1
|
Khade VV, Bhowmick A, Thube AS, Bhat RG. Direct Access to Strained Fused Dihalo-Aziridino Quinoxalinones via C3-Alkylation Followed by Tandem Cyclization. J Org Chem 2023. [PMID: 37262098 DOI: 10.1021/acs.joc.3c00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Quinoxalinones are a privileged class of compounds, and their structural framework is found in many bioactive compounds, natural compounds, and pharmaceuticals. Quinoxalinone is a promising scaffold for different types of functionalization, and the slight modification of the quinoxalinone skeleton is known to offer a wide range of compounds for drug discovery. Owing to the importance of the quinoxalinone scaffold, we have developed a base-mediated protocol for the C3-alkylation of quinoxalinone followed by tandem cyclization to access novel types of strenuous and fused dihalo-aziridino-quinoxalinone heterocycles via the construction of C-C and C-N bonds. The protocol proved to be simple and practical to access desired fused quinoxalinone heterocycles in excellent yields (up to 98% yield). As an application, the highly functionalized fused dihalo-aziridino-quinoxalinone molecule has been further utilized for mono-dehalogenation under visible light irradiation and selective amide reduction. Moreover, the protocol has also been demonstrated on a gram scale.
Collapse
Affiliation(s)
- Vikas V Khade
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, Maharashtra 411008, India
| | - Anindita Bhowmick
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, Maharashtra 411008, India
| | - Archana S Thube
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, Maharashtra 411008, India
| | - Ramakrishna G Bhat
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, Maharashtra 411008, India
| |
Collapse
|
2
|
Identification of Potential Antitubulin Agents with Anticancer Assets from a Series of Imidazo[1,2- a]quinoxaline Derivatives: In Silico and In Vitro Approaches. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020802. [PMID: 36677860 PMCID: PMC9867416 DOI: 10.3390/molecules28020802] [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: 11/01/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Computer-aided drug design is a powerful and promising tool for drug design and development, with a reduced cost and time. In the current study, we rationally selected a library of 34 fused imidazo[1,2-a]quinoxaline derivatives and performed virtual screening, molecular docking, and molecular mechanics for a lead identification against tubulin as an anticancer molecule. The computational analysis and pharmacophoric features were represented as 1A2; this was a potential lead against tubulin, with a maximized affinity and binding score at the colchicine-binding site of tubulin. The efficiency of this lead molecule was further identified using an in vitro assay on a tubulin enzyme and the anticancer potential was established using an MTT assay. Compound 1A2 (IC50 = 4.33-6.11 µM against MCF-7, MDA-MB-231, HCT-116, and A549 cell lines) displayed encouraging results similar to the standard drug colchicine in these in vitro studies, which further confirmed the effectiveness of CADD in new drug developments. Thus, we successfully applied the utility of in silico techniques to identify the best plausible leads from the fused azaheterocycles.
Collapse
|
3
|
Goel KK, Rajput SK, Kumar A, Nandi NK, Joshi G, Kharb R. Imidazoquinoxaline as a Privileged Fused Pharmacophore in Anticancer Drug Development: A Review of Synthetic Strategies and Medicinal Aspects. ChemistrySelect 2022. [DOI: 10.1002/slct.202200834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Kapil Kumar Goel
- Deparment of Pharmaceutical Sciences Gurukul Kangri (Deemed to be University) Haridwar 249404 India
- Amity Institute of Pharmacy Amity University Noida Uttar Pradesh 201301 India
| | - Satyendra Kumar Rajput
- Deparment of Pharmaceutical Sciences Gurukul Kangri (Deemed to be University) Haridwar 249404 India
| | - Ashwani Kumar
- Deparment of Pharmaceutical Sciences Gurukul Kangri (Deemed to be University) Haridwar 249404 India
| | - Nilay Kumar Nandi
- Department of Pharmaceutical Chemistry ISF College of Pharmacy, Ghal Kalan G.T Road, Moga Punjab India- 142001
| | - Gaurav Joshi
- School of Pharmacy Graphic Era Hill University Dehradun 248002 India
| | - Rajeev Kharb
- Amity Institute of Pharmacy Amity University Noida Uttar Pradesh 201301 India
| |
Collapse
|
4
|
Kozyra P, Krasowska D, Pitucha M. New Potential Agents for Malignant Melanoma Treatment-Most Recent Studies 2020-2022. Int J Mol Sci 2022; 23:6084. [PMID: 35682764 PMCID: PMC9180979 DOI: 10.3390/ijms23116084] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Malignant melanoma (MM) is the most lethal skin cancer. Despite a 4% reduction in mortality over the past few years, an increasing number of new diagnosed cases appear each year. Long-term therapy and the development of resistance to the drugs used drive the search for more and more new agents with anti-melanoma activity. This review focuses on the most recent synthesized anti-melanoma agents from 2020-2022. For selected agents, apart from the analysis of biological activity, the structure-activity relationship (SAR) is also discussed. To the best of our knowledge, the following literature review delivers the latest achievements in the field of new anti-melanoma agents.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Danuta Krasowska
- Department of Dermatology, Venerology and Pediatric Dermatology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| |
Collapse
|
5
|
EAPB0503, an Imidazoquinoxaline Derivative Modulates SENP3/ARF Mediated SUMOylation, and Induces NPM1c Degradation in NPM1 Mutant AML. Int J Mol Sci 2022; 23:ijms23073421. [PMID: 35408798 PMCID: PMC8998649 DOI: 10.3390/ijms23073421] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
Nucleophosmin-1 (NPM1) is a pleiotropic protein involved in numerous cellular processes. NPM1 shuttles between the nucleus and the cytoplasm, but exhibits a predominant nucleolar localization, where its fate and functions are exquisitely controlled by dynamic post-translational modifications (PTM). Sentrin/SUMO Specific Peptidase 3 (SENP3) and ARF are two nucleolar proteins involved in NPM1 PTMs. SENP3 antagonizes ARF-mediated NPM1 SUMOylation, to promote ribosomal biogenesis. In Acute Myeloid Leukemia (AML), NPM1 is frequently mutated, and exhibits an aberrant cytoplasmic localization (NPM1c). NPM1c mutations define a separate AML entity with good prognosis in some AML patients, rendering NPM1c as a potential therapeutic target. SENP3-mediated NPM1 de-SUMOylation induces resistance to therapy in NPM1c AML. Here, we demonstrate that the imidazoquinoxaline EAPB0503 prolongs the survival and results in selective reduction in the leukemia burden of NPM1c AML xenograft mice. Indeed, EAPB0503 selectively downregulates HDM2 expression and activates the p53 pathway in NPM1c expressing cells, resulting in apoptosis. Importantly, we unraveled that NPM1c expressing cells exhibit low basal levels of SUMOylation paralleled with high SENP3 and low ARF basal levels. EAPB0503 reverted these molecular players by inducing NPM1c SUMOylation and ubiquitylation, leading to its proteasomal degradation. EAPB0503-induced NPM1c SUMOylation is concurrent with SENP3 downregulation and ARF upregulation in NPM1c expressing cells. Collectively, these results provide a strong rationale for testing therapies modulating NPM1c post-translational modifications in the management of NPM1c AML.
Collapse
|
6
|
Borah B, Chowhan LR. Recent advances in the transition-metal-free synthesis of quinoxalines. RSC Adv 2021; 11:37325-37353. [PMID: 35496411 PMCID: PMC9043781 DOI: 10.1039/d1ra06942j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/30/2021] [Indexed: 01/04/2023] Open
Abstract
Quinoxalines, also known as benzo[a]pyrazines, constitute an important class of nitrogen-containing heterocyclic compounds as a result of their widespread prevalence in natural products, biologically active synthetic drug candidates, and optoelectronic materials. Owing to their importance and chemists' ever-increasing imagination of new transformations of these products, tremendous efforts have been dedicated to finding more efficient approaches toward the synthesis of quinoxaline rings. The last decades have witnessed a marvellous outburst in modifying organic synthetic methods to create them sustainable for the betterment of our environment. The exploitation of transition-metal-free catalysis in organic synthesis leads to a new frontier to access biologically active heterocycles and provides an alternative method from the perspective of green and sustainable chemistry. Despite notable developments achieved in transition-metal catalyzed synthesis, the high cost involved in the preparation of the catalyst, toxicity, and difficulty in removing it from the final products constitute disadvantageous effects on the atom economy and eco-friendly nature of the transformation. In this review article, we have summarized the recent progress achieved in the synthesis of quinoxalines under transition-metal-free conditions and cover the reports from 2015 to date. This aspect is presented alongside the mechanistic rationalization and limitations of the reaction methodologies. The scopes of future developments are also highlighted.
Collapse
Affiliation(s)
- Biplob Borah
- School of Applied Material Sciences, Centre for Applied Chemistry, Central University of Gujarat Gandhinagar-382030 India
| | - L Raju Chowhan
- School of Applied Material Sciences, Centre for Applied Chemistry, Central University of Gujarat Gandhinagar-382030 India
| |
Collapse
|
7
|
Patil VS, Yadavalli SR, Merugu R, S J S, Devunuri N. One-pot, two-step synthesis of substituted triazoloquinoxalinone starting from 3-hydrazineylquinoxalin-2(1H)-one. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1918171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Vikas S Patil
- Division of Chemistry, Department of Sciences & Humanities, Vignan’s Foundation for Science, Technology & Research (VFSTR) Deemed University, Guntur, India
| | - Subba Rao Yadavalli
- Division of Chemistry, Department of Sciences & Humanities, Vignan’s Foundation for Science, Technology & Research (VFSTR) Deemed University, Guntur, India
| | - Ramchander Merugu
- Department of Bio Chemistry, Mahatma Gandhi University, Nalgonda, India
| | - Swamy S J
- Department of Chemistry, Kakatiya University, Warangal, India
| | - Nagaraju Devunuri
- Division of Chemistry, Department of Sciences & Humanities, Vignan’s Foundation for Science, Technology & Research (VFSTR) Deemed University, Guntur, India
| |
Collapse
|
8
|
Patinote C, Deleuze-Masquéfa C, Kaddour KH, Vincent LA, Larive R, Zghaib Z, Guichou JF, Assaf MD, Cuq P, Bonnet PA. Imidazo[1,2-a]quinoxalines for melanoma treatment with original mechanism of action. Eur J Med Chem 2020; 212:113031. [PMID: 33309473 DOI: 10.1016/j.ejmech.2020.113031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 11/14/2020] [Indexed: 10/22/2022]
Abstract
The malignant transformation of melanocytes causes several thousand deaths each year, making melanoma an important public health concern. Melanoma is the most aggressive skin cancer, which incidence has regularly increased over the past decades. We described here the preparation of new compounds based on the 1-(3,4-dihydroxyphenyl)imidazo[1,2-a]quinoxaline structure. Different positions of the quinoxaline moiety were screened to introduce novel substituents in order to study their influence on the biological activity. Several alkylamino or alkyloxy groups were also considered to replace the methylamine of our first generation of Imiqualines. Imidazo[1,2-a]pyrazine derivatives were also designed as potential minimal structure. The investigation on A375 melanoma cells displayed interesting in vitro low nanomolar cytotoxic activity. Among them, 9d (EAPB02303) is particularly remarkable since it is 20 times more potent than vemurafenib, the reference clinical therapy used on BRAF mutant melanoma. Contrary to the first generation, EAPB02303 does not inhibit tubulin polymerization, as confirmed by an in vitro assay and a molecular modelisation study. The mechanism of action for EAPB02303 highlighted by a transcriptomic analysis is clearly different from a panel of 12 well-known anticancer drugs. In vivoEAPB02303 treatment reduced tumor size and weight of the A375 human melanoma xenografts in a dose-dependent manner, correlated with a low mitotic index but not with necrosis.
Collapse
Affiliation(s)
- Cindy Patinote
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France; Société d'Accélération du Transfert de Technologies (SATT AxLR), CSU, 950 rue Saint Priest, 34090, Montpellier, France
| | - Carine Deleuze-Masquéfa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France.
| | - Kamel Hadj Kaddour
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Laure-Anaïs Vincent
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Romain Larive
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Zahraa Zghaib
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Jean-François Guichou
- CNRS, UMR 5048, INSERM, U105, Université de Montpellier, Centre de Biochimie Structurale, 34090, Montpellier, France
| | - Mona Diab Assaf
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Pierre Cuq
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| |
Collapse
|
9
|
Patinote C, Cirnat N, Hadj-Kaddour K, Cuq P, Bonnet PA, Deleuze-Masquéfa C. Substantial Cellular Penetration of Fluorescent Imidazoquinoxalines. J Fluoresc 2020; 30:1499-1512. [PMID: 32778995 DOI: 10.1007/s10895-020-02595-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/27/2020] [Indexed: 11/26/2022]
Abstract
Fluorescent tools have revolutionized our capability to visualize, probe, study, and understand the biological cellular properties, processes and dynamics, enabling researchers to improve their knowledge for example in cancer field. In this paper, we use the peculiar properties of our Imiqualines derivatives to study their cellular penetration and distribution in a human melanoma cell line A375 using confocal microscopy. Preliminary results on colocalization with the potent protein target c-Kit of our lead are also described.
Collapse
Affiliation(s)
- Cindy Patinote
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Natalina Cirnat
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Pierre Cuq
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | |
Collapse
|
10
|
Kalinin AА, Islamova LN, Fazleeva GM. Imidazo[A]Quinoxalines: New Approaches to Synthesis and Biological Activity. Chem Heterocycl Compd (N Y) 2020. [DOI: 10.1007/s10593-020-02716-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
In vitro cytotoxic activity of thiazole-indenoquinoxaline hybrids as apoptotic agents, design, synthesis, physicochemical and pharmacokinetic studies. Bioorg Chem 2020; 100:103951. [DOI: 10.1016/j.bioorg.2020.103951] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/23/2020] [Accepted: 05/14/2020] [Indexed: 01/09/2023]
|
12
|
Wu M, Tian Y, Wang Q, Guo C. Gout: a disease involved with complicated immunoinflammatory responses: a narrative review. Clin Rheumatol 2020; 39:2849-2859. [PMID: 32382830 DOI: 10.1007/s10067-020-05090-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Gout is a disease with acute and/or chronic inflammation and tissue damage induced by the precipitation of monosodium urate crystal (MSU) crystals in bone joints, kidneys, and subcutaneous sites. In recent years, with the continuous research on gout animal models and patient clinical investigations, the mechanism of inflammation activation of gout has been further discovered. Studies have shown that pro-inflammatory factors such as interleukin (IL)-1β, IL-8 and IL-17, NLRP3 inflammasome, and tumor necrosis factor alpha (TNF-α), anti-inflammatory factors such as IL-10, IL-37 are all involved in the MSU-induced gout inflammatory process. And the immune cells in gout, including neutrophils, monocytes/macrophages, and lymphocytes, all play important roles in the pathogenesis of gout. In this review, we mainly emphasize the understanding of various cytokines, inflammasome, and immune cells involved in the onset of gout, in order to provide a systematic and theoretical basis for the novel exploration of inflammatory therapeutic targets for gout.
Collapse
Affiliation(s)
- Meimei Wu
- Southern Medical University Second Clinical College, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China
| | - Ye Tian
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Guangdong Medical University Shenzhen Baoan Clinical College, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Shenzhen University Second affiliated Hospital, Shenzhen, 518101, China
| | - Qianqian Wang
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Guangdong Medical University Shenzhen Baoan Clinical College, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Shenzhen University Second affiliated Hospital, Shenzhen, 518101, China
| | - Chengshan Guo
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China.
- Department of Rheumatology and Immunology, Guangdong Medical University Shenzhen Baoan Clinical College, Shenzhen, 518101, China.
- Department of Rheumatology and Immunology, Shenzhen University Second affiliated Hospital, Shenzhen, 518101, China.
| |
Collapse
|
13
|
Patinote C, Karroum NB, Moarbess G, Cirnat N, Kassab I, Bonnet PA, Deleuze-Masquéfa C. Agonist and antagonist ligands of toll-like receptors 7 and 8: Ingenious tools for therapeutic purposes. Eur J Med Chem 2020; 193:112238. [PMID: 32203790 PMCID: PMC7173040 DOI: 10.1016/j.ejmech.2020.112238] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
The discovery of the TLRs family and more precisely its functions opened a variety of gates to modulate immunological host responses. TLRs 7/8 are located in the endosomal compartment and activate a specific signaling pathway in a MyD88-dependant manner. According to their involvement into various autoimmune, inflammatory and malignant diseases, researchers have designed diverse TLRs 7/8 ligands able to boost or block the inherent signal transduction. These modulators are often small synthetic compounds and most act as agonists and to a much lesser extent as antagonists. Some of them have reached preclinical and clinical trials, and only one has been approved by the FDA and EMA, imiquimod. The key to the success of these modulators probably lies in their combination with other therapies as recently demonstrated. We gather in this review more than 360 scientific publications, reviews and patents, relating the extensive work carried out by researchers on the design of TLRs 7/8 modulators, which are classified firstly by their biological activities (agonist or antagonist) and then by their chemical structures, which total syntheses are not discussed here. This review also reports about 90 clinical cases, thereby showing the biological interest of these modulators in multiple pathologies.
Collapse
Affiliation(s)
- Cindy Patinote
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Nour Bou Karroum
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Georges Moarbess
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Natalina Cirnat
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Issam Kassab
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | | | | |
Collapse
|
14
|
Khan MF, Verma G, Akhtar W, Shaquiquzzaman M, Akhter M, Rizvi MA, Alam MM. Pharmacophore modeling, 3D-QSAR, docking study and ADME prediction of acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
15
|
Chen C, Yu TY, Wu JH, Ariraman M, Juang TY, Abu-Omar MM, Lin CH. Synthesis and Properties of Quinoxaline-Containing Benzoxazines and Polybenzoxazines. ACS OMEGA 2019; 4:9092-9101. [PMID: 31459997 PMCID: PMC6648507 DOI: 10.1021/acsomega.9b01042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/07/2019] [Indexed: 06/10/2023]
Abstract
The object of this work is to prepare quinoxaline-based benzoxazines and evaluate thermal properties of their thermosets. For this object, 4,4'-(quinoxaline-2,3-diyl)diphenol (QDP)/furfurylamine-based benzoxazine (QDP-fu) and 4,4',4″,4‴-([6,6'-biquinoxaline]-2,2',3,3'-tetrayl)tetraphenol (BQTP)/furfurylamine-based benzoxazine (BQTP-fu) were prepared. The structures of QDP-fu and BQTP-fu were successfully confirmed by FTIR and 1H and 13C NMR spectra. We studied the curing behavior of QDP-fu and BQTP-fu and thermal properties of their thermosets. According to DSC thermograms, QDP-fu and BQTP-fu have the attractive onset exothermic temperatures of 181 and 186 °C, respectively. The onset temperature is approximately 45 °C lower than that of a bisphenol A/furfurylamine-based benzoxazines. According to DMA TMA and TGA thermograms, the thermoset of BQTP-fu shows impressive thermal properties, with a T g value of 418 °C, a coefficient of thermal expansion of 39 ppm/°C, a 5% decomposition temperature of 430 °C, and a char yield of 72%.
Collapse
Affiliation(s)
- Chien
Han Chen
- Advanced
Research Center for Green Materials Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Tsung Yen Yu
- Department
of Chemical Engineering, National Chung
Hsing University, Taichung 402, Taiwan
| | - Jen-Hao Wu
- Department of Cosmeceutics and School of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Mathivathanan Ariraman
- Department
of Chemical Engineering, National Chung
Hsing University, Taichung 402, Taiwan
| | - Tzong-Yuan Juang
- Department of Cosmeceutics and School of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Mahdi M. Abu-Omar
- Department
of Chemistry and Biochemistry, University
of California, Santa Barbara, Santa
Barbara, California 93106, United States
| | - Ching-Hsuan Lin
- Advanced
Research Center for Green Materials Science and Technology, National Taiwan University, Taipei 10617, Taiwan
- Department
of Chemical Engineering, National Chung
Hsing University, Taichung 402, Taiwan
| |
Collapse
|
16
|
Design and Synthesis of New Quinoxaline Derivatives as Anticancer Agents and Apoptotic Inducers. Molecules 2019; 24:molecules24061175. [PMID: 30934622 PMCID: PMC6470675 DOI: 10.3390/molecules24061175] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/12/2022] Open
Abstract
The quinoxaline scaffold is a promising platform for the discovery of active chemotherapeutic agents. Three series of quinoxaline derivatives were synthesized and biologically evaluated against three tumor cell lines (HCT116 human colon carcinoma, HepG2, liver hepatocellular carcinoma and MCF-7, human breast adenocarcinoma cell line), in addition to VEGFR-2 enzyme inhibition activity. Compounds VIId, VIIIa, VIIIc, VIIIe and XVa exhibited promising activity against the tested cell lines and weak activity against VEGFR-2. Compound VIIIc induced a significant disruption in the cell cycle profile and cell cycle arrest at the G2/M phase boundary. In further assays, the cytotoxic effect of the highly active compounds was determined using a normal Caucasian fibroblast-like fetal lung cell line (WI-38). Compound VIIIc could be considered as a lead compound that merits further optimization and development as an anti-cancer and an apoptotic inducing candidate against the HCT116 cell line.
Collapse
|
17
|
Babu LT, Jadhav GR, Paira P. Luminescent bis(benzo[ d]thiazolyl)quinoxaline: facile synthesis, nucleic acid and protein BSA interaction, live-cell imaging, biopharmaceutical research and cancer theranostic application. RSC Adv 2019; 9:8748-8752. [PMID: 35517674 PMCID: PMC9061853 DOI: 10.1039/c9ra01498e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/31/2022] Open
Abstract
A series of quinoxaline-2-hydroxyphenylbenzothiazole scaffolds were synthesized and characterized using NMR, UV, fluorescence spectroscopy and LCMS. These newly synthesized compounds were found to be cytotoxic in human epithelioid cervix carcinoma (HeLa) and human colon cancer cell lines (Caco-2). Selectivity of the compounds 7e and 7g are more than 9 fold higher in Caco-2 cells with respect to the normal cell line HEK-293. The most fluorescent compound 7e has displayed high cytoselectivity, significant cellular uptake in HeLa cells and strong binding efficacy with DNA and BSA. The most potent compound 7g has primarily classified as BCS class 4 and BDDCS class 4. A series of luminescent bis(benzo[d]thiazolyl)quinoxalines have been synthesized and their fluorescence properties, anticancer potency, DNA and BSA interactions, cellular uptake, and metabolic stabilities are investigated.![]()
Collapse
Affiliation(s)
- Lavanya Thilak Babu
- Department of Chemistry
- School of Advanced Sciences
- Vellore Institute of Technology
- Vellore-632014
- India
| | - Gajanan Raosaheb Jadhav
- Department of Chemistry
- School of Advanced Sciences
- Vellore Institute of Technology
- Vellore-632014
- India
| | - Priyankar Paira
- Department of Chemistry
- School of Advanced Sciences
- Vellore Institute of Technology
- Vellore-632014
- India
| |
Collapse
|
18
|
Montana M, Mathias F, Terme T, Vanelle P. Antitumoral activity of quinoxaline derivatives: A systematic review. Eur J Med Chem 2018; 163:136-147. [PMID: 30503938 DOI: 10.1016/j.ejmech.2018.11.059] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of death and a major health problem worldwide. While many effective anticancer agents are available, the majority of drugs currently on the market are not specific, raising issues like the common side effects of chemotherapy. However, recent research hold promise for the development of more efficient and safer anticancer drugs. Quinoxaline and its derivatives are becoming recognized as a novel class of chemotherapeutic agents with activity against different tumors. The present review compiles and discusses studies concerning the therapeutic potential of the anticancer activity of quinoxaline derivatives, covering articles published between July 2013 and July 2018.
Collapse
Affiliation(s)
- M Montana
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR, UMR 7273, Laboratoire de Pharmaco-Chimie Radicalaire, Marseille, France; Assistance Publique-Hôpitaux de Marseille (AP-HM), Oncopharma, Hôpital Nord, Marseille, France
| | - F Mathias
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR, UMR 7273, Laboratoire de Pharmaco-Chimie Radicalaire, Marseille, France
| | - T Terme
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR, UMR 7273, Laboratoire de Pharmaco-Chimie Radicalaire, Marseille, France
| | - P Vanelle
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR, UMR 7273, Laboratoire de Pharmaco-Chimie Radicalaire, Marseille, France; Assistance Publique-Hôpitaux de Marseille (AP-HM), Service Central de la qualité et de l'information pharmaceutiques (SCQIP), Marseille, France.
| |
Collapse
|
19
|
El Hajj R, Bou Youness H, Lachaud L, Bastien P, Masquefa C, Bonnet PA, El Hajj H, Khalifeh I. EAPB0503: An Imiquimod analog with potent in vitro activity against cutaneous leishmaniasis caused by Leishmania major and Leishmania tropica. PLoS Negl Trop Dis 2018; 12:e0006854. [PMID: 30462645 PMCID: PMC6248897 DOI: 10.1371/journal.pntd.0006854] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/18/2018] [Indexed: 12/16/2022] Open
Abstract
Cutaneous Leishmaniasis (CL) is a parasitic infection classified by the WHO as one of the most uncontrolled spreading neglected diseases. Syria is endemic for Leishmania tropica and Leishmania major, causing CL in the Eastern Mediterranean. The large-scale displacement of Syrian refugees exacerbated the spread of CL into neighboring countries. Therapeutic interventions against CL include local, systemic and physical treatments. The high risk for drug-resistance to current treatments stresses the need for new therapies. Imiquimod is an immunomodulatory drug with a tested efficacy against L. major species. Yet, Imiquimod efficacy against L. tropica and the molecular mechanisms dictating its potency are still underexplored. In this study, we characterized the effect of Imiquimod against L. tropica and L. major, and characterized the molecular mechanisms dictating its anti-leishmanial efficacy against both strains. We also investigated the potency and molecular mechanisms of an Imiquimod analog, EAPB0503, against these two strains. We have tested the effect of Imiquimod and EAPB0503 on macrophages infected with either L. major, L. tropica strains, or patient-derived freshly isolated L. tropica parasites. The anti-amastigote activity of either drugs was assessed by quantitative real time PCR (RT-PCR) using kinetoplast specific primers, confocal microscopy using the Glycoprotein 63 (Gp63) Leishmania amastigote antibody or by histology staining. The mechanism of action of either drugs on the canonical nuclear factor kappa- B (NF-κB) pathway was determined by western blot, and confocal microscopy. The immune production of cytokines upon treatment of infected macrophages with either drugs was assessed by ELISA. Both drugs reduced amastigote replication. EAPB0503 proved more potent, particularly on the wild type L. tropica amastigotes. Toll-Like Receptor-7 was upregulated, mainly by Imiquimod, and to a lesser extent by EAPB0503. Both drugs activated the NF-κB canonical pathway triggering an immune response and i-NOS upregulation in infected macrophages. Our findings establish Imiquimod as a strong candidate for treating L. tropica and show the higher potency of its analog EAPB0503 against CL. Cutaneous Leishmaniasis (CL) is a parasitic infection caused by Leishmania (L.) parasites. In the Old World and the Near East, CL is mainly caused by L. major and L. tropica. The ongoing Syrian war and the resulting massive population displacement led to an alarming increase in the incidence of CL, in Syria and its surrounding countries. Current therapies against CL lead to partial or complete cure in L. major infections but are less effective against L. tropica. These therapies associate with several limitations, including patients’age, immune system, repetitive painful injections, high cost, poor availability, and mainly systemic toxicity. Therefore, it is of high interest to seek for novel drugs against CL. We assessed the activity of an immunomodulatory drug and its analog against L. major and L. tropica parasites and showed their potency. Importantly, the analog proved more efficient against the wild type L. tropica strain. These results highlight the promising efficacy of immuno-modulatory drugs against CL.
Collapse
Affiliation(s)
- Rana El Hajj
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - Hanady Bou Youness
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - Laurence Lachaud
- Centre Hospitalo-Universitaire, Université de Montpellier, Montpellier, France
| | - Patrick Bastien
- Centre Hospitalo-Universitaire, Université de Montpellier, Montpellier, France
| | - Carine Masquefa
- Faculté de Pharmacie, Université de Montpellier, Montpellier, France
| | | | - Hiba El Hajj
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
- * E-mail: (HEH); (IK)
| | - Ibrahim Khalifeh
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
- * E-mail: (HEH); (IK)
| |
Collapse
|
20
|
Imidazo[1,2- a]quinoxalines Derivatives Grafted with Amino Acids: Synthesis and Evaluation on A375 Melanoma Cells. Molecules 2018; 23:molecules23112987. [PMID: 30445763 PMCID: PMC6278480 DOI: 10.3390/molecules23112987] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022] Open
Abstract
Imiqualines (imidazoquinoxaline derivatives) are anticancer compounds with high cytotoxic activities on melanoma cell lines. The first generation of imiqualines, with two lead compounds (EAPB0203 and EAPB0503), shows remarkable in vitro (IC50 = 1 570 nM and IC50 = 200 nM, respectively, on the A375 melanoma cell line) and in vivo activity on melanoma xenografts. The second generation derivatives, EAPB02302 and EAPB02303, are more active, with IC50 = 60 nM and IC50 = 10 nM, respectively, on A375 melanoma cell line. The aim of this study was to optimize the bioavailability of imiqualine derivatives, without losing their intrinsic activity. For that, we achieved chemical modulation on the second generation of imiqualines by conjugating amino acids on position 4. A new series of twenty-five compounds was efficiently synthesized by using microwave assistance and tested for its activity on the A375 cell line. In the new series, compounds 11a, 9d and 11b show cytotoxic activities less than second generation compounds, but similar to that of the first generation ones (IC50 = 403 nM, IC50 = 128 nM and IC50 = 584 nM, respectively). The presence of an amino acid leads to significant enhancement of the water solubility for improved drugability.
Collapse
|
21
|
Ghanbarimasir Z, Bekhradnia A, Morteza-Semnani K, Rafiei A, Razzaghi-Asl N, Kardan M. Design, synthesis, biological assessment and molecular docking studies of new 2-aminoimidazole-quinoxaline hybrids as potential anticancer agents. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 194:21-35. [PMID: 29310028 DOI: 10.1016/j.saa.2017.12.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/17/2017] [Accepted: 12/20/2017] [Indexed: 06/07/2023]
Abstract
In a search for novel antiproliferative agents, a series of quinoxaline derivatives containing 2-aminoimidazole (8a-8x) were designed and synthesized. The structures of synthesized compounds were confirmed by IR, 1H NMR, 13C NMR, Mass Spectroscopy and analyzed using HSQC, COSY, ROESY, HMBC techniques. The anticancer activity of all derivatives were evaluated for colon cancer and breast cancer cell lines by the MTT assay and acridine orange/ethidium bromide double staining method. The anti-cancer effect in human colon cancer (HCT-116) and breast cancer (MCF-7) cell lines exhibited that compounds 8a, 8s, 8t, 8w, 8x appeared as potent antiproliferative agents and especially inhibited the human colon cancer cell proliferation with percentage of inhibition by over 50%. The most active compound was (E)-4-phenyl-1-((quinoxalin-2-ylmethylene)amino)-1H-imidazol-2-amine (8a) with the highest inhibition for MCF-7 (83.3%) and HCT-116 (70%) cell lines after 48 and 24h, respectively. Molecular docking studies of these derivatives within c-kit active site as a validated target might be suggested them as appropriate candidates for further efforts toward more potent anticancer compounds.
Collapse
Affiliation(s)
- Zahra Ghanbarimasir
- Student Research Committee, Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmadreza Bekhradnia
- Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Katayoun Morteza-Semnani
- Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Rafiei
- Molecular and Cell Biology Research Center, Hemoglobinopathy Institute, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nima Razzaghi-Asl
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mostafa Kardan
- Molecular and Cell Biology Research Center, Hemoglobinopathy Institute, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
22
|
Liquid chromatography-electrospray ionization-tandem mass spectrometry method for quantitative estimation of new imiqualine leads with potent anticancer activities in rat and mouse plasma. Application to a pharmacokinetic study in mice. J Pharm Biomed Anal 2018; 148:369-379. [PMID: 29111492 DOI: 10.1016/j.jpba.2017.10.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/19/2017] [Accepted: 10/22/2017] [Indexed: 11/22/2022]
Abstract
Imidazoquinoxaline derivatives (imiqualines) are a new series of anticancer compounds. Two lead compounds (EAPB0203 and EAPB0503) with remarkable in vitro and in vivo activity on melanoma and T-cell lymphomas have been previously identified. The modulation of the chemical structure of the most active compound, EAPB0503, has led to the synthesis of two compounds, EAPB02302 and EAPB02303, 7 and 40 times more active than EAPB0503 against A375 human melanoma cancer cell line, respectively. The aim of this study was to develop and validate a sensitive and accurate liquid chromatography-electrospray ionization-tandem mass spectrometry method to simultaneously quantify EAPB02303 and its potential active metabolite, EAPB02302, in rat and mouse plasma. Analytes were detected in multiple reaction monitoring acquisition mode using an electrospray ionization detector in positive ion mode. Following a liquid-liquid extraction with ethyl acetate, analytes and internal standard were separated by HPLC reversed-phase on a C18 RP18 Nucleoshell column (2.7μm, 4.6×100mm). The method was validated according to FDA and EMA Bioanalytical Method Validation guidelines. The robustness of the method was assessed by introducing small variations in nine nominal analytical parameters. Statistical interpretation was performed by mean of the Student's t-test. Standard curves were generated via unweighted quadratic regression of calibrators (EAPB02303: 1.95-1000ng/mL, EAPB02302: 7.81-1000ng/mL in rat plasma; EAPB02303: 0.98-1000ng/mL, EAPB02302: 1.95-1000ng/mL in mouse plasma). From the analysis of QC samples, intra- and inter-assay precision and accuracy studies demonstrated %R.S.Ds. <12.5% and percent deviation from nominal concentration <7%. Matrix effects (mean matrix factors from 91.8-108.5% in rat plasma; and from 90.4-102.4% in mouse plasma) and stability assays (recoveries >87%) were acceptable and in accordance with the guidelines. No quantifiable carryover effect was observed. The LLOQs were 1.95ng/mL for EAPB02303 and 7.81ng/mL for EAPB02302 in rat plasma, and 0.98ng/mL and 1.95ng/mL for the two compounds in mouse plasma, respectively. This method was successfully implemented to support a mouse pharmacokinetic study following a single intraperitoneal administration of EAPB02303 in male C57Bl/6 mice. The obtained pharmacokinetic parameters of EAPB02303 would be useful to optimize the dosing and the rhythm of administration for subsequent preclinical in vivo activity studies.
Collapse
|
23
|
Joshi G, Chauhan M, Kumar R, Thakur A, Sharma S, Singh R, Wani AA, Sharon A, Bharatam PV, Kumar R. Cyclocondensation reactions of an electron deactivated 2-aminophenyl tethered imidazole with mono/1,2-biselectrophiles: synthesis and DFT studies on the rationalisation of imidazo[1,2-a]quinoxaline versus benzo[f]imidazo[1,5-a][1,3,5]triazepine selectivity switches. Org Chem Front 2018. [DOI: 10.1039/c8qo00706c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Microwave-assisted cyclocondensation of title compounds afforded unreported imidazo[1,2-a] quinoxaline and benzo[f]imidazo[1,5-a][1,3,5]triazepines in high yields.
Collapse
Affiliation(s)
- Gaurav Joshi
- Department of Pharmaceutical Sciences and Natural Products
- Central University of Punjab
- Bathinda-151001
- India
| | - Monika Chauhan
- Department of Pharmaceutical Sciences and Natural Products
- Central University of Punjab
- Bathinda-151001
- India
| | - Rakesh Kumar
- Department of Pharmaceutical Sciences and Natural Products
- Central University of Punjab
- Bathinda-151001
- India
| | - Ankush Thakur
- Department of Pharmaceutical Sciences and Natural Products
- Central University of Punjab
- Bathinda-151001
- India
| | - Sachin Sharma
- Department of Pharmaceutical Sciences and Natural Products
- Central University of Punjab
- Bathinda-151001
- India
| | - Rajveer Singh
- Department of Pharmaceutical Chemistry
- I.S.F. College of Pharmacy
- Moga
- India
| | - Aabid Abdullah Wani
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- S.A.S. Nagar
- India
| | - Ashoke Sharon
- Department of Chemistry
- Birla Institute of Technology
- Ranchi
- India
| | - Prasad V. Bharatam
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- S.A.S. Nagar
- India
| | - Raj Kumar
- Department of Pharmaceutical Sciences and Natural Products
- Central University of Punjab
- Bathinda-151001
- India
- Department of Pharmaceutical Chemistry
| |
Collapse
|
24
|
Imidazo[1,2- a ]pyrazine, Imidazo[1,5- a ]quinoxaline and Pyrazolo[1,5- a ]quinoxaline derivatives as IKK1 and IKK2 inhibitors. Eur J Med Chem 2017; 138:909-919. [DOI: 10.1016/j.ejmech.2017.07.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/02/2017] [Accepted: 07/13/2017] [Indexed: 01/04/2023]
|
25
|
Courbet A, Bec N, Constant C, Larroque C, Pugniere M, El Messaoudi S, Zghaib Z, Khier S, Deleuze-Masquefa C, Gattacceca F. Imidazoquinoxaline anticancer derivatives and imiquimod interact with tubulin: Characterization of molecular microtubule inhibiting mechanisms in correlation with cytotoxicity. PLoS One 2017; 12:e0182022. [PMID: 28797090 PMCID: PMC5552358 DOI: 10.1371/journal.pone.0182022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 07/11/2017] [Indexed: 11/19/2022] Open
Abstract
Displaying a strong antiproliferative activity on a wide variety of cancer cells, EAPB0203 and EAPB0503 belong to the imidazo[1,2-a]quinoxalines family of imiquimod structural analogues. EAPB0503 has been shown to inhibit tubulin polymerization. The aim of the present study is to characterize the interaction of EAPB0203 and EAPB0503 with tubulin. We combine experimental approaches at the cellular and the molecular level both in vitro and in silico in order to evaluate the interaction of EAPB0203 and EAPB0503 with tubulin. We examine the influence of EAPB0203 and EAPB0503 on the cell cycle and fate, explore the binding interaction with purified tubulin, and use a computational molecular docking model to determine the binding modes to the microtubule. We then use a drug combination study with other anti-microtubule agents to compare the binding site of EAPB0203 and EAPB0503 to known potent tubulin inhibitors. We demonstrate that EAPB0203 and EAPB0503 are capable of blocking human melanoma cells in G2 and M phases and inducing cell death and apoptosis. Second, we show that EAPB0203 and EAPB0503, but also unexpectedly imiquimod, bind directly to purified tubulin and inhibit tubulin polymerization. As suggested by molecular docking and binding competition studies, we identify the colchicine binding site on β-tubulin as the interaction pocket. Furthermore, we find that EAPB0203, EAPB0503 and imiquimod display antagonistic cytotoxic effect when combined with colchicine, and disrupt tubulin network in human melanoma cells. We conclude that EAPB0203, EAPB0503, as well as imiquimod, interact with tubulin through the colchicine binding site, and that the cytotoxic activity of EAPB0203, EAPB0503 and imiquimod is correlated to their tubulin inhibiting effect. These compounds appear as interesting anticancer drug candidates as suggested by their activity and mechanism of action, and deserve further investigation for their use in the clinic.
Collapse
Affiliation(s)
- Alexis Courbet
- University of Montpellier, Montpellier, France
- Department of Biochemistry & Institute for Protein Design, University of Washington, Seattle, WA, United States of America
| | - Nicole Bec
- University of Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Caroline Constant
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
| | - Christian Larroque
- University of Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Martine Pugniere
- University of Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Safia El Messaoudi
- University of Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Zahraa Zghaib
- IBMM, Institut des Biomolécules Max Mousseron, UMR 5247, Université de Montpellier, Montpellier, France
| | - Sonia Khier
- University of Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Carine Deleuze-Masquefa
- IBMM, Institut des Biomolécules Max Mousseron, UMR 5247, Université de Montpellier, Montpellier, France
| | - Florence Gattacceca
- University of Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
26
|
Gu W, Wang S, Jin X, Zhang Y, Hua D, Miao T, Tao X, Wang S. Synthesis and Evaluation of New Quinoxaline Derivatives of Dehydroabietic Acid as Potential Antitumor Agents. Molecules 2017; 22:molecules22071154. [PMID: 28696365 PMCID: PMC6152277 DOI: 10.3390/molecules22071154] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/04/2017] [Indexed: 01/19/2023] Open
Abstract
A series of new quinoxaline derivatives of dehydroabietic acid (DAA) were designed and synthesized as potential antitumor agents. Their structures were characterized by IR, 1H-NMR, 13C-NMR, and MS spectra and elemental analyses. All the new compounds were screened for their in vitro antiproliferative activities against three human cancer cell lines (MCF-7, SMMC-7721 and HeLa) and noncancerous human hepatocyte cells (LO2). A cytotoxic assay manifested that compound 4b showed the most potent cytotoxic activity against the three cancer cell lines, with IC50 values of 1.78 ± 0.36, 0.72 ± 0.09 and 1.08 ± 0.12 μM, respectively, and a substantially lower cytotoxicity to LO2 cells (IC50: 11.09 ± 0.57 μM). Moreover, the cell cycle analysis suggested that compound 4b caused cell cycle arrest of SMMC-7721 cells at the G0/G1 phase. In a Hoechst 33258 staining assay, compound 4b caused considerable morphological changes of the nuclei of SMMC-7721 cells, correlated with cell apoptosis. In addition, an Annexin V-FITC/PI dual staining assay confirmed that compound 4b could induce the apoptosis of SMMC-7721 cells in a dose-dependent manner.
Collapse
Affiliation(s)
- Wen Gu
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Shuang Wang
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Xiaoyan Jin
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Yaliang Zhang
- The State Key Lab of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China.
| | - Dawei Hua
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Tingting Miao
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Xubing Tao
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Shifa Wang
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
27
|
Patinote C, Hadj-Kaddour K, Damian M, Deleuze-Masquéfa C, Cuq P, Bonnet PA. Fluorescence Study of Imidazoquinoxalines. J Fluoresc 2017; 27:1607-1611. [DOI: 10.1007/s10895-017-2097-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/04/2017] [Indexed: 11/28/2022]
|
28
|
Nabbouh AI, Hleihel RS, Saliba JL, Karam MM, Hamie MH, Wu HCJM, Berthier CP, Tawil NM, Bonnet PAA, Deleuze-Masquefa C, El Hajj HA. Imidazoquinoxaline derivative EAPB0503: A promising drug targeting mutant nucleophosmin 1 in acute myeloid leukemia. Cancer 2017; 123:1662-1673. [PMID: 28055106 DOI: 10.1002/cncr.30515] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/10/2016] [Accepted: 11/17/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Nucleophosmin 1 (NPM1) is a nucleocytoplasmic shuttling protein mainly localized in the nucleolus. NPM1 is frequently mutated in acute myeloid leukemia (AML). NPM1c oligomerizes with wild-type nucleophosmin 1 (wt-NPM1), and this leads to its continuous cytoplasmic delocalization and contributes to leukemogenesis. Recent studies have shown that Cytoplasmic NPM1 (NPM1c) degradation leads to growth arrest and apoptosis of NPM1c AML cells and corrects wt-NPM1 normal nucleolar localization. METHODS AML cells expressing wt-NPM1 or NPM1c or transfected with wt-NPM1 or NPM1c as well as wt-NPM1 and NPM1c AML xenograft mice were used. Cell growth was assessed with trypan blue or a CellTiter 96 proliferation kit. The cell cycle was studied with a propidium iodide (PI) assay. Caspase-mediated intrinsic apoptosis was assessed with annexin V/PI, the mitochondrial membrane potential, and poly(adenosine diphosphate ribose) polymerase cleavage. The expression of NPM1, p53, phosphorylated p53, and p21 was analyzed via immunoblotting. Localization was performed with confocal microscopy. The leukemia burden was evaluated by flow cytometry with an anti-human CD45 antibody. RESULTS The imidazoquinoxaline 1-(3-methoxyphenyl)-N-methylimidazo[1,2-a]quinoxalin-4-amine (EAPB0503) induced selective proteasome-mediated degradation of NPM1c, restored wt-NPM1 nucleolar localization in NPM1c AML cells, and thus yielded selective growth arrest and apoptosis. Introducing NPM1c to cells normally harboring wt-NPM1 sensitized them to EAPB0503 and led to their growth arrest. Moreover, EAPB0503 selectively reduced the leukemia burden in NPM1c AML xenograft mice. CONCLUSIONS These findings further reinforce the idea of targeting the NPM1c oncoprotein to eradicate leukemic cells and warrant a broader preclinical evaluation and then a clinical evaluation of this promising drug. Cancer 2017;123:1662-1673. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Ali I Nabbouh
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon.,Max Mousseron Institute of Biomolecules, Faculty of Pharmacy, Montpellier University, Montpellier, France
| | - Rita S Hleihel
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon.,Department of Cell Biology, Anatomy, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Jessica L Saliba
- Department of Biology, Faculty of Science, Lebanese University, Beirut, Lebanon
| | - Martin M Karam
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Microbiology, and Immunology, American University of Beirut, Beirut, Lebanon
| | - Maguy H Hamie
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Microbiology, and Immunology, American University of Beirut, Beirut, Lebanon
| | - Hsin-Chieh J M Wu
- National Institute of Health and Medical Research Unit 944, Collège de France, Paris, France
| | - Caroline P Berthier
- National Institute of Health and Medical Research Unit 944, Collège de France, Paris, France
| | - Nadim M Tawil
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Pierre-Antoine A Bonnet
- Max Mousseron Institute of Biomolecules, Faculty of Pharmacy, Montpellier University, Montpellier, France
| | - Carine Deleuze-Masquefa
- Max Mousseron Institute of Biomolecules, Faculty of Pharmacy, Montpellier University, Montpellier, France
| | - Hiba A El Hajj
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Microbiology, and Immunology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
29
|
Chouchou A, Aubert-Pouëssel A, Dorandeu C, Zghaib Z, Cuq P, Devoisselle JM, Bonnet PA, Bégu S, Deleuze-Masquefa C. Lipid nanocapsules formulation and cellular activities evaluation of a promising anticancer agent: EAPB0503. Int J Pharm Investig 2017; 7:155-163. [PMID: 29692974 PMCID: PMC5903019 DOI: 10.4103/jphi.jphi_53_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective: EAPB0503, lead compound of imiqualines, presented high antitumor activities but also a very low water solubility which was critical for further preclinical studies. To apply to EAPB0503, a robust and safe lipid formulation already used for poor soluble anticancer agents for injectable administration at a concentration higher than 1 mg/mL. Materials and Methods: Physicochemical properties of EAPB0503 were determined to consider an adapted formulation. In a second time, lipid nanocapsules (LNC) formulations based on the phase-inversion process were developed for EAPB0503 encapsulation. Then, EAPB0503 loaded-LNC were tested in vitro on different cell lines and compared to standard EAPB0503 solutions. Results: Optimized EAPB0503 LNC displayed an average size of 111.7 ± 0.9 nm and a low polydispersity index of 0.059 ± 0.002. The obtained loading efficiency was higher than 96% with a drug loading of 1.7 mg/mL. A stability study showed stability during 4 weeks stored at 25°C. In vitro results highlighted similar efficiencies between LNC and standard EAPB0503 solutions prepared in dimethyl sulfoxide. Conclusion: In view of results obtained for loading efficiency and drug loading, the use of a LNC formulation is very interesting to permit the solubilization of a lipophilic drug and to improve its bioavailability. Preliminary tested pharmaceutical formulation applied to EAPB0503 significantly improved its water solubility and will be soon considered for future preclinical in vivo studies.
Collapse
Affiliation(s)
- Adrien Chouchou
- CNRS, ENSCM, IBMM, University of Montpellier, Montpellier, France
| | - Anne Aubert-Pouëssel
- UMR 5253, CNRS, ENSCM, University of Montpellier, Institut Charles Gerhardt Montpellier, Montpellier, France
| | - Christophe Dorandeu
- UMR 5253, CNRS, ENSCM, University of Montpellier, Institut Charles Gerhardt Montpellier, Montpellier, France
| | - Zahraa Zghaib
- CNRS, ENSCM, IBMM, University of Montpellier, Montpellier, France
| | - Pierre Cuq
- CNRS, ENSCM, IBMM, University of Montpellier, Montpellier, France
| | - Jean-Marie Devoisselle
- UMR 5253, CNRS, ENSCM, University of Montpellier, Institut Charles Gerhardt Montpellier, Montpellier, France
| | | | - Sylvie Bégu
- UMR 5253, CNRS, ENSCM, University of Montpellier, Institut Charles Gerhardt Montpellier, Montpellier, France
| | | |
Collapse
|