Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.384
Filtrar
1.
Biochemistry ; 63(20): 2580-2593, 2024 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-39359146

RESUMEN

As a traceless, bioreversible modification, the esterification of carboxyl groups in peptides and proteins has the potential to increase their clinical utility. An impediment is the lack of strategies to quantify esterase-catalyzed hydrolysis rates for esters in esterified biologics. We have developed a continuous Förster resonance energy transfer (FRET) assay for esterase activity based on a peptidic substrate and a protease, Glu-C, that cleaves a glutamyl peptide bond only if the glutamyl side chain is a free acid. Using pig liver esterase (PLE) and human carboxylesterases, we validated the assay with substrates containing simple esters (e.g., ethyl) and esters designed to be released by self-immolation upon quinone methide elimination. We found that simple esters were not cleaved by esterases, likely for steric reasons. To account for the relatively low rate of quinone methide elimination, we extended the mathematics of the traditional Michaelis-Menten model to conclude with a first-order intermediate decay step. By exploring two regimes of our substrate → intermediate → product (SIP) model, we evaluated the rate constants for the PLE-catalyzed cleavage of an ester on a glutamyl side chain (kcat/KM = 1.63 × 103 M-1 s-1) and subsequent spontaneous quinone methide elimination to regenerate the unmodified peptide (kI = 0.00325 s-1; t1/2 = 3.55 min). The detection of esterase activity was also feasible in the human intestinal S9 fraction. Our assay and SIP model increase the understanding of the release kinetics of esterified biologics and facilitate the rational design of efficacious peptide prodrugs.


Asunto(s)
Esterasas , Péptidos , Profármacos , Profármacos/química , Profármacos/metabolismo , Humanos , Animales , Péptidos/química , Péptidos/metabolismo , Porcinos , Esterasas/metabolismo , Esterasas/química , Transferencia Resonante de Energía de Fluorescencia , Hígado/enzimología , Cinética , Hidrólisis , Especificidad por Sustrato , Ésteres/química , Ésteres/metabolismo
2.
Int J Pharm ; 665: 124663, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39265854

RESUMEN

Elucidation of the role of gut microbiota in the metabolism of orally administered drugs may improve therapeutic effectiveness and contribute to the development of personalized medicine. In this study, ten different artificial gut microbiota (AGM), obtained by culturing fecal samples in a continuous fermentation system, were challenged for their metabolizing capacity on a panel of six glucocorticoids selected from either prodrugs or drugs. Data from metabolic stability assays highlighted that, while the hydrolysis-mediated conversion of prodrugs to drugs represented only a minor metabolic pathway, significant differences in the stability of parent compounds and in their conversion rates to multiple reductive metabolites were obtained for the selected drugs. In the latter case, a taxonomic composition-dependent ability to convert parent drugs to metabolites was observed. Indeed, the artificial microbial communities dominated by the genus Bacteroides showed the maximal conversion of parent glucocorticoids to several metabolites. Furthermore, the effect of drugs on AGM was also evaluated through shallow shotgun sequencing and flow cytometry-based total bacterial cell count highlighting that these drugs can affect both the taxonomic composition and growth performances of the human gut microbiota.


Asunto(s)
Heces , Microbioma Gastrointestinal , Glucocorticoides , Microbioma Gastrointestinal/efectos de los fármacos , Glucocorticoides/metabolismo , Glucocorticoides/administración & dosificación , Humanos , Heces/microbiología , Hidrólisis , Administración Oral , Profármacos/metabolismo , Fermentación
3.
Nature ; 632(8023): 39-49, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39085542

RESUMEN

In this Review, we explore natural product antibiotics that do more than simply inhibit an active site of an essential enzyme. We review these compounds to provide inspiration for the design of much-needed new antibacterial agents, and examine the complex mechanisms that have evolved to effectively target bacteria, including covalent binders, inhibitors of resistance, compounds that utilize self-promoted entry, those that evade resistance, prodrugs, target corrupters, inhibitors of 'undruggable' targets, compounds that form supramolecular complexes, and selective membrane-acting agents. These are exemplified by ß-lactams that bind covalently to inhibit transpeptidases and ß-lactamases, siderophore chimeras that hijack import mechanisms to smuggle antibiotics into the cell, compounds that are activated by bacterial enzymes to produce reactive molecules, and antibiotics such as aminoglycosides that corrupt, rather than merely inhibit, their targets. Some of these mechanisms are highly sophisticated, such as the preformed ß-strands of darobactins that target the undruggable ß-barrel chaperone BamA, or teixobactin, which binds to a precursor of peptidoglycan and then forms a supramolecular structure that damages the membrane, impeding the emergence of resistance. Many of the compounds exhibit more than one notable feature, such as resistance evasion and target corruption. Understanding the surprising complexity of the best antimicrobial compounds provides a roadmap for developing novel compounds to address the antimicrobial resistance crisis by mining for new natural products and inspiring us to design similarly sophisticated antibiotics.


Asunto(s)
Antibacterianos , Bacterias , Productos Biológicos , Animales , Humanos , Aminoglicósidos/farmacología , Aminoglicósidos/química , Aminoglicósidos/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/metabolismo , Bacterias/efectos de los fármacos , Bacterias/enzimología , Bacterias/metabolismo , Antibióticos Betalactámicos/química , Antibióticos Betalactámicos/farmacología , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/farmacología , Productos Biológicos/química , Productos Biológicos/farmacología , Productos Biológicos/metabolismo , Diseño de Fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Peptidil Transferasas/antagonistas & inhibidores , Profármacos/farmacología , Profármacos/química , Profármacos/metabolismo , Sideróforos/metabolismo , Sideróforos/química , Sideróforos/farmacología
4.
Sci Rep ; 14(1): 14175, 2024 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898077

RESUMEN

Central nervous system tumors have resisted effective chemotherapy because most therapeutics do not penetrate the blood-tumor-brain-barrier. Nanomedicines between ~ 10 and 100 nm accumulate in many solid tumors by the enhanced permeability and retention effect, but it is controversial whether the effect can be exploited for treatment of brain tumors. PLX038A is a long-acting prodrug of the topoisomerase 1 inhibitor SN-38. It is composed of a 15 nm 4-arm 40 kDa PEG tethered to four SN-38 moieties by linkers that slowly cleave to release the SN-38. The prodrug was remarkably effective at suppressing growth of intracranial breast cancer and glioblastoma (GBM), significantly increasing the life span of mice harboring them. We addressed the important issue of whether the prodrug releases SN-38 systemically and then penetrates the brain to exert anti-tumor effects, or whether it directly penetrates the blood-tumor-brain-barrier and releases the SN-38 cargo within the tumor. We argue that the amount of SN-38 formed systemically is insufficient to inhibit the tumors, and show by PET imaging that a close surrogate of the 40 kDa PEG carrier in PLX038A accumulates and is retained in the GBM. We conclude that the prodrug penetrates the blood-tumor-brain-barrier, accumulates in the tumor microenvironment and releases its SN-38 cargo from within. Based on our results, we pose the provocative question as to whether the 40 kDa nanomolecule PEG carrier might serve as a "Trojan horse" to carry other drugs past the blood-tumor-brain-barrier and release them into brain tumors.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Irinotecán , Profármacos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Irinotecán/farmacocinética , Barrera Hematoencefálica/metabolismo , Ratones , Profármacos/farmacocinética , Profármacos/química , Profármacos/metabolismo , Humanos , Línea Celular Tumoral , Femenino , Ensayos Antitumor por Modelo de Xenoinjerto , Glioblastoma/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Camptotecina/uso terapéutico
5.
Int J Mol Sci ; 25(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38928299

RESUMEN

Bacterial nitroreductase enzymes capable of activating imaging probes and prodrugs are valuable tools for gene-directed enzyme prodrug therapies and targeted cell ablation models. We recently engineered a nitroreductase (E. coli NfsB F70A/F108Y) for the substantially enhanced reduction of the 5-nitroimidazole PET-capable probe, SN33623, which permits the theranostic imaging of vectors labeled with oxygen-insensitive bacterial nitroreductases. This mutant enzyme also shows improved activation of the DNA-alkylation prodrugs CB1954 and metronidazole. To elucidate the mechanism behind these enhancements, we resolved the crystal structure of the mutant enzyme to 1.98 Å and compared it to the wild-type enzyme. Structural analysis revealed an expanded substrate access channel and new hydrogen bonding interactions. Additionally, computational modeling of SN33623, CB1954, and metronidazole binding in the active sites of both the mutant and wild-type enzymes revealed key differences in substrate orientations and interactions, with improvements in activity being mirrored by reduced distances between the N5-H of isoalloxazine and the substrate nitro group oxygen in the mutant models. These findings deepen our understanding of nitroreductase substrate specificity and catalytic mechanisms and have potential implications for developing more effective theranostic imaging strategies in cancer treatment.


Asunto(s)
Metronidazol , Nitroimidazoles , Nitrorreductasas , Nitrorreductasas/metabolismo , Nitrorreductasas/química , Nitrorreductasas/genética , Nitroimidazoles/química , Nitroimidazoles/metabolismo , Metronidazol/química , Metronidazol/metabolismo , Metronidazol/farmacología , Profármacos/metabolismo , Profármacos/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Tomografía de Emisión de Positrones/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Dominio Catalítico , Ingeniería de Proteínas , Modelos Moleculares , Aziridinas/química , Aziridinas/metabolismo
6.
Anal Sci ; 40(8): 1529-1535, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38748393

RESUMEN

This study introduces the α-rhamnose (Rham)-conjugated prodrug of SN-38 (Rham-SN-38) as a promising alternative to irinotecan. α-rhamnosidase, responsible for SN-38 release from Rham-SN-38, does not express in human cells, minimizing individual variability and side effects. The injection of the α-rhamnosidase into the tumor tissues makes it possible, for the first time, to activate the Rham-SN-38. Furthermore, α-rhamnosidase demonstrates significantly higher activity than carboxylesterase, the specific enzyme activating irinotecan. SN-38 release mediated by α-rhamnosidase completes within 2 h, with a kcat/Km value approximately 5.0 × 104-fold higher than that of irinotecan. The 50% inhibition concentration (IC50) of Rham-SN-38 against three types of cancer cells and one normal cell exceeds 4.5 × 103 nM. The addition of α-rhamnosidase significantly increases cytotoxicity, with IC50 comparable to free SN-38. The QIC50, an index reflecting the difference in cytotoxicity with and without α-rhamnosidase, exceeds approximately 1.0 × 102-fold. Rham-SN-38, synthesized in this study, demonstrates significant potential as a prodrug for cancer therapy.


Asunto(s)
Glicósido Hidrolasas , Irinotecán , Profármacos , Irinotecán/farmacología , Irinotecán/química , Humanos , Profármacos/farmacología , Profármacos/química , Profármacos/metabolismo , Profármacos/síntesis química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales , Relación Dosis-Respuesta a Droga , Línea Celular Tumoral
7.
Pharm Res ; 41(6): 1031-1044, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38740664

RESUMEN

Both intrinsic and extrinsic aging lead to a series of morphological changes in the skin including the flattening of the dermal-epidermal junction, increased stratum corneum dryness, reduction in sebaceous gland activity and enzyme activity as well as atrophy of blood vessels. In this study, the impact of these changes on the transport of molecules through the skin was revised. The increase in the number of transdermal formulations on the market in recent decades and life expectancy represent the main reasons for an in-depth discussion of this topic. Furthermore, elderly subjects have often been excluded from clinical trials due to polypharmacy, raising concerns in terms of efficacy and safety. In this way, ex vivo and in vivo studies comparing the transport of molecules through the mature and young skin were analyzed in detail. The reduced water content in mature skin had a significant impact on the transport rate of hydrophilic molecules. The lower enzymatic activity in aged skin, in turn, would explain changes in the activation of prodrugs. Interestingly, greater deposition of nanoparticles was also found in mature skin. In vivo models should be prioritized in future experimental studies as they allow to evaluate both absorption and metabolism simultaneously, providing more realistic information.


Asunto(s)
Administración Cutánea , Envejecimiento , Absorción Cutánea , Piel , Humanos , Piel/metabolismo , Envejecimiento/metabolismo , Animales , Transporte Biológico , Nanopartículas/metabolismo , Nanopartículas/química , Envejecimiento de la Piel , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Profármacos/farmacocinética , Profármacos/metabolismo
8.
J Med Chem ; 67(9): 7470-7486, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38690769

RESUMEN

We assessed factors that determine the tissue-specific bioactivation of ProTide prodrugs by comparing the disposition and activation of remdesivir (RDV), its methylpropyl and isopropyl ester analogues (MeRDV and IsoRDV, respectively), the oral prodrug GS-621763, and the parent nucleotide GS-441524 (Nuc). RDV and MeRDV yielded more active metabolite remdesivir-triphosphate (RDV-TP) than IsoRDV, GS-621763, and Nuc in human lung cell models due to superior cell permeability and higher susceptivity to cathepsin A. Intravenous administration to mice showed that RDV and MeRDV delivered significantly more RDV-TP to the lung than other compounds. Nevertheless, all four ester prodrugs exhibited very low oral bioavailability (<2%), with Nuc being the predominant metabolite in blood. In conclusion, ProTides prodrugs, such as RDV and MeRDV, are more efficient in delivering active metabolites to the lung than Nuc, driven by high cell permeability and susceptivity to cathepsin A. Optimizing ProTides' ester structures is an effective strategy for enhancing prodrug activation in the lung.


Asunto(s)
Adenosina/análogos & derivados , Antivirales , Catepsina A , Pulmón , Profármacos , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacocinética , Profármacos/farmacología , Animales , Ratones , Antivirales/farmacocinética , Antivirales/farmacología , Antivirales/química , Antivirales/metabolismo , Humanos , Catepsina A/metabolismo , Pulmón/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacocinética , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/química , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/química , Alanina/farmacocinética , Alanina/metabolismo , Alanina/farmacología , Permeabilidad , ProTides
9.
Angew Chem Int Ed Engl ; 63(31): e202404243, 2024 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-38747847

RESUMEN

6-Thioguanine (6TG) is a clinically used antitumor agent that was rationally designed as a DNA-targeting antimetabolite, but it also occurs naturally. 6TG is a critical virulence factor produced by Erwinia amylovorans, a notorious plant pathogen that causes fire blight of pome fruit trees. The biosynthesis of the rare thioamide metabolite involves an adenylating enzyme (YcfA) and a sulfur-mobilizing enzyme (YcfC), but the mechanism of sulfur transfer and putative intermediates have remained elusive. Through dissection and in vitro reconstitution of the thionation process using diverse substrates, we uncover an intermediate, prodrug-like thio-conjugate and elucidate the precise enzyme functions. YcfA not only adenylates GMP but also transfers the mercapto group of l-cysteine to the activated carbonyl. A designated C-S lyase (YcfC) then cleaves the resulting S-adduct to yield the thioamide. This pathway is distinct from canonical tRNA sulfur modifications and known enzymatic peptide thionations. By exploring a wide range of substrate surrogates, we exploited the tolerance of the enzyme pair to produce even a seleno analog. This study provides valuable insight into a previously unexplored area of bacterial thioamide formation and lays the groundwork for synthetic biology approaches to produce thioamide antimetabolites.


Asunto(s)
Profármacos , Tioamidas , Profármacos/química , Profármacos/metabolismo , Tioamidas/química , Tioamidas/metabolismo
10.
Biophys Chem ; 310: 107256, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38728807

RESUMEN

Understanding the mechanisms by which drugs interact with cell membranes is crucial for unraveling the underlying biochemical and biophysical processes that occur on the surface of these membranes. Our research focused on studying the interaction between an ester-type derivative of tristearoyl uridine and model cell membranes composed of lipid monolayers at the air-water interface. For that, we selected a specific lipid to simulate nontumorigenic cell membranes, namely 1,2-dihexadecanoyl-sn-glycero-3-phospho-l-serine. We noted significant changes in the surface pressure-area isotherms, with a noticeable shift towards larger areas, which was lower than expected for ideal mixtures, indicating monolayer condensation. Furthermore, the viscoelastic properties of the interfacial film demonstrated an increase in both the elastic and viscous parameters for the mixed film. We also observed structural alterations using vibrational spectroscopy, which revealed an increase in the all-trans to gauche conformers ratio. This confirmed the stiffening effect of the prodrug on the lipid monolayer. In summary, this study indicates that this lipophilic prodrug significantly impacts the lipid monolayer's thermodynamic, rheological, electrical, and molecular characteristics. This information is crucial for understanding how the drug interacts with specific sites on the cellular membrane. It also has implications for drug delivery, as the drug's passage into the cytosol may involve traversing the lipid bilayer.


Asunto(s)
Membrana Celular , Profármacos , Uridina , Profármacos/química , Profármacos/farmacología , Profármacos/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Uridina/química , Uridina/farmacología , Fosfatidilserinas/química , Termodinámica , Propiedades de Superficie , Viscosidad , Elasticidad
11.
Mol Cell ; 84(10): 1948-1963.e11, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38759627

RESUMEN

The yeast glucose-induced degradation-deficient (GID) E3 ubiquitin ligase forms a suite of complexes with interchangeable receptors that selectively recruit N-terminal degron motifs of metabolic enzyme substrates. The orthologous higher eukaryotic C-terminal to LisH (CTLH) E3 complex has been proposed to also recognize substrates through an alternative subunit, WDR26, which promotes the formation of supramolecular CTLH E3 assemblies. Here, we discover that human WDR26 binds the metabolic enzyme nicotinamide/nicotinic-acid-mononucleotide-adenylyltransferase 1 (NMNAT1) and mediates its CTLH E3-dependent ubiquitylation independently of canonical GID/CTLH E3-family substrate receptors. The CTLH subunit YPEL5 inhibits NMNAT1 ubiquitylation and cellular turnover by WDR26-CTLH E3, thereby affecting NMNAT1-mediated metabolic activation and cytotoxicity of the prodrug tiazofurin. Cryoelectron microscopy (cryo-EM) structures of NMNAT1- and YPEL5-bound WDR26-CTLH E3 complexes reveal an internal basic degron motif of NMNAT1 essential for targeting by WDR26-CTLH E3 and degron mimicry by YPEL5's N terminus antagonizing substrate binding. Thus, our data provide a mechanistic understanding of how YPEL5-WDR26-CTLH E3 acts as a modulator of NMNAT1-dependent metabolism.


Asunto(s)
Nicotinamida-Nucleótido Adenililtransferasa , Profármacos , Ubiquitina-Proteína Ligasas , Ubiquitinación , Humanos , Microscopía por Crioelectrón , Células HEK293 , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Nicotinamida-Nucleótido Adenililtransferasa/genética , Profármacos/metabolismo , Unión Proteica , Especificidad por Sustrato , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo
12.
ChemMedChem ; 19(14): e202400127, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38634306

RESUMEN

Hypoxia is a state characterized by low concentration of Oxygen. Hypoxic state is often found in the central region of solid tumors. Hypoxia is associated with abnormal neovascularization resulted in poor blood flow in tissues and increased proliferation of tumor cells, imbalance between O2 supply and O2 consumption in tumor cells, high concentration of proton and strong reducibility. And, these abnormalities enhance the survival potency of the hypoxic tumours and increase the resistance towards chemotherapy and radiotherapy. One of the approach for treating hypoxic region of tumour is to use reducing environment of hypoxic tumours for reducing a molecule (hypoxia activated prodrug, HAP) and as a result the active drug will be released in hypoxic region in a controlled manner from the prodrug and kill the hypoxic tumour. Co(III) and Pt(IV) complexes with monodentate active drug molecule in the axial position can be reduced to Co(II) and Pt(II) moieties and as a result, the axial ligands (active drug) could come out from the metal center and could show its anticancer activity. In this review we have highlighted the research articles where transition metal-based complexes are used as chemotherapeutic hypoxia activated prodrug molecules which are reported in last 5 years.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Profármacos , Profármacos/química , Profármacos/farmacología , Profármacos/síntesis química , Profármacos/metabolismo , Humanos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Elementos de Transición/química , Neoplasias/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Estructura Molecular , Ensayos de Selección de Medicamentos Antitumorales , Animales
13.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38673999

RESUMEN

E. coli nitroreductase A (NfsA) is a candidate for gene-directed prodrug cancer therapy using bioreductively activated nitroaromatic compounds (ArNO2). In this work, we determined the standard redox potential of FMN of NfsA to be -215 ± 5 mV at pH 7.0. FMN semiquinone was not formed during 5-deazaflavin-sensitized NfsA photoreduction. This determines the two-electron character of the reduction of ArNO2 and quinones (Q). In parallel, we characterized the oxidant specificity of NfsA with an emphasis on its structure. Except for negative outliers nitracrine and SN-36506, the reactivity of ArNO2 increases with their electron affinity (single-electron reduction potential, E17) and is unaffected by their lipophilicity and Van der Waals volume up to 386 Å. The reactivity of quinoidal oxidants is not clearly dependent on E17, but 2-hydroxy-1,4-naphthoquinones were identified as positive outliers and a number of compounds with diverse structures as negative outliers. 2-Hydroxy-1,4-naphthoquinones are characterized by the most positive reaction activation entropy and the negative outlier tetramethyl-1,4-benzoquinone by the most negative. Computer modelling data showed that the formation of H bonds with Arg15, Arg133, and Ser40, plays a major role in the binding of oxidants to reduced NfsA, while the role of the π-π interaction of their aromatic structures is less significant. Typically, the calculated hydride-transfer distances during ArNO2 reduction are smallwer than for Q. This explains the lower reactivity of quinones. Another factor that slows down the reduction is the presence of positively charged aliphatic substituents.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Nitrorreductasas , Oxidación-Reducción , Profármacos , Nitrorreductasas/metabolismo , Nitrorreductasas/química , Nitrorreductasas/genética , Profármacos/química , Profármacos/metabolismo , Especificidad por Sustrato , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Potenciometría , Catálisis , Simulación del Acoplamiento Molecular
14.
ACS Nano ; 18(11): 7945-7958, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38452275

RESUMEN

Ferroptosis is a form of regulated cell death accompanied by lipid reactive oxygen species (ROS) accumulation in an iron-dependent manner. However, the efficiency of tumorous ferroptosis was seriously restricted by intracellular ferroptosis defense systems, the glutathione peroxidase 4 (GPX4) system, and the ubiquinol (CoQH2) system. Inspired by the crucial role of mitochondria in the ferroptosis process, we reported a prodrug nanoassembly capable of unleashing potent mitochondrial lipid peroxidation and ferroptotic cell death. Dihydroorotate dehydrogenase (DHODH) inhibitor (QA) was combined with triphenylphosphonium moiety through a disulfide-containing linker to engineer well-defined nanoassemblies (QSSP) within a single-molecular framework. After being trapped in cancer cells, the acidic condition provoked the structural disassembly of QSSP to liberate free prodrug molecules. The mitochondrial membrane-potential-driven accumulation of the lipophilic cation prodrug was delivered explicitly into the mitochondria. Afterward, the thiol-disulfide exchange would occur accompanied by downregulation of reduced glutathione levels, thus resulting in mitochondria-localized GPX4 inactivation for ferroptosis. Simultaneously, the released QA from the hydrolysis reaction of the adjacent ester bond could further devastate mitochondrial defense and evoke robust ferroptosis via the DHODH-CoQH2 system. This subcellular targeted nanoassembly provides a reference for designing ferroptosis-based strategy for efficient cancer therapy through interfering antiferroptosis systems.


Asunto(s)
Ferroptosis , Compuestos Organofosforados , Profármacos , Profármacos/farmacología , Profármacos/metabolismo , Dihidroorotato Deshidrogenasa , Peroxidación de Lípido , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/metabolismo , Disulfuros/metabolismo
15.
Pharm Res ; 41(5): 849-861, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38485855

RESUMEN

PURPOSE: Olmesartan medoxomil (olmesartan-MX), an ester-type prodrug of the angiotensin II receptor blocker (ARB) olmesartan, is predominantly anionic at intestinal pH. Human organic anion transporting polypeptide 2B1 (OATP2B1) is expressed in the small intestine and is involved in the absorption of various acidic drugs. This study was designed to test the hypothesis that OATP2B1-mediated uptake contributes to the enhanced intestinal absorption of olmesartan-MX, even though olmesartan itself is not a substrate of OATP2B1. METHODS: Tetracycline-inducible human OATP2B1- and rat Oatp2b1-overexpressing HEK 293 cell lines (hOATP2B1/T-REx-293 and rOatp2b1/T-REx-293, respectively) were established to characterize OATP2B1-mediated uptake. Rat jejunal permeability was measured using Ussing chambers. ARBs were quantified by liquid chromatography-tandem mass spectrometry. RESULTS: Significant olmesartan-MX uptake was observed in hOATP2B1/T-REx-293 and rOatp2b1/T-REx-293 cells, whereas olmesartan uptake was undetectable or much lower than olmesartan-MX uptake, respectively. Furthermore, olmesartan-MX exhibited several-fold higher uptake in Caco-2 cells and greater permeability in rat jejunum compared to olmesartan. Olmesartan-MX uptake in hOATP2B1/T-REx-293 cells and in Caco-2 cells was significantly decreased by OATP2B1 substrates/inhibitors such as 1 mM estrone-3-sulfate, 100 µM rifamycin SV, and 100 µM fluvastatin. Rat Oatp2b1-mediated uptake and rat jejunal permeability of olmesartan-MX were significantly decreased by 50 µM naringin, an OATP2B1 inhibitor. Oral administration of olmesartan-MX with 50 µM naringin to rats significantly reduced the area under the plasma concentration-time curve of olmesartan to 76.9%. CONCLUSION: Olmesartan-MX is a substrate for OATP2B1, and the naringin-sensitive transport system contributes to the improved intestinal absorption of olmesartan-MX compared with its parent drug, olmesartan.


Asunto(s)
Imidazoles , Absorción Intestinal , Olmesartán Medoxomilo , Transportadores de Anión Orgánico , Profármacos , Tetrazoles , Animales , Humanos , Absorción Intestinal/efectos de los fármacos , Olmesartán Medoxomilo/metabolismo , Profármacos/farmacocinética , Profármacos/metabolismo , Células HEK293 , Tetrazoles/farmacocinética , Tetrazoles/metabolismo , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Masculino , Imidazoles/farmacocinética , Imidazoles/metabolismo , Ratas , Ratas Sprague-Dawley , Yeyuno/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Permeabilidad/efectos de los fármacos , Células CACO-2
16.
Mol Diagn Ther ; 28(2): 141-151, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38302842

RESUMEN

Necrosis is a common feature of solid tumours that offers a unique opportunity for targeted cancer therapy as it is absent from normal healthy tissues. Tumour necrosis provides an ideal environment for germination of the anaerobic bacterium Clostridium from endospores, resulting in tumour-specific colonisation. Two main species, Clostridium novyi-NT and Clostridium sporogenes, are at the forefront of this therapy, showing promise in preclinical models. However, anti-tumour activity is modest when used as a single agent, encouraging development of Clostridium as a tumour-selective gene delivery system. Various methods, such as allele-coupled exchange and CRISPR-cas9 technology, can facilitate the genetic modification of Clostridium, allowing chromosomal integration of transgenes to ensure long-term stability of expression. Strains of Clostridium can be engineered to express prodrug-activating enzymes, resulting in the generation of active drug selectively in the tumour microenvironment (a concept termed Clostridium-directed enzyme prodrug therapy). More recently, Clostridium strains have been investigated in the context of cancer immunotherapy, either in combination with immune checkpoint inhibitors or with engineered strains expressing immunomodulatory molecules such as IL-2 and TNF-α. Localised expression of these molecules using tumour-targeting Clostridium strains has the potential to improve delivery and reduce systemic toxicity. In summary, Clostridium species represent a promising platform for cancer therapy, with potential for localised gene delivery and immunomodulation selectively within the tumour microenvironment. The ongoing clinical progress being made with C. novyi-NT, in addition to developments in genetic modification techniques and non-invasive imaging capabilities, are expected to further progress Clostridium as an option for cancer treatment.


Asunto(s)
Neoplasias , Profármacos , Humanos , Composición de Base , Filogenia , ARN Ribosómico 16S , Análisis de Secuencia de ADN , Neoplasias/genética , Neoplasias/terapia , Clostridium/genética , Clostridium/metabolismo , Profármacos/metabolismo , Técnicas de Transferencia de Gen , Necrosis , Microambiente Tumoral
17.
Bioorg Med Chem ; 99: 117607, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38246114

RESUMEN

Various tenofovir (TFV) prodrugs have been developed by introducing masking groups to the hydroxyls of the monophosphonate group to enhance intestinal absorption efficiency and therapeutic effects. However, the reported TFV prodrugs have drawbacks such as low bioavailability, systemic toxicity caused by their breakdown in non-targeted tissues, and potential low intracellular conversion efficiency. In the present study, we developed a class of TFV monobenzyl ester phosphonoamidate prodrugs without substitutions on the benzene ring. Compared with previous TFV prodrugs, compounds 3a and 3b developed in the present study showed higher anti-hepatitis B virus activity, stronger stability and higher levels of intrahepatic enrichment of the metabolic product (TFV), indicating the potential of these compounds as novel prodrugs with high efficiency and low systemic toxicity for the treatment of hepatitis B.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , Profármacos , Humanos , Tenofovir/farmacología , Tenofovir/metabolismo , Tenofovir/uso terapéutico , Fármacos Anti-VIH/uso terapéutico , Adenina/farmacología , Adenina/uso terapéutico , Profármacos/metabolismo , Anticuerpos , Infecciones por VIH/tratamiento farmacológico
18.
Biotechnol Appl Biochem ; 71(1): 5-16, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37743549

RESUMEN

Suicide gene therapy involves introducing viral or bacterial genes into tumor cells, which enables the conversion of a nontoxic prodrug into a toxic-lethal drug. The application of the bacterial cytosine deaminase (bCD)/5-fluorocytosine (5-FC) approach has been beneficial and progressive within the current field of cancer therapy because of the enhanced bystander effect. The basis of this method is the preferential deamination of 5-FC to 5-fluorouracil by cancer cells expressing cytosine deaminase (CD), which strongly inhibits DNA synthesis and RNA function, effectively targeting tumor cells. However, the poor binding affinity of toward 5-FC compared to the natural substrate cytosine and/or inappropriate thermostability limits the clinical applications of this gene therapy approach. Nowadays, many genetic engineering studies have been carried out to solve and improve the activity of this enzyme. In the current review, we intend to discuss the biotechnological aspects of Escherichia coli CD, including its structure, functions, molecular cloning, and protein engineering. We will also explore its relevance in cancer clinical trials. By examining these aspects, we hope to provide a thorough understanding of E. coli CD and its potential applications in cancer therapy.


Asunto(s)
Citosina Desaminasa , Profármacos , Humanos , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Escherichia coli/metabolismo , Fluorouracilo/química , Flucitosina/farmacología , Flucitosina/metabolismo , Terapia Genética , Profármacos/metabolismo
19.
Xenobiotica ; 54(1): 10-17, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38142303

RESUMEN

1. Carboxylesterase (CES) has been studied extensively, mostly with substrates in the monoester structures. We investigated the relationship between indomethacin diester prodrugs and metabolic activation by microsomes and recombinant human CES.2. Eight indomethacin diester prodrugs were synthesised in two steps. They were used as substrates and hydrolysis rates were calculated.3. As a result, the major hydrolysis enzyme was CES. The hydrolysis rate of recombinant CES2A1 was comparable to that of recombinant CES1A1.4. In this study, by changing the structure of the prodrug to a diester structure, it was found that CES2 activity was equivalent to CES1 activity.5. It should be noted that the use of diester prodrugs in prodrug discovery, where organ-specific hydrolysis reactions are expected, may not yield the expected results.


Asunto(s)
Hidrolasas de Éster Carboxílico , Profármacos , Humanos , Hidrolasas de Éster Carboxílico/metabolismo , Indometacina , Profármacos/química , Profármacos/metabolismo , Carboxilesterasa/metabolismo , Microsomas/metabolismo , Hidrólisis
20.
PLoS One ; 18(11): e0294696, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38032917

RESUMEN

Gene-directed enzyme prodrug therapy is an emerging strategy for cancer treatment based on the delivery of a gene that encodes an enzyme that is able to convert a prodrug into a potent cytotoxin exclusively in target cancer cells. However, it is limited by the lack of suitable enzyme variants and a scarce choice of chemical bonds that could be activated. Therefore, this study is aimed to determine the capability of bacterial amidohydrolases YqfB and D8_RL to activate novel prodrugs and the effect such system has on the viability of eukaryotic cancer cells. We have established cancer cell lines that stably express the bacterial amidohydrolase genes and selected several N4-acylated cytidine derivatives as potential prodrugs. A significant decrease in the viability of HCT116 human colon cancer cell lines expressing either the YqfB or the D8_RL was observed after exposure to the novel prodrugs. The data we acquired suggests that bacterial YqfB and D8_RL amidohydrolases, together with the modified cytidine-based prodrugs, may serve as a promising enzyme-prodrug system for gene-directed enzyme prodrug therapy.


Asunto(s)
Antineoplásicos , Neoplasias del Colon , Profármacos , Humanos , Profármacos/metabolismo , Amidohidrolasas/genética , Citidina/farmacología , Neoplasias del Colon/tratamiento farmacológico , Antineoplásicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...