1
|
Malarz K, Kuczak M, Rurka P, Rawicka P, Boguszewska-Czubara A, Jampilek J, Mularski J, Musiol R, Mrozek-Wilczkiewicz A. Unveiling the role of Ndrg1 gene on the oxidative stress induction behind the anticancer potential of styrylquinazoline derivatives. Sci Rep 2025; 15:16081. [PMID: 40341822 PMCID: PMC12062220 DOI: 10.1038/s41598-025-99277-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
This work presents a multifaceted mechanism of the anticancer action of a 2-styrylquinazoline derivative. Extensive analysis of various aspects related to tyrosine kinase inhibition and effects on cellular targets at both the gene and protein levels revealed the potential of this IS20 compound for future research. This study presents a detailed analysis of the relationship between ABL and SRC kinase affecting the inhibition of the EGFR/mTOR signaling pathway in a non-obvious manner. The study was supported by experiments using various molecular biology techniques to confirm the induction of oxidative stress, inhibition of the cell cycle in the G2/M phase and the triggering of cell death via both the apoptosis and autophagy pathways. The cell models included those with different p53 protein status, which affected the cellular response in the form of altered Ndrg1 expression. Finally, the appropriate physicochemical properties of IS20 for adequate bioavailability and toxicity to the body were observed in an in vivo model.
Collapse
Affiliation(s)
- Katarzyna Malarz
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland.
- Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland.
| | - Michał Kuczak
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland
| | - Patryk Rurka
- Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland
| | - Patrycja Rawicka
- Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4a, Lublin, 20-093, Poland
| | - Josef Jampilek
- Department of Chemical Biology, Palacky University Olomouc, Slechtitelu 27, Olomouc, 779 00, Czech Republic
| | - Jacek Mularski
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland
| | - Anna Mrozek-Wilczkiewicz
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
- Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, Chorzów, 41-500, Poland
| |
Collapse
|
2
|
Mansour MA, AboulMagd AM, Abbas SH, Abdel-Aziz M, Abdel-Rahman HM. Quinazoline-chalcone hybrids as HDAC/EGFR dual inhibitors: Design, synthesis, mechanistic, and in-silico studies of potential anticancer activity against multiple myeloma. Arch Pharm (Weinheim) 2024; 357:e2300626. [PMID: 38297894 DOI: 10.1002/ardp.202300626] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/02/2024]
Abstract
Two new series of quinazoline-chalcone hybrids were designed, synthesized as histone deacetylase (HDAC)/epidermal growth factor receptor (EGFR) dual inhibitors, and screened in vitro against the NCI 60 human cancer cell line panel. The most potent derivative, compound 5e bearing a 3,4,5-trimethoxyphenyl chalcone moiety, showed the most effective growth inhibition value against the panel of NCI 60 human cancer cell lines. Thus, it was selected for further investigation for NCI 5 log doses. Interestingly, this trimethoxy-substituted analog inhibited the proliferation of Roswell Park Memorial Institute (RPMI)-8226 cells by 96%, at 10 µM with IC50 = 9.09 ± 0.34 µM and selectivity index = 7.19 against normal blood cells. To confirm the selectivity of this compound, it was evaluated against a panel of tyrosine kinase enzymes. Mechanistically, it successfully and selectively inhibited HDAC6, HDAC8, and EGFR with IC50 = 0.41 ± 0.015, 0.61 ± 0.027, and 0.09 ± 0.004 µM, respectively. Furthermore, the selected derivative induced apoptosis via the mitochondrial apoptotic pathway by raising the Bax/Bcl-2 ratio and activating caspases 3, 7, and 9. Also, the flow cytometry analysis of RPMI-8226 cells showed that the trimethoxy-substituted analog produced cell cycle arrest in the G1 and S phases at 55.82%. Finally, an in silico study was performed to explore the binding interaction of the most active compound within the zinc-containing binding site of HDAC6 and HDAC8.
Collapse
Affiliation(s)
- Mostafa A Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University in Beni-Suef (NUB), Beni-Suef, Egypt
| | - Asmaa M AboulMagd
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University in Beni-Suef (NUB), Beni-Suef, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Hamdy M Abdel-Rahman
- Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Assiut (BUA), Assiut, Egypt
| |
Collapse
|
3
|
Malarz K, Mularski J, Pacholczyk M, Musiol R. Styrylquinazoline derivatives as ABL inhibitors selective for different DFG orientations. J Enzyme Inhib Med Chem 2023; 38:2201410. [PMID: 37070569 PMCID: PMC10120462 DOI: 10.1080/14756366.2023.2201410] [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: 01/21/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/19/2023] Open
Abstract
Among tyrosine kinase inhibitors, quinazoline-based compounds represent a large and well-known group of multi-target agents. Our previous studies have shown interesting kinases inhibition activity for a series of 4-aminostyrylquinazolines based on the CP-31398 scaffold. Here, we synthesised a new series of styrylquinazolines with a thioaryl moiety in the C4 position and evaluated in detail their biological activity. Our results showed high inhibition potential against non-receptor tyrosine kinases for several compounds. Molecular docking studies showed differential binding to the DFG conformational states of ABL kinase for two derivatives. The compounds showed sub-micromolar activity against leukaemia. Finally, in-depth cellular studies revealed the full landscape of the mechanism of action of the most active compounds. We conclude that S4-substituted styrylquinazolines can be considered as a promising scaffold for the development of multi-kinase inhibitors targeting a desired binding mode to kinases as effective anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Malarz
- Institute of Physics, University of Silesia in Katowice, Chorzów, Poland
| | - Jacek Mularski
- Institute of Chemistry, University of Silesia in Katowice, Chorzów, Poland
| | - Marcin Pacholczyk
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, Chorzów, Poland
| |
Collapse
|
4
|
Wu W, Fan S, Wu X, Fang L, Zhu J. Cobalt Homeostatic Catalysis for Coupling of Enaminones and Oxadiazolones to Quinazolinones. J Org Chem 2023; 88:1945-1962. [PMID: 36705660 DOI: 10.1021/acs.joc.2c01934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Transition metal catalysis has revolutionized modern synthetic chemistry for its diverse modes of coordination reactivity. However, this versatility in reactivity is also the predominant cause of catalyst deactivation, a persisting issue that can significantly compromise its synthetic value. Homeostatic catalysis, a catalytic process that can sustain its productive catalytic cycle even when chemically disturbed, is proposed herein as an effective tactic to address the challenge. In particular, a cobalt homeostatic catalysis process has been developed for the water-tolerant coupling of enaminones and oxadiazolones to quinazolinones. Dynamic covalent bonding serves as a mechanistic handle for the preferred buffering of water onto enaminone and reverse exchange by a released secondary amine, thus securing reversible entry into cobalt's dormant and active states for productive catalysis. Through this homeostatic catalysis mode, a broad structural scope has been achieved for quinazolinones, enabling further elaboration into distinct pharmaceutically active agents.
Collapse
Affiliation(s)
- Weiping Wu
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, China
| | - Shuaixin Fan
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, China
| | - Xuan Wu
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, China
| | - Lili Fang
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, China
| | - Jin Zhu
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, China
| |
Collapse
|
5
|
Hricovíni M, Owens RJ, Bak A, Kozik V, Musiał W, Pierattelli R, Májeková M, Rodríguez Y, Musioł R, Slodek A, Štarha P, Piętak K, Słota D, Florkiewicz W, Sobczak-Kupiec A, Jampílek J. Chemistry towards Biology-Instruct: Snapshot. Int J Mol Sci 2022; 23:14815. [PMID: 36499140 PMCID: PMC9739621 DOI: 10.3390/ijms232314815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
The knowledge of interactions between different molecules is undoubtedly the driving force of all contemporary biomedical and biological sciences. Chemical biology/biological chemistry has become an important multidisciplinary bridge connecting the perspectives of chemistry and biology to the study of small molecules/peptidomimetics and their interactions in biological systems. Advances in structural biology research, in particular linking atomic structure to molecular properties and cellular context, are essential for the sophisticated design of new medicines that exhibit a high degree of druggability and very importantly, druglikeness. The authors of this contribution are outstanding scientists in the field who provided a brief overview of their work, which is arranged from in silico investigation through the characterization of interactions of compounds with biomolecules to bioactive materials.
Collapse
Affiliation(s)
- Miloš Hricovíni
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Raymond J. Owens
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK, University of Oxford, Oxford OX11 0QS, UK
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Andrzej Bak
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Violetta Kozik
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211A, 50 556 Wrocław, Poland
| | - Roberta Pierattelli
- Magnetic Resonance Center and Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Magdaléna Májeková
- Center of Experimental Medicine SAS and Department of Biochemical Pharmacology, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovakia
| | - Yoel Rodríguez
- Department of Natural Sciences, Eugenio María de Hostos Community College, City University of New York, 500 Grand Concourse, Bronx, NY 10451, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Robert Musioł
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Aneta Slodek
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Pavel Štarha
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Karina Piętak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Dagmara Słota
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Wioletta Florkiewicz
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Agnieszka Sobczak-Kupiec
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Josef Jampílek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
6
|
Hamdi A, El-Shafey HW, Othman DI, El-Azab AS, AlSaif NA, A.-M. Abdel-Aziz A. Design, synthesis, antitumor, and VEGFR-2 inhibition activities of novel 4-anilino-2-vinyl-quinazolines: Molecular modeling studies. Bioorg Chem 2022; 122:105710. [DOI: 10.1016/j.bioorg.2022.105710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/09/2022] [Accepted: 02/25/2022] [Indexed: 01/21/2023]
|
7
|
Malarz K, Mularski J, Kuczak M, Mrozek-Wilczkiewicz A, Musiol R. Novel Benzenesulfonate Scaffolds with a High Anticancer Activity and G2/M Cell Cycle Arrest. Cancers (Basel) 2021; 13:cancers13081790. [PMID: 33918637 PMCID: PMC8068801 DOI: 10.3390/cancers13081790] [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: 03/02/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Sulfonates, unlike their derivatives, sulphonamides, have rarely been investigated for their anticancer activity. Unlike the well-known sulphonamides, esters are mainly used as convenient intermediates in a synthesis. Here, we present the first in-depth investigation of quinazoline sulfonates. A small series of derivatives were synthesized and tested for their anticancer activity. Based on their structural similarity, these compounds resemble tyrosine kinase inhibitors and the p53 reactivator CP-31398. Their biological activity profile, however, was more related to sulphonamides because there was a strong cell cycle arrest in the G2/M phase. Further investigation revealed a multitargeted mechanism of the action that corresponded to the p53 protein status in the cell. Although the compounds expressed a high submicromolar activity against leukemia and colon cancers, pancreatic cancer and glioblastoma were also susceptible. Apoptosis and autophagy were confirmed as the cell death modes that corresponded with the inhibition of metabolic activity and the activation of the p53-dependent and p53-independent pathways. Namely, there was a strong activation of the p62 protein and GADD44. Other proteins such as cdc2 were also expressed at a higher level. Moreover, the classical caspase-dependent pathway in leukemia was observed at a lower concentration, which again confirmed a multitargeted mechanism. It can therefore be concluded that the sulfonates of quinazolines can be regarded as promising scaffolds for developing anticancer agents.
Collapse
Affiliation(s)
- Katarzyna Malarz
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland; (M.K.); (A.M.-W.)
- Correspondence: (K.M.); (R.M.)
| | - Jacek Mularski
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland;
| | - Michał Kuczak
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland; (M.K.); (A.M.-W.)
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland;
| | - Anna Mrozek-Wilczkiewicz
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland; (M.K.); (A.M.-W.)
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland;
- Correspondence: (K.M.); (R.M.)
| |
Collapse
|
8
|
Grover P, Bhardwaj M, Kapoor G, Mehta L, Ghai R, Nagarajan K. Advances on Quinazoline Based Congeners for Anticancer Potential. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210212121056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The heterocyclic compounds have a great significance in medicinal chemistry because
they have extensive biological activities. Cancer is globally the leading cause of death
and it is a challenge to develop appropriate treatment for the management of cancer. Continuous
efforts are being made to find a suitable medicinal agent for cancer therapy. Nitrogencontaining
heterocycles have received noteworthy attention due to their wide and distinctive
pharmacological activities. One of the most important nitrogen-containing heterocycles in
medicinal chemistry is ‘quinazoline’ that possesses a wide spectrum of biological properties.
This scaffold is an important pharmacophore and is considered a privileged structure. Various
substituted quinazolines displayed anticancer activity against different types of cancer. This
review highlights the recent advances in quinazoline based molecules as anticancer agents.
Several in-vitro and in-vivo models used along with the results are also included. A subpart briefing natural quinazoline
containing anticancer compounds is also incorporated in the review.
Collapse
Affiliation(s)
- Parul Grover
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - Monika Bhardwaj
- Natural Product Chemistry Division, Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Garima Kapoor
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - Lovekesh Mehta
- Amity Institute of Pharmacy, Amity University, Noida, 201301, India
| | - Roma Ghai
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - K. Nagarajan
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| |
Collapse
|
9
|
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: 25] [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
|
10
|
Wu JT, Lin CL, Huang CJ, Cheng YC, Chien CC, Sung YC. Potential synergistic effects of sorafenib and CP-31398 for treating anaplastic thyroid cancer with p53 mutations. Oncol Lett 2020; 19:3021-3026. [PMID: 32218859 DOI: 10.3892/ol.2020.11377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Thyroid cancer is the most commonly diagnosed endocrine cancer. Anaplastic thyroid cancer (ATC) is the most aggressive type of thyroid cancer and has a poor prognosis. Loss of p53 function has been reported to lead to poorly differentiated thyroid tumors; therefore, mutant p53 protein can be considered a crucial therapeutic target in patients with ATC. Sorafenib, a multi-kinase inhibitor, has been approved for the treatment of metastatic and differentiated thyroid cancer. Combined targeted therapy, including sorafenib, may be clinically significant for patients with ATC harboring p53 mutations. In the present study, CP-31398, a p53-restoring agent, was used to improve the therapeutic efficacy of sorafenib in SW579 cells, an ATC cell line harboring p53 mutations. The molecular function of CP-31398 was evaluated using western blot analysis and a luciferase reporter assay. The decreased viability of SW579 cells, following CP-31398 treatment, was augmented by sorafenib, and CP-31398 enhanced the antimitogenic effect of sorafenib; thus, sorafenib and CP-31398 synergistically inhibited the growth of SW579 cells. These results indicate a potential clinical application of CP-31398 for patients with ATC harboring p53 abnormalities, since these individuals generally respond poorly to sorafenib alone.
Collapse
Affiliation(s)
- Jiin-Torng Wu
- Division of Respiratory Therapy and Chest Medicine, Department of Internal Medicine, Cathay General Hospital, Taipei 11031, Taiwan, R.O.C
| | - Ching-Ling Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Cathay General Hospital, Taipei 11031, Taiwan, R.O.C.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Chi-Jung Huang
- Department of Medical Research, Cathay General Hospital, Taipei 11490, Taiwan, R.O.C.,Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan, R.O.C.,School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan, R.O.C
| | - Yu-Che Cheng
- Department of Medical Research, Cathay General Hospital, Taipei 11490, Taiwan, R.O.C.,School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan, R.O.C.,Institute of Biomedical Engineering, Center for Biocellular Engineering, National Central University, Taoyuan 32001, Taiwan, R.O.C
| | - Chih-Cheng Chien
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan, R.O.C.,Department of Anesthesiology, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Yung-Chuan Sung
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan, R.O.C.,Division of Hematology/Oncology, Department of Internal Medicine, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| |
Collapse
|
11
|
Malarz K, Mularski J, Pacholczyk M, Musiol R. The Landscape of the Anti-Kinase Activity of the IDH1 Inhibitors. Cancers (Basel) 2020; 12:cancers12030536. [PMID: 32110969 PMCID: PMC7139656 DOI: 10.3390/cancers12030536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/20/2020] [Accepted: 02/22/2020] [Indexed: 12/18/2022] Open
Abstract
Isocitrate dehydrogenases constitute a class of enzymes that are crucial for cellular metabolism. The overexpression or mutation of isocitrate dehydrogenases are often found in leukemias, glioblastomas, lung cancers, and ductal pancreatic cancer among others. Mutation R132H, which changes the functionality of an enzyme to produce mutagenic 2-hydroxyglutarate instead of a normal product, is particularly important in this field. A series of inhibitors were described for these enzymes of which ivosidenib was the first to be approved for treating leukemia and bile duct cancers in 2018. Here, we investigated the polypharmacological landscape of the activity for known sulfamoyl derivatives that are inhibitors, which are selective towards IDH1 R132H. These compounds appeared to be effective inhibitors of several non-receptor kinases at a similar level as imatinib and axitinib. The antiproliferative activity of these compounds against a panel of cancer cells was tested and is explained based on the relative expression levels of the investigated proteins. The multitargeted activity of these compounds makes them valuable agents against a wide range of cancers, regardless of the status of IDH1.
Collapse
Affiliation(s)
- Katarzyna Malarz
- August Chełkowski Institute of Physics and Silesian Center for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1, 41-500 Chorzów, Poland
- Correspondence: (K.M.); (R.M.)
| | - Jacek Mularski
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1A, 41-500 Chorzów, Poland;
| | - Marcin Pacholczyk
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland;
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1A, 41-500 Chorzów, Poland;
- Correspondence: (K.M.); (R.M.)
| |
Collapse
|
12
|
Musiol R. Styrylquinoline – A Versatile Scaffold in Medicinal Chemistry. Med Chem 2020; 16:141-154. [PMID: 31161997 DOI: 10.2174/1573406415666190603103012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 03/08/2019] [Accepted: 05/03/2019] [Indexed: 11/22/2022]
Abstract
Background: :
Styrylquinolines are characteristic fully aromatic compounds with flat,
rather lipophilic structures. The first reports on their synthesis and biological activity were published
roughly a century ago. However, their low selectivity, unfavorable toxicity and problems
with their mechanism of action significantly hampered their development. As a result, they have
been abandoned for most of the time since they were discovered.
Objective: :
Their renaissance was observed by the antiretroviral activity of several styrylquinoline
derivatives that have been reported to be HIV integrase inhibitors. Subsequently, other activities
such as their antifungal and anticancer abilities have also been revisited.
Methods:
In the present review, the spectrum of the activity of styrylquinolines and their use in
drug design is presented and analyzed.
Results:
New properties and applications that were reported recently have re-established
styrylquinolines within medicinal and material chemistry. The considerable increase in the number
of published papers regarding their activity spectrum will ensure further discoveries in the field.
Conclusions:
Styrylquinolines have earned a much stronger position in medicinal chemistry due to
the discovery of their new activities, profound mechanisms of action and as drug candidates in
clinical trials.
Collapse
Affiliation(s)
- Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, 75 Pulku Piechoty 1A, 41-500, Chorzow, Poland
| |
Collapse
|
13
|
Glycofullerenes as non-receptor tyrosine kinase inhibitors- towards better nanotherapeutics for pancreatic cancer treatment. Sci Rep 2020; 10:260. [PMID: 31937861 PMCID: PMC6959220 DOI: 10.1038/s41598-019-57155-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/23/2019] [Indexed: 12/27/2022] Open
Abstract
The water-soluble glycofullerenes GF1 and GF2 were synthesized using two-step modified Bingel-Hirsch methodology. Interestingly, we identified buckyballs as a novel class of non-receptor Src kinases inhibitors. The evaluated compounds were found to inhibit Fyn A and BTK proteins with IC50 values in the low micromolar range, with the most active compound at 39 µM. Moreover, we have demonstrated that formation of protein corona on the surface of [60]fullerene derivatives is changing the landscape of their activity, tuning the selectivity of obtained carbon nanomaterials towards Fyn A and BTK kinases. The performed molecular biology studies revealed no cytotoxicity and no influence of engineered carbon nanomaterials on the cell cycle of PANC-1 and AsPC-1 cancer cell lines. Incubation with the tested compounds resulted in the cellular redox imbalance triggering the repair systems and influenced the changing of protein levels.
Collapse
|
14
|
2-Styryl-4-aminoquinazoline derivatives as potent DNA-cleavage, p53-activation and in vivo effective anticancer agents. Eur J Med Chem 2020; 186:111851. [DOI: 10.1016/j.ejmech.2019.111851] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/03/2019] [Accepted: 11/03/2019] [Indexed: 02/06/2023]
|
15
|
Mrozek-Wilczkiewicz A, Kuczak M, Malarz K, Cieślik W, Spaczyńska E, Musiol R. The synthesis and anticancer activity of 2-styrylquinoline derivatives. A p53 independent mechanism of action. Eur J Med Chem 2019; 177:338-349. [DOI: 10.1016/j.ejmech.2019.05.061] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/13/2022]
|