Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 817
Filtrer
1.
Molecules ; 29(10)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38792153

RÉSUMÉ

Breast cancer is associated with high mortality and morbidity rates. As about 20-30% of patients exhibiting ER-positive phenotype are resistant to hormonal treatment with the standard drug tamoxifen, finding new therapies is a necessity. Postbiotics, metabolites, and macromolecules isolated from probiotic bacteria cultures have been proven to have sufficient bioactivity to exert prohealth and anticancer effects, making them viable adjunctive agents for the treatment of various neoplasms, including breast cancer. In the current study, postbiotics derived from L. plantarum and L. rhamnosus cultures were assessed on an in vitro breast cancer model as potential adjunctive agents to therapy utilizing tamoxifen and a candidate aziridine-hydrazide hydrazone derivative drug. Cell viability and cell death processes, including apoptosis, were analyzed for neoplastic MCF-7 cells treated with postbiotics and synthetic compounds. Cell cycle progression and proliferation were analyzed by PI-based flow cytometry and Ki-67 immunostaining. Postbiotics decreased viability and triggered apoptosis in MCF-7, modestly affecting the cell cycle and showing a lack of negative impact on normal cell viability. Moreover, they enhanced the cytotoxic effect of tamoxifen and the new candidate drug toward MCF-7, accelerating apoptosis and the inhibition of proliferation. This illustrates postbiotics' potential as natural adjunctive agents supporting anticancer therapy based on synthetic drugs.


Sujet(s)
Apoptose , Aziridines , Tumeurs du sein , Prolifération cellulaire , Tamoxifène , Humains , Tamoxifène/pharmacologie , Tamoxifène/composition chimique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Cellules MCF-7 , Femelle , Aziridines/pharmacologie , Aziridines/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Hydrazones/pharmacologie , Hydrazones/composition chimique , Probiotiques/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Cycle cellulaire/effets des médicaments et des substances chimiques
2.
Molecules ; 29(7)2024 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-38611710

RÉSUMÉ

A series of optically pure aziridine phosphines and their corresponding phosphine oxides were synthesized through established chemical methodologies. The compounds were systematically investigated for their biological properties. Notably, all synthesized compounds demonstrated moderate antibacterial activity only against the reference strain of Staphylococcus aureus. However, compounds 5 and 7 exhibited noteworthy cell viability inhibition of human cervical epithelioid carcinoma HeLa cells and endometrial adenocarcinoma Ishikawa cells. Further studies of these compounds revealed additional biological effects, including disruption of the cell membrane in high concentrations, cell cycle arrest in the S phase, and the induction of reactive oxygen species (ROS). Comparative analysis of the two classes of chiral organophosphorus derivatives of aziridines indicated that chiral phosphine oxides displayed significantly higher biological activity. Consequently, these findings suggest that chiral phosphine oxides may be potential candidates for the development of anticancer drugs. In light of the significant interest in preparations whose structure is based on a three-membered aziridine ring in terms of potential anticancer therapy, this research fits into the current research trend and should constitute a valuable addition to the current state of knowledge and the existing library of aziridine derivatives with anticancer properties.


Sujet(s)
Aziridines , Phosphines , Humains , Cellules HeLa , Aziridines/pharmacologie , Oxydes
3.
Cells ; 12(14)2023 07 21.
Article de Anglais | MEDLINE | ID: mdl-37508570

RÉSUMÉ

Tumor therapy escape due to undesired side effects induced by treatment, such as prosurvival autophagy or cellular senescence, is one of the key mechanisms of resistance that eventually leads to tumor dormancy and recurrence. Glioblastoma is the most frequent and practically incurable neoplasm of the central nervous system; thus, new treatment modalities have been investigated to find a solution more effective than the currently applied standards based on temozolomide. The present study examined the newly synthesized compounds of aziridine-hydrazide hydrazone derivatives to determine their antineoplastic potential against glioblastoma cells in vitro. Although the output of our investigation clearly demonstrates their proapoptotic activity, the cytotoxic effect appeared to be blocked by treatment-induced autophagy, the phenomenon also detected in the case of temozolomide action. The addition of an autophagy inhibitor, chloroquine, resulted in a significant increase in apoptosis triggered by the tested compounds, as well as temozolomide. The new aziridine-hydrazide hydrazone derivatives, which present cytotoxic potential against glioblastoma cells comparable to or even higher than that of temozolomide, show promising results and, thus, should be further investigated as antineoplastic agents. Moreover, our findings suggest that the combination of an apoptosis inducer with an autophagy inhibitor could optimize chemotherapeutic efficiency, and the addition of an autophagy inhibitor should be considered as an optional adjunctive therapy minimizing the risk of tumor escape from treatment.


Sujet(s)
Antinéoplasiques , Aziridines , Glioblastome , Humains , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Chloroquine/pharmacologie , Hydrazones/pharmacologie , Hydrazines/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Autophagie , Aziridines/pharmacologie , Aziridines/usage thérapeutique
4.
J Enzyme Inhib Med Chem ; 38(1): 2158187, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37070480

RÉSUMÉ

In this study, we report a series of newly synthesised sulphonamides of aziridine-2-carboxylic acid (Az-COOH) ester and amide analogues as potent protein disulphide isomerase (PDI, EC 5.3.4.1) inhibitors. The inhibitory activity on PDI was determined against recombinant human PDIA1 and PDIA3 proteins using an insulin reduction assay. These compounds in low micromolar to low nanomolar concentrations showed the effective in vitro inhibitory properties of PDIA1 with weaker effects on PDIA3. Complexes of 15N- and 15N,13C- uniformly labelled recombinant human PDIA1a with two PDIA1 inhibitors were produced and investigated by a protein nuclear magnetic resonance (NMR) spectroscopy. It was found that both C53 and C56 of the PDIA1 enzyme were involved in covalent binding. Finally, in a range of pharmacological studies, we demonstrated that investigated compounds displayed anti-cancer and anti-thrombotic activity. These findings demonstrate that sulphonamides of Az-COOH derivatives are promising candidates for the development of novel anti-cancer and anti-thrombotic agents.


Sujet(s)
Aziridines , Protein Disulfide-Isomerases , Sulfonamides , Humains , Aziridines/pharmacologie , Protein Disulfide-Isomerases/antagonistes et inhibiteurs , Protein Disulfide-Isomerases/composition chimique , Sulfonamides/pharmacologie
5.
Int J Mol Sci ; 23(20)2022 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-36293216

RÉSUMÉ

The ubiquitin-proteasome pathway (UPP) is the major proteolytic system in the cytosol and nucleus of all eukaryotic cells. The role of proteasome inhibitors (PIs) as critical agents for regulating cancer cell death has been established. Aziridine derivatives are well-known alkylating agents employed against cancer. However, to the best of our knowledge, aziridine derivatives showing inhibitory activity towards proteasome have never been described before. Herein we report a new class of selective and nonPIs bearing an aziridine ring as a core structure. In vitro cell-based assays (two leukemia cell lines) also displayed anti-proliferative activity for some compounds. In silico studies indicated non-covalent binding mode and drug-likeness for these derivatives. Taken together, these results are promising for developing more potent PIs.


Sujet(s)
Antinéoplasiques , Aziridines , Tumeurs , Humains , Inhibiteurs du protéasome/pharmacologie , Inhibiteurs du protéasome/usage thérapeutique , Proteasome endopeptidase complex/métabolisme , Antinéoplasiques/usage thérapeutique , Aziridines/pharmacologie , Aziridines/composition chimique , Tumeurs/métabolisme , Agents alcoylants , Ubiquitines
6.
Cancer Gene Ther ; 29(7): 1021-1032, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-34837065

RÉSUMÉ

Advances in the field of cancer immunotherapy have stimulated renewed interest in adenoviruses as oncolytic agents. Clinical experience has shown that oncolytic adenoviruses are safe and well tolerated but possess modest single-agent activity. One approach to improve the potency of oncolytic viruses is to utilise their tumour selectivity to deliver genes encoding prodrug-activating enzymes. These enzymes can convert prodrugs into cytotoxic species within the tumour; however, these cytotoxins can interfere with viral replication and limit utility. In this work, we evaluated the activity of a nitroreductase (NTR)-armed oncolytic adenovirus ONYX-411NTR in combination with the clinically tested bioreductive prodrug PR-104. Both NTR-expressing cells in vitro and xenografts containing a minor population of NTR-expressing cells were highly sensitive to PR-104. Pharmacologically relevant prodrug exposures did not interfere with ONYX-411NTR replication in vitro. In vivo, prodrug administration increased virus titre and improved virus distribution within tumour xenografts. Colonisation of tumours with high ONYX-411NTR titre resulted in NTR expression and prodrug activation. The combination of ONYX-411NTR with PR-104 was efficacious against HCT116 xenografts, whilst neither prodrug nor virus were active as single agents. This work highlights the potential for future clinical development of NTR-armed oncolytic viruses in combination with bioreductive prodrugs.


Sujet(s)
Aziridines , Tumeurs , Thérapie virale de cancers , Promédicaments , Adenoviridae , Aziridines/pharmacologie , Aziridines/usage thérapeutique , Humains , Tumeurs/thérapie , Moutardes à l'azote , Nitroréductases/génétique , Nitroréductases/métabolisme , Virus oncolytiques , Promédicaments/pharmacologie , Promédicaments/usage thérapeutique
7.
Molecules ; 26(14)2021 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-34299540

RÉSUMÉ

Several phosphorus-substituted N-acylated cyanoaziridines 2 and N-carbamoylated cyanoziridines 5 were prepared in good to high yields. N-Acylated cyanoaziridines 2 were used, after ring expansion, in an efficient synthesis of oxazoline derivative 3a and in a completely regio-controlled reaction in the presence of NaI. Conversely, N-carbamoyl cyanoaziridines 5 reacted with NaI to obtain a regioisomeric mixture of 2-aminocyanooxazolines 7. Mild acidic conditions can be used for the isomerization of N-thiocarbamoyl cyanoaziridine 6a into a 2-aminocyanothiazoline derivative 8a by using BF3·OEt2 as a Lewis acid. Likewise, a one pot reaction of NH-cyanoaziridines 1 with isocyanates obtained 2-iminocyanooxazolidines 9 regioselectively. This synthetic methodology involves the addition of isocyanates to starting cyanoaziridines to obtain N-carbamoyl cyanoaziridines 5, which after the ring opening, reacts with a second equivalent of isocyanate to give the final 2-imino cyanooxazolidines 9. In addition, the cytotoxic effect on the cell lines derived from human lung adenocarcinoma (A549) was also screened. 2-Iminooxazolidines 9 exhibited moderate activity against the A549 cell line in vitro. Furthermore, a selectivity towards cancer cells (A549) over non-malignant cells (MCR-5) was detected.


Sujet(s)
Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacologie , Aziridines/synthèse chimique , Aziridines/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Phosphore/pharmacologie , Cellules A549 , Adénocarcinome pulmonaire/traitement médicamenteux , Phénomènes biochimiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Humains , Tumeurs du poumon/traitement médicamenteux , Structure moléculaire
8.
Vet Microbiol ; 252: 108928, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-33248402

RÉSUMÉ

Binary ethylenimine (BEI) has been widely used as a virucide to inactivate viruses. For regulatory exclusion of a select agent, the United States Federal Select Agent Program (FSAP) requires an inactivation procedure that renders a select agent non-viable but allows the select agent to retain antigenic characteristics for future use must be validated, and the inactivated agent must be confirmed by a viability testing. In this curve-based validation study, we examined impacts of BEI concentration, treatment temperature, and time on our in-house inactivation procedures of Foot-and-Mouth Disease Virus (FMDV), Vesicular Stomatitis Virus (VSV), and Swine Vesicular Disease Virus (SVDV). The inactivation efficacy was confirmed by virus titration and 3 consecutive blind passages on the monolayers of susceptible cells. A linear correlation between the virus titer reduction and BEI concentration, treatment time, and temperature was established. The results confirmed our in-house BEI inactivation procedure of two doses of 1.5 mM BEI treatment at 37 °C, 1st dose for 24 h, then 2nd dose for 6 more hours for a total of 30 h BEI contact time, can ensure complete inactivation of FMDV, VSV, and SVDV.


Sujet(s)
Aziridines/pharmacologie , Entérovirus humain B/effets des médicaments et des substances chimiques , Virus de la fièvre aphteuse/effets des médicaments et des substances chimiques , Fièvre aphteuse/prévention et contrôle , Maladies des porcs/prévention et contrôle , Stomatite vésiculeuse/prévention et contrôle , Virus de la stomatite vésiculeuse de type Indiana/effets des médicaments et des substances chimiques , Animaux , Confinement de risques biologiques/médecine vétérinaire , Suidae , Maladies des porcs/virologie , Stomatite vésiculeuse/virologie , Inactivation virale/effets des médicaments et des substances chimiques
9.
Bioorg Chem ; 105: 104450, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33189994

RÉSUMÉ

In this study, various N-heterocyclic nitro prodrugs (NHN1-16) containing pyrimidine, triazine and piperazine rings were designed and synthesized. The final compounds were identified using FT-IR, 1H NMR, 13C NMR as well as elemental analyses. Enzymatic activities of compounds were conducted by using HPLC analysis to investigate the interaction of substrates with Ssap-NtrB nitroreductase enzyme. MTT assay was performed to evaluate the toxic effect of compounds against Hep3B and PC3 cancer cell lines and healthy HUVEC cell. It was observed that synthesized compounds NHN1-16 exhibited different cytotoxic profiles. Pyrimidine derivative NHN3 and triazine derivative NHN5 can be good drug candidates for prostate cancer with IC50 values of 54.75 µM and 48.9 µM, respectively. Compounds NHN6, NHN10, NHN12, NHN14 and NHN16 were selected as prodrug candidates because of non-toxic properties against three different cell models. The NHN prodrugs and Ssap-NtrB combinations were applied to SRB assay to reveal the prodrug capabilities of these selected compounds. SRB screening results showed that the metabolites of all selected non-toxic compounds showed remarkable cytotoxicity with IC50 values in the range of 1.71-4.72 nM on prostate cancer. Among the tested compounds, especially piperazine derivatives NHN12 and NHN14 showed significant toxic effect with IC50 values of 1.75 nM and 1.79 nM against PC3 cell compared with standart prodrug CB1954 (IC50: 1.71 nM). Novel compounds NHN12 and NHN14 can be considered as promising prodrug candidates for nitroreductase-prodrug based prostate cancer therapy.


Sujet(s)
Antinéoplasiques/composition chimique , Colletotrichum/composition chimique , Composés à cycles fusionnés/composition chimique , Composés hétérocycliques/composition chimique , Composés nitrés/composition chimique , Nitroréductases/antagonistes et inhibiteurs , Promédicaments/composition chimique , Tumeurs de la prostate/traitement médicamenteux , Antinéoplasiques/pharmacologie , Aziridines/pharmacologie , Aziridines/normes , Mélanges complexes/composition chimique , Mélanges complexes/pharmacologie , Tests de criblage d'agents antitumoraux , Activation enzymatique/effets des médicaments et des substances chimiques , Antienzymes/composition chimique , Antienzymes/pharmacologie , Fermentation , Composés à cycles fusionnés/pharmacologie , Composés hétérocycliques/pharmacologie , Humains , Mâle , Polycétides/composition chimique , Promédicaments/pharmacologie , Relation structure-activité
10.
ACS Chem Biol ; 15(2): 325-333, 2020 02 21.
Article de Anglais | MEDLINE | ID: mdl-32017532

RÉSUMÉ

Inhibition of the poly(ADP-ribose) polymerase (PARP) family of enzymes has become an attractive therapeutic strategy in oncology and beyond; however, chemical tools to profile PARP engagement in live cells are lacking. Herein, we report the design and application of PARPYnD, the first photoaffinity probe (AfBP) for PARP enzymes based on triple PARP1/2/6 inhibitor AZ9482, which induces multipolar spindle (MPS) formation in breast cancer cells. PARPYnD is a robust tool for profiling PARP1/2 and is used to profile clinical PARP inhibitor olaparib, identifying several novel off-target proteins. Surprisingly, while PARPYnD can enrich recombinant PARP6 spiked into cellular lysates and inhibits PARP6 in cell-free assays, it does not label PARP6 in intact cells. These data highlight an intriguing biomolecular disparity between recombinant and endogenous PARP6. PARPYnD provides a new approach to expand our knowledge of the targets of this class of compounds and the mechanisms of action of PARP inhibitors in cancer.


Sujet(s)
Aziridines/pharmacologie , Marqueurs de photoaffinité/pharmacologie , Phtalazines/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Aziridines/synthèse chimique , Lignée cellulaire tumorale , Humains , Marqueurs de photoaffinité/synthèse chimique , Phtalazines/synthèse chimique , Pipérazines/pharmacologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly(ADP-ribose) polymerases/métabolisme , Protéomique , Rayons ultraviolets
11.
Int J Mol Sci ; 21(4)2020 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-32093290

RÉSUMÉ

Prodrug activator gene therapy mediated by murine leukemia virus (MLV)-based retroviral replicating vectors (RRV) was previously shown to be highly effective in killing glioma cells both in culture and in vivo. To avoid receptor interference and enable dual vector co-infection with MLV-RRV, we have developed another RRV based on gibbon ape leukemia virus (GALV) that also shows robust replicative spread in a wide variety of tumor cells. We evaluated the potential of GALV-based RRV as a cancer therapeutic agent by incorporating yeast cytosine deaminase (CD) and E. coli nitroreductase (NTR) prodrug activator genes into the vector. The expression of CD and NTR genes from GALV-RRV achieved highly efficient delivery of these prodrug activator genes to RG-2 glioma cells, resulting in enhanced cytotoxicity after administering their respective prodrugs 5-fluorocytosine and CB1954 in vitro. In an immune-competent intracerebral RG-2 glioma model, GALV-mediated CD and NTR gene therapy both significantly suppressed tumor growth with CB1954 administration after a single injection of vector supernatant. However, NTR showed greater potency than CD, with control animals receiving GALV-NTR vector alone (i.e., without CB1954 prodrug) showing extensive tumor growth with a median survival time of 17.5 days, while animals receiving GALV-NTR and CB1954 showed significantly prolonged survival with a median survival time of 30 days. In conclusion, GALV-RRV enabled high-efficiency gene transfer and persistent expression of NTR, resulting in efficient cell killing, suppression of tumor growth, and prolonged survival upon CB1954 administration. This validates the use of therapeutic strategies employing this prodrug activator gene to arm GALV-RRV, and opens the door to the possibility of future combination gene therapy with CD-armed MLV-RRV, as the latter vector is currently being evaluated in clinical trials.


Sujet(s)
Aziridines/pharmacologie , Tumeurs du cerveau/thérapie , Flucytosine/pharmacologie , Thérapie génétique , Vecteurs génétiques , Gliome/thérapie , Tumeurs expérimentales/thérapie , Promédicaments/pharmacologie , Animaux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Cytosine deaminase/biosynthèse , Cytosine deaminase/génétique , Protéines Escherichia coli/biosynthèse , Protéines Escherichia coli/génétique , Gliome/génétique , Gliome/métabolisme , Gliome/anatomopathologie , Virus de la leucémie du gibbon , Tumeurs expérimentales/génétique , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Nitroréductases/biosynthèse , Nitroréductases/génétique , Rats de lignée F344 , Protéines de Saccharomyces cerevisiae/biosynthèse , Protéines de Saccharomyces cerevisiae/génétique
12.
Chem Commun (Camb) ; 56(14): 2194-2197, 2020 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-31971170

RÉSUMÉ

An Ugi, novel pseudo-Knoevenagel, ring expansion cascade reaction was discovered and utilized for the synthesis of aziridinyl succinimides in one-pot. Subsequently, densely functionalized aziridines and maleimides have been designed and synthesized through similar cascade reactions. The target compounds were prepared by means of a mild reaction and a simple operation procedure, which could be applicable to a broad scope of starting materials. This series of novel cascade reactions generates opportunities for the tailored synthesis of a wide range of biologically active scaffolds through tuneable Ugi inputs. Discovery of compound 8i with comparable potency to sorafenib in liver cancer cell lines could provide a new avenue for liver cancer drug discovery.


Sujet(s)
Antinéoplasiques/pharmacologie , Aziridines/pharmacologie , Maléimides/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Aziridines/synthèse chimique , Aziridines/composition chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Humains , Maléimides/synthèse chimique , Maléimides/composition chimique , Structure moléculaire
13.
Bioorg Chem ; 95: 103499, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31838287

RÉSUMÉ

Phosphazine and phosphazide derivatives are described herein as a new class of selective and potent acetylcholinesterase (AChE) inhibitors and ß-amyloid aggregation inhibitors. Phosphazines (5-7) were synthesized smoothly via a redox-condensation reaction of 1,2-bis(diphenylphosphino)ethane with different amines derivatives in the presence of dialkyl azodicarboxylate (Staudinger reaction) while phosphazides (8) via electrophilic attack of azido derivatives. Structures of the synthesized compounds were justified on the basis of compatible elementary and spectroscopic analyses. All the compounds were evaluated for their acetylcholinesterase inhibitory activity. The most three potent compounds (5b-c and 8b) showing AChE IC50 values (29.85-34.96 nM) comparable to that of donepezil (34.42 nM) were subjected to further investigation by testing their butyrylcholinesterase, MMP-2 and self-induced Aß aggregation inhibition activity. Especially, the coumarin phosphazide derivative (8b) presented the best AChE inhibition selectivity index (IC50 = 34.96 nM, AChE/BuChE; 3.81) together with good inhibition ability against MMP-2 (IC50 = 441.33 nM) and self-induced Aß1-42 aggregation (IC50 = 337.77 nM). In addition, the inhibition of metal-induced Aß aggregation by 8b was confirmed by thioflavine T fluorescence. The most potent effect of 8b was observed on the Zn2+-induced Aß42 aggregation. Kinetic study of compound 8b suggested it to be a competitive AChE inhibitor. Also, it specifically chelates metal and is predicted to be permeable to BBB. It also possesses low toxicity on SH-SY5Y neuroblastoma cells with a safety index of 15.37. In addition, it was demonstrated that compound 8b can improve the cognitive impairment of scopolamine-induced model in mice with % alternations and transfer latency time comparable to that of donepezil. Also, a docking study was carried out and it was in accordance with the in vitro results. These promising in vitro and in vivo findings highlight compound 8b as a possible drug candidate in searching for new multifunctional AD drugs.


Sujet(s)
Acetylcholinesterase/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Aziridines/pharmacologie , Anticholinestérasiques/pharmacologie , Phosphonates/pharmacologie , Zidovudine/analogues et dérivés , Maladie d'Alzheimer/métabolisme , Amnésie/induit chimiquement , Amnésie/traitement médicamenteux , Amnésie/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Aziridines/synthèse chimique , Aziridines/composition chimique , Anticholinestérasiques/synthèse chimique , Anticholinestérasiques/composition chimique , Relation dose-effet des médicaments , Humains , Cinétique , Ligands , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Souris , Modèles moléculaires , Structure moléculaire , Phosphonates/synthèse chimique , Phosphonates/composition chimique , Agrégats de protéines/effets des médicaments et des substances chimiques , Scopolamine , Relation structure-activité , Zidovudine/synthèse chimique , Zidovudine/composition chimique , Zidovudine/pharmacologie
14.
Chem Biodivers ; 16(11): e1900375, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-31512351

RÉSUMÉ

New aziridine 2-phosphonic acids were prepared by monohydrolysis of the aziridine 2-phosphonates that were obtained by the modified Gabriel-Cromwell reaction of vinyl phosphonate or α-tosylvinyl phosphonate with a primary amine or a chiral amine. The cellular cytotoxicity of these compounds was tested against the HCT-116 colorectal cancer cell lines and the CCD-18Co normal colon fibroblast lines using the MTT assay. Three of the synthesized phosphonic acid derivatives 2e (ethyl hydrogen {(2S)-1-[(1S)-1-(naphthalen-2-yl)ethyl]aziridin-2-yl}phosphonate), 2h (ethyl hydrogen (1-benzylaziridin-2-yl)phosphonate), and 2i (ethyl hydrogen (1-cyclohexylaziridin-2-yl)phosphonate) showed higher cytotoxicity than the reference cancer treatment agent etoposide. Cell death was through a robust induction of apoptosis even more effectively than etoposide, a well-known apoptosis inducing agent.


Sujet(s)
Antinéoplasiques/pharmacologie , Aziridines/pharmacologie , Acides phosphoreux/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Aziridines/synthèse chimique , Aziridines/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Tests de criblage d'agents antitumoraux , Humains , Structure moléculaire , Acides phosphoreux/synthèse chimique , Acides phosphoreux/composition chimique
15.
Int J Mol Sci ; 20(10)2019 May 15.
Article de Anglais | MEDLINE | ID: mdl-31096565

RÉSUMÉ

Many medicinal plant species are currently threatened in their natural habitats because of the growing demand for phytochemicals worldwide. A sustainable alternative for the production of bioactive plant compounds are plant biofactories based on cell cultures and organs. In addition, plant extracts from biofactories have significant advantages over those obtained from plants, since they are free of contamination by microorganisms, herbicides and pesticides, and they provide more stable levels of active ingredients. In this context, we report the establishment of Satureja khuzistanica cell cultures able to produce high amounts of rosmarinic acid (RA). The production of this phytopharmaceutical was increased when the cultures were elicited with coronatine and scaled up to a benchtop bioreactor. S. khuzistanica extracts enriched in RA were found to reduce the viability of cancer cell lines, increasing the sub-G0/G1 cell population and the activity of caspase-8 in MCF-7 cells, which suggest that S. khuzistanica extracts can induce apoptosis of MCF-7 cells through activation of the extrinsic pathway. In addition, our findings indicate that other compounds in S. khuzistanica extracts may act synergistically to potentiate the anticancer activity of RA.


Sujet(s)
Aziridines/pharmacologie , Cinnamates/métabolisme , Cinnamates/pharmacologie , Cyclohexènes/pharmacologie , Depsides/métabolisme , Depsides/pharmacologie , Espèce en voie de disparition , Extraits de plantes/pharmacologie , Satureja/métabolisme , Adénocarcinome/traitement médicamenteux , Bioréacteurs , Caspase 8/métabolisme , Caspases/métabolisme , Techniques de culture cellulaire , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules HepG2/effets des médicaments et des substances chimiques , Humains , Cellules MCF-7 , Composés phytochimiques/pharmacologie , Plantes médicinales/composition chimique , Satureja/croissance et développement ,
16.
Cancer Chemother Pharmacol ; 83(6): 1183-1189, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30868237

RÉSUMÉ

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.


Sujet(s)
Antinéoplasiques/administration et posologie , Aziridines/administration et posologie , Chromatographie en phase liquide à haute performance , Hématurie/complications , Indolequinones/administration et posologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/pharmacologie , Aziridines/pharmacocinétique , Aziridines/pharmacologie , Lignée cellulaire tumorale , Période , Humains , Techniques in vitro , Indolequinones/pharmacocinétique , Indolequinones/pharmacologie , Plan de recherche
17.
Arch Pharm Res ; 42(9): 815-823, 2019 Sep.
Article de Anglais | MEDLINE | ID: mdl-30879173

RÉSUMÉ

RH1 (2, 5-diaziridinyl-3-(hydroxymethy)-6-methyl-1, 4-benzoquinone) is a bioreductive anticancer drug. The mechanism underlying its therapeutic properties has not yet been elucidated. In this study, we aimed to determine whether RH1 exerts its anticancer effect via p53-mediated apoptosis and senescence in vitro and in vivo. RH1 displayed dose-dependent biphasic effects in vitro, i.e., it induced apoptosis at higher dose and senescence at lower dose accompanied by marked activation of p53. Thus, RH1 primarily induced cell death by apoptosis. The cytotoxicity of RH1 was inhibited in A549 cells treated with the p53-inhibitor pifithrin-α or transfected p53 siRNA and in human colon cancer HCT116 isogenic (p53-/-) cells. At sub-lethal doses of RH1, the cells survived and underwent senescence. The senescent cells showed flattened and enlarged morphology, and exhibited blue color in senescence-associated ß-galactosidase staining. These changes were found to be related to p53. RH1-induced senescence decreased in A549-E6 cells (suppressed p53 level) and HCT 116 p53-/- cells. The growth of A549 xenograft tumors in nude mice was significantly delayed by intraperitoneal injection of RH1, and senescent cells were observed in these xenograft tumors. These results suggest that the in vivo anticancer therapeutic effect of RH1 is mediated by senescence via p53 activation.


Sujet(s)
Antinéoplasiques/pharmacologie , Aziridines/pharmacologie , Benzoquinones/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Cellules A549 , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Humains , Relation structure-activité , Protéine p53 suppresseur de tumeur/génétique
18.
Eur J Med Chem ; 163: 736-746, 2019 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-30576904

RÉSUMÉ

This work reports an efficient diastereoselective synthetic methodology for the preparation of phosphorus substituted cyanoaziridines through the nucleophilic addition of TMSCN, as cyanide source, to the C-N double bond of 2H-azirine derivatives. The aziridine ring, in these novel cyanoaziridines, can be activated by simple N-tosylation or N-acylation. In addition, the cytotoxic effect on cell lines derived from human lung adenocarcinoma (A549) and human embryonic kidney (HEK293) was also screened. N-H and N-Substituted cyanoaziridines showed excellent activity against the A549 cell line in vitro. Moreover, selectivity towards cancer cell (A549) over (HEK293), and non-malignant cells (MCR-5) has been observed.


Sujet(s)
Aziridines/pharmacologie , Phosphonates/pharmacologie , Oxydes/pharmacologie , Cellules A549 , Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacologie , Aziridines/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cyanures , Cellules HEK293 , Humains , Phosphonates/synthèse chimique , Oxydes/synthèse chimique , Relation structure-activité
19.
Curr Eye Res ; 44(4): 413-422, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30444431

RÉSUMÉ

PURPOSE: To determine the contribution of the ON and OFF cone bipolar cell pathways to the electroretinograms (ERGs) elicited by ultraviolet (UV) and middle wavelength light in mice. MATERIALS AND METHODS: The experiments were performed on 8- to 10-week-old C57BL/6J mice. The ERGs elicited by single-flash and flickering UV light stimuli were compared to those elicited by green light stimuli under photopic conditions. Pharmacological agents were used to selectively block the ON and OFF pathways contributing to the ERGs. Saline was used as a control. The flicker ERGs elicited by UV light were compared to the ERGs elicited by green light after the injection of the pharmacological agents to determine the contribution of the cone ON and OFF pathways to the ERGs. RESULTS: The photopic single-flash and flicker ERGs were more sensitive to the UV light stimuli than to those elicited by green light stimuli. The flicker ERG responses elicited by both UV and green light stimuli at stimulus frequencies lower than 15-Hz decreased after L-2-amino-4-phosphobutyric acid was injected. The ERGs elicited by UV light at 30-Hz and by green light at frequencies lower than 15-Hz decreased significantly after the intravitreal injection of cis-2, 3-piperidine-dicarboxylic acid. An analysis of the ON and OFF components of the flicker ERGs showed that there might be pharmacological differences between the UV light-sensitive responses and the green light-sensitive responses. CONCLUSIONS: These results suggest that the UV light-sensitive cones connect to both the ON and OFF bipolar cells differently than that of the green light-sensitive cones.


Sujet(s)
Électrorétinographie , Cellules bipolaires rétiniennes/physiologie , Cellules photoréceptrices en cône de la rétine/physiologie , Amino-butyrates/pharmacologie , Animaux , Aziridines/pharmacologie , Adaptation à l'obscurité/physiologie , Lumière , Souris , Souris de lignée C57BL , Stimulation lumineuse , Cellules bipolaires rétiniennes/effets des médicaments et des substances chimiques , Rayons ultraviolets
20.
Biochem Pharmacol ; 158: 192-200, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30352235

RÉSUMÉ

Gene-directed enzyme-prodrug therapy (GDEPT) employs tumour-tropic vectors including viruses and bacteria to deliver a genetically-encoded prodrug-converting enzyme to the tumour environment, thereby sensitising the tumour to the prodrug. Nitroreductases, able to activate a range of promising nitroaromatic prodrugs to genotoxic metabolites, are of great interest for GDEPT. The bystander effect (cell-to-cell transfer of activated prodrug metabolites) has been quantified for some nitroaromatic prodrugs in mixed multilayer human cell cultures, however while these provide a good model for viral DEPT (VDEPT) they do not inform on the ability of these prodrug metabolites to exit bacterial vectors (relevant to bacterial-DEPT (BDEPT)). To investigate this we grew two Escherichia coli strains in co-culture; an activator strain expressing the nitroreductase E. coli NfsA and a recipient strain containing an SOS-GFP DNA damage responsive gene construct. In this system, induction of GFP by reduced prodrug metabolites can only occur following their transfer from the activator to the recipient cells. We used this to investigate five clinically relevant prodrugs: metronidazole, CB1954, nitro-CBI-DEI, and two dinitrobenzamide mustard prodrug analogues, PR-104A and SN27686. Consistent with the bystander efficiencies previously measured in human cell multilayers, reduced metronidazole exhibited little bacterial cell-to-cell transfer, whereas nitro-CBI-DEI was passed very efficiently from activator to recipient cells post-reduction. However, in contrast with observations in human cell multilayers, the nitrogen mustard prodrug metabolites were not effectively passed between the two bacterial strains, whereas reduced CB1954 was transferred efficiently. Using nitroreductase enzymes that exhibit different biases for the 2- versus 4-nitro substituents of CB1954, we further showed that the 2-nitro reduction products exhibit substantially higher levels of bacterial cell-to-cell transfer than the 4-nitro reduction products, consistent with their relative bystander efficiencies in human cell culture. Overall, our data suggest that prodrugs may differ in their suitability for VDEPT versus BDEPT applications and emphasise the importance of evaluating an enzyme-prodrug partnership in an appropriate context for the intended vector.


Sujet(s)
Escherichia coli/métabolisme , Thérapie génétique/méthodes , Vecteurs génétiques/métabolisme , Nitroréductases/métabolisme , Promédicaments/métabolisme , Aziridines/métabolisme , Aziridines/pharmacologie , Relation dose-effet des médicaments , Évaluation préclinique de médicament/méthodes , Escherichia coli/effets des médicaments et des substances chimiques , Vecteurs génétiques/pharmacologie , Humains , Promédicaments/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...