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.
J Neurooncol ; 109(2): 229-38, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22696210

ABSTRACT

The aim of this study is to investigate the anti-cancer effect of the bispecific diphtheria toxin (DT) based immunotoxin DTATEGF, which targets both the epidermal growth factor (EGF) receptor (EGFR) and the urokinase-type plasminogen activator (uPA) receptor (uPAR) in vitro and in vivo when delivered by convection-enhanced delivery (CED) via an osmotic minipump in a human metastatic non-small cell lung cancer (NSCLC) brain tumor mouse xenograft model. The effects of the bispecific immunotoxin DTATEGF, and monospecific DTAT, DTEGF and control DT at various concentrations were tested for their ability to inhibit the proliferation of human metastatic NSCLC PC9-BrM3 cells in vitro by MTT assay. A xenograft model of human metastatic NSCLC intracranial model was established in nude mice using the human NSCLC PC9-BrM3 cell line genetically marked with a firefly luciferase reporter gene. One microgram of DTATEGF in the treatment group or control DT in the control group was delivered intracranially by CED via an osmotic minipump. The bioluminescent imaging (BLI) was performed at day 7, 14, 1 month, 2 months, and 3 months. Kaplan-Meier survival curves for the two groups were generated. The brain tissue samples were stained by hematoxylin and eosin for histopathological assessment. In vitro, DTATEGF could selectively kill PC9-BrM3 cells and showed an IC(50) less than 0.001 nM, representing a more than 100- to 1000-fold increase in activity as compared to monospecific DTAT and DTEGF. In vivo, mice with tumors were treated intracranially with drug via CED where the results showed the treatment was successful in providing a survival benefit with the median survival of mice treated with DTATEGF being significantly prolonged relative to controls (87 vs. 63 days, P = 0.006). The results of these experiments indicate that DTATEGF kills the NSCLC PC9-BrM3 cell line in vitro, and when it is delivered via CED intracranially, it is highly efficacious against metastatic NSCLC brain tumors. DTATEGF is a safe and effective drug where further preclinical and clinical development is warranted for the management of metastatic brain tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/pathology , Diphtheria Toxin/administration & dosage , Epidermal Growth Factor/metabolism , Animals , Body Weight/drug effects , Brain Neoplasms/mortality , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Delivery Systems , Humans , Kaplan-Meier Estimate , Mice , Mice, Nude , Neoplasm Transplantation , Recombinant Fusion Proteins/metabolism , Time Factors , Xenograft Model Antitumor Assays
2.
Cancer Biother Radiopharm ; 22(4): 488-500, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17803443

ABSTRACT

Studies were performed to determine the suitability of using the polyethylene glycol (PEG)-labeled AHN-12 anti-CD45 monoclonal antibody to deliver the high-energy beta-particle-emitting isotope 90Y to a CD45+ B-cell Daudi lymphoma grown as flank tumors in athymic nude mice. The PEGylated radiolabeled antibody displayed a significantly better antitumor effect in the mouse tumor flank model (p<0.03) and significantly better blood pharmacokinetics in normal rats (p<0.05) than the non-PEGylated radiolabeled antibody. Studies of two different sizes of PEG showed that rats given 43 kDa of PEGylated AHN-12, but not 5 kDa of PEGylated AHN-12, had significantly higher radiolabeled antibody blood levels and, therefore, improved pharmacokinetics, as compared to rodents given non-PEGylated radiolabeled AHN-12 (p<0.05). Surviving mice revealed no signs of kidney, liver, or gastrointestinal damage by histology study. Notably, in vitro studies indicated that PEGylation did not have a major effect on labeling efficiency and the binding of labeled antibody. These findings indicate that PEGylation of radiolabeled anti-CD45 antibody may be a useful and desirable means of extending blood half-life and enhancing efficacy. Also, the final outcome may be impacted by the size of the PEG molecule used for the modification of the blood half-life.


Subject(s)
Antibodies/therapeutic use , Leukocyte Common Antigens/immunology , Neoplasms/immunology , Neoplasms/radiotherapy , Polyethylene Glycols/chemistry , Yttrium Radioisotopes/chemistry , Yttrium Radioisotopes/therapeutic use , Animals , Antibodies/immunology , Antibodies/pharmacology , Body Weight/drug effects , Cell Line, Tumor , Chromatography, Ion Exchange , Humans , Immunotherapy , Leukocyte Common Antigens/metabolism , Mice , Mice, Nude , Molecular Weight , Neoplasms/metabolism , Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 11(10): 3879-88, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15897589

ABSTRACT

A novel bispecific single-chain fusion protein, DT2219, was assembled consisting of the catalytic and translocation domains of diphtheria toxin (DT(390)) fused to two repeating sFv subunits recognizing CD19 and CD22 and expressed in Escherichia coli. Problems with yield, purity, and aggregation in the refolding step were solved by incorporating a segment of human muscle aldolase and by using a sodium N-lauroyl-sarcosine detergent-based refolding procedure. Problems with reduced efficacy were addressed by combining the anti-CD19 and anti-CD22 on the same single-chain molecule. DT2219 had greater anticancer activity than monomeric or bivalent immunotoxins made with anti-CD19 and anti-CD22 sFv alone and it showed a higher level of binding to patient leukemia cells and to CD19(+)CD22(+) Daudi or Raji cells than did anti-CD19 and anti-CD22 parental monoclonal antibodies. The resulting DT2219, mutated to enhance its avidity, was cytotoxic to Daudi cells in vitro (IC(50) = 0.3 nmol/L). In vivo, DT2219 was effective in a flank tumor therapy model in which it significantly inhibited tumor growth (P < 0.05) and in a systemic model in which it significantly prolonged survival of severe combined immunodeficient mice with established Daudi (P < 0.008) compared with controls. DT2219 has broader reactivity in recognizing B-cell malignancies, has more killing power, and requires less toxin than using individual immunotoxin, which warrants further investigation as a new drug for treating B leukemia/lymphoma.


Subject(s)
Antigens, CD19/immunology , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Cell Adhesion Molecules/immunology , Immunotoxins/immunology , Immunotoxins/pharmacology , Lectins/immunology , Animals , Antibodies , Cell Death , Diphtheria Toxin/immunology , Escherichia coli/genetics , Escherichia coli/immunology , Female , Leukemia, B-Cell , Lymphoma, B-Cell , Mice , Mice, Nude , Mice, SCID , Molecular Conformation , Sialic Acid Binding Ig-like Lectin 2 , Survival Analysis , Transplantation, Heterologous , Tumor Cells, Cultured
4.
Leuk Res ; 29(3): 331-41, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15661270

ABSTRACT

A novel bivalent single chain fusion protein, Bic3, was assembled consisting of the catalytic and translocation domains of diphtheria toxin (DT(390)) fused to two repeating sFv molecules recognizing human CD3 epsilon of the human T-cell receptor. Historically, problems with these constructs include low yield, toxicity, and reduced efficacy. Instead of using conventional Gly(4)Ser linkers to connect heavy/light chains, aggregation reducing linkers (ARL) were used which when combined with a new SLS-based refolding method reduced aggregation and enhanced the yield of final product. Toxicity was reduced at least 25-fold by repeating the two sFv molecules and adding a portion of the hinge-CH2-CH3 human constant regions. The resulting Bic3 was just as cytotoxic to HPB-MLT.UM T leukemia cells in vitro (IC(50)=4 pmol) as a monovalent construct made with the same DT and sFv. In vivo, Bic3 was effective in a new and aggressive therapy model in which it significantly prolonged survival of scid mice with established human T-cell leukemia (p<0.0001 compared to controls). Importantly, no toxicity measured by weight loss, enzyme function, or histology was observed at the highest dose of Bic3 tested (2000 ug/kg). Bic3 warrants investigation as a new drug for treating T-cell malignancy and other T-cell related disorders.


Subject(s)
Immunotoxins/adverse effects , Immunotoxins/chemistry , Leukemia, T-Cell/drug therapy , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/chemical synthesis , Animals , Antibodies, Monoclonal , CD3 Complex/immunology , Disease Models, Animal , Humans , Immunoglobulin G , Kidney/pathology , Mice , Mice, SCID , Receptors, Antigen, T-Cell , Recombinant Fusion Proteins/chemistry , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...