1
|
Sulaiman NF, Zulkifli SZ, Saaidin AS, Lekkala R, Izzaty Hassan N, Pungot NH. Exploring β-carboline hybrids and their derivatives: A review on synthesis and anticancer efficiency. Eur J Med Chem 2025; 288:117412. [PMID: 39987835 DOI: 10.1016/j.ejmech.2025.117412] [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: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
β-Carboline is a crucial compound in medicinal chemistry known for its versatile pharmacological activities. Recent research has focused on hybrid molecules incorporating a β-carboline scaffold linked to other pharmacophore moieties. These hybrid compounds have demonstrated diverse therapeutic properties, including anticancer, antianxiety, antimalarial, antidepressant, anti-inflammatory, antileishmanial, and antioxidant effects. This review highlights studies conducted from 2014 to the present with a particular emphasis on the development of β-carboline hybrid compounds and their derivatives as potent anticancer agents. The structure-activity relationship (SAR) analysis reveals that these hybrids exhibit significant cytotoxicity against various cancer cell lines. This review aims to inspire further research into the novel synthesis and evolution of β-carboline hybrids and their derivatives, potentially leading to new therapeutic advancements.
Collapse
Affiliation(s)
- Nur Fatihah Sulaiman
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia
| | - Siti Zafirah Zulkifli
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia; Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Aimi Suhaily Saaidin
- Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Ravindar Lekkala
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bandar Baru Bangi, Selangor, 43600, Malaysia
| | - Nurul Izzaty Hassan
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bandar Baru Bangi, Selangor, 43600, Malaysia
| | - Noor Hidayah Pungot
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia; Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia.
| |
Collapse
|
2
|
Bhandari R, Kaleem M, Rai R, Shraogi N, Patnaik S, Misra A. A sensitive molecular probe exhibiting significant change in their photophysical and morphological behavior upon interaction with Fe 3+ ion. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125671. [PMID: 39742620 DOI: 10.1016/j.saa.2024.125671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/03/2025]
Abstract
An efficient molecular probe 8 has been designed and synthesized. The photophysical, electrochemical and morphological behavior of the probe has been examined in the absence and presence of different ions. The probe 8 at 90 % water fraction in acetonitrile showed aggregation induced emission (AIE). Probe 8 upon interaction with ions binds with Fe3+ ion selectively in a 1:1 stoichiometry and showed fluorescence "turn-Off" response with good limit of detection (LOD = 92.2 nM). The particle size (DLS method) of probe upon increasing water fraction in acetonitrile showed a gradual increase while upon formation of a stable complex, 8 + Fe3+ particle size decreased along with change in morphology of the probe. SEM and TEM studies showed that in pure acetonitrile probe self-assemble into a sheet like structure of uneven surface. While in aggregated state (fw, 90 %) it changes to a uniform hollow rectangular rod shape structure. Further interaction of the probe with Fe3+ ions in aggregated state acquired a well-defined smooth sheet. Electrochemical (CV) studies suggested that the redox property of the probe incurred a marginal change in band gap upon complexation with Fe3+. The cell imaging studies were performed to detect Fe3+ in HeLa cells. The paper strip test and real water sample analysis showed the potential analytical application of probe to detect Fe3+ with a naked-eye sensitive visible color change. The formation of a complex, 8 + Fe3+ involving N and O atoms of the probe molecule was confirmed by 1HNMR and HRMS data.
Collapse
Affiliation(s)
- Rimpi Bhandari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Mohammed Kaleem
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Ravisen Rai
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Nikita Shraogi
- Nano Laboratory, Drug and Chemical Toxicology Group, FEST Division, Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, UP, India
| | - Satyakam Patnaik
- Nano Laboratory, Drug and Chemical Toxicology Group, FEST Division, Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, UP, India
| | - Arvind Misra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| |
Collapse
|
3
|
Mou LL, Wu XM, Bibi A, Wang JX, Zhou CH. A comprehensive insight into naphthalimides as novel structural skeleton of multitargeting promising antibiotics. Future Med Chem 2025; 17:575-590. [PMID: 39957205 PMCID: PMC11901364 DOI: 10.1080/17568919.2025.2463872] [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: 10/20/2024] [Accepted: 01/27/2025] [Indexed: 02/18/2025] Open
Abstract
The globally growing antimicrobial resistance seriously threatens human health, increasing efforts have been devoting to the development of novel antibiotics. Naphthalimides contain a special skeleton of cyclic double imides and the naphthalene framework, this unique structure can exert multitargeting abilities which are helpful to overcome the escalating issue of resistance. Therefore, research in connection with the development of naphthalimides as novel antimicrobial agents is becoming progressively active. It has been revealed that naphthalimides as novel structural skeleton of multitargeting promising antibiotics could not only target DNAs and enzymes, disturb membrane, produce reactive oxygen species, etc. suggesting the multitargeting actions which do not induce resistance, but also show a broad antimicrobial spectrum with safety profile and pharmacokinetic characteristics, implying large potential as a new type of antibiotics via continuous efforts toward antimicrobial naphthalimides. This review presents naphthalimides as a new type of potential antimicrobial agents and discusses rational design strategies, structure-activity relationships, and mechanisms of action, with a comprehensive view to providing a new insight for in the rational design of efficient, broad-spectrum, and low-toxic naphthalimide antibiotics.
Collapse
Affiliation(s)
- Lin-Li Mou
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| | - Xin-Miao Wu
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| | - Aisha Bibi
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| | - Jin-Xin Wang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
- Chongqing Research Institute, Chongqing Werlchem New Materials Technology Co., Ltd., Chongqing, China
| | - Cheng-He Zhou
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| |
Collapse
|
4
|
Wang K, Wang L, Shang Z, Yang X, Li H, Wang X, Zhu M, Meng Q. A series of DNA targeted Cu (II) complexes containing 1,8-naphthalimide ligands: Synthesis, characterization and in vitro anticancer activity. J Inorg Biochem 2024; 261:112721. [PMID: 39236444 DOI: 10.1016/j.jinorgbio.2024.112721] [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: 06/21/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Copper(II) complexes are very promising candidates for platinum-based anticancer agents. Herein, three Cu (II) complexes (1-3) containing 1,8-naphthalimide ligands were synthesized and characterized by FT-IR, elemental analysis, ESI-MS and single crystal X-ray diffraction (complex 3). In addition, a control compound (complex 4) without 1,8-naphthalimide ligand was synthesized and characterized. The in vitro anticancer activity of the synthesized complexes against five cancer cell lines and one normal cell line was evaluated by MTS assay. The results displayed the antitumor activity of complexes 1-3 was controlled by the aliphatic chain length of ligands, their cytotoxicity was in the order 3 > 2 > 1, giving the IC50 values ranging from 2.874 ± 0.155 μM to 31.47 ± 0.29 μM against five cancer cell lines. Complex 4 showed less activity in comparison with complex 1-3. Notably, complexes 1-3 displayed much higher selectivity (SI = 2.65 to 10.16) compared to complex 4 (SI = 1.0), indicated that the introduction of 1,8-naphthalimide group not only increased the activity of this series of compounds but also enhanced their specific selectivity to cancer cells. Compound 3 induced apoptosis in cancer cells and blocked the S-phase and G2/M of cancer cells. The interaction with DNA of complexes 3 and 4 was studied by UV/Vis spectroscopic titrations, competitive DNA-binding experiment, viscometry and CD spectra. The results showed that complex 3 interacted with DNA in an intercalating mode, but the interaction mode of compound 4 with DNA was electrostatic interaction.
Collapse
Affiliation(s)
- Kehua Wang
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China; Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan Normal University, Anshan, Liaoning 114007, PR China
| | - Ling Wang
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Zhuye Shang
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China
| | - Xingzhi Yang
- Center for Natural Drug Activity Screening, Kunming Institute of Botany, Chinese Academy of Sciences, PR China
| | - Hongmei Li
- Center for Natural Drug Activity Screening, Kunming Institute of Botany, Chinese Academy of Sciences, PR China
| | - Xiaochun Wang
- Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan Normal University, Anshan, Liaoning 114007, PR China
| | - Mingchang Zhu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Qingtao Meng
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China.
| |
Collapse
|
5
|
Lee CC, Chang CH, Huang YC, Shih TL. Novel 1,8-Naphthalimide Derivatives Inhibit Growth and Induce Apoptosis in Human Glioblastoma. Int J Mol Sci 2024; 25:11593. [PMID: 39519145 PMCID: PMC11546702 DOI: 10.3390/ijms252111593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Given the rapid advancement of functional 1,8-Naphthalimide derivatives in anticancer research, we synthesized these two novel naphthalimide derivatives with diverse substituents and investigated the effect on glioblastoma multiforme (GBM) cells. Cytotoxicity, apoptosis, cell cycle, topoisomerase II and Western blotting assays were evaluated for these compounds against GBM in vitro. A human GBM xenograft mouse model established by subcutaneously injecting U87-MG cells and the treatment responses were assessed. Both compounds 3 and 4 exhibited significant antiproliferative activities, inducing apoptosis and cell death. Only compound 3 notably induced G2/M phase cell cycle arrest in the U87-MG GBM cells. Both compounds inhibited DNA topoisomerase II activity, resulting in DNA damage. The in vivo antiproliferative potential of compound 3 was further validated in a U87-MG GBM xenograft mouse model, without any discernible loss of body weight or kidney toxicity noted. This study presents novel findings demonstrating that 1,8-Naphthalimide derivatives exhibited significant GBM cell suppression in vitro and in vivo without causing adverse effects on body weight or kidney function. Further experiments, including investigations into mechanisms and pathways, as well as preclinical studies on the pharmacokinetics and pharmacodynamics, may be instrumental to the development of a new anti-GBM compound.
Collapse
Affiliation(s)
- Cheng-Chi Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333423, Taiwan;
| | - Chuan-Hsin Chang
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
- Research Center for Chinese Herbal Medicine, Graduate Institute of Healthy Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
| | - Yin-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333423, Taiwan;
| | - Tzenge-Lien Shih
- Department of Chemistry, Tamkang University, Tamsui Dist., New Taipei City 251301, Taiwan
| |
Collapse
|
6
|
Zuo Y, Lan Y, Gou Z, Chai Y, Yan M. Polysiloxane-Based Fluorescent Probes for Visualizing pH and Thiocyanate during Mitochondrial Autophagy. Anal Chem 2024; 96:16811-16817. [PMID: 39387845 DOI: 10.1021/acs.analchem.4c03499] [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: 10/15/2024]
Abstract
Mitochondrial autophagy, known as mitophagy, is a vital cellular process that involves the selective degradation of damaged or dysfunctional mitochondria through autophagy, which is critical to the functional integrity of the entire mitochondrial network and determines the survival and death of cells. An abnormal pH may lead to an imbalance in mitochondrial homeostasis and the occurrence of mitochondrial autophagic acidification and dysfunction. SCN- is also an important anion in cellular metabolism, and its abnormal concentration may lead to mitochondrial damage. However, so far, there are few reports on the simultaneous realization of pH and SCN- detection in mitochondria. Therefore, to complement the blank in this area, we developed the polysiloxane-based fluorescent probe P0-CMN that is capable of simultaneously visualizing pH and SCN- fluctuation levels in mitochondria. The probe P0-CMN has the desired mitochondrial-targeting properties and sensitivity to pH and SCN-. It is able to simultaneously monitor pH and SCN- changes in mitochondria in a dual-channel mode. In addition, probe P0-CMN can visualize pH changes during mitochondrial autophagy. This work provides an effective strategy for the design of dual-responsive fluorescent probes and further broadens the application of polysiloxane fluorescent materials.
Collapse
Affiliation(s)
- Yujing Zuo
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P.R. China
| | - Ying Lan
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P.R. China
| | - Zhiming Gou
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P.R. China
| | - Yanfu Chai
- School of Mechanical and Electrical Engineering, Shaoxing University, Shaoxing 312000, China
- School of Mechanical and Electrical Engineering, Shaoxing Key Laboratory of Mechanical Components Surface & Interface Science, Shaoxing University, Shaoxing 312000, China
| | - Mei Yan
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P.R. China
| |
Collapse
|
7
|
Wang Q, Liang SM, Mao ZC, Ma XL, Wei JH, Huang RZ, Zhang Y. Design, docking optimization, and evaluation of biotin-PEG4-1,8-naphthalimide as a potent and safe antitumor agent with dual targeting of ferroptosis and DNA. RSC Med Chem 2024; 15:1640-1651. [PMID: 38784471 PMCID: PMC11110740 DOI: 10.1039/d4md00134f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/30/2024] [Indexed: 05/25/2024] Open
Abstract
A set of biotin-polyethylene glycol (PEG)-naphthalimide derivatives 4a-4h with dual targeting of ferroptosis and DNA were designed and optimized using docking simulation as antitumor agents. Docking simulation optimization results indicated that biotin-PEG4-piperazine-1,8-naphthalimide 4d should be the best candidate among these designed compounds 4a-4h, and therefore, we synthesized and evaluated it as a novel antitumor agent. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and MGC-803 and U251 xenograft models identified 4d as a good candidate antitumor agent with potent efficacy and safety profiles, compared with amonafide and temozolomide. The findings of the docking simulations, fluorescence intercalator displacement (FID), western blot, comet, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transmission electron microscopy, and BODIPY-581/591-C11, FerroOrange, and dihydroethidium (DHE) fluorescent probe assays revealed that 4d could induce DNA damage, affect DNA synthesis, and cause cell cycle arrest in the S phase in MGC-803 cells. Also, it could induce lipid peroxidation and thus lead to ferroptosis in MGC-803 cells, indicating that it mainly exerted antitumor effects through dual targeting of ferroptosis and DNA. These results suggested that it was feasible to design, optimize using docking simulation, and evaluate the potency and safety of biotin-PEG-1,8-naphthalimide as a antitumor agent with dual targeting of ferroptosis and DNA, based on a multi-target drug strategy.
Collapse
Affiliation(s)
- Qi Wang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University Guilin 541004 China
| | - Si-Min Liang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University Guilin 541004 China
| | - Zhi-Chen Mao
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University Guilin 541004 China
| | - Xian-Li Ma
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University Guilin 541004 China
| | - Jian-Hua Wei
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University Guilin 541004 China
| | - Ri-Zhen Huang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University Guilin 541004 China
- Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University Guilin 541004 China
| |
Collapse
|
8
|
Morozov BS, Gargiulo F, Ghule S, Lee DJ, Hampel F, Kim HM, Kataev EA. Macrocyclic Conformational Switch Coupled with Pyridinium-Induced PET for Fluorescence Detection of Adenosine Triphosphate. J Am Chem Soc 2024; 146:7105-7115. [PMID: 38417151 DOI: 10.1021/jacs.4c01621] [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: 03/01/2024]
Abstract
The binding of nucleotides is crucial for signal transduction as it induces conformational protein changes, leading to downstream cellular responses. Synthetic receptors that bind nucleotides and transduce the binding event into global conformational rearrangements are highly challenging to design, especially those that operate in an aqueous solution. Much work is focused on evaluating functionalized dyes to detect nucleotides, whereas coupling of a nucleotide-induced conformational switching to a sensing event has not been reported to date. We disclose synthetic receptors that undergo a global conformational rearrangement upon nucleotide binding. Integrating naphthalimide and the pyridinium ion into the structure enables stabilization of the folded conformation and efficient fluorescence quenching. The binding of a nucleotide rearranges the receptor conformation and alters the strong fluorescence enhancement. The methylpyridinium-containing receptor demonstrated high sensing selectivity for adenosine 5'-triphosphate (ATP) and a record 160-fold fluorescence enhancement. It can detect fluctuations of ATP in HeLa cells and possesses low cytotoxicity. The developed systems present an attractive approach for designing ATP-responsive artificial molecular switches that operate in water and integrate a strong fluorescence response.
Collapse
Affiliation(s)
- Boris S Morozov
- Department of Chemistry and Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Fabiano Gargiulo
- Department of Chemistry and Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Swapnil Ghule
- Department of Chemistry and Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Dong Joon Lee
- Department of Chemistry and Department of Energy Systems Research, Ajou University, 16499 Suwon, Republic of Korea
| | - Frank Hampel
- Department of Chemistry and Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Hwan Myung Kim
- Department of Chemistry and Department of Energy Systems Research, Ajou University, 16499 Suwon, Republic of Korea
| | - Evgeny A Kataev
- Department of Chemistry and Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
9
|
Adhikari S, Nath P, Das A, Datta A, Baildya N, Duttaroy AK, Pathak S. A review on metal complexes and its anti-cancer activities: Recent updates from in vivo studies. Biomed Pharmacother 2024; 171:116211. [PMID: 38290253 DOI: 10.1016/j.biopha.2024.116211] [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: 10/21/2023] [Revised: 12/22/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
Research into cancer therapeutics has uncovered various potential medications based on metal-containing scaffolds after the discovery and clinical applications of cisplatin as an anti-cancer agent. This has resulted in many metallodrugs that can be put into medical applications. These metallodrugs have a wider variety of functions and mechanisms of action than pure organic molecules. Although platinum-based medicines are very efficient anti-cancer agents, they are often accompanied by significant side effects and toxicity and are limited by resistance. Some of the most studied and developed alternatives to platinum-based anti-cancer medications include metallodrugs based on ruthenium, gold, copper, iridium, and osmium, which showed effectiveness against many cancer cell lines. These metal-based medicines represent an exciting new category of potential cancer treatments and sparked a renewed interest in the search for effective anti-cancer therapies. Despite the widespread development of metal complexes touted as powerful and promising in vitro anti-cancer therapeutics, only a small percentage of these compounds have shown their worth in vivo models. Metallodrugs, which are more effective and less toxic than platinum-based drugs and can treat drug-resistant cancer cells, are the focus of this review. Here, we highlighted some of the most recently developed Pt, Ru, Au, Cu, Ir, and Os complexes that have shown significant in vivo antitumor properties between 2017 and 2023.
Collapse
Affiliation(s)
- Suman Adhikari
- Department of Chemistry, Govt. Degree Collage, Dharmanagar, Tripura (N) 799253, India.
| | - Priyatosh Nath
- Department of Human Physiology, Tripura University, Suryamaninagar, West Tripura 799022, India
| | - Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Abhijit Datta
- Department of Botany, Ambedkar College, Fatikroy, Unakoti 799290, Tripura, India
| | - Nabajyoti Baildya
- Department of Chemistry, Milki High School, Milki, Malda 732209, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| |
Collapse
|
10
|
Zhang W, Ma Y, Song H, Miao R, Kong J, Zhou M. Deciphering the photophysical properties of naphthalimide derivatives using ultrafast spectroscopy. Phys Chem Chem Phys 2024; 26:4607-4613. [PMID: 38251277 DOI: 10.1039/d3cp05654f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Naphthalimide derivatives composed of donor-acceptor type structures hold significant promise across a wide range of applications. Here, the solvent polarity and viscosity controlled excited-state dynamics of a naphthalimide derivative with a donor-acceptor structure were studied using multiple spectroscopic techniques. From the stationary spectroscopic investigations, large Stokes shift and low fluorescence quantum yield were observed with increasing the solvent polarity, suggesting a more polar excited state relative to the ground state, which is evidenced by the Lippert-Mataga relationship. We also observe an enhanced fluorescence with a prolonged lifetime in a more viscous solution due to the restriction of excited-state molecular rearrangement. These observations result from the emerged twisted intramolecular charge transfer (TICT) state. The ultrafast spectroscopy studies further unravel a solvent polarity dependent excited state evolution from the intramolecular charge transfer state to the TICT state, revealing that the TICT state can be populated only in strong polar solvents. Control experiments by tuning the solvent viscosity in ultrafast experiments were employed to verify the excited state molecular rearrangement subsequently. These observations collectively emphasize how fine-tuning the photophysical properties of naphthalimide derivatives can be achieved through strategic manipulation of solvent polarity and viscosity.
Collapse
Affiliation(s)
- Wei Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Anhui 230026, China.
| | - Yalei Ma
- Laboratory of Applied Surface and Colloids Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, P. R. China.
| | - Hongwei Song
- Department of Chemistry-Angstrom Laboratory, Uppsala University, Box 523, Uppsala 75120, Sweden
| | - Rong Miao
- Laboratory of Applied Surface and Colloids Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, P. R. China.
| | - Jie Kong
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Anhui 230026, China.
| | - Meng Zhou
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Anhui 230026, China.
| |
Collapse
|
11
|
Liang SM, Liang GB, Wang HL, Jiang H, Ma XL, Wei JH, Huang RZ, Zhang Y. Discovery of 4-(N-dithiobenzyl piperazine)-1,8-naphthalimide as a potent multi-target antitumor agent with good efficacy, limited toxicity, and low resistance. Eur J Med Chem 2024; 263:115937. [PMID: 37972528 DOI: 10.1016/j.ejmech.2023.115937] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/04/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
A series of 4-(N-dithiobenzyl piperazine)-1,8-naphthalimide derivatives 4-6 were designed, synthesized, and evaluated as novel multi-target antitumor agents. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) results showed that compounds 5j, 5k, and 6j exhibited superior in vitro antiproliferative activity in MGC-803, HepG-2, SKOV-3, and T24 cancer cell lines and the cisplatin-resistant cell line A549/DDP. HepG-2, SKOV-3, and T24 xenograft assay results revealed that compounds 5j, 5k, and 6j exhibited good antitumor effects compared with amonafide. The pathology results indicated that compound 5j exhibited the least comprehensive toxicity among the three compounds, identifying compound 5j as a good candidate antitumor agent with good efficacy, limited toxicity, and low resistance. Compound 5j was thus chose for further antitumor mechanism investigation. Results from the omics research, confocal immunofluorescence, Western blot, transmission electron microscopy, and flow cytometry indicated that compound 5j exerted antitumor effects through multiple mechanisms, including ferroptosis, autophagy, apoptosis, and cell cycle arrest. These results suggest that screening novel 1,8-naphthalimide-based antitumor agents for good efficacy, limited toxicity, and low resistance based on a multi-target drug strategy is feasible.
Collapse
Affiliation(s)
- Si-Min Liang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China
| | - Gui-Bin Liang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China
| | - Hui-Ling Wang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China
| | - Hong Jiang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China
| | - Xian-Li Ma
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China
| | - Jian-Hua Wei
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China.
| | - Ri-Zhen Huang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China.
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin, 541004, China.
| |
Collapse
|
12
|
Napiórkowska M, Kurpios-Piec D, Kiernozek-Kalińska E, Leśniak A, Klawikowska M, Bujalska-Zadrożny M. New aryl-/heteroarylpiperazine derivatives of 1,7-dimethyl-8,9-diphenyl-4-azatricyclo[5.2.1.0 2,6]dec-8-ene-3,5,10-trione: Synthesis and preliminary studies of biological activities. Bioorg Med Chem 2023; 96:117518. [PMID: 37951135 DOI: 10.1016/j.bmc.2023.117518] [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: 08/28/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
Compounds containing dicarboximide skeleton such as succinimides, maleimides, glutarimides, and phthalimides possess broad biological properties including anti-fungal, antibacterial, antidepressant, or analgesic activities. The piperazine ring is found in a wide range of molecules that have demonstrated a variety of biological functions such as anticancer action and 5-HT receptors agonist/antagonist activity. In the present study, we combined both structures to develop new antitumor agents, a series of piperazine derivatives of 1,7-dimethyl-8,9-diphenyl-4-azatricyclo[5.2.1.02,6]dec-8-ene-3,5,10-trione and evaluated their biological activity. The structures of all tested compounds were confirmed by 1H and 13C NMR and by ESI MS spectral analysis. Their cytotoxicity was assessed in vitro against eight human cancer cell lines, namely prostate (PC3), colon (HCT116, SW480, SW620), leukemia (K562), liver (HepG2), lung (A549) and breast (MDA-Mb-231) in contrast to normal HMEC-1 cell line, by using MTT and Trypan blue method. The tested compounds showed significant activity toward cancer cells. The most pronounced cytotoxic effect was observed in K562 and HCT116 with IC50 values below 10 μM for all studied compounds. Importantly, the most promising derivatives for each cancer cell line (IC50 < 10 μM) exerted a weaker cytotoxic effect toward normal HMEC-1 cells than cancer cells. The evaluation of proapoptotic and inhibitory effects on IL-6 release showed that K562 and HCT116 cells were more sensitive to studied compounds than other cancer cell lines. Furthermore, for all piperazine derivatives, the functional activities at the 5-HT1A, D2 receptors as well as their binding affinities at the 5-HT2A, H1 and M receptors, were determined. The current investigation was able to successfully design compounds with both serotoninergic and anticancer properties. It serves as a good starting point for a multimodal approach for the management of cancer and cancer-related symptoms.
Collapse
Affiliation(s)
- Mariola Napiórkowska
- Chair and Department of Biochemistry, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland.
| | - Dagmara Kurpios-Piec
- Chair and Department of Biochemistry, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Ewelina Kiernozek-Kalińska
- Department of Immunology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str., 02-096 Warsaw, Poland
| | - Anna Leśniak
- Department of Pharmacodynamics, Faculty of Pharmacy, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Małgorzata Klawikowska
- Department of Pharmacodynamics, Faculty of Pharmacy, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Magdalena Bujalska-Zadrożny
- Department of Pharmacodynamics, Faculty of Pharmacy, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| |
Collapse
|
13
|
Rykowski S, Gurda-Woźna D, Fedoruk-Wyszomirska A, Orlicka-Płocka M, Kowalczyk A, Stączek P, Denel-Bobrowska M, Biniek-Antosiak K, Rypniewski W, Wyszko E, Olejniczak AB. Carboranyl-1,8-naphthalimide intercalators induce lysosomal membrane permeabilization and ferroptosis in cancer cell lines. J Enzyme Inhib Med Chem 2023; 38:2171028. [PMID: 36715272 PMCID: PMC9888480 DOI: 10.1080/14756366.2023.2171028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
The synthesis of carborane-1,8-naphthalimide conjugates and evaluation of their DNA-binding ability and anticancer activity were performed. A series of 4-carboranyl-3-nitro-1,8-naphthalimide derivatives, mitonafide and pinafide analogs, were synthesised via amidation and reductive amination reactions, and their calf thymus DNA (ct-DNA)-binding properties were investigated using circular dichroism, UV-vis spectroscopy, and thermal denaturation. Results showed that conjugates 34-37 interacted very strongly with ct-DNA (ΔTm = 10.00-13.00 °C), indicating their ability to intercalate with DNA, but did not inhibit the activity of topoisomerase II. The conjugates inhibited the cell growth of the HepG2 cancer cell line in vitro. The same compounds caused the G2M phase arrest. Cell lines treated with these conjugates showed an increase in reactive oxygen species, glutathione, and Fe2+ levels, lipid peroxidation, and mitochondrial membrane potential relative to controls, indicating the involvement of ferroptosis. Furthermore, these conjugates caused lysosomal membrane permeabilization in HepG2 cells but not in MRC-5 cells.
Collapse
Affiliation(s)
| | - Dorota Gurda-Woźna
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | | | | - Aleksandra Kowalczyk
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Paweł Stączek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | | | | | - Wojciech Rypniewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Eliza Wyszko
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | |
Collapse
|
14
|
Ge C, Liu L, Wang Y, Di X, Luo X, Liu H, Qian Y. Novel 1,8-Naphthalimide Derivatives As Antitumor Agents and Potent Demethylase Inhibitors. ACS Med Chem Lett 2023; 14:1551-1557. [PMID: 37974948 PMCID: PMC10641888 DOI: 10.1021/acsmedchemlett.3c00353] [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: 08/09/2023] [Accepted: 10/09/2023] [Indexed: 11/19/2023] Open
Abstract
Functional 1,8-naphthalimide derivatives are rapidly developing in the field of anticancer research. Herein, we designed and synthesized a series of naphthalimide derivatives with different substituents. Interestingly, 1,8-naphthalimide derivatives 1 and 7 inhibited a human demethylase FTO (the fat mass and obesity-associated protein). Computer simulation studies further indicated that 1 and 7 entered the FTO's structural domain II binding pocket through hydrophobic and hydrogen bonding interactions. Anticancer mechanism studies showed that 1 and 7 induced DNA damage and autophagic cell death in A549 cells. The high antiproliferative activity of 1 and 7 was further confirmed by 3D multicellular A549 tumor spheroid assays. This study focuses on the cytotoxicity and mode of action of naphthalimide derivatives, which not only have potential anticancer activity but also are potent demethylase inhibitors.
Collapse
Affiliation(s)
- Chao Ge
- School
of Chemistry and Materials Science, Jiangsu
Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing 210023, People’s Republic of China
- School
of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, People’s Republic of China
| | - Lu Liu
- School
of Chemistry and Materials Science, Jiangsu
Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing 210023, People’s Republic of China
| | - Yanqing Wang
- School
of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, People’s Republic of China
| | - Xiaojiao Di
- School
of Chemistry and Materials Science, Jiangsu
Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing 210023, People’s Republic of China
| | - Xiangjie Luo
- School
of Chemistry and Materials Science, Jiangsu
Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing 210023, People’s Republic of China
| | - Hongke Liu
- School
of Chemistry and Materials Science, Jiangsu
Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing 210023, People’s Republic of China
| | - Yong Qian
- School
of Chemistry and Materials Science, Jiangsu
Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing 210023, People’s Republic of China
| |
Collapse
|
15
|
Chen XM, Zhou JY, Liu SQ, Song LH, Wang HL, Wang Q, Liang SM, Lu L, Wei JH, Huang R, Zhang Y. Design, synthesis, and antitumor evaluation of morpholine substituted bisnaphthalimides as DNA targeting agents. Bioorg Med Chem Lett 2023; 85:129218. [PMID: 36894107 DOI: 10.1016/j.bmcl.2023.129218] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
A series of mono- and bisnaphthalimides derivatives containing 3-nitro and 4-morpholine moieties were designed, synthesized, and evaluated for their in vitro anticancer activities against four cancer cell lines. Some compounds exhibited relatively good antiproliferative activity on the cell lines tested, in comparison with mitonafide and amonafide. It is noteworthy that bisnaphthalimide A6 was identified as the most potent compound in anti-proliferation against MGC-803 cells, with an IC50 lowered to 0.09 μM, a far greater potency than that of mono-naphthalimide A7, mitonafide, and amonafide. A gel electrophoresis assay revealed that DNA and Topo I were the potential targets of compounds A6 and A7. The treatment of CNE-2 cells with compounds A6 and A7 resulted in an S phase cell cycle arrest, accompanied by the upregulation of the expression levels of the antioncogene p27 and the down-regulation of the expression levels of CDK2 and cyclin E. In addition, compounds A6 and A7-induced apoptosis was further confirmed by flow cytometry, ROS generation assay, and Hoechst 33,258 staining. In particular, in vivo antitumor assay results revealed that bisnaphthalimide A6 exhibited potent anticancer efficiency in an MGC-803 xenograft tumor model, in comparison with mitonafide, and had lower toxicity than mono-naphthalimide A7. In brief, the results suggested that bisnaphthalimide derivatives containing 3-nitro and 4-morpholine moieties might serve as DNA binding agents for the development of new antitumor agents.
Collapse
Affiliation(s)
- Xiao-Man Chen
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Jian-Yu Zhou
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Shuang-Qiang Liu
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Long-Hao Song
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Hui-Ling Wang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Qi Wang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Si-Min Liang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Lin Lu
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Jian-Hua Wei
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China.
| | - Rizhen Huang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China.
| | - Ye Zhang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China.
| |
Collapse
|
16
|
Cao SY, Zhou Y, Ma YX, Cheng SX, Tang GM, Wang YT. Syntheses, crystal structure, luminescent behaviors and Hirshfeld surface of salts with imidazole and benzimidazole-yl scaffolds. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Hassan RA, Hamed MI, Abdou AM, El-Dash Y. Novel antiproliferative agents bearing substituted thieno[2,3-d]pyrimidine scaffold as dual VEGFR-2 and BRAF kinases inhibitors and apoptosis inducers; design, synthesis and molecular docking. Bioorg Chem 2022; 125:105861. [DOI: 10.1016/j.bioorg.2022.105861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 12/15/2022]
|
18
|
Staneva D, Manov H, Vasileva‐Tonkova E, Kukeva R, Stoyanova R, Grabchev I. Enhancing the antibacterial activity of
PAMAM
dendrimer modified with 1,8‐naphthalimides and its copper complex via light illumination. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Desislava Staneva
- Department of Textile, Leader and Fuels University of Chemical Technology and Metallurgy Sofia Bulgaria
| | - Hristo Manov
- Faculty of Chemistry and Pharmacy Sofia University “St. Kliment Ohridski” Sofia Bulgaria
| | | | - Rositsa Kukeva
- Institute of General and Inorganic Chemistry Bulgarian Academy of Sciences Sofia Bulgaria
| | - Radostina Stoyanova
- Institute of General and Inorganic Chemistry Bulgarian Academy of Sciences Sofia Bulgaria
| | - Ivo Grabchev
- Faculty of Medicine Sofia University “St. Kliment Ohridski” Sofia Bulgaria
| |
Collapse
|