Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 82
1.
ACS Chem Neurosci ; 15(9): 1813-1827, 2024 05 01.
Article En | MEDLINE | ID: mdl-38621296

Acetylcholinesterase (AChE) inhibition by organophosphorus (OP) compounds poses a serious health risk to humans. While many therapeutics have been tested for treatment after OP exposure, there is still a need for efficient reactivation against all kinds of OP compounds, and current oxime therapeutics have poor blood-brain barrier penetration into the central nervous system, while offering no recovery in activity from the OP-aged forms of AChE. Herein, we report a novel library of 4-amidophenol quinone methide precursors (QMP) that provide effective reactivation against multiple OP-inhibited forms of AChE in addition to resurrecting the aged form of AChE after exposure to a pesticide or some phosphoramidates. Furthermore, these QMP compounds also reactivate OP-inhibited butyrylcholinesterase (BChE) which is an in vivo, endogenous scavenger of OP compounds. The in vitro efficacies of these QMP compounds were tested for reactivation and resurrection of soluble forms of human AChE and BChE and for reactivation of cholinesterases within human blood as well as blood and brain samples from a humanized mouse model. We identify compound 10c as a lead candidate due to its broad-scope efficacy against multiple OP compounds as well as both cholinesterases. With methylphosphonates, compound 10c (250 µM, 1 h) shows >60% recovered activity from OEt-inhibited AChE in human blood as well as mouse blood and brain, thus highlighting its potential for future in vivo analysis. For 10c, the effective concentration (EC50) is less than 25 µM for reactivation of three different methylphosphonate-inhibited forms of AChE, with a maximum reactivation yield above 80%. Similarly, for OP-inhibited BChE, 10c has EC50 values that are less than 150 µM for two different methylphosphonate compounds. Furthermore, an in vitro kinetic analysis show that 10c has a 2.2- and 92.1-fold superior reactivation efficiency against OEt-inhibited and OiBu-inhibited AChE, respectively, when compared to an oxime control. In addition to 10c being a potent reactivator of AChE and BChE, we also show that 10c is capable of resurrecting (ethyl paraoxon)-aged AChE, which is another current limitation of oximes.


Acetylcholinesterase , Butyrylcholinesterase , Cholinesterase Inhibitors , Cholinesterase Reactivators , Organophosphorus Compounds , Animals , Cholinesterase Inhibitors/pharmacology , Humans , Acetylcholinesterase/metabolism , Acetylcholinesterase/drug effects , Mice , Butyrylcholinesterase/metabolism , Organophosphorus Compounds/pharmacology , Cholinesterase Reactivators/pharmacology , Cholinesterase Reactivators/chemistry , Indolequinones/pharmacology
2.
Bioorg Chem ; 130: 106270, 2023 01.
Article En | MEDLINE | ID: mdl-36399864

Arylboronic acid/esters and phenyl selenides-based quinone methide (QM) precursors were reported to induce DNA interstrand crosslink (ICL) formation upon reaction with the inherently high concentrations of H2O2 in cancer cells. However, some normal cells (such as macrophages) also contain high-levels of H2O2, which may interfere with precursors' selectivity. In order to enhance the spatiotemporal specificity by the photolysis, we developed photo- and H2O2- dual-responsive DNA ICL precursors 1-3, bearing a photo-responsive coumarin moiety and a H2O2 inducible phenyl selenide group. Precursors 1-3 are efficiently activated by photoirradiation and H2O2 to generate reactive QMs crosslinking DNA. Moreover, the reactivity of precursors can be modulated by the introduction of aromatic substituents (OMe, F), and the electron donating group (OMe) displays a more pronounced promoting effect on DNA ICL formation. A subsequent piperidine heat stability study confirmed that the formed QMs primarily alkylate dAs, dGs and dCs in DNA. Furthermore, 1-3 inhibit lung cancer cell (H1299) growth by inducing DNA damage and producing toxic reactive oxygen species (ROS) upon photolysis of released coumarin. This study illustrates the potent cytotoxicity achieved by novel photo/H2O2 dual-responsive QM precursors 1-3, affording a novel strategy for the development of inducible DNA interstrand cross-linkers.


Cross-Linking Reagents , Hydrogen Peroxide , Indolequinones , Coumarins/chemistry , DNA Damage/drug effects , Hydrogen Peroxide/pharmacology , Indolequinones/pharmacology , Photolysis , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Lung Neoplasms/drug therapy , Humans , Cell Line, Tumor
3.
Biomed Pharmacother ; 156: 113893, 2022 Dec.
Article En | MEDLINE | ID: mdl-36279719

Compounds containing Michael acceptor units display a wide variety of biological effects, and have attracted much attention in medicinal chemistry. In this paper, we designed and synthesized a panel of para-quinone methides (p-QMs) derivatives, classified as electron-deficient alkenes, and evaluated their cytotoxicity against cancer cells. These results revealed that drawing substituents into the ortho-position of the phenyl ring could obviously strengthen the cytotoxicity of p-QMs derivatives compared with that of meta- and para-substituents. Further biological studies demonstrated that the cytotoxicity of p-QMs derivatives originated from their ROS-generation abilities, which could further disrupt the redox balance, lipid peroxidation, the loss of MMP, cell cycle arrest at G0/G1 phase and apoptosis. 1h also exhibited potent antitumor activity through inhibiting TrxR and activating Bax and caspase 3 expression in vitro and in vivo, and 1h had certain safety in vivo. Moreover, the electrophilicity of the Michael acceptor, which could covalently modify with the TrxR, play a potent role in the ROS generation. From the perspective of chemistry, we affirmed that p-QMs derivatives could rapidly covalent binding with cysteamine, and the addition product was characterized by 1H NMR. Together, these new p-QMs derivatives may possess potential as leads for development of effective antitumor agents.


Antineoplastic Agents , Indolequinones , Reactive Oxygen Species/metabolism , Indolequinones/pharmacology , Indolequinones/chemistry , Antineoplastic Agents/chemistry , Apoptosis
4.
J Org Chem ; 87(10): 6730-6741, 2022 05 20.
Article En | MEDLINE | ID: mdl-35545917

Multicomponent synthesis of biologically relevant S-benzyl dithiocarbamates from para-quinone methides, amines, and carbon disulfide are described under catalyst and additive-free conditions. The reactions proceeded at room temperature in a short span of time with excellent yields. One of the synthesized compounds, 3e showed considerable acetylcholinesterase (AChE) inhibitory (51.70 + 5.63% at 20 µm) and antioxidant (63.52 ± 1.15 at 20 µm) activities.


Alzheimer Disease , Indolequinones , Acetylcholinesterase , Alzheimer Disease/drug therapy , Amines , Humans , Indolequinones/pharmacology
5.
Chem Commun (Camb) ; 58(42): 6160-6175, 2022 May 24.
Article En | MEDLINE | ID: mdl-35522910

Quinone methides (QMs) are considered to be highly reactive intermediates because of their aromatization both in chemical and biological systems. Being highly accessible, quinone methides (QMs) have been widely exploited and their concurrent use has been manifested for the synthesis of tertiary and quaternary carbon centers of bioactives, drugs and drug-like molecules. In this feature article, the synthetic routes, structure-reactivity relationships and synthetic applications of quinone methides are discussed. Formation of the intermediates during bioactivation of different chemical entities and possible chemical manifestations leading to their toxicity in biological systems are also covered.


Indolequinones , Pharmaceutical Preparations , Humans , Indolequinones/chemical synthesis , Indolequinones/pharmacology , Indolequinones/toxicity , Pharmaceutical Preparations/chemical synthesis
6.
Bioorg Med Chem ; 44: 116281, 2021 08 15.
Article En | MEDLINE | ID: mdl-34216983

Quinone methide (QM) species have been included in the design of various functional molecules. In this review, we present a comprehensive overview of bioanalytical tools based on QM chemistry. In the first part, we focus on self-immolative linkers that have been incorporated into functional molecules such as prodrugs and fluorescent probes. In the latter half, we outline how the highly electrophilic property of QMs, enabling them to react rapidly with neighboring nucleophiles, has been applied to develop inhibitors or labeling probes for enzymes, as well as self-immobilizing fluorogenic probes with high spatial resolution. This review systematically summarizes the versatile QM toolbox available for investigating biological processes.


Alkaline Phosphatase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacology , Indolequinones/pharmacology , beta-Galactosidase/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Animals , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Indolequinones/chemical synthesis , Indolequinones/chemistry , Molecular Structure , beta-Galactosidase/metabolism
7.
Molecules ; 26(11)2021 Jun 02.
Article En | MEDLINE | ID: mdl-34199541

Quinone methide precursors 1a-e, with different alkyl linkers between the naphthol and the naphthalimide chromophore, were synthesized. Their photophysical properties and photochemical reactivity were investigated and connected with biological activity. Upon excitation of the naphthol, Förster resonance energy transfer (FRET) to the naphthalimide takes place and the quantum yields of fluorescence are low (ΦF ≈ 10-2). Due to FRET, photodehydration of naphthols to QMs takes place inefficiently (ΦR ≈ 10-5). However, the formation of QMs can also be initiated upon excitation of naphthalimide, the lower energy chromophore, in a process that involves photoinduced electron transfer (PET) from the naphthol to the naphthalimide. Fluorescence titrations revealed that 1a and 1e form complexes with ct-DNA with moderate association constants Ka ≈ 105-106 M-1, as well as with bovine serum albumin (BSA) Ka ≈ 105 M-1 (1:1 complex). The irradiation of the complex 1e@BSA resulted in the alkylation of the protein, probably via QM. The antiproliferative activity of 1a-e against two human cancer cell lines (H460 and MCF 7) was investigated with the cells kept in the dark or irradiated at 350 nm, whereupon cytotoxicity increased, particularly for 1e (>100 times). Although the enhancement of this activity upon UV irradiation has no imminent therapeutic application, the results presented have importance in the rational design of new generations of anticancer phototherapeutics that absorb visible light.


Antineoplastic Agents/pharmacology , Indolequinones/chemical synthesis , Naphthalimides/chemistry , Naphthols/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Fluorescence Resonance Energy Transfer , Humans , Indolequinones/chemistry , Indolequinones/pharmacology , MCF-7 Cells , Molecular Structure , Photochemical Processes , Quantum Theory
8.
Invest New Drugs ; 39(5): 1232-1241, 2021 10.
Article En | MEDLINE | ID: mdl-33768386

Radiotherapy is an effective treatment modality for breast cancer but, unfortunately, not all patients respond fully with a significant number experiencing local recurrences. Overexpression of thioredoxin and thioredoxin reductase has been reported to cause multidrug and radiation resistance - their inhibition may therefore improve therapeutic efficacy. Novel indolequinone compounds have been shown, in pancreatic cancer models, to inhibit thioredoxin reductase activity and exhibit potent anticancer activity. The present study evaluates, using in vitro breast cancer models, the efficacy of a novel indolequinone compound (IQ9) as a single agent and in combination with ionising radiation using a variety of endpoint assays including cell proliferation, clonogenic survival, enzyme activity, and western blotting. Three triple-negative breast cancer (MDA-MB-231, MDA-MB-468, and MDA-MB-436) and two luminal (MCF-7 and T47D) breast cancer cell lines were used. Results show that treatment with IQ9 significantly inhibited thioredoxin reductase activity, and inhibited cell growth and colony formation of breast cancer cells with IC50 values in the low micromolar ranges. Enhanced radiosensitivity of triple-negative breast cancer cells was observed, with sensitiser enhancement ratios of 1.20-1.43, but with no evident radiosensitisation of luminal breast cancer cell lines. IQ9 upregulated protein expression of thioredoxin reductase in luminal but not in triple-negative breast cancer cells which may explain the observed differential radiosensitisation. This study provides important evidence of the roles of the thioredoxin system as an exploitable radiobiological target in breast cancer cells and highlights the potential therapeutic value of indolequinones as radiosensitisers.***This study was not part of a clinical trial. Clinical trial registration number: N/A.


Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Indolequinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Thioredoxins/drug effects , Triple Negative Breast Neoplasms/pathology
9.
Chembiochem ; 22(5): 894-903, 2021 03 02.
Article En | MEDLINE | ID: mdl-33105515

Quinone methide (QM) chemistry is widely applied including in enzyme inhibitors. Typically, enzyme-mediated bond breaking releases a phenol product that rearranges into an electrophilic QM that in turn covalently modifies protein side chains. However, the factors that govern the reactivity of QM-based inhibitors and their mode of inhibition have not been systematically explored. Foremost, enzyme inactivation might occur in cis, whereby a QM molecule inactivates the very same enzyme molecule that released it, or by trans if the released QMs diffuse away and inactivate other enzyme molecules. We examined QM-based inhibitors for enzymes exhibiting phosphoester hydrolase activity. We tested different phenolic substituents and benzylic leaving groups, thereby modulating the rates of enzymatic hydrolysis, phenolate-to-QM rearrangement, and the electrophilicity of the resulting QM. By developing assays that distinguish between cis and trans inhibition, we have identified certain combinations of leaving groups and phenyl substituents that lead to inhibition in the cis mode, while other combinations gave trans inhibition. Our results suggest that cis-acting QM-based substrates could be used as activity-based probes to identify various phospho- and phosphono-ester hydrolases, and potentially other hydrolases.


Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indolequinones/chemistry , Indolequinones/pharmacology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Hydrolysis , Organophosphates/metabolism
10.
Curr Top Med Chem ; 20(3): 192-208, 2020.
Article En | MEDLINE | ID: mdl-31868148

BACKGROUND: According to the World Health Organization, antimicrobial resistance is one of the most important public health threats of the 21st century. Therefore, there is an urgent need for the development of antimicrobial agents with new mechanism of action, especially those capable of evading known resistance mechanisms. OBJECTIVE: We described the synthesis, in vitro antimicrobial evaluation, and in silico analysis of a series of 1H-indole-4,7-dione derivatives. METHODS: The new series of 1H-indole-4,7-diones was prepared with good yield by using a copper(II)- mediated reaction between bromoquinone and ß-enamino ketones bearing alkyl or phenyl groups attached to the nitrogen atom. The antimicrobial potential of indole derivatives was assessed. Molecular docking studies were also performed using AutoDock 4.2 for Windows. Characterization of all compounds was confirmed by one- and two-dimensional NMR techniques 1H and 13C NMR spectra [1H, 13C - APT, 1H x 1H - COSY, HSQC and HMBC], IR and mass spectrometry analysis. RESULTS: Several indolequinone compounds showed effective antimicrobial profile against Grampositive (MIC = 16 µg.mL-1) and Gram-negative bacteria (MIC = 8 µg.mL-1) similar to antimicrobials current on the market. The 3-acetyl-1-(2,5-dimethylphenyl)-1H-indole-4,7-dione derivative exhibited an important effect against different biofilm stages formed by a serious hospital life-threatening resistant strain of Methicillin-Resistant Staphylococcus aureus (MRSA). A hemocompatibility profile analysis based on in vitro hemolysis assays revealed the low toxicity effects of this new series. Indeed, in silico studies showed a good pharmacokinetics and toxicological profiles for all indolequinone derivatives, reinforcing their feasibility to display a promising oral bioavailability. An elucidation of the promising indolequinone derivatives binding mode was achieved, showing interactions with important sites to biological activity of S. aureus DNA gyrase. These results highlighted 3-acetyl-1-(2-hydroxyethyl)-1Hindole- 4,7-dione derivative as broad-spectrum antimicrobial prototype to be further explored for treating bacterial infections. CONCLUSION: The highly substituted indolequinones were obtained in moderate to good yields. The pharmacological study indicated that these compounds should be exploited in the search for a leading substance in a project aimed at obtaining new antimicrobials effective against Gram-negative bacteria.


Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Indolequinones/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Humans , Indolequinones/chemical synthesis , Indolequinones/chemistry , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure
11.
Drug Dev Res ; 80(7): 970-980, 2019 11.
Article En | MEDLINE | ID: mdl-31348537

In recent studies, we have investigated the monoamine oxidase (MAO) inhibition properties of pyrrolo[3,4-f]indole-5,7-dione and indole-5,6-dicarbonitrile derivatives. Since numerous high potency MAO inhibitors are present among these chemical classes, the present study synthesizes 44 additional derivatives in an attempt to further derive structure-activity relationships (SARs) and to establish optimal substitution patterns for MAO inhibition. The results show that, with the exception of one compound, all indole-5,6-dicarbonitrile derivatives (10) exhibit submicromolar IC50 values for the inhibition of MAO, with the most potent MAO-A inhibitor exhibiting an IC50 value of 0.006 µM while the most potent MAO-B inhibitor exhibits an IC50 value of 0.058 µM. Interestingly, an N-oxide derivative (4c) also proved to be a potent and nonspecific MAO inhibitor. With the exception of one compound, all of the pyrrolo[3,4-f]indole-5,7-diones (28) also exhibit submicromolar IC50 values for the inhibition of an MAO isoform. The most potent inhibitor exhibit an IC50 value of 0.011 µM for MAO-A. This study proposes that high potency MAO inhibitors such as those investigated here, may act as lead compounds for the development of treatments for neurodegenerative and neuropsychiatric disorders such as Parkinson's disease and depression.


Indolequinones/pharmacology , Indoles/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Nitriles/pharmacology , Humans , Indolequinones/chemical synthesis , Indoles/chemical synthesis , Inhibitory Concentration 50 , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Nitriles/chemical synthesis , Structure-Activity Relationship
12.
Org Lett ; 21(11): 4137-4142, 2019 06 07.
Article En | MEDLINE | ID: mdl-31094530

Cyanoalkylated diarylmethanes with biological and pharmacological potentials were synthesized from para-quinone methides and cyanoalkylating reagents through a photocatalytic process. This protocol is operationally simple and mild and has high efficiency, which gave the corresponding products in moderate to good yields. The synthetic utility of this work has been illustrated in the efficient synthesis of GPR40 agonists, which play an important role in FA-induced glucose-sensitive insulin secretion.


Indolequinones/pharmacology , Light , Receptors, G-Protein-Coupled/agonists , Alkylation , Humans , Indolequinones/chemical synthesis , Indolequinones/chemistry , Molecular Structure
13.
Bioorg Med Chem Lett ; 29(11): 1304-1307, 2019 06 01.
Article En | MEDLINE | ID: mdl-30975626

Among the various enzymes, reductases that catalyze one-electron reduction are involved in the selective activation of functional compounds or materials in hypoxia, which is one of the well-known pathophysiological characteristics of solid tumors. Enzymatic one-electron reduction has been recognized as a useful reaction that can be applied in the design of tumor hypoxia-targeting drugs. In this report, we characterized the enzymatic reaction of 5-fluorodeoxyuridine (FdUrd) prodrug bearing an indolequinone unit (IQ-FdUrd), which is a substrate of reductases. IQ-FdUrd was activated to release FdUrd under hypoxic conditions after treatment with cytochrome NADPH P450 reductase. We also confirmed that IQ-FdUrd showed selective cytotoxicity in hypoxic tumor cells.


Cell Hypoxia/drug effects , Floxuridine/pharmacology , Indolequinones/pharmacology , NADPH-Ferrihemoprotein Reductase/metabolism , Prodrugs/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Floxuridine/chemistry , Floxuridine/metabolism , Humans , Indolequinones/chemistry , Indolequinones/metabolism , Molecular Structure , NADP/metabolism , Prodrugs/chemistry , Prodrugs/metabolism , Structure-Activity Relationship
14.
Cancer Chemother Pharmacol ; 83(6): 1183-1189, 2019 06.
Article En | MEDLINE | ID: mdl-30868237

PURPOSE: Despite positive responses in phase II clinical trials, the bioreductive prodrug apaziquone failed to achieve statistically significant activity in non-muscle invasive bladder cancer in phase III trials. Apaziquone was administered shortly after transurethral resection and here we test the hypothesis that haematuria inactivates apaziquone. METHODS: HPLC analysis was used to determine the ability of human whole blood to metabolise apaziquone ex vivo. An in vitro model of haematuria was developed and the response of RT112 and EJ138 cells following a 1-h exposure to apaziquone was determined in the presence of urine plus or minus whole blood or lysed whole blood. RESULTS: HPLC analysis demonstrated that apaziquone is metabolised by human whole blood with a half-life of 78.6 ± 23.0 min. As a model for haematuria, incubation of cells in media containing up to 75% buffered (pH 7.4) urine and 25% whole blood was not toxic to cells for a 1-h exposure period. Whole blood (5% v/v) significantly (p < 0.01) reduced the potency of apaziquone in this experimental model. Lysed whole blood also significantly (p < 0.05) reduced cell growth, although higher concentrations were required to achieve an effect (15% v/v). CONCLUSIONS: The results of this study demonstrate that haematuria can reduce the potency of apaziquone in this experimental model. These findings impact upon the design of further phase III clinical trials and strongly suggest that apaziquone should not be administered immediately after transurethral resection of non-muscle invasive bladder cancer when haematuria is common.


Antineoplastic Agents/administration & dosage , Aziridines/administration & dosage , Chromatography, High Pressure Liquid , Hematuria/complications , Indolequinones/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aziridines/pharmacokinetics , Aziridines/pharmacology , Cell Line, Tumor , Half-Life , Humans , In Vitro Techniques , Indolequinones/pharmacokinetics , Indolequinones/pharmacology , Research Design
15.
J Neurochem ; 149(4): 535-550, 2019 05.
Article En | MEDLINE | ID: mdl-30592774

Targeting epigenetic mechanisms has shown promise against several cancers but has so far been unsuccessful against glioblastoma (GBM). Altered histone 3 lysine 4 methylation and increased lysine-specific histone demethylase 1A (LSD1) expression in GBM tumours nonetheless suggest that epigenetic mechanisms are involved in GBM. We engineered a dual-action prodrug, which is activated by the high hydrogen peroxide levels associated with GBM cells. This quinone methide phenylaminecyclopropane prodrug releases the LSD1 inhibitor 2-phenylcyclopropylamine with the glutathione scavenger para-quinone methide to trigger apoptosis in GBM cells. Quinone methide phenylaminocyclopropane impaired GBM cell behaviours in two-dimensional and three-dimensional assays, and triggered cell apoptosis in several primary and immortal GBM cell cultures. These results support our double-hit hypothesis of potentially targeting LSD1 and quenching glutathione, in order to impair and kill GBM cells but not healthy astrocytes. Our data suggest this strategy is effective at selectively targeting GBM and potentially other types of cancers. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Glioblastoma/pathology , Prodrugs/pharmacology , Aniline Compounds/pharmacology , Cells, Cultured , Cyclopropanes/pharmacology , Drug Design , Glutathione/antagonists & inhibitors , Histone Demethylases/antagonists & inhibitors , Humans , Indolequinones/pharmacology
16.
ACS Chem Neurosci ; 8(10): 2247-2253, 2017 10 18.
Article En | MEDLINE | ID: mdl-28763613

Aminochrome, an orthoquinone formed during the dopamine oxidation of neuromelanin, is neurotoxic because it induces mitochondria dysfunction, protein degradation dysfunction (both autophagy and proteasomal systems), α-synuclein aggregation to neurotoxic oligomers, neuroinflammation, and oxidative and endoplasmic reticulum stress. In this study, we investigated the relationship between aminochrome-induced autophagy/lysosome dysfunction and mitochondrial dysfunction in U373MGsiGST6 cells. Aminochrome (75 µM) induces mitochondrial dysfunction as determined by (i) a significant decrease in ATP levels (70%; P < 0.001) and (ii) a significant decrease in mitochondrial membrane potential (P < 0.001). Interestingly, the pretreatment of U373MGsiGST6 cells with 100 nM bafilomycin-A1, an inhibitor of lysosomal vacuolar-type H+-ATPase, restores ATP levels, mitochondrial membrane potential, and mitophagy, and decreases cell death. These results reveal (i) the importance of macroautophagy/the lysosomal degradation system for the normal functioning of mitochondria and for cell survival, and (ii) aminochrome-induced lysosomal dysfunction depends on the aminochrome-dependent inactivation of the vacuolar-type H+-ATPase, which pumps protons into the lysosomes. This study also supports the proposed protective role of glutathione transferase mu2-2 (GSTM2) in astrocytes against aminochrome toxicity, mediated by mitochondrial and lysosomal dysfunction.


Macrolides/pharmacology , Mitochondria/drug effects , Mitophagy/physiology , Autophagy/physiology , Cell Line, Tumor , Dopamine/metabolism , Humans , Indolequinones/pharmacology , Lysosomes/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , alpha-Synuclein/metabolism
17.
Eur J Med Chem ; 142: 95-130, 2017 Dec 15.
Article En | MEDLINE | ID: mdl-28754470

Natural pentacyclic triterpenoids (PTs) have been often reported to exhibit a wide range of biological activities. Among them, the anticancer and anti-inflammatory activities are the most studied. Over the last two decades, the number of publications reporting the anticancer effects of PTs has risen exponentially, reflecting the increasing interest in these natural products for the development of new antineoplastic drugs. Among of the most investigated PTs regarding their anticancer properties are oleanane-, ursane and friedelane-types, including oleanolic, glycyrrhetinic, ursolic and asiatic acids, and celastrol, among others. The extensive research in this field shows that the anticancer effects of PTs are mediated by several mechanisms, as they modulate a diverse range of molecular targets and signaling pathways, involved in cancer cell proliferation and survival. Considering the anticancer potential of this class of compounds, a number of semisynthetic derivatives has been synthetized aiming to improve their therapeutic activity and pharmacokinetic properties, and decrease their toxicity. Some of these new semisynthetic derivatives have shown improved anticancer activity in various cancer cell lines and animal models compared with the parent compound. Moreover, some of these compounds have been assessed in clinical trials, proving to be safe for human use. This review updates the most recent findings on the semisynthetic derivatives of oleanane-, ursane- and quinone methide friedelane-type PTs with anticancer activity. A brief introduction concerning the PTs and their anticancer activity is given, and the main semisynthetic modifications that have been performed between 2012 and early 2017 are reviewed and discussed.


Antineoplastic Agents/pharmacology , Indolequinones/pharmacology , Neoplasms/drug therapy , Oleanolic Acid/analogs & derivatives , Triterpenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Chemistry Techniques, Synthetic , Humans , Indolequinones/chemical synthesis , Indolequinones/chemistry , Indolequinones/therapeutic use , Oleanolic Acid/chemical synthesis , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Structure-Activity Relationship , Triterpenes/chemical synthesis , Triterpenes/chemistry , Triterpenes/therapeutic use
18.
Expert Opin Drug Metab Toxicol ; 13(7): 783-791, 2017 Jul.
Article En | MEDLINE | ID: mdl-28637373

INTRODUCTION: Apaziquone (also known as EO9 and QapzolaTM) is a prodrug that is activated to DNA damaging species by oxidoreductases (particularly NQO1) and has the ability to kill aerobic and/or hypoxic cancer cells. Areas covered: Whilst its poor pharmacokinetic properties contributed to its failure in phase II clinical trials when administered intravenously, these properties were ideal for loco-regional therapies. Apaziquone demonstrated good anti-cancer activity against non-muscle invasive bladder cancer (NMIBC) when administered intravesically to marker lesions and was well tolerated with no systemic side effects. However, phase III clinical trials did not reach statistical significance for the primary endpoint of 2-year recurrence in apaziquone over placebo although improvements were observed. Post-hoc analysis of the combined study data did indicate a significant benefit for patients treated with apaziquone, especially when the instillation of apaziquone was given 30 min or more after surgery. A further phase III study is ongoing to test the hypotheses generated in the unsuccessful phase III studies conducted to date. Expert opinion: Because of its specific pharmacological properties, Apaziquone is excellently suited for local therapy such as NMIBC. Future studies should include proper biomarkers.


Antineoplastic Agents/administration & dosage , Aziridines/administration & dosage , Indolequinones/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aziridines/pharmacokinetics , Aziridines/pharmacology , Humans , Indolequinones/pharmacokinetics , Indolequinones/pharmacology , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Urinary Bladder Neoplasms/pathology
19.
Free Radic Res ; 51(5): 545-553, 2017 May.
Article En | MEDLINE | ID: mdl-28503967

Serotonin (5-hydroxytryptamine) is a putative substrate for myeloperoxidase, which may convert it into the reactive quinone tryptamine-4,5-dione (TD). In this study, we found that the viability of human SH-SY5Y neuroblastoma cells treated with 25 µM TD was increased to approximately 117%. On the other hand, the cell viability was significantly decreased by exposure to TD (150-200 µM), with an increase in intracellular reactive oxygen species (ROS). Interestingly, pre-treatment of SH-SY5Y cells with 100 µM TD prevented cell death and suppressed intracellular ROS generation evoked by the addition of hydrogen peroxide (H2O2). Expression of the phase-II antioxidant enzyme NAD(P)H: quinone oxidoreductase 1 and haem oxygenase 1 were upregulated by TD at a concentration of 50-100 µM. Nuclear factor erythroid 2-related factor 2 (Nrf2), the regulator of these enzyme, was translocated from the cytosol to the nucleus by 100 µM TD. In summary, moderate concentrations of TD may increase the self-defence capacity of neuronal cells against oxidative stress.


Antioxidants/pharmacology , Indolequinones/pharmacology , Neurons/drug effects , Tryptamines/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Evaluation, Preclinical , Heme Oxygenase-1/metabolism , Humans , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Oxidative Stress , Protein Transport , Reactive Oxygen Species/metabolism
20.
Eur J Med Chem ; 133: 197-207, 2017 Jun 16.
Article En | MEDLINE | ID: mdl-28388522

Quinone methide (QM) formation induced by endogenously generated H2O2 is attractive for biological and biomedical applications. To overcome current limitations due to low biological activity of H2O2-activated QM precursors, we are introducing herein several new arylboronates with electron donating substituents at different positions of benzene ring and/or different neutral leaving groups. The reaction rate of the arylboronate esters with H2O2 and subsequent bisquinone methides formation and DNA cross-linking was accelerated with the application of Br as a leaving group instead of acetoxy groups. Additionally, a donating group placed meta to the nascent exo-methylene group of the quinone methide greatly improves H2O2-induced DNA interstrand cross-link formation as well as enhances the cellular activity. Multiple donating groups decrease the stability and DNA cross-linking capability, which lead to low cellular activity. A cell-based screen demonstrated that compounds 2a and 5a with a OMe or OH group dramatically inhibited the growth of various tissue-derived cancer cells while normal cells were less affected. Induction of H2AX phosphorylation by these compounds in CLL lymphocytes provide evidence for a correlation between cell death and DNA damage. The compounds presented herein showed potent anticancer activities and selectivity, which represent a novel scaffold for anticancer drug development.


Antineoplastic Agents/pharmacology , Benzene Derivatives/pharmacology , DNA/chemistry , Hydrogen Peroxide/metabolism , Indolequinones/pharmacology , Intercalating Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Base Sequence/drug effects , Benzene Derivatives/chemistry , Benzene Derivatives/metabolism , Boronic Acids/chemistry , Boronic Acids/metabolism , Boronic Acids/pharmacology , Cell Line, Tumor , Humans , Indolequinones/chemistry , Indolequinones/metabolism , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
...