1
|
Strzyga-Łach P, Kurpios-Piec D, Chrzanowska A, Szczepaniak J, Bielenica A. 1,3-Disubstituted thiourea derivatives: Promising candidates for medicinal applications with enhanced cytotoxic effects on cancer cells. Eur J Pharmacol 2024; 982:176885. [PMID: 39128803 DOI: 10.1016/j.ejphar.2024.176885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
The distinct chemical structure of thiourea derivatives provides them with an advantage in selectively targeting cancer cells. In our previous study, we selected the most potent compounds, 2 and 8, with 3,4-dichloro- and 3-trifluoromethylphenyl substituents, respectively, across colorectal (SW480 and SW620), prostate (PC3), and leukemia (K-562) cancer cell lines, as well as non-tumor HaCaT cells. Our research has demonstrated their anticancer potential by targeting key molecular pathways involved in cancer progression, including caspase 3/7 activation, NF-κB (Nuclear Factor Kappa-light-chain-enhancer of activated B cells) activation decrease, VEGF (Vascular Endothelial Growth Factor) secretion, ROS (Reactive Oxygen Species) production, and metabolite profile alterations. Notably, these processes exhibited no significant alterations in HaCaT cells. The effectiveness of the studied compounds was also tested on spheroids (3D culture). Both derivatives 2 and 8 increased caspase activity, decreased ROS production and NF-κB activation, and suppressed the release of VEGF in cancer cells. Metabolomic analysis revealed intriguing shifts in cancer cell metabolic profiles, particularly in lipids and pyrimidines metabolism. Assessment of cell viability in 3D spheroids showed that SW620 cells exhibited better sensitivity to compound 2 than 8. In summary, structural modifications of the thiourea terminal components, particularly dihalogenophenyl derivative 2 and para-substituted analog 8, demonstrate their potential as anticancer agents while preserving safety for normal cells.
Collapse
Affiliation(s)
- Paulina Strzyga-Łach
- Chair and Department of Biochemistry, Medical University of Warsaw, Ul. Banacha 1, 02-097, Warsaw, Poland.
| | - Dagmara Kurpios-Piec
- Chair and Department of Biochemistry, Medical University of Warsaw, Ul. Banacha 1, 02-097, Warsaw, Poland.
| | - Alicja Chrzanowska
- Chair and Department of Biochemistry, Medical University of Warsaw, Ul. Banacha 1, 02-097, Warsaw, Poland.
| | - Jarosław Szczepaniak
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences ul., Ciszewskiego 8, 02-786, Warsaw, Poland.
| | - Anna Bielenica
- Chair and Department of Biochemistry, Medical University of Warsaw, Ul. Banacha 1, 02-097, Warsaw, Poland.
| |
Collapse
|
2
|
El-Atawy MA, Alsubaie MS, Alazmi ML, Hamed EA, Hanna DH, Ahmed HA, Omar AZ. Synthesis, Characterization, and Anticancer Activity of New N,N'-Diarylthiourea Derivative against Breast Cancer Cells. Molecules 2023; 28:6420. [PMID: 37687250 PMCID: PMC10490226 DOI: 10.3390/molecules28176420] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/21/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The goal of the current study was to prepare two new homologous series of N,N'-diarylurea and N,N'-diarylthiourea derivatives to investigate the therapeutic effects of these derivatives on the methodologies of inhibition directed on human MCF-7 cancer cells. The molecular structures of the prepared derivatives were successfully revealed through elemental analyses, 1H-NMR, 13C-NMR and FT-IR spectroscopy. The cytotoxic results showed that Diarylthiourea (compound 4) was the most effective in suppressing MCF-7 cell growth when compared to all other prepared derivatives, with the most effective IC50 value (338.33 ± 1.52 µM) after an incubation period of 24 h and no cytotoxic effects on normal human lung cells (wi38 cells). Using the annexin V/PI and comet tests, respectively, treated MCF-7 cells with this IC50 value of the Diarylthiourea 4 compound displayed a considerable increase in early and late apoptotic cells, as well as an intense comet nucleus in comparison to control cells. An arrest of the cell cycle in the S phase was observed via flow cytometry in MCF-7 cells treated with the Diarylthiourea 4 compound, suggesting the onset of apoptosis. Additionally, ELISA research showed that caspase-3 was upregulated in MCF-7 cells treated with compound 4 compared to control cells, suggesting that DNA damage induced by compound 4 may initiate an intrinsic apoptotic pathway and activate caspase-3. These results contributed to recognizing that the successfully prepared Diarylthiourea 4 compound inhibited the proliferation of MCF-7 cancer cells by arresting the S cell cycle and caspase-3 activation via an intrinsic apoptotic route. These results, however, need to be verified through in vivo studies utilizing an animal model.
Collapse
Affiliation(s)
- Mohamed A. El-Atawy
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
- Chemistry Department, Faculty of Science, Taibah University, Yanbu 46423, Saudi Arabia
| | - Mai S. Alsubaie
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| | - Mohammed L. Alazmi
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| | - Ezzat A. Hamed
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| | - Demiana H. Hanna
- Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt;
| | - Hoda A. Ahmed
- Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt;
| | - Alaa Z. Omar
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| |
Collapse
|
3
|
Strzyga-Łach P, Chrzanowska A, Kiernozek-Kalińska E, Żyżyńska-Granica B, Podsadni K, Podsadni P, Bielenica A. Proapoptotic effects of halogenated bis-phenylthiourea derivatives in cancer cells. Arch Pharm (Weinheim) 2023; 356:e2300105. [PMID: 37401845 DOI: 10.1002/ardp.202300105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
New halogenated thiourea derivatives were synthesized via the reaction of substituted phenylisothiocyanates with aromatic amines. Their cytotoxic activity was examined in in vitro studies against solid tumors (SW480, SW620, PC3), a hematological malignance (K-562), and normal keratinocytes (HaCaT). Most of the compounds were more effective against SW480 (1a, 3a, 3b, 5j), K-562 (2b, 3a, 4a), or PC3 (5d) cells than cisplatin, with favorable selectivity. Their anticancer mechanisms were studied by Annexin V-fluorescein-5-isothiocyanate apoptosis, caspase-3/caspase-7 assessment, cell cycle analysis, interleukin-6 (IL-6) release inhibition, and reactive oxygen species (ROS) generation assay. Thioureas 1a, 2b, 3a, and 4a were the most potent activators of early apoptosis in K-562 cells, and substances 1a, 3b, 5j triggered late-apoptosis or necrosis in SW480 cells. This proapoptotic effect was proved by the significant increase of caspase-3/caspase-7 activation. Cell cycle analysis revealed that derivatives 1a, 3a, 5j increased the number of SW480 and K-562 cells in the sub-G1 and/or G0/G1 phases, and one evoked cycle arrest at the G2 phase. The most potent thioureas inhibited IL-6 cytokine secretion from PC3 cells and both colon cancer cell lines. Apoptosis-inducing compounds also increased ROS production in all tumor cell cultures, which may enhance their anticancer properties.
Collapse
Affiliation(s)
- Paulina Strzyga-Łach
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Alicja Chrzanowska
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Katarzyna Podsadni
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Podsadni
- Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Anna Bielenica
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
4
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 189] [Impact Index Per Article: 189.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
5
|
Puri S, Sawant S, Juvale K. A comprehensive review on the indazole based derivatives as targeted anticancer agents. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
|
6
|
Yıldız ML, Demir Y, Küfrevioğlu ÖI. Screening of in vitro and in silico effect of Fluorophenylthiourea compounds on glucose 6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase enzymes. J Mol Recognit 2022; 35:e2987. [PMID: 36326002 DOI: 10.1002/jmr.2987] [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: 07/25/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 01/05/2023]
Abstract
Inhibition studies of enzymes in the pentose phosphate pathway (PPP) have recently emerged as a promising technique for pharmacological intervention in several illnesses. Glucose 6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD) are the most important enzymes of the PPP. For this purpose, in the current study, we examined the effect of some fluorophenylthiourea on G6PD and 6PGD enzyme activity. These compounds exhibited moderate inhibitory activity against G6PD and 6PGD with KI values ranging from 21.60 ± 8.42 to 39.70 ± 11.26 μM, and 15.82 ± 1.54 to 29.97 ± 5.72 μM, respectively. 2,6-difluorophenylthiourea displayed the most potent inhibitory effect for G6PD, and 2-fluorophenylthiourea demonstrated the most substantial inhibitory effect for 6PGD. Furthermore, the molecular docking analyses of the fluorophenylthioureas, competitive inhibitors, were performed to understand the binding interactions at the enzymes' binding site.
Collapse
Affiliation(s)
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, Turkey
| | | |
Collapse
|
7
|
Koopaei NN, Shademani M, Yazdi NS, Tahmasvand R, Dehbid M, Koopaei MN, Azizian H, Mousavi Z, Almasirad A, Salimi M. Design and synthesis of novel ureido and thioureido conjugated hydrazone derivatives with potent anticancer activity. BMC Chem 2022; 16:81. [PMID: 36320042 PMCID: PMC9624014 DOI: 10.1186/s13065-022-00873-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Background Compounds possessing urea/thiourea moiety have a wide range of biological properties including anticancer activity. On the other hand, taking advantage of the low toxicity and structural diversity of hydrazone derivatives, they are presently being considered for designing chemical compounds with hydrazone moiety in the field of cancer treatment. With this in mind, a series of novel ureido/thioureido derivatives possessing a hydrazone moiety bearing nitro and chloro substituents (4a–4i) have been designed, synthesized, characterized and evaluated for their in vitro cytotoxic effect on HT-29 human colon carcinoma and HepG2 hepatocarcinoma cell lines. Results Two compounds (4c and 4e) having the chloro phenylurea group hybridized with phenyl hydrazone bearing nitro or chloro moieties demonstrated potent anticancer effect with the IC50 values between 2.2 and 4.8 µM at 72 h. The mechanism of action of compound 4c was revealed in hepatocellular carcinoma cells as an inducer of apoptosis in a caspase-independent pathway. Conclusion Taken together, the current work presented compound 4c as a potential lead compound in developing future hepatocellular carcinoma chemotherapy drugs. Methods The compounds were synthesized and then characterized by physical and spectral data (FT-IR, 1H-NMR, 13C-NMR, Mass). The anticancer activity was assessed using MTT assay, flowcytometry, annexin-V, DAPI staining and Western blot analysis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13065-022-00873-3.
Collapse
Affiliation(s)
- Nasrin Nassiri Koopaei
- grid.411463.50000 0001 0706 2472Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, P.O. Box 1941933111, Tehran, Iran
| | - Mehrasa Shademani
- grid.420169.80000 0000 9562 2611Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran ,grid.411463.50000 0001 0706 2472Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nasrin Shirzad Yazdi
- grid.411463.50000 0001 0706 2472Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, P.O. Box 1941933111, Tehran, Iran ,grid.412571.40000 0000 8819 4698Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raheleh Tahmasvand
- grid.420169.80000 0000 9562 2611Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Mina Dehbid
- grid.411872.90000 0001 2087 2250Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mansur Nassiri Koopaei
- grid.411705.60000 0001 0166 0922Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design & Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Azizian
- grid.411746.10000 0004 4911 7066Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Science, Tehran, Iran
| | - Zahra Mousavi
- grid.411463.50000 0001 0706 2472Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Almasirad
- grid.411463.50000 0001 0706 2472Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, P.O. Box 1941933111, Tehran, Iran
| | - Mona Salimi
- grid.420169.80000 0000 9562 2611Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| |
Collapse
|
8
|
Tan C, Yang SJ, Zhao DH, Li J, Yin LQ. Antihypertensive activity of indole and indazole analogues: A review. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
9
|
Tandon N, Luxami V, Kant D, Tandon R, Paul K. Current progress, challenges and future prospects of indazoles as protein kinase inhibitors for the treatment of cancer. RSC Adv 2021; 11:25228-25257. [PMID: 35478899 PMCID: PMC9037120 DOI: 10.1039/d1ra03979b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/29/2021] [Indexed: 01/19/2023] Open
Abstract
The indazole core is an interesting pharmacophore due to its applications in medicinal chemistry. In the past few years, this moiety has been used for the synthesis of kinase inhibitors. Many researchers have demonstrated the use of indazole derivatives as specific kinase inhibitors, including tyrosine kinase and serine/threonine kinases. A number of anticancer drugs with an indazole core are commercially available, e.g. axitinib, linifanib, niraparib, and pazopanib. Indazole derivatives are applied for the targeted treatment of lung, breast, colon, and prostate cancers. In this review, we compile the current development of indazole derivatives as kinase inhibitors and their application as anticancer agents in the past five years.
Collapse
Affiliation(s)
- Nitin Tandon
- School of Chemical Engineering and Physical Sciences, Lovely Professional University Phagwara-144411 India
| | - Vijay Luxami
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology Patiala-147004 India
| | - Divya Kant
- School of Chemical Engineering and Physical Sciences, Lovely Professional University Phagwara-144411 India
| | - Runjhun Tandon
- School of Chemical Engineering and Physical Sciences, Lovely Professional University Phagwara-144411 India
| | - Kamaldeep Paul
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology Patiala-147004 India
| |
Collapse
|
10
|
Shang C, Hou Y, Meng T, Shi M, Cui G. The Anticancer Activity of Indazole Compounds: A Mini Review. Curr Top Med Chem 2021; 21:363-376. [PMID: 33238856 DOI: 10.2174/1568026620999201124154231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 11/22/2022]
Abstract
The incidence and mortality of cancer continue to grow since the current medical treatments often fail to produce a complete and durable tumor response and ultimately give rise to therapy resistance and tumor relapse. Heterocycles with potential therapeutic values are of great pharmacological importance, and among them, indazole moiety is a privileged structure in medicinal chemistry. Indazole compounds possess potential anticancer activity, and indazole-based agents such as, axitinib, lonidamine and pazopanib have already been employed for cancer therapy, demonstrating indazole compounds as useful templates for the development of novel anticancer agents. The aim of this review is to present the main aspects of exploring anticancer properties, such as the structural modifications, the structure-activity relationship and mechanisms of action, making an effort to highlight the importance and therapeutic potential of the indazole compounds in the present anticancer agents.
Collapse
Affiliation(s)
- Congshan Shang
- Medical College, Xi'an Peihua University, Xi'an 710025, Shaanxi, China
| | - Yani Hou
- Medical College, Xi'an Peihua University, Xi'an 710025, Shaanxi, China
| | - Tingting Meng
- Medical College, Xi'an Peihua University, Xi'an 710025, Shaanxi, China
| | - Min Shi
- Medical College, Xi'an Peihua University, Xi'an 710025, Shaanxi, China
| | - Guoyan Cui
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, Shaanxi, China
| |
Collapse
|
11
|
Mary YS, Mary YS, Bielenica A, Armaković S, Armaković SJ, Chandramohan V, Dammalli M. Investigation of the reactivity properties of a thiourea derivative with anticancer activity by DFT and MD simulations. J Mol Model 2021; 27:217. [PMID: 34218339 DOI: 10.1007/s00894-021-04835-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/23/2021] [Indexed: 11/27/2022]
Abstract
Spectroscopic analysis of 1-(2-fluorophenyl)-3-[3-(trifluoromethyl)phenyl]thiourea (FPTT) is reported. Experimental and theoretical analyses of FPTT, with molecular dynamics (MD) simulations, are reported for finding different parameters like identification of suitable excipients, interactions with water, and sensitivity towards autoxidation. Molecular dynamics and docking show that FPTT can act as a potential inhibitor for new drug. Additionally, local reactivity, interactivity with water, and compatibility of FPTT molecule with frequently used excipients have been studied by combined application of density functional theory (DFT) and MD simulations. Analysis of local reactivity has been performed based on selected fundamental quantum-molecular descriptors, while interactivity with water was studied by calculations of radial distribution functions (RDFs). Compatibility with excipients has been assessed through calculations of solubility parameters, applying MD simulations. Graphical abstract Reactive sites identified.
Collapse
Affiliation(s)
| | | | - Anna Bielenica
- Department of Biochemistry, Medical University of Warsaw, 02-097, Warszawa, Poland
| | - Stevan Armaković
- Faculty of Sciences, Department of Physics, University of Novi Sad, Trg D. Obradovića 4, Novi Sad, 21000, Serbia
| | - Sanja J Armaković
- Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, University of Novi Sad, Trg D. Obradovića 3, Novi Sad, 21000, Serbia
| | - Vivek Chandramohan
- Department of Biotechnology, Siddaganga Institute of Technology, Tumakuru, Karnataka, 572103, India
| | - Manjunath Dammalli
- Department of Biotechnology, Siddaganga Institute of Technology, Tumakuru, Karnataka, 572103, India
| |
Collapse
|
12
|
Upadhyay N, Tilekar K, Safuan S, Kumar AP, Stalin J, Ruegg C, Ramaa C S. Recent Anti‐angiogenic Drug Discovery Efforts To Combat Cancer. ChemistrySelect 2021. [DOI: 10.1002/slct.202101792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Neha Upadhyay
- Department of Pharmaceutical Chemistry Bharati Vidyapeeth's College of Pharmacy Sector 8, CBD Belapur Navi Mumbai 400614 India
| | - Kalpana Tilekar
- Department of Pharmaceutical Chemistry Bharati Vidyapeeth's College of Pharmacy Sector 8, CBD Belapur Navi Mumbai 400614 India
| | - Sabreena Safuan
- Pusat pengajian sains School of Health Sciences Universiti Sains Malaysia Malaysia 16150 Kubang Kerian Kelantan
| | - Alan P. Kumar
- Department of Pharmacology National University of Singapore Singapore
| | - Jimmy Stalin
- Department of Oncology Microbiology, and Immunology University of Fribourg Chemin du Musée 18, PER17, CH 1700 Fribourg Switzerland
| | - Curzio Ruegg
- Department of Oncology Microbiology, and Immunology University of Fribourg Chemin du Musée 18, PER17, CH 1700 Fribourg Switzerland
| | - Ramaa C S
- Department of Pharmaceutical Chemistry Bharati Vidyapeeth's College of Pharmacy Sector 8, CBD Belapur Navi Mumbai 400614 India
| |
Collapse
|
13
|
Guo T, Ma S. Recent Advances in the Discovery of Multitargeted Tyrosine Kinase Inhibitors as Anticancer Agents. ChemMedChem 2020; 16:600-620. [PMID: 33179854 DOI: 10.1002/cmdc.202000658] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/28/2020] [Indexed: 12/18/2022]
Abstract
The treatment of cancer has been one of the most significant challenges for the medical field. Further research on the signal transduction pathway of tumor cells is driving the rapid development of antitumor agents targeting tyrosine kinases. However, most of the currently approved tyrosine kinase inhibitors based on the "single target/single drug" design are becoming less and less effective in the treatment of complex, heterogeneous, and multigenic cancers; this also results in resistance to chemotherapy. In contrast, multitargeted tyrosine kinase inhibitors (MT-TKIs) can effectively block multiple pathways of intracellular signal transduction. Therefore, they have therapeutic advantages over single-targeted inhibitors and have become a hotspot in antitumor drug research in recent years. This minireview summarizes recent advances in the discovery of MT-TKIs based on their chemical structures. In particular, we describe the kinase inhibitory and antitumor activity of promising compounds, as well as their structure - activity relationships (SARs).
Collapse
Affiliation(s)
- Ting Guo
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, West Wenhua Road 44, Jinan, 250012, P. R. China
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, West Wenhua Road 44, Jinan, 250012, P. R. China
| |
Collapse
|
14
|
Discovery of novel anti-angiogenesis agents. Part 11: Development of PROTACs based on active molecules with potency of promoting vascular normalization. Eur J Med Chem 2020; 205:112654. [DOI: 10.1016/j.ejmech.2020.112654] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
|
15
|
El-Damasy AK, Jin H, Seo SH, Bang EK, Keum G. Design, synthesis, and biological evaluations of novel 3-amino-4-ethynyl indazole derivatives as Bcr-Abl kinase inhibitors with potent cellular antileukemic activity. Eur J Med Chem 2020; 207:112710. [PMID: 32961435 DOI: 10.1016/j.ejmech.2020.112710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 10/23/2022]
Abstract
Breakpoint cluster region-Abelson (Bcr-Abl) kinase is a key driver in the pathophysiology of chronic myelogenous leukemia (CML). Broadening the chemical diversity of Bcr-Abl kinase inhibitors with novel chemical entities possessing favorable target potency and cellular efficacy is a current medical demand for CML treatment. In this respect, a new series of ethynyl bearing 3-aminoindazole based Bcr-Abl inhibitors has been designed, synthesized, and biologically evaluated. The target compounds were designed based on introducing the key structural features of ponatinib, alkyne spacer and diarylamide, into the previously reported indazole II to improve its Bcr-Abl inhibitory activity and overcome its poor cellular potency. All target compounds elicited potent activity against Bcr-AblWT with sub-micromolar IC50 values ranging 4.6-667 nM. In addition, certain derivatives exhibited promising potency over the clinically imatinib-resistant Bcr-AblT315I. Among the target molecules, compounds 9c, 9h and 10c stood as the most potent derivatives with IC50 values of 15.4 nM, 4.6 nM, and 25.8 nM, respectively, against Bcr-AblWT. Interestingly, 9h showed 2 folds and 3.6 times superior potency to the lead indazole II and 10c, respectively, against Bcr-AblT315I. Molecular docking of 9h pointed out its possibility to be a type II kinase inhibitor. Furthermore, all compounds, except 9b, showed highly potent antiproliferative activity against the Bcr-Abl positive leukemia K562 cell (MTT assay) surpassing the modest activity of lead indazole II. Moreover, the most potent members 9h and 10c exerted potent antileukemic activity against NCI leukemia panel, particularly K562 cell (SRB assay) with GI50 less than 10 nM, being superior to the FDA approved drug imatinib. Further biochemical hERG and cellular toxicity, phosphorylation assay, and NanoBRET target engagement of 9h underscored its merits as a promising candidate for CML therapy.
Collapse
Affiliation(s)
- Ashraf K El-Damasy
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Heewon Jin
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, 151-747, Republic of Korea
| | - Seon Hee Seo
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Gyochang Keum
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
16
|
Gingipalli L, Boerth J, Emmons D, Grebe T, Hatoum-Mokdad H, Peng B, Sha L, Tentarelli S, Wang H, Wu Y, Zheng X, Edmondson S, Gopalsamy A. Photoredox Catalysis: 1,4-Conjugate Addition of N-Methyl Radicals to Electron-Deficient Olefins via Decarboxylation of N-Substituted Acetic Acids. Org Lett 2020; 22:3418-3422. [DOI: 10.1021/acs.orglett.0c00873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Lakshmaiah Gingipalli
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Jeffrey Boerth
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - David Emmons
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Tyler Grebe
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Holia Hatoum-Mokdad
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Bo Peng
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Li Sha
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Sharon Tentarelli
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Haixia Wang
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Ye Wu
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - XiaoLan Zheng
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Scott Edmondson
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Ariamala Gopalsamy
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| |
Collapse
|
17
|
Rathi E, Kumar A, Suvarna, Kini G. Design of Potential Inhibitors and Prediction of their Activity by the Structural Insight of VEGFR2 Inhibitors: Atom‐based 3D‐QSAR, Fingerprint‐based 2D QSAR and Off‐target analysis. ChemistrySelect 2020. [DOI: 10.1002/slct.201903898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ekta Rathi
- Department of Pharmaceutical Chemistry Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhav Nagar, Manipal Karnataka India- 576104
| | - Avinash Kumar
- Department of Pharmaceutical Chemistry Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhav Nagar, Manipal Karnataka India- 576104
| | - Suvarna
- Department of Pharmaceutical Chemistry Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhav Nagar, Manipal Karnataka India- 576104
| | - G. Kini
- Department of Pharmaceutical Chemistry Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhav Nagar, Manipal Karnataka India- 576104
| |
Collapse
|
18
|
Alimohammadi A, Mostafavi H, Mahdavi M. Thiourea Derivatives Based on the Dapsone‐Naphthoquinone Hybrid as Anticancer and Antimicrobial Agents: In Vitro Screening and Molecular Docking Studies. ChemistrySelect 2020. [DOI: 10.1002/slct.201903179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Aazam Alimohammadi
- Department of Organic Chemistry & Biochemistry University of Tabriz 29 BahmanBoulvard Tabriz 5166614766 Iran
| | - Hossein Mostafavi
- Department of Organic Chemistry & Biochemistry University of Tabriz 29 BahmanBoulvard Tabriz 5166614766 Iran
| | - Majid Mahdavi
- Biology, Faculty of Natural Sciences University of Tabriz 29 Bahman Boulvard Tabriz 5166614766 Iran
| |
Collapse
|
19
|
de Oliveira Viana J, Monteiro AFM, Filho JMB, Scotti L, Scotti MT. The Azoles in Pharmacochemistry: Perspectives on the Synthesis of New Compounds and Chemoinformatic Contributions. Curr Pharm Des 2020; 25:4702-4716. [DOI: 10.2174/1381612825666191125090700] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022]
Abstract
:
Due to their versatile biological activity, Azoles are widely studied in pharmacochemistry. It is possible
to use them in many applications and in studies aimed at discovering antiparasitic, antineoplastic, antiviral,
antimicrobial compounds; and in the production of materials for treatment of varied pathologies. Based on their
biological activity, our review presents several studies that involve this class of organic compounds. A bibliographic
survey of this type can effectively contribute to pharmaceutical sciences, stimulating the discovery of new
compounds, and structural improvements to biological profiles of interest. In this review, articles are discussed
involving the synthesis of new compounds and chemoinformatic contributions. Current applications of azoles in
both the pharmaceutical and agri-business sectors are well known, yet as this research highlights, azole compounds
can also bring important contributions to the fight against many diseases. Among the heterocyclics, azoles
are increasingly studied by research groups around the world for application against tuberculosis, HIV, fungal and
bacterial infections; and against parasites such as leishmaniasis and trypanosomiasis. Our hope is that this work
will help arouse the interest of research groups planning to develop new bioactives to fight against these and
other diseases.
Collapse
Affiliation(s)
- Jéssika de Oliveira Viana
- Natural and Synthetic Bioactive Products Program (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa- PB, Brazil
| | - Alex France Messias Monteiro
- Natural and Synthetic Bioactive Products Program (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa- PB, Brazil
| | - José Maria Barbosa Filho
- Natural and Synthetic Bioactive Products Program (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa- PB, Brazil
| | - Luciana Scotti
- Natural and Synthetic Bioactive Products Program (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa- PB, Brazil
| | - Marcus Tullius Scotti
- Natural and Synthetic Bioactive Products Program (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa- PB, Brazil
| |
Collapse
|
20
|
Liao B, Peng L, Zhou J, Mo H, Zhao J, Yang Z, Guo X, Zhang P, Zhang X, Zhu Z. Synthesis and Activity Evaluation of Nasopharyngeal Carcinoma Inhibitors Based on 6-(Pyrimidin-4-yl)-1H-indazole. Chem Biodivers 2019; 16:e1800598. [PMID: 30788913 DOI: 10.1002/cbdv.201800598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/18/2019] [Indexed: 11/07/2022]
Abstract
Human nasopharyngeal carcinoma is a common head and neck malignancy with high incidence in Southeast Asia and Southern China. It is necessary to develop safe, effective and inexpensive anticancer agents to improve the therapeutics of patients with nasopharyngeal carcinoma. A series of small molecular compounds based on 6-(pyrimidin-4-yl)-1H-indazole were synthesized and evaluated for antiproliferative activities against human nasopharyngeal carcinoma cell lines SUNE1. Compounds 6b, 6c, 6e and 6l showed potent antiproliferative activities similar to positive control drug cisplatin in vitro with lower nephrotoxicity than it. N-[4-(1H-Indazol-6-yl)pyrimidin-2-yl]benzene-1,3-diamine (6l) was selected for further study. It was found that 6l induced mitochondria-mediated apoptosis and G2 /M phase arrest in SUNE1 cells. Furthermore, compound 6l at 10 mg/kg can suppress the growth of an implanted SUNE1 xenograft with a TGI% (tumor growth inhibition) value of 50 % and did not cause serious side effects in BALB/c nude mice. This study suggests that 6-(pyrimidin-4-yl)-1H-indazole derivatives are a series of small molecule compounds with anti-nasopharyngeal carcinoma activities.
Collapse
Affiliation(s)
- Bohong Liao
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Lingrong Peng
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Jin Zhou
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Huiting Mo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Jialan Zhao
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Zike Yang
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Xiaowen Guo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Peiquan Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xin Zhang
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Zhibo Zhu
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| |
Collapse
|
21
|
Zeidan MA, Mostafa AS, Gomaa RM, Abou-zeid LA, El-Mesery M, El-Sayed MAA, Selim KB. Design, synthesis and docking study of novel picolinamide derivatives as anticancer agents and VEGFR-2 inhibitors. Eur J Med Chem 2019; 168:315-329. [DOI: 10.1016/j.ejmech.2019.02.050] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/03/2019] [Accepted: 02/16/2019] [Indexed: 10/27/2022]
|
22
|
Discovery of novel anti-angiogenesis agents. Part 9: Multiplex inhibitors suppressing compensatory activations of RTKs. Eur J Med Chem 2019; 164:440-447. [DOI: 10.1016/j.ejmech.2018.12.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/25/2018] [Accepted: 12/25/2018] [Indexed: 11/21/2022]
|
23
|
Zhang SG, Liang CG, Zhang WH. Recent Advances in Indazole-Containing Derivatives: Synthesis and Biological Perspectives. Molecules 2018; 23:E2783. [PMID: 30373212 PMCID: PMC6278422 DOI: 10.3390/molecules23112783] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/14/2018] [Accepted: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
Indazole-containing derivatives represent one of the most important heterocycles in drug molecules. Diversely substituted indazole derivatives bear a variety of functional groups and display versatile biological activities; hence, they have gained considerable attention in the field of medicinal chemistry. This review aims to summarize the recent advances in various methods for the synthesis of indazole derivatives. The current developments in the biological activities of indazole-based compounds are also presented.
Collapse
Affiliation(s)
- Shu-Guang Zhang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing 210095, China.
| | - Chao-Gen Liang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing 210095, China.
| | - Wei-Hua Zhang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
24
|
Bielenica A, Sanna G, Madeddu S, Giliberti G, Stefańska J, Kozioł AE, Savchenko O, Strzyga-Łach P, Chrzanowska A, Kubiak-Tomaszewska G, Struga M. Disubstituted 4-Chloro-3-nitrophenylthiourea Derivatives: Antimicrobial and Cytotoxic Studies. Molecules 2018; 23:molecules23102428. [PMID: 30248936 PMCID: PMC6222614 DOI: 10.3390/molecules23102428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/23/2022] Open
Abstract
4-Chloro-3-nitrophenylthioureas 1–30 were synthesized and tested for their antimicrobial and cytotoxic activities. Compounds exhibited high to moderate antistaphylococcal activity against both standard and clinical strains (MIC values 2–64 μg/mL). Among them derivatives with electron-donating alkyl substituents at the phenyl ring were the most promising. Moreover, compounds 1–6 and 8–19 were cytotoxic against MT-4 cells and various other cell lines derived from human hematological tumors (CC50 ≤ 10 μM). The influence of derivatives 11, 13 and 25 on viability, mortality and the growth rate of immortalized human keratinocytes (HaCaT) was observed.
Collapse
Affiliation(s)
- Anna Bielenica
- Department of Biochemistry, Medical University, 02-097 Warszawa, Poland.
| | - Giuseppina Sanna
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliary, Cittadella Universitaria, 09042 Monserrato, Italy.
| | - Silvia Madeddu
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliary, Cittadella Universitaria, 09042 Monserrato, Italy.
| | - Gabriele Giliberti
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliary, Cittadella Universitaria, 09042 Monserrato, Italy.
| | - Joanna Stefańska
- Department of Pharmaceutical Microbiology, Medical University, 02-007 Warszawa, Poland.
| | - Anna E Kozioł
- Faculty of Chemistry, Maria Curie-Sklodowska University, 20-031 Lublin, Poland.
| | | | | | - Alicja Chrzanowska
- Department of Biochemistry, Medical University, 02-097 Warszawa, Poland.
| | - Grażyna Kubiak-Tomaszewska
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warszawa, Poland.
| | - Marta Struga
- Department of Biochemistry, Medical University, 02-097 Warszawa, Poland.
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warszawa, Poland.
| |
Collapse
|
25
|
Dong J, Zhang Q, Wang Z, Huang G, Li S. Recent Advances in the Development of Indazole-based Anticancer Agents. ChemMedChem 2018; 13:1490-1507. [PMID: 29863292 DOI: 10.1002/cmdc.201800253] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/25/2018] [Indexed: 12/20/2022]
Abstract
Cancer is one of the leading causes of human mortality globally; therefore, intensive efforts have been made to seek new active drugs with improved anticancer efficacy. Indazole-containing derivatives are endowed with a broad range of biological properties, including anti-inflammatory, antimicrobial, anti-HIV, antihypertensive, and anticancer activities. In recent years, the development of anticancer drugs has given rise to a wide range of indazole derivatives, some of which exhibit outstanding activity against various tumor types. The aim of this review is to outline recent developments concerning the anticancer activity of indazole derivatives, as well as to summarize the design strategies and structure-activity relationships of these compounds.
Collapse
Affiliation(s)
- Jinyun Dong
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Qijing Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Zengtao Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Guang Huang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Shaoshun Li
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| |
Collapse
|
26
|
Shan Y, Wang B, Zhang J. New strategies in achieving antiangiogenic effect: Multiplex inhibitors suppressing compensatory activations of RTKs. Med Res Rev 2018; 38:1674-1705. [DOI: 10.1002/med.21517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/19/2018] [Accepted: 05/19/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Yuanyuan Shan
- Department of Pharmacy; The First Affiliated Hospital of Xi'an Jiaotong University; Xi'an China
| | - Binghe Wang
- Department of Chemistry; Center for Diagnostics and Therapeutics; Georgia State University; Atlanta GA USA
| | - Jie Zhang
- School of Pharmacy, Health Science Center; Xi'an Jiaotong University; Xi'an China
| |
Collapse
|