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.
Clin Cancer Res ; 5(11): 3632-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589780

ABSTRACT

The efficacy of chemotherapy has been improved by regimens that combine several cytotoxic drugs with different mechanisms of action and/or different dose-limiting toxicities. Here we demonstrate clearly, and for the first time, that combined therapy using an anticarcinoma immunoconjugate, BR96-doxorubicin, and the cytotoxic drug paclitaxel results in a significant increase in antitumor activity over that of either agent alone. Synergistic activity was seen at doses of BR96-doxorubicin that were minimally active as single agents. A dramatic increase in regression rates was seen when a regimen that combined BR96-doxorubicin and paclitaxel was used to treat both paclitaxel-sensitive and paclitaxel-insensitive carcinomas. Importantly, combined therapy resulted in increased antitumor activity against lung, colon, and breast tumors xenografted in athymic mice and large, paclitaxel-insensitive colon tumors xenografted in athymic rats that also express the Lewis(y) target antigen in normal tissues.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/therapeutic use , Neoplasms/drug therapy , Paclitaxel/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasms/pathology , Paclitaxel/administration & dosage , Rats , Rats, Nude , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured
2.
J Exp Med ; 190(10): 1535-40, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10562327

ABSTRACT

The 4-1BB receptor (CDw137), a member of the tumor necrosis factor receptor superfamily, has been shown to costimulate the activation of T cells. Here we show that anti-mouse 4-1BB monoclonal antibodies (mAbs) inhibit thymus-dependent antibody production by B cells. Injection of anti-4-1BB mAbs into mice being immunized with cellular or soluble protein antigens induced long-term anergy of antigen-specific T cells. The immune response to the type II T cell-independent antigen trinintrophenol-conjugated Ficoll, however, was not suppressed. Inhibition of humoral immunity occurred only when anti-4-1BB mAb was given within 1 wk after immunization. Anti-4-1BB inhibition was observed in mice lacking functional CD8(+) T cells, indicating that CD8(+) T cells were not required for the induction of anergy. Analysis of the requirements for the anti-4-1BB-mediated inhibition of humoral immunity revealed that suppression could not be adoptively transferred with T cells from anti-4-1BB-treated mice. Transfer of BALB/c splenic T cells from sheep red blood cell (SRBC)-immunized and anti-4-1BB-treated mice together with normal BALB/c B cells into C.B-17 severe combined immunodeficient mice failed to generate an anti-SRBC response. However, B cells from the SRBC-immunized, anti-4-1BB-treated BALB/c mice, together with normal naive T cells, exhibited a normal humoral immune response against SRBC after transfer, demonstrating that SRBC-specific B cells were left unaffected by anti-4-1BB mAbs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Formation , Immune Tolerance , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes/physiology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antigens, CD , Erythrocytes/immunology , Female , Ficoll/analogs & derivatives , Ficoll/immunology , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Trinitrobenzenes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9
3.
Cancer Res ; 57(1): 100-5, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8988048

ABSTRACT

The internalizing anti-Le(y) monoclonal antibody (MAb) BR64 was conjugated to the anticancer drug doxorubicin (DOX) using an acid-labile hydrazone bond to the DOX and either a disulfide or thioether bond to the MAb. The resulting disulfide (BR64-SS-DOX) and thioether (BR64-S-DOX) conjugates were evaluated for stability, potency, and antigen-specific activity in both in vitro and in vivo model systems. The BR64-SS-DOX conjugates demonstrated antigen-specific activity both in vitro and when evaluated against antigen-expressing, DOX-sensitive human carcinoma xenografts. However, the stability and potency of disulfide conjugates were poor, and in vivo activity superior to unconjugated DOX was seen only at doses approaching the maximum tolerated dose. Furthermore, BR64-SS-DOX conjugates were not active against antigen-expressing, DOX-insensitive colon tumor xenografts. In contrast, the BR64-S-DOX conjugates demonstrated good stability both in vitro and in vivo. The increased stability of the BR64-S-DOX conjugates resulted in the delivery of more biologically active DOX to tumors with a concomitant increase in potency and efficacy over that which could be achieved with either unconjugated DOX or BR64-SS-DOX conjugates. Delivery of DOX by BR64-SS-DOX conjugates resulted in complete regressions and cures of both DOX-sensitive lung xenografts and DOX-intensitive colon tumor xenografts. These results demonstrate the importance of linker stability when delivering drugs such as DOX to carcinomas via internalizing antibodies and are likely to have direct relevance to the clinical utility of MAb-directed delivery.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Doxorubicin/pharmacology , Immunoconjugates/pharmacology , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Screening Assays, Antitumor , Epitopes/immunology , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Transplantation, Heterologous , Tumor Cells, Cultured
4.
Arch Oral Biol ; 37(2): 139-45, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1622340

ABSTRACT

Platelet-derived growth factor (PDGF) and insulin-like growth factor-I (IGF-I) interact to stimulate proliferation of fibroblasts in culture. Glucocorticoids variably effect the response of cultured fibroblasts to polypeptide growth factors. This study determined the effects of dexamethasone on growth-factor stimulation of gingival, periodontal ligament and pulp fibroblast proliferation in vitro. Cultures of quiescent, low-passage, human fibroblasts were treated for varying periods of time with transforming growth factor-beta 2 (TGF-beta 2), PDGF and IGF-I: (1) alone, (2) in combination with each other, (3) singly plus dexamethasone, (4) in combination plus dexamethasone. Combinations of human, recombinant PDGF and IGF-I (10-1000 ng/ml) induced significantly higher rates of cell proliferation than either factor alone. Dexamethasone at doses ranging from 10(-5) to 10(-8) M substantially enhanced cell proliferation induced by these combinations and by PDGF without IGF-I but not IGF-I alone. By 6 days after a single application, 2-3 times as many cells were present in the PDGF and dexamethasone cultures as compared to PDGF without IGF-I. TGF-beta 2 specifically blocked the effects of dexamethasone added to PDGF-stimulated cells. Collagen and non-collagenous protein synthesis was not affected by the addition of PDGF and IGF-I with or without dexamethasone. These data suggest that dexamethasone may substitute for IGF-I in PDGF stimulation of cell proliferation.


Subject(s)
Dental Pulp/drug effects , Dexamethasone/pharmacology , Gingiva/drug effects , Periodontal Ligament/drug effects , Platelet-Derived Growth Factor/pharmacology , Cell Division/drug effects , Cells, Cultured , Collagen/biosynthesis , Dental Pulp/cytology , Dental Pulp/metabolism , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gingiva/cytology , Gingiva/metabolism , Humans , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Mitogens/administration & dosage , Mitogens/pharmacology , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Platelet-Derived Growth Factor/administration & dosage , Proline/metabolism , Prostaglandins E/biosynthesis , Transforming Growth Factor beta/administration & dosage , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...