1
|
Zhang J, Zhang R, Li W, Ma XC, Qiu F, Sun CP. IκB kinase β (IKKβ): Structure, transduction mechanism, biological function, and discovery of its inhibitors. Int J Biol Sci 2023; 19:4181-4203. [PMID: 37705738 PMCID: PMC10496512 DOI: 10.7150/ijbs.85158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/26/2023] [Indexed: 09/15/2023] Open
Abstract
The effective approach to discover innovative drugs will ask natural products for answers because of their complex and changeable structures and multiple biological activities. Inhibitory kappa B kinase beta (IKKβ), known as IKK2, is a key regulatory kinase responsible for the activation of NF-κB through its phosphorylation at Ser177 and Ser181 to promote the phosphorylation of inhibitors of kappa B (IκBs), triggering their ubiquitination and degradation to active the nuclear factor kappa-B (NF-κB) cascade. Chemical inhibition of IKKβ or its genetic knockout has become an effective method to block NF-κB-mediated proliferation and migration of tumor cells and inflammatory response. In this review, we summarized the structural feature and transduction mechanism of IKKβ and the discovery of inhibitors from natural resources (e.g. sesquiterpenoids, diterpenoids, triterpenoids, flavonoids, and alkaloids) and chemical synthesis (e.g. pyrimidines, pyridines, pyrazines, quinoxalines, thiophenes, and thiazolidines). In addition, the biosynthetic pathway of novel natural IKKβ inhibitors and their biological potentials were discussed. This review will provide inspiration for the structural modification of IKKβ inhibitors based on the skeleton of natural products or chemical synthesis and further phytochemistry investigations.
Collapse
Affiliation(s)
- Juan Zhang
- School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- College of Pharmacy, Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518061, China
| | - Rui Zhang
- School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Li
- School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan
| | - Xiao-Chi Ma
- College of Pharmacy, Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Feng Qiu
- School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Cheng-Peng Sun
- School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- College of Pharmacy, Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
2
|
Reja S, Sarkar K, Mukherjee D, Fayaz T, Kumar P, Das P, Sanphui P, Das RK. 3, 3′-[succinylbis(diazaneyl)]bis(N,N,N-trimethylpropan-1-ammonium) perchlorate: Synthesis, characterization, computational studies and in vitro anticancer activity against the human colon carcinoma cell line (HT-29). J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134377] [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]
|
3
|
Di Francesco B, Verzella D, Capece D, Vecchiotti D, Di Vito Nolfi M, Flati I, Cornice J, Di Padova M, Angelucci A, Alesse E, Zazzeroni F. NF-κB: A Druggable Target in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:3557. [PMID: 35884618 PMCID: PMC9319319 DOI: 10.3390/cancers14143557] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is an aggressive hematological malignancy that relies on highly heterogeneous cytogenetic alterations. Although in the last few years new agents have been developed for AML treatment, the overall survival prospects for AML patients are still gloomy and new therapeutic options are still urgently needed. Constitutive NF-κB activation has been reported in around 40% of AML patients, where it sustains AML cell survival and chemoresistance. Given the central role of NF-κB in AML, targeting the NF-κB pathway represents an attractive strategy to treat AML. This review focuses on current knowledge of NF-κB's roles in AML pathogenesis and summarizes the main therapeutic approaches used to treat NF-κB-driven AML.
Collapse
|
4
|
An T, Yin H, Lu Y, Liu F. The Emerging Potential of Parthenolide Nanoformulations in Tumor Therapy. Drug Des Devel Ther 2022; 16:1255-1272. [PMID: 35517982 PMCID: PMC9063801 DOI: 10.2147/dddt.s355059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/05/2022] [Indexed: 12/21/2022] Open
Abstract
Plant-derived sesquiterpene lactones are promising natural sources for the discovery of anti-cancer drugs. As an extensively studied sesquiterpene lactone, the tumor suppression effect of parthenolide (PTL) has been clarified by targeting a number of prominent signaling pathways and key protein regulators in carcinogenesis. Notably, PTL was also the first small molecule reported to eradicate cancer stem cells. Nevertheless, the clinical application of PTL as an antitumor agent remains limited, owing to some disadvantages such as low water solubility and poor bioavailability. Thus, nanomedicine has attracted much interest because of its great potential for transporting poorly soluble drugs to desired body sites. In view of the significant advantages over their free small-molecule counterparts, nanoparticle delivery systems appear to be a potential solution for addressing the delivery of hydrophobic drugs, including PTL. In this review, we summarized the key anticancer mechanisms underlined by PTL as well as engineered PTL nanoparticles synthesized to date. Therefore, PTL nanoformulations could be an alternative strategy to maximize the therapeutic value of PTL.
Collapse
Affiliation(s)
- Tao An
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, People's Republic of China
| | - Huanhuan Yin
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, People's Republic of China
| | - Yanting Lu
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Feng Liu
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, People's Republic of China.,Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center (SDATC), Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
5
|
Chen R, Zhou C, Pang X, Liu J, Gu Y, Liu J, Li Z. Design, Synthesis, Anti-cancer Activities and Computational Analysis of Novel Diamides Conformationally Restricted by Cyclopropane. CHINESE J ORG CHEM 2022. [DOI: 10.6023/cjoc202106053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
6
|
Morais PAB, Francisco CS, de Paula H, Ribeiro R, Eloy MA, Javarini CL, Neto ÁC, Júnior VL. Semisynthetic Triazoles as an Approach in the Discovery of Novel Lead Compounds. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210126100227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Historically, medicinal chemistry has been concerned with the approach of organic
chemistry for new drug synthesis. Considering the fruitful collections of new molecular entities,
the dedicated efforts for medicinal chemistry are rewarding. Planning and search for new
and applicable pharmacologic therapies involve the altruistic nature of the scientists. Since
the 19th century, notoriously applying isolated and characterized plant-derived compounds in
modern drug discovery and various stages of clinical development highlight its viability and
significance. Natural products influence a broad range of biological processes, covering transcription,
translation, and post-translational modification, being effective modulators of most
basic cellular processes. The research of new chemical entities through “click chemistry”
continuously opens up a map for the remarkable exploration of chemical space towards leading
natural products optimization by structure-activity relationship. Finally, in this review, we expect to gather a
broad knowledge involving triazolic natural product derivatives, synthetic routes, structures, and their biological activities.
Collapse
Affiliation(s)
- Pedro Alves Bezerra Morais
- Centro de Ciencias Exatas, Naturais e da Saude, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Carla Santana Francisco
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Heberth de Paula
- Centro de Ciencias Exatas, Naturais e da Saude, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Rayssa Ribeiro
- Programa de Pos- Graduacao em Agroquimica, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Mariana Alves Eloy
- Programa de Pos- Graduacao em Agroquimica, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Clara Lirian Javarini
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Álvaro Cunha Neto
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Valdemar Lacerda Júnior
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| |
Collapse
|
7
|
Mandal SK, Debnath U, Kumar A, Thomas S, Mandal SC, Choudhury MD, Palit P. Natural Sesquiterpene Lactones in the Prevention and Treatment of Inflammatory Disorders and cancer: A Systematic Study of this Emerging Therapeutic Approach based on Chemical and Pharmacological Aspect. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817999200421144007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background and Introduction:
Sesquiterpene lactones are a class of secondary metabolite
that contains sesquiterpenoids and lactone ring as pharmacophore moiety. A large group of bioactive
secondary metabolites such as phytopharmaceuticals belong to this category. From the Asteraceae
family-based medicinal plants, more than 5,000 sesquiterpene lactones have been reported so
far. Sesquiterpene lactone-based pharmacophore moieties hold promise for broad-spectrum biological
activities against cancer, inflammation, parasitic, bacterial, fungal, viral infection and other functional
disorders. Moreover, these moiety based phytocompounds have been highlighted with a new
dimension in the natural drug discovery program worldwide after the 2015 Medicine Nobel Prize
achieved by the Artemisinin researchers.
Objective:
These bitter substances often contain an α, β-unsaturated-γ-lactone as a major structural
backbone, which in recent studies has been explored to be associated with anti-tumor, cytotoxic, and
anti-inflammatory action. Recently, the use of sesquiterpene lactones as phytomedicine has been
increased. This study will review the prospect of sesquiterpene lactones against inflammation and
cancer.
Methods:
Hence, we emphasized on the different features of this moiety by incorporating its structural
diversity on biological activities to explore structure-activity relationships (SAR) against inflammation
and cancer.
Results:
How the dual mode of action such as anti-inflammatory and anti-cancer has been exhibitedby
these phytopharmaceuticals will be forecasted in this study. Furthermore, the correlation of
anti-inflammatory and anti-cancer activity executed by the sesquiterpene lactones for fruitful phytotherapy
will also be revealed in the present review in the milieu of pharmacophore activity relation
and pharmacodynamics study as well.
Conclusion:
So, these metabolites are paramount in phytopharmacological aspects. The present discussion
on the future prospect of this moiety based on the reported literature could be a guide for
anti-inflammatory and anti-cancer drug discovery programs for the upcoming researchers.
Collapse
Affiliation(s)
- Sudip Kumar Mandal
- Department of Pharmaceutical Chemistry, Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur- 713206, India
| | - Utsab Debnath
- School of Pharmaceutical Technology, Adamas University, Kolkata 700126, India
| | - Amresh Kumar
- Department of Life Science and Bioinformatics, Biotech Hub, Assam University, Silchar, Assam-788011, India
| | - Sabu Thomas
- Mahatma Gandhi University, Kottayam-686560, Kerala, India
| | - Subhash Chandra Mandal
- Department of Pharmaceutical Technology, Pharmacognosy and Phytotherapy Research Laboratory, Jadavpur University, Kolkata 700032, India
| | - Manabendra Dutta Choudhury
- Department of Life Science and Bioinformatics, Biotech Hub, Assam University, Silchar, Assam-788011, India
| | - Partha Palit
- Department of Pharmaceutical Sciences, Drug Discovery Research Laboratory, Division of Pharmacognosy, Assam University (A Central University), Silchar-788011, India
| |
Collapse
|
8
|
Synthesis, characterization of some pyrazine derivatives as anti-cancer agents: In vitro and in Silico approaches. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
9
|
Qiu J, Yuan CM, Wen M, Li YN, Chen J, Jian JY, Huang LJ, Gu W, Li YM, Hao XJ. Design, synthesis, and cytotoxic activities of novel hybrids of parthenolide and thiazolidinedione via click chemistry. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2020; 22:425-433. [PMID: 31012734 DOI: 10.1080/10286020.2019.1597055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/14/2019] [Indexed: 06/09/2023]
Abstract
A series of novel parthenolide-thiazolidinedione hybrids have been synthesized via a click chemistry-mediated coupling between parthenolide and thiazolidinedione, and evaluated for their cytotoxic activities. The results indicated that all the hybrids showed moderate cytotoxic effects on human cancer cell lines, including human erythroleukemia cell line (HEL), prostate (PC3), and breast (MDA-MB-231) by MTT assay. In particular, compound VI-6 exhibited the best cytotoxic activities against the MDA-MB-231 cells with IC50 value of 2.07 µM, which was about eight times more active than that of the original compound (PTL). These interesting results might be used to develop novel lead scaffolds for potential anticancer agents.
Collapse
Affiliation(s)
- Jie Qiu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Chun-Mao Yuan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Min Wen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Ya-Nan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Juan Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Jun-You Jian
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Lie-Jun Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Wei Gu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Yan-Mei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Xiao-Jiang Hao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| |
Collapse
|
10
|
Freund RRA, Gobrecht P, Fischer D, Arndt HD. Advances in chemistry and bioactivity of parthenolide. Nat Prod Rep 2020; 37:541-565. [DOI: 10.1039/c9np00049f] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
(−)-Parthenolide is a germacrane sesquiterpene lactone, available in ample amounts from the traditional medical plant feverfew (Tanacetum parthenium).
Collapse
Affiliation(s)
- Robert R. A. Freund
- Institut für Organische Chemie und Makromolekulare Chemie
- Friedrich-Schiller-Universität
- D-07743 Jena
- Germany
| | - Philipp Gobrecht
- Lehrstuhl für Zellphysiologie
- Ruhr-Universität Bochum
- D-44780 Bochum
- Germany
| | - Dietmar Fischer
- Lehrstuhl für Zellphysiologie
- Ruhr-Universität Bochum
- D-44780 Bochum
- Germany
| | - Hans-Dieter Arndt
- Institut für Organische Chemie und Makromolekulare Chemie
- Friedrich-Schiller-Universität
- D-07743 Jena
- Germany
| |
Collapse
|
11
|
Qu W, Yang Q, Wang G, Wang Z, Huang P, Huang W, Zhang R, Yan D. Amphiphilic irinotecan–melampomagnolide B conjugate nanoparticles for cancer chemotherapy. RSC Adv 2020; 10:8958-8966. [PMID: 35496516 PMCID: PMC9050120 DOI: 10.1039/d0ra00912a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Melampomagnolide B (MMB) is a natural sesquiterpene lactone product structurally related to parthenolide (PTL). Although MMB has been widely used to treat various types of cancers, such as glioma, leukemia and colon cancer, the effective delivery of MMB to cancer cells remains a challenge. An amphiphilic drug–drug conjugate (ADDC) strategy has been proposed and developed as a promising drug self-delivery system for cancer therapy because of its simple preparation, carrier-free nature, and high therapeutic activity. Herein, we present a new ADDC, which is synthesized by linking the hydrophilic anticancer drug irinotecan (Ir) and the hydrophobic anticancer drug MMB through a carbonate bond. The obtained amphiphilic irinotecan–melampomagnolide B conjugate (Ir–C–MMB) can self-assemble in water into stable nanoparticles with an average diameter of around 122.1 nm. After cellular uptake, the carbonate bond between the hydrophilic drug and hydrophobic drug can be cleaved to release free Ir and MMB under acidic conditions, which exhibit a synergistic effect in tumor cells. MTT results reveal that the Ir–C–MMB nanoparticles can effectively inhibit proliferation of cancer cells. The apoptosis data indicate that the apoptosis rate of cells treated with Ir–C–MMB nanoparticles is about 50% within 24 h, which is much higher than that of free Ir or MMB. Our results suggest that this ADDC strategy could be used as a drug delivery platform for MMB and its derivatives, and that it offers effective synergistic therapeutic efficacy. Ir–C–MMB nanoparticles can be easily fabricated using an ADDC strategy, and used as a MMB drug self-delivery platform for synergistic cancer therapy.![]()
Collapse
Affiliation(s)
- Wenhao Qu
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Quanjun Yang
- Department of Pharmacy
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Guanchun Wang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Zhaohong Wang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Ping Huang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Wei Huang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Rong Zhang
- Department of Obstetrics and Gynecology
- Fengxian Hospital
- Southern Medical University
- Shanghai 201499
- China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| |
Collapse
|
12
|
Naaz F, Ahmad F, Lone BA, Pokharel YR, Fuloria NK, Fuloria S, Ravichandran M, Pattabhiraman L, Shafi S, Shahar Yar M. Design and synthesis of newer 1,3,4-oxadiazole and 1,2,4-triazole based Topsentin analogues as anti-proliferative agent targeting tubulin. Bioorg Chem 2019; 95:103519. [PMID: 31884140 DOI: 10.1016/j.bioorg.2019.103519] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/01/2019] [Accepted: 12/12/2019] [Indexed: 01/14/2023]
Abstract
A set of two series of 1,3,4-oxadiazole (11a-n) and 1,2,4-Triazole (12a, c, e, g, h, j-n) based topsentin analogues were prepared by replacing imizadole moiety of topsentin through a multistep synthesis starting from indole. All the compounds synthesized were submitted for single dose (10 µM) screening against a NCI panel of 60-human cancer cell lines. Among all cancer cell lines, colon (HCC-2998) and Breast (MCF-7, T-47D) cancer cell lines were found to be more susceptible for this class of compounds. Among the compounds tested, compounds 11a, 11d, 11f, 12e and 12h, were exhibited good anti-proliferative activity against various cancer cell lines. Compounds 11d, 12e and 12h demonstrated better activity with IC50 2.42 µM, 3.06 µM, and 3.30 µM respectively against MCF-7 human cancer cell line than that of the standard drug doxorubicin IC50 6.31 µM. Furthermore, 11d induced cell cycle arrest at G0/G1 phase and also disrupted mitochondrial membrane potential with reducing cell migration potential of MCF-7 cells in dose dependent manner. In vitro microtubule polymerization assays found that compound 11d disrupt tubulin dynamics by inhibiting tubulin polymerization with IC50 3.89 μM compared with standard nocodazole (IC50 2.49 μM). In silico docking studies represented that 11d was binding at colchicine binding site of β-tubulin. Compound 11d emerged as lead molecule from the library of compounds tested and this may serve as a template for further drug discovery.
Collapse
Affiliation(s)
- Fatima Naaz
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Faiz Ahmad
- Faculty of Life Sciences and Biology, South Asian University, New Delhi 110021, India
| | - Bilal Ahmad Lone
- Faculty of Life Sciences and Biology, South Asian University, New Delhi 110021, India
| | - Yuba Raj Pokharel
- Faculty of Life Sciences and Biology, South Asian University, New Delhi 110021, India
| | - Neeraj Kumar Fuloria
- Faculty of Pharmacy, AIMST University, Semeling Campus, Jalan Bedong-Semeling, Bedong, Kedah Darul Aman 08100, Malaysia
| | - Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Semeling Campus, Jalan Bedong-Semeling, Bedong, Kedah Darul Aman 08100, Malaysia
| | - Manickam Ravichandran
- Faculty of Applied Science, AIMST University, Semeling Campus, Jalan Bedong-Semeling, Bedong, Kedah Darul Aman 08100, Malaysia
| | - Lalitha Pattabhiraman
- Faculty of Medical Sciences, AIMST University, Semeling Campus, Jalan Bedong-Semeling, Bedong, Kedah Darul Aman 08100, Malaysia
| | - Syed Shafi
- Department of Chemistry, School of Chemical and Life Science, Jamia Hamdard, New Delhi, India.
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
13
|
Govindaraju S, Roshini A, Lee MH, Yun K. Kaempferol conjugated gold nanoclusters enabled efficient for anticancer therapeutics to A549 lung cancer cells. Int J Nanomedicine 2019; 14:5147-5157. [PMID: 31371953 PMCID: PMC6636439 DOI: 10.2147/ijn.s209773] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/15/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Kaempferol (K) is a recognized anticancer drug that can conjugate with small-size gold nanoclusters (AuNCs). Materials and methods: K-AuNCs were synthesized and their use as an anticancer drug was explored using A549 lung cancer cells. Colony formation and cell migration assays were carried out. The morphology of the K-AuNCs treated A549 cells was explored using bio-atomic force microscopy. Results: The K-AuNCs were 1-3 nm in diameter and emitted strong fluorescent at 650 nm following excitation at 550 nm. The stretching and bending nature of the K-AuNCs were analyzed by the Fourier transform infrared spectroscopy. The presence of kaempferol in the AuNCs were confirmed by the PL spectroscopy. Conclusion: The synthesized K-AuNCs mainly targeted and damaged the nuclei of the cancer cells. This composite nanocluster was less toxicity to the normal human cell and higher toxicity to the A549 lunch cancer cell and these material is potential for anticancer drug delivery and bio imaging applications.
Collapse
Affiliation(s)
- Saravanan Govindaraju
- Department of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Arivazhagan Roshini
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Min-Ho Lee
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Kyusik Yun
- Department of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Republic of Korea
| |
Collapse
|
14
|
Lamture G, Crooks PA, Borrelli MJ. Actinomycin-D and dimethylamino-parthenolide synergism in treating human pancreatic cancer cells. Drug Dev Res 2018; 79:287-294. [PMID: 30295945 PMCID: PMC6193836 DOI: 10.1002/ddr.21441] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022]
Abstract
Preclinical Research & Development Pancreatic cancer is the third leading cause of death in the US with a poor 5-year survival rate of 8.5%. A novel anti-cancer drug, dimethylamino parthenolide (DMAPT), is the water-soluble analog of the natural sesquiterpene lactone, parthenolide. The putative modes of action of DMAPT are inhibition of the Nuclear chain factor kappa-light-chain enhancer of activated B cells (NFκB) pathway and depletion of glutathione levels; the latter causing cancer cells to be more susceptible to oxidative stress-induced cell death. Actinomycin-D (ActD) is a polypeptide antibiotic that binds to DNA, and inhibits RNA and protein synthesis by inhibiting RNA polymerase II. A phase 2 clinical trial indicated that ActD could be a potent drug against pancreatic cancer; however, it was not a favored drug due to toxicity issues. New drug entities and methods of drug delivery, used alone or in combination, are needed to treat pancreatic cancer more effectively. Thus, it was postulated that combining DMAPT and ActD would result in synergistic inhibition of Panc-1 pancreatic cancer cell growth because DMAPT's inhibition of NFκB would enhance induction of apoptosis by ActD, via phosphorylation of c-Jun, by minimizing NFκB inhibition of c-Jun phosphorylation. Combining these two drugs induced a higher level of cell death than each drug alone. A fixed drug ratio of DMAPT: ActD (1,200:1) was used. Data from metabolic (MTT) and colony formation assays were analyzed for synergism with CompuSyn software, which utilizes the Chou-Talalay equation. The analyses indicated synergism and moderate synergism at combination concentrations of DMAPT/ActD of 12/0.01 and 18/0.015 μM, respectively.
Collapse
Affiliation(s)
- Gauri Lamture
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Peter A Crooks
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | |
Collapse
|
15
|
Janganati V, Ponder J, Balasubramaniam M, Bhat-Nakshatri P, Bar EE, Nakshatri H, Jordan CT, Crooks PA. MMB triazole analogs are potent NF-κB inhibitors and anti-cancer agents against both hematological and solid tumor cells. Eur J Med Chem 2018; 157:562-581. [PMID: 30121494 DOI: 10.1016/j.ejmech.2018.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 10/28/2022]
Abstract
Triazole derivatives of melampomagnolide B (MMB) have been synthesized via click chemistry methodologies and screened against a panel of 60 human cancer cell lines. Several derivatives showed promising anti-cancer activity, affording growth inhibition (GI50) values in the nanomolar range (GI50 = 0.02-0.99 μM). Lead compound 7h exhibited EC50 values of 400 nM and 700 nM, respectively, against two AML clinical specimens. Compound 7h was significantly more potent than parthenolide as an inhibitor of p65 phosphorylation in both hematological and solid tumor cell lines, indicating its ability to inhibit the NF-κB pathway. In TMD-231 breast cancer cells, treatment with 7h reduced DNA binding activity of NF-κB through inhibition of IKK-β mediated p65 phosphorylation and caused elevation of basal IκBα levels through inhibition of constitutive IκBα turnover and NF-κB activation. Molecular docking and dynamic modeling studies indicated that 7h interacts with the kinase domain of the monomeric IKKβ subunit, leading to inhibition of IKKβ activation, and compromising phosphorylation of downstream targets of the NF-κB pathway; dynamic modeling studies show that this interaction also causes unwinding of the α-helix of the NEMO binding site on IKKβ. Molecular docking studies with 10, a water-soluble analog of 7h, demonstrate that this analog interacts with the dimerization/oligomerization domain of monomeric IKKβ and may inhibit oligomer formation and subsequent autophosphorylation. Sesquiterpene lactones 7h and 10 are considered ideal candidates for potential clinical development.
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Jessica Ponder
- Division of Hematology, University of Colorado, Aurora, CO, 80045, USA; Department of Toxicology, University of Colorado, Aurora, CO, 80045, USA
| | | | - Poornima Bhat-Nakshatri
- Department of Surgery, Indiana School of Medicine, Indianapolis, IN, 46202, USA; Department of Biochemistry and Molecular Biology, Indiana School of Medicine, Indianapolis, IN, 46202, USA
| | - Eli E Bar
- Department of Neurological Surgery, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana School of Medicine, Indianapolis, IN, 46202, USA; Department of Biochemistry and Molecular Biology, Indiana School of Medicine, Indianapolis, IN, 46202, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Aurora, CO, 80045, USA; Department of Toxicology, University of Colorado, Aurora, CO, 80045, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
16
|
Ding Y, Guo H, Ge W, Chen X, Li S, Wang M, Chen Y, Zhang Q. Copper(I) oxide nanoparticles catalyzed click chemistry based synthesis of melampomagnolide B-triazole conjugates and their anti-cancer activities. Eur J Med Chem 2018; 156:216-229. [PMID: 30006167 DOI: 10.1016/j.ejmech.2018.06.058] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/15/2018] [Accepted: 06/23/2018] [Indexed: 01/07/2023]
Abstract
A series of thirty one melampomagnolide B-triazole conjugates was synthesized via Copper(I) oxide nanoparticles catalyzed click chemistry. These conjugates were evaluated for their anti-cancer activities against a panel of five human cancer cell lines. The most active compound 6e showed high activity against HCT116 cell line with IC50 value of 0.43 μM, which demonstrated 11.5-fold improvement compared to that of the parent compound melampomagnolide B (IC50 = 4.93 μM). Compound 6e showed significant efficacy of inducing apoptosis, inhibiting proliferation and migration of HCT116 cells. The preliminary molecular mechanism of 6e was also investigated. On the base of these results, compound 6e might be considered as a promising candidate for further evaluation as a potential anti-cancer drug.
Collapse
Affiliation(s)
- Yahui Ding
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Hongyu Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Weizhi Ge
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Xinyi Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Shengzu Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Mengmeng Wang
- Accendatech Company, Ltd., Tianjin, 300384, People's Republic of China
| | - Yue Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Quan Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| |
Collapse
|