1
|
Patil SM, Nikalje P, Gavande N, Asgaonkar KD, Rathod V. An Insight into the Structure-activity Relationship of Benzimidazole and Pyrazole Derivatives as Anticancer Agents. Curr Top Med Chem 2025; 25:350-377. [PMID: 39484762 DOI: 10.2174/0115680266343336241021080438] [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: 07/10/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Cancer is a leading cause of death worldwide, driving the urgent need for new and effective treatments. Benzimidazole and pyrazole derivatives have gained attention for their potential as anticancer agents due to their diverse biological activities. The development of resistance in cancer cells, toxicity concerns, and inconsistent efficacy across different types of cancer are a few of the challenges. To overcome these challenges, optimisation of these nuclei using the structure-activity relationships is necessary. OBJECTIVES This review aimed to examine various benzimidazole, pyrazole, and their hybrid derivatives by focusing on their structure-activity relationships (SAR) as anticancer agents. Results of the most potent and least potent benzimidazole, pyrazole compounds, and their hybrid derivatives published by researchers were compiled. METHODS The findings of different researchers working on benzimidazole and pyrazole nuclei were reviewed and analysed for different targets and cell lines. Moreover, substitutions on different positions of pyrazole, benzimidazole, and their hybrid were summarised to derive an optimised pharmacophore. RESULTS Based on our analysis of existing studies, we anticipate that this review will guide researchers in creating potent pyrazole, benzimidazole, and hybrid derivatives crucial for combating cancer effectively. CONCLUSION Structure-Activity Relationship (SAR) studies can help in developing pyrazolebenzimidazole hybrids that are more powerful and selective in targeting specific aspects of cancer.
Collapse
Affiliation(s)
- Shital M Patil
- Department of Chemistry, AISSMS College of Pharmacy, Kennedy Road, Pune-01, India
| | - Piyush Nikalje
- Department of Chemistry, AISSMS College of Pharmacy, Kennedy Road, Pune-01, India
| | - Navnath Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, Michigan, 48201, USA
| | - Kalyani D Asgaonkar
- Department of Chemistry, AISSMS College of Pharmacy, Kennedy Road, Pune-01, India
| | - Vaishnavi Rathod
- Department of Chemistry, AISSMS College of Pharmacy, Kennedy Road, Pune-01, India
| |
Collapse
|
2
|
Hassan SM, Farid A, Panda SS, Bekheit MS, Dinkins H, Fayad W, Girgis AS. Indole Compounds in Oncology: Therapeutic Potential and Mechanistic Insights. Pharmaceuticals (Basel) 2024; 17:922. [PMID: 39065774 PMCID: PMC11280311 DOI: 10.3390/ph17070922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer remains a formidable global health challenge, with current treatment modalities such as chemotherapy, radiotherapy, surgery, and targeted therapy often hindered by low efficacy and adverse side effects. The indole scaffold, a prominent heterocyclic structure, has emerged as a promising candidate in the fight against cancer. This review consolidates recent advancements in developing natural and synthetic indolyl analogs, highlighting their antiproliferative activities against various cancer types over the past five years. These analogs are categorized based on their efficacy against common cancer types, supported by biochemical assays demonstrating their antiproliferative properties. In this review, emphasis is placed on elucidating the mechanisms of action of these compounds. Given the limitations of conventional cancer therapies, developing targeted therapeutics with enhanced selectivity and reduced side effects remains a critical focus in oncological research.
Collapse
Affiliation(s)
- Sara M. Hassan
- Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Alyaa Farid
- Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Siva S. Panda
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt;
| | - Holden Dinkins
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt;
| |
Collapse
|
3
|
Dube ZF, Soremekun OS, Ntombela T, Alahmdi MI, Abo-Dya NE, Sidhom PA, Shawky AM, Shibl MF, Ibrahim MA, Soliman ME. Inherent efficacies of pyrazole-based derivatives for cancer therapy: the interface between experiment and in silico. Future Med Chem 2023; 15:1719-1738. [PMID: 37772542 DOI: 10.4155/fmc-2023-0142] [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] [Indexed: 09/30/2023] Open
Abstract
There has been an increasing trend in the design of novel pyrazole derivatives for desired biological applications. For a cost-effective strategy, scientists have implemented various computational drug design tools to go hand in hand with experiments for the design and discovery of potentially effective pyrazole-based therapeutics. This review highlights the milestones of pyrazole-containing inhibitors and the use of molecular modeling techniques in conjunction with experimental studies to provide a view of the binding mechanism of these compounds. The review focuses on the established targets that play a key role in cancer therapy, including proteins involved in tubulin polymerization, carbonic anhydrase and tyrosine kinase. Overall, using both experimental and computational methods in drug design represents a promising approach to cancer therapy.
Collapse
Affiliation(s)
- Zanele F Dube
- Molecular Bio-Computational & Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Opeyemi S Soremekun
- Molecular Bio-Computational & Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, South Kensington, London, SW7 2BX, UK
| | - Thandokuhle Ntombela
- Catalysis & Peptide Research Unit, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Mohammed Issa Alahmdi
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Ahmed M Shawky
- Science & Technology Unit, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Mohamed F Shibl
- Renewable Energy Program, Center for Sustainable Development, College of Arts & Sciences, Qatar University, Doha, 2713, Qatar
| | - Mahmoud Aa Ibrahim
- Molecular Bio-Computational & Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Mahmoud Es Soliman
- Molecular Bio-Computational & Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| |
Collapse
|
4
|
Zhang Y, Wu C, Zhang N, Fan R, Ye Y, Xu J. Recent Advances in the Development of Pyrazole Derivatives as Anticancer Agents. Int J Mol Sci 2023; 24:12724. [PMID: 37628906 PMCID: PMC10454718 DOI: 10.3390/ijms241612724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Pyrazole derivatives, as a class of heterocyclic compounds, possess unique chemical structures that confer them with a broad spectrum of pharmacological activities. They have been extensively explored for designing potent and selective anticancer agents. In recent years, numerous pyrazole derivatives have been synthesized and evaluated for their anticancer potential against various cancer cell lines. Structure-activity relationship studies have shown that appropriate substitution on different positions of the pyrazole ring can significantly enhance anticancer efficacy and tumor selectivity. It is noteworthy that many pyrazole derivatives have demonstrated multiple mechanisms of anticancer action by interacting with various targets including tubulin, EGFR, CDK, BTK, and DNA. Therefore, this review summarizes the current understanding on the structural features of pyrazole derivatives and their structure-activity relationships with different targets, aiming to facilitate the development of potential pyrazole-based anticancer drugs. We focus on the latest research advances in anticancer activities of pyrazole compounds reported from 2018 to present.
Collapse
Affiliation(s)
- Yingqian Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (C.W.); (N.Z.); (R.F.); (Y.Y.)
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou 311121, China
| | - Chenyuan Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (C.W.); (N.Z.); (R.F.); (Y.Y.)
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou 311121, China
| | - Nana Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (C.W.); (N.Z.); (R.F.); (Y.Y.)
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou 311121, China
| | - Rui Fan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (C.W.); (N.Z.); (R.F.); (Y.Y.)
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou 311121, China
| | - Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (C.W.); (N.Z.); (R.F.); (Y.Y.)
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou 311121, China
| | - Jun Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
5
|
Hu B, Yan W, Jiang P, Jiang L, Yuan X, Lin J, Jiao Y, Jin Y. Switchable synthesis of natural-product-like lawsones and indenopyrazoles through regioselective ring-expansion of indantrione. Commun Chem 2023; 6:17. [PMID: 36697885 PMCID: PMC9849474 DOI: 10.1038/s42004-022-00807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023] Open
Abstract
Lawsones and indenopyrazoles are the prevalent structural motifs and building blocks in pharmaceuticals and bioactive molecules, but their synthesis has always remained challenging as no comprehensive protocol has been outlined to date. Herein, a metal-free, ring-expansion reaction of indantrione with diazomethanes, generated in situ from the N-tosylhydrazones, has been developed for the synthesis of lawsone and indenopyrazole derivatives in acetonitrile and alcohol solvents, respectively. It provides these valuable lawsone and pyrazole skeletons in good yields and high levels of diastereoselectivity from simple and readily available starting materials. DFT calculations were used to explore the mechanism in different solutions. The synthetic application example also showed the prospects of this method for the preparation of valuable compounds.
Collapse
Affiliation(s)
- Bingwei Hu
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Wenxin Yan
- School of Chemistry and Chemical Engineering, Key Laboratory of Theoretical Organic Chemistry and Functional Molecular, Ministry of Education, Hunan University of Science and Technology, 411201, Xiangtan, China
| | - Peiyun Jiang
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Ling Jiang
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Xu Yuan
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Jun Lin
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Yinchun Jiao
- School of Chemistry and Chemical Engineering, Key Laboratory of Theoretical Organic Chemistry and Functional Molecular, Ministry of Education, Hunan University of Science and Technology, 411201, Xiangtan, China.
| | - Yi Jin
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China.
| |
Collapse
|
6
|
Synthesis of new morpholine-benzimidazole-pyrazole hybrids as tubulin polymerization inhibiting anticancer agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
7
|
Constantinescu T, Mihis AG. Two Important Anticancer Mechanisms of Natural and Synthetic Chalcones. Int J Mol Sci 2022; 23:11595. [PMID: 36232899 PMCID: PMC9570335 DOI: 10.3390/ijms231911595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
ATP-binding cassette subfamily G and tubulin pharmacological mechanisms decrease the effectiveness of anticancer drugs by modulating drug absorption and by creating tubulin assembly through polymerization. A series of natural and synthetic chalcones have been reported to have very good anticancer activity, with a half-maximal inhibitory concentration lower than 1 µM. By modulation, it is observed in case of the first mechanism that methoxy substituents on the aromatic cycle of acetophenone residue and substitution of phenyl nucleus by a heterocycle and by methoxy or hydroxyl groups have a positive impact. To inhibit tubulin, compounds bind to colchicine binding site. Presence of methoxy groups, amino groups or heterocyclic substituents increase activity.
Collapse
Affiliation(s)
- Teodora Constantinescu
- Department of Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University, 400012 Cluj-Napoca, Romania
| | - Alin Grig Mihis
- Advanced Materials and Applied Technologies Laboratory, Institute of Research-Development-Innovation in Applied Natural Sciences, “Babes-Bolyai” University, Fantanele Str. 30, 400294 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Qin R, You FM, Zhao Q, Xie X, Peng C, Zhan G, Han B. Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: from molecular mechanisms to potential therapeutic targets. J Hematol Oncol 2022; 15:133. [PMID: 36104717 PMCID: PMC9471064 DOI: 10.1186/s13045-022-01350-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/03/2022] [Indexed: 12/11/2022] Open
Abstract
Regulated cell death (RCD) is a critical and active process that is controlled by specific signal transduction pathways and can be regulated by genetic signals or drug interventions. Meanwhile, RCD is closely related to the occurrence and therapy of multiple human cancers. Generally, RCD subroutines are the key signals of tumorigenesis, which are contributed to our better understanding of cancer pathogenesis and therapeutics. Indole alkaloids derived from natural sources are well defined for their outstanding biological and pharmacological properties, like vincristine, vinblastine, staurosporine, indirubin, and 3,3′-diindolylmethane, which are currently used in the clinic or under clinical assessment. Moreover, such compounds play a significant role in discovering novel anticancer agents. Thus, here we systemically summarized recent advances in indole alkaloids as anticancer agents by targeting different RCD subroutines, including the classical apoptosis and autophagic cell death signaling pathways as well as the crucial signaling pathways of other RCD subroutines, such as ferroptosis, mitotic catastrophe, necroptosis, and anoikis, in cancer. Moreover, we further discussed the cross talk between different RCD subroutines mediated by indole alkaloids and the combined strategies of multiple agents (e.g., 3,10-dibromofascaplysin combined with olaparib) to exhibit therapeutic potential against various cancers by regulating RCD subroutines. In short, the information provided in this review on the regulation of cell death by indole alkaloids against different targets is expected to be beneficial for the design of novel molecules with greater targeting and biological properties, thereby facilitating the development of new strategies for cancer therapy.
Collapse
|
9
|
Li MM, Huang H, Pu Y, Tian W, Deng Y, Lu J. A close look into the biological and synthetic aspects of fused pyrazole derivatives. Eur J Med Chem 2022; 243:114739. [PMID: 36126386 DOI: 10.1016/j.ejmech.2022.114739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022]
Abstract
The fusion of pyrazole scaffold with other skeletons creates a class of attractive molecules, demonstrating significant biological and chemical potentiality in the development of medicinal chemistry. Over the past few decades, numerous biologically active molecules featuring fused pyrazole moieties have been excavated and synthesized, some of which represented by sildenafil have been marketed as drugs, and the biological importance together with chemical synthesis strategies of fused pyrazole compounds, including structural modification based on lead compounds, have been steadily progressing. In this review, we focused our attention on the biological importance of fused pyrazoles and highlighted recent progress in the synthesis of this framework over the past 10 years. What' s more, the limitations, challenges, and future prospects were proposed, wishing to provide references for the development of pyrazole fused frameworks in the field of medicinal chemistry. Contents.
Collapse
Affiliation(s)
- Mei-Mei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Hui Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yiru Pu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wanrong Tian
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yun Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
| |
Collapse
|
10
|
Mor S, Khatri M, Punia R, Kumar D, Jindal DK, Basu B, Jakhar K. Synthesis and in vitro anticancer evaluation of 8b-hydroxy-1-(6-substitutedbenzo[d]thiazol-2-yl)-3-(3-substitutedphenyl)-1,8b-dihydroindeno[1,2-c]pyrazol-4(3aH)-ones. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Ebenezer O, Shapi M, Tuszynski JA. A Review of the Recent Developments of Molecular Hybrids Targeting Tubulin Polymerization. Int J Mol Sci 2022; 23:4001. [PMID: 35409361 PMCID: PMC8999808 DOI: 10.3390/ijms23074001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
Microtubules are cylindrical protein polymers formed from αβ-tubulin heterodimers in the cytoplasm of eukaryotic cells. Microtubule disturbance may cause cell cycle arrest in the G2/M phase, and anomalous mitotic spindles will form. Microtubules are an important target for cancer drug action because of their critical role in mitosis. Several microtubule-targeting agents with vast therapeutic advantages have been developed, but they often lead to multidrug resistance and adverse side effects. Thus, single-target therapy has drawbacks in the effective control of tubulin polymerization. Molecular hybridization, based on the amalgamation of two or more pharmacophores of bioactive conjugates to engender a single molecular structure with enhanced pharmacokinetics and biological activity, compared to their parent molecules, has recently become a promising approach in drug development. The practical application of combined active scaffolds targeting tubulin polymerization inhibitors has been corroborated in the past few years. Meanwhile, different designs and syntheses of novel anti-tubulin hybrids have been broadly studied, illustrated, and detailed in the literature. This review describes various molecular hybrids with their reported structural-activity relationships (SARs) where it is possible in an effort to generate efficacious tubulin polymerization inhibitors. The aim is to create a platform on which new active scaffolds can be modeled for improved tubulin polymerization inhibitory potency and hence, the development of new therapeutic agents against cancer.
Collapse
Affiliation(s)
- Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Michael Shapi
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Jack A. Tuszynski
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada
- DIMEAS, Politecnico di Torino, 10129 Turin, Italy
| |
Collapse
|
12
|
Shankaraiah N, Tokala R, Bora D. Contribution of Knoevenagel Condensation Products towards Development of Anticancer Agents: An Updated Review. ChemMedChem 2022; 17:e202100736. [PMID: 35226798 DOI: 10.1002/cmdc.202100736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/23/2022] [Indexed: 11/10/2022]
Abstract
Knoevenagel condensation is an entrenched, prevailing, prominent arsenal following greener principles in the generation of α, β-unsaturated ketones/carboxylic acids by involving carbonyl functionalities and active methylenes. This reaction has proved to be a major driving force in many multicomponent reactions indicating the prolific utility towards the development of biologically fascinating molecules. This eminent reaction was acclimatised on different pharmacophoric aldehydes (benzimidazole, β-carboline, phenanthrene, indole, imidazothiadiazole, pyrazole etc.) and active methylenes (oxindole, barbituric acid, Meldrum's acid, thiazolidinedione etc.) to generate the library of chemical compounds. Their potential was also explicit to understand the significance of functionalities involved, which thereby evoke further developments in drug discovery. Furthermore, most of these reaction products exhibited remarkable anticancer activity in nanomolar to micromolar ranges by targeting different cancer targets like DNA, microtubules, Topo-I/II, and kinases (PIM, PARP, NMP, p300/CBP) etc. This review underscores the efficiency of the Knoevenagel condensation explored in the past six-year to generate molecules of pharmacological interest, predominantly towards cancer. The present review also provides the aspects of structure-activity relationships, mode of action and docking study with possible interaction with the target protein.
Collapse
Affiliation(s)
- Nagula Shankaraiah
- National Institute of Pharmaceutical Education and Research NIPER, Department of Medicinal Chemistry, Balanagar, 500037, Hyderabad, INDIA
| | - Ramya Tokala
- NIPER Hyderabad: National Institute of Pharmaceutical Education and Research Hyderabad, Medicinal Chemistry, INDIA
| | - Darshana Bora
- NIPER Hyderabad: National Institute of Pharmaceutical Education and Research Hyderabad, Medicinal Chemistry, INDIA
| |
Collapse
|
13
|
Hong Y, Zhu YY, He Q, Gu SX. Indole derivatives as tubulin polymerization inhibitors for the development of promising anticancer agents. Bioorg Med Chem 2022; 55:116597. [PMID: 34995858 DOI: 10.1016/j.bmc.2021.116597] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/18/2021] [Accepted: 12/27/2021] [Indexed: 01/01/2023]
Abstract
The α- and β-tubulins are the major polypeptide components of microtubules (MTs), which are attractive targets for anticancer drug development. Indole derivatives display a variety of biological activities including antitumor activity. In recent years, a great number of indole derivatives as tubulin polymerization inhibitors have sprung up, which encourages medicinal chemists to pursue promising inhibitors with improved antitumor activities, excellent physicochemical, pharmacokinetic and pharmacodynamic properties. In this review, the recent progress from 2010 to present in the development of indole derivatives as tubulin polymerization inhibitors was summarized and reviewed, which would provide useful clues and inspirations for further design of outstanding tubulin polymerization inhibitors.
Collapse
Affiliation(s)
- Yu Hong
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yuan-Yuan Zhu
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Qiuqin He
- Department of Chemistry, Fudan University, Shanghai 200433, China.
| | - Shuang-Xi Gu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, Wuhan 430205, China.
| |
Collapse
|
14
|
Mor S, Khatri M. Synthesis, antimicrobial evaluation, α-amylase inhibitory ability and molecular docking studies of 3-alkyl-1-(4-(aryl/heteroaryl)thiazol-2-yl)indeno[1,2-c]pyrazol-4(1H)-ones. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Özdemir A, Ciftci H, Sever B, Tateishi H, Otsuka M, Fujita M, Altıntop MD. A New Series of Indeno[1,2- c]pyrazoles as EGFR TK Inhibitors for NSCLC Therapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020485. [PMID: 35056800 PMCID: PMC8778314 DOI: 10.3390/molecules27020485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 02/05/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death throughout the world. Due to the shortcomings of traditional chemotherapy, targeted therapies have come into prominence for the management of NSCLC. In particular, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) therapy has emerged as a first-line therapy for NSCLC patients with EGFR-activating mutations. In this context, new indenopyrazoles, which were prepared by an efficient microwave-assisted method, were subjected to in silico and in vitro assays to evaluate their potency as EGFR TK-targeted anti-NSCLC agents. Compound 4 was the most promising antitumor agent towards A549 human lung adenocarcinoma cells, with an IC50 value of 6.13 µM compared to erlotinib (IC50 = 19.67 µM). Based on its low cytotoxicity to peripheral blood mononuclear cells (PBMCs), it can be concluded that compound 4 exerts selective antitumor action. This compound also inhibited EGFR TK with an IC50 value of 17.58 µM compared to erlotinib (IC50 = 0.04 µM) and induced apoptosis (56.30%). Taking into account in silico and in vitro data, compound 4 stands out as a potential EGFR TKI for the treatment of NSCLC.
Collapse
Affiliation(s)
- Ahmet Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey;
- Correspondence: (A.Ö.); (M.F.); (M.D.A.); Tel.: +90-222-335-0580 (ext. 3780) (A.Ö.); +81-96-371-4622 (M.F.); +90-222-335-0580 (ext. 3807) (M.D.A.)
| | - Halilibrahim Ciftci
- Department of Drug Discovery, Science Farm Ltd., Kumamoto 862-0976, Japan; (H.C.); (M.O.)
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey;
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
| | - Hiroshi Tateishi
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
| | - Masami Otsuka
- Department of Drug Discovery, Science Farm Ltd., Kumamoto 862-0976, Japan; (H.C.); (M.O.)
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
- Correspondence: (A.Ö.); (M.F.); (M.D.A.); Tel.: +90-222-335-0580 (ext. 3780) (A.Ö.); +81-96-371-4622 (M.F.); +90-222-335-0580 (ext. 3807) (M.D.A.)
| | - Mehlika Dilek Altıntop
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey;
- Correspondence: (A.Ö.); (M.F.); (M.D.A.); Tel.: +90-222-335-0580 (ext. 3780) (A.Ö.); +81-96-371-4622 (M.F.); +90-222-335-0580 (ext. 3807) (M.D.A.)
| |
Collapse
|
16
|
Chaudhari P, Bari S, Surana S, Shirkhedkar A, Wakode S, Shelar S, Racharla S, Ugale V, Ghodke M. Logical synthetic strategies and structure-activity relationship of indolin-2-one hybrids as small molecule anticancer agents: An overview. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131280] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Khetmalis YM, Shivani M, Murugesan S, Chandra Sekhar KVG. Oxindole and its derivatives: A review on recent progress in biological activities. Biomed Pharmacother 2021; 141:111842. [PMID: 34174506 DOI: 10.1016/j.biopha.2021.111842] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Oxindole has been shown to be a pharmacologically advantageous scaffold having many biological properties that are relevant to medicinal chemistry. The simplicity and widespread occurrence of this scaffold in plant-based alkaloids have further reinforced oxindole's merit in the domain of novel drug discovery. First extracted from Uncaria tomentosa, commonly the known as cat claw's plant which was found abundantly in the Amazon rainforest, molecules with the oxindole moiety have been shown to be common in a wide variety of compounds extracted from plant sources. The role of oxindole as a chemical scaffold for fabricating and designing biological drugs agents can be ascribed to its ability to be modified by a number of chemical groups to generate novel biological functions. This review is aimed at providing a description of the general chemistry based on existing corresponding structure-activity relationships (SARs) and compile all recent developmentary studies on oxindole-derived compounds as a successful pharmaceutical agent. A substantial group of oxindole derivatives are chiefly being tested as anticancer agents, however, a several oxindole derivatives have been shown to possesses antimicrobial, α-glucosidase inhibitory, antiviral, antileishmanial, antitubercular, antioxidative, tyrosinase inhibitory, PAK4 inhibitory, antirheumatoid arthritis and intraocular pressure reducing activities, to name a few. In this review we show the potential value of developing newer oxindole derivatives with an improved range of pharmacological implications as well as identifying drugs possessing oxindole core, that are showing and serving increased efficacy in clinical practice.
Collapse
Affiliation(s)
- Yogesh Mahadu Khetmalis
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Mithula Shivani
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 33303, Rajasthan, India
| | - Kondapalli Venkata Gowri Chandra Sekhar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India.
| |
Collapse
|
18
|
Khan I, Ganapathi T, Rehman MMU, Shareef MA, Kumar CG, Kamal A. New indenopyrazole linked oxadiazole conjugates as anti-pancreatic cancer agents: Design, synthesis, in silico studies including 3D-QSAR analysis. Bioorg Med Chem Lett 2021; 44:128094. [PMID: 33964437 DOI: 10.1016/j.bmcl.2021.128094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 11/25/2022]
Abstract
To continue the quest of newer anticancer agents, herein a novel class of 1,4-Dihydroindenopyrazole linked oxadiazole conjugates 9(a-r) was designed, synthesized and experimented for their anti-proliferative activities against four different cancer cell lines (human) such as MDA MB-231 (breast), PANC-1 (pancreatic), MCF-7 (breast), and Caco-2 (Colorectal) by using MTT assay. Among the series compound 9h and 9 m demonstrated significant potency against the PANC-1 (human pancreatic cancer cells) with IC50 value 7.4 μM and 4.3 μM respectively. While compound 9 m was found to be equipotent to standard Gomitabine (IC50 = 4.2 μM). The detailed biological assays revealed S phase cell cycle arrest and their ability to propagate apoptosis by activating caspase 3 and 9 enzymes which was confirmed by Annexin-FITC assay and caspase assay. Moreover, docking study suggested their binding modes and interactions with caspase-3. In addition, in silico studies revealed that they exhibit good pharmacokinetics and drug likeliness properties. Furthermore, 3D-QSAR was carried out to achieve a pharmacophoric model with CoMFA (q2 = 0.631, r2 = 0.977) and CoMSIA (q2 = 0.686, r2 = 0.954) on PANC-1 cancer cells which were established, generated and validated to be reliable models for further design and optimization of newer molecules with enhanced anticancer activity.
Collapse
Affiliation(s)
- Irfan Khan
- Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Thipparapu Ganapathi
- Stem Cell Research Division, National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Hyderabad 500007, Telangana, India.
| | - Md Muzaffar-Ur- Rehman
- Catalysis and Fine Chemicals Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, India.
| | - Mohd Adil Shareef
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India; Centre for Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Tarnaka 500007, Hyderabad, India.
| | - C Ganesh Kumar
- Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Ahmed Kamal
- Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India; School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
19
|
Abstract
The heterocyclic compounds are the building blocks for the synthesis of the different biologically
active compounds in the organic chemistry. Heterocyclic compounds have versatile synthetic
applicability and biological activity. Pyrazole carboxylic acid derivatives are significant scaffold
structures in heterocyclic compounds due to biologic activities such as antimicrobial, anticancer, inflammatory,
antidepressant, antifungal anti-tubercular and antiviral, etc. The aim of this mini-review
is an overview synthesis of pyrazole carboxylic acid derivatives and their biologic applications. The
summarized literature survey presents biological activities of pyrazole carboxylic acid derivatives
and their various synthetic methods in detail. This mini-review can be a guide to many scientists in
medicinal chemistry.
Collapse
Affiliation(s)
- Adnan Cetin
- Department of Sciences, Faculty of Education, University of Mus Alparslan, Mus, Turkey
| |
Collapse
|
20
|
Mor S, Khatri M, Punia R, Sindhu S. Recent Progress on Anticancer Agents Incorporating Pyrazole Scaffold. Mini Rev Med Chem 2021; 22:115-163. [PMID: 33823764 DOI: 10.2174/1389557521666210325115218] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/01/2021] [Accepted: 02/07/2021] [Indexed: 11/22/2022]
Abstract
The search of new anticancer agents is considered as a dynamic field of medicinal chemistry. In recent years, the synthesis of compounds with anticancer potential has increased and a large number of structurally varied compounds displaying potent anticancer activities have been published. Pyrazole is an important biologically active scaffold that possessed nearly all types of biological activities. The aim of this review is to collate literature work reported by researchers to provide an overview on in vivo and in vitro anticancer activities of pyrazole based derivatives among the diverse biological activities displayed by them and also presents recent efforts made on this heterocyclic moiety regarding anticancer activities. This review has been driven from the increasing number of publications, on this issue, which have been reported in the literature since the ending of the 20th century (from 1995-to date).
Collapse
Affiliation(s)
- Satbir Mor
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar-125001, Haryana. India
| | - Mohini Khatri
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar-125001, Haryana. India
| | - Ravinder Punia
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar-125001, Haryana. India
| | - Suchita Sindhu
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar-125001, Haryana. India
| |
Collapse
|
21
|
Sana S, Reddy VG, Srinivasa Reddy T, Tokala R, Kumar R, Bhargava SK, Shankaraiah N. Cinnamide derived pyrimidine-benzimidazole hybrids as tubulin inhibitors: Synthesis, in silico and cell growth inhibition studies. Bioorg Chem 2021; 110:104765. [PMID: 33677248 DOI: 10.1016/j.bioorg.2021.104765] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/12/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022]
Abstract
An approach in modern medicinal chemistry to discover novel bioactive compounds is by mimicking diverse complementary pharmacophores. In extension of this strategy, a new class of piperazine-linked cinnamide derivatives of benzimidazole-pyrimidine hybrids have been designed and synthesized. Their in vitro cytotoxicity profiles were explored on selected human cancer cell lines. Specifically, structural comparison of target hybrids with tubulin-DAMA-colchicine and tubulin-nocodazole complexes has exposed a deep position of benzimidazole ring into the αT5 loop. All the synthesized compounds were demonstrated modest to interesting cytotoxicity against different cancer cell lines. The utmost cytotoxicity has shown with an amine linker of benzimidazole-pyrimidine series, with specificity toward A549 (lung cancer) cell line. The most potent compound in this series was 18i, which inhibited cancer cell growth at micromolar concentrations ranging 2.21-7.29 µM. Flow cytometry studies disclosed that 18i inhibited the cells in G2/M phase of cell cycle. The potent antitumor activity of 18i resulted from enhanced microtubule disruption at a similar level as nocodazole on β-tubulin antibody, explored using immunofluorescence staining. The most active compound 18i also inhibited tubulin polymerization with an IC50 of 5.72 ± 0.51 µM. In vitro biological analysis of 18i presented apoptosis induction on A549 cells with triggering of ROS generation and loss of mitochondrial membrane potential, resulting in DNA injury. In addition, 18i displayed impairment in cellular migration and inhibited the colony formation. Notably, the safety profile of most potent compound 18i was revealed by screening against normal human pulmonary epithelial cells (L132: IC50: 69.25 ± 5.95 μM). The detailed binding interactions of 18i with tubulin was investigated by employing molecular docking, superimposition and free energy analyses. Thus remarks made in this study established that pyrimidine-benzimidazole hybrids as a new class of tubulin polymerization inhibitors with significant anticancer activity.
Collapse
Affiliation(s)
- Sravani Sana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Velma Ganga Reddy
- Centre for Advanced Materials & Industrial Chemistry (CAMIC), School of Science, RMIT University, GPO Box 2476, Melbourne 3001, Australia.
| | - T Srinivasa Reddy
- Centre for Advanced Materials & Industrial Chemistry (CAMIC), School of Science, RMIT University, GPO Box 2476, Melbourne 3001, Australia
| | - Ramya Tokala
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Rahul Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Suresh K Bhargava
- Centre for Advanced Materials & Industrial Chemistry (CAMIC), School of Science, RMIT University, GPO Box 2476, Melbourne 3001, Australia
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
22
|
Cui YJ, Liu C, Ma CC, Ji YT, Yao YL, Tang LQ, Zhang CM, Wu JD, Liu ZP. SAR Investigation and Discovery of Water-Soluble 1-Methyl-1,4-dihydroindeno[1,2- c]pyrazoles as Potent Tubulin Polymerization Inhibitors. J Med Chem 2020; 63:14840-14866. [PMID: 33201714 DOI: 10.1021/acs.jmedchem.0c01345] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Taking the previously discovered 1-methyl-1,4-dihydroindeno[1,2c]pyrazol derivative LL01 as a lead, systematic structural modifications were made at the phenolic 6- and 7-positions and the aniline at the 3-position of the indenopyrazole core to investigate the SARs and to improve water solubility. Among the designed indenopyrazoles ID01-ID33, a series of potent MTAs were identified. As the hydrochloride salt(s), ID09 and ID33 showed excellent aqueous solubility and favorable Log P value and displayed noteworthily low nanomolar potency against a variety of tumor cells, including those taxol-resistant ones. They inhibited tubulin polymerization, disrupted cellular microtubule networks by targeting the colchicine site, and promoted HepG2 cell cycle arrest and cell apoptosis. In the HepG2 xenograft mouse model, ID09 and ID33 effectively inhibited tumor growth at an oral dose of 25 mg/kg. At an intravenous (iv) injection dose of 10 mg/kg every other day, ID09 suppressed tumor growth by 68% without obvious toxicity.
Collapse
Affiliation(s)
- Ying-Jie Cui
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Chao Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Chen-Chen Ma
- Central Laboratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250012, P. R. China
| | - Ya-Ting Ji
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Yi-Li Yao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Long-Qian Tang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Cheng-Mei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Jing-De Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| |
Collapse
|
23
|
Noha RM, Abdelhameid MK, Ismail MM, Mohammed MR, Salwa E. Design, synthesis and screening of benzimidazole containing compounds with methoxylated aryl radicals as cytotoxic molecules on (HCT-116) colon cancer cells. Eur J Med Chem 2020; 209:112870. [PMID: 33158579 DOI: 10.1016/j.ejmech.2020.112870] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
A novel series of benzimidazole derivatives with methoxylated aryl groups was designed and synthesized as molecules with potential cytotoxic activity. In vitro cytotoxic activity over HCT-116 cells showed that N-(benzimidazothiazolone)acetamides 11a, 11b and 11c were found to be the most cytotoxic compounds compared camptothecin (CPT). The tested compounds had a dual topoisomerase I-β (Topo I-β) and tubulin inhibiting activities when compared to CPT and Podophyllotoxin (Podo) where, compounds l0a, l0b, 11a and 11b exhibited a potent inhibitory activity on Topo I-β enzyme in nano-molar concentration, on the other hand, compounds 12b and 13b exhibited the best inhibitory activity β-tubulin polymerization. Results of the cell cycle analysis as well as the results of annexin-V on HCT-116 cells showed that benzimidazothiazoles 12b and 13b had a pro-apoptotic activity higher than CPT by 1.33- and 1.30-folds, respectively. Moreover, the concentration of p53, Bax/Bcl-2 ratio and caspase 3/7 increased in compounds l0b, 11b, l2b, 13b, especially, compounds 11b and 13b exhibited an increased level of these mediators than CPT. Finally, compound 11b regulated the radiosensitizing activity of the HCT-116 cells by modulating the chromosomal instability.
Collapse
Affiliation(s)
- Ryad M Noha
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science & Technology, Giza, P.O. Box 77, Egypt
| | - Mohammed K Abdelhameid
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11561, Egypt.
| | - M Mohsen Ismail
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science & Technology, Giza, P.O. Box 77, Egypt; Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11561, Egypt
| | - Manal R Mohammed
- Department of Radiation Biology, National Center for Radiation Research and Technology, Cairo, 11787, Egypt
| | - Elmeligie Salwa
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11561, Egypt
| |
Collapse
|
24
|
Bai L, Fei WD, Gu YY, He M, Du F, Zhang WY, Yang LL, Liu YJ. Liposomes encapsulated iridium(III) polypyridyl complexes enhance anticancer activity in vitro and in vivo. J Inorg Biochem 2020; 205:111014. [PMID: 32044395 DOI: 10.1016/j.jinorgbio.2020.111014] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
Three iridium(III) complexes [Ir(ppy)2(CPIP)](PF6) (Ir-1, ppy = 2-phenylpyridine, CPIP = 2-(4-chlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline), [Ir(ppy)2(DCPIP)](PF6) (Ir-2, DCPIP = 2-(3,4-dichlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ir(ppy)2(TCPIP)](PF6) (Ir-3, TCPIP = 2,3,5-trichlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline) were synthesized and characterized. The complexes Ir-1, Ir-2 and Ir-3 were encapsulated in liposomes to form Ir-1-Lipo, Ir-2-Lipo and Ir-3-Lipo. Morphology, size distribution, and zeta potential of liposomes were examined by transmission electron microscopy (TEM) and Zetasizer. The cytotoxic activity in vitro of Ir-1, Ir-2 and Ir-3 against cancer A549, HTC-116, HepG2, BEL-7402, Eca-109, B16, HeLa SGC-7901 and normal NIH3T3 cells was evaluated by 3-(4,5-dimethylthiazole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT) method. Ir-2 and Ir-3 show no cytotoxic activity against the selected cancer cells, and Ir-1 displays moderate cytotoxic effect on the cell growth in A549 cells. However, Ir-1, Ir-2 and Ir-3 were encapsulated in liposomes, the cytotoxic activity was greatly enhanced. In particular, Ir-1-Lipo and Ir-2-Lipo can effectively inhibit the cell growth in A549 cells with a low IC50 value of 3.1 ± 0.3 and 1.2 ± 0.4 μM. The apoptosis was assayed by flow cytometry. Ir-1, Ir-2 and Ir-3 reveal weak apoptotic effect, whereas Ir-1-Lipo, Ir-2-Lipo and Ir-3-Lipo induce an apoptotic percentage of 55.6%, 69.3% and 16.7% in A549 cells, respectively. Specially, in the assay of antitumor activity in vivo, the inhibiting percentage of tumor growth induced by Ir-2 is 27.65%, while inhibiting percentage of tumor growth caused by Ir-2-Lipo is 57.45%. Obviously, the liposomes can enhance anticancer activity in vitro and in vivo compared with the complexes. The results show that the iridium(III) complexes encapsulated liposomes induce apoptosis in A549 cells through ROS-mediated lysosome-mitochondria dysfunction pathway and target the microtubules.
Collapse
Affiliation(s)
- Lan Bai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Wei-Dong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, PR China
| | - Yi-Ying Gu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Miao He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Fan Du
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Wen-Yao Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lin-Lin Yang
- Department of Pediatrics, Guangdong Women and Children Hospital, Guangzhou 510000, PR China.
| | - Yun-Jun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
25
|
Reddy VG, Reddy TS, Jadala C, Reddy MS, Sultana F, Akunuri R, Bhargava SK, Wlodkowic D, Srihari P, Kamal A. Pyrazolo-benzothiazole hybrids: Synthesis, anticancer properties and evaluation of antiangiogenic activity using in vitro VEGFR-2 kinase and in vivo transgenic zebrafish model. Eur J Med Chem 2019; 182:111609. [DOI: 10.1016/j.ejmech.2019.111609] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022]
|
26
|
Khan I, Shareef MA, Kumar CG. An overview on the synthetic and medicinal perspectives of indenopyrazoles. Eur J Med Chem 2019; 178:1-12. [DOI: 10.1016/j.ejmech.2019.05.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/25/2019] [Accepted: 05/25/2019] [Indexed: 02/07/2023]
|
27
|
Shareef MA, Sirisha K, Khan I, Sayeed IB, Jadav SS, Ramu G, Kumar CG, Kamal A, Babu BN. Design, synthesis, and antimicrobial evaluation of 1,4-dihydroindeno[1,2- c]pyrazole tethered carbohydrazide hybrids: exploring their in silico ADMET, ergosterol inhibition and ROS inducing potential. MEDCHEMCOMM 2019; 10:806-813. [PMID: 31191871 PMCID: PMC6540956 DOI: 10.1039/c9md00155g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/29/2019] [Indexed: 11/21/2022]
Abstract
A series of new 1,4-dihydroindeno[1,2-c]pyrazole tethered carbohydrazide hybrids (5a-u) were designed, synthesized and evaluated for their antimicrobial activity. Compounds 5d, 5g, 5j, 5k and 5q demonstrated significant activity against the entire panel of test pathogens. Further, compounds 5d and 5g exhibited significant anti-Candida activity. These potential hybrids (5d and 5g) also exhibited promising ergosterol biosynthesis inhibition against Candida albicans, which was further validated through molecular docking studies. Furthermore, compounds 5d and 5g caused intracellular ROS accumulation in C. albicans MTCC 3017 and were non-toxic to normal human lung cell line MRC5. In silico studies revealed that they demonstrated drug likeness and an appreciable pharmacokinetic profile. Overall, the findings demonstrate that 5d and 5g may be considered as promising leads for further development of new antifungal drugs.
Collapse
Affiliation(s)
- Mohd Adil Shareef
- Department of Fluoro-Agrochemicals , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad , India .
- Academy of Scientific and Innovative Research , New Delhi 110 025 , India
| | - K Sirisha
- Academy of Scientific and Innovative Research , New Delhi 110 025 , India
- Organic Synthesis and Process Chemistry Division , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad 500007 , India
| | - Irfan Khan
- Academy of Scientific and Innovative Research , New Delhi 110 025 , India
- Organic Synthesis and Process Chemistry Division , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad 500007 , India
| | - Ibrahim Bin Sayeed
- Academy of Scientific and Innovative Research , New Delhi 110 025 , India
- Organic Synthesis and Process Chemistry Division , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad 500007 , India
| | - Surender Singh Jadav
- Department of Fluoro-Agrochemicals , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad , India .
| | - Gopathi Ramu
- Department of Fluoro-Agrochemicals , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad , India .
- Academy of Scientific and Innovative Research , New Delhi 110 025 , India
| | - C Ganesh Kumar
- Organic Synthesis and Process Chemistry Division , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad 500007 , India
| | - Ahmed Kamal
- School of Pharmaceutical Education and Research , Jamia Hamdard University , New Delhi 110062 , India .
| | - Bathini Nagendra Babu
- Department of Fluoro-Agrochemicals , CSIR-Indian Institute of Chemical Technology , Tarnaka , Hyderabad , India .
- Academy of Scientific and Innovative Research , New Delhi 110 025 , India
| |
Collapse
|
28
|
Khan I, Kanugala S, Shareef MA, Ganapathi T, Shaik AB, Shekar KC, Kamal A, Kumar CG. Synthesis of new bis‐pyrazole linked hydrazides and their in vitro evaluation as antimicrobial and anti‐biofilm agents: A mechanistic role on ergosterol biosynthesis inhibition inCandida albicans. Chem Biol Drug Des 2019; 94:1339-1351. [DOI: 10.1111/cbdd.13509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/19/2019] [Accepted: 02/09/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Irfan Khan
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research Ghaziabad India
| | - Sirisha Kanugala
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research Ghaziabad India
| | - Mohd. Adil Shareef
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research Ghaziabad India
| | - Thipparapu Ganapathi
- Stem Cell Research DivisionDepartment of Biochemistry ICMR‐National Institute of Nutrition Hyderabad India
| | - Anver Basha Shaik
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Kunta Chandra Shekar
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Ahmed Kamal
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research Ghaziabad India
- School of Pharmaceutical Education and Research Jamia Hamdard University New Delhi India
| | - Chityal Ganesh Kumar
- Department of Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research Ghaziabad India
| |
Collapse
|
29
|
Design, synthesis, in silico pharmacokinetics prediction and biological evaluation of 1,4-dihydroindeno[1,2-c]pyrazole chalcone as EGFR /Akt pathway inhibitors. Eur J Med Chem 2019; 163:636-648. [DOI: 10.1016/j.ejmech.2018.12.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 01/07/2023]
|
30
|
Gezegen H, Tutar U, Hepokur C, Ceylan M. Synthesis and biological evaluation of novel indenopyrazole derivatives. J Biochem Mol Toxicol 2019; 33:e22285. [PMID: 30672630 DOI: 10.1002/jbt.22285] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/03/2019] [Indexed: 12/15/2022]
Abstract
A series of novel indenopyrazole derivatives 2a-j and 3a-j were synthesized from the reaction of 1-(4-(hydroxy(1-oxo-1,3-dihydro-2 H-inden-2-ylidene)methyl)phenyl)-3-phenylurea derivatives 1a-j with hydrazine and phenylhydrazine, respectively. The obtained novel indenopyrazoles ( 2a-j and 3a-j) were evaluated for anticancer activity against HeLa and C6 cell lines. Antiproliferative activity was determined by the BrdU proliferation ELISA assay; 2a, 2b, 2d, 2h, and 3h were found to be the most active compounds. The compounds were also screened for antimicrobial activity, and all compounds showed moderate activity against used microorganisms.
Collapse
Affiliation(s)
- Hayreddin Gezegen
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Cumhuriyet University, Sivas, Turkey
| | - Uğur Tutar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Cumhuriyet University, Sivas, Turkey
| | - Ceylan Hepokur
- Department of Biochemistry, Faculty of Pharmacy, Cumhuriyet University, Sivas, Turkey
| | - Mustafa Ceylan
- Department of Chemistry, Faculty of Arts and Sciences, Gaziosmanpasa University, Tokat, Turkey
| |
Collapse
|
31
|
Sonawane V, Mohd Siddique MU, Jadav SS, Sinha BN, Jayaprakash V, Chaudhuri B. Cink4T, a quinazolinone-based dual inhibitor of Cdk4 and tubulin polymerization, identified via ligand-based virtual screening, for efficient anticancer therapy. Eur J Med Chem 2019; 165:115-132. [PMID: 30665142 DOI: 10.1016/j.ejmech.2019.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/05/2019] [Accepted: 01/05/2019] [Indexed: 12/15/2022]
Abstract
Inhibition of cyclin dependent kinase 4 (Cdk4) prevents cancer cells from entering the early G0/G1 phase of the cell division cycle whereas inhibiting tubulin polymerization blocks cancer cells' ability to undergo mitosis (M) late in the cell cycle. We had reported earlier that two non-planar and relatively non-toxic fascaplysin derivatives, an indole and a tryptoline, inhibit Cdk4 with IC50 values of 6.2 and 10 μM, respectively. Serendipitously, we had also found that they inhibited tubulin polymerization. The molecules were efficacious in mouse tumor models. We have now identified Cink4T in a 59-compound quinazolinone library, designed on the basis of ligand-based virtual screening, as a compound that inhibits Cdk4 and tubulin. Its IC50 value for Cdk4 inhibition is 0.47 μM and >50 μM for inhibition of Cdk1, Cdk2, Cdk6, Cdk9. Cink4T inhibits tubulin polymerization with an IC50 of 0.6 μM. Molecular modelling studies on Cink4T with Cdk4 and tubulin crystal structures lend support to these observations. Cancer cell cycle analyses confirm that Cink4T blocks cells at both G0/G1 and M phases as it should if it were to inhibit both Cdk4 and tubulin polymerization. Our results show, for the very first time, that virtual screening can be used to design novel inhibitors that can potently block two crucial phases of the cell division cycle.
Collapse
Affiliation(s)
- Vinay Sonawane
- Leicester School of Pharmacy, De Montfort University, Leicester, LE1 9BH, UK
| | - Mohd Usman Mohd Siddique
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | | | - Barij Nayan Sinha
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Venkatesan Jayaprakash
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India.
| | - Bhabatosh Chaudhuri
- Leicester School of Pharmacy, De Montfort University, Leicester, LE1 9BH, UK.
| |
Collapse
|
32
|
Wang Y, Wang X, Lin J, Yao B, Wang G, Zhao Y, Zhang X, Lin B, Liu Y, Cheng M, Liu Y. Ynesulfonamide-Based Silica Gel and Alumina-Mediated Diastereoselective Cascade Cyclizations to Spiro[indoline-3,3′-pyrrolidin]-2-ones under Neat Conditions. Adv Synth Catal 2018. [DOI: 10.1002/adsc.201701576] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Yanshi Wang
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
| | - Xiaoyu Wang
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
| | - Jingsheng Lin
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
- Wuya College of Innovation; Shenyang Pharmaceutical University; Shenyang 110016 People's Republic of China
| | - Bo Yao
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
| | - Guanghui Wang
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
- Wuya College of Innovation; Shenyang Pharmaceutical University; Shenyang 110016 People's Republic of China
| | - Yuandong Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
- Wuya College of Innovation; Shenyang Pharmaceutical University; Shenyang 110016 People's Republic of China
| | - Xinhang Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
- Wuya College of Innovation; Shenyang Pharmaceutical University; Shenyang 110016 People's Republic of China
| | - Bin Lin
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
| | - Yang Liu
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
| | - Yongxiang Liu
- Key Laboratory of Structure-Based Drug Design and Discovery Shenyang Pharmaceutical University; Ministry of Education; Shenyang 110016 People's Republic of China
- Institute of Drug Research in Medicine Capital of China; Benxi 117000 People's Republic of China
- Wuya College of Innovation; Shenyang Pharmaceutical University; Shenyang 110016 People's Republic of China
| |
Collapse
|
33
|
Yuan S, Qiao T, Li X, Zhuang X, Chen W, Chen X, Zhang Q. Toll-like receptor 9 activation by CpG oligodeoxynucleotide 7909 enhances the radiosensitivity of A549 lung cancer cells via the p53 signaling pathway. Oncol Lett 2018. [PMID: 29541253 DOI: 10.3892/ol.2018.7916] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Unmethylated cytosine-phosphorothioate-guanine (CpG)-containing oligodeoxynucleotides (ODNs) are synthetic DNA sequences that mimic bacterial DNA, and are known to serve as ligands for Toll-like receptor 9 (TLR9). The interaction between a CpG ODNs with TLR9 activates the complex downstream cascade that contributes to exerting its function. In the present study, the results of clonogenic assays demonstrated that the activation of TLR9 by CpG ODNs significantly increased the radiosensitivity of A549 lung cancer cells, with a sensitivity enhancement ratio (SER) of 1.28. When the expression of TLR9 was effectively silenced, CpG ODNs used alone were identified to produce SERs as low as 1.01. Flow cytometry demonstrated that the interaction between TLR9 and CpG ODN 7909 alone did not significantly affect the rate of apoptosis, but may significantly enhance the radiation-induced apoptosis of A549 cells. Western blot analysis revealed that TLR9 activation by CpG ODN 7909 increased the levels of mitogen-activated protein kinase 14, cellular tumor antigen p53, B-cell lymphoma 2 associated X protein and genome polyprotein, and decreased Bcl-2 expression levels, whereas these effects were not observed in CpG ODN 7909-treated cells in which TLR9 was knocked down. These results suggest that CpG ODN 7909 may enhance radiosensitivity through TLR9 activation, and partially via the p53 pathway in A549 lung cancer cells.
Collapse
Affiliation(s)
- Sujuan Yuan
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Tiankui Qiao
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Xuan Li
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Xibing Zhuang
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Wei Chen
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Xue Chen
- Department of Radiology, Shanghai Cancer Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Qi Zhang
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
34
|
Jiang YY, Dou GY, Xu K, Zeng CC. Bromide-catalyzed electrochemical trifluoromethylation/cyclization of N-arylacrylamides with low catalyst loading. Org Chem Front 2018. [DOI: 10.1039/c8qo00645h] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
An electro-generated trifluoromethyl radical mediated by a bromide ion is efficiently applied to the trifluoromethylarylation/cyclization of N-arylacrylamides.
Collapse
Affiliation(s)
- Yang-Ye Jiang
- Beijing Key Laboratory of Environmental and Viral Oncology
- College of Life Science & Bioengineering
- Beijing University of Technology
- Beijing 100124
- China
| | - Gui-Yuan Dou
- Beijing Key Laboratory of Environmental and Viral Oncology
- College of Life Science & Bioengineering
- Beijing University of Technology
- Beijing 100124
- China
| | - Kun Xu
- Beijing Key Laboratory of Environmental and Viral Oncology
- College of Life Science & Bioengineering
- Beijing University of Technology
- Beijing 100124
- China
| | - Cheng-Chu Zeng
- Beijing Key Laboratory of Environmental and Viral Oncology
- College of Life Science & Bioengineering
- Beijing University of Technology
- Beijing 100124
- China
| |
Collapse
|