1
|
Omidkhah N, Gheisari A, Oskuei SR, Chamani J, Hadizadeh F, Atarodi A, Ghodsi R. Design, synthesis, biological evaluation and multi spectroscopic studies of novel 2-styrylquinoline-carboxamide derivatives as potential DNA intercalating anticancer agents. Bioorg Chem 2025; 154:108098. [PMID: 39753040 DOI: 10.1016/j.bioorg.2024.108098] [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: 10/14/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025]
Abstract
In this study, novel 2-styrylquinoline derivatives possessing a planar aromatic system and a flexible side chain with an amino substituent were designed and synthesized as DNA-intercalating antitumor agents. The cytotoxic activity of the synthesized compounds was evaluated against four cancer cell lines including MCF-7 (breast cancer cells), A549 (lung epithelial cancer cells), HCT116 (colon cancer cells) and normal cell line L929 (mouse fibroblast cell line). The results displayed that the anti-cancer activity of the target quinolines is sensitive to the lipophilic nature of the C-6 and C-7 quinoline substituents. The anticancer activity of most of the target quinolines against MCF-7 and A549 cells was more than those of HCT116. Compound 3h possessing two methyl groups at the C-6 and C-7 of quinoline ring displayed the most cytotoxicity with IC50 value of 5.7 µM against A549 cancer cells. Interaction of compound 3h with calf thymus DNA (ctDNA) was investigated by means of UV absorption spectrophotometry, fluorescence spectroscopy, circular dichroism (CD), Resonance light scattering (RLS), viscos metric techniques and also by docking and molecular dynamic studies. RLS intensity, fluorescence quenching of ctDNA and fluorescence quenching EtBr-ctDNA and AO-ctDNA complexes augmented with increasing of compound 3h concentration, significant increasing in viscosity and melting point of ctDNA in the presence of compound 3h, absorbance increasing of ctDNA-compound 3h complex by increasing of NaCl and KI concentrations, higher Ksv value for dsctDNA (3.03 × 104 M-1) compared to ssctDNA (1.31 × 104 M-1), circular dichroism (CD) studies, docking and molecular dynamic studies revealed that compound 3h can interact with ctDNA through intercalation into DNA.
Collapse
Affiliation(s)
- Negar Omidkhah
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Gheisari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Rahimzadeh Oskuei
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshidkhan Chamani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Atarodi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Anticancer Activities of 9-chloro-6-(piperazin-1-yl)-11H-indeno[1,2-c] quinolin-11-one (SJ10) in Glioblastoma Multiforme (GBM) Chemoradioresistant Cell Cycle-Related Oncogenic Signatures. Cancers (Basel) 2022; 14:cancers14010262. [PMID: 35008426 PMCID: PMC8750065 DOI: 10.3390/cancers14010262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) remains to be the most frequent malignant tumor of the central nervous system (CNS), which accounts for approximately 54% of all primary brain gliomas. Current treatment modalities for GBM include surgical resection, followed by radiotherapy and chemotherapy with temozolomide (TMZ). However, due to its genetic heterogeneity, GBM tumors always recur, due to treatment reasistance. The aim of this study was to identify molecular gene signatures, responsible for cancer initiation, progression, resistances and to treatment, metastasis, and also evaluate the potency of our novel compounds SJ10 as potential target for CCNB1/CDC42/MAPK7/CD44 oncogenic signatures. Accordingly, we used computational simulation and identify these signatures as regulators of the cell cycle in GBM, which leads to cancer development and metastasis. We also showed the antiproliferative and cytotoxic effects of SJ10 compound against a panel of NCI-60 cancer cell lines. This suggests the potential of the compounds to inhibit CCNB1/CDC42/MAPK7/CD44 in GBM. Abstract Current anticancer treatments are inefficient against glioblastoma multiforme (GBM), which remains one of the most aggressive and lethal cancers. Evidence has shown the presence of glioblastoma stem cells (GSCs), which are chemoradioresistant and associated with high invasive capabilities in normal brain tissues. Moreover, accumulating studies have indicated that radiotherapy contributes to abnormalities in cell cycle checkpoints, including the G1/S and S phases, which may potentially lead to resistance to radiation. Through computational simulations using bioinformatics, we identified several GBM oncogenes that are involved in regulating the cell cycle. Cyclin B1 (CCNB1) is one of the cell cycle-related genes that was found to be upregulated in GBM. Overexpression of CCNB1 was demonstrated to be associated with higher grades, proliferation, and metastasis of GBM. Additionally, increased expression levels of CCNB1 were reported to regulate activation of mitogen-activated protein kinase 7 (MAPK7) in the G2/M phase, which consequently modulates mitosis; additionally, in clinical settings, MAPK7 was demonstrated to promote resistance to temozolomide (TMZ) and poor patient survival. Therefore, MAPK7 is a potential novel drug target due to its dysregulation and association with TMZ resistance in GBM. Herein, we identified MAPK7/extracellular regulated kinase 5 (ERK5) genes as being overexpressed in GBM tumors compared to normal tissues. Moreover, our analysis revealed increased levels of the cell division control protein homolog (CDC42), a protein which is also involved in regulating the cell cycle through the G1 phase in GBM tissues. This therefore suggests crosstalk among CCNB1/CDC42/MAPK7/cluster of differentiation 44 (CD44) oncogenic signatures in GBM through the cell cycle. We further evaluated a newly synthesized small molecule, SJ10, as a potential target agent of the CCNB1/CDC42/MAPK7/CD44 genes through target prediction tools and found that SJ10 was indeed a target compound for the above-mentioned genes; in addition, it displayed inhibitory activities against these oncogenes as observed from molecular docking analysis.
Collapse
|
3
|
Lawal B, Kuo YC, Tang SL, Liu FC, Wu ATH, Lin HY, Huang HS. Transcriptomic-Based Identification of the Immuno-Oncogenic Signature of Cholangiocarcinoma for HLC-018 Multi-Target Therapy Exploration. Cells 2021; 10:2873. [PMID: 34831096 PMCID: PMC8616156 DOI: 10.3390/cells10112873] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinomas (CHOLs), hepatobiliary malignancies, are characterized by high genetic heterogeneity, a rich tumor microenvironment, therapeutic resistance, difficulty diagnosing, and poor prognoses. Current knowledge of genetic alterations and known molecular markers for CHOL is insufficient, necessitating the need for further evaluation of the genome and RNA expression data in order to identify potential therapeutic targets, clarify the roles of these targets in the tumor microenvironment, and explore novel therapeutic drugs against the identified targets. Consequently, in our attempt to explore novel genetic markers associated with the carcinogenesis of CHOL, five genes (SNX15, ATP2A1, PDCD10, BET1, and HMGA2), collectively termed CHOL-hub genes, were identified via integration of differentially expressed genes (DEGs) from relatively large numbers of samples from CHOL GEO datasets. We further explored the biological functions of the CHOL-hub genes and found significant enrichment in several biological process and pathways associated with stem cell angiogenesis, cell proliferation, and cancer development, while the interaction network revealed high genetic interactions with a number of onco-functional genes. In addition, we established associations between the CHOL-hub genes and tumor progression, metastasis, tumor immune and immunosuppressive cell infiltration, dysfunctional T-cell phenotypes, poor prognoses, and therapeutic resistance in CHOL. Thus, we proposed that targeting CHOL-hub genes could be an ideal therapeutic approach for treating CHOLs, and we explored the potential of HLC-018, a novel benzamide-linked small molecule, using molecular docking of ligand-receptor interactions. To our delight, HLC-018 was well accommodated with high binding affinities to binding pockets of CHOL-hub genes; more importantly, we found specific interactions of HLC-018 with the conserved sequence of the AT-hook DNA-binding motif of HMGA2. Altogether, our study provides insights into the immune-oncogenic phenotypes of CHOL and provides valuable information for our ongoing experimental validation.
Collapse
Affiliation(s)
- Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Sung-Ling Tang
- Department of Pharmacy Practice, Tri-Service General Hospital, School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Feng-Cheng Liu
- Department of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
| | - Alexander T. H. Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Hung-Yun Lin
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
4
|
Lawal B, Kuo YC, Sumitra MR, Wu ATH, Huang HS. In vivo Pharmacokinetic and Anticancer Studies of HH-N25, a Selective Inhibitor of Topoisomerase I, and Hormonal Signaling for Treating Breast Cancer. J Inflamm Res 2021; 14:4901-4913. [PMID: 34588796 PMCID: PMC8473721 DOI: 10.2147/jir.s329401] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/11/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Breast cancer is the most frequently diagnosed cancer globally, and the leading cause of cancer-associated mortality among women. The efficacy of most clinical chemotherapies is often limited by poor pharmacokinetics and the development of drug resistance by tumors. In a continuing effort to explore small molecules as alternative therapies, we herein evaluated the therapeutic potential of HH-N25, a novel nitrogen-substituted anthra[1,2-c][1,2,5]thiadiazole-6,11-dione derivative. METHODS We evaluated the in vivo pharmacokinetic properties and maximum tolerated dose (MTD) of HH-N25 in rats. We also characterized the compound for in vitro and in vivo anticancer activities and its inhibitory effects against DNA topoisomerases and hormonal signaling in breast cancer. Furthermore, we used molecular docking to analyse the ligand-receptor interactions between the compound and the targets. RESULTS The maximum serum concentration (Cmax), half-life (t1/2 beta), mean residence time (MRT), oral clearance (CL/f), and apparent volume of distribution (VD/f) of HH-N25 were 1446.67 ± 312.05 ng/mL, 4.51 ± 0.27 h, 2.56 ± 0.16 h, 8.32 ± 1.45 mL/kg/h, and 1.26 ± 0.15 mL/kg, respectively, after single-dose iv administration at 3 mg/kg body weight. HH-N25 had potent anticancer activity against a panel of human breast cancer cell lines with 50% inhibitory concentrations (IC50) ranging 0.045±0.01~4.21±0.05 µM. The drug also demonstrated marked in vivo anticancer activity at a tolerated dose and prolonged the survival duration of mice without unacceptable toxicities based on body weight changes in human tumor xenograft models. In addition, HH-N25 exhibited a dose-dependent inhibition of topoisomerase I and ligand-mediated activities of progesterone and androgen receptors. CONCLUSION HH-N25 represents a new molecular entity that selective suppressed TOP1 and hormonal signaling, and shows potent antitumor activities in human breast cancer cells in vitro and in vivo. HH-N25 thus represents a promising anticancer agent that warrants further preclinical and clinical exploration.
Collapse
Affiliation(s)
- Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Maryam Rachmawati Sumitra
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Alexander T H Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 11490, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei, 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| |
Collapse
|
5
|
Design and synthesis of novel conformationally constrained 7,12-dihydrodibenzo[b,h][1,6] naphthyridine and 7H-Chromeno[3,2-c] quinoline derivatives as topoisomerase I inhibitors: In vitro screening, molecular docking and ADME predictions. Bioorg Chem 2021; 115:105174. [PMID: 34314913 DOI: 10.1016/j.bioorg.2021.105174] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 12/25/2022]
Abstract
Novel non-camptothecin (non-CPT) class of conformationally constrained, hitherto unknown 7,12-dihydrodibenzo[b,h][1,6] naphthyridine and 7H-Chromeno[3,2-c] quinoline derivatives have been designed, synthesized and evaluated for anti-cancer activity. In vitro anti-proliferation evaluation against human cancer cell lines (A549 and MCF-7) exhibited significant cytotoxicity. Among the derivatives (8-24), 8 (IC50 0.44 μM and IC50 0.62 μM) and 12 (IC50 0.69 μM and IC50 0.54 μM) were identified as the most promising candidate against A-549 and MCF-7 cancer cell lines respectively. Topo I inhibitory activity of 8 and 12 suggested that, they may be developed as potential anti-cancer molecules in future and rationalized by docking analysis with effective binding modes. Further, in silico ADME prediction studies of all derivatives were found promising, signifying the drug like properties. In precise, the present investigation displays a new strategy to synthesize and emphasis on anticancer activities of conformationally constrained dibenzo[b,h][1,6] naphthyridine derivatives and Chromeno[3,2-c] quinoline derivatives in the context of cancer drug development and refinement.
Collapse
|
6
|
Bernal CC, Bohórquez ARR, Henao JA, Macías MA. Synthesis, thermal stabilities and solid state analysis of few selected tetrahydro-7H-indeno[2,1-c]quinoline derivatives. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
7
|
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: 41] [Impact Index Per Article: 10.3] [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
|
8
|
Mekheimer RA, Al-Sheikh MA, Medrasi HY, Sadek KU. Advancements in the synthesis of fused tetracyclic quinoline derivatives. RSC Adv 2020; 10:19867-19935. [PMID: 35520416 PMCID: PMC9054245 DOI: 10.1039/d0ra02786c] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/07/2020] [Indexed: 11/21/2022] Open
Abstract
Fused tetracyclic systems containing a quinoline nucleus represent an important class of heterocyclic bioactive natural products and pharmaceuticals because of their significant and wide-spectrum biological properties. Several of these compounds have been obtained with diverse pharmacological and biological activities, such as antiplasmodial, antifungal, antibacterial, potent antiparasitic, antiproliferative, anti-tumor and anti-inflammatory activities. This information will be beneficial for medicinal chemists in the field of drug discovery to design and synthesize new fused tetracyclic quinolines as potent therapeutical agents. This review article provides a comprehensive report regarding the methods developed for the synthesis of fused tetracyclic quinolines reported so far (till October 2019). The article includes synthesis by one-pot domino reaction, microwave synthesis using a catalyst, using ionic liquids, photocatalytic synthesis (UV radiation), Pfitzinger reaction, I2-catalyzed cyclization reaction, Wittig reaction, cascade reaction, imino Diels-Alder reaction, Friedel-Crafts reaction, CDC reaction, solvent-free reactions and using small chiral organic molecules as catalysts. To the best of our knowledge, this is the first review focused on the synthesis of fused tetracyclic quinolines along with mechanistic aspects.
Collapse
Affiliation(s)
- Ramadan A Mekheimer
- Department of Chemistry, Faculty of Science, Minia University Minia 61519 Egypt
| | - Mariam A Al-Sheikh
- Department of Chemistry, Faculty of Science, University of Jeddah Jeddah 21589 Saudi Arabia
| | - Hanadi Y Medrasi
- Department of Chemistry, Faculty of Science, University of Jeddah Jeddah 21589 Saudi Arabia
| | - Kamal U Sadek
- Department of Chemistry, Faculty of Science, Minia University Minia 61519 Egypt
| |
Collapse
|
9
|
Ye F, Tran C, Jullien L, Le Saux T, Haddad M, Michelet V, Ratovelomanana-Vidal V. Synthesis of Fluorescent Azafluorenones and Derivatives via a Ruthenium-Catalyzed [2 + 2 + 2] Cycloaddition. Org Lett 2018; 20:4950-4953. [PMID: 30070483 DOI: 10.1021/acs.orglett.8b02085] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An original and mild synthetic route for the preparation of novel azafluorenones and derivatives via a ruthenium-mediated [2 + 2 + 2] cycloaddition of α,ω-diynes and cyanamides has been developed. This atom-economical catalytic process demonstrated remarkable regioselectivities to access fluorescent azafluorenone derivatives. The photophysical properties of azafluorenone derivatives have been evaluated, and photoluminescence phenomena at solid and liquid states have been highlighted.
Collapse
Affiliation(s)
- Fei Ye
- PSL Research University, Chimie ParisTech - CNRS, Institut de Recherche de Chimie Paris , Paris 75005 , France
| | - Christine Tran
- PSL Research University, Chimie ParisTech - CNRS, Institut de Recherche de Chimie Paris , Paris 75005 , France
| | - Ludovic Jullien
- PASTEUR, Chemistry Department , École Normale Supérieure, PSL University, Sorbonne University, CNRS , Paris 75005 , France
| | - Thomas Le Saux
- PASTEUR, Chemistry Department , École Normale Supérieure, PSL University, Sorbonne University, CNRS , Paris 75005 , France
| | - Mansour Haddad
- PSL Research University, Chimie ParisTech - CNRS, Institut de Recherche de Chimie Paris , Paris 75005 , France
| | - Véronique Michelet
- PSL Research University, Chimie ParisTech - CNRS, Institut de Recherche de Chimie Paris , Paris 75005 , France
| | | |
Collapse
|
10
|
Ye F, Boukattaya F, Haddad M, Ratovelomanana-Vidal V, Michelet V. Synthesis of 2-aminopyridines via ruthenium-catalyzed [2+2+2] cycloaddition of 1,6- and 1,7-diynes with cyanamides: scope and limitations. NEW J CHEM 2018. [DOI: 10.1039/c7nj04933a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A direct route to access 2-aminopyridines using Ru-catalyzed-[2+2+2] cycloaddition of 1,n-diynes with cyanamides is described in excellent yields and regioselectivities.
Collapse
Affiliation(s)
- Fei Ye
- PSL
- Research University
- Chimie ParisTech – CNRS
- Institut de Recherche de Chimie Paris
- 75005 Paris
| | - Fatma Boukattaya
- PSL
- Research University
- Chimie ParisTech – CNRS
- Institut de Recherche de Chimie Paris
- 75005 Paris
| | - Mansour Haddad
- PSL
- Research University
- Chimie ParisTech – CNRS
- Institut de Recherche de Chimie Paris
- 75005 Paris
| | | | - Véronique Michelet
- PSL
- Research University
- Chimie ParisTech – CNRS
- Institut de Recherche de Chimie Paris
- 75005 Paris
| |
Collapse
|
11
|
Ye F, Haddad M, Michelet V, Ratovelomanana-Vidal V. Access toward Fluorenone Derivatives through Solvent-Free Ruthenium Trichloride Mediated [2 + 2 + 2] Cycloadditions. Org Lett 2016; 18:5612-5615. [DOI: 10.1021/acs.orglett.6b02840] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Fei Ye
- PSL Research University, Chimie ParisTech, CNRS, Institut de Recherche de Chimie
Paris, Paris 75005, France
| | - Mansour Haddad
- PSL Research University, Chimie ParisTech, CNRS, Institut de Recherche de Chimie
Paris, Paris 75005, France
| | - Véronique Michelet
- PSL Research University, Chimie ParisTech, CNRS, Institut de Recherche de Chimie
Paris, Paris 75005, France
| | | |
Collapse
|