Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 352
Filtrar
1.
Colloids Surf B Biointerfaces ; 238: 113870, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38555763

RESUMEN

Antibiotic resistance has become an urgent threat to health care in recent years. The use of drug delivery systems provides advantages over conventional administration of antibiotics and can slow the development of antibiotic resistance. In the current study, we developed a toxin-triggered liposomal antibiotic delivery system, in which the drug release is enabled by the leukotoxin (LtxA) produced by the Gram-negative pathogen, Aggregatibacter actinomycetemcomitans. LtxA has previously been shown to mediate membrane disruption by promoting a lipid phase change in nonlamellar lipids, such as 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-methyl (N-methyl-DOPE). In addition, LtxA has been observed to bind strongly and nearly irreversibly to membranes containing large amounts of cholesterol. Here, we designed a liposomal delivery system composed of N-methyl-DOPE and cholesterol to take advantage of these interactions. Specifically, we hypothesized that liposomes composed of N-methyl-DOPE and cholesterol, encapsulating antibiotics, would be sensitive to LtxA, enabling controlled antibiotic release. We observed that liposomes composed of N-methyl-DOPE were sensitive to the presence of low concentrations of LtxA, and cholesterol increased the extent and kinetics of content release. The liposomes were stable under various storage conditions for at least 7 days. Finally, we showed that antibiotic release occurs selectively in the presence of an LtxA-producing strain of A. actinomycetemcomitans but not in the presence of a non-LtxA-expressing strain. Together, these results demonstrate that the designed liposomal vehicle enables toxin-triggered delivery of antibiotics to LtxA-producing strains of A. actinomycetemcomitans.


Asunto(s)
Aggregatibacter actinomycetemcomitans , Antibacterianos , Liposomas , Liposomas/química , Antibacterianos/farmacología , Antibacterianos/química , Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Liberación de Fármacos , Colesterol/química , Colesterol/metabolismo , Pruebas de Sensibilidad Microbiana , Exotoxinas/metabolismo , Exotoxinas/química , Fosfatidiletanolaminas/química , Sistemas de Liberación de Medicamentos
2.
Mol Cells ; 46(12): 764-777, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38052492

RESUMEN

Recombinant immunotoxins (RITs) are fusion proteins consisting of a targeting domain linked to a toxin, offering a highly specific therapeutic strategy for cancer treatment. In this study, we engineered and characterized RITs aimed at mesothelin, a cell surface glycoprotein overexpressed in various malignancies. Through an extensive screening of a large nanobody library, four mesothelin-specific nanobodies were selected and genetically fused to a truncated Pseudomonas exotoxin (PE24B). Various optimizations, including the incorporation of furin cleavage sites, maltose-binding protein tags, and tobacco etch virus protease cleavage sites, were implemented to improve protein expression, solubility, and purification. The RITs were successfully overexpressed in Escherichia coli, achieving high solubility and purity post-purification. In vitro cytotoxicity assays on gastric carcinoma cell lines NCI-N87 and AGS revealed that Meso(Nb2)-PE24B demonstrated the highest cytotoxic efficacy, warranting further characterization. This RIT also displayed selective binding to human and monkey mesothelins but not to mouse mesothelin. The competitive binding assays between different RIT constructs revealed significant alterations in IC50 values, emphasizing the importance of nanobody specificity. Finally, a modification in the endoplasmic reticulum retention signal at the C-terminus further augmented its cytotoxic activity. Our findings offer valuable insights into the design and optimization of RITs, showcasing the potential of Meso(Nb2)-PE24B as a promising therapeutic candidate for targeted cancer treatment.


Asunto(s)
Antineoplásicos , Toxinas Bacterianas , Inmunotoxinas , Neoplasias , Anticuerpos de Dominio Único , Animales , Ratones , Humanos , Exotoxinas/genética , Exotoxinas/farmacología , Exotoxinas/química , Inmunotoxinas/genética , Inmunotoxinas/farmacología , Inmunotoxinas/química , Mesotelina , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/farmacología , Toxinas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Dominio Catalítico , Línea Celular Tumoral , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Neoplasias/tratamiento farmacológico
3.
Ther Deliv ; 14(1): 31-60, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36950853

RESUMEN

Pseudomonas aeruginosa exotoxin A-based immunotoxins (PE-ITs) are fusion proteins that harness targeting and toxin moieties. Structural optimizations in PE and targeting moieties were implemented to lower their immunogenicity and alleviate undesirable side effects. PE moiety was engineered to lack its cell-binding domain and T cell epitope regions, whereas single chain (scFv) and disulfide Fv portions (dsFv), nanobodies, and monobodies were utilized as targeting moieties. This review discusses applications of PE-ITs on different types of cancer, structural optimizations to reduce PE-ITs drawbacks, and recent modifications applied for efficient therapeutic delivery. Finally, we draw attention to the possibility of combining radiotherapy, radionuclides, and RGDs with PE-IT to improve overall response rates of IT-based treatments and reduce cancer cell resistance.


Exotoxin A-immunotoxins are proteins that have been used in cancer treatments. The building components of these proteins are very poisonous to both cancer and normal cells. Also, unfavorable body reactions and side effects were seen with their usage. To allow the safe use of these proteins, changes were made in their building components. These changes made them damaging only to cancer cells while being safe to normal non-cancerous cells. This review will talk about the use of exotoxin A-Immunotoxins in different cancer treatments, and how they are created to limit the poisonous effect of their building components to only cancer cells.


Asunto(s)
Toxinas Bacterianas , Inmunotoxinas , Neoplasias , Humanos , Inmunotoxinas/uso terapéutico , Inmunotoxinas/química , Exotoxinas/uso terapéutico , Exotoxinas/química , Neoplasias/tratamiento farmacológico , Pseudomonas aeruginosa , Exotoxina A de Pseudomonas aeruginosa
4.
Comput Methods Programs Biomed ; 222: 106952, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35724475

RESUMEN

The leukotoxin (LtxA) of Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) is a protein exotoxin belonging to the repeat-in-toxin family (RTX). Numerous studies have demonstrated that LtxA may play a critical role in the pathogenicity of A. actinomycetemcomitans since hyper-leukotoxic strains have been associated with severe disease. Accordingly, considerable effort has been made to elucidate the mechanisms by which LtxA interacts with host cells and induce their death. However, these attempts have been hampered by the unavailability of a tertiary structure of the toxin, which limits the understanding of its molecular properties and mechanisms. In this paper, we used homology and template free modeling algorithms to build the complete tertiary model of LtxA at atomic level in its calcium-bound Holo-state. The resulting model was refined by energy minimization, validated by Molprobity and ProSA tools, and subsequently subjected to a cumulative 600ns of all-atom classical molecular dynamics simulation to evaluate its structural aspects. The druggability of the proposed model was assessed using Fpocket and FTMap tools, resulting in the identification of four putative cavities and fifteen binding hotspots that could be targeted by rational drug design tools to find new ligands to inhibit LtxA activity.


Asunto(s)
Aggregatibacter actinomycetemcomitans , Exotoxinas , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/metabolismo , Simulación por Computador , Exotoxinas/química , Exotoxinas/metabolismo , Exotoxinas/farmacología
5.
Anticancer Res ; 41(8): 3741-3746, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34281832

RESUMEN

BACKGROUND/AIM: Reports on over-expression of the epidermal growth factor receptor (EGFR) in bladder cancer and its function in tumorigenesis have suggested to target this antigen. MATERIALS AND METHODS: We generated the targeted toxin EGF-PE40 consisting of the human epidermal growth factor (EGF) as the binding domain and PE40, a truncated version of Pseudomonas Exotoxin A, as the toxin domain. EGF-PE40 was tested on EGFR-expressing bladder cancer cells in view of binding via flow cytometry, and cytotoxicity via WST viability assay. Induction of apoptosis was examined by western blot. RESULTS: The targeted toxin specifically triggered cytotoxicity in the bladder cancer cells with 50% inhibitory concentration (IC50) values in the low nanomolar or picomolar range, and was about 1,250- to 1,500-fold more cytotoxic than the EGFR inhibitor erlotinib. Cytotoxicity of EGF-PE40 was based on the induction of apoptosis. CONCLUSION: EGF-PE40 represents a promising candidate for the future treatment of bladder cancer.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Factor de Crecimiento Epidérmico/química , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , ADP Ribosa Transferasas/química , Animales , Toxinas Bacterianas/química , Células CHO , Línea Celular Tumoral , Cricetulus , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Exotoxinas/química , Humanos , Factores de Virulencia/química , Exotoxina A de Pseudomonas aeruginosa
6.
Int Immunopharmacol ; 96: 107759, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34162138

RESUMEN

Recombinant immunotoxins are fusion proteins composed of a peptide toxin and a specific targeting domain through genetic recombination. They are engineered to recognize disease-specific target receptors and kill the cell upon internalization. Full-sized monoclonal antibodies, smaller antibody fragments and ligands, such as a cytokine or a growth factor, have been commonly used as the targeting domain, while bacterial Pseudomonas aeruginosa exotoxin (PE) is the usual toxin fusion partner, due to its natural cytotoxicity and other unique advantages. PE-based recombinant immunotoxins have shown remarkable efficacy in the treatment of tumors and autoimmune diseases. At the same time, efforts are underway to address major challenges, including immunogenicity, nonspecific cytotoxicity and poor penetration, which limit their clinical applications. Recent strategies for structural optimization of PE-based immunotoxins, combined with mutagenesis approaches, have reduced the immunogenicity and non-specific cytotoxicity, thus increasing both their safety and efficacy. This review highlights novel insights and design concepts that were used to advance immunotoxins for the treatment of hematological and solid tumors and also presents future development prospect of PE-based recombinant immunotoxins that are expected to play an important role in cancer therapy.


Asunto(s)
Exotoxinas/uso terapéutico , Inmunotoxinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Pseudomonas aeruginosa/química , Animales , Ensayos Clínicos como Asunto , Exotoxinas/química , Exotoxinas/farmacología , Humanos , Inmunotoxinas/química , Inmunotoxinas/inmunología , Inmunotoxinas/farmacología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico
7.
Int J Mol Med ; 48(1)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34080646

RESUMEN

Glioblastomas (GBMs) are refractory to current treatments and novel therapeutic approaches need to be explored. Pro­apoptotic tumor necrosis factor­related apoptosis­inducing ligand (TRAIL) is tumor­specific and has been shown to induce apoptosis and subsequently kill GBM cells. However, approximately 50% of GBM cells are resistant to TRAIL and a combination of TRAIL with other therapeutics is necessary to induce mechanism­based cell death in TRAIL­resistant GBMs. The present study examined the ability of the tumor cell surface receptor, interleukin (IL)­13 receptor α2 (IL13Rα2)­ and epidermal growth factor receptor (EGFR)­targeted pseudomonas exotoxin (PE) to sensitize TRAIL­resistant GBM cells and assessed the dual effects of interleukin 13­PE (IL13­PE) or EGFR nanobody­PE (ENb­PE) and TRAIL for the treatment of a broad range of brain tumors with a distinct TRAIL therapeutic response. Receptor targeted toxins upregulated TRAIL death receptors (DR4 and DR5) and suppressed the expression of anti­apoptotic FLICE­inhibitory protein (FLIP) and X­linked inhibitor of apoptosis protein (XIAP). This also led to the induction of the cleavage of caspase­8 and caspase­9 and resulted in the sensitization of highly resistant established GBM and patient­derived GBM stem cell (GSC) lines to TRAIL­mediated apoptosis. These findings provide a mechanism­based strategy that may provide options for the cell­mediated delivery of bi­functional therapeutics to target a wide spectrum of TRAIL­resistant GBMs.


Asunto(s)
Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Glioblastoma , Subunidad alfa2 del Receptor de Interleucina-13 , Interleucina-13/farmacología , Proteínas de Neoplasias , Pseudomonas/química , Anticuerpos de Dominio Único/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF , Toxinas Bacterianas/química , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Exotoxinas/química , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Inmunotoxinas/química , Inmunotoxinas/farmacología , Interleucina-13/química , Subunidad alfa2 del Receptor de Interleucina-13/genética , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Anticuerpos de Dominio Único/química , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
8.
J Biol Chem ; 295(50): 17241-17250, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33051210

RESUMEN

Leukocidin ED (LukED) is a pore-forming toxin produced by Staphylococcus aureus, which lyses host cells and promotes virulence of the bacteria. LukED enables S. aureus to acquire iron by lysing erythrocytes, which depends on targeting the host receptor Duffy antigen receptor for chemokines (DARC). The toxin also targets DARC on the endothelium, contributing to the lethality observed during bloodstream infection in mice. LukED is comprised of two monomers: LukE and LukD. LukE binds to DARC and facilitates hemolysis, but the closely related Panton-Valentine leukocidin S (LukS-PV) does not bind to DARC and is not hemolytic. The interaction of LukE with DARC and the role this plays in hemolysis are incompletely characterized. To determine the domain(s) of LukE that are critical for DARC binding, we studied the hemolytic function of LukE-LukS-PV chimeras, in which areas of sequence divergence (divergence regions, or DRs) were swapped between the toxins. We found that two regions of LukE's rim domain contribute to hemolysis, namely residues 57-75 (DR1) and residues 182-196 (DR4). Interestingly, LukE DR1 is sufficient to render LukS-PV capable of DARC binding and hemolysis. Further, LukE, by binding DARC through DR1, promotes the recruitment of LukD to erythrocytes, likely by facilitating LukED oligomer formation. Finally, we show that LukE targets murine Darc through DR1 in vivo to cause host lethality. These findings expand our biochemical understanding of the LukE-DARC interaction and the role that this toxin-receptor pair plays in S. aureus pathophysiology.


Asunto(s)
Proteínas Bacterianas , Sistema del Grupo Sanguíneo Duffy , Eritrocitos , Exotoxinas , Proteínas Hemolisinas , Receptores de Superficie Celular , Staphylococcus aureus , Animales , Humanos , Ratones , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sistema del Grupo Sanguíneo Duffy/química , Sistema del Grupo Sanguíneo Duffy/genética , Sistema del Grupo Sanguíneo Duffy/metabolismo , Eritrocitos/química , Eritrocitos/metabolismo , Exotoxinas/química , Exotoxinas/genética , Exotoxinas/metabolismo , Dominios Proteicos , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Staphylococcus aureus/química , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
9.
Bioconjug Chem ; 31(10): 2421-2430, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-32996763

RESUMEN

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.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Antineoplásicos/farmacología , Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Inmunotoxinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Anticuerpos de Dominio Único/farmacología , Factores de Virulencia/farmacología , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , Antineoplásicos/química , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Exotoxinas/química , Exotoxinas/genética , Femenino , Humanos , Inmunotoxinas/química , Inmunotoxinas/genética , Modelos Moleculares , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Factores de Virulencia/química , Factores de Virulencia/genética , Exotoxina A de Pseudomonas aeruginosa
10.
J Nutr Biochem ; 86: 108484, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32827665

RESUMEN

Linoleic acid (LA) is the most abundant polyunsaturated fatty acid found in the Western diet. Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. Previous studies of the EpOMEs and DiHOMEs have focused on their roles in cytotoxic processes, primarily in the inhibition of the neutrophil respiratory burst. More recent research has suggested the DiHOMEs may be important lipid mediators in pain perception, altered immune response and brown adipose tissue activation by cold and exercise. The purpose of this review is to summarize the current understanding of the physiological and pathophysiological roles and modes of action of the EpOMEs and DiHOMEs in health and disease.


Asunto(s)
Ácido Linoleico/metabolismo , Ácidos Oléicos/metabolismo , Ácidos Esteáricos/metabolismo , Tejido Adiposo Pardo/metabolismo , Analgésicos/farmacología , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Endocrino/efectos de los fármacos , Epóxido Hidrolasas/metabolismo , Exotoxinas/química , Humanos , Sistema Inmunológico/efectos de los fármacos , Inflamación , Lípidos/química , Pulmón/efectos de los fármacos , Ratones , Neutrófilos/metabolismo , Oxidación-Reducción , Manejo del Dolor , Estallido Respiratorio
11.
Front Immunol ; 11: 1261, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32695104

RESUMEN

Immunotoxins are cytolytic fusion proteins developed for cancer therapy, composed of an antibody fragment that binds to a cancer cell and a protein toxin fragment that kills the cell. Pseudomonas exotoxin A (PE) is a potent toxin that is used for the killing moiety in many immunotoxins. Moxetumomab Pasudotox (Lumoxiti) contains an anti-CD22 Fv and a 38 kDa portion of PE. Lumoxiti was discovered in the Laboratory of Molecular Biology at the U.S. National Cancer Institute and co-developed with Medimmune/AstraZeneca to treat hairy cell leukemia. In 2018 Lumoxiti was approved by the US Food and Drug Administration for the treatment of drug-resistant Hairy Cell Leukemia. Due to the bacterial origin of the killing moiety, immunotoxins containing PE are highly immunogenic in patients with normal immune systems, but less immunogenic in patients with hematologic malignancies, whose immune systems are often compromised. LMB-100 is a de-immunized variant of the toxin with a humanized antibody that targets mesothelin and a PE toxin that was rationally designed for diminished reactivity with antibodies and B cell receptors. It is now being evaluated in clinical trials for the treatment of mesothelioma and pancreatic cancer and is showing somewhat diminished immunogenicity compared to its un modified parental counterpart. Here we review the immunogenicity of the original and de-immunized PE immunotoxins in mice and patients, the development of anti-drug antibodies (ADAs), their impact on drug availability and their effect on clinical efficacy. Efforts to mitigate the immunogenicity of immunotoxins and its impact on immunogenicity will be described including rational design to identify, remove, or suppress B cell or T cell epitopes, and combination of immunotoxins with immune modulating drugs.


Asunto(s)
Exotoxinas/inmunología , Inmunotoxinas/inmunología , Pseudomonas/inmunología , ADP Ribosa Transferasas/inmunología , Animales , Formación de Anticuerpos/inmunología , Toxinas Bacterianas/inmunología , Ensayos Clínicos como Asunto , Epítopos de Linfocito B/inmunología , Exotoxinas/química , Exotoxinas/farmacocinética , Humanos , Inmunoensayo , Inmunomodulación/efectos de los fármacos , Inmunotoxinas/química , Inmunotoxinas/farmacocinética , Mesotelina , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes/inmunología , Relación Estructura-Actividad , Resultado del Tratamiento , Factores de Virulencia/inmunología , Exotoxina A de Pseudomonas aeruginosa
12.
J Am Chem Soc ; 142(25): 10899-10904, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32479075

RESUMEN

Optimization of small-molecule probes or drugs is a synthetically lengthy, challenging, and resource-intensive process. Lack of automation and reliance on skilled medicinal chemists is cumbersome in both academic and industrial settings. Here, we demonstrate a high-throughput hit-to-lead process based on the biocompatible sulfur(VI) fluoride exchange (SuFEx) click chemistry. A high-throughput screening hit benzyl (cyanomethyl)carbamate (Ki = 8 µM) against a bacterial cysteine protease SpeB was modified with a SuFExable iminosulfur oxydifluoride [RN═S(O)F2] motif, rapidly diversified into 460 analogs in overnight reactions, and the products were directly screened to yield drug-like inhibitors with 480-fold higher potency (Ki = 18 nM). We showed that the improved molecule is active in a bacteria-host coculture. Since this SuFEx linkage reaction succeeds on picomole scale for direct screening, we anticipate our methodology can accelerate the development of robust biological probes and drug candidates.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Exotoxinas/antagonistas & inhibidores , Compuestos de Azufre/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Química Clic , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/toxicidad , Descubrimiento de Drogas , Exotoxinas/química , Exotoxinas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Jurkat , Microsomas Hepáticos/metabolismo , Prueba de Estudio Conceptual , Unión Proteica
13.
Infect Genet Evol ; 85: 104424, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32561294

RESUMEN

Pseudomonas aeruginosa (PA) is an opportunistic human pathogen responsible for causing serious infections in patients with cystic fibrosis. Infections caused by PA are difficult to treat and eradicate due to intrinsic and added resistance to antibiotic therapy. Therefore, it is necessary to establish effective prevention strategies against this infectious agent. In this study, a combination of immunoinformatic tools was applied to predict immunogenic and immunodominant regions in the structure of exotoxins commonly secreted as virulence factors in PA infection (ExoA, ExoS, ExoT, ExoU and ExoY). The peptides derived from exotoxins were evaluated for the potential affinity for human leukocyte antigen (HLA) I and HLA-II molecules, antigenicity score and toxicity profile. From an initial screening of 941 peptides, 13 (1.38%) were successful in all analyzes. The peptides with relevant immunogenic properties were mainly those derived from Exo A (10 / 76.9%). All peptides selected in the last analysis present a high population coverage rate based on the interaction of HLA alleles (95.36 ± 7.83%). Therefore, the peptides characterized in this study are recommended for in vitro and in vivo studies and can provide the basis for the rational design of a vaccine against PA.


Asunto(s)
Exotoxinas/química , Exotoxinas/inmunología , Antígenos HLA/química , Péptidos/química , Péptidos/inmunología , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/inmunología , Secuencia de Aminoácidos , Toxinas Bacterianas/inmunología , Simulación por Computador , Epítopos/química , Epítopos/inmunología , Antígenos HLA/inmunología , Humanos , Inmunogenicidad Vacunal , Simulación del Acoplamiento Molecular , Conformación Proteica , Infecciones por Pseudomonas/inmunología , Vacunas/química , Vacunas/inmunología , Factores de Virulencia/química , Factores de Virulencia/inmunología
14.
Hepatology ; 71(5): 1696-1711, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31520528

RESUMEN

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.


Asunto(s)
ADP Ribosa Transferasas/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Carcinoma Hepatocelular/terapia , Exotoxinas/uso terapéutico , Glipicanos/antagonistas & inhibidores , Inmunotoxinas/uso terapéutico , Neoplasias Hepáticas/terapia , Anticuerpos de Dominio Único/uso terapéutico , Factores de Virulencia/uso terapéutico , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/farmacología , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacología , Línea Celular Tumoral , Exotoxinas/química , Exotoxinas/farmacología , Humanos , Inmunotoxinas/química , Inmunotoxinas/farmacología , Ratones , Ratones Desnudos , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/farmacología , Factores de Virulencia/química , Factores de Virulencia/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
15.
Adv Mater ; 32(7): e1907348, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31879981

RESUMEN

Functional amyloids produced in bacteria as nanoscale inclusion bodies are intriguing but poorly explored protein materials with wide therapeutic potential. Since they release functional polypeptides under physiological conditions, these materials can be potentially tailored as mimetic of secretory granules for slow systemic delivery of smart protein drugs. To explore this possibility, bacterial inclusion bodies formed by a self-assembled, tumor-targeted Pseudomonas exotoxin (PE24) are administered subcutaneously in mouse models of human metastatic colorectal cancer, for sustained secretion of tumor-targeted therapeutic nanoparticles. These proteins are functionalized with a peptidic ligand of CXCR4, a chemokine receptor overexpressed in metastatic cancer stem cells that confers high selective cytotoxicity in vitro and in vivo. In the mouse models of human colorectal cancer, time-deferred anticancer activity is detected after the subcutaneous deposition of 500 µg of PE24-based amyloids, which promotes a dramatic arrest of tumor growth in the absence of side toxicity. In addition, long-term prevention of lymphatic, hematogenous, and peritoneal metastases is achieved. These results reveal the biomedical potential and versatility of bacterial inclusion bodies as novel tunable secretory materials usable in delivery, and they also instruct how therapeutic proteins, even with high functional and structural complexity, can be packaged in this convenient format.


Asunto(s)
Amiloide/metabolismo , Antineoplásicos/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Portadores de Fármacos/química , Cuerpos de Inclusión/metabolismo , Nanopartículas/química , Amiloide/administración & dosificación , Amiloide/efectos adversos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Proteínas Bacterianas/química , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Exotoxinas/química , Exotoxinas/metabolismo , Células HeLa , Humanos , Cuerpos de Inclusión/química , Ratones , Conformación Molecular , Terapia Molecular Dirigida , Metástasis de la Neoplasia/prevención & control , Células Madre Neoplásicas/metabolismo , Péptidos/química , Péptidos/metabolismo , Ingeniería de Proteínas , Receptores CXCR4/química , Proteínas Recombinantes/química
16.
ACS Appl Mater Interfaces ; 12(5): 5381-5388, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-31840972

RESUMEN

Five peptide ligands of four different cell surface receptors (nucleolin, CXCR1, CMKLR1, and CD44v6) have been evaluated as targeting moieties for triple-negative human breast cancers. Among them, the peptide F3, derived from phage display, promotes the fast and efficient internalization of a genetically fused green fluorescent protein (GFP) inside MDA-MB-231 cancer stem cells in a specific receptor-dependent fashion. The further engineering of this protein into the modular construct F3-RK-GFP-H6 and the subsequent construct F3-RK-PE24-H6 resulted in self-assembling polypeptides that organize as discrete and regular nanoparticles. These materials, 15-20 nm in size, show enhanced nucleolin-dependent cell penetrability. We show that the F3-RK-PE24-H6, based on the Pseudomonas aeruginosa exotoxin A (PE24) as a core functional domain, is highly cytotoxic over target cells. The combination of F3, the cationic peptide (RK)n, and the toxin domain PE24 in such unusual presentation appears as a promising approach to cell-targeted drug carriers in breast cancers and addresses selective drug delivery in otherwise difficult-to-treat triple-negative breast cancers.


Asunto(s)
Portadores de Fármacos/química , Nanoestructuras/química , Péptidos/química , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Exotoxinas/química , Exotoxinas/farmacología , Femenino , Humanos , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Pseudomonas aeruginosa/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Factores de Virulencia/química , Factores de Virulencia/farmacología , Exotoxina A de Pseudomonas aeruginosa
17.
J Cell Physiol ; 235(4): 3711-3720, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31578716

RESUMEN

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.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Toxinas Bacterianas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Citotoxinas/farmacocinética , Exotoxinas/farmacología , Nanopartículas del Metal/química , Factores de Virulencia/farmacología , ADP Ribosa Transferasas/química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/química , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Caspasa 3/genética , Caspasas/genética , Proliferación Celular/efectos de los fármacos , Citotoxinas/química , Escherichia coli/genética , Exotoxinas/química , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Microscopía Electrónica de Transmisión , Proteínas Proto-Oncogénicas c-bcl-2/genética , Plata/química , Plata/farmacología , Factores de Virulencia/química , Proteína X Asociada a bcl-2/genética , Exotoxina A de Pseudomonas aeruginosa
18.
J Pharm Sci ; 109(1): 104-115, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669121

RESUMEN

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.


Asunto(s)
Antineoplásicos/metabolismo , Inmunotoxinas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Ensayos Clínicos como Asunto/métodos , Citosol/efectos de los fármacos , Citosol/metabolismo , Exotoxinas/administración & dosificación , Exotoxinas/química , Exotoxinas/metabolismo , Humanos , Inmunotoxinas/administración & dosificación , Inmunotoxinas/química , Unión Proteica/fisiología , Estructura Secundaria de Proteína
19.
Toxins (Basel) ; 11(12)2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31835552

RESUMEN

The repeats-in-toxin (RTX) family represents a unique class of bacterial exoproteins. The first family members described were toxins from Gram-negative bacterial pathogens; however, additional members included exoproteins with diverse functions. Our review focuses on well-characterized RTX family toxins from Aggregatibacteractinomycetemcomitans (LtxA), Mannheimiahaemolytica (LktA), Bordetella pertussis (CyaA), uropathogenic Escherichia coli (HlyA), and Actinobacillus pleuropneumoniae (ApxIIIA), as well as the studies that have honed in on a single host cell receptor for RTX toxin interactions, the ß2 integrins. The ß2 integrin family is composed of heterodimeric members with four unique alpha subunits and a single beta subunit. ß2 integrins are only found on leukocytes, including neutrophils and monocytes, the first responders to inflammation following bacterial infection. The LtxA, LktA, HlyA, and ApxIIIA toxins target the shared beta subunit, thereby targeting all types of leukocytes. Specific ß2 integrin family domains are required for the RTX toxin's cytotoxic activity and are summarized here. Research examining the domains of the RTX toxins required for cytotoxic and hemolytic activity is also summarized. RTX toxins attack and kill phagocytic immune cells expressing a single integrin family, providing an obvious advantage to the pathogen. The critical question that remains, can the specificity of the RTX-ß2 integrin interaction be therapeutically targeted?


Asunto(s)
Proteínas Bacterianas , Toxinas Bacterianas , Citotoxinas , Exotoxinas , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidad , Citotoxinas/química , Citotoxinas/toxicidad , Exotoxinas/química , Exotoxinas/toxicidad , Interacciones Huésped-Patógeno , Humanos , Monocitos/inmunología , Neutrófilos/inmunología , Dominios Proteicos , Receptores de Superficie Celular/inmunología
20.
ACS Nano ; 13(10): 11912-11920, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31560513

RESUMEN

Nosocomial infections transmitted through airborne, droplet, aerosol, and particulate-transported modes pose substantial infection risks to patients and healthcare employees. In this study, we demonstrate a self-cleaning filter comprised of laser-induced graphene (LIG), a porous conductive graphene foam formed through photothermal conversion of a polyimide film by a commercial CO2 laser cutter. LIG was shown to capture particulates and bacteria. The bacteria cannot proliferate even when submerged in culture medium. Through a periodic Joule-heating mechanism, the filter readily reaches >300 °C. This destroys any microorganisms including bacteria, along with molecules that can cause adverse biological reactions and diseases. These molecules include pyrogens, allergens, exotoxins, endotoxins, mycotoxins, nucleic acids, and prions. Capitalizing on the high surface area and thermal stability of LIG, the utility of graphene for reduction of nosocomial infection in hospital settings is suggested.


Asunto(s)
Filtros de Aire , Grafito/química , Exotoxinas/química , Micotoxinas/química , Ácidos Nucleicos/química , Priones/química , Pirógenos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...