Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Bioconjug Chem ; 26(6): 1120-8, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-25997032

ABSTRACT

Recombinant immunotoxins (RITs) are fusion proteins that join antibodies to protein toxins for targeted cell killing. RITs armed with Pseudomonas exotoxin A (PE) are undergoing clinical trials for the treatment of cancer. The current design of PE-based RITs joins an antibody fragment to the catalytic domain of PE using a polypeptide linker that is cleaved by the protease furin. Intracellular cleavage of native PE by furin is required for cytotoxicity, yet the PE cleavage site has been shown to be a poor furin substrate. Here we describe the rational design of more efficiently cleaved furin linkers in PE-based RITs, and experiments evaluating their effects on cleavage and cytotoxicity. We found that changes to the furin site could greatly influence both cleavage and cytotoxicity, but the two parameters were not directly correlated. Furthermore, the effects of alterations to the furin linker were not universal. Identical mutations in the anti-CD22 RIT HA22-LR often displayed different cytotoxicity from mutations in the anti-mesothelin RIT SS1-LR/GGS, underscoring the prominent role of the target site in their intoxication pathways. Combining several beneficial mutations in HA22-LR resulted in a variant (HA22-LR/FUR) with a remarkably enhanced cleavage rate and improved cytotoxicity against five B cell lines and similar or enhanced cytotoxicity in five out of six hairy cell leukemia patient samples. This result informs the design of protease-sensitive linkers and suggests that HA22-LR/FUR may be a candidate for further preclinical development.


Subject(s)
ADP Ribose Transferases/chemistry , ADP Ribose Transferases/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Exotoxins/chemistry , Exotoxins/pharmacology , Furin/metabolism , Immunotoxins/chemistry , Immunotoxins/pharmacology , Virulence Factors/chemistry , Virulence Factors/pharmacology , ADP Ribose Transferases/metabolism , Amino Acid Sequence , Antineoplastic Agents/metabolism , Bacterial Toxins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Exotoxins/metabolism , Humans , Immunotoxins/metabolism , Leukemia/drug therapy , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Virulence Factors/metabolism , Pseudomonas aeruginosa Exotoxin A
2.
Proc Natl Acad Sci U S A ; 109(51): E3597-603, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213206

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins that are being developed for cancer treatment. We have produced RITs that contain PE38, a portion of the bacterial protein Pseudomonas exotoxin A. Because the toxin is bacterial, it often induces neutralizing antibodies, which limit the number of treatment cycles and the effectiveness of the therapy. Because T cells are essential for antibody responses to proteins, we adopted an assay to map the CD4(+) T-cell epitopes in PE38. We incubated peripheral blood mononuclear cells with an immunotoxin to stimulate T-cell expansion, followed by exposure to overlapping peptide fragments of PE38 and an IL-2 ELISpot assay to measure responses. Our observation of T-cell responses in 50 of 50 individuals correlates with the frequency of antibody formation in patients with normal immune systems. We found a single, highly immunodominant epitope in 46% (23/50) of the donors. The immunodominant epitope is DRB1-restricted and was observed in subjects with different HLA alleles, indicating promiscuity. We identified two amino acids that, when deleted or mutated to alanine, eliminated the immunodominant epitope, and we used this information to construct mutant RITs that are highly cytotoxic and do not stimulate T-cell responses in many donors.


Subject(s)
ADP Ribose Transferases/chemistry , Bacterial Toxins/chemistry , Epitopes, T-Lymphocyte/chemistry , Exotoxins/chemistry , Protein Engineering/methods , Virulence Factors/chemistry , Antibodies/chemistry , CD4-Positive T-Lymphocytes/cytology , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/chemistry , Epitopes, T-Lymphocyte/immunology , Gene Deletion , Genetic Variation , Humans , Immune System , Interleukin-2/metabolism , Leukocytes, Mononuclear/cytology , Molecular Conformation , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Pseudomonas aeruginosa Exotoxin A
3.
Proc Natl Acad Sci U S A ; 108(14): 5742-7, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21436054

ABSTRACT

Many nonhuman proteins have useful pharmacological activities, but are infrequently effective in humans because of their high immunogenicity. A recombinant immunotoxin (HA22, CAT8015, moxetumomab pasudotox) composed of an anti-CD22 antibody variable fragment fused to PE38, a 38-kDa portion of Pseudomonas exotoxin A, has produced many complete remissions in drug-resistant hairy-cell leukemia when several cycles of the agent can be given, but has much less activity when antibodies develop. We have pursued a strategy to deimmunize recombinant immunotoxins by identifying and removing B-cell epitopes. We previously reported that we could eliminate most B-cell epitopes using a combination of point mutations and deletions. Here we show the location and amino acid composition of all of the B-cell epitopes in the remaining 25-kDa portion of Pseudomonas exotoxin. Using this information, we eliminated these epitopes to produce an immunotoxin (HA22-LR-8M) that is fully cytotoxic against malignant B-cell lines, has high cytotoxic activity against cells directly isolated from patients with chronic lymphocytic leukemia, and has excellent antitumor activity in mice. HA22-LR-8M does not induce antibody formation in mice when given repeatedly by intravenous injection and does not induce a secondary antibody response when given to mice previously exposed to HA22. HA22-LR-8M also has greatly reduced antigenicity when exposed to sera from patients who have produced antibodies to HA22. The properties of HA22-LR-8M make it an excellent candidate for further clinical development.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Epitopes, B-Lymphocyte/genetics , Exotoxins/metabolism , Immunization, Passive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Models, Molecular , Protein Engineering/methods , Recombinant Fusion Proteins/immunology , Virulence Factors/metabolism , ADP Ribose Transferases/genetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Bacterial Toxins/genetics , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Exotoxins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Statistics, Nonparametric , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
4.
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
5.
BMC Res Notes ; 12(1): 293, 2019 May 27.
Article in English | MEDLINE | ID: mdl-31133049

ABSTRACT

OBJECTIVE: Recombinant immunotoxins (RITs) are antibody-toxin fusion proteins that can selectively eliminate populations of cells expressing specific surface receptors. They are in evaluation as therapeutic agents for cancer. RITs based on Pseudomonas exotoxin A (PE) are in use clinically for the treatment of hairy cell leukemia, and under trial for the treatment of other cancers. In an effort to improve the efficacy of PE-based RITs, we evaluated the potential of combination therapy with several common antibiotics (tetracycline, chloramphenicol, streptomycin, linezolid, fusidic acid, and kanamycin) on human cell lines HEK293, OVCAR8, and CA46. Antibiotics were selected based on their potential to inhibit mitochondrial protein synthesis and disrupt energy metabolism in cancer cells. RESULTS: Tetracycline, chloramphenicol, linezolid, and fusidic acid alone killed cultured human cells at high concentrations. At high but nontoxic concentrations of each antibiotic, only chloramphenicol treatment of the Burkitt's lymphoma cell line CA46 showed enhanced cytotoxicity when paired with an anti-transferrin receptor/PE RIT. This result, however, could not be replicated in additional Burkitt's lymphoma cell lines Ramos and Raji. Although the six antibiotics we tested are not promising candidates for RIT combination therapy, we suggest that fusidic acid could be considered independently as a potential cancer therapeutic.


Subject(s)
ADP Ribose Transferases/pharmacology , Anti-Bacterial Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Fusidic Acid/pharmacology , Immunotoxins/pharmacology , Virulence Factors/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Chloramphenicol/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , HEK293 Cells , Humans , Inhibitory Concentration 50 , Kanamycin/pharmacology , Linezolid/pharmacology , Lymphocytes/drug effects , Lymphocytes/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Streptomycin/pharmacology , Tetracycline , Pseudomonas aeruginosa Exotoxin A
6.
PLoS One ; 13(6): e0198699, 2018.
Article in English | MEDLINE | ID: mdl-29912917

ABSTRACT

Small proteins are a new and expanding area of research. Many characterized small proteins are composed of a single hydrophobic α-helix, and the functional requirements of their limited amino acid sequence are not well understood. One hydrophobic small protein, CydX, has been shown to be a component of the cytochrome bd oxidase complex in Escherichia coli, and is required for enzyme function. To investigate small protein sequence specificity, an alanine scanning mutagenesis on the small protein CydX was conducted using mutant alleles expressed from the E. coli chromosome at the wild-type locus. The resulting mutant strains were assayed for CydX function. No single amino acid was required to maintain wild-type resistance to ß-mercaptoethanol. However, substitutions of 10-amino acid blocks indicated that the N-terminus of the protein was required for wild-type CydX activity. A series of double mutants showed that multiple mutations at the N-terminus led to ß-mercaptoethanol sensitivity in vivo. Triple mutants showed both in vivo and in vitro phenotypes. Together, these data provide evidence suggesting a high level of functional plasticity in CydX, in which multiple amino acids may work cooperatively to facilitate CydX function.


Subject(s)
Cytochromes/genetics , Electron Transport Chain Complex Proteins/genetics , Escherichia coli Proteins/genetics , Oxidoreductases/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Chromosomes, Bacterial/genetics , Cytochrome b Group , Cytochromes/isolation & purification , Cytochromes/metabolism , Cytochromes/physiology , Electron Transport Chain Complex Proteins/isolation & purification , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Chain Complex Proteins/physiology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/physiology , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , Immunoblotting , Mutation/genetics , Oxidoreductases/isolation & purification , Oxidoreductases/metabolism , Oxidoreductases/physiology
7.
Proteins ; 66(3): 646-54, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17173282

ABSTRACT

The arabinose-binding pockets of wild type AraC dimerization domains crystallized in the absence of arabinose are occupied with the side chains of Y31 from neighboring domains. This interaction leads to aggregation at high solution concentrations and prevents determination of the structure of truely apo AraC. In this work we found that the aggregation does not significantly occur at physiological concentrations of AraC. We also found that the Y31V mutation eliminates the self-association, but does not affect regulation properties of the protein. At the same time, the mutation allows crystallization of the dimerization domain of the protein with only solvent in the arabinose-binding pocket. Using a distance difference method suitable for detecting and displaying even minor structural variation among large groups of similar structures, we find that there is no significant structural change in the core of monomers of the AraC dimerization domain resulting from arabinose, fucose, or tyrosine occupancy of the ligand-binding pocket. A slight change is observed in the relative orientation of monomers in the dimeric form of the domain upon the binding of arabinose but its significance cannot yet be assessed.


Subject(s)
AraC Transcription Factor/chemistry , AraC Transcription Factor/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , AraC Transcription Factor/genetics , Arabinose/chemistry , Arabinose/metabolism , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , Dimerization , Escherichia coli Proteins/genetics , Models, Molecular , Operon , Plasmids , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
8.
Protein Sci ; 15(12): 2828-35, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17132863

ABSTRACT

Deletion of the regulatory N-terminal arms of the AraC protein from its dimerization domain fragments increases the susceptibility of the dimerization domain to form a series of higher order polymers by indefinite self-association. We investigated how the normal presence of the arm inhibits this self-association. One possibility is that arms can act as an entropic bristles to interfere with the approach of other macromolecules, thereby decreasing collision frequencies. We examined the repulsive effect of flexible arms by measuring the rate of trypsin cleavage of a specially constructed ubiquitin-arm protein. Adding an arm to ubiquitin or increasing its length produced only a modest repulsive effect. This suggests that arms such as the N-terminal arm of AraC do not reduce self-association by entropic exclusion. We consequently tested the hypothesis that the arm on AraC reduces self-association by binding to the core of the dimerization domain even in the absence of arabinose. The behaviors of dimerization domain mutants containing deletions or alterations in the N-terminal arms substantiate this hypothesis. Apparently, interactions between the N-terminal arm and the dimerization domain core position the arm to interfere with the protein-protein contacts necessary for self-association.


Subject(s)
AraC Transcription Factor/chemistry , AraC Transcription Factor/metabolism , Binding Sites , Dimerization , Entropy , Escherichia coli , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/physiology , Protein Binding , Protein Engineering/methods , Protein Folding , Protein Structure, Tertiary/physiology , Solubility , Trypsin/metabolism
9.
BMC Biochem ; 4: 1, 2003 Feb 19.
Article in English | MEDLINE | ID: mdl-12625842

ABSTRACT

BACKGROUND: Pigment epithelium-derived factor (PEDF) has binding affinity for cell-surface receptors in retinoblastoma cells and for glycosaminoglycans. We investigated the effects of glycosaminoglycans on PEDF-receptor interactions. RESULTS: 125I-PEDF formed complexes with protease-resistant components of medium conditioned by human retinoblastoma Y-79 cells. Using specific glycosaminoglycan degrading enzymes in spectrophotometric assays and PEDF-affinity chromatography, we detected heparin and heparan sulfate-like glycosaminoglycans in the Y-79 conditioned media, which had binding affinity for PEDF. The Y-79 conditioned media significantly enhanced the binding of 125I-PEDF to Y-79 cell-surface receptors. However, enzymatic and chemical depletion of sulfated glycosaminoglycans from the Y-79 cell cultures by heparitinase and chlorate treatments decreased the degree of 125I-PEDF binding to cell-surface receptors. CONCLUSIONS: These data indicate that retinoblastoma cells secrete heparin/heparan sulfate with binding affinity for PEDF, which may be important in efficient cell-surface receptor binding.


Subject(s)
Glycosaminoglycans/pharmacology , Heparitin Sulfate/metabolism , Receptors, Neuropeptide/metabolism , Retinal Neoplasms/chemistry , Retinal Neoplasms/pathology , Retinoblastoma/chemistry , Retinoblastoma/pathology , Chlorates/pharmacology , Culture Media, Conditioned/chemistry , Glycosaminoglycans/metabolism , Heparin/metabolism , Humans , Iodine Radioisotopes/metabolism , Iodine Radioisotopes/pharmacology , Lyases/pharmacology , Pigment Epithelium of Eye/drug effects , Receptors, Cell Surface/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Tumor Cells, Cultured
10.
Mol Cancer Ther ; 12(1): 48-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23136186

ABSTRACT

SS1P is a recombinant immunotoxin (RIT) engineered for the targeted elimination of malignant cells that express the tumor-associated antigen mesothelin. It is composed of an antimesothelin antibody variable fragment (Fv) linked to a cytotoxic fragment of Pseudomonas exotoxin A (PE) that includes domains II and III of native PE. The clinical use of SS1P is limited by its propensity to induce neutralizing antibodies and to cause a dose-limiting capillary leak syndrome (CLS) in patients. In this article, we describe a reengineered SS1P with improved properties that overcome these deficits. The redesign of SS1P consists of (i) removing the bulk of PE domain II (residues 251-273 and 284-394 of native PE), leaving only an 11-residue furin cleavage site, (ii) adding a Gly-Gly-Ser peptide linker after the furin cleavage site, and (iii) replacing eight highly solvent-exposed residues in the catalytic domain of PE. The new molecule, SS1-LR/GGS/8M, has cytotoxic activity comparable with SS1P on several mesothelin-expressing cell lines and remarkably improved activity on primary cells from patients with mesothelioma. In a mouse xenograft tumor model, high doses of SS1-LR/GGS/8M elicit antitumor activity superior to the activity of SS1P at its maximum-tolerated dose. In addition, SS1-LR/GGS/8M has greatly decreased ability to cause CLS in a rat model and reduced antigenicity or reactivity with antibodies to the sera of patients previously treated with SS1P.


Subject(s)
Antineoplastic Agents/pharmacology , GPI-Linked Proteins/metabolism , Mesothelioma/drug therapy , Recombinant Fusion Proteins/pharmacology , Amino Acid Substitution , Animals , Antigens, Neoplasm , Antineoplastic Agents/adverse effects , Antineoplastic Agents/immunology , Capillary Leak Syndrome/chemically induced , Cell Line, Tumor , Female , GPI-Linked Proteins/immunology , Half-Life , Humans , Inhibitory Concentration 50 , Maximum Tolerated Dose , Mesothelin , Mesothelioma/pathology , Mice , Mice, Nude , Mutagenesis, Site-Directed , Rats , Rats, Wistar , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Protein Eng Des Sel ; 25(1): 1-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22101015

ABSTRACT

Recombinant immunotoxins (RITs) are anti-cancer agents that combine the Fv of an antibody against cancer cells with a protein toxin from bacteria or plants. Since RITs contain a non-human protein, immunogenicity can be an obstacle in their development. In this study, we have explored the hypothesis that increasing stability can reduce the immunogenicity of a RIT using HA22-LR, which is composed of an anti-CD22 Fv fused to domain III of Pseudomonas exotoxin A. We introduced a disulfide bond into domain III by identifying and mutating two structurally adjacent residues to cysteines at sites suggested by computer modeling. This RIT, HA22-LR-DB, displays a remarkable increase in thermal stability and an enhanced resistance to trypsin degradation. In addition, HA22-LR-DB retains cytotoxic and anti-tumor activity, while exhibiting significantly lower immunogenicity in mice. This study demonstrates that it is possible to design mutations in a protein molecule that will increase the stability of the protein and thereby reduce its immunogenicity.


Subject(s)
Disulfides/immunology , Immunotoxins/immunology , Protein Engineering/methods , Recombinant Proteins/immunology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , ADP Ribose Transferases/pharmacology , Animals , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Disulfides/chemistry , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Exotoxins/genetics , Exotoxins/immunology , Exotoxins/pharmacology , Female , Hot Temperature , Humans , Immunotoxins/genetics , Immunotoxins/pharmacology , Lymphoma/drug therapy , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Mutation , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Time Factors , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
12.
FEBS J ; 278(23): 4683-700, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21585657

ABSTRACT

Pseudomonas exotoxin A (PE) is a highly toxic protein secreted by the opportunistic pathogen Pseudomonas aeruginosa. The modular structure and corresponding mechanism of action of PE make it amenable to extensive modifications that can redirect its potent cytotoxicity from disease to a therapeutic function. In combination with a variety of artificial targeting elements, such as receptor ligands and antibody fragments, PE becomes a selective agent for the elimination of specific cell populations. This review summarizes our current understanding of PE, its intoxication pathway, and the ongoing efforts to convert this toxin into a treatment for cancer.


Subject(s)
ADP Ribose Transferases/chemistry , ADP Ribose Transferases/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Exotoxins/chemistry , Exotoxins/immunology , Immunotoxins/chemistry , Immunotoxins/immunology , Neoplasms/therapy , Virulence Factors/chemistry , Virulence Factors/immunology , ADP Ribose Transferases/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Humans , Immunotoxins/pharmacology , Neoplasms/immunology , Protein Conformation , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Virulence Factors/pharmacology , Pseudomonas aeruginosa Exotoxin A
13.
J Immunother ; 33(3): 297-304, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20445350

ABSTRACT

Recombinant immunotoxins (RITs) are genetically engineered proteins designed to kill cancer cells. The RIT HA22 contains the Fv portion of an anti-CD22 antibody fused to a 38 kDa fragment of Pseudomonas exotoxin A (PE38). As PE38 is a bacterial protein, patients frequently produce antibodies that neutralize its activity, preventing retreatment. We have earlier shown in mice that PE38 contains 7 major B-cell epitopes located in domains II and III of the protein. Here we present a new mutant RIT, HA22-LR-6X, in which we removed most B-cell epitopes by deleting domain II and mutating 6 residues in domain III. HA22-LR-6X is cytotoxic to several lymphoma cell lines, has very low nonspecific toxicity, and retains potent antitumor activity in mice with CA46 lymphomas. To assess its immunogenicity, we immunized 3 MHC-divergent strains of mice with 5 microg doses of HA22-LR-6X, and found that HA22-LR-6X elicited significantly lower antibody responses than HA22 or other mutant RITs with fewer epitopes removed. Furthermore, large (50 microg) doses of HA22-LR-6X induced markedly lower antibody responses than 5 microg of HA22, indicating that high doses can be administered with low immunogenicity. Our experiments show that we have correctly identified and removed B-cell epitopes from PE38, producing a highly active immunotoxin with low immunogenicity and low animal toxicity. Future studies will determine if these properties carry over to humans with cancer.


Subject(s)
Burkitt Lymphoma/immunology , Immunotoxins/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Antibodies/blood , Antibodies/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Burkitt Lymphoma/pathology , Burkitt Lymphoma/prevention & control , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Exotoxins/genetics , Exotoxins/immunology , Female , Humans , Immunization/methods , Immunotoxins/administration & dosage , Immunotoxins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Mice, SCID , Recombinant Proteins/immunology , Species Specificity , Virulence Factors/genetics , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
14.
J Biol Chem ; 283(48): 33310-20, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-18805795

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a multifunctional serpin with antitumorigenic, antimetastatic, and differentiating activities. PEDF is found within tissues rich in the glycosaminoglycan hyaluronan (HA), and its amino acid sequence contains putative HA-binding motifs. We show that PEDF coprecipitation with glycosaminoglycans in media conditioned by human retinoblastoma Y-79 cells decreased after pretreatments with hyaluronidase, implying an association between HA and PEDF. Direct binding of human recombinant PEDF to highly purified HA was demonstrated by coprecipitation in the presence of cetylpyridinium chloride. Binding of PEDF to HA was concentration-dependent and saturable. The PEDF-HA interactions were sensitive to increasing NaCl concentrations, indicating an ionic nature of these interactions and having affinity higher than PEDF-heparin. Competition assays showed that PEDF can bind heparin and HA simultaneously. PEDF chemically modified with fluorescein retained the capacity for interacting with HA but lacked heparin affinity, suggesting one or more distinct HA-binding regions on PEDF. The HA-binding region was examined by site-directed mutagenesis. Single-point and cumulative alterations at basic residues within the putative HA-binding motif K189A/K191A/R194A/K197A drastically reduced the HA-binding activity without affecting heparin- or collagen I binding of PEDF. Cumulative alterations at sites critical for heparin binding (K146A/K147A/R149A) decreased HA affinity but not collagen I binding. Thus these clusters of basic residues (BXBXXBXXB and BX3AB2XB motifs) in PEDF are functional regions for binding HA. In the spatial PEDF structure they are located in distinct areas away from the collagen-binding site. The HA-binding activity of PEDF may contribute to deposition in the extracellular matrix and to its reported antitumor/antimetastatic effects.


Subject(s)
Extracellular Matrix/chemistry , Eye Proteins/chemistry , Hyaluronic Acid/chemistry , Nerve Growth Factors/chemistry , Peptide Mapping , Serpins/chemistry , Amino Acid Motifs/physiology , Amino Acid Substitution , Animals , Cell Line, Tumor , Cricetinae , Extracellular Matrix/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Heparin/chemistry , Heparin/metabolism , Humans , Hyaluronic Acid/metabolism , Mutagenesis, Site-Directed , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Organ Specificity/physiology , Protein Binding/physiology , Rats , Serpins/genetics , Serpins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL