1
|
Kaya B, Smith H, Chen Y, Azad MG, Russell TM, Richardson V, Dharmasivam M, Richardson DR. Innovative N-Acridine Thiosemicarbazones and Their Zn(II) Complexes Transmetallate with Cu(II): Redox Activity and Suppression of Detrimental Oxy-Myoglobin Oxidation. Inorg Chem 2024; 63:20840-20858. [PMID: 39404641 DOI: 10.1021/acs.inorgchem.4c03642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The coordination chemistry and electrochemistry of novel N-acridine thiosemicarbazones (NATs) were investigated along with their redox activity, antiproliferative efficacy, transmetalation, and dissociation properties. The ability of NAT Fe(III) complexes to inhibit detrimental oxy-myoglobin (oxy-Mb) oxidation was also examined. The NATs act as tridentate ligands with a 2:1 L/Zn(II) complex crystal structure, revealing a distorted octahedral geometry, where both ligands bind Zn(II) in a meridional conformation. The NAT Fe(III) complexes exhibited fully reversible one-electron FeIII/II couples with more positive potentials than the Fe(III) complexes of a related clinically trialed thiosemicarbazone (e.g., [Fe(DpC)2]+) due to the electron-donating capacity of acridine. Surprisingly, the NAT-Zn(II) complexes showed generally greater or similar antiproliferative activity than their ligands, Cu(II), or Fe(III) complexes. This may be explained by (1) formation of a highly lipophilic Zn(II) complex that acts as a chaperone to promote cellular uptake and (2) the capacity of the Zn(II) complex to dissociate or undergo transmetalation to the redox-active Cu(II) complex. Of the NAT-Fe(III) complexes, [Fe(AOBP)2]+ demonstrated a significant (p < 0.0001) improvement in preventing oxy-Mb oxidation than the Fe(III) complex of the clinically trialed thiosemicarbazone, DpC. This article advances our understanding of NAT coordination chemistry, electrochemistry, and the intriguing biological activity of their complexes.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Henry Smith
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Yanbing Chen
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
2
|
Fonte M, Rôla C, Santana S, Prudêncio M, Almeida J, Ferraz R, Prudêncio C, Teixeira C, Gomes P. Repurposing antiplasmodial leads for cancer: Exploring the antiproliferative effects of N-cinnamoyl-aminoacridines. Bioorg Med Chem Lett 2024; 111:129894. [PMID: 39043264 DOI: 10.1016/j.bmcl.2024.129894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/26/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
Drug repurposing and rescuing have been widely explored as cost-effective approaches to expand the portfolio of chemotherapeutic agents. Based on the reported antitumor properties of both trans-cinnamic acids and quinacrine, an antimalarial aminoacridine, we explored the antiproliferative properties of two series of N-cinnamoyl-aminoacridines recently identified as multi-stage antiplasmodial leads. The compounds were evaluated in vitro against three cancer cell lines (MKN-28, Huh-7, and HepG2), and human primary dermal fibroblasts. One of the series displayed highly selective antiproliferative activity in the micromolar range against the three cancer cell lines tested, without any toxicity to non-carcinogenic cells.
Collapse
Affiliation(s)
- Mélanie Fonte
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal
| | - Catarina Rôla
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Sofia Santana
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Joana Almeida
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Ricardo Ferraz
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Cristina Prudêncio
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Cátia Teixeira
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Gyros Protein Technologies Inc., Tucson, AZ, USA
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal.
| |
Collapse
|
3
|
Jung HM, Ha JH, dela Cerna MVC, Burlison JA, Choi J, Kim BR, Bang JK, Ryu KS, Lee D. An Innovative Inhibitor with a New Chemical Moiety Aimed at Biliverdin IXβ Reductase for Thrombocytopenia and Resilient against Cellular Degradation. Pharmaceutics 2024; 16:1148. [PMID: 39339186 PMCID: PMC11435328 DOI: 10.3390/pharmaceutics16091148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Biliverdin IXβ reductase (BLVRB) has emerged as a promising therapeutic target for thrombocytopenia due to its involvement in reactive oxygen species (ROS) mechanisms. During the pursuit of inhibitors targeting BLVRB, olsalazine (OSA) became apparent as one of the most potent candidates. However, the direct application of OSA as a BLVRB inhibitor faces challenges, as it is prone to degradation into 5-aminosalicylic acid through cleavage of the diazenyl bond by abundant azoreductase (AzoR) enzymes in gut microbiota and eukaryotic cells. To overcome this obstacle, we devised olsalkene (OSK), an inhibitor where the diazenyl bond in OSA has been substituted with an alkene bond. OSK not only matches the efficacy of OSA but also demonstrates improved stability against degradation by AzoR, presenting a promising solution to this limitation. Furthermore, we have found that both OSK and OSA inhibit BLVRB, regardless of the presence of nicotinamide adenine dinucleotide phosphate, unlike other known inhibitors. This discovery opens new avenues for investigating the roles of BLVRB in blood disorders, including thrombocytopenia.
Collapse
Affiliation(s)
- Hoe-Myung Jung
- Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si 28119, Republic of Korea; (H.-M.J.); (J.C.); (B.-R.K.); (J.K.B.)
- Department of Bio-Analytical Science, University of Science & Technology, Daejoen 34113, Republic of Korea
| | - Jung-Hye Ha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea;
| | - Mark Vincent C. dela Cerna
- Department of Biochemistry, Chemistry and Physics, Georgia Southern University, 11935 Abercorn Street, Savannah, GA 31419, USA;
| | - Joseph A. Burlison
- Department of Medicine, James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock St., Louisville, KY 40202, USA;
| | - Joonhyeok Choi
- Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si 28119, Republic of Korea; (H.-M.J.); (J.C.); (B.-R.K.); (J.K.B.)
| | - Bo-Ram Kim
- Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si 28119, Republic of Korea; (H.-M.J.); (J.C.); (B.-R.K.); (J.K.B.)
| | - Jeong Kyu Bang
- Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si 28119, Republic of Korea; (H.-M.J.); (J.C.); (B.-R.K.); (J.K.B.)
- Department of Bio-Analytical Science, University of Science & Technology, Daejoen 34113, Republic of Korea
- Dandicure Inc., Ochang-Eup, Cheongju-Si 28119, Republic of Korea
| | - Kyoung-Seok Ryu
- Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si 28119, Republic of Korea; (H.-M.J.); (J.C.); (B.-R.K.); (J.K.B.)
- Department of Bio-Analytical Science, University of Science & Technology, Daejoen 34113, Republic of Korea
| | - Donghan Lee
- Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si 28119, Republic of Korea; (H.-M.J.); (J.C.); (B.-R.K.); (J.K.B.)
| |
Collapse
|
4
|
Huang SC, Chen CW, Satange R, Hsieh CC, Chang CC, Wang SC, Peng CL, Chen TL, Chiang MH, Horng YC, Hou MH. Targeting DNA junction sites by bis-intercalators induces topological changes with potent antitumor effects. Nucleic Acids Res 2024; 52:9303-9316. [PMID: 39036959 PMCID: PMC11347135 DOI: 10.1093/nar/gkae643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/21/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
Targeting inter-duplex junctions in catenated DNA with bidirectional bis-intercalators is a potential strategy for enhancing anticancer effects. In this study, we used d(CGTATACG)2, which forms a tetraplex base-pair junction that resembles the DNA-DNA contact structure, as a model target for two alkyl-linked diaminoacridine bis-intercalators, DA4 and DA5. Cross-linking of the junction site by the bis-intercalators induced substantial structural changes in the DNA, transforming it from a B-form helical end-to-end junction to an over-wounded side-by-side inter-duplex conformation with A-DNA characteristics and curvature. These structural perturbations facilitated the angled intercalation of DA4 and DA5 with propeller geometry into two adjacent duplexes. The addition of a single carbon to the DA5 linker caused a bend that aligned its chromophores with CpG sites, enabling continuous stacking and specific water-mediated interactions at the inter-duplex contacts. Furthermore, we have shown that the different topological changes induced by DA4 and DA5 lead to the inhibition of topoisomerase 2 activities, which may account for their antitumor effects. Thus, this study lays the foundations for bis-intercalators targeting biologically relevant DNA-DNA contact structures for anticancer drug development.
Collapse
Affiliation(s)
- Shih-Chun Huang
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Chia-Wei Chen
- Department of Chemistry, National Changhua University of Education, Changhua 50058, Taiwan
| | - Roshan Satange
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | | | - Chih-Chun Chang
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Shun-Ching Wang
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Chi-Li Peng
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Tai-Lin Chen
- Post Baccalaureate Medicine, School of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Ming-Hsi Chiang
- Institute of Chemistry, Academia Sinica, Taipei 11528, Taiwan
| | - Yih-Chern Horng
- Department of Chemistry, National Changhua University of Education, Changhua 50058, Taiwan
| | - Ming-Hon Hou
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
5
|
Kaya B, Smith H, Chen Y, Azad MG, M Russell T, Richardson V, Bernhardt PV, Dharmasivam M, Richardson DR. Targeting lysosomes by design: novel N-acridine thiosemicarbazones that enable direct detection of intracellular drug localization and overcome P-glycoprotein (Pgp)-mediated resistance. Chem Sci 2024:d4sc04339a. [PMID: 39165729 PMCID: PMC11331336 DOI: 10.1039/d4sc04339a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024] Open
Abstract
Innovative N-acridine thiosemicarbazones (NATs) were designed along with their iron(iii), copper(ii), and zinc(ii) complexes. Lysosomal targeting was promoted by specifically incorporating the lysosomotropic Pgp substrate, acridine, into the thiosemicarbazone scaffold to maintain the tridentate N, N, S-donor system. The acridine moiety enables a significant advance in thiosemicarbazone design, since: (1) it enables tracking of the drugs by confocal microscopy using its inherent fluorescence; (2) it is lysosomotropic enabling lysosomal targeting; and (3) as acridine is a P-glycoprotein (Pgp) substrate, it facilitates lysosomal targeting, resulting in the drug overcoming Pgp-mediated resistance. These new N-acridine analogues are novel, and this is the first time that acridine has been specifically added to the thiosemicarbazone framework to achieve the three important properties above. These new agents displayed markedly greater anti-proliferative activity against resistant Pgp-expressing cells than very low Pgp-expressing cells. The anti-proliferative activity of NATs against multiple Pgp-positive cancer cell-types (colon, lung, and cervical carcinoma) was abrogated by the third generation Pgp inhibitor, Elacridar, and also Pgp siRNA that down-regulated Pgp. Confocal microscopy demonstrated that low Pgp in KB31 (-Pgp) cells resulted in acridine's proclivity for DNA intercalation promoting NAT nuclear-targeting. In contrast, high Pgp in KBV1 (+Pgp) cells led to NAT lysosomal sequestration, preventing its nuclear localisation. High Pgp expression in KBV1 (+Pgp) cells resulted in co-localization of NATs with the lysosomal marker, LysoTracker™, that was significantly (p < 0.001) greater than the positive control, the di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) Zn(ii) complex, [Zn(DpC)2]. Incorporation of acridine into the thiosemicarbazone scaffold led to Pgp-mediated transport into lysosomes to overcome Pgp-resistance.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Henry Smith
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Yanbing Chen
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland Brisbane 4072 Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine Nagoya 466-8550 Japan
| |
Collapse
|
6
|
Zhou HY, Dong L. Synthesis of acridones via Ir(III)-catalyzed amination annulation of oxazoles with anthranils. Org Biomol Chem 2024; 22:4036-4040. [PMID: 38698770 DOI: 10.1039/d4ob00377b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
An unprecedented Ir(III)-catalyzed C-H activation/amination/annulation of 2-phenyloxazoles with anthranils for the highly selective preparation of acridone derivatives in one-pot under controlled conditions is reported. This protocol is characterized by atom economy and high regioselectivity. A wide range of anthranils with 2-phenyloxazoles were well tolerated and afforded the desired products in moderate to good yields, in which the anthranil serves as a convenient amination reagent.
Collapse
Affiliation(s)
- Han-Yi Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Lin Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Galenko EE, Novikov MS, Bunev AS, Khlebnikov AF. Acridine-Isoxazole and Acridine-Azirine Hybrids: Synthesis, Photochemical Transformations in the UV/Visible Radiation Boundary Region, and Anticancer Activity. Molecules 2024; 29:1538. [PMID: 38611817 PMCID: PMC11013717 DOI: 10.3390/molecules29071538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Easy-to-handle N-hydroxyacridinecarbimidoyl chloride hydrochlorides were synthesized as convenient nitrile oxide precursors in the preparation of 3-(acridin-9/2-yl)isoxazole derivatives via 1,3-dipolar cycloaddition with terminal alkynes, 1,1-dichloroethene, and acrylonitrile. Azirines with an acridin-9/2-yl substituent attached directly or via the 1,2,3-triazole linker to the azirine C2 were also synthesized. The three-membered rings of the acridine-azirine hybrids were found to be resistant to irradiation in the UV/visible boundary region, despite their long-wave absorption at 320-420 nm, indicating that the acridine moiety cannot be used as an antenna to transfer light energy to generate nitrile ylides from azirines for photoclick cycloaddition. The acridine-isoxazole hybrids linked at the C9-C3 or C2-C3 atoms under blue light irradiation underwent the addition of such hydrogen donor solvents, such as, toluene, o-xylene, mesitylene, 4-chlorotoluene, THF, 1,4-dioxane, or methyl tert-butyl ether (MTBE), to the acridine system to give the corresponding 9-substituted acridanes in good yields. The synthesized acridine-azirine, acridine-isoxazole, and acridane-isoxazole hybrids exhibited cytotoxicity toward both all tested cancer cell lines (HCT 116, MCF7, and A704) and normal cells (WI-26 VA4).
Collapse
Affiliation(s)
- Ekaterina E. Galenko
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya Naberezhnaya, St. Petersburg 199034, Russia; (E.E.G.); (M.S.N.)
| | - Mikhail S. Novikov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya Naberezhnaya, St. Petersburg 199034, Russia; (E.E.G.); (M.S.N.)
| | - Alexander S. Bunev
- Medicinal Chemistry Center, Togliatti State University, Togliatti 445020, Russia;
| | - Alexander F. Khlebnikov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya Naberezhnaya, St. Petersburg 199034, Russia; (E.E.G.); (M.S.N.)
| |
Collapse
|
8
|
Fonte M, Teixeira C, Gomes P. Improved synthesis of antiplasmodial 4-aminoacridines and 4,9-diaminoacridines. RSC Adv 2024; 14:6253-6261. [PMID: 38375018 PMCID: PMC10875606 DOI: 10.1039/d4ra00091a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/07/2024] [Indexed: 02/21/2024] Open
Abstract
Acridines are one of the most important nitrogen-containing heterocycle systems and have many applications in the therapeutic field. However, the synthesis of acridine-based scaffolds is not always straightforward. Herein, we report the optimization of two multi-step synthetic routes towards 4,9-diaminoacridines and 4-aminoacridines, which have shown promising antiplasmodial properties. The improved synthesis pathways make use of greener, simpler, and more efficient methods, with less reaction steps and increased overall yields, which were doubled in some cases. These are impactful results towards future approaches to the chemical synthesis of acridine-based compounds.
Collapse
Affiliation(s)
- Mélanie Fonte
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto Portugal
| | - Cátia Teixeira
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto Portugal
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto Portugal
| |
Collapse
|
9
|
Schlosser J, Fedorova O, Fedorov Y, Ihmels H. Photoinduced in situ generation of DNA-targeting ligands: DNA-binding and DNA-photodamaging properties of benzo[ c]quinolizinium ions. Beilstein J Org Chem 2024; 20:101-117. [PMID: 38264449 PMCID: PMC10804566 DOI: 10.3762/bjoc.20.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024] Open
Abstract
The photoreactions of selected styrylpyridine derivatives to the corresponding benzo[c]quinolizinium ions are described. It is shown that these reactions are more efficient in aqueous solution (97-44%) than in organic solvents (78-20% in MeCN). The quinolizinium derivatives bind to DNA by intercalation with binding constants of 6-11 × 104 M-1, as shown by photometric and fluorimetric titrations as well as by CD- and LD-spectroscopic analyses. These ligand-DNA complexes can also be established in situ upon irradiation of the styrylpyridines and formation of the intercalator directly in the presence of DNA. In addition to the DNA-binding properties, the tested benzo[c]quinolizinium derivatives also operate as photosensitizers, which induce DNA damage at relative low concentrations and short irradiation times, even under anaerobic conditions. Investigations of the mechanism of the DNA damage revealed the involvement of intermediate hydroxyl radicals and C-centered radicals. Under aerobic conditions, singlet oxygen only contributes to marginal extent to the DNA damage.
Collapse
Affiliation(s)
- Julika Schlosser
- Department of Chemistry and Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cµ), University of Siegen, Adolf-Reichwein-Str. 2, D-57068 Siegen, Germany
| | - Olga Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova str. 28, 119991 Moscow, Russia
| | - Yuri Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova str. 28, 119991 Moscow, Russia
| | - Heiko Ihmels
- Department of Chemistry and Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cµ), University of Siegen, Adolf-Reichwein-Str. 2, D-57068 Siegen, Germany
| |
Collapse
|
10
|
Lee YC, Chiou JT, Wang LJ, Chen YJ, Chang LS. Amsacrine downregulates BCL2L1 expression and triggers apoptosis in human chronic myeloid leukemia cells through the SIDT2/NOX4/ERK/HuR pathway. Toxicol Appl Pharmacol 2023; 474:116625. [PMID: 37451322 DOI: 10.1016/j.taap.2023.116625] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Accumulating evidence indicates that the anticancer activity of acridine derivatives is mediated through the regulation of anti-apoptotic and pro-apoptotic BCL2 protein expression. Therefore, we investigated whether the cytotoxicity of amsacrine with an acridine structural scaffold in human chronic myeloid leukemia (CML) K562 cells was mediated by BCL2 family proteins. Amsacrine induced apoptosis, mitochondrial depolarization, and BCL2L1 (also known as BCL-XL) downregulation in K562 cells. BCL2L1 overexpression inhibited amsacrine-induced cell death and mitochondrial depolarization. Amsacrine treatment triggered SIDT2-mediated miR-25 downregulation, leading to increased NOX4-mediated ROS production. ROS-mediated inactivation of ERK triggered miR-22 expression, leading to increased HuR mRNA decay. As HuR is involved in stabilizing BCL2L1 mRNA, downregulation of BCL2L1 was noted in K562 cells after amsacrine treatment. In contrast, amsacrine-induced BCL2L1 downregulation was alleviated by restoring ERK phosphorylation and HuR expression. Altogether, the results of this study suggest that amsacrine triggers apoptosis in K562 cells by inhibiting BCL2L1 expression through the SIDT2/NOX4/ERK-mediated downregulation of HuR. Furthermore, a similar pathway also explains the cytotoxicity of amsacrine in CML MEG-01 and KU812 cells.
Collapse
Affiliation(s)
- Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Ying-Jung Chen
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
11
|
Alam M, Ahmed S, Abid M, Hasan GM, Islam A, Hassan MI. Therapeutic targeting of microtubule affinity-regulating kinase 4 in cancer and neurodegenerative diseases. J Cell Biochem 2023; 124:1223-1240. [PMID: 37661636 DOI: 10.1002/jcb.30468] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/12/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
Microtubule affinity-regulating kinase 4 (MARK4) is a member of the Ser/Thr protein kinase family, phosphorylates the microtubule-connected proteins and plays a vital role in causing cancers and neurodegenerative diseases. This kinase modulates multiple signaling pathways, including mammalian target of rapamycin, nuclear factor-κB, and Hippo-signaling, presumably responsible for cancer and Alzheimer's. MARK4 acts as a negative controller of the Hippo-kinase cassette for promoting YAP/TAZ action, and the loss of MARK4 detains the tumorigenic properties of cancer cells. MARK4 is involved in tau hyperphosphorylation that consequently affects neurodegeneration. MARK4 is a promising drug target for cancer, diabetes, and Alzheimer's. Developing the potent and selective inhibitors of MAKR4 are promising in the therapeutic management of associated diseases. Despite its great significance, a few reviews are available to discuss its structure, function and clinical significance. In the current review, we aimed to provide detailed information on the structural features of MARK4 targeted in drug development and its role in various signaling pathways related to cancer and neurodegenerative diseases. We further described the therapeutic potential of MARK4 inhibitors in preventing numerous diseases. Finally, the updated information on MARK4 will be helpful in the further development of effective therapeutic molecules.
Collapse
Affiliation(s)
- Manzar Alam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Sarfraz Ahmed
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Abid
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
12
|
Voura M, Anwar S, Sigala I, Parasidou E, Fragoulidou S, Hassan MI, Sarli V. Synthesis, Structural Modification, and Bioactivity Evaluation of Substituted Acridones as Potent Microtubule Affinity-Regulating Kinase 4 Inhibitors. ACS Pharmacol Transl Sci 2023; 6:1052-1074. [PMID: 37470016 PMCID: PMC10353068 DOI: 10.1021/acsptsci.3c00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Indexed: 07/21/2023]
Abstract
Acridones present numerous pharmacological activities, including inhibition of microtubule affinity-regulating kinase 4 (MARK4) kinase activity. To investigate structure-activity relationships and develop potent MARK4 inhibitors, derivatives of 2-methylacridone were synthesized and tested for their activity against MARK4 kinase. Selective substitutions at the nitrogen atom were accomplished by treating 2-methylacridone with alkyl halides in the presence of K2CO3. In addition, amidation of acridone acetic acid 11 with piperazine or tryptophan methyl ester followed by derivatization with various amines gave a series of new acridone derivatives. Among the tested compounds, six were identified as possessing high inhibitory activity against MARK4. The molecular modeling studies showed that the derivatives bearing piperazine or tryptophan bind well to the ATP-binding site of MARK4. The antiproliferative activity of six active compounds was evaluated against HeLa and U87MG cancer cells. Tryptophan derivatives 23a, 23b, and 23c showed significant cytotoxicity against both cell lines with EC50 values ranging from 2.13 to 4.22 μM, while derivatives bearing piperazine were found to be not cytotoxic. Additionally, compound 23a decreased the proliferation of human MDA-MB-435 and U251 cancer cells in the low micromolar range; however, it also affects the non-cancerous HGF cells. Due to their high binding affinity against MARK4, the synthesized compounds could be potential agents to target MARK4 against cancer and tauopathies.
Collapse
Affiliation(s)
- Maria Voura
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Saleha Anwar
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ioanna Sigala
- Laboratory
of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleftheria Parasidou
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Souzanna Fragoulidou
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Md. Imtaiyaz Hassan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Vasiliki Sarli
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
13
|
Sánchez JD, Alcántara AR, González JF, Sánchez-Montero JM. Advances in the discovery of heterocyclic-based drugs against Alzheimer's disease. Expert Opin Drug Discov 2023; 18:1413-1428. [PMID: 37800875 DOI: 10.1080/17460441.2023.2264766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Alzheimer's disease is a multifactorial neurodegenerative disorder characterized by beta-amyloid accumulation and tau protein hyperphosphorylation. The disease involves interconnected mechanisms, which can be clustered into two target-packs based on the affected proteins. Pack-1 focuses on beta-amyloid accumulation, oxidative stress, and metal homeostasis dysfunction, and Pack-2 involves tau protein, calcium homeostasis, and neuroinflammation. Against this background heterocyclic system, there is a powerful source of pharmacophores to develop effective small drugs to treat multifactorial diseases like Alzheimer's. AREAS COVERED This review highlights the most promising heterocyclic systems as potential hit candidates with multi-target capacity for the development of new drugs targeting Alzheimer's disease. The selection of these heterocyclic systems was based on two crucial factors: their synthetic versatility and their well-documented biological properties of therapeutic potential in neurodegenerative diseases. EXPERT OPINION The synthesis of small drugs against Alzheimer's disease requires a multifactorial approach that targets the key pathological proteins. In this context, the utilization of heterocyclic systems, with well-established synthetic processes and facile functionalization, becomes a crucial element in the design phases. Furthermore, the selection of hit heterocyclic should be guided by a full understanding of their biological activities. Thus, the identification of promising heterocyclic scaffolds with known biological effects increases the potential to develop effective molecules against Alzheimer's disease.
Collapse
Affiliation(s)
- Juan D Sánchez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Andrés R Alcántara
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan F González
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - José María Sánchez-Montero
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
14
|
Liu L, Zhang Y, Zhao W, Wen J, Dong C, Hu C, Li J. Photoredox-Catalyzed Cascade sp 2 C-H Bond Functionalization to Construct Substituted Acridine with Diarylamine and Hypervalent Iodine(III) Reagents. Org Lett 2023; 25:592-596. [PMID: 36656299 DOI: 10.1021/acs.orglett.2c04114] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A photocatalyzed cascade double C-C formation via sp2 C-H bond activation of diarylamines with hypervalent iodine diazo reagents was developed. A variety of diarylamines and hypervalent iodine(III) reagents were tolerated well, and a range of substituted acridines with yields ranging from moderate to excellent was provided efficiently. The protocol introduces diazo groups onto diarylamines and enables subsequent late-stage assembly point functionalization with the diazonium structure, forming two new C-C bonds in a sequential fashion.
Collapse
Affiliation(s)
- Li Liu
- School of Pharmacy, Changzhou University, Changzhou 213164, China.,Analysis and Testing Center, NERC Biomass of Changzhou University, Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, Changzhou University, Changzhou 213164, China
| | - Yage Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Wenyan Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jinxia Wen
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Chunping Dong
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Caijuan Hu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jian Li
- School of Pharmacy, Changzhou University, Changzhou 213164, China.,Analysis and Testing Center, NERC Biomass of Changzhou University, Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, Changzhou University, Changzhou 213164, China
| |
Collapse
|
15
|
Różycka D, Kowalczyk A, Denel-Bobrowska M, Kuźmycz O, Gapińska M, Stączek P, Olejniczak AB. Acridine/Acridone-Carborane Conjugates as Strong DNA-Binding Agents with Anticancer Potential. ChemMedChem 2023; 18:e202200666. [PMID: 36734215 DOI: 10.1002/cmdc.202200666] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/04/2023]
Abstract
Synthesis of acridine derivatives that act as DNA-targeting anticancer agents is an evolving field and has resulted in the introduction of several drugs into clinical trials. Carboranes can be of importance in designing biologically active compounds due to their specific properties. Therefore, a series of novel acridine analogs modified with carborane clusters were synthesized. The DNA-binding ability of these analogs was evaluated on calf thymus DNA (ct-DNA). Results of these analyses showed that 9-[(1,7-dicarba-closo-dodecaborane-1-yl)propylamino]acridine (30) interacted strongly with ct-DNA, indicating its ability to intercalate into DNA, whereas 9-[(1,7-dicarba-closo-dodecaborane-1-yl)propanamido]acridine (29) changed the B-form of ct-DNA to the Z form. Compound 30 demonstrated cytotoxicity, was able to inhibit cell proliferation, arrest the cell cycle in the S phase in the HeLa cancer cell line, and induced the production of reactive oxygen species (ROS). In addition, it was specifically localized in lysosomes and was a weak inhibitor of Topo IIα.
Collapse
Affiliation(s)
- Daria Różycka
- Screening Laboratory, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., Łódź, 93-232, Poland
| | - Aleksandra Kowalczyk
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., Łódź, 90-237, Poland
| | - Marta Denel-Bobrowska
- Screening Laboratory, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., Łódź, 93-232, Poland
| | - Olga Kuźmycz
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., Łódź, 90-237, Poland
| | - Magdalena Gapińska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology Environmental Protection, University of Lodz, 12/16 Banacha St., Łódź, 90-237, Poland
| | - Paweł Stączek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., Łódź, 90-237, Poland
| | - Agnieszka B Olejniczak
- Screening Laboratory, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., Łódź, 93-232, Poland
| |
Collapse
|
16
|
Okoro CO, Fatoki TH. A Mini Review of Novel Topoisomerase II Inhibitors as Future Anticancer Agents. Int J Mol Sci 2023; 24:ijms24032532. [PMID: 36768852 PMCID: PMC9916523 DOI: 10.3390/ijms24032532] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Several reviews of inhibitors of topoisomerase II have been published, covering research before 2018. Therefore, this review is focused primarily on more recent publications with relevant points from the earlier literature. Topoisomerase II is an established target for anticancer drugs, which are further subdivided into poisons and catalytic inhibitors. While most of the topoisomerase II-based drugs in clinical use are mostly topoisomerase II poisons, their mechanism of action has posed severe concern due to DNA damaging potential, including the development of multi-drug resistance. As a result, we are beginning to see a gradual paradigm shift towards non-DNA damaging agents, such as the lesser studied topoisomerase II catalytic inhibitors. In addition, this review describes some novel selective catalytic topoisomerase II inhibitors. The ultimate goal is to bring researchers up to speed by curating and delineating new scaffolds as the leads for the optimization and development of new potent, safe, and selective agents for the treatment of cancer.
Collapse
|
17
|
Excited-State Dynamics of Proflavine after Intercalation into DNA Duplex. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238157. [PMID: 36500248 PMCID: PMC9738913 DOI: 10.3390/molecules27238157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Proflavine is an acridine derivative which was discovered as one of the earliest antibacterial agents, and it has been proven to have potential application to fields such as chemotherapy, photobiology and solar-energy conversion. In particular, it is well known that proflavine can bind to DNA with different modes, and this may open addition photochemical-reaction channels in DNA. Herein, the excited-state dynamics of proflavine after intercalation into DNA duplex is studied using femtosecond time-resolved spectroscopy, and compared with that in solution. It is demonstrated that both fluorescence and the triplet excited-state generation of proflavine were quenched after intercalation into DNA, due to ultrafast non-radiative channels. A static-quenching mechanism was identified for the proflavine-DNA complex, in line with the spectroscopy data, and the excited-state deactivation mechanism was proposed.
Collapse
|
18
|
Kondaka K, Gabriel I. Targeting DNA Topoisomerase II in Antifungal Chemotherapy. Molecules 2022; 27:molecules27227768. [PMID: 36431868 PMCID: PMC9698242 DOI: 10.3390/molecules27227768] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Topoisomerase inhibitors have been in use clinically for the treatment of several diseases for decades. Although those enzymes are significant molecular targets in antibacterial and anticancer chemotherapy very little is known about the possibilities to target fungal topoisomerase II (topo II). Raising concern for the fungal infections, lack of effective drugs and a phenomenon of multidrug resistance underlie a strong need to expand the range of therapeutic options. In this review paper, we discussed the usefulness of fungal topo II as a molecular target for new drug discovery. On the basis of previously published data, we described structural and biochemical differences between fungal and human enzymes as well as a molecular basis of differential sensitivity to known anticancer drugs targeting the latter. This review focuses especially on highlighting the differences that may underlie the selectivity of action of new inhibitors. Distinct sites within fungal topo II in comparison with human counterparts are observed and should be further studied to understand the significance of those sites and their possible usage in design of new drugs.
Collapse
Affiliation(s)
| | - Iwona Gabriel
- Correspondence: ; Tel.: +48-58-348-6078; Fax: +48-58-347-1144
| |
Collapse
|
19
|
Elmusa M, Elmusa S, Mert S, Kasımoğulları R, Türkan F, Atalar MN, Bursal E. One-pot three-component synthesis of novel pyrazolo-acridine derivatives and assessment of their acetylcholinesterase inhibitory properties: An in vitro and in silico study. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Goni LKMO, Jafar Mazumder MA, Tripathy DB, Quraishi MA. Acridine and Its Derivatives: Synthesis, Biological, and Anticorrosion Properties. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7560. [PMID: 36363152 PMCID: PMC9658428 DOI: 10.3390/ma15217560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
The phenomenon of corrosion threatens metallic components, human safety, and the economy. Despite being eco-friendly and promising as a corrosion inhibitor, acridine has not been explored to its full potential. In this review, we have discussed multiple biological activities that acridines have been found to show in a bid to prove that they are environmentally benign and much less toxic than many inhibitors. Some synthetic routes to acridines and substituted acridines have also been discussed. Thereafter, a multitude of acridines and substituted acridines as corrosion inhibitors of different metals and alloys in various corrosive media have been highlighted. A short mechanistic insight into how acridine-based compounds function as corrosion inhibitors have also been included. We believe this review will generate an impression that there is still much to learn about previously reported acridines. In the wake of recent surges to find efficient and non-toxic corrosion inhibitors, acridines and their analogs could be an appropriate answer.
Collapse
Affiliation(s)
- Lipiar K. M. O. Goni
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| | - Mohammad A. Jafar Mazumder
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| | - Divya B. Tripathy
- School of Basic and Applied Sciences, Galgotias University, Greater Noida 210310, India
| | - Mumtaz A. Quraishi
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| |
Collapse
|
21
|
Kostelansky F, Miletin M, Havlinova Z, Szotakova B, Libra A, Kucera R, Novakova V, Zimcik P. Thermal stabilisation of the short DNA duplexes by acridine-4-carboxamide derivatives. Nucleic Acids Res 2022; 50:10212-10229. [PMID: 36156152 PMCID: PMC9561273 DOI: 10.1093/nar/gkac777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/12/2022] Open
Abstract
The short oligodeoxynucleotide (ODN) probes are suitable for good discrimination of point mutations. However, the probes suffer from low melting temperatures. In this work, the strategy of using acridine-4-carboxamide intercalators to improve thermal stabilisation is investigated. The study of large series of acridines revealed that optimal stabilisation is achieved upon decoration of acridine by secondary carboxamide carrying sterically not demanding basic function bound through a two-carbon linker. Two highly active intercalators were attached to short probes (13 or 18 bases; designed as a part of HFE gene) by click chemistry into positions 7 and/or 13 and proved to increase the melting temperate (Tm) of the duplex by almost 8°C for the best combination. The acridines interact with both single- and double-stranded DNAs with substantially preferred interaction for the latter. The study of interaction suggested higher affinity of the acridines toward the GC- than AT-rich sequences. Good discrimination of two point mutations was shown in practical application with HFE gene (wild type, H63D C > G and S65C A > C mutations). Acridine itself can also serve as a fluorophore and also allows discrimination of the fully matched sequences from those with point mutations in probes labelled only with acridine.
Collapse
Affiliation(s)
| | - Miroslav Miletin
- Faculty of Pharmacy in Hradec Králové, Charles University, Ak. Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic
| | - Zuzana Havlinova
- Generi Biotech, Machkova 587, Hradec Kralove, 500 11, Czech Republic
| | - Barbora Szotakova
- Generi Biotech, Machkova 587, Hradec Kralove, 500 11, Czech Republic
| | - Antonin Libra
- Generi Biotech, Machkova 587, Hradec Kralove, 500 11, Czech Republic
| | - Radim Kucera
- Faculty of Pharmacy in Hradec Králové, Charles University, Ak. Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic
| | - Veronika Novakova
- Faculty of Pharmacy in Hradec Králové, Charles University, Ak. Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic
| | - Petr Zimcik
- To whom correspondence should be addressed. Tel: +420 495067257;
| |
Collapse
|
22
|
Synthesis and Evaluation of Antiproliferative Activity, Topoisomerase IIα Inhibition, DNA Binding and Non-Clinical Toxicity of New Acridine-Thiosemicarbazone Derivatives. Pharmaceuticals (Basel) 2022; 15:ph15091098. [PMID: 36145320 PMCID: PMC9506480 DOI: 10.3390/ph15091098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we report the synthesis of twenty new acridine–thiosemicarbazone derivatives and their antiproliferative activities. Mechanisms of action such as the inhibition of topoisomerase IIα and the interaction with DNA have been studied for some of the most active derivatives by means of both in silico and in vitro methods, and evaluations of the non-clinical toxicities (in vivo) in mice. In general, the compounds showed greater cytotoxicity against B16-F10 cells, with the highest potency for DL-08 (IC50 = 14.79 µM). Derivatives DL-01 (77%), DL-07 (74%) and DL-08 (79%) showed interesting inhibition of topoisomerase IIα when compared to amsacrine, at 100 µM. In silico studies proposed the way of bonding of these compounds and a possible stereoelectronic reason for the absence of enzymatic activity for CL-07 and DL-06. Interactions with DNA presented different spectroscopic effects and indicate that the compound CL-07 has higher affinity for DNA (Kb = 4.75 × 104 M−1; Ksv = 2.6 × 103 M−1). In addition, compounds selected for non-clinical toxicity testing did not show serious signs of toxicity at the dose of 2000 mg/kg in mice; cytotoxic tests performed on leukemic cells (K-562) and its resistant form (K-562 Lucena 1) identified moderate potency for DL-01 and DL-08, with IC50 between 11.45 and 17.32 µM.
Collapse
|
23
|
Gu Y, Yuan L, Li M, Wang X, Rao D, Bai X, Shi K, Xu H, Hou S, Yao H. Co-immobilized bienzyme of horseradish peroxidase and glucose oxidase on dopamine-modified cellulose-chitosan composite beads as a high-efficiency biocatalyst for degradation of acridine. RSC Adv 2022; 12:23006-23016. [PMID: 36105961 PMCID: PMC9379555 DOI: 10.1039/d2ra04091c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Co-immobilized bienzyme biocatalysts are attracting increasing interest in the field of wastewater treatment due to their widespread application. In this study, we successfully prepared a co-immobilized bienzyme biocatalyst by immobilizing horseradish peroxidase (HRP) and glucose oxidase (GOD) on dopamine (DA) modified cellulose (Ce)-chitosan (Cs) composite beads via covalent binding, designated as Ce-Cs@DA/HRP-GOD beads, and found that the bienzyme biocatalyst had a good ability to catalytically degrade acridine in wastewater. SEM, EPR, FTIR, and XRD were used to characterise the structure and properties of the Ce-Cs@DA/HRP-GOD beads. The co-immobilized bienzyme biocatalyst with a small amount of HRP exhibited better degradation efficiency for acridine (99.5%, 8 h) in simulated wastewater compared to the Ce-Cs@DA/HRP (93.8%, 8 h) and Ce-Cs@DA/GOD (15.8%, 8 h) beads alone. In addition, a reusability study showed that the co-immobilized bienzyme biocatalyst maintained a degradation rate of 61.2% after six cycles of acridine degradation. The good biodegradability and reusability of the biocatalyst might be due to the synergistic effect of bienzyme HRP-GOD, including the strong covalent bonding. Accordingly, the co-immobilized bienzyme biocatalyst based on the cascade reaction may pave the way for efficient and eco-friendly treatment of industrial wastewater.
Collapse
Affiliation(s)
- Yaohua Gu
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Lin Yuan
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Mingming Li
- Urology Surgery, General Hospital of Ningxia Medical University Yinchuan 750004 P. R. China
| | - Xinyu Wang
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Deyu Rao
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Xiaoyan Bai
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Keren Shi
- State Key Laboratory of High-efficiency Coal Utilization and Green Chemical Engineering, Ningxia University Yinchuan 750021 P. R. China
| | - Haiming Xu
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Shaozhang Hou
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| | - Huiqin Yao
- Key Laboratory of Environmental Factors and Chronic Disease Control, College of Public Health and Management, School of Basic Medicine, Ningxia Medical University Yinchuan 750004 P. R. China +86-951-698-0689
| |
Collapse
|
24
|
A comprehensive review on acridone based derivatives as future anti-cancer agents and their structure activity relationships. Eur J Med Chem 2022; 239:114527. [PMID: 35717872 DOI: 10.1016/j.ejmech.2022.114527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/03/2022]
Abstract
The development of drug resistance and severe side-effects has reduced the clinical efficacy of the existing anti-cancer drugs available in the market. Thus, there is always a constant need to develop newer anti-cancer drugs with minimal adverse effects. Researchers all over the world have been focusing on various alternative strategies to discover novel, potent, and target specific molecules for cancer therapy. In this direction, several heterocyclic compounds are being explored but amongst them one promising heterocycle is acridone which has attracted the attention of medicinal chemists and gained huge biological importance as acridones are found to act on different therapeutically proven molecular targets, overcome ABC transporters mediated drug resistance and DNA intercalation in cancer cells. Some of these acridone derivatives have reached clinical studies as these heterocycles have shown huge potential in cancer therapeutics and imaging. Here, the authors have attempted to compile and make some recommendations of acridone based derivatives concerning their cancer biological targets and in vitro-cytotoxicity based on drug design and novelty to increase their therapeutic potential. This review also provides some important insights on the design, receptor targeting and future directions for the development of acridones as possible clinically effective anti-cancer agents.
Collapse
|
25
|
Acridine Based N-Acylhydrazone Derivatives as Potential Anticancer Agents: Synthesis, Characterization and ctDNA/HSA Spectroscopic Binding Properties. Molecules 2022; 27:molecules27092883. [PMID: 35566236 PMCID: PMC9100673 DOI: 10.3390/molecules27092883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
A series of novel acridine N-acylhydrazone derivatives have been synthesized as potential topoisomerase I/II inhibitors, and their binding (calf thymus DNA—ctDNA and human serum albumin—HSA) and biological activities as potential anticancer agents on proliferation of A549 and CCD-18Co have been evaluated. The acridine-DNA complex 3b (-F) displayed the highest Kb value (Kb = 3.18 × 103 M−1). The HSA-derivatives interactions were studied by fluorescence quenching spectra. This method was used for the calculation of characteristic binding parameters. In the presence of warfarin, the binding constant values were found to decrease (KSV = 2.26 M−1, Kb = 2.54 M−1), suggesting that derivative 3a could bind to HSA at Sudlow site I. The effect of tested derivatives on metabolic activity of A549 cells evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide or MTT assay decreased as follows 3b(-F) > 3a(-H) > 3c(-Cl) > 3d(-Br). The derivatives 3c and 3d in vitro act as potential dual inhibitors of hTopo I and II with a partial effect on the metabolic activity of cancer cells A594. The acridine-benzohydrazides 3a and 3c reduced the clonogenic ability of A549 cells by 72% or 74%, respectively. The general results of the study suggest that the novel compounds show potential for future development as anticancer agents.
Collapse
|
26
|
Synthesis of novel 1,2,3-triazole based acridine and benzothiazole scaffold N-glycosides with anti-proliferative activity, docking studies, and comparative computational studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131941] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
27
|
9-Aminoacridine Inhibits Ribosome Biogenesis by Targeting Both Transcription and Processing of Ribosomal RNA. Int J Mol Sci 2022; 23:ijms23031260. [PMID: 35163183 PMCID: PMC8836032 DOI: 10.3390/ijms23031260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Aminoacridines, used for decades as antiseptic and antiparasitic agents, are prospective candidates for therapeutic repurposing and new drug development. Although the mechanisms behind their biological effects are not fully elucidated, they are most often attributed to the acridines’ ability to intercalate into DNA. Here, we characterized the effects of 9-aminoacridine (9AA) on pre-rRNA metabolism in cultured mammalian cells. Our results demonstrate that 9AA inhibits both transcription of the ribosomal RNA precursors (pre-rRNA) and processing of the already synthesized pre-rRNAs, thereby rapidly abolishing ribosome biogenesis. Using a fluorescent intercalator displacement assay, we further show that 9AA can bind to RNA in vitro, which likely contributes to its ability to inhibit post-transcriptional steps in pre-rRNA maturation. These findings extend the arsenal of small-molecule compounds that can be used to block ribosome biogenesis in mammalian cells and have implications for the pharmacological development of new ribosome biogenesis inhibitors.
Collapse
|
28
|
Majhi S. Recent developments in the synthesis and anti-cancer activity of acridine and xanthine-based molecules. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2021-0216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Abstract
Cancer is the uncontrolled growth and development of abnormal cells which is a major cause of death in both advanced and emerging countries. Although currently chemotherapy is most broadly used among an extensive range of anti-cancer therapies, it includes many demerits, such as highly toxic, side-effects, expensive and partial lack of targeting specificity. So the design and synthesis of new molecules that perform specifically on target proteins in tumor cells is a focus of contemporary research. So many researchers aim for new drugs that will be more efficient, more selective, and less toxic. Because of the interesting structures and significant biological profile, naturally occurring acridines and xanthines as well as their analogues have attracted considerable interest in researchers and technologists. Natural and synthetic acridine derivatives form a significant category of heterocycles having nitrogen that is of considerable interest for organic chemists and biological communities due to their attractive anti-cancer activity. Another important class of therapeutic agents with diverse biological properties including cytotoxic effects is xanthine derivatives which are collectively called xanthines (a group of alkaloids). Among many significant molecules based on the structure of the purine, there is a group of natural xanthines, involving theobromine, caffeine, and theophylline and analogues of xanthine display anti-cancer activity. Hence the present chapter wishes to concentrate the attention on the synthesis and anti-cancer activity of acridine and xanthine-based compounds brilliantly.
Collapse
Affiliation(s)
- Sasadhar Majhi
- Department of Chemistry (UG & PG Dept.) , Triveni Devi Bhalotia College, Kazi Nazrul University , Raniganj , West Bengal 713347 , India
| |
Collapse
|
29
|
Kim M, Ha JH, Choi J, Kim BR, Gapsys V, Lee KO, Jee JG, Chakrabarti KS, de Groot BL, Griesinger C, Ryu KS, Lee D. Repositioning Food and Drug Administration-Approved Drugs for Inhibiting Biliverdin IXβ Reductase B as a Novel Thrombocytopenia Therapeutic Target. J Med Chem 2021; 65:2548-2557. [PMID: 34957824 DOI: 10.1021/acs.jmedchem.1c01664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Biliverdin IXβ reductase B (BLVRB) has recently been proposed as a novel therapeutic target for thrombocytopenia through its reactive oxygen species (ROS)-associated mechanism. Thus, we aim at repurposing drugs as new inhibitors of BLVRB. Based on IC50 (<5 μM), we have identified 20 compounds out of 1496 compounds from the Food and Drug Administration (FDA)-approved library and have clearly mapped their binding sites to the active site. Furthermore, we show the detailed BLVRB-binding modes and thermodynamic properties (ΔH, ΔS, and KD) with nuclear magnetic resonance (NMR) and isothermal titration calorimetry together with complex structures of eight water-soluble compounds. We anticipate that the results will serve as a novel platform for further in-depth studies on BLVRB effects for related functions such as ROS accumulation and megakaryocyte differentiation, and ultimately treatments of platelet disorders.
Collapse
Affiliation(s)
- Myeongkyu Kim
- Protein Structure Research Team, Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si, Chungcheongbuk-Do 28119, South Korea.,Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Jung-Hye Ha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation(DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Joonhyeok Choi
- Protein Structure Research Team, Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si, Chungcheongbuk-Do 28119, South Korea
| | - Bo-Ram Kim
- Protein Structure Research Team, Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si, Chungcheongbuk-Do 28119, South Korea
| | - Vytautas Gapsys
- Department of Theoretical and Computational Biophysics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Ko On Lee
- Protein Structure Research Team, Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si, Chungcheongbuk-Do 28119, South Korea
| | - Jun-Goo Jee
- Research Institute of Pharmaceutical Sciences College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu 41566, South Korea
| | | | - Bert L de Groot
- Department of Theoretical and Computational Biophysics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Christian Griesinger
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Kyoung-Seok Ryu
- Protein Structure Research Team, Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si, Chungcheongbuk-Do 28119, South Korea
| | - Donghan Lee
- Department of Medicine, James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock Street, Louisville, Kentucky 40202, United States
| |
Collapse
|
30
|
Li Q, Moussallem C, Castet F, Muccioli L, Dourges MA, Toupance T, Nicolas Y. Direct Triple Annulations: A Way to Design Large Triazastarphenes with Intertwined Hexagonal Packing. Org Lett 2021; 24:344-348. [PMID: 34908427 DOI: 10.1021/acs.orglett.1c04001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A new straightforward synthetic strategy has been elaborated to achieve star-shaped triazatrinaphthylene and, for the first time, triazatrianthrylene derivatives. Their solution- and solid-state properties were thoroughly characterized by cyclic voltammetry, UV-vis absorption spectroscopy, X-ray diffraction, and density functional theory calculations. Original hexagonal molecular arrangements were found in the crystal phase, which opens a new pathway for designing materials with improved three-dimensional charge-transport properties.
Collapse
Affiliation(s)
- Qian Li
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, 351 Cours de la Libération, F-33405 Cedex Talence, France
| | - Chady Moussallem
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, 351 Cours de la Libération, F-33405 Cedex Talence, France.,Université Libanaise, Faculté des Sciences, Laboratoire de Chimie, Campus Michael Slayman, 1352 Rasmaska, Lebanon
| | - Frédéric Castet
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, 351 Cours de la Libération, F-33405 Cedex Talence, France
| | - Luca Muccioli
- Department of Industrial Chemistry "Toso Montanari″, University of Bologna, Viale Risorgimento 4, 40136 Bologna, Italy
| | - Marie-Anne Dourges
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, 351 Cours de la Libération, F-33405 Cedex Talence, France
| | - Thierry Toupance
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, 351 Cours de la Libération, F-33405 Cedex Talence, France
| | - Yohann Nicolas
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, 351 Cours de la Libération, F-33405 Cedex Talence, France
| |
Collapse
|
31
|
Rząd K, Paluszkiewicz E, Neubauer D, Olszewski M, Kozłowska-Tylingo K, Kamysz W, Gabriel I. The Effect of Conjugation with Octaarginine, a Cell-Penetrating Peptide on Antifungal Activity of Imidazoacridinone Derivative. Int J Mol Sci 2021; 22:ijms222413190. [PMID: 34947987 PMCID: PMC8705783 DOI: 10.3390/ijms222413190] [Citation(s) in RCA: 5] [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: 11/02/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/31/2022] Open
Abstract
Acridine cell-penetrating peptide conjugates are an extremely important family of compounds in antitumor chemotherapy. These conjugates are not so widely analysed in antimicrobial therapy, although bioactive peptides could be used as nanocarriers to smuggle antimicrobial compounds. An octaarginine conjugate of an imidazoacridinone derivative (Compound 1-R8) synthetized by us exhibited high antifungal activity against reference and fluconazole-resistant clinical strains (MICs ≤ 4 μg mL−1). Our results clearly demonstrate the qualitative difference in accumulation of the mother compound and Compound 1-R8 conjugate into fungal cells. Only the latter was transported and accumulated effectively. Microscopic and flow cytometry analysis provide some evidence that the killing activity of Compound 1-R8 may be associated with a change in the permeability of the fungal cell membrane. The conjugate exhibited low cytotoxicity against human embryonic kidney (HEK-293) and human liver (HEPG2) cancer cell lines. Nevertheless, the selectivity index value of the conjugate for human pathogenic strains remained favourable and no hemolytic activity was observed. The inhibitory effect of the analysed compound on yeast topoisomerase II activity suggested its molecular target. In summary, conjugation with R8 effectively increased imidazoacridinone derivative ability to enter the fungal cell and achieve a concentration inside the cell that resulted in a high antifungal effect.
Collapse
Affiliation(s)
- Kamila Rząd
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland; (K.R.); (E.P.); (M.O.); (K.K.-T.)
| | - Ewa Paluszkiewicz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland; (K.R.); (E.P.); (M.O.); (K.K.-T.)
| | - Damian Neubauer
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Gen. J. Hallera 107th Avenue, 80-416 Gdańsk, Poland; (D.N.); (W.K.)
| | - Mateusz Olszewski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland; (K.R.); (E.P.); (M.O.); (K.K.-T.)
| | - Katarzyna Kozłowska-Tylingo
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland; (K.R.); (E.P.); (M.O.); (K.K.-T.)
| | - Wojciech Kamysz
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Gen. J. Hallera 107th Avenue, 80-416 Gdańsk, Poland; (D.N.); (W.K.)
| | - Iwona Gabriel
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland; (K.R.); (E.P.); (M.O.); (K.K.-T.)
- Correspondence: ; Tel.: +48-58-348-6078; Fax: +48-58-347-1144
| |
Collapse
|
32
|
Li ZY, Xu GS, Song YL, Li X. Structural optimizations and bioevaluation of N-substituted acridone derivatives as strong topoisomerase II inhibitors. Bioorg Chem 2021; 119:105543. [PMID: 34929515 DOI: 10.1016/j.bioorg.2021.105543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/28/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022]
Abstract
Previously, an array of N-substituted acridone derivatives have been reported as potent topoisomerase II (topo II) inhibitors, and preliminary structure-activity relationship (SAR) outcomes revealed that the linker between 1-NH and N-methyl piperazine motif of the tricyclic acridone scaffold significantly affected their anti-proliferative potencies. To further explore the SARs of acridone-derived topo II inhibitors, a wider range of novel acridone derivatives were herein synthesized via two rounds of structural optimizations on two validated hits, E17 and E24. Initially, the linker length was optimized, and then influences of N-methyl piperazinyl moiety and disposition of three N atoms on the bioactivity were investigated. As a result, a newly developed topo II inhibitor 6 h was found to be more potent than E17 and E24, thereby serving as a tool compound for the follow-up mechanistic study. Compound 6 h functioned as a strong topo IIα/β inhibitor, caused obvious DNA damage, and induced apoptosis by triggering the loss of mitochondrial membrane potential (Δψm). Further molecular docking and MD study illustrated the favorable interactions of 6 h with both topo IIα and topo IIβ subtypes.
Collapse
Affiliation(s)
- Zhi-Ying Li
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, China
| | - Guang-Sen Xu
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, China
| | - Yu-Liang Song
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, China
| | - Xun Li
- Institute of Materia Medica, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong 250117, China; Key Laboratory of Forensic Toxicology, Ministry of Public Security, Beijing 100192, China.
| |
Collapse
|
33
|
Suganya S, Saravanan K, Jaganathan R, Kumaradhas P. Salt formation, hydrogen-bonding patterns and supramolecular architectures of acridine with salicylic and hippuric acid molecules. Acta Crystallogr C 2021; 77:790-799. [PMID: 34864722 DOI: 10.1107/s2053229621011189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/25/2021] [Indexed: 11/10/2022] Open
Abstract
The intermolecular interactions and salt formation of acridine with 4-aminosalicylic acid, 5-chlorosalicylic acid and hippuric acid were investigated. The salts obtained were acridin-1-ium 4-aminosalicylate (4-amino-2-hydroxybenzoate), C13H10N+·C7H6NO3- (I), acridin-1-ium 5-chlorosalicylate (5-chloro-2-hydroxybenzoate), C13H10N+·C7H4ClO3- (II), and acridin-1-ium hippurate (2-benzamidoacetate) monohydrate, C13H10N+·C9H8NO3-·H2O (III). Acridine is involved in strong intermolecular interactions with the hydroxy group of the three acids, enabling it to form supramolecular assemblies. Hirshfeld surfaces, fingerprint plots and enrichment ratios were generated and investigated, and the intermolecular interactions were analyzed, revealing their quantitative contributions in the crystal packing of salts I, II and III. A quantum theory of atoms in molecules (QTAIM) analysis shows the charge-density distribution of the intermolecular interactions. The isosurfaces of the noncovalent interactions were studied, which allows visualization of where the hydrogen-bonding and dispersion interactions contribute within the crystal.
Collapse
Affiliation(s)
- Suresh Suganya
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem 636 011, India
| | | | - Ramakrishnan Jaganathan
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem 636 011, India
| | - Poomani Kumaradhas
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem 636 011, India
| |
Collapse
|
34
|
Tayde DT, Navgire ME, Lande MK. One Pot Three Component Synthesis of Substituted 3,4,6,7-Tetrahydro-3,3,6,6-Tetramethyl-9,10-Diphenylacridine-1,8(2H,5H,9H,10H)-Diones Catalyzed by Mesostrucrted In 2O 3-SiO 2. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.2006249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Deepak T. Tayde
- Department of Chemistry, M.J.M Arts, Commerce & Science College, Karanajali, India
| | - Madhukar E. Navgire
- Department of Chemistry, Jijamata College of Science and Arts, Bhende, India
| | - Machhindra K. Lande
- Department of Chemistry, Dr. Babasaheb Ambedkar, Marathwada University, Aurangabad, India
| |
Collapse
|
35
|
Electrochemical DNA Sensor Based on Acridine Yellow Adsorbed on Glassy Carbon Electrode. SENSORS 2021; 21:s21227763. [PMID: 34833839 PMCID: PMC8621912 DOI: 10.3390/s21227763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Electrochemical DNA sensors offer unique opportunities for the sensitive detection of specific DNA interactions. In this work, a voltametric DNA sensor is proposed on the base of glassy carbon electrode modified with carbon black, adsorbed acridine yellow and DNA for highly sensitive determination of doxorubicin antitumor drug. The signal recorded by cyclic voltammetry was attributed to irreversible oxidation of the dye. Its value was altered by aggregation of the hydrophobic dye molecules on the carbon black particles. DNA molecules promote disaggregation of the dye and increased the signal. This effect was partially suppressed by doxorubicin compensate for the charge of DNA in the intercalation. Sensitivity of the signal toward DNA and doxorubicin was additionally increased by treatment of the layer with dimethylformamide. In optimal conditions, the linear range of doxorubicin concentrations determined was 0.1 pM–1.0 nM, and the detection limit was 0.07 pM. No influence of sulfonamide medicines and plasma electrolytes on the doxorubicin determination was shown. The DNA sensor was tested on two medications (doxorubicin-TEVA and doxorubicin-LANS) and showed recoveries of 102–105%. The DNA sensor developed can find applications in the determination of drug residues in blood and for the pharmacokinetics studies.
Collapse
|
36
|
Li ZY, Xu GS, Li X. A Unique Topoisomerase II Inhibitor with Dose-Affected Anticancer Mechanisms and Less Cardiotoxicity. Cells 2021; 10:cells10113138. [PMID: 34831359 PMCID: PMC8621772 DOI: 10.3390/cells10113138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Type II DNA topoisomerase (topo II) is an essential nuclear enzyme and a well-validated anticancer drug target. Previously, we have carried out several rounds of structural optimizations on our in-house topo II inhibitor E17, which was shown to have superior anticancer activity and less risk of multidrug resistance (MDR). Among the newly developed acridone derivatives, 6h displayed significant anticancer efficacy with unique mechanisms of action. At low concentrations, it arrested cancer cell cycles and triggered cell apoptosis, which is similar to the action of the well-known topo II inhibitor VP16. By contrast, 6h showed significant and long-term anti-proliferative activity at relatively high concentrations, with negligible influence on apoptosis. In addition, 6h exhibited no serious cardiotoxicity compared to doxorubicin (DOXO), a widely used topo II-targeting antineoplastic drug in clinic, but with damaging myocardial side effects. Collectively, our present work has supported the therapeutic value of 6h as a promising chemotherapy for cancers.
Collapse
Affiliation(s)
- Zhi-Ying Li
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Z.-Y.L.); (G.-S.X.)
| | - Guang-Sen Xu
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Z.-Y.L.); (G.-S.X.)
| | - Xun Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
- Key Laboratory of Forensic Toxicology, Ministry of Public Security, Beijing 100192, China
- Correspondence:
| |
Collapse
|
37
|
Franco Pinto J, Fillion A, Duchambon P, Bombard S, Granzhan A. Acridine-O 6-benzylguanine hybrids: Synthesis, DNA binding, MGMT inhibition and antiproliferative activity. Eur J Med Chem 2021; 227:113909. [PMID: 34731767 DOI: 10.1016/j.ejmech.2021.113909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/24/2021] [Accepted: 10/06/2021] [Indexed: 12/13/2022]
Abstract
O6-Methylguanine-DNA-methyltransferase (MGMT) is a key DNA repair enzyme involved in chemoresistance to DNA-alkylating anti-cancer drugs such as Temozolomide (TMZ) through direct repair of drug-induced O6-methylguanine residues in DNA. MGMT substrate analogues, such as O6-benzylguanine (BG), efficiently inactivate MGMT in vitro and in cells; however, these drugs failed to reach the clinic due to adverse side effects. Here, we designed hybrid drugs combining a BG residue covalently linked to a DNA-interacting moiety (6-chloro-2-methoxy-9-aminoacridine). Specifically, two series of hybrids, encompassing three compounds each, were obtained by varying the position of the attachment point of BG (N9 of guanine vs. the benzyl group) and the length and nature of the linker. UV/vis absorption and fluorescence data indicate that all six hybrids adopt an intramolecularly stacked conformation in aqueous solutions in a wide range of temperatures. All hybrids interact with double-stranded DNA, as clearly evidenced by spectrophotometric titrations, without intercalation of the acridine ring and do not induce thermal stabilization of the duplex. All hybrids, as well as the reference DNA intercalator (6-chloro-2-methoxy-9-aminoacridine 8), irreversibly inhibit MGMT in vitro with variable efficiency, comparable to that of BG. In a multidrug-resistant glioblastoma cell line T98G, benzyl-linked hybrids 7a-c and the N9-linked hybrid 19b are moderately cytotoxic (GI50 ≥ 15 μM after 96 h), while N9-linked hybrids 19a and 19c are strongly cytotoxic (GI50 = 1-2 μM), similarly to acridine 8 (GI50 = 0.6 μM). Among all compounds, hybrids 19a and 19c, similarly to BG, display synergic cytotoxic effect upon co-treatment with subtoxic doses of TMZ, with combination index (CI) values as low as 0.2-0.3. In agreement with in vitro results, compound 19a inactivates cellular MGMT but, unlike BG, does not induce significant levels of DNA damage, either alone or in combination with TMZ, as indicated by the results of γH2AX immunostaining experiments. Instead, and unlike BG, compound 19a alone induces significant apoptosis of T98G cells, which is not further increased in a combination with TMZ. These results indicate that molecular mechanisms underlying the cytotoxicity of 19a and its combination with TMZ are distinct from that of BG. The strongly synergic properties of this combination represent an interesting therapeutic opportunity in treating TMZ-resistant cancers.
Collapse
Affiliation(s)
- Jaime Franco Pinto
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405, Orsay, France; CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405, Orsay, France
| | - Alexandra Fillion
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405, Orsay, France; CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405, Orsay, France
| | - Patricia Duchambon
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405, Orsay, France; CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405, Orsay, France
| | - Sophie Bombard
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405, Orsay, France; CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405, Orsay, France.
| | - Anton Granzhan
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405, Orsay, France; CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405, Orsay, France.
| |
Collapse
|
38
|
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]
|
39
|
Anokhina VS, Miller BL. Targeting Ribosomal Frameshifting as an Antiviral Strategy: From HIV-1 to SARS-CoV-2. Acc Chem Res 2021; 54:3349-3361. [PMID: 34403258 DOI: 10.1021/acs.accounts.1c00316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Treatment of HIV-1 has largely involved targeting viral enzymes using a cocktail of inhibitors. However, resistance to these inhibitors and toxicity in the long term have pushed the field to identify new therapeutic targets. To that end, -1 programmed ribosomal frameshifting (-1 PRF) has gained attention as a potential node for therapeutic intervention. In this process, a ribosome moves one nucleotide backward in the course of translating a mRNA, revealing a new reading frame for protein synthesis. In HIV-1, -1 PRF allows the virus to regulate the ratios of enzymatic and structural proteins as needed for correct viral particle assembly. Two RNA structural elements are central to -1 PRF in HIV: a slippery sequence and a highly conserved stable hairpin called the HIV-1 frameshifting stimulatory signal (FSS). Dysregulation of -1 PRF is deleterious for the virus. Thus, -1 PRF is an attractive target for new antiviral development. It is important to note that HIV-1 is not the only virus exploiting -1 PRF for regulating production of its proteins. Coronaviruses, including the COVID-19 pandemic virus SARS-CoV-2, also rely on -1 PRF. In SARS-CoV-2 and other coronaviruses, -1 PRF is required for synthesis of RNA-dependent RNA polymerase and several other nonstructural proteins. Coronaviruses employ a more complex RNA structural element for regulating -1 PRF called a pseudoknot. The purpose of this Account is primarily to review the development of molecules targeting HIV-1 -1 PRF. These approaches are case studies illustrating how the entire pipeline from screening to the generation of high-affinity leads might be implemented. We consider both target-based and function-based screening, with a particular focus on our group's approach beginning with a resin-bound dynamic combinatorial library (RBDCL) screen. We then used rational design approaches to optimize binding affinity, selectivity, and cellular bioavailability. Our tactic is, to the best of our knowledge, the only study resulting in compounds that bind specifically to the HIV-1 FSS RNA and reduce infectivity of laboratory and drug-resistant strains of HIV-1 in human cells. Lessons learned from strategies targeting -1 PRF HIV-1 might provide solutions in the development of antivirals in areas of unmet medical need. This includes the development of new frameshift-altering therapies for SARS-CoV-2, approaches to which are very recently beginning to appear.
Collapse
Affiliation(s)
- Viktoriya S. Anokhina
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, New York 14642, United States
| | - Benjamin L. Miller
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, New York 14642, United States
- Department of Dermatology, University of Rochester, Rochester, New York 14642, United States
| |
Collapse
|
40
|
Lauria A, La Monica G, Bono A, Martorana A. Quinoline anticancer agents active on DNA and DNA-interacting proteins: From classical to emerging therapeutic targets. Eur J Med Chem 2021; 220:113555. [PMID: 34052677 DOI: 10.1016/j.ejmech.2021.113555] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/25/2022]
Abstract
Quinoline is one of the most important and versatile nitrogen heterocycles embodied in several biologically active molecules. Within the numerous quinolines developed as antiproliferative agents, this review is focused on compounds interfering with DNA structure or with proteins/enzymes involved in the regulation of double helix functional processes. In this light, a special focus is given to the quinoline compounds, acting with classical/well-known mechanisms of action (DNA intercalators or Topoisomerase inhibitors). In particular, the quinoline drugs amsacrine and camptothecin (CPT) have been studied as key lead compounds for the development of new agents with improved PK and tolerability properties. Moreover, notable attention has been paid to the quinoline molecules, which are able to interfere with emerging targets involved in cancer progression, as G-quadruplexes or the epigenetic ones (e.g.: histone deacetylase, DNA and histones methyltransferase). The antiproliferative and the enzymatic inhibition data of the reviewed compounds have been analyzed. Furthermore, concerning the SAR (structure-activity relationship) aspects, the most recurrent ligand-protein interactions are summarized, underling the structural requirements for each kind of mechanism of action.
Collapse
Affiliation(s)
- Antonino Lauria
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Gabriele La Monica
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Alessia Bono
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Annamaria Martorana
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy.
| |
Collapse
|
41
|
Antifungal Activity of Capridine β as a Consequence of Its Biotransformation into Metabolite Affecting Yeast Topoisomerase II Activity. Pathogens 2021; 10:pathogens10020189. [PMID: 33572407 PMCID: PMC7916213 DOI: 10.3390/pathogens10020189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/21/2022] Open
Abstract
In the last few years, increasing importance is attached to problems caused by fungal pathogens. Current methods of preventing fungal infections remain unsatisfactory. There are several antifungal compounds which are highly effective in some cases, however, they have limitations in usage: Nephrotoxicity and other adverse effects. In addition, the frequent use of available fungistatic drugs promotes drug resistance. Therefore, there is an urgent need for the development of a novel antifungal drug with a different mechanism of action, blocking of the fungal DNA topoisomerases activity appear to be a promising idea. According to previous studies on the m-AMSA moderate inhibitory effect on fungal topoisomerase II, we have decided to study Capridine β (also acridine derivative) antifungal activity, as well as its inhibitory potential on yeast topoisomerase II (yTOPOII). Results indicated that Capridine β antifungal activity depends on the kind of strains analyzed (MICs range 0.5–64 μg mL−1) and is related to its biotransformation in the cells. An investigation of metabolite formation, identified as Capridine β reduction product (IE1) by the fungus Candida albicans was performed. IE1 exhibited no activity against fungal cells due to an inability to enter the cells. Although no antifungal activity was observed, in contrast to Capridine β, biotransformation metabolite totally inhibited the yTOPOII-mediated relaxation at concentrations lower than detected for m-AMSA. The closely related Capridine β only slightly diminished the catalytic activity of yTOPOII.
Collapse
|
42
|
Biological perspective of a triazine derivative with isatin/chalcone/acridone: DFT and docking investigations. Struct Chem 2021. [DOI: 10.1007/s11224-020-01609-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
43
|
Bortolozzi R, Ihmels H, Schulte R, Stremmel C, Viola G. Synthesis, DNA-binding and antiproliferative properties of diarylquinolizinium derivatives. Org Biomol Chem 2021; 19:878-890. [PMID: 33410854 DOI: 10.1039/d0ob02298e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A series of ten 2,7- and 2,8-diarylquinolizinium derivatives was synthesized and their DNA-binding and cytotoxic properties were investigated. Except for one nitro-substituted derivative all tested diarylquinolizinium ions bind to DNA with sufficient affinity (2 × 104 M-1-2 × 105 M-1). It was shown with photometric, fluorimetric and polarimetric titrations as well as with flow-LD analysis that the ligands bind mainly by intercalation to duplex DNA, however, depending on the ligand-DNA ratio, groove binding and backbone association were also observed with some derivatives. The biological activity was further investigated with tests of cytotoxicity and antiproliferative properties towards non-tumor cells and selected cancer cells, along with cell cycle analysis and an annexin-V assay. Notably, substrates that carry donor-functionalities in the 4-position of the phenyl substituents revealed a strong, and in some cases selective, antiproliferative activity as quantified by the growth inhibition, GI50, at very low micromolar and even submicromolar level both in leukemia and solid tumors.
Collapse
Affiliation(s)
- Roberta Bortolozzi
- Department of Women's and Child's health, Oncohematology laboratory, University of Padova, Via Giustiniani 2, I-35128 Padova, Italy. giampietro,
| | - Heiko Ihmels
- Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Str. 2, 57068 Siegen, Germany.
| | - Robin Schulte
- Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Str. 2, 57068 Siegen, Germany.
| | - Christopher Stremmel
- Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Str. 2, 57068 Siegen, Germany.
| | - Giampietro Viola
- Department of Women's and Child's health, Oncohematology laboratory, University of Padova, Via Giustiniani 2, I-35128 Padova, Italy. giampietro,
| |
Collapse
|
44
|
Fonte M, Tassi N, Gomes P, Teixeira C. Acridine-Based Antimalarials-From the Very First Synthetic Antimalarial to Recent Developments. Molecules 2021; 26:molecules26030600. [PMID: 33498868 PMCID: PMC7865557 DOI: 10.3390/molecules26030600] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
Malaria is among the deadliest infectious diseases in the world caused by Plasmodium parasites. Due to the high complexity of the parasite’s life cycle, we partly depend on antimalarial drugs to fight this disease. However, the emergence of resistance, mainly by Plasmodium falciparum, has dethroned most of the antimalarials developed to date. Given recent reports of resistance to artemisinin combination therapies, first-line treatment currently recommended by the World Health Organization, in Western Cambodia and across the Greater Mekong sub-region, it seems very likely that artemisinin and its derivatives will follow the same path of other antimalarial drugs. Consequently, novel, safe and efficient antimalarial drugs are urgently needed. One fast and low-cost strategy to accelerate antimalarial development is by recycling classical pharmacophores. Quinacrine, an acridine-based compound and the first clinically tested synthetic antimalarial drug with potent blood schizonticide but serious side effects, has attracted attention due to its broad spectrum of biological activity. In this sense, the present review will focus on efforts made in the last 20 years for the development of more efficient, safer and affordable antimalarial compounds, through recycling the classical quinacrine drug.
Collapse
|
45
|
Rząd K, Paluszkiewicz E, Gabriel I. A new 1-nitro-9-aminoacridine derivative targeting yeast topoisomerase II able to overcome fluconazole-resistance. Bioorg Med Chem Lett 2021; 35:127815. [PMID: 33486051 DOI: 10.1016/j.bmcl.2021.127815] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/05/2021] [Accepted: 01/17/2021] [Indexed: 02/02/2023]
Abstract
Fungal resistance remains a significant threat and a leading cause of death worldwide. Thus, overcoming microbial infections have again become a serious clinical problem. Although acridine derivatives are widely analyzed as anticancer agents, only a few reports have demonstrated their antifungal activity. In an effort to develop biologically active antifungals, twelve novel C-857 (9-(2'-hydroxyethylamino)-1-nitroacridine) and C-1748 (9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine) derivatives were synthesized. The evaluation of biological properties suggests that starting compounds: C-1748, C-857 and IE3 (2-[(4-methyl-1-nitroacridin-9-yl)amino]ethyl lysinate), IE4 (2-[(1-nitroacridin-9-yl)amino]ethyl lysinate) antifungal mode of action differ from that determined for IE5 (N'-{3-[(4-methyl-1-nitroacridin-9-yl)amino]propyl}lysinamide), IE6 (N'-{3-[(1-nitroacridin-9-yl)amino]propyl}lysinamide) and IE10 (3,3'-Bis-(1-nitroacridin-9-ylamino)-aminoethylaminoethylaminoethylamine). Although MIC values determined for the latter were higher, in contrast to C-857 and C-1748, newly synthesized IE5, IE6 and IE10 reduced C. albicans hyphal growth in different inducing media. Those compounds also exhibited antibiofilm activity, whereas IE10 was the most effective. Moreover, only IE6 exhibited antifungal activity against fluconazole resistant C. albicans strains with MICs values in the range of 16-64 μg mL-1. Our results also indicate that, in contrast to other analyzed derivatives, novel synthetized compounds IE6 and IE10 with antifungal activity target yeast topoisomerase II activity.
Collapse
Affiliation(s)
- Kamila Rząd
- Department of Pharmaceutical Technology and Biochemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland
| | - Ewa Paluszkiewicz
- Department of Pharmaceutical Technology and Biochemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland
| | - Iwona Gabriel
- Department of Pharmaceutical Technology and Biochemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., 80-233 Gdańsk, Poland.
| |
Collapse
|
46
|
Binas O, de Jesus V, Landgraf T, Völklein AE, Martins J, Hymon D, Kaur Bains J, Berg H, Biedenbänder T, Fürtig B, Lakshmi Gande S, Niesteruk A, Oxenfarth A, Shahin Qureshi N, Schamber T, Schnieders R, Tröster A, Wacker A, Wirmer‐Bartoschek J, Wirtz Martin MA, Stirnal E, Azzaoui K, Richter C, Sreeramulu S, José Blommers MJ, Schwalbe H. 19 F NMR-Based Fragment Screening for 14 Different Biologically Active RNAs and 10 DNA and Protein Counter-Screens. Chembiochem 2021; 22:423-433. [PMID: 32794266 PMCID: PMC7436455 DOI: 10.1002/cbic.202000476] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/11/2020] [Indexed: 11/17/2022]
Abstract
We report here the nuclear magnetic resonance 19 F screening of 14 RNA targets with different secondary and tertiary structure to systematically assess the druggability of RNAs. Our RNA targets include representative bacterial riboswitches that naturally bind with nanomolar affinity and high specificity to cellular metabolites of low molecular weight. Based on counter-screens against five DNAs and five proteins, we can show that RNA can be specifically targeted. To demonstrate the quality of the initial fragment library that has been designed for easy follow-up chemistry, we further show how to increase binding affinity from an initial fragment hit by chemistry that links the identified fragment to the intercalator acridine. Thus, we achieve low-micromolar binding affinity without losing binding specificity between two different terminator structures.
Collapse
Affiliation(s)
- Oliver Binas
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Vanessa de Jesus
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Tom Landgraf
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Albrecht Eduard Völklein
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Jason Martins
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Daniel Hymon
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Jasleen Kaur Bains
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Hannes Berg
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Thomas Biedenbänder
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Boris Fürtig
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Santosh Lakshmi Gande
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Anna Niesteruk
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Andreas Oxenfarth
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Nusrat Shahin Qureshi
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Tatjana Schamber
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Robbin Schnieders
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Alix Tröster
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Anna Wacker
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Julia Wirmer‐Bartoschek
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Maria Alexandra Wirtz Martin
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Elke Stirnal
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Kamal Azzaoui
- Saverna TherapeuticsGewerbestrasse 244123AllschwilSwitzerland
| | - Christian Richter
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | - Sridhar Sreeramulu
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| | | | - Harald Schwalbe
- Institute for Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Strasse 760438Frankfurt am MainGermany
| |
Collapse
|
47
|
Bortolami M, Rocco D, Messore A, Di Santo R, Costi R, Madia VN, Scipione L, Pandolfi F. Acetylcholinesterase inhibitors for the treatment of Alzheimer's disease - a patent review (2016-present). Expert Opin Ther Pat 2021; 31:399-420. [PMID: 33428491 DOI: 10.1080/13543776.2021.1874344] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction - AD, the most common form of dementia, has a multifactorial etiology, and the current therapy (AChEIs and memantine) is unable to interrupt its progress and fatal outcome. This is reflected in the research programs that are oriented toward the development of new therapeutics able to operate on multiple targets involved in the disease progression.Areas covered - The patents from 2016 to present regarding the use of AChEIs in AD, concerns the development of new AChEIs, multitarget or multifunctional ligands, or the associations of currently used AChEIs with other compounds acting on different targets involved in the AD.Expert opinion - The development of new multitarget AChEIs promises to identify compounds with great therapeutic potential but requires more time and effort in order to obtain drugs with the optimal pharmacodynamic profile. Otherwise, the research on new combinations of existing drugs, with known pharmacodynamic and ADME profile, could shorten the time and reduce the costs to develop a new therapeutic treatment for AD. From the analyzed data, it seems more likely that a response to the urgent need to develop effective treatments for AD therapy could come more quickly from studies on drug combinations than from the development of new AChEIs.
Collapse
Affiliation(s)
- Martina Bortolami
- Department of Scienze Di Base E Applicate per l'Ingegneria, Sapienza University of Rome, Rome, Italy
| | - Daniele Rocco
- Department of Scienze Di Base E Applicate per l'Ingegneria, Sapienza University of Rome, Rome, Italy
| | - Antonella Messore
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Roberto Di Santo
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Roberta Costi
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Valentina Noemi Madia
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Luigi Scipione
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabiana Pandolfi
- Department of Scienze Di Base E Applicate per l'Ingegneria, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
48
|
Chen JN, Wu XK, Lu CH, Li X. Structure-activity relationship of novel acridone derivatives as antiproliferative agents. Bioorg Med Chem 2021; 29:115868. [PMID: 33191085 DOI: 10.1016/j.bmc.2020.115868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/12/2020] [Accepted: 11/01/2020] [Indexed: 10/23/2022]
Abstract
Unlike other DNA topoisomerase II (topo II) inhibitors, our recently identified acridone derivative E17 exerted strong cytotoxic activity by inhibiting topo II without causing topo II degradation and DNA damage, which promoted us to explore more analogues of E17 by expanding its chemical diversification and enrich the structure-activity relationship (SAR) outcomes of acridone-oriented chemotypes. To achieve this goal, 42 novel acridone derivatives were synthesized and evaluated for their antiproliferative efficacies. SAR investigations revealed that orientation and spatial topology of R3 substituents make greater contributions to the bioactivity, exemplified by compounds E24, E25 and E27, which has provided valuable information for guiding further development of acridone derivatives as promising drug candidates.
Collapse
Affiliation(s)
- Ji-Ning Chen
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Xing-Kang Wu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Chun-Hua Lu
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Xun Li
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China; Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong 250002, PR China
| |
Collapse
|
49
|
Thangamani M, Srinivasan K. Scandium(III) Triflate-Catalyzed Reaction of Aroyl-Substituted Donor-Acceptor Cyclopropanes with 1-Naphthylamines: Access to Dibenzo[ c, h]acridines. J Org Chem 2021; 86:1172-1177. [PMID: 33236912 DOI: 10.1021/acs.joc.0c02105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The reaction of aroyl-substituted donor-acceptor (D-A) cyclopropanes with two equivalents of 1-naphthylamines in the presence of a catalytic amount of scandium(III) triflate provides access to dibenzo[c,h]acridines. The key steps of the transformation are the formation of nucleophilic ring-opening products from the D-A cyclopropanes and 1-naphthylamines and their subsequent fragmentation and cyclization. The method has a reasonable substrate scope, and the products are formed in 50-70% yields.
Collapse
Affiliation(s)
- Murugesan Thangamani
- School of Chemistry, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620 024, India
| | - Kannupal Srinivasan
- School of Chemistry, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620 024, India
| |
Collapse
|
50
|
Grindstone Chemistry: Design, One-Pot Synthesis, and Promising Anticancer Activity of Spiro[acridine-9,2'-indoline]-1,3,8-trione Derivatives against the MCF-7 Cancer Cell Line. Molecules 2020; 25:molecules25245862. [PMID: 33322433 PMCID: PMC7763815 DOI: 10.3390/molecules25245862] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022] Open
Abstract
In this study, the synthesis of one-pot 10-phenyl-3,4,6,7-tetrahydro-1H-spiro [acridine-9,2'-indoline]-1,3,8-trione derivatives was achieved via a four-component cyclocondensation reaction, which was carried out in solvent-free conditions, and using p-toluenesulfonic acid (p-TSA) as a catalyst. The product was confirmed by FT-IR, 1H-NMR, 13C-NMR, mass spectra, and elemental analysis. Furthermore, the anticancer activity was screened for all compounds. Among these compounds, compound 1c was more effective (GI50 0.01 µm) against MCF-7 cancer cell lines than standard and other compounds. Therefore, the objective of this study was achieved with a few promising molecules having been demonstrated to be potential anticancer agents.
Collapse
|