Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 471
Filter
1.
Toxicon ; 230: 107157, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37196787

ABSTRACT

Crotamine, one of the major toxins present in the venom of the South American rattlesnake Crotalus durissus terrificus, exhibits potent cytotoxic properties and has been suggested for cancer therapy applications. However, its selectivity for cancer cells needs to be improved. This study designed and produced a novel recombinant immunotoxin, HER2(scFv)-CRT, composed of crotamine and single-chain Fv (scFv) derived from trastuzumab targeting human epidermal growth factor receptor 2 (HER2). The recombinant immunotoxin was expressed in Escherichia coli and purified using various chromatographic techniques. The cytotoxicity of HER2(scFv)-CRT was assessed in three breast cancer cell lines, demonstrating enhanced specificity and toxicity in HER2-expressing cells. These findings suggest that the crotamine-based recombinant immunotoxin has the potential to expand the repertoire of recombinant immunotoxin applications in cancer therapy.


Subject(s)
Crotalid Venoms , Immunotoxins , Neoplasms , Animals , Humans , Crotalid Venoms/chemistry , Crotalus , Immunotoxins/metabolism , Neoplasms/drug therapy , Cell Line, Tumor
2.
Exp Gerontol ; 174: 112130, 2023 04.
Article in English | MEDLINE | ID: mdl-36822486

ABSTRACT

Senescent cells were recently shown to play a role in aging-related malfunctions and pathologies. This consensus has been facilitated by evidence from senolytic model mice capable of eliminating senescent cells in tissues using well-characterized senescent markers, such as p16INK4a (hereafter p16). However, since the incomplete or artificial gene expression regulatory regions of manipulated marker genes affect their cognate expression, it currently remains unclear whether these models accurately reflect physiological senescence. We herein describe a novel approach to eliminate p16-expressing cells from mice at any given point in time, generating a new type of knock-in model, p16hCD2 mice and a toxin-conjugated anti-human CD2 antibody (hCD2-SAP) as an inducer. p16hCD2 mice possess an intact Cdkn2a locus that includes a p16 coding region and human CD2 (hCD2) expression unit. We confirmed cognate p16-associated hCD2 expression in mouse embryonic fibroblasts (MEFs) and in several tissues, such as the spleen, liver, and skin. We detected chronological increases in the hCD2-positive population in T lymphocytes that occurred in a p16-dependent manner, which reflected physiological aging. We then confirmed the high sensitivity of hCD2-SAP to hCD2 and validated its efficacy to remove p16-positive cells, particularly in T lymphocytes. The multiple administration of hCD2-SAP for a prolonged p16-positive cell deficiency partially restored aging-related phenotypes in T lymphocytes, such as the contraction of the CD4+ naïve population and expansion of senescence-associated T cells. Our novel approach of targeting p16-positive senescent cells will provide novel insights into the mechanisms underlying physiological aging in vivo.


Subject(s)
Immunotoxins , T-Lymphocytes , Mice , Animals , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Immunotoxins/genetics , Immunotoxins/metabolism , Senotherapeutics , Fibroblasts/metabolism , Phenotype , CD4-Positive T-Lymphocytes
3.
J Mol Model ; 28(12): 392, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36400988

ABSTRACT

Breast cancer remains the most frequently diagnosed cancer and the principal cause of mortality by malignancy in women. HER2 positive subtype includes 15-20% of breast cancer cases. This receptor could be an appropriate mark for targeting breast cancer cells. Immunotherapy methods compared to current cancer treatment methods have the lowest side effects. DELTA-stichotoxin-Hmg2a is isolated from the sea anemone and kills cells through pore formation. In the current study, we designed and evaluated an immunotoxin composed of pertuzumab and DELTA-stichotoxin-Hmg2a-derived scFv by bioinformatics tools. The designed immunotoxin was constructed using the amino acid sequences. Then, secondary structure and physico-chemical features were studied, and the tertiary structure of the immunotoxin was built according to the homology modeling methods. The validation and allergenicity of the model were assessed. The immunotoxin and receptor were docked and molecular dynamics simulation indicated the construct stability. The analysis results indicated that the construct is a stable protein that could have a natural-like structure and would not be an allergen, so this immunotoxin could effectively target HER2 receptors. Therefore, our designed immunotoxin could be an appropriate immunotoxin against HER2-positive breast cancer and could be a challenging topic for future in vitro and in vivo studies.


Subject(s)
Breast Neoplasms , HMGB3 Protein , Immunotoxins , Humans , Female , Immunotoxins/chemistry , Immunotoxins/metabolism , Immunotoxins/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Immunotherapy
4.
J Nanobiotechnology ; 20(1): 387, 2022 Aug 23.
Article in English | MEDLINE | ID: mdl-35999603

ABSTRACT

The plant toxin ricin, especially its cytotoxic A chain (RTA), can be genetically engineered with targeting ligands to develop specific anti-cancer recombinant immunotoxins (RITs). Here, we used affibody molecules targeting two cancer biomarkers, the receptors HER2 and EGFR, along with the KDEL signal peptide to construct two cancer-specific ricin-based RITs, HER2Afb-RTA-KDEL and EGFRAfb-RTA-KDEL. The affibodies successfully provided target-specificity and subsequent receptor-mediated endocytosis and the KDEL signal peptide routed the RITs through the retrograde transport pathway, effectively delivering RTA to the cytosol as well as avoiding the alternate recycling pathway that typical cancer cells frequently have. The in vivo efficacy of RITs was enhanced by introducing the albumin binding domain (AlBD) to construct AlBD/HER2Afb/RTA-KDEL. Systemic administration of AlBD-containing RITs to tumor-bearing mice significantly suppressed tumor growth without any noticeable side-effects. Collectively, combining target-selective affibody molecules, a cytotoxic RTA, and an intracellularly designating peptide, we successfully developed cancer-specific and efficacious ricin-based RITs. This approach can be applied to develop novel protein-based "magic bullets" to effectively suppress tumors that are resistant to conventional anti-cancer drugs.


Subject(s)
Immunotoxins , Neoplasms , Ricin , Animals , Apoptosis , Endocytosis , Immunotoxins/metabolism , Immunotoxins/pharmacology , Mice , Neoplasms/drug therapy , Protein Sorting Signals , Ricin/pharmacology , Ricin/toxicity
5.
Toxicol In Vitro ; 83: 105417, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35718257

ABSTRACT

Cancer stem cells (CSCs) are self-renewing multipotent cells that play a vital role in the development of cancer drug resistance conditions. Various therapies like conventional, targeted, and radiotherapies have been broadly used in targeting and killing these CSCs. Among these, targeted therapy selectively targets CSCs and leads to overcoming disease recurrence conditions in cancer patients. Immunotoxins (ITs) are protein-based therapeutics with selective targeting capabilities. These chimeric molecules are composed of two functional moieties, i.e., a targeting moiety for cell surface binding and a toxin moiety that induces the programmed cell death upon internalization. Several ITs have been constructed recently, and their preclinical and clinical efficacies have been evaluated. In this review, we comprehensively discussed the recent preclinical and clinical advances as well as significant challenges in ITs targeting CSCs, which might reduce the burden of drug resistance conditions in cancer patients from bench to bedside.


Subject(s)
Immunotoxins , Neoplasms , Apoptosis , Drug Resistance, Neoplasm , Humans , Immunotoxins/metabolism , Immunotoxins/pharmacology , Immunotoxins/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplastic Stem Cells
6.
Elife ; 112022 05 17.
Article in English | MEDLINE | ID: mdl-35579416

ABSTRACT

Typhoid toxin is an essential virulence factor for Salmonella Typhi, the cause of typhoid fever in humans. This toxin has an unusual biology in that it is produced by Salmonella Typhi only when located within host cells. Once synthesized, the toxin is secreted to the lumen of the Salmonella-containing vacuole from where it is transported to the extracellular space by vesicle carrier intermediates. Here, we report the identification of the typhoid toxin sorting receptor and components of the cellular machinery that packages the toxin into vesicle carriers, and exports it to the extracellular space. We found that the cation-independent mannose-6-phosphate receptor serves as typhoid toxin sorting receptor and that the coat protein COPII and the GTPase Sar1 mediate its packaging into vesicle carriers. Formation of the typhoid toxin carriers requires the specific environment of the Salmonella Typhi-containing vacuole, which is determined by the activities of specific effectors of its type III protein secretion systems. We also found that Rab11B and its interacting protein Rip11 control the intracellular transport of the typhoid toxin carriers, and the SNARE proteins VAMP7, SNAP23, and Syntaxin 4 their fusion to the plasma membrane. Typhoid toxin's cooption of specific cellular machinery for its transport to the extracellular space illustrates the remarkable adaptation of an exotoxin to exert its function in the context of an intracellular pathogen.


Subject(s)
Immunotoxins , Typhoid Fever , Humans , Immunotoxins/metabolism , Salmonella , Salmonella typhi/metabolism
7.
Adv Mater ; 33(48): e2103114, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34585802

ABSTRACT

Bifunctional proteins (BFPs) are a class of therapeutic agents produced through genetic engineering and protein engineering, and are increasingly used to treat various human diseases, including cancer. These proteins usually have two or more biological functions-specifically recognizing different molecular targets to regulate the related signaling pathways, or mediating effector molecules/cells to kill tumor cells. Unlike conventional small-molecule or single-target drugs, BFPs possess stronger biological activity but lower systemic toxicity. Hence, BFPs are considered to offer many benefits for the treatment of heterogeneous tumors. In this review, the authors briefly describe the unique structural feature of BFP molecules and innovatively divide them into bispecific antibodies, cytokine-based BFPs (immunocytokines), and protein toxin-based BFPs (immunotoxins) according to their mode of action. In addition, the latest advances in the development of BFPs are discussed and the potential limitations or problems in clinical applications are outlined. Taken together, future studies need to be centered on understanding the characteristics of BFPs for optimizing and designing more effective such drugs.


Subject(s)
Antibodies, Bispecific/therapeutic use , Cytokines/therapeutic use , Immunotoxins/therapeutic use , Neoplasms/therapy , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Immunotoxins/genetics , Immunotoxins/metabolism , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Protein Engineering , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment
8.
Toxins (Basel) ; 13(8)2021 08 13.
Article in English | MEDLINE | ID: mdl-34437438

ABSTRACT

Actinoporins (APs) are soluble pore-forming proteins secreted by sea anemones that experience conformational changes originating in pores in the membranes that can lead to cell death. The processes involved in the binding and pore-formation of members of this protein family have been deeply examined in recent years; however, the intracellular responses to APs are only beginning to be understood. Unlike pore formers of bacterial origin, whose intracellular impact has been studied in more detail, currently, we only have knowledge of a few poorly integrated elements of the APs' intracellular action. In this review, we present and discuss an updated landscape of the studies aimed at understanding the intracellular pathways triggered in response to APs attack with particular reference to sticholysin II, the most active isoform produced by the Caribbean Sea anemone Stichodactyla helianthus. To achieve this, we first describe the major alterations these cytolysins elicit on simpler cells, such as non-nucleated mammalian erythrocytes, and then onto more complex eukaryotic cells, including tumor cells. This understanding has provided the basis for the development of novel applications of sticholysins such as the construction of immunotoxins directed against undesirable cells, such as tumor cells, and the design of a cancer vaccine platform. These are among the most interesting potential uses for the members of this toxin family that have been carried out in our laboratory.


Subject(s)
Cell Death/drug effects , Cnidarian Venoms/metabolism , Cnidarian Venoms/toxicity , Immunotoxins/chemistry , Immunotoxins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Sea Anemones/chemistry , Animals
9.
Toxicol Lett ; 350: 30-39, 2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34147605

ABSTRACT

Bisphenol F (BPF) is a member of endocrine disrupting chemicals (EDCs). As a substitute of bisphenol A (BPA), BPF is widely used in various consumer products, leading to an increased risk of people's exposure. However, there are few studies on the immunotoxicity and mechanism of BPF. This study aimed to investigate the effect of BPF on the secretion of pro-inflammatory cytokines by macrophages and explore its mechanism. In our study, RAW264.7 macrophages were treated with different concentrations of BPF (0, 5, 10 and 20 µM) for 24 h. The results showed that the secretion of pro-inflammatory cytokines (IL-6, TNF-α and IL-1ß) and the production of lactate were increased in a dose-dependent manner. BPFalso led to the activation of the PI3K-AKT signaling pathway. After pretreatment with glycolysis inhibitor (2-DG) and exposure to BPF (20 µM), the secretion of pro-inflammatory cytokines induced by BPF was inhibited. PI3K inhibitor (LY294002) and estrogen receptor (ER) antagonist (ICI 182,780) could also inhibit the above effects induced by BPF (20 µM). In conclusion, our results suggested that BPF can enhance glycolysis through ER mediated PI3K-AKT signaling pathway, and the enhanced glycolysis further promoted the secretion of pro-inflammatory cytokines. Our research provides basic data for future studies on bisphenol exposure and immunotoxicity.


Subject(s)
Benzhydryl Compounds/toxicity , Cytokines/metabolism , Glycolysis/drug effects , Inflammation/metabolism , Macrophages/drug effects , Phenols/toxicity , Signal Transduction/drug effects , Benzhydryl Compounds/metabolism , Cells, Cultured/drug effects , Humans , Immunotoxins/metabolism , Phenols/metabolism
10.
J Cell Physiol ; 236(6): 4303-4312, 2021 06.
Article in English | MEDLINE | ID: mdl-33421131

ABSTRACT

The overexpression of epidermal growth factor receptor (EGFR) could result in the development of solid tumors of prostate, breast, gastric, colorectal, ovarian, and head and neck, leading to carcinoma. Antibody therapies are ideal methods to overcome malignant diseases. However, immunoribonucleases are a new generation of antibodies in which an RNase binds to a specific antibody and shows a stronger ability to terminate cancer cells. In this study, we engineered Rana pipiens RNase to bind to the scFv of human antiepidermal growth factor receptor antibody. The molecular dynamic simulations confirmed protein stability and the ability of scFv-ranpirnase (rantoxin) to bind to epidermal growth factor receptor protein. Then, the rantoxin construct was synthesized in a pCDNA 3.1 Neo vector. CHO-K1 cells were used as expression hosts and the construct was transfected. Cells were selected by antibiotic therapies using neomycin, 120 mg/ml, and the high-yield colony was screened by real-time polymerase chain reaction (PCR) methods. Then, the recombinant protein production was confirmed using the sodium dodecyl sulfate polyacrylamide gel electrophoresis and western blot analyses. The molecular dynamic simulation (MDS) confirmed that the I467, S468, Q408, and H409 amino acids of EGFR bonded well to rantoxin. As revealed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analyses, the rantoxin production and PCR analysis showed that the T3 colony can produce rantoxin messenger RNA fourfold higher than the GAPDH gene. The immunotoxin function was assessed in A431 cancer cells and EGFR-negative HEK293 cells, and IC50  values were estimated to be 22.4 ± 3 and >620.4 ± 5 nM, respectively. The results indicated that the immunotoxins produced in this study had the potential for use as anticancer drugs.


Subject(s)
Amphibian Proteins/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Immunotoxins/pharmacology , Protein Engineering , Ribonucleases/pharmacology , Single-Chain Antibodies/pharmacology , Skin Neoplasms/drug therapy , Amphibian Proteins/genetics , Amphibian Proteins/metabolism , Animals , Antineoplastic Agents, Immunological/metabolism , Apoptosis/drug effects , Binding Sites, Antibody , CHO Cells , Cell Line, Tumor , Cricetulus , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , ErbB Receptors/metabolism , HEK293 Cells , Humans , Immunotoxins/genetics , Immunotoxins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Rana pipiens , Ribonucleases/genetics , Ribonucleases/metabolism , Single-Chain Antibodies/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/pathology
11.
Clin Transl Oncol ; 23(8): 1549-1560, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33474678

ABSTRACT

OBJECTIVES: Epithelial growth factor receptor (EGFR), as a malignancy marker, is overly expressed in multiple solid tumors including colorectal neoplasms, one of the most prevalent malignancies worldwide. The main objective of this study is to enhance the efficacy of anti-tumor therapy targeting EGFR by constructing a novel EGFR-specific immunotoxin (C-CUS245C) based on Cetuximab and recombinant Cucurmosin (CUS245C). METHODS: E. coli BL21 (DE3) PlysS (E. coli) was used to express CUS245C with a cysteine residue inserting to the C-terminus of Cucurmosin. Then immobilized metal ion affinity chromatography (IMAC) was used to purify CUS245C. The chemical conjugation method was used for the preparation of C-CUS245C. Then dialysis and IMAC were used to purify C-CUS245C. Western blot as well as SDS-PAGE was carried out to characterize the formation of C-CUS245C. At last the anti-colorectal cancer activity of C-CUS245C was investigated in vitro and in vivo. RESULTS: CUS245C with high purity could be obtained from the prokaryotic system. C-CUS245C was successfully constructed and highly purified. The cytotoxicity assays in vitro showed a significant proliferation inhibition of C-CUS245C on EGFR-positive cells for 120 h with IC50 values less than 0.1 pM. Besides, the anti-tumor efficacy of C-CUS245C was remarkably more potent than that of Cetuximab, CUS245C, and C + CUS245C (P < 0.001). Whereas the cytotoxicity of C-CUS245C could hardly be detected on EGFR-null cell line. Our results also showed that C-CUS245C had efficacy of anti-colorectal cancer in mouse xenograft model, indicating the therapeutic potential of C-CUS245C for the targeted therapy of colorectal neoplasms. CONCLUSIONS: C-CUS245C exhibits potent and EGFR-specific cytotoxicity. Insertional mutagenesis technique is worthy to be adopted in the preparation of immunotoxin. Immunotoxin can be highly purified through dialysis followed by IMAC.


Subject(s)
Cetuximab/therapeutic use , Colorectal Neoplasms/therapy , Immunotoxins/therapeutic use , Molecular Targeted Therapy/methods , Plant Proteins/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab/pharmacology , Chromatography, Affinity/methods , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Escherichia coli/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Immunotoxins/chemistry , Immunotoxins/isolation & purification , Immunotoxins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis, Insertional/methods , Plant Proteins/chemistry , Plant Proteins/metabolism , Time Factors , Xenograft Model Antitumor Assays
12.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228031

ABSTRACT

Cholesterol seems to play a central role in the augmentation of saporin-based immunotoxin (IT) cytotoxicity by triterpenoid saponins. Endolysosomal escape has been proposed as one mechanism for the saponin-mediated enhancement of targeted toxins. We investigated the effects of lipid depletion followed by repletion on Saponinum album (SA)-induced endolysosomal escape of Alexa Fluor labelled saporin and the saporin-based immunotoxin OKT10-SAP, directed against CD38, in Daudi lymphoma cells. Lipid deprived cells showed reduced SA-induced endolysosomal escape at two concentrations of SA, as determined by a flow cytometric method. The repletion of membrane cholesterol by low density lipoprotein (LDL) restored SA-induced endolysosomal escape at a concentration of 5 µg/mL SA but not at 1 µg/mL SA. When LDL was used to restore the cholesterol levels in lipid deprived cells, the SA augmentation of OKT10-SAP cytotoxicity was partially restored at 1 µg/mL SA and fully restored at 5 µg/mL SA. These results suggest that different mechanisms of action might be involved for the two different concentrations of SA and that endosomal escape may not be the main mechanism for the augmentation of saporin IT cytotoxicity by SA at the sub-lytic concentration of 1 µg/mL SA.


Subject(s)
Cholesterol/chemistry , Endosomes/drug effects , Immunotoxins/metabolism , Lysosomes/drug effects , Saponins/pharmacology , Saporins/metabolism , ADP-ribosyl Cyclase 1/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cholesterol/metabolism , Cholesterol, LDL/pharmacology , Dose-Response Relationship, Drug , Endosomes/chemistry , Endosomes/metabolism , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Humans , Immunotoxins/chemistry , Lymphocytes/chemistry , Lymphocytes/drug effects , Lymphocytes/metabolism , Lysosomes/chemistry , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Saporins/chemistry , Sulfonic Acids/chemistry , Triterpenes/pharmacology
13.
Toxins (Basel) ; 12(10)2020 10 15.
Article in English | MEDLINE | ID: mdl-33076544

ABSTRACT

Toxins, while harmful and potentially lethal, have been engineered to develop potent therapeutics including cytotoxins and immunotoxins (ITs), which are modalities with highly selective targeting capabilities. Currently, three cytotoxins and IT are FDA-approved for treatment of multiple forms of hematological cancer, and additional ITs are tested in the clinical trials or at the preclinical level. For next generation of ITs, as well as antibody-mediated drug delivery systems, specific targeting by monoclonal antibodies is critical to enhance efficacies and reduce side effects, and this methodological field remains open to discover potent therapeutic monoclonal antibodies. Here, we describe our application of engineered toxin termed a cell-based IT screening system. This unique screening strategy offers the following advantages: (1) identification of monoclonal antibodies that recognize cell-surface molecules, (2) selection of the antibodies that are internalized into the cells, (3) selection of the antibodies that induce cytotoxicity since they are linked with toxins, and (4) determination of state-specific activities of the antibodies by differential screening under multiple experimental conditions. Since the functional monoclonal antibodies with internalization capacities have been identified successfully, we have pursued their subsequent modifications beyond antibody drug conjugates, resulting in development of immunoliposomes. Collectively, this screening system by using engineered toxin is a versatile platform, which enables straight-forward and rapid selection for discovery of novel functional antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Membrane/metabolism , High-Throughput Screening Assays , Immunoconjugates/pharmacology , Immunotoxins/pharmacology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Specificity , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Biological Transport , Cell Membrane/immunology , Cytotoxicity, Immunologic , Diphtheria Toxin/immunology , Diphtheria Toxin/metabolism , Diphtheria Toxin/pharmacology , Exotoxins/immunology , Exotoxins/metabolism , Exotoxins/pharmacology , Humans , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/metabolism , Immunotoxins/genetics , Immunotoxins/immunology , Immunotoxins/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2/pharmacology , Liposomes , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
14.
Biomolecules ; 10(9)2020 09 17.
Article in English | MEDLINE | ID: mdl-32957689

ABSTRACT

Cancer cells frequently upregulate surface receptors that promote growth and survival. These receptors constitute valid targets for intervention. One strategy involves the delivery of toxic payloads with the goal of killing those cancer cells with high receptor levels. Delivery can be accomplished by attaching a toxic payload to either a receptor-binding antibody or a receptor-binding ligand. Generally, the cell-binding domain of the toxin is replaced with a ligand or antibody that dictates a new binding specificity. The advantage of this "immunotoxin" approach lies in the potency of these chimeric molecules for killing cancer cells. However, receptor expression on normal tissue represents a significant obstacle to therapeutic intervention.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotoxins/immunology , Neoplasms/immunology , Receptors, Cell Surface/immunology , Toxins, Biological/immunology , Animals , Antibodies, Monoclonal/metabolism , Cell Survival/drug effects , Cell Survival/immunology , Humans , Immunotoxins/metabolism , Immunotoxins/therapeutic use , Ligands , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Toxins, Biological/metabolism
15.
Proc Natl Acad Sci U S A ; 117(36): 22090-22100, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32839344

ABSTRACT

The application of proteinaceous toxins for cell ablation is limited by their high on- and off-target toxicity, severe side effects, and a narrow therapeutic window. The selectivity of targeting can be improved by intein-based toxin reconstitution from two dysfunctional fragments provided their cytoplasmic delivery via independent, selective pathways. While the reconstitution of proteins from genetically encoded elements has been explored, exploiting cell-surface receptors for boosting selectivity has not been attained. We designed a robust splitting algorithm and achieved reliable cytoplasmic reconstitution of functional diphtheria toxin from engineered intein-flanked fragments upon receptor-mediated delivery of one of them to the cells expressing the counterpart. Retargeting the delivery machinery toward different receptors overexpressed in cancer cells enables selective ablation of specific subpopulations in mixed cell cultures. In a mouse model, the transmembrane delivery of a split-toxin construct potently inhibits the growth of xenograft tumors expressing the split counterpart. Receptor-mediated delivery of engineered split proteins provides a platform for precise therapeutic and experimental ablation of tumors or desired cell populations while also greatly expanding the applicability of the intein-based protein transsplicing.


Subject(s)
Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Cytoplasm/metabolism , Drug Delivery Systems/methods , Inteins , Neoplasms/drug therapy , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Line, Tumor , Cytoplasm/genetics , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/chemistry , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Female , Heterografts , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Immunotoxins/genetics , Immunotoxins/metabolism , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Protein Domains , Protein Transport , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
16.
Anticancer Res ; 40(7): 3793-3799, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32620618

ABSTRACT

BACKGROUND/AIM: Head and neck squamous cell carcinoma (HNSCC) is one of the most common types of cancer worldwide. Our study focused on the axon guidance receptor roundabout guidance receptor 1 (ROBO1) as a target for monoclonal antibody therapy of HNSCC. We previously showed that saporin-conjugated anti-ROBO1 (B5209B) immunotoxin (IT-ROBO1) enhanced cytotoxic effects on HNSCC cells in combination with the photosensitizer aluminum phthalocyanine disulphonate (AlPcS2a) and illumination. We examined the effects of this combination therapy in a mouse xenograft model. MATERIALS AND METHODS: IT-ROBO1 was intraperitoneally administered to HSQ-89 (derived from Japanese maxillary sinus squamous carcinoma, RCB0789; RIKEN, Tsukuba, Japan) xenografted mice. After 3 days, AlPcS2a was injected subcutaneously around the tumor and the area was illuminated at 650 nm for 30 min. The growth of the tumor was evaluated and the effects on the tumor were examined. RESULTS: Pronounced anti-tumor effects were elicited by the administration of IT-ROBO1 and AlPcS2a with light illumination on tumor size and pathological characteristics. CONCLUSION: The results showed that photosensitizer treatment with illumination robustly enhanced the antitumor effect of the IT-ROBO1 immunotoxin.


Subject(s)
Head and Neck Neoplasms/drug therapy , Immunotoxins/metabolism , Maxillary Sinus/drug effects , Nerve Tissue Proteins/metabolism , Photosensitizing Agents/pharmacology , Receptors, Immunologic/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Maxillary Sinus/metabolism , Mice , Mice, Inbred BALB C , Squamous Cell Carcinoma of Head and Neck/metabolism , Xenograft Model Antitumor Assays , Roundabout Proteins
17.
Toxicon ; 180: 49-61, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32268155

ABSTRACT

Mycotoxins-contaminated milk could threaten human health; therefore, it is necessary to demonstrate the toxicological effect of mycotoxins in milk. Most recently, researchers have paid more attention to the immunotoxic effects of the individual cereal-contaminating mycotoxins, namely, zearalenone and deoxynivalenol. However, there is scant information about the intestinal immunotoxicity of aflatoxin M1 (AFM1), let alone that of a combination of AFM1 and ochratoxin A (OTA), which often co-occur in milk. To reveal the inflammatory response caused by these mycotoxins, expression of inflammation-related genes in differentiated Caco-2 cells was analyzed, demonstrating a synergistic effect of the mixture of AFM1 (4 µg/mL) and OTA (4 µg/mL). Integrative transcriptomic and proteomic analyses were also performed. A cross-omics analysis identified several mechanisms underlying this synergy: (i) compared with stimulation with either compound alone, combined use resulted in stronger induction of proteins involved in immunity-related pathways; (ii) combination of the two agents targeted different points in the same pathways; and (iii) combination of the two agents activated specific inflammation-related pathways. These results suggested that combined use of AFM1 and OTA might exacerbate intestinal inflammation, indicating that regulatory authorities should pay more attention to food contamination by multiple mycotoxins when performing risk assessments.


Subject(s)
Aflatoxin M1/metabolism , Immunotoxins/metabolism , Intestines/drug effects , Ochratoxins/metabolism , Proteome/metabolism , Aflatoxin M1/genetics , Animals , Caco-2 Cells , Cell Differentiation , Food Contamination , Gene Expression Profiling , Humans , Immunotoxins/genetics , Milk , Mycotoxins , Proteomics , Transcriptome , Zearalenone
18.
PLoS Pathog ; 16(1): e1008223, 2020 01.
Article in English | MEDLINE | ID: mdl-31905218

ABSTRACT

Epstein-Barr virus (EBV) infection is closely linked to several human malignancies including endemic Burkitt's lymphoma, Hodgkin's lymphoma and nasopharyngeal carcinomas (NPC). Latent membrane protein 2 (LMP-2) of EBV plays a pivotal role in pathogenesis of EBV-related tumors and thus, is a potential target for diagnosis and targeted therapy of EBV LMP-2+ malignant cancers. Affibody molecules are developing as imaging probes and tumor-targeted delivery of small molecules. In this study, four EBV LMP-2-binding affibodies (ZEBV LMP-212, ZEBV LMP-2132, ZEBV LMP-2137, and ZEBV LMP-2142) were identified by screening a phage-displayed LMP-2 peptide library for molecular imaging and targeted therapy in EBV xenograft mice model. ZEBV LMP-2 affibody has high binding affinity for EBV LMP-2 and accumulates in mouse tumor derived from EBV LMP-2+ xenografts for 24 h after intravenous (IV) injection. Subsequent fusion of Pseudomonas exotoxin PE38KDEL to the ZEBV LMP-2 142 affibody led to production of Z142X affitoxin. This fused Z142X affitoxin exhibits high cytotoxicity specific for EBV+ cells in vitro and significant antitumor effect in mice bearing EBV+ tumor xenografts by IV injection. The data provide the proof of principle that EBV LMP-2-speicifc affibody molecules are useful for molecular imaging diagnosis and have potentials for targeted therapy of LMP-2-expressing EBV malignancies.


Subject(s)
Herpesvirus 4, Human , Immunotoxins/therapeutic use , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/therapy , Viral Matrix Proteins/metabolism , Animals , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Immunotoxins/metabolism , Mice , Mice, Inbred BALB C , Molecular Imaging , Nasopharyngeal Carcinoma/diagnostic imaging , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/diagnostic imaging , Nasopharyngeal Neoplasms/virology , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/therapy , Peptide Library , Protein Binding , Viral Matrix Proteins/antagonists & inhibitors , Xenograft Model Antitumor Assays
19.
Immunol Cell Biol ; 98(3): 187-202, 2020 03.
Article in English | MEDLINE | ID: mdl-31916611

ABSTRACT

The stage-specific embryonic antigen-4 (SSEA-4) is a cell surface glycosphingolipid antigen expressed in early stages of human development. This surface marker is downregulated during the differentiation process but is found re-expressed in several types of tumors, including breast cancer. This feature makes SSEA-4 an attractive target for the development of therapeutic antibodies against tumors. In this work, we first studied the binding and intracellular fate of the monoclonal antibody MC-813-70 directed against SSEA-4. MC-813-70 was found to be rapidly internalized into triple-negative breast cancer cells following binding to its target at the plasma membrane, and to accumulate in acidic organelles, most likely lysosomes. Given the internalization feature of MC-813-70, we next tested whether the antibody was able to selectively deliver the saporin toxin inside SSEA-4-expressing cells. Results show that the immunotoxin complex was properly endocytosed and able to reduce cell viability of breast cancer cells in vitro, either alone or in combination with chemotherapeutic drugs. Our findings indicate that the MC-813-70 antibody has the potential to be developed as an alternative targeted therapeutic agent for cancer cells expressing the SSEA-4 glycolipid.


Subject(s)
Immunotoxins/pharmacology , Saporins/pharmacology , Stage-Specific Embryonic Antigens/immunology , Triple Negative Breast Neoplasms/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Female , Humans , Immunotoxins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Saporins/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
20.
J Pharm Sci ; 109(1): 104-115, 2020 01.
Article in English | MEDLINE | ID: mdl-31669121

ABSTRACT

Immunotoxins (ITs) are attractive anticancer modalities aimed at cancer-specific delivery of highly potent cytotoxic protein toxins. An IT consists of a targeting domain (an antibody, cytokine, or another cell-binding protein) chemically conjugated or recombinantly fused to a highly cytotoxic payload (a bacterial and plant toxin or human cytotoxic protein). The mode of action of ITs is killing designated cancer cells through the effector function of toxins in the cytosol after cellular internalization via the targeted cell-specific receptor-mediated endocytosis. Although numerous ITs of diverse structures have been tested in the past decades, only 3 ITs-denileukin diftitox, tagraxofusp, and moxetumomab pasudotox-have been clinically approved for treating hematological cancers. No ITs against solid tumors have been approved for clinical use. In this review, we discuss critical research and development issues associated with ITs that limit their clinical success as well as strategies to overcome these obstacles. The issues include off-target and on-target toxicities, immunogenicity, human cytotoxic proteins, antigen target selection, cytosolic delivery efficacy, solid-tumor targeting, and developability. To realize the therapeutic promise of ITs, novel strategies for safe and effective cytosolic delivery into designated tumors, including solid tumors, are urgently needed.


Subject(s)
Antineoplastic Agents/metabolism , Immunotoxins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Clinical Trials as Topic/methods , Cytosol/drug effects , Cytosol/metabolism , Exotoxins/administration & dosage , Exotoxins/chemistry , Exotoxins/metabolism , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Protein Binding/physiology , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...