Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Int J Oncol ; 55(1): 309-319, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31180549

ABSTRACT

Fusion toxins consisting of an affinity protein fused to toxic polypeptides derived from Pseudomonas exotoxin A (ETA) are promising agents for targeted cancer therapy. In this study, we examined whether fusion toxins consisting of an albumin binding domain­derived affinity protein (ADAPT) interacting with human epidermal growth factor receptor 2 (HER2), coupled to the ETA­derived polypeptides PE38X8 or PE25, with or without an albumin binding domain (ABD) for half­life extension, can be used for specific killing of HER2­expressing cells. The fusion toxins could easily be expressed in a soluble form in Escherichia coli and purified to homogeneity. All constructs had strong affinity for HER2 (KD 10 to 26 nM) and no tendency for aggregation could be detected. The fusion toxins including the ABD showed strong interaction with human and mouse serum albumin [equilibrium dissociation constant (KD) 1 to 3 nM and 2 to 10 nM, respectively]. The in vitro investigation of the cytotoxic potential revealed IC50­values in the picomolar range for cells expressing high levels of HER2. The specificity was also demonstrated, by showing that free HER2 receptors on the target cells are required for fusion toxin activity. In mice, the fusion toxins containing the ABD exhibited an appreciably longer time in circulation. The uptake was highest in liver and kidney. Fusion with PE25 was associated with the highest hepatic uptake. Collectively, the results suggest that fusion toxins consisting of ADAPTs and ETA­derivatives are promising agents for targeted cancer therapy.


Subject(s)
ADP Ribose Transferases/administration & dosage , Bacterial Toxins/administration & dosage , Exotoxins/administration & dosage , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/administration & dosage , Virulence Factors/administration & dosage , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , Albumins/administration & dosage , Albumins/chemistry , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Cell Line, Tumor , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/pharmacokinetics , Female , Humans , Mice , Neoplasms/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacokinetics , Protein Binding , Protein Domains , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Surface Plasmon Resonance , Tissue Distribution , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
2.
J Control Release ; 164(1): 58-64, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23075769

ABSTRACT

The use of cell-penetrating peptides (CPPs), such as polyarginine, has been shown to facilitate the import of drugs and other cargos into cells. However, a major obstacle limiting their use as delivery agents is their entrapment following internalization into endocytic vesicles, leading to either their recycling out of cells or their degradation in lysosomes. To address this challenge, we fused a CPP sequence to the translocation domain of Pseudomonas aeruginosa exotoxin A (ETA) to facilitate the endosomal escape of imported CPP-containing protein constructs. Specifically, a fusion protein incorporating ten arginines linked to residues 253 to 412 of ETA (ETA(253-412)) was tested for its ability to effectively route a protein cargo (enhanced green fluorescent protein, eGFP) to the cytosol of cells. Using flow cytometry and fluorescence live-cell imaging, we observed a 5-fold improvement of cellular uptake as well as a 40-fold increase in cytosolic delivery of the CPP-ETA(253-412)-eGFP construct in relation to CPP-eGFP. Furthermore, analysis of intracellular routing events indicated that the incorporation of ETA(253-412) within the CPP-containing protein fusion construct avoided lysosomal degradation by re-directing the construct from early endosomes to the ER lumen and finally to the cytosol. Studies using inhibitors of vesicular transport confirmed that the ER lumen is a key compartment reached by the CPP-ETA(253-412)-eGFP construct before accessing the cytosol. Together, these findings suggest that incorporating a CPP motif and the ETA translocation domain into protein constructs can facilitate their cytosolic delivery.


Subject(s)
ADP Ribose Transferases/administration & dosage , Bacterial Toxins/administration & dosage , Cell-Penetrating Peptides/administration & dosage , Cytosol/metabolism , Drug Carriers/administration & dosage , Exotoxins/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Virulence Factors/administration & dosage , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Cell Culture Techniques , Cell Tracking , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacokinetics , Drug Carriers/pharmacokinetics , Exotoxins/genetics , Exotoxins/pharmacokinetics , Flow Cytometry , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
3.
J Biol Chem ; 287(10): 7367-73, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22228764

ABSTRACT

Members of the type 2 ribosome-inactivating proteins (RIPs) family (e.g. ricin, abrin) are potent cytotoxins showing a strong lethal activity toward eukaryotic cells. Type 2 RIPs contain two polypeptide chains (usually named A, for "activity", and B, for "binding") linked by a disulfide bond. The intoxication of the cell is a consequence of a reductive process in which the toxic domain is cleaved from the binding domain by oxidoreductases located in the lumen of the endoplasmic reticulum (ER). The best known example of type 2 RIPs is ricin. Protein disulfide isomerase (PDI) was demonstrated to be involved in the process of ricin reduction; however, when PDI is depleted from cell fraction preparations ricin reduction can still take place, indicating that also other oxidoreductases might be implicated in this process. We have investigated the role of TMX, a transmembrane thioredoxin-related protein member of the PDI family, in the cell intoxication operated by type 2 RIPs ricin and abrin. Overexpressing TMX in A549 cells resulted in a dramatic increase of ricin or abrin cytotoxicity compared with control mock-treated cells. Conversely, no difference in cytotoxicity was observed after treatment of A549 cells or control cells with saporin or Pseudomonas exotoxin A whose intracellular mechanism of activation is not dependent upon reduction (saporin) or only partially dependent upon it (Pseudomonas exotoxin A). Moreover, the silencing of TMX in the prostatic cell line DU145 reduced the sensitivity of the cells to ricin intoxication further confirming a role for this enzyme in intracellular ricin activation.


Subject(s)
Abrin/pharmacokinetics , Chemical Warfare Agents/pharmacokinetics , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Ricin/pharmacokinetics , Thioredoxins/metabolism , ADP Ribose Transferases/pharmacokinetics , ADP Ribose Transferases/pharmacology , Abrin/pharmacology , Bacterial Toxins/pharmacokinetics , Bacterial Toxins/pharmacology , Chemical Warfare Agents/pharmacology , Endoplasmic Reticulum/genetics , Exotoxins/pharmacokinetics , Exotoxins/pharmacology , Humans , Jurkat Cells , Membrane Proteins/genetics , Oxidation-Reduction/drug effects , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Synthesis Inhibitors/pharmacokinetics , Protein Synthesis Inhibitors/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacokinetics , Ribosome Inactivating Proteins, Type 1/pharmacology , Ricin/pharmacology , Saporins , Thioredoxins/genetics , Virulence Factors/pharmacokinetics , Virulence Factors/pharmacology , Pseudomonas aeruginosa Exotoxin A
4.
Mol Cancer Ther ; 10(10): 1829-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21862685

ABSTRACT

A novel anticancer agent was constructed by fusing a gene encoding the scFV that targets both glycosylated and unglycosylated forms of CD133 to a gene fragment encoding deimmunized PE38KDEL. The resulting fusion protein, dCD133KDEL, was studied to determine its ability to bind and kill tumor-initiating cells in vitro and in vivo. The anti-CD133 scFV selectively bound HEK293 cells transfected with the CD133 receptor gene. Time course viability studies showed that dCD133KDEL selectively inhibited NA-SCC and UMSCC-11B, 2 head and neck squamous cell carcinomas that contain a CD133 expressing subpopulation. Importantly, the drug did not inhibit the viability of hematopoietic lineages measured by long-term culture-initiating cell and colony-forming assays from sorted human CD34+ progenitor cells. In addition to in vitro studies, in vivo tumor initiation experiments confirmed that CD133-sorted cells implanted into the flanks of nude mice grew faster and larger than unsorted cells. In contrast, cells that were pretreated with dCD133KDEL before implantation showed the slowest and lowest incidence of tumors. Furthermore, UMSCC-11B-luc tumors treated with multiple intratumoral injections of dCD133KDEL showed marked growth inhibition, leading to complete degradation of the tumors that was not observed with an irrelevant control-targeted toxin. Experiments in immunocompetent mice showed that toxin deimmunization resulted in a 90% reduction in circulating antitoxin levels. These studies show that dCD133KDEL is a novel anticancer agent effective at inhibiting cell proliferation, tumor initiation, and eliminating established tumors by targeting the CD133 subpopulation. This agent shows significant promise for potential development as a clinically useful therapy.


Subject(s)
Antigens, CD/immunology , Carcinoma, Squamous Cell/drug therapy , Glycoproteins/immunology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Immunotoxins/pharmacology , Neoplastic Stem Cells/drug effects , Peptides/immunology , Recombinant Fusion Proteins/pharmacology , AC133 Antigen , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , ADP Ribose Transferases/pharmacology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Bacterial Toxins/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Exotoxins/genetics , Exotoxins/pharmacokinetics , Exotoxins/pharmacology , Female , Glycoproteins/biosynthesis , Glycoproteins/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/pharmacology , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Peptides/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Transfection , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
5.
FEBS J ; 277(18): 3735-49, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20718861

ABSTRACT

To assess Pseudomonas exotoxin A (ETA) compartmentalization, processing and cytotoxicity in vivo, we have studied the fate of internalized ETA with the use of the in vivo rodent liver model following toxin administration, cell-free hepatic endosomes, and pure in vitro protease assays. ETA taken up into rat liver in vivo was rapidly associated with plasma membranes (5-30 min), internalized within endosomes (15-60 min), and later translocated into the cytosolic compartment (30-90 min). Coincident with endocytosis of intact ETA, in vivo association of the catalytic ETA-A subunit and low molecular mass ETA-A fragments was observed in the endosomal apparatus. After an in vitro proteolytic assay with an endosomal lysate and pure proteases, the ETA-degrading activity was attributed to the luminal species of endosomal acidic cathepsins B and D, with the major cleavages generated in vitro occurring mainly within domain III of ETA-A. Cell-free endosomes preloaded in vivo with ETA intraluminally processed and extraluminally released intact ETA and ETA-A in vitro in a pH-dependent and ATP-dependent manner. Rat hepatic cells underwent in vivo intrinsic apoptosis at a late stage of ETA infection, as assessed by the mitochondrial release of cytochrome c, caspase-9 and caspase-3 activation, and DNA fragmentation. In an in vitro assay, intact ETA induced ADP-ribosylation of EF-2 and mitochondrial release of cytochrome c, with the former effect being efficiently increased by a cathepsin B/cathepsin D pretreatment. The data show a novel processing pathway for internalized ETA, involving cathepsins B and D, resulting in the production of ETA fragments that may participate in cytotoxicity and mitochondrial dysfunction.


Subject(s)
ADP Ribose Transferases/metabolism , Apoptosis/drug effects , Bacterial Toxins/metabolism , Cathepsins/metabolism , Exotoxins/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Pseudomonas aeruginosa/metabolism , Virulence Factors/metabolism , ADP Ribose Transferases/pharmacokinetics , Animals , Bacterial Toxins/pharmacokinetics , Cathepsin B/metabolism , Cathepsin D/metabolism , Endocytosis , Endosomes/enzymology , Endosomes/metabolism , Exotoxins/pharmacokinetics , Hydrolysis , Liver/enzymology , Liver/metabolism , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Mitochondria, Liver/metabolism , Peptide Elongation Factor 2/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley , Time Factors , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
6.
Int J Pharm ; 374(1-2): 145-52, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19446771

ABSTRACT

We previously reported the development of PE38KDEL-loaded anti-HER2 poly(lactic-co-glycolic acid) (PLGA) nanoparticles that bind and internalize in HER2-overexpressing breast cancer cells, enabling potent anti-tumor activity. To overcome the problems associated with the short half-lives of this drug delivery system, we have constructed PE38KDEL-loaded anti-HER2 PEGylated liposomes (PE-HER-liposomes). PE-HER-liposomes were constructed with Fab' of recombinant humanized anti-HER2 monoclonal antibody (anti-HER2 Fab') covalently linked to PEGylated liposomes containing PE38KDEL (PE-liposomes). We attached anti-HER2 Fab' to the terminus of PEG (polyethylene glycol) on PEGylated liposomes. Incorporation of pyridylthiopropionoylamino-PEG-distearoylphosphatidylethanolamine (PDP-PEG-DSPE) into PEGylated liposomes followed by mild thiolysis of the PDP groups resulted in the formation of reactive thiol groups at the periphery of the liposomes. Efficient attachment of maleimide-derivatized anti-HER2 Fab' took place under mild conditions. The characterization of PE-HER-liposomes, such as particle size, was evaluated by dynamic light-scattering detector. The Micro BCA method was used to determine the encapsulation efficiency of PE38KDEL and the quantity of conjugated Fab'. Flow cytometry and confocal microscopy showed that PE-HER-liposomes possessed receptor-specific binding and internalization for HER2-overexpressing SK-BR3 cells. Remarkably, PE-HER-liposomes were more cytotoxic than non-targeted PE-liposomes in HER2-overexpressing breast cancer cells. In conclusion, PE-HER-liposomes could serve as a promising therapeutic candidate for the treatment of HER2-overexpressing breast cancers.


Subject(s)
ADP Ribose Transferases/pharmacokinetics , Bacterial Toxins/pharmacokinetics , Breast Neoplasms/drug therapy , Exotoxins/pharmacokinetics , Virulence Factors/pharmacokinetics , ADP Ribose Transferases/immunology , ADP Ribose Transferases/pharmacology , Bacterial Toxins/immunology , Bacterial Toxins/pharmacology , Breast Neoplasms/immunology , Cell Line, Tumor , Drug Delivery Systems , Exotoxins/immunology , Exotoxins/pharmacology , Female , Flow Cytometry , Half-Life , Humans , Immunoglobulin Fab Fragments/immunology , Liposomes , Particle Size , Polyethylene Glycols/chemistry , Receptor, ErbB-2/immunology , Sulfhydryl Compounds/chemistry , Virulence Factors/immunology , Virulence Factors/pharmacology , Pseudomonas aeruginosa Exotoxin A
7.
Blood ; 113(16): 3792-800, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-18988862

ABSTRACT

Immunotoxins based on Pseudomonas exotoxin A (PE) are promising anticancer agents that combine a variable fragment (Fv) from an antibody to a tumor-associated antigen with a 38-kDa fragment of PE (PE38). The intoxication pathway of PE immunotoxins involves receptor-mediated internalization and trafficking through endosomes/lysosomes, during which the immunotoxin undergoes important proteolytic processing steps but must otherwise remain intact for eventual transport to the cytosol. We have investigated the proteolytic susceptibility of PE38 immunotoxins to lysosomal proteases and found that cleavage clusters within a limited segment of PE38. We subsequently generated mutants containing deletions in this region using HA22, an anti-CD22 Fv-PE38 immunotoxin currently undergoing clinical trials for B-cell malignancies. One mutant, HA22-LR, lacks all identified cleavage sites, is resistant to lysosomal degradation, and retains excellent biologic activity. HA22-LR killed chronic lymphocytic leukemia cells more potently and uniformly than HA22, suggesting that lysosomal protease digestion may limit immunotoxin efficacy unless the susceptible domain is eliminated. Remarkably, mice tolerated doses of HA22-LR at least 10-fold higher than lethal doses of HA22, and these higher doses exhibited markedly enhanced antitumor activity. We conclude that HA22-LR advances the therapeutic efficacy of HA22 by using an approach that may be applicable to other PE-based immunotoxins.


Subject(s)
ADP Ribose Transferases/pharmacology , Antibodies, Monoclonal/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Immunoglobulin Variable Region/pharmacology , Immunotoxins/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Sialic Acid Binding Ig-like Lectin 2 , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , ADP Ribose Transferases/adverse effects , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Bacterial Toxins/adverse effects , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Clinical Trials as Topic , Endosomes/metabolism , Exotoxins/adverse effects , Exotoxins/genetics , Exotoxins/pharmacokinetics , Female , Humans , Immunoglobulin Variable Region/adverse effects , Immunoglobulin Variable Region/genetics , Immunotoxins/adverse effects , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Virulence Factors/adverse effects , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
8.
Basic Clin Pharmacol Toxicol ; 99(6): 398-404, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17169119

ABSTRACT

LHRH-PE40, a recombinant DNA-derived protein composed of LHRH and Pseudomonas aeruginosa exotoxin A, is being developed for the treatment of malignant tumours. This experiment was designed to assess its preclinical safety. Reproductive toxicity studies, pharmacokinetic studies, single- and repeat-dose intraperitoneal or intravenous toxicity studies in mice, rats and monkeys were conducted to assess the toxicity of LHRH-PE40. In intravenous single-dose studies in mice, the LD50 was 731.26 microg/kg and 676.03 microg/kg in male and female mice respectively. In single-dose studies and repeat-dose range-finding studies in rats, dose-limited severe vascular leakage syndromes occurred. In repeat-dose long-term studies, except drug-related vascular leakage syndromes, other drug-related changes included decreased testis weight and testis atrophy. In single-dose and repeat-dose studies in monkeys, dose-limited acute tubular necrosis of the kidneys was the chief finding. In reproductive studies, drug-related changes were decreased food intakes, decreased testis weight and uterus weight, decreased foetus weight and increased foetus mortality, increased maternal and F1 offspring mortality and decreased maternal and F1 offspring body weight. Pharmacokinetic studies showed a similar half-time of distribution and clearance in mice and monkeys. Tissue distribution showed a high concentration in the kidneys and a low concentration in the brain. LHRH-PE40 induced vascular leak syndromes in rats and acute tubular necrosis in monkeys. It also led to testicle atrophy in rats and overt productive toxicity to parents and F1 generations in mice. Because of these findings, it should be monitored carefully in human clinical trials for things such as respiratory, urinary and reproductive toxicities.


Subject(s)
ADP Ribose Transferases/toxicity , Antineoplastic Agents/toxicity , Bacterial Toxins/toxicity , Exotoxins/toxicity , Gonadotropin-Releasing Hormone/toxicity , Recombinant Fusion Proteins/toxicity , Virulence Factors/toxicity , ADP Ribose Transferases/immunology , ADP Ribose Transferases/pharmacokinetics , Animals , Antibodies/blood , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Bacterial Toxins/immunology , Bacterial Toxins/pharmacokinetics , Drug Evaluation, Preclinical , Exotoxins/immunology , Exotoxins/pharmacokinetics , Female , Gonadotropin-Releasing Hormone/immunology , Gonadotropin-Releasing Hormone/pharmacokinetics , Kidney/drug effects , Kidney/pathology , Lung/drug effects , Lung/pathology , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Mice, Nude , Neoplasms/drug therapy , Organ Size/drug effects , Pregnancy , Rats , Rats, Wistar , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Reproduction/drug effects , Testis/drug effects , Testis/growth & development , Testis/pathology , Uterus/drug effects , Uterus/growth & development , Virulence Factors/immunology , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
9.
Clin Cancer Res ; 12(10): 3145-51, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707614

ABSTRACT

PURPOSE: To determine if the tumor-targeted cytotoxin interleukin 13 bound to Pseudomonas exotoxin (IL13-PE) could be delivered to the brainstem safely at therapeutic doses while monitoring its distribution in real-time using a surrogate magnetic resonance imaging tracer, we used convection-enhanced delivery to perfuse rat and primate brainstems with IL13-PE and gadolinium-bound albumin (Gd-albumin). EXPERIMENTAL DESIGN: Thirty rats underwent convective brainstem perfusion of IL13-PE (0.25, 0.5, or 10 microg/mL) or vehicle. Twelve primates underwent convective brainstem perfusion of either IL13-PE (0.25, 0.5, or 10 microg/mL; n = 8), co-infusion of 125I-IL13-PE and Gd-albumin (n = 2), or co-infusion of IL13-PE (0.5 microg/mL) and Gd-albumin (n = 2). The animals were permitted to survive for up to 28 days before sacrifice and histologic assessment. RESULTS: Rats showed no evidence of toxicity at all doses. Primates showed no toxicity at 0.25 or 0.5 microg/mL but showed clinical and histologic toxicity at 10 microg/mL. Quantitative autoradiography confirmed that Gd-albumin precisely tracked IL13-PE anatomic distribution and accurately showed the volume of distribution. CONCLUSIONS: IL13-PE can be delivered safely and effectively to the primate brainstem at therapeutic concentrations and over clinically relevant volumes using convection-enhanced delivery. Moreover, the distribution of IL13-PE can be accurately tracked by co-infusion of Gd-albumin using real-time magnetic resonance imaging.


Subject(s)
ADP Ribose Transferases/pharmacokinetics , Bacterial Toxins/pharmacokinetics , Blood-Brain Barrier , Brain Stem/chemistry , Exotoxins/pharmacokinetics , Interleukin-13/pharmacokinetics , Virulence Factors/pharmacokinetics , ADP Ribose Transferases/adverse effects , Animals , Autoradiography , Bacterial Toxins/adverse effects , Brain Stem Neoplasms/drug therapy , Contrast Media/administration & dosage , Convection , Dose-Response Relationship, Drug , Exotoxins/adverse effects , Gadolinium/administration & dosage , Glioma/drug therapy , Interleukin-13/adverse effects , Macaca mulatta , Macromolecular Substances , Magnetic Resonance Imaging , Male , Rats , Rats, Sprague-Dawley , Serum Albumin/administration & dosage , Virulence Factors/adverse effects , Pseudomonas aeruginosa Exotoxin A
10.
World J Gastroenterol ; 10(19): 2870-3, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15334689

ABSTRACT

AIM: To explore the ability of recombinant toxin luteinizing hormone releasing hormone-Pseudomonas aeruginosa exotoxin 40 (LHRH-PE40) and LHRH binding to LHRH receptor (LHRHR) on the membrane surface of human liver cancer HEPG cells. METHODS: LHRH was labeled by using (125)I with enzymatic reaction. The affinity and receptor volume of LHRH-PE40 and LHRH binding to LHRHR on the membrane surface of human liver cancer cells were measured with radioligand receptor assay. RESULTS: The specific activity of LHRH labeled with (125)I was 2.7 x 10(4) kBq/microL, and its radiochemical purity reached to 99.2-99.7%. The binding of (125)I to LHRH was maximal for 240 min in the warm cultivation, and this binding was stabilized. The inhibiting rates of (125)I-LHRH and LHRH on the proliferation of human liver cancer HEPG cells were not significantly different. On the basis of the saturation curve of (125)I-LHRH binding to the membrane LHRHR of HEPG cells, (125)I-LHRH of 1 x 10(5) cpm was selected for radioligand receptor assay. The affinity constants (Kd) of LHRH-PE40 and LHRH binding to the membrane LHRHR of HEPG cells were 0.43+/-0.12 nmol/L and 4.86+/-1.47 nmol/L, respectively, and their receptor volumes were 0.37+/-0.15 micromol/g and 0.42+/-0.13 micromol/g, respectively. The binding of LHRH-PE40 to the membrane protein of normal liver cells was not observed. CONCLUSION: The recombinant toxin LHRH-PE40 binding to the membrane surface of LHRHR of human liver cancer HEPG cells was very strong, while the specific binding of it to normal liver cells was not observed. The results provide an important experimental basis for the clinical application of LHRH-PE.


Subject(s)
ADP Ribose Transferases/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Bacterial Toxins/pharmacokinetics , Exotoxins/pharmacokinetics , Gonadotropin-Releasing Hormone/metabolism , Liver Neoplasms/metabolism , Receptors, LHRH/metabolism , Virulence Factors/pharmacokinetics , ADP Ribose Transferases/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Binding, Competitive , Cell Division/drug effects , Cell Line, Tumor , Exotoxins/pharmacology , Humans , Kinetics , Liver Neoplasms/pathology , Virulence Factors/pharmacology , Pseudomonas aeruginosa Exotoxin A
11.
Mol Cancer Ther ; 1(2): 79-84, 2001 Dec.
Article in English | MEDLINE | ID: mdl-12467225

ABSTRACT

We have used protein engineering to generate a stable bivalent fragment variable (Fv) molecule from the antimesothelin antibody SS, in which the VH and VL domains of the Fv are linked to each other by a disulfide bond, and the two Fvs are connected by a flexible 15-amino acid (Gly4-Ser)3 linker. The SS (dsFv)2 molecule is fused to a M(r) 38,000 truncated form of Pseudomonas exotoxin to generate a bivalent, disulfide stabilized, (dsFv)2 immunotoxin. The immunotoxin was expressed in Escherichia coli, refolded in vitro, and purified to approximately 95% purity with a high yield of > 10%. Binding studies demonstrated that the (dsFv)2 molecule has 40 times higher apparent affinity for recombinant mesothelin than a monovalent dsFv molecule. The (dsFv)2 immunotoxin was 4-10-fold more cytotoxic to three mesothelin antigen-positive cell lines than the monovalent dsFv immunotoxin. However, when tested in mice bearing tumor cells expressing mesothelin, the antitumor activity of the bivalent immunotoxin is very similar to the activity of the lower affinity monovalent immunotoxin. Our data indicate that increasing affinity of an antibody fragment does not necessarily lead to higher antitumor activity of an immunotoxin in vivo.


Subject(s)
ADP Ribose Transferases/pharmacology , Bacterial Toxins/pharmacology , Disulfides/chemistry , Exotoxins/pharmacology , Immunotoxins/pharmacology , Mesothelioma/immunology , Ovarian Neoplasms/immunology , Virulence Factors/pharmacology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , Animals , Antibodies, Monoclonal , Antigens, Neoplasm/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Escherichia coli/genetics , Escherichia coli/metabolism , Exotoxins/genetics , Exotoxins/pharmacokinetics , Female , GPI-Linked Proteins , Gene Expression , Humans , Immunoglobulin Fragments/immunology , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Membrane Glycoproteins/metabolism , Mesothelin , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Plasmids , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL