51
|
Huang Q, Chen H, Ren Y, Wang Z, Zeng P, Li X, Wang J, Zheng X. Anti-hepatocellular carcinoma activity and mechanism of chemopreventive compounds: ursolic acid derivatives. PHARMACEUTICAL BIOLOGY 2016; 54:3189-3196. [PMID: 27564455 DOI: 10.1080/13880209.2016.1214742] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 07/13/2016] [Indexed: 06/06/2023]
Abstract
CONTEXT Hepatocellular carcinoma (HCC) is a common cancer around the world, with high mortality rate. Currently, there is no effective drug for the therapy of HCC. Ursolic acid (UA) is a natural product which exists in various medicinal herbs and fruits, exhibiting multiple biological effects such as its outstanding anticancer and hepatoprotective activity, which has drawn many pharmacists' attention. OBJECTIVE This paper summarizes the current status of the hepatoprotective activity of UA analogues and explains the related mechanism, providing a clear direction for the development of novel anti-HCC drugs. METHODS All of the data resources were derived from PubMed. By comparing the IC50 values and analyzing the structure-activity relationships, we listed compounds with good pharmacological activity from the relevant literature, and summarized their anti-HCC mechanism. RESULTS From the database, 58 new UA derivatives possessing wonderful anticancer and hepatoprotective effects were listed, and the relevant anti-HCC mechanism were discussed. CONCLUSION UA's anti-HCC effect is the result of combined action of many mechanisms. These 58 new UA derivatives, particularly compounds 45 and 53, can be used as potential drugs for the treatment of liver cancer.
Collapse
Affiliation(s)
- Qiuxia Huang
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
| | - Hongfei Chen
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
| | - Yuyan Ren
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
| | - Zhe Wang
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
| | - Peiyu Zeng
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
- c Research Interest Group of Pharmacy , University of South China , Hengyang , China
| | - Xuan Li
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
- c Research Interest Group of Pharmacy , University of South China , Hengyang , China
| | - Juan Wang
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
| | - Xing Zheng
- a Department of Pharmacy & Pharmacology , University of South China , Hengyang , China
- b Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , Hengyang , China
| |
Collapse
|
52
|
Nguyen BCQ, Takahashi H, Uto Y, Shahinozzaman MD, Tawata S, Maruta H. 1,2,3-Triazolyl ester of Ketorolac: A "Click Chemistry"-based highly potent PAK1-blocking cancer-killer. Eur J Med Chem 2016; 126:270-276. [PMID: 27889630 DOI: 10.1016/j.ejmech.2016.11.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/04/2016] [Accepted: 11/18/2016] [Indexed: 10/20/2022]
Abstract
An old anti-inflammatory/analgesic drug called Toradol is a racemic form of Ketorolac (50% R-form and 50% S-form) that blocks the oncogenic RAC-PAK1-COX-2 (cyclooxygenase-2) signaling, through the direct inhibition of RAC by the R-form and of COX-2 by the S-form, eventually down-regulating the production of prostaglandins. However, due to its COOH moiety which is clearly repulsive to negatively-charged phospholipid-based plasma membrane, its cell-permeability is rather poor (the IC50 against the growth of human cancer cells such as A549 is around 13 μM). In an attempt to boost its anti-cancer activity, hopefully by increasing its cell-permeability through abolishing the negative charge, yet keeping its water-solubility, here we synthesized a 1,2,3-triazolyl ester of Toradol through "Click Chemistry". The resultant water-soluble "azo" derivative called "15K" was found to be over 500 times more potent than Toradol with the IC50 around 24 nM against the PAK1-dependent growth of A549 cancer cells, inactivating PAK1 in cell culture with the apparent IC50 around 65 nM, and inhibiting COX-2 in vitro with the IC50 around 6 nM. Furthermore, the Click Chemistry boosts the anti-cancer activity of Ketorolac by 5000 times against the PAK1-independent growth of B16F10 melanoma cells. Using a multi-drug-resistant (MDR) cancer cell line (EMT6), we found that the esterization of Ketorolac boosts its cell-permeability by at least 10 folds. Thus, the Click Chemistry dramatically boosts the anti-cancer activity of Ketorolac, at least in three ways: increasing its cell-permeability, the anti-PAK1 activity of R-form and anti-COX-2 activity of S-form. The resultant "15K" is so far among the most potent PAK1-blockers, and therefore would be potentially useful for the therapy of many different PAK1-dependent diseases/disorders such as cancers.
Collapse
Affiliation(s)
- Binh Cao Quan Nguyen
- PAK Research Center (Lab), Okinawa, Japan; Kagoshima University, Kagoshima, Japan
| | | | | | - M D Shahinozzaman
- PAK Research Center (Lab), Okinawa, Japan; Kagoshima University, Kagoshima, Japan
| | | | - Hiroshi Maruta
- PAK Research Center (Lab), Okinawa, Japan; PAK Research Center (Office), Melbourne, Australia.
| |
Collapse
|
53
|
Mendes VIS, Bartholomeusz GA, Ayres M, Gandhi V, Salvador JAR. Synthesis and cytotoxic activity of novel A-ring cleaved ursolic acid derivatives in human non-small cell lung cancer cells. Eur J Med Chem 2016; 123:317-331. [PMID: 27484517 PMCID: PMC5652311 DOI: 10.1016/j.ejmech.2016.07.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 12/19/2022]
Abstract
Ursolic acid (UA) is a pentacyclic triterpenoid with recognized anticancer properties. We prepared a series of new A-ring cleaved UA derivatives and evaluated their antiproliferative activity in non-small cell lung cancer (NSCLC) cell lines using 2D and 3D culture models. Compound 17, bearing a cleaved A-ring with a secondary amide at C3, was found to be the most active compound, with potency in 2D systems. Importantly, even in 3D systems, the effect was maintained albeit a slight increase in the IC50. The molecular mechanism underlying the anticancer activity was further investigated. Compound 17 induced apoptosis via activation of caspase-8 and caspase-7 and via decrease of Bcl-2. Moreover, induction of autophagy was also detected with increased levels of Beclin-1 and LC3A/B-II and decreased levels of mTOR and p62. DNA synthetic capacity and cell cycle profiles were not affected by the drug, but total RNA synthesis was modestly but significantly decreased. Given its activity and mechanism of action, compound 17 might represent a potential candidate for further cancer research.
Collapse
Affiliation(s)
- Vanessa I. S. Mendes
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal. ; Fax: + 351 239 488 503; Tel: + 351 239 488 400
- Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Geoffrey A. Bartholomeusz
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. ; Fax: + 1 713-794-4316; Tel: + 1 713-792-2989
| | - Mary Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. ; Fax: + 1 713-794-4316; Tel: + 1 713-792-2989
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. ; Fax: + 1 713-794-4316; Tel: + 1 713-792-2989
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal. ; Fax: + 351 239 488 503; Tel: + 351 239 488 400
- Center for Neuroscience and Cell Biology, Coimbra, Portugal
| |
Collapse
|
54
|
Hajipour AR, Karimzadeh M, Fakhari F, Karimi H. CuFeO 2/tetrabutylammonium bromide catalyzes selective synthesis of 1,4-disubstituted 1,2,3-triazoles in neat water at room temperature. Appl Organomet Chem 2016. [DOI: 10.1002/aoc.3526] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Abdol R. Hajipour
- Pharmaceutical Research Laboratory, Department of Chemistry; Isfahan University of Technology; Isfahan 84156 Islamic Republic of Iran
- Department of Neuroscience; University of Wisconsin, Medical School; 1300 University Avenue Madison WI 53706-1532 USA
| | - Morteza Karimzadeh
- Pharmaceutical Research Laboratory, Department of Chemistry; Isfahan University of Technology; Isfahan 84156 Islamic Republic of Iran
| | - Farzaneh Fakhari
- Pharmaceutical Research Laboratory, Department of Chemistry; Isfahan University of Technology; Isfahan 84156 Islamic Republic of Iran
| | - Hirbod Karimi
- Pharmaceutical Research Laboratory, Department of Chemistry; Isfahan University of Technology; Isfahan 84156 Islamic Republic of Iran
| |
Collapse
|
55
|
Akhtar J, Khan AA, Ali Z, Haider R, Shahar Yar M. Structure-activity relationship (SAR) study and design strategies of nitrogen-containing heterocyclic moieties for their anticancer activities. Eur J Med Chem 2016; 125:143-189. [PMID: 27662031 DOI: 10.1016/j.ejmech.2016.09.023] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/04/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
Abstract
The present review article offers a detailed account of the design strategies employed for the synthesis of nitrogen-containing anticancer agents. The results of different studies describe the N-heterocyclic ring system is a core structure in many synthetic compounds exhibiting a broad range of biological activities. Benzimidazole, benzothiazole, indole, acridine, oxadiazole, imidazole, isoxazole, pyrazole, triazoles, quinolines and quinazolines including others drugs containing pyridazine, pyridine and pyrimidines are covered. The following studies of these compounds suggested that these compounds showed their antitumor activities through multiple mechanisms including inhibiting protein kinase (CDK, MK-2, PLK1, kinesin-like protein Eg5 and IKK), topoisomerase I and II, microtubule inhibition, and many others. Our concise representation exploits the design and anticancer potency of these compounds. The direct comparison of anticancer activities with the standard enables a systematic analysis of the structure-activity relationship among the series.
Collapse
Affiliation(s)
- Jawaid Akhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi, 110062, India
| | - Ahsan Ahmed Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi, 110062, India
| | - Zulphikar Ali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi, 110062, India
| | - Rafi Haider
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi, 110062, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi, 110062, India.
| |
Collapse
|
56
|
Pham Thi T, Le Nhat TG, Ngo Hanh T, Luc Quang T, Pham The C, Dang Thi TA, Nguyen HT, Nguyen TH, Hoang Thi P, Van Nguyen T. Synthesis and cytotoxic evaluation of novel indenoisoquinoline-substituted triazole hybrids. Bioorg Med Chem Lett 2016; 26:3652-7. [PMID: 27342752 DOI: 10.1016/j.bmcl.2016.05.092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 01/14/2023]
Abstract
The synthesis of various substituted triazole-indenoisoquinoline hybrids was performed based on a CuI-catalyzed 1,3-cycloaddition between propargyl-substituted derivatives and the azide-containing indenoisoquinoline. Besides, a variety of N-(alkyl)propargylindenoisoquinolines was used as substrates for the construction of triazole-indenoisoquinoline-AZT conjugated via a click chemistry-mediated coupling with 3'-azido-3'-deoxythymidine (AZT). Thus, twenty three new indenoisoquinoline-substituted triazole hybrids were successfully prepared and evaluated as cytotoxic agents, revealing an interesting anticancer activity of four triazole linker-indenoisoquinoline-AZT hybrids in KB and HepG2 cancer cell lines.
Collapse
Affiliation(s)
- Tham Pham Thi
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam; Thuyloi University, 175, Tay Son, Hanoi, Vietnam
| | - Thuy Giang Le Nhat
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Thuong Ngo Hanh
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Tan Luc Quang
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam; Hanoi Pedagogical University No. 2, Vietnam
| | - Chinh Pham The
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam; Thainguyen University of Science, Tanthinh, Thainguyen, Vietnam
| | - Tuyet Anh Dang Thi
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Ha Thanh Nguyen
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Thu Ha Nguyen
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Phuong Hoang Thi
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Tuyen Van Nguyen
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
57
|
Nguyen BCQ, Tawata S. The Chemistry and Biological Activities of Mimosine: A Review. Phytother Res 2016; 30:1230-42. [PMID: 27213712 DOI: 10.1002/ptr.5636] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/05/2016] [Accepted: 04/12/2016] [Indexed: 12/14/2022]
Abstract
Mimosine [β-[N-(3-hydroxy-4-oxypyridyl)]-α-aminopropionic acid] is a non-protein amino acid found in the members of Mimosoideae family. There are a considerable number of reports available on the chemistry, methods for estimation, biosynthesis, regulation, and degradation of this secondary metabolite. On the other hand, over the past years of active research, mimosine has been found to have various biological activities such as anti-cancer, antiinflammation, anti-fibrosis, anti-influenza, anti-virus, herbicidal and insecticidal activities, and others. Mimosine is a leading compound of interest for use in the development of RAC/CDC42-activated kinase 1 (PAK1)-specific inhibitors for the treatment of various diseases/disorders, because PAK1 is not essential for the growth of normal cells. Interestingly, the new roles of mimosine in malignant glioma treatment, regenerative dentistry, and phytoremediation are being emerged. These identified properties indicate an exciting future for this amino acid. The present review is focused on the chemistry and recognized biological activities of mimosine in an attempt to draw a link between these two characteristics. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Binh Cao Quan Nguyen
- Department of Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan.,PAK Research Center, Okinawa, 903-0213, Japan
| | - Shinkichi Tawata
- PAK Research Center, Okinawa, 903-0213, Japan.,Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus, Senbaru 1, Nishihara-cho, Okinawa, 903-0213, Japan
| |
Collapse
|
58
|
Lone AM, Dar NJ, Hamid A, Shah WA, Ahmad M, Bhat BA. Promise of Retinoic Acid-Triazolyl Derivatives in Promoting Differentiation of Neuroblastoma Cells. ACS Chem Neurosci 2016; 7:82-9. [PMID: 26551203 DOI: 10.1021/acschemneuro.5b00267] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Retinoic acid induces differentiation in various types of cells including skeletal myoblasts and neuroblasts and maintains differentiation of epithelial cells. The present study demonstrates synthesis and screening of a library of retinoic acid-triazolyl derivatives for their differentiation potential on neuroblastoma cells. Click chemistry approach using copper(I)-catalyzed azide-alkyne cycloaddition was adopted for the preparation of these derivatives. The neurite outgrowth promoting potential of retinoic acid-triazolyl derivatives was studied on neuroblastoma cells. Morphological examination revealed that compounds 8a, 8e, 8f, and 8k, among the various derivatives screened, exhibited promising neurite-outgrowth inducing activity at a concentration of 10 μM compared to undifferentiated and retinoic acid treated cells. Further on, to confirm this differentiation potential of these compounds, neuroblastoma cells were probed for expression of neuronal markers such as NF-H and NeuN. The results revealed a marked increase in the NF-H and NeuN protein expression when treated with 8a, 8e, 8f, and 8k compared to undifferentiated and retinoic acid treated cells. Thus, these compounds could act as potential leads in inducing neuronal differentiation for future studies.
Collapse
Affiliation(s)
| | - Nawab John Dar
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Abid Hamid
- Cancer
Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Wajaht Amin Shah
- Department
of Chemistry, University of Kashmir, Srinagar 190006, India
| | - Muzamil Ahmad
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Bilal A. Bhat
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| |
Collapse
|
59
|
Nguyen BCQ, Taira N, Maruta H, Tawata S. Artepillin C and Other Herbal PAK1-blockers: Effects on Hair Cell Proliferation and Related PAK1-dependent Biological Function in Cell Culture. Phytother Res 2015; 30:120-7. [PMID: 26537230 DOI: 10.1002/ptr.5510] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/14/2015] [Accepted: 10/14/2015] [Indexed: 12/28/2022]
Abstract
PAK1 (RAC/CDC42-activated kinase 1) is the major oncogenic kinase, and a number of herbal PAK1-blockers such as propolis and curcumin have been shown to be anti-oncogenic and anti-melanogenic as well as anti-alopecia (promoting hair growth). Previously, we found several distinct PAK1-inhibitors in Okinawa plants including Alpinia zerumbet (alpinia). Thus, here, we tested the effects of these herbal compounds and their derivatives on the growth of cancer or normal hair cells, and melanogenesis in cell culture of A549 lung cancer, hair follicle dermal papilla cell, and B16F10 melanoma. Among these herbal PAK1-inhibitors, cucurbitacin I from bitter melon (Goya) turned out to be the most potent to inhibit the growth of human lung cancer cells with the IC50 around 140 nM and to promote the growth of hair cells with the effective dose around 10 nM. Hispidin, a metabolite of 5,6-dehydrokawain from alpinia, inhibited the growth of cancer cells with the IC50 of 25 μM as does artepillin C, the major anti-cancer ingredient in Brazilian green propolis. Mimosine tetrapeptides (MFWY, MFYY, and MFFY) and hispidin derivatives (H1-3) also exhibited a strong anti-cancer activity with the IC50 ranging from 16 to 30 μM. Mimosine tetrapeptides and hispidin derivatives strongly suppressed the melanogenesis in melanoma cells.
Collapse
Affiliation(s)
- Binh Cao Quan Nguyen
- Department of Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-8580, Japan
| | - Nozomi Taira
- Department of Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-8580, Japan
| | | | - Shinkichi Tawata
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus, Senbaru 1, Nishihara-cho, Okinawa, 903-0213, Japan
| |
Collapse
|
60
|
Xu B, Chu F, Zhang Y, Wang X, Li Q, Liu W, Xu X, Xing Y, Chen J, Wang P, Lei H. A Series of New Ligustrazine-Triterpenes Derivatives as Anti-Tumor Agents: Design, Synthesis, and Biological Evaluation. Int J Mol Sci 2015; 16:21035-55. [PMID: 26404253 PMCID: PMC4613240 DOI: 10.3390/ijms160921035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 01/11/2023] Open
Abstract
A series of novel ligustrazine-triterpenes derivatives was designed, synthesized and screened for their cytotoxicity against five cancer cell lines (Bel-7402, HepG2, HT-29, Hela, and MCF-7) and Madin-Darby canine kidney (MDCK). Current study suggested that most of the ligustrazine-triterpenes conjunctions showed better cytotoxicity than the starting materials. In particular, compound 4a exhibited better cytotoxic activity (IC50 < 5.23 μM) against Bel-7402, HT-29, MCF-7, Hela, and HepG2 than the standard anticancer drug cisplatin (DDP). The cytotoxicity selectivity detection revealed that 4a exhibited low cytotoxicity (IC50 > 20 μM) towards MDCK cells. A combination of fluorescence staining observation and flow cytometric analysis indicated that 4a could induce HepG2 cell apoptosis. Further studies suggested that 4a-induced apoptosis is mediated through depolarization of the mitochondrial membrane potential and increase of intracellular free Ca2+ concentration. In addition, the structure-activity relationships of these derivatives were briefly discussed.
Collapse
Affiliation(s)
- Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Fuhao Chu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Yuzhong Zhang
- Department of Pathology, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Xiaobo Wang
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Qiang Li
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Wei Liu
- School of Management, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Xin Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Yanyi Xing
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Jing Chen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
61
|
Fu HJ, Zhao Y, Zhou YR, Bao BH, Du Y, Li JX. Ursolic acid derivatives as bone anabolic agents targeted to tryptophan hydroxylase 1 (Tph-1). Eur J Pharm Sci 2015; 76:33-47. [PMID: 25930119 DOI: 10.1016/j.ejps.2015.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/15/2015] [Accepted: 04/26/2015] [Indexed: 01/28/2023]
Abstract
Tryptophan hydroxylase 1 (Tph-1) initiates the biosynthesis of peripheral serotonin. As peripheral serotonin suppresses bone formation, inhibitor of Tph-1 provides a useful tool to discover anabolic agents for osteoporosis. In the present study, series of ursolic acid (UA) derivatives were synthesized, and their inhibitory activity on serotonin biosynthesis and cytotoxicity were evaluated. Among the derivatives, 8d with potent inhibitory activity on serotonin was applied for further research. The data revealed that 8d significantly inhibited protein and mRNA expressions of Tph-1, and an SPR study indicated that 8d directly interacted to Tph-1 with a binding affinity of KD=15.09μM. Oral administration of 8d significantly prevented bone loss via suppressing serotonin biosynthesis without estrogenic side-effects in ovariectomized (OVX) rats.
Collapse
Affiliation(s)
- Hai-Jian Fu
- State Key Lab of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yang Zhao
- State Key Lab of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yu-Ren Zhou
- State Key Lab of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Bei-Hua Bao
- State Key Lab of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yun Du
- State Key Lab of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Jian-Xin Li
- State Key Lab of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, PR China.
| |
Collapse
|
62
|
Hua SX, Huang RZ, Ye MY, Pan YM, Yao GY, Zhang Y, Wang HS. Design, synthesis and in vitro evaluation of novel ursolic acid derivatives as potential anticancer agents. Eur J Med Chem 2015; 95:435-52. [PMID: 25841199 DOI: 10.1016/j.ejmech.2015.03.051] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/20/2015] [Accepted: 03/21/2015] [Indexed: 10/23/2022]
Abstract
A series of novel ursolic acid (UA) derivatives modified at the C-3 and the C-28 positions were designed and synthesized in an attempt to develop potential antitumor agents. The in vitro cytotoxicity were evaluated against five cancer cell lines (MGC-803, HCT-116, T24, HepG2 and A549 cell lines) and a normal cell (HL-7702) by MTT assay. The screening results indicated that some of these target compounds displayed moderate to high levels of antiproliferative activities compared with ursolic acid and 5-fluorouracil (5-FU), and exhibited much lower cytotoxicity than 5-FU, indicating that the targeted compounds had selective and significant effect on the cell lines. The induction of apoptosis and affects on the cell cycle distribution of compound 6r were investigated by acridine orange/ethidium bromide staining, Hoechst 33258 staining, JC-1 mitochondrial membrane potential staining and flow cytometry, which revealed that the antitumor activity of 6r was possibly achieved through the induction of cell apoptosis by G1 cell-cycle arrest. Western blot and qRT-PCR (quantitative real-time PCR) experiments demonstrated that compound 6r may induce apoptosis through both of intrinsic and extrinsic apoptosis pathway.
Collapse
Affiliation(s)
- Shi-Xian Hua
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China
| | - Ri-Zhen Huang
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China
| | - Man-Yi Ye
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China
| | - Ying-Ming Pan
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China
| | - Gui-Yang Yao
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China
| | - Ye Zhang
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China; Department of Chemistry & Pharmaceutical Science, Guilin Normal College, Xinyi Road 15, Guangxi 541001, PR China.
| | - Heng-Shan Wang
- State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmaceutical Science of Guangxi Normal University, Yucai Road 15, Guilin 541004, Guangxi, PR China.
| |
Collapse
|
63
|
Chen H, Gao Y, Wang A, Zhou X, Zheng Y, Zhou J. Evolution in medicinal chemistry of ursolic acid derivatives as anticancer agents. Eur J Med Chem 2015; 92:648-55. [PMID: 25617694 PMCID: PMC4336574 DOI: 10.1016/j.ejmech.2015.01.031] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/28/2014] [Accepted: 01/15/2015] [Indexed: 12/12/2022]
Abstract
Currently, there is a renewed interest in common dietaries and plant-based traditional medicines for the prevention and treatment of cancer. In the search for potential anticancer agents from natural sources, ursolic acid (UA), a pentacyclic triterpenoid widely found in various medicinal herbs and fruits, exhibits powerful biological effects including its attractive anticancer activity against various types of cancer cells. However, the limited solubility, rapid metabolism and poor bioavailability of UA restricted its further clinical applications. In the past decade, with substantial progress toward the development of new chemical entities for the treatment of cancer, numerous UA derivatives have been designed and prepared to overcome its disadvantages. Despite extensive effort, discovery of effective UA derivatives has so far met with only limited success. This review summarizes the current status of the structural diversity and evolution in medicinal chemistry of UA analogues and provides a detailed discussion of future direction for further research in the chemical modifications of UA.
Collapse
Affiliation(s)
- Haijun Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China; Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Basic Science Building 3.314, Galveston, TX 77555, United States
| | - Yu Gao
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Ailan Wang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Xiaobin Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yunquan Zheng
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Basic Science Building 3.314, Galveston, TX 77555, United States.
| |
Collapse
|
64
|
Gu W, Hao Y, Zhang G, Wang SF, Miao TT, Zhang KP. Synthesis, in vitro antimicrobial and cytotoxic activities of new carbazole derivatives of ursolic acid. Bioorg Med Chem Lett 2015; 25:554-7. [PMID: 25537271 DOI: 10.1016/j.bmcl.2014.12.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/07/2014] [Accepted: 12/09/2014] [Indexed: 12/12/2022]
Abstract
A series of new carbazole derivatives of ursolic acid were designed and synthesized in an attempt to develop potent antimicrobial or antitumor agents. Their structures were confirmed by using IR, HRMS and (1)H NMR analysis. All the synthesized compounds were evaluated for their antimicrobial activity against four bacterial and three fungal strains using serial dilution method. Compounds 3a, 3b, 4a, 4b and 5a-f exhibited significant antibacterial activity against at least one tested bacteria with MIC values of 3.9-15.6μg/ml. In addition, the in vitro cytotoxicity of these compounds were also assayed against two human tumor cell lines (SMMC-7721 and HepG2) using MTT colorimetric method. From the results, compounds 5a-e and 5h displayed pronounced cytotoxic activity with IC50 values below 10μM. Specially, compound 5e was found to be the most potent compound with IC50 values of 1.08±0.22 and 1.26±0.17μM against SMMC-7721 and HepG2 cells, respectively, comparable to those of doxorubicin. In addition, compound 5e showed reduced cytotoxicity against noncancerous LO2 cells with IC50 value of 5.75±0.48μM.
Collapse
Affiliation(s)
- Wen Gu
- Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China.
| | - Yun Hao
- Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Guang Zhang
- Jiangsu Key Lab of Molecular Medicine, State Key Lab of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, PR China
| | - Shi-Fa Wang
- Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Ting-Ting Miao
- Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Kang-Ping Zhang
- Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| |
Collapse
|
65
|
Dong H, Yang X, Xie J, Xiang L, Li Y, Ou M, Chi T, Liu Z, Yu S, Gao Y, Chen J, Shao J, Jia L. UP12, a novel ursolic acid derivative with potential for targeting multiple signaling pathways in hepatocellular carcinoma. Biochem Pharmacol 2015; 93:151-62. [PMID: 25522955 DOI: 10.1016/j.bcp.2014.11.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 01/10/2023]
Abstract
Targeting cancer cell glucose metabolism is a promising strategy for cancer therapy. In past approaches to cancer drug discovery, ursolic acid (UA) has been chemically modified to improve its antitumor activities and bioavailability. Here, a novel ursolic acid (UA) derivative UP12 was developed via computer-aided drug design to explore potent anti-cancer agents and to examine possible mechanisms. The structural docking analyses suggested that UP12 could bind to the active sites of glucokinase (GK), glucose transporter 1 (GLUT1) and ATPase, which are the main enzymes involved in cancer glucose metabolism. We further investigated the synergistic effect between UP12 and glycolysis inhibitor 2-deoxy-d-glucose (2-DG) in inhibiting glucose metabolism of cancer cells. The pharmacological results showed that the combination enhanced depletion of intracellular ATP and decrease in lactate production, and pushed more cancer cells arrested in the S and G2/M cycle phases. The combination selectively down-regulated the expression of Bcl-2 and HKII proteins, up-regulated the expression of Bax and p53, and collectively resulted in enhanced apoptosis related to caspase-3, -8, and -9 activities, in addition to inhibition on the cell mitochondrial membrane potential. The animal studies further demonstrated that the combination exhibited significant antitumor activity without obvious toxicity. In summary, UP12 can interfere cancer cell metabolism pathway and further enhance the therapeutic effects of 2-DG likely through synergistic suppression of cancer cell glucose metabolism, making UP12 a likely new candidate for anti-cancer drug development.
Collapse
Affiliation(s)
- Haiyan Dong
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Xiang Yang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Jingjing Xie
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Liping Xiang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Yuanfang Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Minrui Ou
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Ting Chi
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Zhenhua Liu
- Department of Medical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Suhong Yu
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China
| | - Jianzhong Chen
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350001, China
| | - Jingwei Shao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China; Biopharmaceutical Photocatalysis, State Key Laboratory of Photocatalysis on Energy and Environment, Fuzhou University, Fuzhou 350002, China.
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350002, China; Biopharmaceutical Photocatalysis, State Key Laboratory of Photocatalysis on Energy and Environment, Fuzhou University, Fuzhou 350002, China.
| |
Collapse
|
66
|
Synthesis and cytotoxic evaluation of novel amide–triazole-linked triterpenoid–AZT conjugates. Tetrahedron Lett 2015. [DOI: 10.1016/j.tetlet.2014.11.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
67
|
Dangroo NA, Dar AA, Dar BA. An efficient protocol for domino one pot synthesis of 1,2,3-triazoles from natural organic acids and phenols. Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.09.123] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
68
|
Khaybullin RN, Zhang M, Fu J, Liang X, Li T, Katritzky AR, Okunieff P, Qi X. Design and synthesis of isosteviol triazole conjugates for cancer therapy. Molecules 2014; 19:18676-89. [PMID: 25405286 PMCID: PMC5753759 DOI: 10.3390/molecules191118676] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 01/26/2023] Open
Abstract
One of the keys for successfully developing drugs against the broad spectrum of cancer cell types is structural diversity. In the current study, we focused on a family of isosteviol derivatives as potential novel antitumor agents. Isosteviol is a tetracyclic diterpenoid obtained by acid hydrolysis of steviol glycoside extracts isolated from abundant Stevia rebaudiana plants. In this work, we have designed and synthesized a panel of isosteviol triazole conjugates using "click" chemistry methodology. Evaluation of these compounds against a series of cancer cell lines derived from primary and metastatic tumors demonstrated that these conjugates exhibit cytotoxic activities with IC50 in the low μM range. In addition, their anti-proliferative activities are cancer cell type specific. Taken together, our studies underscore the importance of structural diversity in achieving cancer cell type specific drug development.
Collapse
Affiliation(s)
- Ravil N Khaybullin
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Mei Zhang
- Department of Radiation Oncology, College of Medicine, University of Florida Health Cancer Center, Gainesville, FL 32610, USA
| | - Junjie Fu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Xiao Liang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Tammy Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Alan R Katritzky
- Center for Heterocyclic Compounds, Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Paul Okunieff
- Department of Radiation Oncology, College of Medicine, University of Florida Health Cancer Center, Gainesville, FL 32610, USA
| | - Xin Qi
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
69
|
Dang Thi TA, Kim Tuyet NT, Pham The C, Thanh Nguyen H, Ba Thi C, Doan Duy T, D'hooghe M, Van Nguyen T. Synthesis and cytotoxic evaluation of novel ester-triazole-linked triterpenoid-AZT conjugates. Bioorg Med Chem Lett 2014; 24:5190-4. [PMID: 25442310 DOI: 10.1016/j.bmcl.2014.09.079] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 12/26/2022]
Abstract
Betulinic acid and analogous naturally occurring triterpenoid acids were transformed into the corresponding propargyl esters and subsequently deployed as substrates for a click chemistry-mediated coupling with azidothymidine (AZT) en route to novel 1,2,3-triazole-tethered triterpenoid-AZT conjugates. Twelve new hybrids were thus prepared and assessed in terms of their cytotoxic activity, revealing an interesting anticancer activity of five triterpenoid-AZT hybrids on KB and Hep-G2 tumor cell lines.
Collapse
Affiliation(s)
- Tuyet Anh Dang Thi
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Nguyen Thi Kim Tuyet
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Chinh Pham The
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Ha Thanh Nguyen
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Cham Ba Thi
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Tien Doan Duy
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
| | - Tuyen Van Nguyen
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
70
|
Li JF, Huang RZ, Yao GY, Ye MY, Wang HS, Pan YM, Xiao JT. Synthesis and biological evaluation of novel aniline-derived asiatic acid derivatives as potential anticancer agents. Eur J Med Chem 2014; 86:175-88. [DOI: 10.1016/j.ejmech.2014.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 07/27/2014] [Accepted: 08/01/2014] [Indexed: 10/24/2022]
|
71
|
Anikina LV, Shemyakina DA, Pavlogradskaya LV, Nedugov AN, Glushkov VA. Synthesis of ferrocene conjugates with di- and triterpenes by click chemistry method. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2014. [DOI: 10.1134/s1070428014080181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|