@article{MTMT:34840288, title = {A Novel 2-Methoxyestradiol Derivative: Disrupting Mitosis-Inhibiting Cell Motility and Inducing Apoptosis in HeLa Cells In Vitro}, url = {https://m2.mtmt.hu/api/publication/34840288}, author = {Njangiru, Isaac Kinyua and Bózsity-Faragó, Noémi and Resch, Vivien Erzsébet and Paragi, Gábor and Frank, Éva and Balogh, György Tibor and Zupkó, István and Minorics, Renáta}, doi = {10.3390/pharmaceutics16050622}, journal-iso = {PHARMACEUTICS}, journal = {PHARMACEUTICS}, volume = {16}, unique-id = {34840288}, issn = {1999-4923}, abstract = {The clinical application of 2-methoxyestradiol (2ME) in cancer therapy has been limited by its low solubility and rapid metabolism. Derivatives of 2ME have been synthesised to enhance bioavailability and decrease hepatic metabolism. Compound 4a, an analog of 2ME, has demonstrated exceptional pharmacological activity, in addition to promising pharmacokinetic profile. Our study, therefore, aimed at exploring the anticancer effects of 4a on the cervical cancer cell line, HeLa. Compound 4a exhibited a significant and dose-dependent antimetastatic and antiinvasive impact on HeLa cells, as determined by wound-healing and Boyden chamber assays, respectively. Hoechst/Propidium iodide (HOPI) double staining showcased a substantial induction of apoptosis via 4a, with minimal necrotic effect. Flow cytometry revealed a significant G2/M phase arrest, accompanied by a noteworthy rise in the sub-G1 cell population, indicating apoptosis, 18 h post-treatment. Moreover, a cell-independent tubulin polymerisation assay illustrated compound 4a’s ability to stabilise microtubules by promoting tubulin polymerisation. Molecular modelling experiments depicted that 4a interacts with the colchicine-binding site, nestled between the α and β tubulin dimers. Furthermore, 4a displayed an affinity for binding to and activating ER-α, as demonstrated by the luciferase reporter assay. These findings underscore the potential of 4a in inhibiting HPV18+ cervical cancer proliferation and cellular motility.}, year = {2024}, eissn = {1999-4923}, orcid-numbers = {Paragi, Gábor/0000-0001-5408-1748; Frank, Éva/0000-0002-1332-0551; Balogh, György Tibor/0000-0003-3347-1880; Zupkó, István/0000-0003-3243-5300; Minorics, Renáta/0000-0001-9685-813X} } @article{MTMT:34789041, title = {Synthesis of Estrone Heterodimers and Evaluation of Their In Vitro Antiproliferative Activity}, url = {https://m2.mtmt.hu/api/publication/34789041}, author = {Bózsity-Faragó, Noémi and Nagy, Viktória and Szabó, Johanna and Pálházi, Balázs and Kele, Zoltán and Resch, Vivien Erzsébet and Paragi, Gábor and Zupkó, István and Minorics, Renáta and Mernyák, Erzsébet}, doi = {10.3390/ijms25084274}, journal-iso = {INT J MOL SCI}, journal = {INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES}, volume = {25}, unique-id = {34789041}, issn = {1661-6596}, abstract = {Directed structural modifications of natural products offer excellent opportunities to develop selectively acting drug candidates. Natural product hybrids represent a particular compound group. The components of hybrids constructed from different molecular entities may result in synergic action with diminished side effects. Steroidal homo- or heterodimers deserve special attention owing to their potentially high anticancer effect. Inspired by our recently described antiproliferative core-modified estrone derivatives, here, we combined them into heterodimers via Cu(I)-catalyzed azide–alkyne cycloaddition reactions. The two trans-16-azido-3-(O-benzyl)-17-hydroxy-13α-estrone derivatives were reacted with 3-O-propargyl-D-secoestrone alcohol or oxime. The antiproliferative activities of the four newly synthesized dimers were evaluated against a panel of human adherent gynecological cancer cell lines (cervical: Hela, SiHa, C33A; breast: MCF-7, T47D, MDA-MB-231, MDA-MB-361; ovarian: A2780). One heterodimer (12) exerted substantial antiproliferative activity against all investigated cell lines in the submicromolar or low micromolar range. A pronounced proapoptotic effect was observed by fluorescent double staining and flow cytometry on three cervical cell lines. Additionally, cell cycle blockade in the G2/M phase was detected, which might be a consequence of the effect of the dimer on tubulin polymerization. Computational calculations on the taxoid binding site of tubulin revealed potential binding of both steroidal building blocks, mainly with hydrophobic interactions and water bridges.}, year = {2024}, eissn = {1422-0067}, orcid-numbers = {Resch, Vivien Erzsébet/0000-0003-0044-5731; Paragi, Gábor/0000-0001-5408-1748; Zupkó, István/0000-0003-3243-5300; Minorics, Renáta/0000-0001-9685-813X; Mernyák, Erzsébet/0000-0003-4494-1817} } @article{MTMT:34779108, title = {Diterpenes Isolated from Three Different Plectranthus Sensu Lato Species and Their Antiproliferative Activities against Gynecological and Glioblastoma Cancer Cells}, url = {https://m2.mtmt.hu/api/publication/34779108}, author = {Gáborová, Mária and Vágvölgyi, Máté and Tayeb, Bizhar Ahmed and Minorics, Renáta and Zupkó, István and Jurček, Ondřej and Béni, Szabolcs and Kubínová, Renata and Balogh, György Tibor and Hunyadi, Attila}, doi = {10.1021/acsomega.4c00800}, journal-iso = {ACS OMEGA}, journal = {ACS OMEGA}, volume = {9}, unique-id = {34779108}, issn = {2470-1343}, year = {2024}, eissn = {2470-1343}, pages = {18495-18504}, orcid-numbers = {Vágvölgyi, Máté/0000-0002-2233-9422; Tayeb, Bizhar Ahmed/0000-0001-5197-564X; Minorics, Renáta/0000-0001-9685-813X; Zupkó, István/0000-0003-3243-5300; Béni, Szabolcs/0000-0001-7056-6825; Balogh, György Tibor/0000-0003-3347-1880; Hunyadi, Attila/0000-0003-0074-3472} } @article{MTMT:34666945, title = {Synthesis and Study of the Structure–Activity Relationship of Antiproliferative N-Substituted Isosteviol-Based 1,3-Aminoalcohols}, url = {https://m2.mtmt.hu/api/publication/34666945}, author = {Ozsvár, Dániel and Bózsity-Faragó, Noémi and Zupkó, István and Szakonyi, Zsolt}, doi = {10.3390/ph17020262}, journal-iso = {PHARMACEUTICALS-BASE}, journal = {PHARMACEUTICALS}, volume = {17}, unique-id = {34666945}, abstract = {Starting from isosteviol, a series of diterpenoid 1,3-aminoalcohol derivatives were prepared via stereoselective transformations. The acid-catalysed hydrolysis and rearrangement of natural stevioside produced isosteviol, which was transformed into the key intermediate methyl ester. In the next step, an 1,3-aminoalcohol library was prepared by the reductive amination of the intermediate 3-hydroxyaldehyde obtained from isosteviol in a two-step synthesis. To study the effect of the carboxylate ester function at position 4, the free carboxylic acid, benzyl ester and acryloyl ester analogues were prepared as elongated derivatives in comparison with our earlier results in this field. The antiproliferative activity of compounds against human tumour cell lines (A2780, HeLa, MCF-7 and MDA-MB-231) was investigated. In our preliminary study, the 1,3-aminoalcohol function with N-benzyl or (1H-imidazol-1-yl)-propyl substitution and benzyl ester moiety seemed essential for the reliable antiproliferative activity. The results obtained could be a good starting point to further functionalisation towards more efficient antiproliferative diterpenes.}, year = {2024}, eissn = {1424-8247}, orcid-numbers = {Zupkó, István/0000-0003-3243-5300; Szakonyi, Zsolt/0000-0003-2432-8409} } @article{MTMT:34577084, title = {Stereoselective synthesis and antiproliferative activity of allo-gibberic acid-based 1,3-aminoalcohols regioisomers}, url = {https://m2.mtmt.hu/api/publication/34577084}, author = {Khdar, Zein Alabdeen and Le Minh, Tam and Schelz, Zsuzsanna and Zupkó, István and Szakonyi, Zsolt}, doi = {10.1039/D3MD00665D}, journal-iso = {RSC MED CHEM}, journal = {RSC MEDICINAL CHEMISTRY}, volume = {15}, unique-id = {34577084}, abstract = {A new library of allo -gibberic acid-based aminoalcohol regioisomers was synthesised stereoselectively starting from commercially available gibberellic acid, which yields allo -gibberic acid under mild acidic conditions. The successful formation of hydroxymethyl...}, year = {2024}, eissn = {2632-8682}, pages = {874-887}, orcid-numbers = {Le Minh, Tam/0000-0002-8296-887X; Schelz, Zsuzsanna/0000-0002-8519-4830; Zupkó, István/0000-0003-3243-5300; Szakonyi, Zsolt/0000-0003-2432-8409} } @article{MTMT:34541373, title = {Thymoquinone-protoflavone hybrid molecules as potential antitumor agents}, url = {https://m2.mtmt.hu/api/publication/34541373}, author = {AHMED, Sara Hassan Hassan and Tayeb, Bizhar Ahmed and Gonda, Tímea and Girst, Gábor and Szőri, Kornél and Berkecz, Róbert and Zupkó, István and Minorics, Renáta and Hunyadi, Attila}, doi = {10.1371/journal.pone.0291567}, journal-iso = {PLOS ONE}, journal = {PLOS ONE}, volume = {19}, unique-id = {34541373}, issn = {1932-6203}, abstract = {We describe herein the synthesis of eight new ester-coupled hybrid compounds from thymoquinone and protoflavone building blocks, and their bioactivity testing against multiple cancer cell lines. Among the hybrids, compound 14 showed promising activities in all cell lines studied. The highest activities were recorded against breast cancer cell lines with higher selectivity to MDA-MB-231 as compared to MCF-7. Even though the hybrids were found to be completely hydrolysed in 24 h under cell culture conditions, compound 14 demonstrated a ca. three times stronger activity against U-87 glioblastoma cells than a 1:1 mixture of its fragments. Further, compound 14 showed good tumour selectivity: it acted 4.4-times stronger on U-87 cells than on MRC-5 fibroblasts. This selectivity was much lower, only ca. 1.3-times, when the cells were co-treated with a 1:1 mixture of its non-coupled fragments. Protoflavone-thymoquinone hybrids may therefore serve as potential new antitumor leads particularly against glioblastoma.}, year = {2024}, eissn = {1932-6203}, orcid-numbers = {Tayeb, Bizhar Ahmed/0000-0001-5197-564X; Szőri, Kornél/0000-0003-3337-8635; Berkecz, Róbert/0000-0002-9076-2177; Zupkó, István/0000-0003-3243-5300; Minorics, Renáta/0000-0001-9685-813X; Hunyadi, Attila/0000-0003-0074-3472} } @article{MTMT:34499406, title = {CuAAC-Based Synthesis, Copper-Catalyzed Aldehyde-Forming Hydrolytic Fission and Antiproliferative Evaluation of Novel Ferrocenoylamino-Substituted Triazole-Tethered Quinine–Chalcone Hybrids}, url = {https://m2.mtmt.hu/api/publication/34499406}, author = {Dembo, António and Ferenczi, Etelka and Jernei, Tamás and Bor, Andrea and Schelz, Zsuzsanna and Zupkó, István and Varga, Szilárd and Csámpai, Antal}, doi = {10.3390/molecules29020375}, journal-iso = {MOLECULES}, journal = {MOLECULES}, volume = {29}, unique-id = {34499406}, issn = {1420-3049}, abstract = {A series of novel triazole-tethered ferrocenoylamino-substituted cinchona–chalcone hybrids along with two representative benzoylamino-substituted reference compounds were prepared by three methods of CuAAC chemistry. In line with the limited success or complete failure of attempted conversions with low catalyst loadings, by means of DFT modeling studies, we demonstrated that a substantial part of the Cu(I) ions can be chelated and thus trapped in the aroylamino-substituted cinchona fragment and all of the accessible coordinating sites of the chalcone residues. Accordingly, increased amounts of catalysts were used to achieve acceptable yields; however, the cycloadditions with para-azidochalcones were accompanied by partial or complete aldehyde-forming hydrolytic fission of the enone C=C bond in a substituent-, solvent- and copper load-dependent manner. The experienced hydrolytic stability of the hybrids obtained by cycloadditions with ortho-azidochalcones was interpreted in terms of relative energetics, DFT reactivity indices and MO analysis of simplified models of two isomer copper–enone complexes. The novel hybrids were evaluated on HeLa, MDA-MB-231 and A2780 cell lines and showed substantial activity at low-to-submicromolar concentrations. An organometallic model carrying 3,4,5-trimethoxyphenyl residue in the enone part with a para-disubstituted benzene ring in the central skeletal region was identified as the most potent antiproliferative lead, characterized by submicromolar IC50 values measured on the three investigated cells. The biological assays also disclosed that this ferrocenoylamino-containing lead compound displays a ca. two- to five-fold more substantial antiproliferative effect than its benzoylamino-substituted counterpart.}, year = {2024}, eissn = {1420-3049}, orcid-numbers = {Schelz, Zsuzsanna/0000-0002-8519-4830; Zupkó, István/0000-0003-3243-5300; Varga, Szilárd/0000-0001-9611-5168; Csámpai, Antal/0000-0003-2107-7309} } @article{MTMT:34475300, title = {New 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinazoline and 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinoline Derivatives: Synthesis and Biological Evaluation as Novel Anticancer Agents by Targeting G-Quadruplex}, url = {https://m2.mtmt.hu/api/publication/34475300}, author = {Guillon, Jean and Le Borgne, Marc and Milano, Vittoria and Guédin-Beaurepaire, Aurore and Moreau, Stéphane and Pinaud, Noël and Ronga, Luisa and Savrimoutou, Solène and Albenque-Rubio, Sandra and Marchivie, Mathieu and Kalout, Haouraa and Walker, Charley and Chevallier, Louise and Buré, Corinne and Largy, Eric and Gabelica, Valérie and Mergny, Jean-Louis and Baylot, Virginie and Ferrer, Jacky and Idrissi, Yamina and Chevret, Edith and Cappellen, David and Desplat, Vanessa and Schelz, Zsuzsanna and Zupkó, István}, doi = {10.3390/ph17010030}, journal-iso = {PHARMACEUTICALS-BASE}, journal = {PHARMACEUTICALS}, volume = {17}, unique-id = {34475300}, abstract = {The syntheses of novel 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinazolines 12 and 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinolines 13 are reported here in six steps starting from various halogeno-quinazoline-2,4-(1H,3H)-diones or substituted anilines. The antiproliferative activities of the products were determined in vitro against a panel of breast (MCF-7 and MDA-MB-231), human adherent cervical (HeLa and SiHa), and ovarian (A2780) cell lines. Disubstituted 6- and 7-phenyl-bis(3-dimethylaminopropyl)aminomethylphenyl-quinazolines 12b, 12f, and 12i displayed the most interesting antiproliferative activities against six human cancer cell lines. In the series of quinoline derivatives, 6-phenyl-bis(3-dimethylaminopropyl)aminomethylphenylquinoline 13a proved to be the most active. G-quadruplexes (G4) stacked non-canonical nucleic acid structures found in specific G-rich DNA, or RNA sequences in the human genome are considered as potential targets for the development of anticancer agents. Then, as small aza-organic heterocyclic derivatives are well known to target and stabilize G4 structures, their ability to bind G4 structures have been determined through FRET melting, circular dichroism, and native mass spectrometry assays. Finally, telomerase inhibition ability has been also assessed using the MCF-7 cell line.}, year = {2024}, eissn = {1424-8247}, orcid-numbers = {Guillon, Jean/0000-0001-8577-3894; Le Borgne, Marc/0000-0003-1398-075X; Pinaud, Noël/0000-0001-7646-5312; Mergny, Jean-Louis/0000-0003-3043-8401; Chevret, Edith/0000-0002-9724-8437; Schelz, Zsuzsanna/0000-0002-8519-4830; Zupkó, István/0000-0003-3243-5300} } @{MTMT:34556245, title = {Gyomorfekély ellenes hatással rendelkező, hagyma leveléből kivont poliszacharidok, eljárás előállításukra és alkalmazásuk}, url = {https://m2.mtmt.hu/api/publication/34556245}, author = {Csupor, Dezső and Zupkó, István and Hohmann, Judit and Kiss, Tivadar and Horváth, Attila}, unique-id = {34556245}, year = {2023}, orcid-numbers = {Csupor, Dezső/0000-0002-4088-3333; Zupkó, István/0000-0003-3243-5300; Hohmann, Judit/0000-0002-2887-6392; Kiss, Tivadar/0000-0003-3538-377X} } @article{MTMT:34443794, title = {Surface Modification of Titanate Nanotubes with a Carboxylic Arm for Further Functionalization Intended to Pharmaceutical Applications}, url = {https://m2.mtmt.hu/api/publication/34443794}, author = {Saker, Ranim and Jójártné Laczkovich, Orsolya and Regdon, Géza (ifj.) and Takács, Tamás and Szenti, Imre and Bózsity-Faragó, Noémi and Zupkó, István and Sovány, Tamás}, doi = {10.3390/pharmaceutics15122780}, journal-iso = {PHARMACEUTICS}, journal = {PHARMACEUTICS}, volume = {15}, unique-id = {34443794}, issn = {1999-4923}, abstract = {Nanotechnology is playing a significant role in modern life with tremendous potential and promising results in almost every domain, especially the pharmaceutical one. The impressive performance of nanomaterials is shaping the future of science and revolutionizing the traditional concepts of industry and research. Titanate nanotubes (TNTs) are one of these novel entities that became an appropriate choice to apply in several platforms due to their remarkable properties such as preparation simplicity, high stability, good biocompatibility, affordability and low toxicity. Surface modification of these nanotubes is also promoting their superior characters and contributing more to the enhancement of their performance. In this research work, an attempt was made to functionalize the surface of titanate nanotubes with carboxylic groups to increase their surface reactivity and widen the possibility of bonding different molecules that could not be bonded directly. Three carboxylic acids were investigated (trichloroacetic acid, citric acid and acrylic acid), and the prepared composites were examined using FT-IR and Raman spectroscopy, scanning electron microscopy (SEM), transmission electron microscopy (TEM) and dynamic light scattering (DLS). The toxicity of these functionalized TNTs was also investigated using adherent cancer cell lines and fibroblasts to determine their safety profile and to draw the basic lines for their intended future application. Based on the experimental results, acrylic acid could be the suitable choice for permanent surface modification with multiple carboxylic groups due to its possibility to be polymerized, thus presenting the opportunity to link additional molecules of interest such as polyethylene glycol (PEG) and/or other molecules at the same time.}, year = {2023}, eissn = {1999-4923}, orcid-numbers = {Regdon, Géza (ifj.)/0000-0002-6921-3069; Zupkó, István/0000-0003-3243-5300; Sovány, Tamás/0000-0003-3392-7788} }