies, Neoplasm/physiology , Antigens, Neoplasm/immunology , Binding Sites, Antibody , Binding, Competitive , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Compartmentation , Cell Line , Cell Membrane/metabolism , Hodgkin Disease/immunology , Hodgkin Disease/metabolism , Humans , Immune Sera/pharmacology , Precipitin Tests , Sialyltransferases/antagonists & inhibitors
6.
Neuro Oncol ; 8(3): 261-79, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16775224

ABSTRACT

Little is known about the immune performance and interactions of CNS microglia/macrophages in glioma patients. We found that microglia/macrophages were the predominant immune cell infiltrating gliomas ( approximately 1% of total cells); others identified were myeloid dendritic cells (DCs), plasmacytoid DCs, and T cells. We isolated and analyzed the immune functions of CD11b/c+CD45+ glioma-infiltrating microglia/macrophages (GIMs) from postoperative tissue specimens of glioma patients. Although GIMs expressed substantial levels of Toll-like receptors (TLRs), they did not appear stimulated to produce pro-inflammatory cytokines (tumor necrosis factor alpha, interleukin 1, or interleukin 6), and in vitro, lipopolysaccharides could bind TLR-4 but could not induce GIM-mediated T-cell proliferation. Despite surface major histocompatibility complex class II expression, they lacked expression of the costimulatory molecules CD86, CD80, and CD40 critical for T-cell activation. Ex vivo, we demonstrate a corresponding lack of effector/activated T cells, as glioma-infiltrating CD8+ T cells were phenotypically CD8+CD25-. By contrast, there was a prominent population of regulatory CD4 T cells (CD4+CD25+FOXP3+) infiltrating the tumor. We conclude that while GIMs may have a few intact innate immune functions, their capacity to be stimulated via TLRs, secrete cytokines, upregulate costimulatory molecules, and in turn activate antitumor effector T cells is not sufficient to initiate immune responses. Furthermore, the presence of regulatory T cells may also contribute to the lack of effective immune activation against malignant human gliomas.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Antigen-Presenting Cells/immunology , Brain Neoplasms/immunology , Glioma/immunology , Macrophages/immunology , Microglia/immunology , Antibodies, Neoplasm/physiology , Antibody-Producing Cells/immunology , Antibody-Producing Cells/pathology , Antigen-Presenting Cells/pathology , Biomarkers, Tumor/immunology , Brain Neoplasms/pathology , Glioma/pathology , Humans , Macrophages/pathology , Microglia/pathology , Neoplasm Invasiveness
7.
Eur J Cancer ; 42(14): 2326-34, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16908131

ABSTRACT

Malignant rhabdoid tumours (MRT) are highly aggressive cancers of early childhood that arise in different organs or tissues. The unifying criterion for these tumours is the presence of inactivating mutations of the hSNF5/INI1 tumour suppressor gene which encodes a core subunit of the chromatin remodelling SWI/SNF complex. Using a variety of markers we analysed the phenotypic traits of MON and DEV cell lines derived respectively from an undifferentiated abdominal MRT and from a brain MRT. DEV cells express spontaneously a wide range of neural and glial markers. It can be induced to differentiate into the neural lineage following hSNF5/INI1 expression with appearance of neurite processes, strong increase of neural markers and decrease of glial markers. A less pronounced neural differentiation is also observed with MON cells, which possess more primitive polyphenotypic features with positivity for markers from the three embryonic layers. Finally, we show that the neural differentiation of rat PC12 cells in the presence of nerve growth factor (NGF) is strongly impaired when hSNF5/INI1 expression is inhibited by RNA interference. Altogether these results indicate that hSNF5/INI1 is an essential subunit for SWI/SNF-dependant induction of neural differentiation programs. Further experiments should enable documentation of whether it provides instructive or permissive signals for differentiation.


Subject(s)
Cell Transformation, Neoplastic/pathology , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins/physiology , Genes, Tumor Suppressor/physiology , Rhabdoid Tumor/pathology , Transcription Factors/physiology , Animals , Antibodies, Neoplasm/physiology , Blotting, Western , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Flow Cytometry , Mice , Phenotype , RNA, Neoplasm , SMARCB1 Protein , Transcription Factors/metabolism , Transfection
8.
Cancer Res ; 52(10): 2923-30, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1581910

ABSTRACT

Cells of cloned lines of human squamous lung carcinomas elaborate large glycoproteins that are associated with their tumorigenic potential. Two groups of clones (called Le(a)-X-positive and Le(a)-X-negative) were studied that either do or do not express the Le(a)-X oligosaccharide associated with large glycoproteins and mucins secreted by these clones. Le(a)-X-positive cells elaborate a mucin gel complex associated with their apical surfaces, which appears as a mosaic of extracellular plates. Clones of this type are tumorigenic in nude rodents when injected s.c. or when introduced into the lungs via intrabronchial aerosol. By contrast, the Le(a)-X-negative clones do not form extracellular plates and are not tumorigenic in the lungs or subcutaneously. We demonstrate that the extracellular plates of Le(a)-X-positive cells exclude antibodies from interacting with the underlying squamous lung carcinoma cells and may therefore exert an immunoprotective effect. In support of this possibility it was found that: (a) There is a substantial inflammatory cell infiltrate associated with regressing nodules of Le(a)-X-negative cells in nude rodent lung and subcutaneous nodules, while there is no observable infiltration associated with progressing Le(a)-X-positive tumors. (b) In the brain (an immunoprivileged site) tumors develop and progress when either Le(a)-X-negative or -positive cells are introduced.


Subject(s)
Antibodies, Neoplasm/physiology , Carcinoma, Squamous Cell/pathology , Lewis Blood Group Antigens/immunology , Lung Neoplasms/pathology , Mucins/physiology , Oligosaccharides/biosynthesis , Animals , Antibodies, Neoplasm/immunology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Bronchial Neoplasms/pathology , Carbohydrate Sequence , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Cell Communication/physiology , Clone Cells , Female , Gels , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Molecular Sequence Data , Mucins/biosynthesis , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Transplantation , Rats , Rats, Nude , Transplantation, Heterologous , Tumor Cells, Cultured
9.
J Clin Oncol ; 14(4): 1383-400, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8648397

ABSTRACT

PURPOSE: To review antibody structure, function, and production; suitable radioisotopes for radioimmunotherapy; challenges facing the field; recent clinical results; toxicity; and future directions. DESIGN: The radioimmunotherapy literature was reviewed, with an emphasis on clinical results and future directions. RESULTS: The highest complete response rates (overall, approximately 50%) have been achieved in patients with B-cell non-Hodgkin's lymphoma. Challenges that currently face radioimmunotherapy include circulating free antigen, binding of antibodies to nonspecific Fc receptors, insufficient tumor penetration, antigenic heterogeneity and insufficient antigen expression, antigenic modulation, and development of human antimouse antibodies. Possible approaches to these challenges, including high-dose radioimmunotherapy and chemotherapy followed by autologous bone marrow transplantation, the use of radionuclides such as yttrium 90 (90Y) and copper 67 (67Cu), and the development of humanized and bifunctional antibodies, are under investigation. CONCLUSION: Although radioimmunotherapy is a relatively new field, substantial progress has been made. Additional research will ultimately resolve many of the challenges that currently face radioimmunotherapy and hopefully lead to the cure of some currently incurable malignancies.


Subject(s)
Neoplasms/radiotherapy , Radioimmunotherapy , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/physiology , Clinical Trials as Topic , Humans , Leukemia/radiotherapy , Lymphoma/radiotherapy , Neoplasms/immunology , Radioimmunotherapy/adverse effects , Radioimmunotherapy/methods , Radioimmunotherapy/trends , Radioisotopes/therapeutic use , Radiotherapy Dosage
10.
Mol Immunol ; 26(7): 593-9, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2550816

ABSTRACT

A novel murine B lymphoma expressing membrane-associated IgA was isolated and used to compare mechanisms of signal transduction by sIgM and sIgA. Like other isotypes so far studied, crosslinking of sIgA by anti-immunoglobulin antibodies stimulates hydrolysis of inositol phospholipids and causes elevation of intracellular free calcium. Furthermore, signals generated through sIgA are coupled to elevation of c-fos proto-oncogene expression. Coupling appears to be through the protein kinase C rather than through the Ca2+ component of sIg signalling as phorbol diester, but the Ca2+ ionophore cannot mediate this effect. Thus these results, coupled with those from earlier studies, show that early signal transduction through surface immunoglobulin appears to be similar regardless of the particular isotype involved in binding ligand.


Subject(s)
Antibodies, Neoplasm/physiology , Immunoglobulin A/physiology , Lymphoma/immunology , Receptors, Antigen, B-Cell/physiology , Signal Transduction , Animals , B-Lymphocytes/immunology , Calcium/metabolism , Cell Division , Cell Line , Mice , Mice, Inbred BALB C , Phosphatidylinositols/metabolism , Proto-Oncogenes
11.
Mol Immunol ; 32(17-18): 1311-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8643100

ABSTRACT

A monoclonal human IgG1, Campath-1H, was digested with glycosidases to assess the effect of carbohydrate on the functional activities of an IgG1. Removal of the complete carbohydrate moiety abolished complement lysis activity and antibody-dependent cell-mediated cytotoxicity, but left antigen binding activity and protein A binding activity intact. Removal of terminal sialic acid residues through glycopeptidase F digestion was not found to affect any of the tested IgG activities. Removal of the majority of the galactose residues from desialylated Campath-1H was found to reduce but not abolish complement lysis activity. Other activities were not affected by degalactosylation. This indicates a rare separation of complement lysis activity and antibody-dependent cell-mediated cytotoxicity of IgG in the way they behave under controlled conditions. This paper underlines the overall importance of carbohydrate in IgG function and stresses the relative contributions of some of the carbohydrate residues.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/physiology , Antibodies, Neoplasm/metabolism , Antibodies, Neoplasm/physiology , Galactose/metabolism , Sialic Acids/metabolism , Alemtuzumab , Amidohydrolases/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/chemistry , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Carbohydrate Sequence , Complement System Proteins/physiology , Cricetinae , Cytotoxicity Tests, Immunologic , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunoglobulin G/physiology , Molecular Sequence Data , Multiple Myeloma/immunology , N-Acetylneuraminic Acid , Neuraminidase/pharmacology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Structure-Activity Relationship , Tumor Cells, Cultured
12.
J Invest Dermatol ; 107(2): 164-70, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8757757

ABSTRACT

The low cost, high versatility, and reliable production of bacterially produced recombinant antibody fragments speeds up the development of tumor-targeting agents. High-quality recombinant anti-melanoma antibodies are much sought after in the scientific community. We cloned the murine antibody 225.28S, currently used in radioimmunoimaging of human melanoma lesions, in single-chain Fv configuration (scFv) for soluble expression in bacteria. The recombinant antibody fragment conserved the binding specificity of the parental antibody. In order to arm the scFv(225.28S) with biologically useful effector functions, we developed vectors for soluble expression of scFv(225.28S) in bacteria that allow both covalent and noncovalent chemical antibody modification at positions that do not interfere with antigen binding. An expression vector was developed that appends a cysteine residue at the C-terminal extremity of the recombinant antibody, thus allowing reaction with thiol-specific reagents, including 99mTc labeling, at a position that does not interfere with antigen binding. The scFv(225.28S) was also successfully expressed with a casein kinase II substrate tag that enables efficient and stable 32P labeling. For noncovalent antibody modification, we developed an expression vector that appends the human calmodulin gene at the C-terminal extremity of scFv(225.28S). The calmodulin domain is poorly immunogenic and can be targeted with chemically modified high-affinity calmodulin ligands. The recombinant anti-human melanoma antibodies described in this article should prove useful "building blocks" for the development of anti-melanoma diagnostic and therapeutic strategies.


Subject(s)
Antibodies, Monoclonal/physiology , Antibodies, Neoplasm/physiology , Melanoma/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/isolation & purification , Antibody Specificity , Base Sequence , Cloning, Molecular , Humans , Immunoglobulin G/immunology , Mice , Molecular Probes/genetics , Molecular Sequence Data , Recombinant Proteins , Sequence Tagged Sites
13.
Clin Exp Metastasis ; 20(2): 103-9, 2003.
Article in English | MEDLINE | ID: mdl-12705631

ABSTRACT

Cancer metastasis involves distinct steps that depend on complicated tumor-host interactions. The hematogenous dissemination of tumor cells may be facilitated by factors that promote the arrest and adherence of cancer cells in capillaries. We examined whether anti-tumor monoclonal immunoglobulin M (IgM) antibodies promoted the hematogenous dissemination of B16 melanoma cells in syngeneic mice. IgM monoclonal antibodies were generated that selectively bind to B16 melanoma cells as compared to syngeneic fibroblasts, lymphocytes or Lewis lung carcinoma cells. Incubation of B16-BL6 or B16-F0 melanoma cells with these IgM anti-tumor antibodies significantly increased the number of lung colonies as compared with control antibodies. Moreover, intraperitoneal injection of specific antibody also significantly increased lung colonization. All anti-tumor antibodies promoted the aggregation of B16 melanoma cells. A chemically generated immunoglobulin G (IgG)-like fragment of an anti-tumor IgM antibody displayed greatly reduced tumor aggregation and, in contrast to intact IgM, did not significantly increase lung colonization of B16 melanoma cells. Neither intact IgM nor the IgG-like fragment enhanced the in vitro invasiveness of B16 melanoma cells across Matrigel-coated membranes. Our results, therefore, suggest that besides their beneficial anti-tumor effects, anti-tumor IgM antibodies may also promote the hematogenous dissemination of cancer cells.


Subject(s)
Antibodies, Neoplasm/physiology , Immunoglobulin M/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Neoplastic Cells, Circulating/immunology , Animals , Antibodies, Monoclonal , Cell Aggregation , Disease Models, Animal , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin G/immunology , In Vitro Techniques , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Invasiveness , Tumor Cells, Cultured
14.
Leuk Res ; 12(11-12): 929-39, 1988.
Article in English | MEDLINE | ID: mdl-3146004

ABSTRACT

An antigen identified by murine monoclonal antibody YB5.B8 has previously been detected only on acute non-lymphoblastic leukaemia (ANLL) cells and tissue mast cells. We now report that the YB5.B8 antigen is present on a minor population (up to 3%) of normal bone marrow mononuclear cells which overlaps the set of progenitor cells capable of forming haemopoietic colonies in vitro. The results indicate that the antigen is a normal haemopoietic progenitor cell marker which is selectively retained on mast cells during maturation, and that leukaemias which express the antigen are not necessarily committed to the mast cell lineage. Furthermore, the antibody was capable of partially inhibiting the formation of haemopoietic colonies in vitro, indicating an important functional role for the antigen. This is consistent with the observation, reported in the accompanying paper, that expression of the YB5.B8 antigen is strongly correlated with poor response to therapy in patients with ANLL.


Subject(s)
Antibodies, Monoclonal/physiology , Antigens, Neoplasm/immunology , Binding Sites, Antibody , Growth Inhibitors/physiology , Hematopoietic Stem Cells/metabolism , Antibodies, Neoplasm/physiology , Bone Marrow/metabolism , Cell Division , Cell Separation , Colony-Forming Units Assay , Flow Cytometry , Hematopoietic Stem Cells/physiology , Humans , Leukemia, Mast-Cell/immunology , Leukemia, Myeloid/immunology , Rosette Formation
15.
Med Hypotheses ; 60(3): 360-72, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12581613

ABSTRACT

Immunoengineering is a term coined to represent the mostly future ability to use or target the immune system's natural components, with emphasis on the regulatory components, to up or down regulate the immune system's attack against specific proteins associated with an unwanted pathology or immune occurrence. It will constitute manipulating parts of the immune system, mostly those specific for the disease associated antigen(s) and generally of a regulatory nature, in various immunological locale or the whole body compartment, to achieve a disease free state for the patient. The number of practical applications awaiting the mastery of immune components as regulatory therapeutics is enormous and immunoengineering should provide treatments in a wide range of disease categories. HIV is a disease where this discipline could provide a quick cure, even eradication of the virus. A potential cheap solution to HIV infection, based on using immunoengineering and adaptable to the infrastructure problems of the Third World is highlighted in the following because of the health emergency that exists in the Third World.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/therapy , Antibodies, Monoclonal/physiology , Antibodies, Neoplasm/physiology , Bone Marrow Transplantation , Immunotherapy/methods , Organ Transplantation , Alemtuzumab , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/metabolism , Antibodies, Neoplasm/therapeutic use , HIV Infections/therapy , Humans , Immunization , Models, Theoretical , T-Lymphocytes/immunology
18.
Vet Immunol Immunopathol ; 141(1-2): 46-57, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21371757

ABSTRACT

BACKGROUND: The cancer stem cell hypothesis proposes that tumours are maintained by a population of cancer stem cells (CSC), which must be eradicated to prevent disease relapse after treatment. Cells expressing high levels of CD44 have been identified as candidate CSC in a variety of human tumours. This study sought to investigate CD44 expression and its potential as a CSC marker in canine cancer. METHODS: CD44 expression in several canine cancer cell lines was determined by flow cytometry. Cells with low and high levels of CD44 expression were examined for differences in growth characteristics, colony forming ability, drug sensitivity and cell cycle profile. RESULTS: CD44(High) cells demonstrated enhanced growth and colony forming capacity, under both adherent and low-density serum free ("tumoursphere") conditions. However, no difference in sensitivity to doxorubicin was seen between the two populations. Moreover, whilst most CD44(Low) cells were in resting or G1 growth phase, an increased proportion of CD44(High) cells were in G2M phase of the cell cycle. Upon proliferation in culture, both populations gave rise to progeny with a full spectrum of CD44 expression. CONCLUSION: CD44 expression is associated with proliferation in cultured canine cancer cells, but transient and fluctuating expression may limit its utility as a CSC marker.


Subject(s)
Cell Proliferation , Dog Diseases/immunology , Hyaluronan Receptors/physiology , Neoplasms/immunology , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/physiology , Antigens, Surface/immunology , Cell Cycle/immunology , Cell Cycle/physiology , Cell Line, Tumor , Dog Diseases/physiopathology , Dogs , Flow Cytometry/veterinary , Gene Expression Regulation, Neoplastic/immunology , Hyaluronan Receptors/biosynthesis , Neoplasms/physiopathology , Tumor Stem Cell Assay/veterinary
19.
Curr Pharm Des ; 16(21): 2369-76, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20618157

ABSTRACT

In order to reduce systemic toxicity and effectively deliver macromolecular drug into tumor cells, a system termed "ATTEMPTS" (antibody targeted, [protamine] triggered, electrically modified prodrug-type strategy) was developed in our laboratory. This approach was adapted from our previously reported heparin/protamine-based system for controlled delivery of protease drugs such as tissue- specific plasminogen activator (tPA). In this "ATTEMPTS" system, the cell-permeable protein drugs are synthesized by conjugating proteins to cell-penetrating peptides (CPPs). Cell penetration ability of such CPP-protein conjugates would initially be disabled, acting as a "prodrug", by forming polyelectrolyte complexes with a functionalized heparin-antibody moiety. The complexes would accumulate in tumor sites by the antibody targeting function, and then the local release of CPP-protein conjugates would be triggered by protamine. We applied this system to the macromolecular anticancer agents, such as the protein drugs (gelonin and asparaginase) as well as the polymerdrugs (polyrotaxane-doxorubicin and polyrotaxane-camptothecin). Both in vitro and preliminary in vivo studies demonstrated the regulable cell penetration behavior based on the competitive ionic interactions between CPP/heparin and heparin/protamine. Thus, this ATTEMPTS approach provides a multi-functionalized system incorporating the features of targeting, prodrug-like, triggerable release, and cell penetration ability for the delivery of macromolecular anticancer agents. A summary of our work on "ATTEMPTS" is presented in this review.


Subject(s)
Antineoplastic Agents/chemistry , Drug Delivery Systems/methods , Prodrugs/chemistry , Animals , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/physiology , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Heparin/administration & dosage , Humans , Macromolecular Substances/administration & dosage , Macromolecular Substances/chemistry , Prodrugs/administration & dosage , Protamines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL