1
|
Angajala G, Aruna V, Pavan P, Reddy PG. Ultrasound promoted montmorillonite K-10 catalyzed synthesis, characterization, molecular modelling, SAR and hypoglycemic studies of new rhodanine bejeweled acridine analogues. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
2
|
Kozurkova M. Acridine derivatives as inhibitors/poisons of topoisomerase II. J Appl Toxicol 2021; 42:544-552. [PMID: 34514603 DOI: 10.1002/jat.4238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/18/2022]
Abstract
The potential of acridines (amsacrine) as a topoisomerase II inhibitor or poison was first discovered in 1984, and since then, a considerable number of acridine derivatives have been tested as topoisomerase inhibitors/poisons, containing different substituents on the acridine chromophore. This review will discuss a series of studies published over the course of the last decade, which have investigated various novel acridine derivatives against topoisomerase II activity.
Collapse
Affiliation(s)
- Maria Kozurkova
- Department of Biochemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University, Kosice, Slovak Republic.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
3
|
Tolan HEM, Radwan MAA, Soliman HA, Awad HM, El-Sayed WA. Synthesis and Anti-Proliferative Activity of New Acridinyl and Benzothiazolyl-Based Triazole Glycosides via Click Cycloaddition and Their Tetrazolyl Analogs. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162020060345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
4
|
Synthesis and Biological Evaluation of Some 1,8-Naphthalimide-Acridinyl Hybrids. J CHEM-NY 2020. [DOI: 10.1155/2020/7989852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In the present study, the synthesis of three 1,8-naphthalimide-acridinyl hybrids (2a, 2b, and 5b) using N-amido-1,8-naphthalimides (1 and 4) and acridinyl isothiocyanates is reported. The newly synthesized hybrids were evaluated for their anticancer activity in six human cancer cell lines (HL-60, MT-4, HepG2, HeLa, SK-OV-3, and MCF-7). Their inhibition activity against DNA-topoisomerase I (Topo I) and Electrophorus electricus acetylcholinesterase (AChE) was also studied. The results indicate that 2b displayed good cytotoxicity for MT-4, HepG2, HeLa, and SK-OV-3 with the IC50 values of 14.66 ± 0.31, 27.32 ± 2.67, 17.51 ± 0.34, and 32.26 ± 1.74 μM, respectively. All compounds, especially 2b, exhibited obvious bands corresponding to DNA fragments at 0.5 mM concentration, further confirming the pharmacological mechanism related to the Topo I inhibitory activities. In addition, compound 2a exhibited higher inhibition activity against AChE than 2b and 5b, with IC50 values of 0.32 ± 0.04 mM, and the acridinyl ring may contribute to the activity of 2a.
Collapse
|
5
|
Kłosiński K, Girek M, Czarnecka K, Pasieka Z, Skibiński R, Szymański P. Biological assessment of new tetrahydroacridine derivatives with fluorobenzoic moiety in vitro on A549 and HT-29 cell lines and in vivo on animal model. Hum Cell 2020; 33:859-867. [PMID: 32449113 PMCID: PMC7324419 DOI: 10.1007/s13577-020-00376-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022]
Abstract
A new series of tetrahydroacridine derivatives with the fluorobenzoyl moiety was synthesized and evaluated for cytotoxic activity against lung cancer cell lines A549 and colorectal cancer HT29. The cytotoxic activity of the compounds was compared on the somatic cell line-EAhy926. Compounds showed high cytotoxic activity on A549 cells (IC50 183.26-68.07 μM) and HT29 cells (IC50 68.41-19.70 μM), higher than controls-etoposide (IC50 451.47 μM) toward A549 and 5-fluorouracil (IC50 1626.85 μM) against HT29. Derivative 4 was the most cytotoxic to A549, whereas for the cell lines HT29 compound 6. Selected compounds showed similar cytotoxicity to the EAhy926 cell line (IC50 about 50 μM). In the hyaluronidase inhibition assay, all compounds exhibited anti-inflammatory activity, including 4 exhibiting the best inhibitory activity-IC50 of 52.27 μM when the IC50 heparin was 56.41 μM. Mathematical modeling was performed to determine LD50 after intraperitoneal, oral, intravenous and subcutaneous administration and to predict potential mutagenicity and carcinogenicity of the compounds analyzed. Obtained results showed that tested derivatives are slightly toxic compounds, and LD50 values (mg/kg) ranged from 680 to 1200 (oral rat model), the analyzed compounds have low mutagenic potential, and differences between derivatives are insignificant and very low probability of carcinogenicity. To confirm mathematical calculations, an in vivo test was carried out on a laboratory mouse model for two selected compounds. It allowed to qualify compounds: 6 to category 4 of the GHS scale, and 4 to category 3 of the GHS scale.
Collapse
Affiliation(s)
- Karol Kłosiński
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136, Lodz, Poland
| | - Małgorzata Girek
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151, Lodz, Poland
| | - Zbigniew Pasieka
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136, Lodz, Poland
| | - Robert Skibiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 4, 20-090, Lublin, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151, Lodz, Poland.
| |
Collapse
|
6
|
Dallavalle S, Dobričić V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, Tsakovska I, Zidar N, Fruttero R. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat 2020; 50:100682. [PMID: 32087558 DOI: 10.1016/j.drup.2020.100682] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is the dominant cause of the failure of cancer chemotherapy. The design of antitumor drugs that are able to evade MDR is rapidly evolving, showing that this area of biomedical research attracts great interest in the scientific community. The current review explores promising recent approaches that have been developed with the aim of circumventing or overcoming MDR. Encouraging results have been obtained in the investigation of the MDR-modulating properties of various classes of natural compounds and their analogues. Inhibition of P-gp or downregulation of its expression have proven to be the main mechanisms by which MDR can be surmounted. The use of hybrid molecules that are able to simultaneously interact with two or more cancer cell targets is currently being explored as a means to circumvent drug resistance. This strategy is based on the design of hybrid compounds that are obtained either by merging the structural features of separate drugs, or by conjugating two drugs or pharmacophores via cleavable/non-cleavable linkers. The approach is highly promising due to the pharmacokinetic and pharmacodynamic advantages that can be achieved over the independent administration of the two individual components. However, it should be stressed that the task of obtaining successful multivalent drugs is a very challenging one. The conjugation of anticancer agents with nitric oxide (NO) donors has recently been developed, creating a particular class of hybrid that can combat tumor drug resistance. Appropriate NO donors have been shown to reverse drug resistance via nitration of ABC transporters and by interfering with a number of metabolic enzymes and signaling pathways. In fact, hybrid compounds that are produced by covalently attaching NO-donors and antitumor drugs have been shown to elicit a synergistic cytotoxic effect in a variety of drug resistant cancer cell lines. Another strategy to circumvent MDR is based on nanocarrier-mediated transport and the controlled release of chemotherapeutic drugs and P-gp inhibitors. Their pharmacokinetics are governed by the nanoparticle or polymer carrier and make use of the enhanced permeation and retention (EPR) effect, which can increase selective delivery to cancer cells. These systems are usually internalized by cancer cells via endocytosis and accumulate in endosomes and lysosomes, thus preventing rapid efflux. Other modalities to combat MDR are described in this review, including the pharmaco-modulation of acridine, which is a well-known scaffold in the development of bioactive compounds, the use of natural compounds as means to reverse MDR, and the conjugation of anticancer drugs with carriers that target specific tumor-cell components. Finally, the outstanding potential of in silico structure-based methods as a means to evaluate the ability of antitumor drugs to interact with drug transporters is also highlighted in this review. Structure-based design methods, which utilize 3D structural data of proteins and their complexes with ligands, are the most effective of the in silico methods available, as they provide a prediction regarding the interaction between transport proteins and their substrates and inhibitors. The recently resolved X-ray structure of human P-gp can help predict the interaction sites of designed compounds, providing insight into their binding mode and directing possible rational modifications to prevent them from becoming P-gp drug substrates. In summary, although major efforts were invested in the search for new tools to combat drug resistant tumors, they all require further implementation and methodological development. Further investigation and progress in the abovementioned strategies will provide significant advances in the rational combat against cancer MDR.
Collapse
Affiliation(s)
- Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Loretta Lazzarato
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Elena Gazzano
- Department of Oncology, Università degli Studi di Torino, Via Santena 5/bis, 10126 Turin, Italy
| | - Miguel Machuqueiro
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, C8 Building, Campo Grande, 1749-016, Lisbon, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| | - Ilza Pajeva
- QSAR and Molecular Modelling Department, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 105, 1113 Sofia, Bulgaria
| | - Ivanka Tsakovska
- QSAR and Molecular Modelling Department, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 105, 1113 Sofia, Bulgaria
| | - Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Roberta Fruttero
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Turin, Italy.
| |
Collapse
|
7
|
Skok Ž, Zidar N, Kikelj D, Ilaš J. Dual Inhibitors of Human DNA Topoisomerase II and Other Cancer-Related Targets. J Med Chem 2019; 63:884-904. [DOI: 10.1021/acs.jmedchem.9b00726] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Žiga Skok
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Nace Zidar
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Danijel Kikelj
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Janez Ilaš
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
8
|
da Silva Filho FA, de Freitas Souza T, Ribeiro AG, Alves JEF, de Oliveira JF, de Lima Souza TRC, de Moura RO, do Carmo Alves de Lima M, de Carvalho Junior LB, de Almeida SMV. Topoisomerase inhibition and albumin interaction studies of acridine-thiosemicarbazone derivatives. Int J Biol Macromol 2019; 138:582-589. [PMID: 31323270 DOI: 10.1016/j.ijbiomac.2019.07.097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/24/2019] [Accepted: 07/15/2019] [Indexed: 10/26/2022]
Abstract
In the present study, acridine-thiosemicarbazones (ATD) derivatives were tested for their interaction properties with BSA through UV-Vis absorption and fluorescence spectroscopic studies. Both hyperchromic and hypochromic effects, as well as red or blue shifts were demonstrated after the derivatives were added to the BSA. Values for the binding constant (Kb) ranged from 1.62 × 104 to 8.71 × 105 M-1 and quenching constant (KSV) from 3.46 × 102 to 7.83 × 103 M-1 indicating a good affinity to BSA protein. Complementary, two compounds were selected to assess their inhibition activity against topoisomerase IIα enzyme, of which derivative 3a presented the best result. Moreover, to evaluate protein-ligand interactions, as well as the antitopoisomerase potential of these compounds, tests of molecular modeling were performed between all compounds using the albumin and Topoisomerase IIα/DNA complex. Finally, in silico studies showed that all derivatives used in this research displayed good oral bioavailability potential.
Collapse
Affiliation(s)
- Francivaldo Araújo da Silva Filho
- Universidade de Pernambuco (UPE), campus Garanhuns, Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Garanhuns, PE, Brazil
| | - Thais de Freitas Souza
- Universidade de Pernambuco (UPE), campus Garanhuns, Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Garanhuns, PE, Brazil
| | - Amélia Galdino Ribeiro
- Laboratório de Química e Inovação Terapêutica (LQIT), Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Jamerson Ferreira de Oliveira
- Laboratório de Química e Inovação Terapêutica (LQIT), Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Ricardo Olímpio de Moura
- Departamento de Ciências Farmacêuticas, Centro de Ciências Biológicas e da Saúde, Universidade Estadual da Paraíba - Bodocongo, Campina Grande, PB, Brazil
| | - Maria do Carmo Alves de Lima
- Laboratório de Química e Inovação Terapêutica (LQIT), Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Sinara Mônica Vitalino de Almeida
- Universidade de Pernambuco (UPE), campus Garanhuns, Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Garanhuns, PE, Brazil; Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
9
|
Girek M, Kłosiński K, Grobelski B, Pizzimenti S, Cucci MA, Daga M, Barrera G, Pasieka Z, Czarnecka K, Szymański P. Novel tetrahydroacridine derivatives with iodobenzoic moieties induce G0/G1 cell cycle arrest and apoptosis in A549 non-small lung cancer and HT-29 colorectal cancer cells. Mol Cell Biochem 2019; 460:123-150. [PMID: 31313023 PMCID: PMC6745035 DOI: 10.1007/s11010-019-03576-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022]
Abstract
A series of nine tetrahydroacridine derivatives with iodobenzoic moiety were synthesized and evaluated for their cytotoxic activity against cancer cell lines—A549 (human lung adenocarcinoma), HT-29 (human colorectal adenocarcinoma) and somatic cell line—EA.hy926 (human umbilical vein cell line). All compounds displayed high cytotoxicity activity against A549 (IC50 59.12–14.87 µM) and HT-29 (IC50 17.32–5.90 µM) cell lines, higher than control agents—etoposide and 5-fluorouracil. Structure–activity relationship showed that the position of iodine in the substituent in the para position and longer linker most strongly enhanced the cytotoxic effect. Among derivatives, 1i turned out to be the most cytotoxic and displayed IC50 values of 14.87 µM against A549 and 5.90 µM against HT-29 cell lines. In hyaluronidase inhibition assay, all compounds presented anti-inflammatory activity, however, slightly lower than reference compound. ADMET prediction showed that almost all compounds had good pharmacokinetic profiles. 1b, 1c and 1f compounds turned out to act against chemoresistance in cisplatin-resistant 253J B-V cells. Compounds intercalated into DNA and inhibited cell cycle in G0/G1 phase—the strongest inhibition was observed for 1i in A549 and 1c in HT-29. Among compounds, the highest apoptotic effect in both cell lines was observed after treatment with 1i. Compounds caused DNA damage and H2AX phosphorylation, which was detected in A549 and HT-29 cells. All research confirmed anticancer properties of novel tetrahydroacridine derivatives and explained a few pathways of their mechanism of cytotoxic action.
Collapse
Affiliation(s)
- Małgorzata Girek
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Karol Kłosiński
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Pabianicka 62, 93-513, Lodz, Poland
| | - Bartłomiej Grobelski
- Animal House, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Marie Angele Cucci
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Martina Daga
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Zbigniew Pasieka
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Pabianicka 62, 93-513, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland.
| |
Collapse
|
10
|
Chen R, Huo L, Jaiswal Y, Huang J, Zhong Z, Zhong J, Williams L, Xia X, Liang Y, Yan Z. Design, Synthesis, Antimicrobial, and Anticancer Activities of Acridine Thiosemicarbazides Derivatives. Molecules 2019; 24:molecules24112065. [PMID: 31151235 PMCID: PMC6600397 DOI: 10.3390/molecules24112065] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Acridine and thiourea derivatives are important compounds in medicinal chemistry due to their diverse biological properties including anticancer and antimicrobial effects. However, literature reveals some side effects associated with use of acridines. It is suggested that hybrid molecules may reduce the side effects and enhance the beneficial properties due to synergistic activity. The objectives of the present study are to synthesize and evaluate the anticancer and antimicrobial properties of new hybrids of acridine thiosemicarbazides derivatives. Results: The structures of the synthesized compounds 4a-4e were elucidated by MS and NMR spectra. In antimicrobial assay, Compound 4c exhibited potent antimicrobial activity compared to the other four compounds. In anticancer studies, we observed that compounds 4a, 4b, 4d and 4e exhibited high cytotoxicity against the MT-4 cell line, with IC50 values of 18.42 ± 1.18, 15.73 ± 0.90, 10.96 ± 0.62 and 11.63 ± 0.11 μM, respectively. The evaluation of anticancer effects, and the associated mechanism reveals that, the anticancer activities may be related to Topo I inhibitory activity, apoptosis and cell-cycle. Molecular docking studies revealed that the presence of planar naphtho-fused rings and a flexible thiourea group together, could improve DNA-intercalation and inhibition of DNA-Topo I activity. Conclusions: The results of this study demonstrate that the rational design of target derivatives as novel antimicrobial or antitumor leads is feasible.
Collapse
Affiliation(s)
- Rui Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
- Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning 530222, China.
| | - Lini Huo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
| | - Yogini Jaiswal
- Center for Excellence in Post-Harvest Technologies, North Carolina A&T State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC-28081, USA.
| | - Jiayong Huang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
| | - Zhenguo Zhong
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
| | - Jing Zhong
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
| | - Leonard Williams
- Center for Excellence in Post-Harvest Technologies, North Carolina A&T State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC-28081, USA.
| | - Xing Xia
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
| | - Yan Liang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| | - Zhenshuo Yan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China.
| |
Collapse
|
11
|
Janockova J, Korabecny J, Plsikova J, Babkova K, Konkolova E, Kucerova D, Vargova J, Koval J, Jendzelovsky R, Fedorocko P, Kasparkova J, Brabec V, Rosocha J, Soukup O, Hamulakova S, Kuca K, Kozurkova M. In vitro investigating of anticancer activity of new 7-MEOTA-tacrine heterodimers. J Enzyme Inhib Med Chem 2019; 34:877-897. [PMID: 30938202 PMCID: PMC6450562 DOI: 10.1080/14756366.2019.1593159] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
A combination of biochemical, biophysical and biological techniques was used to study calf thymus DNA interaction with newly synthesized 7-MEOTA-tacrine thiourea 12-17 and urea heterodimers 18-22, and to measure interference with type I and II topoisomerases. Their biological profile was also inspected in vitro on the HL-60 cell line using different flow cytometric techniques (cell cycle distribution, detection of mitochondrial membrane potential dissipation, and analysis of metabolic activity/viability). The compounds exhibited a profound inhibitory effect on topoisomerase activity (e.g. compound 22 inhibited type I topoisomerase at 1 µM concentration). The treatment of HL-60 cells with the studied compounds showed inhibition of cell growth especially with hybrids containing thiourea (14-17) and urea moieties (21 and 22). Moreover, treatment of human dermal fibroblasts with the studied compounds did not indicate significant cytotoxicity. The observed results suggest beneficial selectivity of the heterodimers as potential drugs to target cancer cells.
Collapse
Affiliation(s)
- Jana Janockova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic.,b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic
| | - Jan Korabecny
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic.,c Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences , University of Defence , Hradec Kralove , Czech Republic
| | - Jana Plsikova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic.,d Associated Tissue Bank, Faculty of Medicine , P.J. Šafárik University , Kosice , Slovak Republic
| | - Katerina Babkova
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic.,c Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences , University of Defence , Hradec Kralove , Czech Republic
| | - Eva Konkolova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Dana Kucerova
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Jana Vargova
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Jan Koval
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Rastislav Jendzelovsky
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Peter Fedorocko
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Jana Kasparkova
- f Department of Biophysics, Faculty of Science , Palacke University , Olomouc , Czech Republic
| | - Viktor Brabec
- f Department of Biophysics, Faculty of Science , Palacke University , Olomouc , Czech Republic
| | - Jan Rosocha
- d Associated Tissue Bank, Faculty of Medicine , P.J. Šafárik University , Kosice , Slovak Republic
| | - Ondrej Soukup
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic.,c Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences , University of Defence , Hradec Kralove , Czech Republic
| | - Slavka Hamulakova
- g Department of Organic Chemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Kamil Kuca
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic
| | - Maria Kozurkova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic.,b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic
| |
Collapse
|
12
|
Venkatapathy K, Magesh CJ, Lavanya G, Perumal PT, Sathishkumar R. A nanocrystalline CdS thin film as a heterogeneous, recyclable catalyst for effective synthesis of dihydropyrimidinones and a new class of carbazolyl dihydropyrimidinones via an improved Biginelli protocol. NEW J CHEM 2019. [DOI: 10.1039/c9nj02139f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An improved Biginelli protocol for the efficient synthesis of a new class of carbazolyl dihydropyrimidinones mediated by CdS thin film nanoparticles deposited on the inner walls of a glass reactor as a recyclable, heterogeneous catalyst.
Collapse
Affiliation(s)
| | | | - G. Lavanya
- Department of Chemistry
- AAGA College
- Cheyyar
- India
| | - P. T. Perumal
- B. S. Abdur Rahman Crescent Insititute of Science & Technology
- Chennai
- India
| | | |
Collapse
|
13
|
Chemical structures and biological activities of bis- and tetrakis-acridine derivatives: A review. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.05.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
14
|
|
15
|
Almeida SMVD, Lafayette EA, Silva WL, Lima Serafim VD, Menezes TM, Neves JL, Ruiz ALTG, Carvalho JED, Moura ROD, Beltrão EIC, Carvalho Júnior LBD, Lima MDCAD. New spiro-acridines: DNA interaction, antiproliferative activity and inhibition of human DNA topoisomerases. Int J Biol Macromol 2016; 92:467-475. [DOI: 10.1016/j.ijbiomac.2016.07.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/09/2016] [Accepted: 07/15/2016] [Indexed: 01/30/2023]
|
16
|
New benzimidazole acridine derivative induces human colon cancer cell apoptosis in vitro via the ROS-JNK signaling pathway. Acta Pharmacol Sin 2015; 36:1074-84. [PMID: 26235743 DOI: 10.1038/aps.2015.44] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/20/2015] [Indexed: 02/06/2023]
Abstract
AIM To investigate the mechanisms underlying anticancer action of the benzimidazole acridine derivative N-{(1H-benzo[d]imidazol-2-yl)methyl}-2-butylacridin-9-amine(8m) against human colon cancer cells in vitro. METHODS Human colon cancer cell lines SW480 and HCT116 were incubated in the presence of 8m, and then the cell proliferation and apoptosis were measured. The expression of apoptotic/signaling genes and proteins was detected using RT-PCR and Western blotting. ROS generation and mitochondrial membrane depolarization were visualized with fluorescence microscopy. RESULTS 8m dose-dependently suppressed the proliferation of SW480 and HCT116 cells with IC50 values of 6.77 and 3.33 μmol/L, respectively. 8m induced apoptosis of HCT116 cells, accompanied by down-regulation of Bcl-2, up-regulation of death receptor-5 (DR5), truncation of Bid, cleavage of PARP, and activation of caspases (including caspase-8 and caspase-9 as well as the downstream caspases-3 and caspase-7). Moreover, 8m selectively activated JNK and p38 without affecting ERK in HCT116 cells. Knockout of JNK1, but not p38, attenuated 8m-induced apoptosis. In addition, 8m induced ROS production and mitochondrial membrane depolarization in HCT116 cells. Pretreatment with the antioxidants N-acetyl cysteine or glutathione attenuated 8m-induced apoptosis and JNK activation in HCT116 cells. CONCLUSION The new benzimidazole acridine derivative, 8m exerts anticancer activity against human colon cancer cells in vitro by inducing both intrinsic and extrinsic apoptosis pathways via the ROS-JNK1 pathway.
Collapse
|
17
|
Solmont K, Boufroura H, Souibgui A, Fornarelli P, Gaucher A, Mahuteau-Betzer F, Ben Hassine B, Prim D. Divergent strategy for the synthesis of original dihydrobenzo- and dihydronaphtho-acridines. Org Biomol Chem 2015; 13:6269-77. [PMID: 25965873 DOI: 10.1039/c5ob00456j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A straightforward access to numerous novel substituted dihydrobenzo- and dihydronaphthoacridines is described using a unique molecular platform in two key steps. A large range of carbon-based substituents such as aromatic, vinyl, alkynyl fragments through Pd-catalysed couplings has been installed. The molecular diversity is extended to the introduction of aza-heterocycles and further authorizes the installation of alkylamino chains by means of Cu-promoted C-N bond formation. Possible access to quinolinium salts is also described. The methodology revealed convenient preparation of a wide panel of molecules that display various rigidity/flexibility and lipophilic/hydrophilic balances. Finally, the influence of structural modulations on the photophysical properties of these novel architectures is also studied. It is noteworthy that styryl and alkynyl derivatives are emissive in water (ϕF up to 12%).
Collapse
Affiliation(s)
- Kathleen Solmont
- Université de Versailles Saint-Quentin-en-Yvelines, Institut Lavoisier de Versailles, UMR CNRS 8180, 45, Avenue des Etats-Unis, 78035 Versailles cedex, France. damien.prim@.uvsq.fr
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Inhibition of DNA topoisomerases I and II and growth inhibition of HL-60 cells by novel acridine-based compounds. Eur J Pharm Sci 2015; 76:192-202. [PMID: 25960253 DOI: 10.1016/j.ejps.2015.04.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/04/2015] [Accepted: 04/25/2015] [Indexed: 11/20/2022]
Abstract
HL-60 cancer cells were treated with a series of novel acridine derivatives (derivatives 1-4) in order to test the compounds' ability to inhibit both cancer cell growth and topoisomerase I and II activity. Binding studies of derivatives 1-4 with calf thymus DNA were also performed using a number of techniques (UV-Vis and fluorescence spectroscopy, thermal denaturation, linear dichroism and viscometry) to determine the nature of the interaction between the compounds and ctDNA. The binding constants for the complexes of the studied acridine derivatives with DNA were calculated from UV-Vis spectroscopic titrations (K=3.1×10(4)-2.0×10(3)M(-1)). Some of the compounds showed a strong inhibitory effect against Topo II at the relatively low concentration of 5μM. Topo I/II inhibition mode assays were also performed and verified that the novel compounds are topoisomerase suppressors rather than poisons. The biological activities of derivatives were studied using MTT assay and flow cytometric methods (detection of mitochondrial membrane potential, measurement of cell viability) after 24 and 48h incubation. The ability of derivatives to impair cell proliferation was tested by an analysis of cell cycle distribution.
Collapse
|
19
|
|
20
|
|
21
|
Rescifina A, Zagni C, Varrica MG, Pistarà V, Corsaro A. Recent advances in small organic molecules as DNA intercalating agents: synthesis, activity, and modeling. Eur J Med Chem 2014; 74:95-115. [PMID: 24448420 DOI: 10.1016/j.ejmech.2013.11.029] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/26/2013] [Accepted: 11/28/2013] [Indexed: 11/28/2022]
Abstract
The interaction of small molecules with DNA plays an essential role in many biological processes. As DNA is often the target for majority of anticancer and antibiotic drugs, study about the interaction of drug and DNA has a key role in pharmacology. Moreover, understanding the interactions of small molecules with DNA is of prime significance in the rational design of more powerful and selective anticancer agents. Two of the most important and promising targets in cancer chemotherapy include DNA alkylating agents and DNA intercalators. For these last the DNA recognition is a critical step in their anti-tumor action and the intercalation is not only one kind of the interactions in DNA recognition but also a pivotal step of several clinically used anti-tumor drugs such as anthracyclines, acridines and anthraquinones. To push clinical cancer therapy, the discovery of new DNA intercalators has been considered a practical approach and a number of intercalators have been recently reported. The intercalative binding properties of such molecules can also be harnessed as diagnostic probes for DNA structure in addition to DNA-directed therapeutics. Moreover, the problem of intercalation site formation in the undistorted B-DNA of different length and sequence is matter of tremendous importance in molecular modeling studies and, nowadays, three models of DNA intercalation targets have been proposed that account for the binding features of intercalators. Finally, despite DNA being an important target for several drugs, most of the docking programs are validated only for proteins and their ligands. Therefore, a default protocol to identify DNA binding modes which uses a modified canonical DNA as receptor is needed.
Collapse
Affiliation(s)
- Antonio Rescifina
- Dipartimento di Scienze del Farmaco, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| | - Chiara Zagni
- Dipartimento di Scienze del Farmaco, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Maria Giulia Varrica
- Dipartimento di Scienze del Farmaco, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Venerando Pistarà
- Dipartimento di Scienze del Farmaco, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Antonino Corsaro
- Dipartimento di Scienze del Farmaco, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
22
|
Shapiro AB. A high-throughput-compatible, fluorescence anisotropy-based assay for ATP-dependent supercoiled DNA relaxation by human topoisomerase IIα. Biochem Pharmacol 2013; 85:1269-77. [PMID: 23415903 DOI: 10.1016/j.bcp.2013.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 11/29/2022]
Abstract
A novel, high-throughput-compatible assay for the ATP-dependent supercoiled DNA relaxing activity of human topoisomerase IIα (hTopoIIα) is described. The principle of detection is the preferential binding of the oligodeoxyribonucleotide BODIPY-TMR-5'-TTCTTCTTCT-3' to relaxed double-stranded plasmid containing the triplex forming sequence (TTC)9 versus the supercoiled plasmid. Binding of the oligonucleotide to the plasmid increases the fluorescence anisotropy of the BODIPY-TMR label. Optimization of the assay conditions was conducted to maximize the signal and the activity of the topoisomerase. The multiwell assay plate-based fluorescence anisotropy assay gave the same values for the potencies of several previously reported inhibitors of hTopoIIα as a gel electrophoresis-based assay of DNA relaxation.
Collapse
Affiliation(s)
- Adam B Shapiro
- Bioscience Department, Infection Innovative Medicines, AstraZeneca R&D Boston, Waltham, MA 02451 USA.
| |
Collapse
|
23
|
Mendonça EAM, Lira MCB, Rabello MM, Cavalcanti IMF, Galdino SL, Pitta IR, do Carmo A. Lima M, Pitta MGR, Hernandes MZ, Santos-Magalhães NS. Enhanced antiproliferative activity of the new anticancer candidate LPSF/AC04 in cyclodextrin inclusion complexes encapsulated into liposomes. AAPS PharmSciTech 2012; 13:1355-66. [PMID: 23054982 DOI: 10.1208/s12249-012-9853-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 09/10/2012] [Indexed: 11/30/2022] Open
Abstract
LPSF/AC04 (5Z)-[5-acridin-9-ylmethylene-3-(4-methyl-benzyl)-thiazolidine-2,4-dione] is an acridine-based derivative, part of a series of new anticancer agents synthesized for the purpose of developing more effective and less toxic anticancer drugs. However, the use of LPSF/AC04 is limited due to its low solubility in aqueous solutions. To overcome this problem, we investigated the interaction of LPSF/AC04 with hydroxypropyl-β-cyclodextrin (HP-β-CyD) and hydroxypropyl-γ-cyclodextrin (HP-γ-CyD) in inclusion complexes and determine which of the complexes formed presents the most significant interactions. In this paper, we report the physical characterization of the LPSF/AC04-HP-CyD inclusion complexes by thermogravimetric analysis, differential scanning calorimetry, infrared spectroscopy absorption, Raman spectroscopy, (1)HNMR, scanning electron microscopy, and by molecular modeling approaches. In addition, we verified that HP-β-CyD complexation enhances the aqueous solubility of LPSF/AC04, and a significant increase in the antiproliferative activity of LPSF/AC04 against cell lines can be achieved by the encapsulation into liposomes. These findings showed that the nanoencapsulation of LPSF/AC04 and LPSF/AC04-HP-CyD inclusion complexes in liposomes leads to improved drug penetration into the cells and, as a result, an enhancement of cytotoxic activity. Further in vivo studies comparing free and encapsulated LPSF/AC04 will be undertaken to support this investigation.
Collapse
|
24
|
|
25
|
Linero FN, Sepúlveda CS, Giovannoni F, Castilla V, García CC, Scolaro LA, Damonte EB. Host cell factors as antiviral targets in arenavirus infection. Viruses 2012; 4:1569-91. [PMID: 23170173 PMCID: PMC3499820 DOI: 10.3390/v4091569] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 08/31/2012] [Accepted: 09/04/2012] [Indexed: 12/11/2022] Open
Abstract
Among the members of the Arenaviridae family, Lassa virus and Junin virus generate periodic annual outbreaks of severe human hemorrhagic fever (HF) in endemic areas of West Africa and Argentina, respectively. Given the human health threat that arenaviruses represent and the lack of a specific and safe chemotherapy, the search for effective antiviral compounds is a continuous demanding effort. Since diverse host cell pathways and enzymes are used by RNA viruses to fulfill their replicative cycle, the targeting of a host process has turned an attractive antiviral approach in the last years for many unrelated virus types. This strategy has the additional benefit to reduce the serious challenge for therapy of RNA viruses to escape from drug effects through selection of resistant variants triggered by their high mutation rate. This article focuses on novel strategies to identify inhibitors for arenavirus therapy, analyzing the potential for antiviral developments of diverse host factors essential for virus infection.
Collapse
Affiliation(s)
- Florencia N Linero
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires/IQUIBICEN (CONICET), Ciudad Universitaria, Pabellón 2, Piso 4, Buenos Aires 1428, Argentina.
| | | | | | | | | | | | | |
Collapse
|
26
|
Bailly C. Contemporary challenges in the design of topoisomerase II inhibitors for cancer chemotherapy. Chem Rev 2012; 112:3611-40. [PMID: 22397403 DOI: 10.1021/cr200325f] [Citation(s) in RCA: 218] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Christian Bailly
- Centre de Recherche et Développement, Institut de Recherche Pierre Fabre, Toulouse, France.
| |
Collapse
|
27
|
Yu XM, Ramiandrasoa F, Guetzoyan L, Pradines B, Quintino E, Gadelle D, Forterre P, Cresteil T, Mahy JP, Pethe S. Synthesis and biological evaluation of acridine derivatives as antimalarial agents. ChemMedChem 2012; 7:587-605. [PMID: 22331612 DOI: 10.1002/cmdc.201100554] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/13/2012] [Indexed: 11/11/2022]
Abstract
New N-alkylaminoacridine derivatives attached to nitrogen heterocycles were synthesized, and their antimalarial potency was examined. They were tested in vitro against the growth of Plasmodium falciparum, including chloroquine (CQ)-susceptible and CQ-resistant strains. This biological evaluation has shown that the presence of a heterocyclic ring significantly increases the activity against P. falciparum. The best compound shows a nanomolar IC(50) value toward parasite proliferation on both CQ-susceptible and CQ-resistant strains. The antimalarial activity of these new acridine derivatives can be explained by the two mechanisms studied in this work. First, we showed the capacity of these compounds to inhibit heme biocrystallization, a detoxification process specific to the parasite and essential for its survival. Second, in our search for alternative targets, we evaluated the in vitro inhibitory activity of these compounds toward Sulfolobus shibatae topoisomerase VI-mediated DNA relaxation. The preliminary results obtained reveal that all tested compounds are potent DNA intercalators, and significantly inhibit the activity of S. shibatae topoisomerase VI at concentrations ranging between 2.0 and 2.5 μM.
Collapse
Affiliation(s)
- Xiao-Min Yu
- Université Paris-Sud 11, Institut de Chimie Moléculaire et des Matériaux d'Orsay, Equipe de Chimie Bioorganique et Bioinorganique, Orsay 91405 CEDEX, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sepúlveda CS, García CC, Fascio ML, D'Accorso NB, Docampo Palacios ML, Pellón RF, Damonte EB. Inhibition of Junin virus RNA synthesis by an antiviral acridone derivative. Antiviral Res 2011; 93:16-22. [PMID: 22027649 DOI: 10.1016/j.antiviral.2011.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/06/2011] [Accepted: 10/07/2011] [Indexed: 01/31/2023]
Abstract
There are no specific approved drugs for the treatment of agents of viral hemorrhagic fevers (HF) and antiviral therapies against these viruses are urgently needed. The present study characterizes the potent and selective antiviral activity against the HF causing arenavirus Junin virus (JUNV) of the compound 10-allyl-6-chloro-4-methoxy-9(10H)-acridone, designated 3f. The effectiveness of 3f to inhibit JUNV multiplication was not importantly affected by the initial multiplicity of infection, with similar effective concentration 50% (EC(50)) values in virus yield inhibition assays performed in Vero cells in the range of 0.2-40 plaque forming units (PFU)/cell. Mechanistic studies demonstrated that 3f did not affect the initial steps of adsorption and internalization. The subsequent process of viral RNA synthesis was strongly inhibited, as quantified by real time RT-PCR in compound-treated cells relative to non-treated cells. The addition of exogenous guanosine rescued the infectivity and RNA synthesis of JUNV in 3f-treated cells in a dose-dependent manner, but the reversal was partial, suggesting that the reduction of the GTP pool contributed to the antiviral activity of 3f, but it was not the main operative mechanism. The comparison of 3f with two other viral RNA inhibitors, ribavirin and mycophenolic acid, showed that ribavirin did not act against JUNV through the cellular enzyme inosine monophosphate dehydrogenase (IMPDH) inhibition whereas the anti-JUNV activity of mycophenolic acid was mainly targeted at this enzyme.
Collapse
Affiliation(s)
- Claudia S Sepúlveda
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428 Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
29
|
Cholewiński G, Dzierzbicka K, Kołodziejczyk AM. Natural and synthetic acridines/acridones as antitumor agents: their biological activities and methods of synthesis. Pharmacol Rep 2011; 63:305-36. [PMID: 21602588 DOI: 10.1016/s1734-1140(11)70499-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 08/13/2010] [Indexed: 10/25/2022]
Abstract
Acridine derivatives constitute a class of compounds that are being intensively studied as potential anticancer drugs. Acridines are well-known for their high cytotoxic activity; however, their clinical application is limited or even excluded because of side effects. Numerous synthetic methods are focused on the preparation of target acridine skeletons or modifications of naturally occurring compounds, such as acridone alkaloids, that exhibit promising anticancer activities. They have been examined in vitro and in vivo to test their importance for cancer treatment and to establish the mechanism of action at both the molecular and cellular level, which is necessary for the optimization of their properties so that they are suitable in chemotherapy. In this article, we review natural and synthetic acridine/acridone analogs, their application as anticancer drugs and methods for their preparation.
Collapse
Affiliation(s)
- Grzegorz Cholewiński
- Department of Organic Chemistry, Gdansk University of Technology, Narutowicza 11/12, PL 80-233 Gdańsk, Poland.
| | | | | |
Collapse
|
30
|
Kolokythas G, Daniilides K, Pouli N, Marakos P, Pratsinis H, Kletsas D. Design, synthesis, and cytotoxic activity evaluation of new linear pyranoxanthone aminoderivatives. J Heterocycl Chem 2011. [DOI: 10.1002/jhet.670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
31
|
Janovec L, Kožurková M, Sabolová D, Ungvarský J, Paulíková H, Plšíková J, Vantová Z, Imrich J. Cytotoxic 3,6-bis((imidazolidinone)imino)acridines: Synthesis, DNA binding and molecular modeling. Bioorg Med Chem 2011; 19:1790-801. [DOI: 10.1016/j.bmc.2011.01.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 01/04/2011] [Accepted: 01/08/2011] [Indexed: 11/27/2022]
|
32
|
Eang R, Girbal-Neuhauser E, Xu B, Gairin JE. Characterization and differential expression of a newly identified phosphorylated isoform of the human 20S proteasome beta7 subunit in tumor vs. normal cell lines. Fundam Clin Pharmacol 2009; 23:215-24. [PMID: 19645816 DOI: 10.1111/j.1472-8206.2009.00665.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The search of new pharmacological targets with original mechanism of action within the ubiquitin-proteasome pathway is still a goal to be reached in oncopharmacology. Modification by phosphorylation/dephosphorylation has been found to be involved in cancer and to regulate functional activity of proteasome. Until now, phosphorylated forms of alpha subunits of the 20S human proteasome have been mostly reported. Here, we have rationally designed a polyclonal antibody specifically directed against a phosphorylated peptide sequence bearing the beta7 subunit Ser249 residue of the human 20S proteasome. This anti-beta7 phosphoSer249 antibody appeared to be a probe of choice to detect the presence of a phosphorylated isoform of the beta7 subunit of the human 20S proteasome using mono or two-dimensional gel electrophoresis. PhosphoSer249 was sensitive to acid phosphatase treatment of native 20S proteasome. Dephosphorylation affected the peptidylglutamyl-peptide hydrolyzing activity whereas the chymotrypsin-like and trypsin-like activities remained unchanged. A comparative analysis between human normal and tumor cells showed a differential expression of the phosphoSer249 beta7 isoform with a significantly lower detection in the proteasome isolated from tumor cells, suggesting its possible use as a biomarker.
Collapse
Affiliation(s)
- Rothmony Eang
- Centre de Recherche en Pharmacologie-Santé, UMR 2587 CNRS-Pierre Fabre, ISTMT, 3 rue des satellites, 31400 Toulouse, France
| | | | | | | |
Collapse
|
33
|
Vantová Z, Paulíková H, Sabolová D, Kožurková M, Sucháňová M, Janovec L, Kristian P, Imrich J. Cytotoxic activity of acridin-3,6-diyl dithiourea hydrochlorides in human leukemia line HL-60 and resistant subline HL-60/ADR. Int J Biol Macromol 2009; 45:174-80. [DOI: 10.1016/j.ijbiomac.2009.04.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/20/2009] [Accepted: 04/27/2009] [Indexed: 10/20/2022]
|
34
|
Abstract
This mini review covers the drug discovery aspect of both proteasome activators and inhibitors. The proteasome is involved in many essential cellular functions, such as regulation of cell cycle, cell differentiation, signal transduction pathways, antigen processing for appropriate immune responses, stress signaling, inflammatory responses, and apoptosis. Due to the importance of the proteasome in cellular functions, inhibition or activation of the proteasome could become a useful therapeutic strategy for a variety of diseases. Many proteasome inhibitors have been identified and can be classified into two groups according to their source: chemically synthesized small molecules and compounds derived from natural products. A successful example of development of a proteasome inhibitor as a clinically useful drug is the peptide boronate, PS341 (Bortezomib), was approved for the treatment of multiple myeloma. In contrast to proteasome inhibitors, small molecules that can activate or enhance proteasome activity are rare and are not well studied. The fact that over-expression of the cellular proteasome activator PA28 exhibited beneficial effects on the Huntington's disease neuronal model cells raised the prospect that small molecule proteasome activators could become useful therapeutics. The beneficial effect of oleuropein, a small molecule proteasome activator, on senescence of human fibroblasts also suggested that proteasome activators might have the potential to be developed into anti-aging agents.
Collapse
Affiliation(s)
- Li Huang
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
35
|
Deciphering the antitumoral activity of quinacrine: Binding to and inhibition of Bcl-xL. Bioorg Med Chem Lett 2009; 19:1592-5. [DOI: 10.1016/j.bmcl.2009.02.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 02/03/2009] [Accepted: 02/04/2009] [Indexed: 11/22/2022]
|
36
|
Oppegard LM, Ougolkov AV, Luchini DN, Schoon RA, Goodell JR, Kaur H, Billadeau DD, Ferguson DM, Hiasa H. Novel acridine-based compounds that exhibit an anti-pancreatic cancer activity are catalytic inhibitors of human topoisomerase II. Eur J Pharmacol 2009; 602:223-9. [PMID: 19071108 PMCID: PMC2637346 DOI: 10.1016/j.ejphar.2008.11.044] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 11/03/2008] [Accepted: 11/18/2008] [Indexed: 10/21/2022]
Abstract
We have identified a small library of novel substituted 9-aminoacridine derivatives that inhibit cell proliferation of pancreatic cancer cell lines by inducing apoptosis [Goodell, J.R. et al., 2008. J. Med. Chem. 51, 179-182.]. To further investigate their antiproliferative activities, we have assessed the antiproliferative activity of these acridine-based compounds against several pancreatic cancer cell lines. All four compounds used in this study inhibited the proliferation of pancreatic cancer cell lines in vitro. In addition, we have employed a xenograft tumor model and found that these compounds also inhibit the proliferation of pancreatic cancer in vivo. In light of the potential importance of the anticancer activity of these acridine-based compounds, we have conducted a series of biochemical assays to determine the effect of these compounds on human topoisomerase II. Unlike amsacrine, these compounds do not poison topoisomerase II. Similar to amsacrine, however, these compounds intercalate into DNA in a way that they would alter the apparent topology of the DNA substrate. Thus, inhibition of the relaxation activity of topoisomerase II by these compounds has been reexamined using a DNA strand passage assay. We have found that these compounds, indeed, inhibit the catalytic activity of topoisomerase II. Thus, these novel acridine-based compounds with anti-pancreatic cancer activity are catalytic inhibitors, not poisons, of human topoisomerase II.
Collapse
Affiliation(s)
- Lisa M. Oppegard
- Department of Pharmacology, University of Minnesota Medical School-Twin Cities, Minneapolis, Minnesota 55455
| | | | | | | | | | | | | | | | - Hiroshi Hiasa
- Department of Pharmacology, University of Minnesota Medical School-Twin Cities, Minneapolis, Minnesota 55455
| |
Collapse
|
37
|
Physalin B, a novel inhibitor of the ubiquitin-proteasome pathway, triggers NOXA-associated apoptosis. Biochem Pharmacol 2008; 76:453-62. [DOI: 10.1016/j.bcp.2008.05.031] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 05/16/2008] [Accepted: 05/20/2008] [Indexed: 11/17/2022]
|