1
|
Levchenko K, Pokhodylo N. Multigram Synthesis of Bicyclic α- and β-Prolines via Intramolecular C(sp 3)-H γ-Lactamization as a Key Step. J Org Chem 2025. [PMID: 40402669 DOI: 10.1021/acs.joc.5c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
A multigram synthesis of novel bicyclic α- and β-prolines was achieved via a C(sp3)-H amidation reaction as a key step. This step prepared seven bicyclic γ-lactams in high yield and stereoselective control. The scalable and reproducible protocol allowed the conversion of γ-lactams into α- and β-prolines in amounts up to 45 g under mild conditions, providing access to various isomeric bicyclic proline derivatives for potential use in drug design as medicinal building blocks.
Collapse
Affiliation(s)
- Kostiantyn Levchenko
- Enamine Ltd., 78 Winston Churchill Str., Kyiv 02094, Ukraine
- Department of Organic Chemistry, Ivan Franko National University of Lviv, Kyryla i Mefodiya 6, Lviv 79005, Ukraine
| | - Nazariy Pokhodylo
- Department of Organic Chemistry, Ivan Franko National University of Lviv, Kyryla i Mefodiya 6, Lviv 79005, Ukraine
| |
Collapse
|
2
|
Derkach NO, Levchenko KV, Iermolenko IA, Ostapchuk EN, Lega DA, Makhankova VG, Rozhenko AB, Volochnyuk DM, Ryabukhin SV. Multigram Synthesis of 3,3-Spiro-α-prolines. J Org Chem 2024; 89:18159-18178. [PMID: 39656886 DOI: 10.1021/acs.joc.4c02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
A series of novel spirocyclic α-proline building blocks with a spiro conjunction in position 3 of the pyrrolidine ring was prepared to employ two convenient and practical synthetic approaches. Both alternative routes utilize simple and readily available starting materials─cyclic ketones and esters─and comprise 6 and 7 steps, respectively. The methodologies feature distinct advantages, using routine organic chemistry transformations, and are suitable for producing multigram amounts of the target prolines. The approach also became valuable for spirocyclic pyroglutamic acids.
Collapse
Affiliation(s)
| | - Kostiantyn V Levchenko
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Department of Organic Chemistry, Ivan Franko National University of Lviv, 6 Kyryla i Mefodia Street, 79005 Lviv, Ukraine
| | - Ievgenii A Iermolenko
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Institute of Organic Chemistry of the National Academy of Sciences of Ukraine, 5 Akademik Kukhar Street, 02094 Kyiv, Ukraine
| | - Eugeniy N Ostapchuk
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
| | - Dmitry A Lega
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
| | - Valeriya G Makhankova
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
| | - Alexander B Rozhenko
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
- Institute of Organic Chemistry of the National Academy of Sciences of Ukraine, 5 Akademik Kukhar Street, 02094 Kyiv, Ukraine
| | - Dmytro M Volochnyuk
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
- Institute of Organic Chemistry of the National Academy of Sciences of Ukraine, 5 Akademik Kukhar Street, 02094 Kyiv, Ukraine
| | - Serhiy V Ryabukhin
- Enamine Ltd., 78 Winston Churchill Street, 02094 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
- Institute of Organic Chemistry of the National Academy of Sciences of Ukraine, 5 Akademik Kukhar Street, 02094 Kyiv, Ukraine
| |
Collapse
|
3
|
In silico study of novel niclosamide derivatives, SARS-CoV-2 nonstructural proteins catalytic residue-targeting small molecules drug candidates. ARAB J CHEM 2023; 16:104654. [PMID: 36777994 PMCID: PMC9904858 DOI: 10.1016/j.arabjc.2023.104654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-mediated coronavirus disease 2019 (COVID-19) infection remains a global pandemic and health emergency with overwhelming social and economic impacts throughout the world. Therapeutics for COVID-19 are limited to only remdesivir; therefore, there is a need for combined, multidisciplinary efforts to develop new therapeutic molecules and explore the effectiveness of existing drugs against SARS-CoV-2. In the present study, we reported eight (SCOV-L-02, SCOV-L-09, SCOV-L-10, SCOV-L-11, SCOV-L-15, SCOV-L-18, SCOV-L-22, and SCOV-L-23) novel structurally related small-molecule derivatives of niclosamide (SCOV-L series) for their targeting potential against angiotensin-converting enzyme-2 (ACE2), type II transmembrane serine protease (TMPRSS2), and SARS-COV-2 nonstructural proteins (NSPs) including NSP5 (3CLpro), NSP3 (PLpro), and RdRp. Our correlation analysis suggested that ACE2 and TMPRSS2 modulate host immune response via regulation of immune-infiltrating cells at the site of tissue/organs entries. In addition, we identified some TMPRSS2 and ACE2 microRNAs target regulatory networks in SARS-CoV-2 infection and thus open up a new window for microRNAs-based therapy for the treatment of SARS-CoV-2 infection. Our in vitro study revealed that with the exception of SCOV-L-11 and SCOV-L-23 which were non-active, the SCOV-L series exhibited strict antiproliferative activities and non-cytotoxic effects against ACE2- and TMPRSS2-expressing cells. Our molecular docking for the analysis of receptor-ligand interactions revealed that SCOV-L series demonstrated high ligand binding efficacies (at higher levels than clinical drugs) against the ACE2, TMPRSS2, and SARS-COV-2 NSPs. SCOV-L-18, SCOV-L-15, and SCOV-L-09 were particularly found to exhibit strong binding affinities with three key SARS-CoV-2's proteins: 3CLpro, PLpro, and RdRp. These compounds bind to the several catalytic residues of the proteins, and satisfied the criteria of drug-like candidates, having good adsorption, distribution, metabolism, excretion, and toxicity (ADMET) pharmacokinetic profile. Altogether, the present study suggests the therapeutic potential of SCOV-L series for preventing and managing SARs-COV-2 infection and are currently under detailed investigation in our lab.
Collapse
|
4
|
Avan İ, Akbulut VM. Synthesis and Antioxidant Evaluation of O-Methylated Emodacidamides: Starting from Parietin, a Secondary Metabolite of Lichen Xanthoria parietina. Chem Biodivers 2023; 20:e202200771. [PMID: 36512748 DOI: 10.1002/cbdv.202200771] [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: 08/11/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Polyhydroxy-anthraquinones bearing amino acids are found rather seldom in nature. Emodacidamides, isolated from a marine-derived fungus, Penicillium sp. SCSIO sof101 by Luo et al. (2017) are the first natural example of amino acid conjugated anthraquinone. In this study, O-methylated emodacidamides and emodinic acid-anilides were synthesized starting from parietin, extracted from the lichen Xanthoria parietina (L.) Th. Fr. The structural elucidations of prepared compounds were confirmed by 1D and 2D NMR analyses including HSQC and HMBC techniques. In addition, all newly synthesized compounds were evaluated for the antioxidant activities with free radical 1,1-diphenyl-2-picrylhydrazyl (DPPH) scavenging. The synthesized compounds showed low to moderate antioxidant and DPPH scavenging activities. The antioxidant activities were supported within quantum chemical calculations using the DFT-B3LYP/6-311++G(d,p) level of theory. It is observed that the antioxidant activity of emodacidamides mostly depends on the phenolic groups on anthraquinone ring. The phenolic groups on other substituents help to improve antioxidant activity and also the position of hydroxy group is a decisive factor for antioxidant ability.
Collapse
Affiliation(s)
- İlker Avan
- Eskisehir Technical University, Faculty of Science, Department of Chemistry, Eskişehir, Türkiye
| | - Volkan Mustafa Akbulut
- Eskisehir Technical University, Faculty of Science, Department of Chemistry, Eskişehir, Türkiye
| |
Collapse
|
5
|
A Preclinical Investigation of GBM-N019 as a Potential Inhibitor of Glioblastoma via Exosomal mTOR/CDK6/STAT3 Signaling. Cells 2021; 10:cells10092391. [PMID: 34572040 PMCID: PMC8471927 DOI: 10.3390/cells10092391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive brain malignancies with high incidences of developing treatment resistance, resulting in poor prognoses. Glioma stem cell (GSC)-derived exosomes are important players that contribute to GBM tumorigenesis and aggressive properties. Herein, we investigated the inhibitory roles of GBM-N019, a novel small molecule on the transfer of aggressive and invasive properties through the delivery of oncogene-loaded exosomes from GSCs to naïve and non-GSCs. Our results indicated that GBM-N019 significantly downregulated the expressions of the mammalian target of rapamycin (mTOR), signal transducer and activator of transcription 3 (STAT3), and cyclin-dependent kinase 6 (CDK6) signaling networks with concomitant inhibitory activities against viability, clonogenicity, and migratory abilities of U251 and U87MG cells. Treatments with GBM-N019 halted the exosomal transfer of protein kinase B (Akt), mTOR, p-mTOR, and Ras-related protein RAB27A to the naïve U251 and U87MG cells, and rescued the cells from invasive and stemness properties that were associated with activation of these oncogenes. GBM-N019 also synergized with and enhanced the anti-GBM activities of palbociclib in vitro and in vivo. In conclusion, our results suggested that GBM-N019 possesses good translational relevance as a potential anti-glioblastoma drug candidate worthy of consideration for clinical trials against recurrent glioblastomas.
Collapse
|
6
|
Xu Q, Deng H, Li X, Quan ZS. Application of Amino Acids in the Structural Modification of Natural Products: A Review. Front Chem 2021; 9:650569. [PMID: 33996749 PMCID: PMC8118163 DOI: 10.3389/fchem.2021.650569] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/02/2021] [Indexed: 01/11/2023] Open
Abstract
Natural products and their derivatives are important sources for drug discovery; however, they usually have poor solubility and low activity and require structural modification. Amino acids are highly soluble in water and have a wide range of activities. The introduction of amino acids into natural products is expected to improve the performance of these products and minimize their adverse effects. Therefore, this review summarizes the application of amino acids in the structural modification of natural products and provides a theoretical basis for the structural modification of natural products in the future. The articles were divided into six types based on the backbone structures of the natural products, and the related applications of amino acids in the structural modification of natural products were discussed in detail.
Collapse
Affiliation(s)
- Qian Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Jilin, China
| | - Hao Deng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Jilin, China
| | - Xiaoting Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Jilin, China
- Department of Pharmaceutical Analysis, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Jilin, China
| |
Collapse
|
7
|
Ali AAA, Lee YR, Wu AT, Yadav VK, Yu DS, Huang HS. Structure-based strategies for synthesis, lead optimization and biological evaluation of N-substituted anthra[1,2-c][1,2,5]thiadiazole-6,11-dione derivatives as potential multi-target anticancer agents. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.10.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
8
|
Akhtar MJ, Yar MS, Sharma VK, Khan AA, Ali Z, Haider MDR, Pathak A. Recent Progress of Benzimidazole Hybrids for Anticancer Potential. Curr Med Chem 2021; 27:5970-6014. [PMID: 31393240 DOI: 10.2174/0929867326666190808122929] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/16/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022]
Abstract
This review presents the detailed account of factors leading to cancer and design strategy for the synthesis of benzimidazole derivatives as anticancer agents. The recent survey for cancer treatment in Cancer facts and figures 2017 American Chemical Society has shown progressive development in fighting cancer. Researchers all over the world in both developed and developing countries are in a continuous effort to tackle this serious concern. Benzimidazole and its derivatives showed a broad range of biological activities due to their resemblance with naturally occurring nitrogenous base i.e. purine. The review discussed benzimidazole derivatives showing anticancer properties through a different mechanism viz. intercalation, alkylating agents, topoisomerases, DHFR enzymes, and tubulin inhibitors. Benzimidazole derivatives act through a different mechanism and the substituents reported from the earlier and recent research articles are prerequisites for the synthesis of targeted based benzimidazole derivatives as anticancer agents. The review focuses on an easy comparison of the substituent essential for potency and selectivity through SAR presented in figures. This will further provide a better outlook or fulfills the challenges faced in the development of novel benzimidazole derivatives as anticancer.
Collapse
Affiliation(s)
- Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Hamdard University,
Hamdard Nagar, New Delhi-110062, India,Department of Pharmaceutical Chemistry, Indo Soviet Friendship College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road MOGA-142001, Punjab, India
| | - Mohammad Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Hamdard University,
Hamdard Nagar, New Delhi-110062, India
| | - Vinod Kumar Sharma
- School of Pharmacy, Bharat Institute of Technology, NH58, Partapur Bypass Meerut-250103, India
| | - Ahsan Ahmed Khan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Hamdard University,
Hamdard Nagar, New Delhi-110062, India
| | - Zulphikar Ali
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Hamdard University,
Hamdard Nagar, New Delhi-110062, India
| | - M D Rafi Haider
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Hamdard University,
Hamdard Nagar, New Delhi-110062, India
| | - Ankita Pathak
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Hamdard University,
Hamdard Nagar, New Delhi-110062, India
| |
Collapse
|
9
|
Chen X, Tang WJ, Shi JB, Liu MM, Liu XH. Therapeutic strategies for targeting telomerase in cancer. Med Res Rev 2019; 40:532-585. [PMID: 31361345 DOI: 10.1002/med.21626] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
Telomere and telomerase play important roles in abnormal cell proliferation, metastasis, stem cell maintenance, and immortalization in various cancers. Therefore, designing of drugs targeting telomerase and telomere is of great significance. Over the past two decades, considerable knowledge regarding telomere and telomerase has been accumulated, which provides theoretical support for the design of therapeutic strategies such as telomere elongation. Therefore, the development of telomere-based therapies such as nucleoside analogs, non-nucleoside small molecules, antisense technology, ribozymes, and dominant negative human telomerase reverse transcriptase are being prioritized for eradicating a majority of tumors. While the benefits of telomere-based therapies are obvious, there is a need to address the limitations of various therapeutic strategies to improve the possibility of clinical applications. In this study, current knowledge of telomere and telomerase is discussed, and therapeutic strategies based on recent research are reviewed.
Collapse
Affiliation(s)
- Xing Chen
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Wen-Jian Tang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Jing Bo Shi
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Ming Ming Liu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Xin-Hua Liu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
10
|
Synthesis and electrochemical, spectral, and biological evaluation of novel 9,10-anthraquinone derivatives containing piperidine unit as potent antiproliferative agents. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2018.07.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
11
|
Identification of a new class of WNT1 inhibitor: Cancer cells migration, G-quadruplex stabilization and target validation. Oncotarget 2018; 7:67986-68001. [PMID: 27626678 PMCID: PMC5356533 DOI: 10.18632/oncotarget.6622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 11/16/2015] [Indexed: 12/31/2022] Open
Abstract
Developing the Wnt pathway inhibitors has been considered as a therapeutic approach for cancers and other Wnt-related diseases. Previously we found that the G-rich sequence of WNT1 promoter is capable of forming G-quadruplex structure and stabilizing agents for Wnt1-mediated signaling pathway. Using a established cell-based drug screen system that enabled the evaluation of WNT1 expression activity in a G-quadruplex structure dependent manner, we evaluated a series of 6-substituted 9-chloro-11H-indeno[1,2-c]quinolin-11-one derivatives that potentially inhibit the Wnt1-mediated signaling pathway. The most potent compound SJ26 showed repression of WNT1 activity in a G-quadruplex structure-dependent manner. Moreover, compound SJ26 inhibited the WNT1-mediated downstream signaling pathway and suppressed migration activity of cancer cells. Thus, we have identified a tetracyclic azafluorenone, SJ26, that is capable of binding to G-quadruplex DNA structure, repressing WNT1 expression, and inhibiting cell migration.
Collapse
|
12
|
Kosbar TR, Abou-Zeid L, Sofan MA. Synthesis, Biological Evaluation, and Molecular Docking Studies of Novel Pyrazolo[3,4-d
]pyrimidines as Potential Telomerase Inhibitors. J Heterocycl Chem 2018. [DOI: 10.1002/jhet.3094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Tamer R. Kosbar
- Department of Chemistry, Faculty of Science; Damietta University; 34517 New Damietta Egypt
| | - Laila Abou-Zeid
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy; Mansoura University; 35516 Mansoura Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy; Delta University; Mansoura Egypt
| | - Mamdouh A. Sofan
- Department of Chemistry, Faculty of Science; Damietta University; 34517 New Damietta Egypt
| |
Collapse
|
13
|
Lee CC, Chen CL, Liu FL, Chiou CY, Chen TC, Wu CC, Sun WH, Chang DM, Huang HS. Development of 1-Amino-4-(phenylamino)anthraquinone-2-sulfonate Sodium Derivatives as a New Class of Inhibitors of RANKL-Induced Osteoclastogenesis. Arch Pharm (Weinheim) 2016; 349:342-55. [DOI: 10.1002/ardp.201500475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/20/2016] [Accepted: 03/23/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Chia-Chung Lee
- Graduate Institute of Cancer Biology and Drug Discovery; College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- School of Pharmacy; National Defense Medical Center; Taipei Taiwan
- Rheumatology/Immunology/Allergy; Taipei Veterans General Hospital; Taipei Taiwan
| | - Chun-Liang Chen
- Graduate Institute of Cancer Biology and Drug Discovery; College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- School of Pharmacy; National Defense Medical Center; Taipei Taiwan
- Rheumatology/Immunology/Allergy; Taipei Veterans General Hospital; Taipei Taiwan
| | - Fei-Lan Liu
- Rheumatology/Immunology/Allergy; Taipei Veterans General Hospital; Taipei Taiwan
| | - Chung-Yu Chiou
- Graduate Institute of Cancer Biology and Drug Discovery; College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- School of Pharmacy; National Defense Medical Center; Taipei Taiwan
| | - Tsung-Chih Chen
- Graduate Institute of Cancer Biology and Drug Discovery; College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- School of Pharmacy; National Defense Medical Center; Taipei Taiwan
| | - Cheng-Chi Wu
- Rheumatology/Immunology/Allergy; Taipei Veterans General Hospital; Taipei Taiwan
- Graduate Institute of Life Sciences; National Defense Medical Center; Taipei Taiwan
| | - Wei-Hsin Sun
- Department of Life Sciences; National Central University; Jhongli City Taiwan
| | - Deh-Ming Chang
- Rheumatology/Immunology/Allergy; Taipei Veterans General Hospital; Taipei Taiwan
- Graduate Institute of Life Sciences; National Defense Medical Center; Taipei Taiwan
| | - Hsu-Shan Huang
- Graduate Institute of Cancer Biology and Drug Discovery; College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- School of Pharmacy; National Defense Medical Center; Taipei Taiwan
- Graduate Institute of Life Sciences; National Defense Medical Center; Taipei Taiwan
| |
Collapse
|
14
|
Wang JQ, Zhao ZZ, Bo HB, Chen QZ. Synthesis, characterization, and antitumor properties of ruthenium(II) anthraquinone complexes. J COORD CHEM 2016. [DOI: 10.1080/00958972.2015.1120291] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Jin-Quan Wang
- School of Life Science and Biopharmaceutical, Guangdong Pharmaceutical University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Biotechnology Candidate Drug Research, Guangzhou, PR China
| | - Zi-Zhuo Zhao
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Hua-Ben Bo
- School of Life Science and Biopharmaceutical, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Qi-Zhu Chen
- School of Life Science and Biopharmaceutical, Guangdong Pharmaceutical University, Guangzhou, PR China
| |
Collapse
|
15
|
Lee YR, Chen TC, Lee CC, Chen CL, Ahmed Ali AA, Tikhomirov A, Guh JH, Yu DS, Huang HS. Ring fusion strategy for synthesis and lead optimization of sulfur-substituted anthra[1,2-c][1,2,5]thiadiazole-6,11-dione derivatives as promising scaffold of antitumor agents. Eur J Med Chem 2015; 102:661-76. [DOI: 10.1016/j.ejmech.2015.07.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/07/2015] [Accepted: 07/31/2015] [Indexed: 12/25/2022]
|
16
|
Langie SA, Koppen G, Desaulniers D, Al-Mulla F, Al-Temaimi R, Amedei A, Azqueta A, Bisson WH, Brown D, Brunborg G, Charles AK, Chen T, Colacci A, Darroudi F, Forte S, Gonzalez L, Hamid RA, Knudsen LE, Leyns L, Lopez de Cerain Salsamendi A, Memeo L, Mondello C, Mothersill C, Olsen AK, Pavanello S, Raju J, Rojas E, Roy R, Ryan E, Ostrosky-Wegman P, Salem HK, Scovassi I, Singh N, Vaccari M, Van Schooten FJ, Valverde M, Woodrick J, Zhang L, van Larebeke N, Kirsch-Volders M, Collins AR. Causes of genome instability: the effect of low dose chemical exposures in modern society. Carcinogenesis 2015; 36 Suppl 1:S61-S88. [PMID: 26106144 PMCID: PMC4565613 DOI: 10.1093/carcin/bgv031] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 12/08/2014] [Accepted: 12/11/2014] [Indexed: 12/17/2022] Open
Abstract
Genome instability is a prerequisite for the development of cancer. It occurs when genome maintenance systems fail to safeguard the genome's integrity, whether as a consequence of inherited defects or induced via exposure to environmental agents (chemicals, biological agents and radiation). Thus, genome instability can be defined as an enhanced tendency for the genome to acquire mutations; ranging from changes to the nucleotide sequence to chromosomal gain, rearrangements or loss. This review raises the hypothesis that in addition to known human carcinogens, exposure to low dose of other chemicals present in our modern society could contribute to carcinogenesis by indirectly affecting genome stability. The selected chemicals with their mechanisms of action proposed to indirectly contribute to genome instability are: heavy metals (DNA repair, epigenetic modification, DNA damage signaling, telomere length), acrylamide (DNA repair, chromosome segregation), bisphenol A (epigenetic modification, DNA damage signaling, mitochondrial function, chromosome segregation), benomyl (chromosome segregation), quinones (epigenetic modification) and nano-sized particles (epigenetic pathways, mitochondrial function, chromosome segregation, telomere length). The purpose of this review is to describe the crucial aspects of genome instability, to outline the ways in which environmental chemicals can affect this cancer hallmark and to identify candidate chemicals for further study. The overall aim is to make scientists aware of the increasing need to unravel the underlying mechanisms via which chemicals at low doses can induce genome instability and thus promote carcinogenesis.
Collapse
Affiliation(s)
- Sabine A.S. Langie
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium
- Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
- Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
- Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia
- University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
- Medical Physics & Applied Radiation Sciences, McMaster University, Hamilton, Ontario L8S4L8, Canada
- Department of Cardiac, Thoracic and Vascular Sciences, Unit of Occupational Medicine, University of Padova, Padova 35128, Italy
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow 226003, Uttar Pradesh, India
- Department of Toxicology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, 6200MD, PO Box 61, Maastricht, The Netherlands
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA
- Laboratory for Analytical and Environmental Chemistry, Vrije Universiteit Brussel, Brussels 1050, Belgium
- Study Centre for Carcinogenesis and Primary Prevention of Cancer, Ghent University, Ghent 9000, Belgium
- Department of Nutrition, University of Oslo, Oslo 0316, Norway
| | - Gudrun Koppen
- *To whom correspondence should be addressed. Tel: +32 14335165; Fax: +32 14580523
| | - Daniel Desaulniers
- Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Dustin Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
| | - Amelia K. Charles
- Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Firouz Darroudi
- Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Laetitia Gonzalez
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Roslida A. Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia
| | - Lisbeth E. Knudsen
- University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark
| | - Luc Leyns
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | | | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Carmel Mothersill
- Medical Physics & Applied Radiation Sciences, McMaster University, Hamilton, Ontario L8S4L8, Canada
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
| | - Sofia Pavanello
- Department of Cardiac, Thoracic and Vascular Sciences, Unit of Occupational Medicine, University of Padova, Padova 35128, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Emilio Rojas
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Elizabeth Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Patricia Ostrosky-Wegman
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow 226003, Uttar Pradesh, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Frederik J. Van Schooten
- Department of Toxicology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, 6200MD, PO Box 61, Maastricht, The Netherlands
| | - Mahara Valverde
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA
| | - Nik van Larebeke
- Laboratory for Analytical and Environmental Chemistry, Vrije Universiteit Brussel, Brussels 1050, Belgium
- Study Centre for Carcinogenesis and Primary Prevention of Cancer, Ghent University, Ghent 9000, Belgium
| | | | | |
Collapse
|
17
|
Chen CL, Liu FL, Lee CC, Chen TC, Chang WW, Guh JH, Ahmed Ali AA, Chang DM, Huang HS. Ring fusion strategy for the synthesis of anthra[2,3-d]oxazole-2-thione-5,10-dione homologues as DNA topoisomerase inhibitors and as antitumor agents. Eur J Med Chem 2014; 87:30-8. [DOI: 10.1016/j.ejmech.2014.09.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 08/22/2014] [Accepted: 09/04/2014] [Indexed: 10/24/2022]
|
18
|
Thiazolidine-2,4-diones as multi-targeted scaffold in medicinal chemistry: Potential anticancer agents. Eur J Med Chem 2014; 87:814-33. [DOI: 10.1016/j.ejmech.2014.10.025] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/22/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022]
|
19
|
Chen TC, Wu CL, Lee CC, Chen CL, Yu DS, Huang HS. Structure-based hybridization, synthesis and biological evaluation of novel tetracyclic heterocyclic azathioxanthone analogues as potential antitumor agents. Eur J Med Chem 2014; 103:615-27. [PMID: 25799376 DOI: 10.1016/j.ejmech.2014.09.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/08/2014] [Accepted: 09/06/2014] [Indexed: 01/27/2023]
Abstract
A series of tetracyclic heterocyclic azathioxanthones were synthesized and evaluated for cell proliferations, topoisomerase inhibitions, and NCI-60 cell panel assay, respectively. Compounds 5, 7, 8, 16, and 19 were selected for topoisomerase assay after MTT assay. 7 not only showed cytotoxic effect (IC50 = 2.84 ± 0.64 μM) in PC-3 cells but also revealed topoisomerases inhibition with IC50 (10-25 μM) and increased apoptotic cleavage of PARP and caspase 3 activity. The overall of novel azathioxanthones with different cytostatic and cytotoxic activities should be further developed as new potential candidates for anticancer drugs.
Collapse
Affiliation(s)
- Tsung-Chih Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Chia-Lun Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Chia-Chung Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Chun-Liang Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Dah-Shyong Yu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan; Uro-Oncology Laboratory, Division of Urology, Department of Surgery, Tri-Service General Hospital, Taipei 114, Taiwan.
| | - Hsu-Shan Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan.
| |
Collapse
|
20
|
Chen TC, Yu DS, Huang KF, Fu YC, Lee CC, Chen CL, Huang FC, Hsieh HH, Lin JJ, Huang HS. Structure-based design, synthesis and biological evaluation of novel anthra[1,2-d]imidazole-6,11-dione homologues as potential antitumor agents. Eur J Med Chem 2013; 69:278-93. [DOI: 10.1016/j.ejmech.2013.06.058] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 06/16/2013] [Accepted: 06/29/2013] [Indexed: 12/13/2022]
|
21
|
Lee CC, Chang DM, Huang KF, Chen CL, Chen TC, Lo Y, Guh JH, Huang HS. Design, synthesis and antiproliferative evaluation of fluorenone analogs with DNA topoisomerase I inhibitory properties. Bioorg Med Chem 2013; 21:7125-33. [PMID: 24094433 DOI: 10.1016/j.bmc.2013.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/03/2013] [Accepted: 09/03/2013] [Indexed: 11/29/2022]
Abstract
A series of 2,7-diamidofluorenones were designed, synthesized, and screened by SRB assay. Some synthesized compounds exhibited antitumor activities in submicromolar range. Ten compounds (3a, 3b, 3c, 3g, 3j, 3l, 4a, 4h, 4i, and 4j) were also selected by NCI screening system and 3c (GI50=1.66 μM) appeared to be the most active agent of this series. Furthermore, 3c attenuated topoisomerase I-mediated DNA relaxation at low micromolar concentrations. These results indicated that fluorenones have potential to be further developed into anticancer drugs.
Collapse
Affiliation(s)
- Chia-Chung Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Synthesis, cytotoxic activity and DNA-interaction studies of novel anthraquinone–thiosemicarbazones with tautomerizable methylene group. Eur J Med Chem 2013; 64:228-38. [DOI: 10.1016/j.ejmech.2013.03.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/27/2013] [Accepted: 03/31/2013] [Indexed: 11/19/2022]
|
23
|
Urine telomerase for diagnosis and surveillance of bladder cancer. Adv Urol 2012; 2012:693631. [PMID: 22888342 PMCID: PMC3410307 DOI: 10.1155/2012/693631] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/31/2012] [Accepted: 06/04/2012] [Indexed: 12/18/2022] Open
Abstract
Bladder cancer has increased incidence during last decades. For those patients with nonmuscle involved tumors, noninvasive diagnosis test and surveillance methods must be designed to avoid current cystoscopies that nowadays are done regularly in a lot of patients. Novel urine biomarkers have been developed during last years. Telomerase is important in cancer biology, improving the division capacity of cancer cells. Even urinary telomerase could be a potentially useful urinary tumor marker; its use for diagnosis of asymptomatic and symptomatic patients or its impact during surveillance is still unknown. Moreover, there will need to be uniformity and standardization in the assays before it can become useful in clinical practice. It does not seem to exist a real difference between the most classical assays for the detection of urine telomerase (TRAP and hTERT). However, the new detection methods with modified TeloTAGGG telomerase or with gold nanoparticles must also be taken into consideration for the correct development of this diagnosis method. Maybe the target population would be the high-risk groups within screening programs. To date there is no enough evidence to use it alone and to eliminate cystoscopies from the diagnosis and surveillance of these patients. The combination with cytology or FISH is still preferred.
Collapse
|