Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.201
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(13): e23759, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38949635

ABSTRACT

The epidermal growth factor receptor (EGFR) is an important target for cancer therapies. Many head and neck cancer (HNC) cells have been reported to overexpress EGFR; therefore, anti-EGFR therapies have been attempted in patients with HNC. However, its clinical efficacy is limited owing to the development of drug resistance. In this study, we developed an EGFR-targeting immunotoxin consisting of a clinically proven anti-EGFR IgG (cetuximab; CTX) and a toxin fragment (LR-LO10) derived from Pseudomonas exotoxin A (PE) using a novel site-specific conjugation technology (peptide-directed photo-crosslinking reaction), as an alternative option. The immunotoxin (CTX-LR-LO10) showed specific binding to EGFR and properties of a typical IgG, such as stability, interactions with receptors of immune cells, and pharmacokinetics, and inhibited protein synthesis via modification of elongation factor-2. Treatment of EGFR-positive HNC cells with the immunotoxin resulted in apoptotic cell death and the inhibition of cell migration and invasion. The efficacy of CTX-LR-LO10 was evaluated in xenograft mouse models, and the immunotoxin exhibited much stronger tumor suppression than CTX or LR-LO10. Transcriptome analyses revealed that the immunotoxins elicited immune responses and altered the expression of genes related to its mechanisms of action. These results support the notion that CTX-LR-LO10 may serve as a new therapeutic agent targeting EGFR-positive cancers.


Subject(s)
ADP Ribose Transferases , ErbB Receptors , Exotoxins , Head and Neck Neoplasms , Immunoglobulin G , Immunotoxins , Pseudomonas aeruginosa Exotoxin A , Virulence Factors , Humans , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , ErbB Receptors/immunology , Animals , Immunotoxins/pharmacology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Mice , Immunoglobulin G/pharmacology , Cell Line, Tumor , Exotoxins/pharmacology , Xenograft Model Antitumor Assays , Cetuximab/pharmacology , Mice, Nude , Bacterial Toxins , Apoptosis/drug effects , Mice, Inbred BALB C , Female , Cell Movement/drug effects , Antineoplastic Agents/pharmacology
2.
Proc Natl Acad Sci U S A ; 119(24): e2200200119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35675429

ABSTRACT

The human transferrin receptor (TFR) is overexpressed in most breast cancers, including preneoplastic ductal carcinoma in situ (DCIS). HB21(Fv)-PE40 is a single-chain immunotoxin (IT) engineered by fusing the variable region of a monoclonal antibody (HB21) against a TFR with a 40 kDa fragment of Pseudomonas exotoxin (PE). In humans, the administration of other TFR-targeted immunotoxins intrathecally led to inflammation and vascular leakage. We proposed that for treatment of DCIS, intraductal (i.duc) injection of HB21(Fv)-PE40 could avoid systemic toxicity while retaining its potent antitumor effects on visible and occult tumors in the entire ductal tree. Pharmacokinetic studies in mice showed that, in contrast to intravenous injection, IT was undetectable by enzyme-linked immunosorbent assay in blood following i.duc injection of up to 3.0 µg HB21(Fv)-PE40. We demonstrated the antitumor efficacy of HB21(Fv)-PE40 in two mammary-in-duct (MIND) models, MCF7 and SUM225, grown in NOD/SCID/gamma mice. Tumors were undetectable by In Vivo Imaging System (IVIS) imaging in intraductally treated mice within 1 wk of initiation of the regimen (IT once weekly/3 wk, 1.5 µg/teat). MCF7 tumor-bearing mice remained tumor free for up to 60 d of observation with i.duc IT, whereas the HB21 antibody alone or intraperitoneal IT treatment had minimal/no antitumor effects. These and similar findings in the SUM225 MIND model were substantiated by analysis of mammary gland whole mounts, histology, and immunohistochemistry for the proteins Ki67, CD31, CD71 (TFR), and Ku80. This study provides a strong preclinical foundation for conducting feasibility and safety trials in patients with stage 0 breast cancer.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Exotoxins , Immunotoxins , Molecular Targeted Therapy , Receptors, Transferrin , Virulence Factors , ADP Ribose Transferases/administration & dosage , ADP Ribose Transferases/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Bacterial Toxins/administration & dosage , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/therapy , Exotoxins/administration & dosage , Female , Humans , Immunotoxins/administration & dosage , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Transferrin/metabolism , Virulence Factors/administration & dosage , Pseudomonas aeruginosa Exotoxin A
3.
J Chem Inf Model ; 63(5): 1578-1591, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36802593

ABSTRACT

Exotoxin A (ETA) is an extracellular secreted toxin and a single-chain polypeptide with A and B fragments that is produced by Pseudomonas aeruginosa. It catalyzes the ADP-ribosylation of a post-translationally modified histidine (diphthamide) on eukaryotic elongation factor 2 (eEF2), which results in the inactivation of the latter and the inhibition of protein biosynthesis. Studies show that the imidazole ring of diphthamide plays an important role in the ADP-ribosylation catalyzed by the toxin. In this work, we employ different in silico molecular dynamics (MD) simulation approaches to understand the role of diphthamide versus unmodified histidine in eEF2 on the interaction with ETA. Crystal structures of the eEF2-ETA complexes with three different ligands NAD+, ADP-ribose, and ßTAD were selected and compared in the diphthamide and histidine containing systems. The study shows that NAD+ bound to ETA remains very stable in comparison with other ligands, enabling the transfer of ADP-ribose to the N3 atom of the diphthamide imidazole ring in eEF2 during ribosylation. We also show that unmodified histidine in eEF2 has a negative impact on ETA binding and is not a suitable target for the attachment of ADP-ribose. Analyzing of radius of gyration and COM distances for NAD+, ßTAD, and ADP-ribose complexes revealed that unmodified His affects the structure and destabilizes the complex with all different ligands throughout the MD simulations.


Subject(s)
Histidine , Molecular Dynamics Simulation , Peptide Elongation Factor 2/chemistry , Histidine/chemistry , NAD/metabolism , Adenosine Diphosphate Ribose/metabolism , Pseudomonas aeruginosa , Pseudomonas aeruginosa Exotoxin A
4.
J Invertebr Pathol ; 197: 107884, 2023 03.
Article in English | MEDLINE | ID: mdl-36642365

ABSTRACT

The cellular immune response of the greater wax moth Galleria mellonella to Pseudomonas aeruginosa exotoxin A was investigated for the first time. The insects were challenged with a sublethal dose of exoA, and then hemocyte parameters were assessed. The analysis showed a statistically significant decrease in the total hemocyte count (THC), which was associated with significant decreases in the number of granulocytes and plasmatocytes. In turn, no statistically significant changes were observed in the number of spherulocytes and oenocytoides. Fluorescent staining indicated that cells collected from the exoA-challenged larvae exhibited features characteristic for apoptotic and autophagic cell death, e.g. cytoplasm vacuolization and chromatin condensation. The flow cytometry analysis revealed a significant increase in the number of phosphatidylserine- and active caspase 3-positive hemocytes challenged with exoA, which proved apoptosis induction. Our results will help in understanding the role of exotoxin A during P. aeruginosa infections not only in insects but also in mammals, including humans.


Subject(s)
Hemocytes , Moths , Humans , Animals , Virulence Factors , Larva , Insecta , Apoptosis , Pseudomonas aeruginosa , Mammals , Pseudomonas aeruginosa Exotoxin A
5.
J Invertebr Pathol ; 187: 107706, 2022 01.
Article in English | MEDLINE | ID: mdl-34919944

ABSTRACT

The role of Pseudomonas aeruginosa exotoxin A in the modulation of humoral immune response parameters in the hemolymph of Galleria mellonella larvae was investigated. Our results indicate that exoA can play a role of a virulence factor by inhibiting insect PO, lysozyme, and antibacterial activity and decreasing the apoLp-III protein level significantly. No peptide bands with molecular mass below 6.5 kDa were detected in the hemolymph of exoA-treated larvae. We provided evidence for involvement of exoA in the pathogenicity of P. aeruginosa against G. mellonella and the usefulness of the insect as a model for analysis of P. aeruginosa toxins.


Subject(s)
Moths , ADP Ribose Transferases , Animals , Bacterial Toxins , Exotoxins , Hemolymph , Host-Pathogen Interactions , Immunity , Larva , Pseudomonas aeruginosa , Virulence Factors , Pseudomonas aeruginosa Exotoxin A
6.
Proc Natl Acad Sci U S A ; 116(10): 4575-4582, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30760587

ABSTRACT

The tumor microenvironment plays a critical role in controlling tumor progression and immune surveillance. We produced an immunotoxin (2E4-PE38) that kills mouse cells expressing CD25 by attaching the Fv portion of monoclonal antibody 2E4 (anti-mouse CD25) to a 38-kDa portion of Pseudomonas exotoxin A. We employed three mouse cancer tumor models (AB1 mesothelioma, 66c14 breast cancer, and CT26M colon cancer). Tumors were implanted at two sites on BALB/c mice. On days 5 and 9, one tumor was directly injected with 2E4-PE38, and the other was not treated; 2E4-PE38 produced complete regressions of 85% of injected AB1 tumors, 100% of 66c14 tumors, and 100% of CT26M tumors. It also produced complete regressions of 77% of uninjected AB1 tumors, 47% of 66c14 tumors, and 92% of CT26M tumors. Mice with complete regressions of 66c14 tumors were immune to rechallenge with 66c14 cells. Mice with complete regressions of AB1 or CT26M tumors developed cross-tumor immunity rejecting both tumor types. Injection of anti-CD25 antibody or a mutant inactive immunotoxin were generally ineffective. Tumors were analyzed 3 days after 2E4-PE38 injection. The number of regulatory T cells (Tregs) was significantly reduced in the injected tumor but not in the spleen. Injected tumors contained an increase in CD8 T cells expressing IFN-γ, the activation markers CD69 and CD25, and macrophages and conventional dendritic cells. Treatment with antibodies to CD8 abolished the antitumor effect. Selective depletion of Tregs in tumors facilitates the development of a CD8 T cell-dependent antitumor effect in three mouse models.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Immunotoxins/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , ADP Ribose Transferases/immunology , Animals , Bacterial Toxins/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Exotoxins/immunology , Female , Humans , Immunity/drug effects , Interferon-gamma/immunology , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/cytology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
7.
Hepatology ; 71(5): 1696-1711, 2020 05.
Article in English | MEDLINE | ID: mdl-31520528

ABSTRACT

BACKGROUND AND AIMS: Treatment of hepatocellular carcinomas using our glypican-3 (GPC3)-targeting human nanobody (HN3) immunotoxins causes potent tumor regression by blocking protein synthesis and down-regulating the Wnt signaling pathway. However, immunogenicity and a short serum half-life may limit the ability of immunotoxins to transition to the clinic. APPROACH AND RESULTS: To address these concerns, we engineered HN3-based immunotoxins to contain various deimmunized Pseudomonas exotoxin (PE) domains. This included HN3-T20, which was modified to remove T-cell epitopes and contains a PE domain II truncation. We compared them to our previously reported B-cell deimmunized immunotoxin (HN3-mPE24) and our original HN3-immunotoxin with a wild-type PE domain (HN3-PE38). All of our immunotoxins displayed high affinity to human GPC3, with HN3-T20 having a KD value of 7.4 nM. HN3-T20 retained 73% enzymatic activity when compared with the wild-type immunotoxin in an adenosine diphosphate-ribosylation assay. Interestingly, a real-time cell growth inhibition assay demonstrated that a single dose of HN3-T20 at 62.5 ng/mL (1.6 nM) was capable of inhibiting nearly all cell proliferation during the 10-day experiment. To enhance HN3-T20's serum retention, we tested the effect of adding a streptococcal albumin-binding domain (ABD) and a llama single-domain antibody fragment specific for mouse and human serum albumin. For the detection of immunotoxin in mouse serum, we developed a highly sensitive enzyme-linked immunosorbent assay and found that HN3-ABD-T20 had a 45-fold higher serum half-life than HN3-T20 (326 minutes vs. 7.3 minutes); consequently, addition of an ABD resulted in HN3-ABD-T20-mediated tumor regression at 1 mg/kg. CONCLUSION: These data indicate that ABD-containing deimmunized HN3-T20 immunotoxins are high-potency therapeutics ready to be evaluated in clinical trials for the treatment of liver cancer.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Carcinoma, Hepatocellular/therapy , Exotoxins/therapeutic use , Glypicans/antagonists & inhibitors , Immunotoxins/therapeutic use , Liver Neoplasms/therapy , Single-Domain Antibodies/therapeutic use , Virulence Factors/therapeutic use , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/pharmacology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Cell Line, Tumor , Exotoxins/chemistry , Exotoxins/pharmacology , Humans , Immunotoxins/chemistry , Immunotoxins/pharmacology , Mice , Mice, Nude , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Virulence Factors/chemistry , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
8.
Mol Pharm ; 18(6): 2285-2297, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33998814

ABSTRACT

Cholecystokinin-2 receptor (CCK2R) has been proven to be a specific biomarker for colorectal malignancies. Immunotoxins are a valuable class of immunotherapy agents consisting of a targeting element and a bacterial or plant toxin. Previous work demonstrated that targeting CCK2R is a good therapeutic strategy for the treatment of colorectal cancer (CRC). In the present study, we developed a new version of CCK2R-targeting immunotoxin GD9P using a targeted peptide, GD9, as the binding motif and a truncated Pseudomonas exotoxin A (PE38) as the cytokiller. BALB/c nude mice were treated with different doses of GD9P, and pharmacodynamics, pharmacokinetic, and toxicological data were obtained throughout this study. Compared to the parental immunotoxin rCCK8PE38, GD9P exhibited about 1.5-fold yield, higher fluorescence intensity, and increased antitumor activity against human CRC in vitro and in vivo. The IC50 values of GD9P in vitro ranged from 1.61 to 4.55 nM. Pharmacokinetic studies were conducted in mice with a T1/2 of 69.315 min. When tumor-bearing nude mice were treated with GD9P at doses ≥2 mg/kg for five doses, a rapid shrinkage in tumor volume and, in some cases, complete remission was observed. A preliminary safety evaluation demonstrated a good safety profile of GD9P as a Pseudomonas exotoxin A-based immunotherapy. The therapy in combination with oxaliplatin can increase the antitumor efficacy and reduce the toxic side effects caused by chemotherapy. In conclusion, the data support the use of GD9P as a promising immunotherapy targeting CCK2R-expressing colorectal malignancies.


Subject(s)
ADP Ribose Transferases/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Colorectal Neoplasms/drug therapy , Exotoxins/pharmacology , Receptor, Cholecystokinin B/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Virulence Factors/pharmacology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Bacterial Toxins/genetics , Bacterial Toxins/therapeutic use , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Exotoxins/genetics , Exotoxins/therapeutic use , Humans , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Tissue Distribution , Toxicity Tests, Acute , Virulence Factors/genetics , Virulence Factors/therapeutic use , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
9.
Microb Cell Fact ; 20(1): 104, 2021 May 24.
Article in English | MEDLINE | ID: mdl-34030723

ABSTRACT

Protein Glycan Coupling Technology (PGCT) uses purposely modified bacterial cells to produce recombinant glycoconjugate vaccines. This vaccine platform holds great potential in this context, namely due to its modular nature, the simplified production process in comparison to traditional chemical conjugation methods, and its amenability to scaled-up operations. As a result, a considerable reduction in production time and cost is expected, making PGCT-made vaccines a suitable vaccine technology for low-middle income countries, where vaccine coverage remains predominantly low and inconsistent. This work aims to develop an integrated whole-process automated platform for the screening of PGCT-made glycoconjugate vaccine candidates. The successful translation of a bench scale process for glycoconjugate production to a microscale automated setting was achieved. This was integrated with a numerical computational software that allowed hands-free operation and a platform adaptable to biological variation over the course of a production process. Platform robustness was proven with both technical and biological replicates and subsequently the platform was used to screen for the most favourable conditions for production of a pneumococcal serotype 4 vaccine candidate. This work establishes an effective automated platform that enabled the identification of the most suitable E. coli strain and genetic constructs to be used in ongoing early phase research and be further brought into preclinical trials.


Subject(s)
ADP Ribose Transferases/metabolism , Automation/methods , Bacterial Toxins/metabolism , Biotechnology/methods , Escherichia coli/metabolism , Exotoxins/metabolism , High-Throughput Screening Assays/methods , Polysaccharides, Bacterial/metabolism , Vaccines, Conjugate/biosynthesis , Virulence Factors/metabolism , Bacterial Vaccines/biosynthesis , Glycosylation , Humans , Pneumococcal Vaccines/biosynthesis , Technology, Pharmaceutical/methods , Pseudomonas aeruginosa Exotoxin A
10.
BMC Infect Dis ; 21(1): 300, 2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33761869

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa is the leading cause of nosocomial infections, especially in people with a compromised immune system. Targeting virulence factors by neutralizing antibodies is a novel paradigm for the treatment of antibiotic-resistant pseudomonas infections. In this respect, exotoxin A is one of the most potent virulence factors in P. aeruginosa. The present study was carried out to identify a novel human scFv antibody against the P. aeruginosa exotoxin A domain I (ExoA-DI) from a human scFv phage library. METHODS: The recombinant ExoA-DI of P. aeruginosa was expressed in E. coli, purified by Ni-NTA column, and used for screening of human antibody phage library. A novel screening procedure was conducted to prevent the elimination of rare specific clones. The phage clone with high reactivity was evaluated by ELISA and western blot. RESULTS: Based on the results of polyclonal phage ELISA, the fifth round of biopanning leads to the isolation of several ExoA-DI reactive clones. One positive clone with high affinity was selected by monoclonal phage ELISA and used for antibody expression. The purified scFv showed high reactivity with the recombinant domain I and full-length native exotoxin A. CONCLUSIONS: The purified anti-exotoxin A scFv displayed high specificity against exotoxin A. The human scFv identified in this study could be the groundwork for developing a novel therapeutic agent to control P. aeruginosa infections.


Subject(s)
ADP Ribose Transferases/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Pseudomonas aeruginosa/immunology , Single-Chain Antibodies/immunology , Virulence Factors/immunology , ADP Ribose Transferases/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Bacterial Toxins/genetics , Escherichia coli/genetics , Exotoxins/genetics , Humans , Peptide Library , Pseudomonas aeruginosa/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/isolation & purification , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
11.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204265

ABSTRACT

Human epidermal growth factor receptor 2 (HER-2) is overexpressed in many malignant tumors. The anti-HER2 antibody trastuzumab has been approved for treating HER2-positive early and metastatic breast cancers. Pseudomonas exotoxin A (PE), a bacterial toxin of Pseudomonas aeruginosa, consists of an A-domain with enzymatic activity and a B-domain with cell binding activity. Recombinant immunotoxins comprising the HER2(scFv) single-chain Fv from trastuzumab and the PE24B catalytic fragment of PE display promising cytotoxic effects, but immunotoxins are typically insoluble when expressed in the cytoplasm of Escherichia coli, and thus they require solubilization and refolding. Herein, a recombinant immunotoxin gene was fused with maltose binding protein (MBP) and overexpressed in a soluble form in E. coli. Removal of the MBP yielded stable HER2(scFv)-PE24B at 91% purity; 0.25 mg of pure HER2(scFv)-PE24B was obtained from a 500 mL flask culture. Purified HER2(scFv)-PE24B was tested against four breast cancer cell lines differing in their surface HER2 level. The immunotoxin showed stronger cytotoxicity than HER2(scFv) or PE24B alone. The IC50 values for HER2(scFv)-PE24B were 28.1 ± 2.5 pM (n = 9) and 19 ± 1.4 pM (n = 9) for high HER2-positive cell lines SKBR3 and BT-474, respectively, but its cytotoxicity was lower against MDA-MB-231 and MCF7. Thus, fusion with MBP can facilitate the soluble expression and purification of scFv immunotoxins.


Subject(s)
ADP Ribose Transferases , Antineoplastic Agents, Immunological/pharmacology , Bacterial Toxins , Exotoxins , Immunotoxins/pharmacology , Maltose-Binding Proteins , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies , Virulence Factors , ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Cell Line, Tumor , Cell Survival , Dose-Response Relationship, Drug , Escherichia coli/genetics , Escherichia coli/metabolism , Exotoxins/genetics , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Humans , Immunotoxins/genetics , Immunotoxins/isolation & purification , Maltose-Binding Proteins/genetics , Mass Spectrometry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Single-Chain Antibodies/genetics , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
12.
J Cell Physiol ; 235(4): 3711-3720, 2020 04.
Article in English | MEDLINE | ID: mdl-31578716

ABSTRACT

Bacterial toxins have received a great deal of attention in the development of antitumor agents. Currently, these protein toxins were used in the immunotoxins as a cancer therapy strategy. Despite the successful use of immunotoxins, immunotherapy strategies are still expensive and limited to hematologic malignancies. In the current study, for the first time, a nano-toxin comprised of truncated pseudomonas exotoxin (PE38) loaded silver nanoparticles (AgNPs) were prepared and their cytotoxicity effect was investigated on human breast cancer cells. The PE38 protein was cloned into pET28a and expressed in Escherichia coli, BL21 (DE3), and purified using metal affinity chromatography and was analyzed by 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis. AgNPs were biologically prepared using cell-free supernatant of E. Coli K12 strain. Nanoparticle formation was characterized by energy dispersive spectroscopy, transmission electron microscopy, and dynamic light scattering. The PE38 protein was loaded on AgNPs and prepared the PE38-AgNPs nano-toxin. Additionally, in vitro release indicated a partial slow release of toxin in about 100 hr. The nano-toxin exhibited dose-dependent cytotoxicity on MCF-7 cells. Also, real-time polymerase chain reaction results demonstrated the ability of nano-toxin to upregulate Bax/Bcl-2 ratio and caspase-3, -8, -9, and P53 apoptotic genes in the MCF-7 tumor cells. Apoptosis induction was determined by Annexin-V/propidium flow cytometry and caspases activity assay after treatment of cancer cells with the nano-toxin. In general, in the current study, the nano-toxin exhibit an inhibitory effect on the viability of breast cancer cells through apoptosis, which suggests that AgNPs could be used as a delivery system for targeting of toxins to cancer cells.


Subject(s)
ADP Ribose Transferases/pharmacology , Bacterial Toxins/pharmacology , Breast Neoplasms/drug therapy , Cytotoxins/pharmacokinetics , Exotoxins/pharmacology , Metal Nanoparticles/chemistry , Virulence Factors/pharmacology , ADP Ribose Transferases/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Bacterial Toxins/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Caspase 3/genetics , Caspases/genetics , Cell Proliferation/drug effects , Cytotoxins/chemistry , Escherichia coli/genetics , Exotoxins/chemistry , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Microscopy, Electron, Transmission , Proto-Oncogene Proteins c-bcl-2/genetics , Silver/chemistry , Silver/pharmacology , Virulence Factors/chemistry , bcl-2-Associated X Protein/genetics , Pseudomonas aeruginosa Exotoxin A
13.
Cancer Sci ; 111(12): 4548-4557, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33058342

ABSTRACT

Drug resistance represents an obstacle in colorectal cancer (CRC) treatment because of its association with poor prognosis. rBC2LCN is a lectin isolated from Burkholderia that binds cell surface glycans that have fucose moieties. Because fucosylation is enhanced in many types of cancers, this lectin could be an efficient drug carrier if CRC cells specifically present such glycans. Therefore, we examined the therapeutic efficacy and toxicity of lectin drug conjugate therapy in CRC mouse xenograft models. The affinity of rBC2LCN for human CRC cell lines HT-29, LoVo, LS174T, and DLD-1 was assessed in vitro. The cytocidal efficacy of a lectin drug conjugate, rBC2LCN-38 kDa domain of pseudomonas exotoxin A (PE38) was evaluated by MTT assay. The therapeutic effects and toxicity for each CRC cell line-derived mouse xenograft model were compared between the intervention and control groups. LS174T and DLD-1 cell lines showed a strong affinity for rBC2LCN. In the xenograft model, the tumor volume in the rBC2LCN-PE38 group was significantly reduced compared with that using control treatment alone. However, the HT-29 cell line showed weak affinity and poor therapeutic efficacy. No significant toxicities or adverse responses were observed. In conclusion, we demonstrated that rBC2LCN lectin binds CRC cells and that rBC2LCN-PE38 significantly suppresses tumor growth in vivo. In addition, the efficacy of the drug conjugate correlated with its binding affinity for each CRC cell line. These results suggest that lectin drug conjugate therapy has potential as a novel targeted therapy for CRC cell surface glycans.


Subject(s)
ADP Ribose Transferases/therapeutic use , Adenocarcinoma/drug therapy , Bacterial Toxins/therapeutic use , Colorectal Neoplasms/drug therapy , Exotoxins/therapeutic use , Immunoconjugates/therapeutic use , Lectins/therapeutic use , Virulence Factors/therapeutic use , ADP Ribose Transferases/adverse effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Bacterial Toxins/adverse effects , Burkholderia cenocepacia/chemistry , Cell Line, Tumor , Cell Survival , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Carriers , Exotoxins/adverse effects , Fucose/metabolism , Fucosyltransferases/metabolism , HT29 Cells , Heterografts , Humans , Immunoconjugates/adverse effects , In Vitro Techniques , Lectins/isolation & purification , Lectins/metabolism , Mice , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/therapeutic use , Tumor Burden , Virulence Factors/adverse effects , Pseudomonas aeruginosa Exotoxin A
14.
Bioconjug Chem ; 31(10): 2421-2430, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32996763

ABSTRACT

Immunotoxins are emerging candidates for cancer therapeutics. These biomolecules consist of a cell-targeting protein combined to a polypeptide toxin. Associations of both entities can be achieved either chemically by covalent bonds or genetically creating fusion proteins. However, chemical agents can affect the activity and/or stability of the conjugate proteins, and additional purification steps are often required to isolate the final conjugate from unwanted byproducts. As for fusion proteins, they often suffer from low solubility and yield. In this report, we describe a straightforward conjugation process to generate an immunotoxin using coassociating peptides (named K3 and E3), originating from the tetramerization domain of p53. To that end, a nanobody targeting the human epidermal growth factor receptor 2 (nano-HER2) and a protein toxin fragment from Pseudomonas aeruginosa exotoxin A (TOX) were genetically fused to the E3 and K3 peptides. Entities were produced separately in Escherichia coli in soluble forms and at high yields. The nano-HER2 fused to the E3 or K3 helixes (nano-HER2-E3 and nano-HER2-K3) and the coassembled immunotoxins (nano-HER2-K3E3-TOX and nano-HER2-E3K3-TOX) presented binding specificity on HER2-overexpressing cells with relative binding constants in the low nanomolar to picomolar range. Both toxin modules (E3-TOX and K3-TOX) and the combined immunotoxins exhibited similar cytotoxicity levels compared to the toxin alone (TOX). Finally, nano-HER2-K3E3-TOX and nano-HER2-E3K3-TOX evaluated on various breast cancer cells were highly potent and specific to killing HER2-overexpressing breast cancer cells with IC50 values in the picomolar range. Altogether, we demonstrate that this noncovalent conjugation method using two coassembling peptides can be easily implemented for the modular engineering of immunotoxins targeting different types of cancers.


Subject(s)
ADP Ribose Transferases/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Immunotoxins/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Virulence Factors/pharmacology , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Antineoplastic Agents/chemistry , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Breast Neoplasms/drug therapy , Cell Line, Tumor , Exotoxins/chemistry , Exotoxins/genetics , Female , Humans , Immunotoxins/chemistry , Immunotoxins/genetics , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
15.
Cell Microbiol ; 21(10): e13074, 2019 10.
Article in English | MEDLINE | ID: mdl-31231933

ABSTRACT

Cholera toxin (Ctx) is an AB-type protein toxin that acts as an adenosine diphosphate (ADP)-ribosyltransferase to disrupt intracellular signalling in the target cell. It moves by vesicle carriers from the cell surface to the endoplasmic reticulum (ER) of an intoxicated cell. The catalytic CtxA1 subunit then dissociates from the rest of the toxin, unfolds, and activates the ER-associated degradation system for export to the cytosol. Translocation occurs through an unusual ratchet mechanism in which the cytosolic chaperone Hsp90 couples CtxA1 refolding with CtxA1 extraction from the ER. Here, we report that Hsp90 recognises two peptide sequences from CtxA1: an N-terminal RPPDEI sequence (residues 11-16) and an LDIAPA sequence in the C-terminal region (residues 153-158) of the 192 amino acid protein. Peptides containing either sequence effectively blocked Hsp90 binding to full-length CtxA1. Both sequences were necessary for the ER-to-cytosol export of CtxA1. Mutagenesis studies further demonstrated that the RPP residues in the RPPDEI motif are required for CtxA1 translocation to the cytosol. The LDIAPA sequence is unique to CtxA1, but we identified an RPPDEI-like motif at the N- or C-termini of the A chains from four other ER-translocating toxins that act as ADP-ribosyltransferases: pertussis toxin, Escherichia coli heat-labile toxin, Pseudomonas aeruginosa exotoxin A, and Salmonella enterica serovar Typhimurium ADP-ribosylating toxin. Hsp90 plays a functional role in the intoxication process for most, if not all, of these toxins. Our work has established a defined RPPDEI binding motif for Hsp90 that is required for the ER-to-cytosol export of CtxA1 and possibly other toxin A chains as well.


Subject(s)
Cholera Toxin/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , HSP90 Heat-Shock Proteins/metabolism , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , Amino Acid Motifs/genetics , Animals , Bacterial Toxins/genetics , CHO Cells , Cholera Toxin/chemistry , Cholera Toxin/genetics , Cholera Toxin/isolation & purification , Cricetulus , Enterotoxins/genetics , Escherichia coli Proteins/genetics , Exotoxins/genetics , Gene Expression , Mutagenesis , Pertussis Toxin/genetics , Protein Binding , Protein Transport/genetics , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
16.
J Immunol ; 200(6): 2038-2045, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29431691

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins being developed for cancer treatment. They are composed of an Ab fragment that targets a cancer Ag and a cytotoxic portion of Pseudomonas exotoxin A. They are effective for patients with hematologic malignancies with defective immunity, but their efficacy against solid tumors is limited by anti-drug Ab (ADA) responses in immune-competent patients. Pre-existing Abs or immune memory owing to previous toxin exposure represent additional hurdles because they induce rapid and strong ADA responses. Here, we evaluated the efficacy of methotrexate (MTX) to prevent ADA formation against the mesothelin-targeting RIT LMB-100 in naive mice and in mice with pre-existing Abs. We found that low-dose MTX combined with LMB-100 completely suppressed the formation of ADAs in a dose- and frequency-dependent manner. Suppression of the immune response restored blood levels of LMB-100 and prevented its neutralization. Furthermore, combination of MTX with LMB-100 did not compromise the immune response against a second Ag given after stopping MTX, indicating specific immune tolerance. Adoptive transfer of splenocytes suppressed Ab responses to LMB-100 in recipient mice, indicating a durable immune tolerance. We conclude that combination of MTX and LMB-100 is effective at preventing immune responses in a durable, Ag-specific manner. We propose combining low-dose MTX in immune-competent cancer patients receiving RIT therapy to prevent immunogenicity. This approach could be applied to other immunogenic therapeutic agents and to proteins for which there is pre-existing immunity.


Subject(s)
Immune Tolerance/drug effects , Immunity, Humoral/drug effects , Immunotoxins/immunology , Methotrexate/pharmacology , Recombinant Proteins/immunology , ADP Ribose Transferases/immunology , Adoptive Transfer/methods , Animals , Antibodies, Monoclonal/immunology , Antibody Formation/drug effects , Bacterial Toxins/immunology , Cells, Cultured , Exotoxins/immunology , Female , GPI-Linked Proteins/pharmacology , Immune Tolerance/immunology , Immunity, Humoral/immunology , Immunotherapy/methods , Mesothelin , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
17.
Immunol Rev ; 270(1): 152-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26864110

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins designed to treat cancer. They are made up of an Fv or Fab that targets an antigen on a cancer cell fused to a 38-kDa portion of Pseudomonas exotoxin A (PE38). Because PE38 is a bacterial protein, it is highly immunogenic in patients with solid tumors that have normal immune systems, but much less immunogenic in patients with hematologic malignancies where the immune system is suppressed. RITs have shown efficacy in refractory hairy cell leukemia and in some children with acute lymphoblastic leukemia, but have been much less effective in solid tumors, because neutralizing antibodies develop and prevent additional treatment cycles. In this paper we will (i) review data from clinical trials describing the immunogenicity of PE38 in different patient populations; (ii) review results from clinical trials using different immunosuppressive drugs; and (iii) describe our efforts to make new less-immunogenic RITs by identifying and removing T- and B-cell epitopes to hide the RIT from the immune system.


Subject(s)
Immunotoxins/immunology , Immunotoxins/therapeutic use , Recombinant Fusion Proteins , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/immunology , Antibody Formation , Antigens/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Clinical Trials as Topic , Drug Administration Routes , Drug Therapy, Combination , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/immunology , Genetic Engineering , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/therapeutic use , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Immunotherapy , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Immunotoxins/chemistry , Immunotoxins/genetics , Mesothelin , Mice , Neoplasms/immunology , Neoplasms/therapy , Polyethylene Glycols , Sequence Deletion , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
18.
Cancer Invest ; 37(10): 546-557, 2019.
Article in English | MEDLINE | ID: mdl-31597492

ABSTRACT

Immunotoxin targeted therapy is a promising way of cancer therapy that is made from a toxin attached to an antibody which target a specific protein presented on cancer cells. In this study, we introduce immunotoxins comprising of truncated pseudomonas exotoxin A (PEA) and diphtheria toxin (DT) conjugated to trastuzumab. The effectiveness of 20 and 30 µg/ml immunotoxins and trastuzumab were studied on SK-BR-3 and BT-474 HER2/neu positive breast cancer cell lines by a cell death assay test. The produced immunotoxins have the potential to reduce the therapeutic dose of the trastuzumab and in the same time achieve higher efficiency.


Subject(s)
ADP Ribose Transferases/pharmacology , Bacterial Toxins/pharmacology , Breast Neoplasms/drug therapy , Diphtheria/metabolism , Exotoxins/pharmacology , Immunotoxins/pharmacology , Pseudomonas/metabolism , Virulence Factors/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Female , Humans , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Pseudomonas aeruginosa Exotoxin A
19.
Int J Mol Sci ; 20(10)2019 May 15.
Article in English | MEDLINE | ID: mdl-31096563

ABSTRACT

High immunogenicity and systemic toxicity are the main obstacles limiting the clinical use of the therapeutic agents based on Pseudomonas aeruginosa exotoxin A. In this work, we studied the immunogenicity, general toxicity and antitumor effect of the targeted toxin DARPin-LoPE composed of HER2-specific DARPin and a low immunogenic exotoxin A fragment lacking immunodominant human B lymphocyte epitopes. The targeted toxin has been shown to effectively inhibit the growth of HER2-positive human ovarian carcinoma xenografts, while exhibiting low non-specific toxicity and side effects, such as vascular leak syndrome and liver tissue degradation, as well as low immunogenicity, as was shown by specific antibody titer. This represents prospects for its use as an agent for targeted therapy of HER2-positive tumors.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Heterografts , Immunotoxins/immunology , Immunotoxins/pharmacology , Muscle Proteins/immunology , Nuclear Proteins/immunology , Ovarian Neoplasms/drug therapy , Receptor, ErbB-2/immunology , ADP Ribose Transferases/immunology , ADP Ribose Transferases/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Bacterial Toxins/immunology , Bacterial Toxins/pharmacology , Biomarkers, Tumor , Carcinoma/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Epitopes, B-Lymphocyte/genetics , Exotoxins/immunology , Exotoxins/pharmacology , Female , Humans , Inhibitory Concentration 50 , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Muscle Proteins/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/pathology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Spleen/pathology , Virulence Factors/immunology , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
20.
World J Microbiol Biotechnol ; 35(6): 94, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31187291

ABSTRACT

Pseudomonas aeruginosa is the major infectious agent of concern for cystic fibrosis (CF) patients. Therefore, it is necessary to develop appropriate strategies for preventing colonization by this bacterium and/or neutralizing virulence factors. In this study, we formulated the encapsulation of exotoxin A into PLGA nanoparticles. The biological activities of the nanovaccine candidate were also characterized. Based on the results, ETA-PLGA can act as a suitable immunogen to stimulate the humoral and cellular immune response. The antibodies raised against ETA-PLGA significantly decreased bacterial titer in the spleens of the immunized mice after challenge with PAO1 strain, compared to the control groups. The encapsulation of PLGA into ETA led to a significantly higher production of INF-γ, TNF-α, IL-4, and IL-17A cytokine responses compared to the ETA group. ETA-PLGA enhanced IgG responses in immunized mice compared to ETA antigen. We concluded that encapsulation of Pseudomonas aeruginosa ETA to PLGA nanoparticles can increase its functional activity by decreasing the bacterial dissemination.


Subject(s)
ADP Ribose Transferases/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Immunization , Nanoconjugates , Polylactic Acid-Polyglycolic Acid Copolymer/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/pathogenicity , Vaccines, Conjugate , Virulence Factors/immunology , ADP Ribose Transferases/therapeutic use , Animals , Bacterial Toxins/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Exotoxins/therapeutic use , Female , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Nanoparticles , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use , Pseudomonas Infections/immunology , Spleen/immunology , Spleen/microbiology , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL