Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 31(10): 3016-25, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11592078

ABSTRACT

The Her2/neu (c-erbB-2) oncogene encodes a 185-kDa protein tyrosine kinase which is overexpressed in 20% of breast adenocarcinomas and is recognized by a humanized anti-Her2/neu monoclonal antibody (mAb) (rhu4D5 or Herceptin). Natural killer (NK) cells are capable of mediating antibody-dependent cell cytotoxicity (ADCC) against antibody-coated targets via their expression of a low-affinity receptor for IgG (FcgammaRIII or CD16). NK cells can be expanded in cancer patients via the administration of low-dose interleukin-2 (IL-2) and become potent cytotoxic effectors following exposure to high doses of IL-2. We tested IL-2-activated NK cells against Her2/neu+ (MCF-7Her2/neu) and Her2/neu- (MDA-468) breast cancer cell lines in a 4-h 51Cr-release cytotoxicity assay in the presence or absence of rhu4D5 mAb (effector : target ratio = 10 : 1). Specific lysis of rhu4D5-coated MCF-7Her2/neu and MDA-468 target cells by IL-2-activated NK cells was 35% and 3%, respectively (p < 0.05). Lysis was less than 5% when targets were treated with either the non-humanized mu4D5 mAb or control huIgG. Lysis of rhu4D5-coated MCF-7Her2/neu cells was inhibited by 80 % when NK cells were pre-treated with an anti-Fc receptor antibody prior to use in the cytotoxicity assay. Enhanced ADCC of MCF-7Her2/neu target cells was seen when the effector cells consisted of mononuclear cells obtained from a patient demonstrating significant expansion of NK cells secondary to therapy with low-dose IL-2. Serum from patients receiving infusions of rhu4D5 mAb could substitute for exogenous antibody in the ADCC assay. NK cells activated by rhu4D5-coated tumor cells in the presence of IL-2 also produced large amounts of IFN-gamma with concomitant up-regulation of cell-surface activation markers CD25 and CD69. These results lend support to the concurrent use of rhu4D5 mAb and IL-2 therapy in patients with cancers that express the Her2/neu oncogene.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/therapy , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Animals , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD56 Antigen/analysis , Cytokines/biosynthesis , Humans , Killer Cells, Natural/drug effects , Lymphocyte Activation , Mice , Receptor, ErbB-2/analysis , Receptors, IgG/physiology , Trastuzumab , Tumor Cells, Cultured
2.
Transplantation ; 71(1): 170-4, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11211189

ABSTRACT

BACKGROUND: Despite advances in antiviral chemotherapy, herpes simplex virus type 1 (HSV-1), continues to complicate the clinical course of many allograft recipients. We have previously demonstrated that the experimental immunosuppressive agent leflunomide inhibits production of cytomegalovirus by interference with virion assembly. We test the hypothesis that this agent exerts similar antiviral activity against HSV-1. METHODS AND RESULTS: Plaque assay of virus yield from endothelial or Vero cells after inoculation with each of four clinical HSV-1 isolates demonstrated a dose-dependent reduction of virus production in the presence of pharmacologic concentrations of A77 1726, the active metabolite of leflunomide. DNA dot blot and biochemical assay of viral DNA polymerase activity indicated that A77 does not inhibit viral DNA synthesis. Rather, as visualized by transmission electron microscopic method, this agent seems to disrupt virion assembly by preventing nucleocapsid tegumentation. CONCLUSIONS: These findings, in demonstrating that leflunomide exerts antiviral activity against HSV-1 by mechanisms similar to those we have previously shown with cytomegalovirus, imply that this agent may possess broad spectrum activity against other herpesviruses.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Immunosuppressive Agents/pharmacology , Isoxazoles/pharmacology , Animals , Chlorocebus aethiops , Humans , Leflunomide , Vero Cells/virology
3.
Blood ; 96(4): 1465-73, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942393

ABSTRACT

The administration of therapeutic doses of recombinant cytokines to patients with malignant disease can be complicated by systemic toxicities, which in their most severe form may present as a systemic inflammatory response. The combination of interleukin (IL)-18 and IL-12 has synergistic antitumor activity in vivo yet has been associated with significant toxicity. The effects of IL-18 plus IL-12 were examined in a murine model, and it was found that the daily, simultaneous administration of IL-18 and IL-12 resulted in systemic inflammation and 100% mortality within 4 to 8 days depending on the strain employed. Mice treated with IL-18 plus IL-12 exhibited unique pathologic findings as well as elevated serum levels of proinflammatory cytokines and acute-phase reactants. The actions of tumor necrosis factor-alpha did not contribute to the observed toxicity, nor did T or B cells. However, toxicity and death from treatment with IL-18 plus IL-12 could be completely abrogated by elimination of natural killer (NK) cells or macrophages. Subsequent studies in genetically altered mice revealed that NK-cell interferon-gamma mediated the fatal toxicity via the signal transducer and activator of transcription pathway of signal transduction. These data may provide insights into methods of ameliorating cytokine-induced shock in humans. (Blood. 2000;96:1465-1473)


Subject(s)
Inflammation/chemically induced , Inflammation/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-12/toxicity , Interleukin-18/immunology , Interleukin-18/toxicity , Killer Cells, Natural/immunology , Signal Transduction/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Trans-Activators/immunology
4.
J Immunol ; 162(8): 4943-51, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10202041

ABSTRACT

The mechanism of cytokine-induced shock remains poorly understood. The combination of IL-2 and IL-12 has synergistic antitumor activity in vivo, yet has been associated with significant toxicity. We examined the effects of IL-2 plus IL-12 in a murine model and found that the daily, simultaneous administration of IL-2 and IL-12 resulted in shock and 100% mortality within 4 to 12 days depending on the strain employed. Mice treated with IL-2 plus IL-12 exhibited NK cell apoptosis, pulmonary edema, degenerative lesions of the gastrointestinal tract, and elevated serum levels of proinflammatory cytokines and acute phase reactants. The actions of TNF-alpha, IFN-gamma, macrophage-inflammatory protein-1alpha, IL-1, IL-1-converting enzyme, Fas, perforin, inducible nitric oxide synthase, and STAT1 did not contribute to the observed toxicity, nor did B or T cells. However, toxicity and death from treatment with IL-2 plus IL-12 could be completely abrogated by elimination of NK cells. These results suggest that the fatal systemic inflammatory response induced by this cytokine treatment is critically dependent upon NK cells, but does not appear to be mediated by the known effector molecules of this cellular compartment. These data may provide insight into the pathogenesis of cytokine-induced shock in humans.


Subject(s)
Interleukins/adverse effects , Killer Cells, Natural/immunology , Shock, Septic/etiology , Shock, Septic/mortality , Animals , Cell Separation , Cytokines/biosynthesis , Cytokines/blood , Drug Therapy, Combination , Female , Interferon-gamma/physiology , Interleukin-1/physiology , Interleukin-12/administration & dosage , Interleukin-12/adverse effects , Interleukin-15/administration & dosage , Interleukin-15/adverse effects , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Interleukins/administration & dosage , Liver/pathology , Lung/pathology , Macrophage Activation , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Mice, Transgenic , Monocytes/immunology , Shock, Septic/immunology , Shock, Septic/pathology , Spleen/pathology , Tumor Necrosis Factor-alpha/physiology
5.
Infect Immun ; 63(9): 3715-7, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7642312

ABSTRACT

Nitric oxide production by mouse macrophages treated with group B streptococci and gamma interferon was inhibited by cytochalasin B or by antibody neutralization of macrophage-derived tumor necrosis factor alpha. Phagocytosis-induced tumor necrosis factor alpha is responsible for group B streptococcus-induced nitric oxide production in interferon-treated macrophages.


Subject(s)
Interferon-gamma/pharmacology , Macrophages/metabolism , Nitric Oxide/physiology , Streptococcus agalactiae/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cells, Cultured , Cytochalasin B/pharmacology , Mice , Mice, Inbred BALB C , Phagocytosis , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...