1
|
Elagawany M, Abdel Ghany LMA, Ibrahim TS, Alharbi AS, Abdel-Aziz MS, El-labbad EM, Ryad N. Development of certain benzylidene coumarin derivatives as anti-prostate cancer agents targeting EGFR and PI3Kβ kinases. J Enzyme Inhib Med Chem 2024; 39:2311157. [PMID: 38348846 PMCID: PMC10866054 DOI: 10.1080/14756366.2024.2311157] [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: 12/04/2023] [Accepted: 01/22/2024] [Indexed: 02/15/2024] Open
Abstract
Novel coumarin derivatives were synthesised and tested for their cytotoxicity against human cancer cells (PC-3 and MDA-MB-231). Compounds 5, 4b, and 4a possessed potent cytotoxic activity against PC-3 cells with IC50 3.56, 8.99, and 10.22 µM, respectively. Compound 4c displayed cytotoxicity more than erlotinib in the MDA-MB-231 cells with IC50 8.5 µM. Moreover, compound 5 exhibited potent inhibitory activity on EFGR with IC50 0.1812 µM, as well as PI3Kβ inhibitory activity that was twofold higher than LY294002, suggesting that this compound has a dual EGFR and PI3Kβ inhibiting activity. Docking aligns with the in vitro results and sheds light on the molecular mechanisms underlying dual targeting. Furthermore, compound 5 decreased AKT and m-TOR expression in PC-3 cells, showing that it specifically targets these cells via the EGFR/PI3K/Akt/m-TOR signalling pathway. Simultaneously, compound 5 caused cell cycle arrest at S phase and induced activation of both intrinsic and extrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Mohamed Elagawany
- Department of Pharmaceutical Chemistry, Damanhour University, Damanhour, Buhaira, Egypt
| | - Lina M. A. Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza, Egypt
| | - Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrhman S. Alharbi
- Department of Chemistry, College of Science and Arts, Shaqra University, Sajir, Shaqra, Saudi Arabia
| | - Mohamed S. Abdel-Aziz
- Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
| | - Eman M. El-labbad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Noha Ryad
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza, Egypt
| |
Collapse
|
2
|
Honndorf VS, Wiehr S, Rolle AM, Schmitt J, Kreft L, Quintanilla-Martinez L, Kohlhofer U, Reischl G, Maurer A, Boldt K, Schwarz M, Schmidt H, Pichler BJ. Preclinical evaluation of the anti-tumor effects of the natural isoflavone genistein in two xenograft mouse models monitored by [18F]FDG, [18F]FLT, and [64Cu]NODAGA-cetuximab small animal PET. Oncotarget 2017; 7:28247-61. [PMID: 27070087 PMCID: PMC5053724 DOI: 10.18632/oncotarget.8625] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/28/2016] [Indexed: 12/11/2022] Open
Abstract
The natural phytoestrogen genistein is known as protein kinase inhibitor and tumor suppressor in various types of cancers. We studied its antitumor effect in two different xenograft models using positron emission tomography (PET) in vivo combined with ex vivo histology and nuclear magnetic resonance (NMR) metabolic fingerprinting.
Collapse
Affiliation(s)
- Valerie S Honndorf
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | - Stefan Wiehr
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | - Anna-Maria Rolle
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | - Julia Schmitt
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | - Luisa Kreft
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | | | - Ursula Kohlhofer
- Institute of Pathology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| | - Karsten Boldt
- Medical Proteome Center, Institute for Ophthalmic Research, Eberhard Karls University, Tuebingen, Germany
| | - Michael Schwarz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Toxicology, Eberhard Karls University, Tuebingen, Germany
| | - Holger Schmidt
- Department of Radiology, Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany
| |
Collapse
|
3
|
He Y, Schmidt MA, Erwin C, Guo J, Sun R, Pendarvis K, Warner BW, Herman EM. Transgenic Soybean Production of Bioactive Human Epidermal Growth Factor (EGF). PLoS One 2016; 11:e0157034. [PMID: 27314851 PMCID: PMC4912142 DOI: 10.1371/journal.pone.0157034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/24/2016] [Indexed: 12/16/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition of premature infants that results from the gut microbiome invading immature intestinal tissues. This results in a life-threatening disease that is frequently treated with the surgical removal of diseased and dead tissues. Epidermal growth factor (EGF), typically found in bodily fluids, such as amniotic fluid, salvia and mother's breast milk, is an intestinotrophic growth factor and may reduce the onset of NEC in premature infants. We have produced human EGF in soybean seeds to levels biologically relevant and demonstrated its comparable activity to commercially available EGF. Transgenic soybean seeds expressing a seed-specific codon optimized gene encoding of the human EGF protein with an added ER signal tag at the N' terminal were produced. Seven independent lines were grown to homozygous and found to accumulate a range of 6.7 +/- 3.1 to 129.0 +/- 36.7 μg EGF/g of dry soybean seed. Proteomic and immunoblot analysis indicates that the inserted EGF is the same as the human EGF protein. Phosphorylation and immunohistochemical assays on the EGF receptor in HeLa cells indicate the EGF protein produced in soybean seed is bioactive and comparable to commercially available human EGF. This work demonstrates the feasibility of using soybean seeds as a biofactory to produce therapeutic agents in a soymilk delivery platform.
Collapse
Affiliation(s)
- Yonghua He
- School of Plant Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Monica A. Schmidt
- School of Plant Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Christopher Erwin
- St. Louis Children's Hospital and Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jun Guo
- St. Louis Children's Hospital and Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Raphael Sun
- St. Louis Children's Hospital and Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ken Pendarvis
- School of Animal & Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Brad W. Warner
- St. Louis Children's Hospital and Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Eliot M. Herman
- School of Plant Sciences, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
4
|
Aggarwal S, Sinha D, Tiwari AK, Pooja P, Kaul A, Singh G, Mishra AK. Studies for development of novel quinazolinones: new biomarker for EGFR. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2015; 143:309-318. [PMID: 25766241 DOI: 10.1016/j.saa.2015.01.069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/17/2014] [Accepted: 01/27/2015] [Indexed: 06/04/2023]
Abstract
The binding capabilities of a series of novel quinazolinone molecules were established and stated in a comprehensive computational methodology as well as by in vitro analysis. The main focus of this work was to achieve more insight of the interactions with crystal structure of PDB ID: 1M17 and predict their binding mode to EGFR. Three molecules were screened for further examination, which were synthesized and characterized using spectroscopic techniques. The persuasive affinity of these molecules towards EGFR inhibition (IC50 for QT=45nM) was established and validated from specific kinase assay including the cell viability spectrophotometric assay (QT=12nM). Drug likeliness property were also considered by analysing, the ADME of these molecules by using scintigraphic techniques. The result showed antitumour activity of QT (4.17 tumour/muscle at 4h). Further photo physical properties were also analysed to see in vitro HSA binding to QT.
Collapse
Affiliation(s)
- Swati Aggarwal
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Delhi 110054, India; Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Deepa Sinha
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Delhi 110054, India; Department of Applied Sciences, HMRITM (Affiliated to GGSIP University), Delhi 110036, India
| | - Anjani Kumar Tiwari
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Delhi 110054, India.
| | - Pooja Pooja
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Delhi 110054, India; Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Ankur Kaul
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Delhi 110054, India
| | - Gurmeet Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Anil Kumar Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Delhi 110054, India.
| |
Collapse
|
5
|
Genistein-induced G2/M cell cycle arrest of human intestinal colon cancer Caco-2 cells is associated with Cyclin B1 and Chk2 down-regulation. Cytotechnology 2013; 65:973-8. [PMID: 23794041 DOI: 10.1007/s10616-013-9592-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 05/31/2013] [Indexed: 12/17/2022] Open
Abstract
Genistein is an isoflavonic phyto-oestrogen contained in soya beans. It is thought to display anti-cancer effects. This study was designed to investigate its effect on human intestinal colon cancer Caco-2 cells. MTT assay, flow cytometric analysis and western blotting were used to investigate the effect of genistein on cell proliferation, cell cycle progression and protein alterations of selected cell cycle-related proteins in Caco-2 cells. Our results showed that genistein and daidzein significantly suppressed cell proliferation. Genistein treatment was demonstrated to modulate cell cycle distribution through accumulation of cells at G2/M phase, with a significant decreasing effect of Cyclin B1 and Serine/threonine-protein kinase 2 (Chk2) proteins expression. However, daidzein did not alter the cell cycle progression in Caco-2 cells. All these observation strongly indicate that genistein has anti-proliferative effect in human intestinal colon cancer Caco-2 cells through the down-regulation of cell cycle check point proteins, Cyclin B1 and Chk2.
Collapse
|
6
|
Hirata M, Kanai Y, Naka S, Yoshimoto M, Kagawa S, Matsumuro K, Katsuma H, Yamaguchi H, Magata Y, Ohmomo Y. A useful EGFR-TK ligand for tumor diagnosis with SPECT: development of radioiodinated 6-(3-morpholinopropoxy)-7-ethoxy-4-(3'-iodophenoxy)quinazoline. Ann Nucl Med 2013; 27:431-43. [PMID: 23494210 PMCID: PMC3672506 DOI: 10.1007/s12149-013-0703-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/06/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Epidermal growth factor receptor tyrosine kinase (EGFR-TK) represents an attractive target for tumor diagnosis agents. Previously, radioiodinated 4-(3-iodophenoxy)-6,7-diethoxyquinazoline (PHY) was reported to possess good characteristics as a tumor imaging agent. We have explored the feasibility of developing tumor diagnosis ligands superior to radioiodinated PHY. METHODS New phenoxyquinazoline derivatives were designed with various side chains introduced to the 6th position of PHY. The IC50 values of the new derivatives to interrupt EGFR-TK phosphorylation were evaluated and compared to well-known EGFR-TK inhibitors. Tumor uptake studies of the new (125)I-labeled derivatives were conducted with A431 tumor-bearing mice. Selectivity and binding characteristics were analyzed by in vitro blocking studies and a binding assay. Furthermore, SPECT/CT scans were performed using A431 tumor-bearing mice. RESULTS Six quinazoline derivatives were designed and synthesized, and among these, 6a-d were found to have relatively high EGFR-TK inhibitory potency. In tumor uptake studies, [(125)I]6a ([(125)I]PYK) was found to have the highest tumor uptake and longest retention in tumors. In contrast, [(125)I]PYK was rapidly cleared from peripheral tissues, resulting in a high tumor-to-tissue ratio 24 h after injection. Moreover, the EGFR-TK selectivity of [(125)I]PYK was confirmed by pretreatment experiments with specific EGFR-TK inhibitors. Furthermore, [(125)I]PYK provided clear SPECT images of tumors. CONCLUSIONS Radioiodinated PYK, one of the newly synthesized quinazoline derivatives, was found to be a desirable ligand for EGFR-TK SPECT imaging. [(125)I]PYK showed high tumor accumulation and selective EGFR-TK binding and also succeeded in delivering high contrast imaging of tumors. These favorable characteristics of [(125)I]PYK suggest that the (123)I-labeled counterpart, [(123)I]PYK, would have great potential for diagnostic SPECT tumor imaging.
Collapse
Affiliation(s)
- Masahiko Hirata
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
| | - Yasukazu Kanai
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Osaka Suita, 565-0871 Japan
| | - Sadahiro Naka
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Osaka Suita, 565-0871 Japan
| | - Mitsuyoshi Yoshimoto
- Functional Imaging Division, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Chiba Kashiwa, 277-8577 Japan
| | - Shinya Kagawa
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
- Research Institute, Shiga Medical Center 5-4-30 Moriyama, Shiga Moriyama, 524-8524 Japan
| | - Keiji Matsumuro
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
| | - Hideyuki Katsuma
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
| | - Hiroshi Yamaguchi
- Department of Molecular Imaging, Applied Medical Photonics Laboratory, Medical Photonics Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192 Japan
| | - Yasuhiro Magata
- Department of Molecular Imaging, Applied Medical Photonics Laboratory, Medical Photonics Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192 Japan
| | - Yoshiro Ohmomo
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Osaka Takatsuki, 569-1094 Japan
| |
Collapse
|
7
|
Collins-Burow BM, Antoon JW, Frigo DE, Elliott S, Weldon CB, Boue SM, Beckman BS, Curiel TJ, Alam J, McLachlan JA, Burow ME. Antiestrogenic activity of flavonoid phytochemicals mediated via the c-Jun N-terminal protein kinase pathway. Cell-type specific regulation of estrogen receptor alpha. J Steroid Biochem Mol Biol 2012; 132:186-93. [PMID: 22634477 PMCID: PMC4083692 DOI: 10.1016/j.jsbmb.2012.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 05/12/2012] [Accepted: 05/14/2012] [Indexed: 12/14/2022]
Abstract
Flavonoid phytochemicals act as both agonists and antagonists of the human estrogen receptors (ERs). While a number of these compounds act by directly binding to the ER, certain phytochemicals, such as the flavonoid compounds chalcone and flavone, elicit antagonistic effects on estrogen signaling independent of direct receptor binding. Here we demonstrate both chalcone and flavone function as cell type-specific selective ER modulators. In MCF-7 breast carcinoma cells chalcone and flavone suppress ERα activity through stimulation of the stress-activated members of the mitogen-activated protein kinase (MAPK) family: c-Jun N-terminal kinase (JNK)1 and JNK2. The use of dominant-negative mutants of JNK1 or JNK2 in stable transfected cells established that the antiestrogenic effects of chalcone and flavone required intact JNK signaling. We further show that constitutive activation of the JNK pathway partially suppresses estrogen (E2)-mediated gene expression in breast, but not endometrial carcinoma cells. Our results demonstrate a role for stress-activated MAPKs in the cell type-specific regulation of ERα function.
Collapse
Affiliation(s)
- Bridgette M. Collins-Burow
- Tulane University Medical Center, New Orleans, Louisiana 70112
- Center for Bioenvironmental Research at Tulane and Xavier Universities, New Orleans, Louisiana 70112
- Department of Medicine, Section of Hematology & Medical Oncology, New Orleans, Louisiana 70112
| | - James W. Antoon
- Tulane University Medical Center, New Orleans, Louisiana 70112
- Department of Medicine, Section of Hematology & Medical Oncology, New Orleans, Louisiana 70112
- Department of Pharmacology, New Orleans, Louisiana 70112
| | - Daniel E. Frigo
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Steven Elliott
- Tulane University Medical Center, New Orleans, Louisiana 70112
- Department of Medicine, Section of Hematology & Medical Oncology, New Orleans, Louisiana 70112
| | - Christopher B. Weldon
- Tulane University Medical Center, New Orleans, Louisiana 70112
- Department of Medicine, Section of Hematology & Medical Oncology, New Orleans, Louisiana 70112
| | - Stephen M. Boue
- U. S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, New Orleans, LA 70179
| | - Barbara S. Beckman
- Center for Bioenvironmental Research at Tulane and Xavier Universities, New Orleans, Louisiana 70112
- Department of Medicine, Section of Hematology & Medical Oncology, New Orleans, Louisiana 70112
| | - Tyler J. Curiel
- Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio
| | - Jawed Alam
- Alton Ochsner Medical Foundation, Department of Molecular Genetics, New Orleans, Louisiana 70121
| | - John A. McLachlan
- Tulane University Medical Center, New Orleans, Louisiana 70112
- Center for Bioenvironmental Research at Tulane and Xavier Universities, New Orleans, Louisiana 70112
| | - Matthew E. Burow
- Tulane University Medical Center, New Orleans, Louisiana 70112
- Center for Bioenvironmental Research at Tulane and Xavier Universities, New Orleans, Louisiana 70112
- Department of Medicine, Section of Hematology & Medical Oncology, New Orleans, Louisiana 70112
- To whom correspondence and requests for reprints should be addressed: Matthew E. Burow, Tulane University Health Sciences Center, Department of Medicine, Section of Hematology & Medical Oncology, 1430 Tulane Ave. SL-78, New Orleans, LA 70112, Phone: 504-988-6688, Fax: 504-988-5483,
| |
Collapse
|
8
|
Synthesis and evaluation of radioiodinated phenoxyquinazoline and benzylaminoquinazoline derivatives as new EGF receptor tyrosine kinase imaging ligands for tumor diagnosis using SPECT. Ann Nucl Med 2012; 26:381-9. [PMID: 22354330 DOI: 10.1007/s12149-012-0583-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/07/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Epidermal growth factor receptor tyrosine kinase (EGFR-TK) represents an attractive target for tumor diagnosis agents. Previously, the radioiodinated 4-(3-iodoanilino)-6,7-diethoxyquinazoline ([(125)I]m-IPQ) has been reported to possess good characteristics as a tumor imaging agent; however, it was also found to have low in vivo stability. To improve the in vivo stability, m-IPQ derivatives, 4-(3-iodophenoxy)-6,7-diethoxyquinazoline (PHY) and 4-(3-iodobenzylamino)-6,7-diethoxyquinazoline (BAY) were designed and synthesized, and the biological studies of [(125)I]PHY and [(125)I]BAY were performed to evaluate these new ligands as in vivo tumor diagnosis agents. METHODS PHY and BAY were synthesized according to previous reports. The EGFR-TK inhibitory potency of these new compounds was measured and compared to other EGFR-TK inhibitors. Radiolabeled [(125)I]PHY and [(125)I]BAY were synthesized by an iododestannylation reaction. Biodistribution studies of these radioligands were conducted in normal mice and tumor-bearing mice. Furthermore, selectivity and binding characteristics of [(125)I]PHY were analyzed by in vitro blocking studies and a binding assay. RESULTS The new derivatives were found to have high inhibitory potency against EGFR-TK (PHY: IC50 = 12.7 ± 7.2 nM, BAY: IC50 = 51.0 ± 8.9 nM). [(125)I]PHY and [(125)I]BAY were conveniently synthesized from tributylstannyl precursors. In in vivo biodistribution studies, [(125)I]PHY and [(125)I]BAY were observed to have lower uptake in the stomach, an indication of deiodination, than [(125)I]m-IPQ. Moreover, [(125)I]PHY showed high uptake and prolonged retention in tumors and low accumulation in blood and muscle tissue resulting in a good tumor-to-blood ratio (0.94-1.50) and tumor-to-muscle ratio (1.02-1.95). The EGFR-TK selectivity of [(125)I]PHY was confirmed by pretreatment experiments with specific EGFR-TK inhibitors. CONCLUSION New radioiodinated quinazoline derivatives were synthesized, which were found to have improved in vivo stability. In particular, [(125)I]PHY showed higher tumor accumulation than the other ligands which was indicative of selective binding to EGFR-TK. These desirable characteristics for [(125)I]PHY suggest that the (123)I-labeled counterpart, [(123)I]PHY, could be a possible candidate for cancer diagnosis radiopharmaceutical.
Collapse
|
9
|
Evaluation of radioiodinated quinazoline derivative as a new ligand for EGF receptor tyrosine kinase activity using SPECT. Ann Nucl Med 2010; 25:117-24. [DOI: 10.1007/s12149-010-0432-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 09/22/2010] [Indexed: 11/26/2022]
|
10
|
Yang SNY, Osman N, Burch ML, Little PJ. Factors affecting proteoglycan synthesis and structure that modify the interaction with lipoproteins. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/clp.09.37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
11
|
Cao C, Li S, Dai X, Chen Y, Feng Z, Zhao Y, Wu J. Genistein inhibits proliferation and functions of hypertrophic scar fibroblasts. Burns 2008; 35:89-97. [PMID: 18976864 DOI: 10.1016/j.burns.2008.03.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 03/10/2008] [Indexed: 11/25/2022]
Abstract
Hypertrophic scarring is abnormal proliferation of dermal fibroblasts and excessive deposition of extracellular matrix. To date, despite many studies, treatments have not been satisfactory. Genistein, a potent, specific inhibitor of tyrosine protein kinases (TPKs), has been proved to inhibit many kinds of tumour and some fibrotic diseases. The purpose of this study was to investigate the effects of genistein on the proliferation and functions of hypertrophic scar fibroblasts (HSFBs) and the mechanism by which genistein inhibits TPK signal transduction. The first effect was observed by methyl-thiazol-diphenyl-tetrazolium assay and the second by [gamma-(32)p] adenosine triphosphate incorporation assay. The results demonstrated that genistein inhibits the proliferation and function of HSFBs and changes the TPK signal transduction pathway, which can provide an experimental basis for treating HS with genistein.
Collapse
Affiliation(s)
- Chuan Cao
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Rayalam S, Della-Fera MA, Yang JY, Park HJ, Ambati S, Baile CA. Resveratrol potentiates genistein's antiadipogenic and proapoptotic effects in 3T3-L1 adipocytes. J Nutr 2007; 137:2668-73. [PMID: 18029481 DOI: 10.1093/jn/137.12.2668] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Genistein (G) and resveratrol (R) individually inhibit adipogenesis in 3T3-L1 adipocytes and induce apoptosis in cancer cells. We investigated whether the combination of G and R resulted in enhanced effects on adipogenesis, lipolysis, and apoptosis in 3T3-L1 cells. Preadipocytes and mature adipocytes were treated with G and R individually at 50 and 100 micromol/L (G100; R100) and in combination. Both in preadipocytes and mature adipocytes, G and R individually decreased cell viability dose-dependently, but G100 + R100 further decreased viability by 59 +/- 0.97% (P < 0.001) and 69.7 +/- 1.2% (P < 0.001) after 48 h compared with G100 and R100, respectively. G100 + R100 induced apoptosis 242 +/- 8.7% (P < 0.001) more than the control after 48 h, whereas G100 and R100 individually increased apoptosis only 46 +/- 9.2 and 46 +/- 7.9%, respectively. G and R did not modulate mitogen-activated protein kinase expression by themselves, but G100 + R100 increased Jun-N-terminal kinase phosphorylation by 38.8 +/- 4.4% (P < 0.001) and decreased extracellular signal-regulating kinase phosphorylation by 48 +/- 3.4% (P < 0.001). Individually, G and R at 25 micromol/L (G25; R25) decreased lipid accumulation by 30 +/- 1.7% and 20.07 +/- 4.27%, respectively (P < 0.001). However, G25 + R25 decreased lipid accumulation by 77.9 +/- 3.4% (P < 0.001). Lipolysis assay revealed that neither G25 nor R25 induced lipolysis, whereas G25 + R25 significantly increased lipolysis by 25.5 +/- 4.6%. The adipocyte-specific proteins PPARgamma and CCAAT/enhancer binding protein-alpha were downregulated after treatment with G + R, but no effect was observed with individual compounds. These results indicate that G and R in combination produce enhanced effects on inhibiting adipogenesis, inducing apoptosis, and promoting lipolysis in 3T3-L1 adipocytes. Thus, the combination of G and R is more potent in exerting antiobesity effects than the individual compounds.
Collapse
Affiliation(s)
- Srujana Rayalam
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602-2771, USA
| | | | | | | | | | | |
Collapse
|
13
|
Gopalakrishnan A, Xu CJ, Nair SS, Chen C, Hebbar V, Kong ANT. Modulation of activator protein-1 (AP-1) and MAPK pathway by flavonoids in human prostate cancer PC3 cells. Arch Pharm Res 2006; 29:633-44. [PMID: 16964758 DOI: 10.1007/bf02968247] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In last couple of decades the use of natural compounds like flavonoids as chemopreventive agents has gained much attention. Our current study focuses on identifying chemopreventive flavonoids and their mechanism of action on human prostate cancer cells. Human prostate cancer cells (PC3), stably transfected with activator protein 1 (AP-1) luciferase reporter gene were treated with four main classes of flavonoids namely flavonols, flavones, flavonones, and isoflavones. The maximum AP-1 luciferase induction of about 3 fold over control was observed with 20 microM concentrations of quercetin, chrysin and genistein and 50 microM concentration of kaempferol. At higher concentrations, most of the flavonoids demonstrated inhibition of AP-1 activity. The MTS assay for cell viability at 24 h showed that even at a very high concentration (500 microM), cell death was minimal for most of the flavonoids. To determine the role of MAPK pathway in the induction of AP-1 by flavonoids, Western blot of phospho MAPK proteins was performed. Four out of the eight flavonoids namely kaempferol, apigenin, genistein and naringenin were used for the Western Blot analysis. Induction of phospho-JNK and phospho-ERK activity was observed after two hour incubation of PC3-AP1 cells with flavonoids. However no induction of phospho-p38 activity was observed. Furthermore, pretreating the cells with specific inhibitors of JNK reduced the AP-1 luciferase activity that was induced by genistein while pretreatment with MEK inhibitor reduced the AP-1 luciferase activity induced by kaempferol. The pharmacological inhibitors did not affect the AP-1 luciferase activity induced by apigenin and naringenin. These results suggest the possible involvement of JNK pathway in genistein induced AP-1 activity while the ERK pathway seems to play an important role in kaempferol induced AP-1 activity.
Collapse
Affiliation(s)
- Avanthika Gopalakrishnan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway NJ-08854, USA
| | | | | | | | | | | |
Collapse
|
14
|
Frey RS, Singletary KW. Genistein activates p38 mitogen-activated protein kinase, inactivates ERK1/ERK2 and decreases Cdc25C expression in immortalized human mammary epithelial cells. J Nutr 2003; 133:226-31. [PMID: 12514295 DOI: 10.1093/jn/133.1.226] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Genistein (4',5,7-trihydroxyisoflavone) is an isoflavonoid present in soybeans that exhibits anticarcinogenic effects in breast, colon and prostate cancer cells. We recently reported that genistein treatment of the immortalized but nonmalignant human mammary epithelial cell line MCF-10F resulted in growth arrest of MCF-10F cells in the G2 phase of the cell cycle, a large induction of the Tyr15 phosphorylation of Cdc2 (along with decreased activity of Cdc2), increased expression of p21(waf/cip1) and decreased expression of the cell cycle phosphatase Cdc25C. In the present study of MCF-10F cells, genistein rapidly and significantly activated p38, inactivated ERK1/ERK2 and had no effect on SAPK/JNK activity. We also showed that p38 is involved in genistein-induced changes in Cdc2 phosphorylation and that the downregulation of Cdc25C expression by genistein is through the p38 pathway. Finally, we provided evidence that the p38 pathway is involved in genistein-inhibited cell proliferation. These data suggest an important interplay between the p38 pathway and G2 cell cycle checkpoint control and provide insights into possible mechanisms whereby this isoflavone may inhibit early events in mammary carcinogenesis.
Collapse
Affiliation(s)
- Randall S Frey
- University of Illinois, Department of Food Science and Human Nutrition, Urbana 61801, USA
| | | |
Collapse
|
15
|
Lee YJ, Aroor AR, Shukla SD. Temporal activation of p42/44 mitogen-activated protein kinase and c-Jun N-terminal kinase by acetaldehyde in rat hepatocytes and its loss after chronic ethanol exposure. J Pharmacol Exp Ther 2002; 301:908-14. [PMID: 12023518 DOI: 10.1124/jpet.301.3.908] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Several cell-damaging effects of ethanol are due to its major metabolite acetaldehyde but its mechanisms are not known. We have studied the effect of acetaldehyde on p42/44 mitogen-activated protein kinase (MAPK) and p46/p54 c-Jun N-terminal kinase (JNK 1/2) in rat hepatocytes. Acetaldehyde caused peak activation of p42/44 MAPK at 10 min followed by JNK activation at 1 h. These responses were acetaldehyde dose-dependent (0.2-5 mM). There was a consistently higher activation of p46 JNK than p54 JNK. Ethanol also activated both p42/44 MAPK and p46/p54 JNK. The activation of JNK by ethanol, however, was not significantly affected by treatment of hepatocytes with 4-methylpyrazole, an alcohol dehydrogenase inhibitor. Cells treated with 200 mM ethanol for 1 h accumulated 0.35 +/- 0.02 mM acetaldehyde, but the magnitude of JNK activation was greater than that expected with 0.35 mM acetaldehyde. Thus, ethanol-activated JNK may be both acetaldehyde-dependent and -independent. The activation of JNK by ethanol or acetaldehyde was insensitive to the treatment of hepatocytes with genistein (tyrosine kinase inhibitor) and 2-[1-(3-dimethylaminopropyl)-1H-indol-3-yl]-3-(1H-indol-3-yl)maleimide (GF109203X) (protein kinase C inhibitor). Remarkably, in contrast to the above-mentioned effects on normal hepatocytes, acetaldehyde was unable to increase JNK activity in hepatocytes isolated from rats chronically fed ethanol for 6 weeks and indicated a loss of this acetaldehyde response. Thus, temporal activation of the p42/44 MAPK and p46/p54 JNK, the greater activation of p46 JNK than p54 JNK, and loss of JNK activation after chronic ethanol exposure indicate that these kinases are differentially affected by ethanol metabolite acetaldehyde.
Collapse
Affiliation(s)
- Youn J Lee
- Department of Pharmacology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | | | | |
Collapse
|
16
|
Dampier K, Hudson EA, Howells LM, Manson MM, Walker RA, Gescher A. Differences between human breast cell lines in susceptibility towards growth inhibition by genistein. Br J Cancer 2001; 85:618-24. [PMID: 11506505 PMCID: PMC2364092 DOI: 10.1054/bjoc.2001.1980] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2000] [Revised: 05/22/2001] [Accepted: 05/24/2001] [Indexed: 01/08/2023] Open
Abstract
Genistein is thought to contribute to the putative breast cancer preventive activity of soya. The mechanisms by which it arrests the growth of breast cells are incompletely understood. In order to explore generic features of the modulation of human breast cell growth by genistein, its effects on cell lines MCF-7, ZR-75.1, T47-D, MDA-MB 468, MDA-MB 231 and HBL 100 were compared. Genistein at 1 microM stimulated growth only in MCF-7 cells. At 10 microM it arrested the growth of all 6 cell types, however that of T47-D and HBL 100 cells only in medium with reduced (2%) fetal calf serum. Genistein induced apoptosis in only MDA-MB 468 cells. It arrested cells in the G2 stage of the cell cycle in all cell lines except ZR-75.1. Cells differed in their susceptibility towards inhibition by genistein of phorbol ester-induced proto-oncogene c-fos levels, transcription factor activator protein-1 (AP-1) activity and extracellular signal-regulated kinase (ERK) activity. Genistein augmented anisomycin-induced levels of proto-oncogene c-jun in ZR 75.1 and MCF-7 cells. The results suggest that induction of apoptosis, G2 cell cycle arrest and inhibition of c-fos expression, AP-1 transactivation and ERK phosphorylation may contribute to the growth-inhibitory effect of genistein in some breast cell types, but none of these effects of genistein constitutes a generic mode of growth-arresting action.
Collapse
Affiliation(s)
- K Dampier
- MRC Toxicology Unit, University of Leicester, PO Box 138, Leicester LE1 9HN, UK
| | | | | | | | | | | |
Collapse
|