1
|
Abusharkh KAN, Comert Onder F, Çınar V, Onder A, Sıkık M, Hamurcu Z, Ozpolat B, Ay M. Novel benzothiazole/benzothiazole thiazolidine-2,4-dione derivatives as potential FOXM1 inhibitors: In silico, synthesis, and in vitro studies. Arch Pharm (Weinheim) 2024:e2400504. [PMID: 39318080 DOI: 10.1002/ardp.202400504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024]
Abstract
The oncogenic transcription factor FOXM1 overexpressed in breast and other solid cancers, is a key driver of tumor growth and progression through complex interactions, making it an attractive molecular target for the development of targeted therapies. Despite the availability of small-molecule inhibitors, their limited specificity, potency, and efficacy hinder clinical translation. To identify effective FOXM1 inhibitors, we synthesized novel benzothiazole derivatives (KC10-KC13) and benzothiazole hybrids with thiazolidine-2,4-dione (KC21-KC36). These compounds were evaluated for FOXM1 inhibition. Molecular docking and molecular dynamics simulation analysis revealed their binding patterns and affinities for the FOXM1-DNA binding domain. The interactions with key amino acids such as Asn283, His287, and Arg286, crucial for FOXM1 inhibition, have been determined with the synthesized compounds. Additionally, the molecular modeling study indicated that KC12, KC21, and KC30 aligned structurally and interacted similarly to the reference compound FDI-6. In vitro studies with the MDA-MB-231 breast cancer cell line demonstrated that KC12, KC21, and KC30 significantly inhibited FOXM1, showing greater potency than FDI-6, with IC50 values of 6.13, 10.77, and 12.86 µM, respectively, versus 20.79 µM for FDI-6. Our findings suggest that KC12, KC21, and KC30 exhibit strong activity as FOXM1 inhibitors and may be suitable for in vivo animal studies.
Collapse
Affiliation(s)
- Khaled A N Abusharkh
- Department of Chemistry, School of Graduate Studies, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
- Department of Chemistry, Natural Products and Drug Research Laboratory, Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
- Department of Chemistry and Chemical Technology, Faculty of Science and Technology, Al-Quds University, East Jerusalem, Palestine
| | - Ferah Comert Onder
- Department of Medical Biology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Venhar Çınar
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Alper Onder
- Department of Chemistry, Natural Products and Drug Research Laboratory, Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Merve Sıkık
- Department of Medical System Biology, School of Graduate Studies, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Zuhal Hamurcu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Mehmet Ay
- Department of Chemistry, Natural Products and Drug Research Laboratory, Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| |
Collapse
|
2
|
Maji S, Debnath B, Panda S, Manna T, Maity A, Dayaramani R, Nath R, Khan SA, Akhtar MJ. Anticancer Potential of the S-Heterocyclic Ring Containing Drugs and its Bioactivation to Reactive Metabolites. Chem Biodivers 2024; 21:e202400473. [PMID: 38723201 DOI: 10.1002/cbdv.202400473] [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: 02/23/2024] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Sulfur-containing heterocyclic derivatives have been disclosed for binding with a wide range of cancer-specific protein targets. Various interesting derivatives of sulfur-containing heterocyclics such as benzothiazole, thiazole, thiophene, thiazolidinedione, benzothiophene, and phenothiazine, etc have been shown to inhibit diverse signaling pathways implicated in cancer. Significant progress has also been made in molecular targeted therapy against specific enzymes such as kinase receptors due to potential binding interactions inside the ATP pocket. Sulfur-containing heterocyclic ring metal complexes i. e., benzothiazole, thiazole, thiophene, benzothiophene and phenothiazines are among the most promising active anticancer compounds. However, sulfur heteroaromatic rings, particularly thiophene, are of high structural alert due to their metabolism to reactive metabolites. The mere presence of a structural alert itself does not determine compound toxicity therefore, this review focuses on some specific findings that shed light on factors influencing the toxicity. In the current review, synthetic strategies of introducing the sulfur core ring in the synthesized derivatives are discussed with their structure-activity relationships to enhance our understanding of toxicity mechanisms and develop safer therapeutic options. The sulfur-containing marketed anticancer drugs included in this review direct the synthesis of novel compounds and will help in the development of potent, safer sulfur-based anticancer drugs in near future.
Collapse
Affiliation(s)
- Sumit Maji
- Department of Pharmacy, Bharat Technology, Uluberia-711316, Howrah, West Bengal, India
| | - Biplab Debnath
- Department of Pharmacy, Bharat Technology, Uluberia-711316, Howrah, West Bengal, India
| | - Shambo Panda
- Department of Pharmacy, Bharat Technology, Uluberia-711316, Howrah, West Bengal, India
| | - Tanusree Manna
- Department of Pharmacy, Bharat Technology, Uluberia-711316, Howrah, West Bengal, India
| | - Arindam Maity
- JIS University, Agarpara Campus, Kolkata-81, Nilgunj Road, Agarpara, Kolkata-700109, India
| | - Richa Dayaramani
- Silver Oak Institute of Pharmacy and Research, Silver Oak University, Ahmedabad, India
| | - Rajarshi Nath
- Department of Pharmacy, Bharat Technology, Uluberia-711316, Howrah, West Bengal, India
- JIS University, Agarpara Campus, Kolkata-81, Nilgunj Road, Agarpara, Kolkata-700109, India
| | - Shah Alam Khan
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PO 620, PC 130, Azaiba, Bousher, Muscat, Sultanate of Oman
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PO 620, PC 130, Azaiba, Bousher, Muscat, Sultanate of Oman
| |
Collapse
|
3
|
Muller C, Lacroix-Malgras V, Kluza J, Laine W, Güler Y, Bost F, Boisbrun M, Mazerbourg S, Flament S. The troglitazone derivative EP13 disrupts energy metabolism through respiratory chain complex I inhibition in breast cancer cells and potentiates the antiproliferative effect of glycolysis inhibitors. Cancer Cell Int 2024; 24:132. [PMID: 38594745 PMCID: PMC11005237 DOI: 10.1186/s12935-024-03319-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The metabolism of cancer cells generally differs from that of normal cells. Indeed, most cancer cells have a high rate of glycolysis, even at normal oxygen concentrations. These metabolic properties can potentially be exploited for therapeutic intervention. In this context, we have developed troglitazone derivatives to treat hormone-sensitive and triple-negative breast cancers, which currently lack therapeutic targets, have an aggressive phenotype, and often have a worse prognosis than other subtypes. Here, we studied the metabolic impact of the EP13 compound, a desulfured derivative of Δ2-troglitazone that we synthetized and is more potent than its parent compounds. METHODS EP13 was tested on two triple-negative breast cancer cell lines, MDA-MB-231 and Hs578T, and on the luminal cell line MCF-7. The oxygen consumption rate (OCR) of the treated cell lines, Hs578T mammospheres and isolated mitochondria was measured using the XFe24 Seahorse analyser. ROS production was quantified using the MitoSOX fluorescent probe. Glycolytic activity was evaluated through measurement of the extracellular acidification rate (ECAR), glucose consumption and lactate production in extracellular medium. The synergistic effect of EP13 with glycolysis inhibitors (oxamate and 2-deoxyglucose) on cell cytotoxicity was established using the Chou-Talalay method. RESULTS After exposure to EP13, we observed a decrease in the mitochondrial oxygen consumption rate in MCF7, MDA-MB-231 and Hs578T cells. EP13 also modified the maximal OCR of Hs578T spheroids. EP13 reduced the OCR through inhibition of respiratory chain complex I. After 24 h, ATP levels in EP13-treated cells were not altered compared with those in untreated cells, suggesting compensation by glycolysis activity, as shown by the increase in ECAR, the glucose consumption and lactate production. Finally, we performed co-treatments with EP13 and glycolysis inhibitors (oxamate and 2-DG) and observed that EP13 potentiated their cytotoxic effects. CONCLUSION This study demonstrates that EP13 inhibits OXPHOS in breast cancer cells and potentiates the effect of glycolysis inhibitors.
Collapse
Affiliation(s)
- Claire Muller
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France
| | | | - Jérôme Kluza
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pour la Recherche Sur le Cancer de Lille, UMR 9020 - UMR-S 1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - William Laine
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pour la Recherche Sur le Cancer de Lille, UMR 9020 - UMR-S 1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Yonca Güler
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France
| | - Frédéric Bost
- Inserm U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire, Team Cancer Metabolism, Environment, F-06200, Nice, France
| | | | - Sabine Mazerbourg
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France.
- CRAN, UMR 7039, Faculté des Sciences et Technologies, BP 70239, 54506, Vandœuvre-lès-Nancy, France.
| | | |
Collapse
|
4
|
Modh DH, Kulkarni VM. Anticancer Drug Discovery By Structure-Based Repositioning Approach. Mini Rev Med Chem 2024; 24:60-91. [PMID: 37165589 DOI: 10.2174/1389557523666230509123036] [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: 12/04/2022] [Revised: 03/07/2023] [Accepted: 03/28/2023] [Indexed: 05/12/2023]
Abstract
Despite the tremendous progress that has occurred in recent years in cell biology and oncology, in chemical, physical and computer sciences, the disease cancer has continued as the major cause of death globally. Research organizations, academic institutions and pharmaceutical companies invest huge amounts of money in the discovery and development of new anticancer drugs. Though much effort is continuing and whatever available approaches are being attempted, the success of bringing one effective drug into the market has been uncertain. To overcome problems associated with drug discovery, several approaches are being attempted. One such approach has been the use of known, approved and marketed drugs to screen these for new indications, which have gained considerable interest. This approach is known in different terms as "drug repositioning or drug repurposing." Drug repositioning refers to the structure modification of the active molecule by synthesis, in vitro/ in vivo screening and in silico computational applications where macromolecular structure-based drug design (SBDD) is employed. In this perspective, we aimed to focus on the application of repositioning or repurposing of essential drug moieties present in drugs that are already used for the treatment of some diseases such as diabetes, human immunodeficiency virus (HIV) infection and inflammation as anticancer agents. This review thus covers the available literature where molecular modeling of drugs/enzyme inhibitors through SBDD is reported for antidiabetics, anti-HIV and inflammatory diseases, which are structurally modified and screened for anticancer activity using respective cell lines.
Collapse
Affiliation(s)
- Dharti H Modh
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Erandwane, Pune, 411038, Maharashtra, India
| | - Vithal M Kulkarni
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Erandwane, Pune, 411038, Maharashtra, India
| |
Collapse
|
5
|
Steiniger KA, Lambert TH. Olefination of carbonyls with alkenes enabled by electrophotocatalytic generation of distonic radical cations. SCIENCE ADVANCES 2023; 9:eadg3026. [PMID: 37058559 PMCID: PMC10104462 DOI: 10.1126/sciadv.adg3026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/16/2023] [Indexed: 05/29/2023]
Abstract
The conversion of carbonyls to olefins is a transformation of great importance for complex molecule synthesis. Standard methods use stoichiometric reagents that have poor atom economy and require strongly basic conditions, which limit their functional group compatibility. An ideal solution would be to catalytically olefinate carbonyls under nonbasic conditions using simple and widely available alkenes, yet no such broadly applicable reaction is known. Here, we demonstrate a tandem electrochemical/electrophotocatalytic reaction to olefinate aldehydes and ketones with a broad range of unactivated alkenes. This method involves the oxidation-induced denitrogenation of cyclic diazenes to form 1,3-distonic radical cations that rearrange to yield the olefin products. This olefination reaction is enabled by an electrophotocatalyst that inhibits back-electron transfer to the radical cation intermediate, thus allowing for the selective formation of olefin products. The method is compatible with a wide range of aldehydes, ketones, and alkene partners.
Collapse
|
6
|
Dupommier D, Boisbrun M, Monard G, Comoy C. Unexplored Vinylic-Substituted 5-Benzylidenethiazolidine-2,4-diones: Synthesis and DFT/NMR Stereochemical Assignment. J Org Chem 2023; 88:3724-3739. [PMID: 36847759 DOI: 10.1021/acs.joc.2c02996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
By exploring an efficient and versatile method for the 6-functionalization of its scaffold, we investigated the opening of a new chemical space around benzylidenethiazolidine-2,4-dione (BTZD). The 6-chloro- and 6-formyl BTZD obtained in two steps starting from 5-lithioTZD were selected as key intermediates and involved in a Pd-catalyzed cross-coupling or Wittig olefination. A variety of aryl, heteroaryl, or alkenyl substituents was successfully introduced on the vinylic position of BTZD, and particular attention was paid to elucidate the stereochemistry of the benzylidene derivatives by using a combined DFT/NMR study.
Collapse
Affiliation(s)
| | | | - Gerald Monard
- Université de Lorraine, CNRS, LPCT, Nancy F-54000, France
| | - Corinne Comoy
- Université de Lorraine, CNRS, L2CM, Nancy F-54000, France
| |
Collapse
|
7
|
Cortelazzo-Polisini E, Boisbrun M, Gansmüller AH, Comoy C. Photoisomerization of Arylidene Heterocycles: Toward the Formation of Fused Heterocyclic Quinolines. J Org Chem 2022; 87:9699-9713. [PMID: 35801862 DOI: 10.1021/acs.joc.2c00748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report herein the photoinduced isomerization of a series of arylidene heterocycles 1. The photoreaction mechanism was investigated by a combined UV-vis/photo-NMR spectroscopic study, and we showed that Ar-TZDs exhibit a positive P-type photochromism, which limits their isomerization efficiency. By exploring the solvatochromism in a series of solvents, the conditions favoring the conversion toward one or the other stereoisomer have been studied, in particular by choosing the appropriate wavelengths. Finally, the extension of this photoisomerization study was proposed with a convenient preparation of various fused heterocyclic quinolines in good overall yields.
Collapse
Affiliation(s)
| | | | | | - Corinne Comoy
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France
| |
Collapse
|
8
|
AB186 Inhibits Migration of Triple-Negative Breast Cancer Cells and Interacts with α-Tubulin. Int J Mol Sci 2022; 23:ijms23126859. [PMID: 35743305 PMCID: PMC9225035 DOI: 10.3390/ijms23126859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 01/27/2023] Open
Abstract
Breast cancer is one of the leading causes of cancer-related death among females worldwide. A major challenge is to develop innovative therapy in order to treat breast cancer subtypes resistant to current treatment. In the present study, we examined the effects of two Troglitazone derivatives Δ2-TGZ and AB186. Previous studies showed that both compounds induce apoptosis, nevertheless AB186 was a more potent agent. The kinetic of cellular events was investigated by real-time cell analysis system (RTCA) in MCF-7 (hormone dependent) and MDA-MB-231 (triple negative) breast cancer (TNBC) cells, followed by cell morphology analysis by immuno-localization. Both compounds induced a rapid modification of both impedance-based signals and cellular morphology. This process was associated with an inhibition of cell migration measured by wound healing and transwell assays in TNBC MDA-MB-231 and Hs578T cells. In order to identify cytoplasmic targets of AB186, we performed surface plasmon resonance (SPR) and pull-down analyses. Subsequently, 6 cytoskeleton components were identified as potential targets. We further validated α-tubulin as one of the direct targets of AB186. In conclusion, our results suggested that AB186 could be promising to develop novel therapeutic strategies to treat aggressive forms of breast cancer such as TNBC.
Collapse
|
9
|
Islam MM, Goertzen A, Singh PK, Saha R. Exploring the metabolic landscape of pancreatic ductal adenocarcinoma cells using genome-scale metabolic modeling. iScience 2022; 25:104483. [PMID: 35712079 PMCID: PMC9194136 DOI: 10.1016/j.isci.2022.104483] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/08/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major research focus because of its poor therapy response and dismal prognosis. PDAC cells adapt their metabolism to the surrounding environment, often relying on diverse nutrient sources. Because traditional experimental techniques appear exhaustive to find a viable therapeutic strategy, a highly curated and omics-informed PDAC genome-scale metabolic model was reconstructed using patient-specific transcriptomics data. From the model-predictions, several new metabolic functions were explored as potential therapeutic targets in addition to the known metabolic hallmarks of PDAC. Significant downregulation in the peroxisomal beta oxidation pathway, flux modulation in the carnitine shuttle system, and upregulation in the reactive oxygen species detoxification pathway reactions were observed. These unique metabolic traits of PDAC were correlated with potential drug combinations targeting genes with poor prognosis in PDAC. Overall, this study provides a better understanding of the metabolic vulnerabilities in PDAC and will lead to novel effective therapeutic strategies.
Collapse
Affiliation(s)
- Mohammad Mazharul Islam
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Andrea Goertzen
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Pankaj K. Singh
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Rajib Saha
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
10
|
Tilekar K, Shelke O, Upadhyay N, Lavecchia A, Ramaa CS. Current status and future prospects of molecular hybrids with thiazolidinedione (TZD) scaffold in anticancer drug discovery. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
11
|
Marc G, Stana A, Pîrnău A, Vlase L, Oniga S, Oniga O. Regioselectivity evaluation of the (Z)-5-(4-hydroxybenzylidene)-thiazolidine-2,4‑dione alkylation in alkaline environment. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
12
|
Alegaon SG, U V, Alagawadi KR, Kumar D, Kavalapure RS, Ranade SD, Priya A S, Jalalpure SS. Synthesis, molecular docking and ADME studies of thiazole-thiazolidinedione hybrids as antimicrobial agents. J Biomol Struct Dyn 2021; 40:6211-6227. [PMID: 33538239 DOI: 10.1080/07391102.2021.1880479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
New thiazole-thiazolidinedione hybrids (5a-k) were efficiently synthesized and evaluated for their in-vitro antimicrobial activity against four fungal and bacterial strains. The chemical structures of the compounds were elucidated by FTIR, 1H NMR, and 13C NMR spectral data. Most of the synthesized compounds were sensitive against gram positive, gram negative bacterial and fungal strains. Among the synthesized molecules, compounds 5h, and 5i exhibited promising inhibitory activity against all selected fungal strains and gram positive bacteria namely, Staphylococcus aureus, and Enterococcus faecalis. The molecular docking results predicted that the thiazole-thiazolidinedione derivatives bind to the active site protein ATP-binding pocket from E. coli, S. aureus and C. albicans with good interaction energy scores. Ct-DNA was used to evaluate the binding interactions of the selected compounds by means of absorption spectroscopy. To further characterize the drug-likeness and ADME properties were calculated using the Qikprop, the result of present study suggests that thiazole-thiazolidinedione hybrid could be an interesting approach for the design of new antimicrobial agents.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shankar G Alegaon
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Venkatasubramanian U
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - K R Alagawadi
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Dushyant Kumar
- National Institute of Traditional Medicine (NITM-ICMR), Belagavi, Karnataka India
| | - Rohini S Kavalapure
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Shriram D Ranade
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Soundarya Priya A
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sunil S Jalalpure
- Department of Pharmacognosy and Phytochemistry KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India.,Dr. Prabhakar Kore Basic Science Research Center, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| |
Collapse
|
13
|
Kulkarni PS, Karale SN, Khandebharad AU, Agrawal BR, Sarda SR. Design, Synthesis, and Biological Evaluation of Newer Arylidene Incorporated 4-Thiazolidinones Derivatives as Potential Antimicrobial Agents. Polycycl Aromat Compd 2020. [DOI: 10.1080/10406638.2020.1823861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
| | - Sanjay N Karale
- Department of Chemistry, Dr. B.A.M. University, Aurangabad, Maharashtra, India
| | | | | | - Swapnil R. Sarda
- Department of Chemistry, J. E. S. College, Jalna, Maharashtra, India
| |
Collapse
|
14
|
Qiu J, Yuan CM, Wen M, Li YN, Chen J, Jian JY, Huang LJ, Gu W, Li YM, Hao XJ. Design, synthesis, and cytotoxic activities of novel hybrids of parthenolide and thiazolidinedione via click chemistry. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2020; 22:425-433. [PMID: 31012734 DOI: 10.1080/10286020.2019.1597055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/14/2019] [Indexed: 06/09/2023]
Abstract
A series of novel parthenolide-thiazolidinedione hybrids have been synthesized via a click chemistry-mediated coupling between parthenolide and thiazolidinedione, and evaluated for their cytotoxic activities. The results indicated that all the hybrids showed moderate cytotoxic effects on human cancer cell lines, including human erythroleukemia cell line (HEL), prostate (PC3), and breast (MDA-MB-231) by MTT assay. In particular, compound VI-6 exhibited the best cytotoxic activities against the MDA-MB-231 cells with IC50 value of 2.07 µM, which was about eight times more active than that of the original compound (PTL). These interesting results might be used to develop novel lead scaffolds for potential anticancer agents.
Collapse
Affiliation(s)
- Jie Qiu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Chun-Mao Yuan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Min Wen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Ya-Nan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Juan Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Jun-You Jian
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Lie-Jun Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Wei Gu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Yan-Mei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Xiao-Jiang Hao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| |
Collapse
|
15
|
Dupommier D, Muller C, Comoy C, Mazerbourg S, Bordessa A, Piquard E, Pawlak M, Piquard F, Martin H, De Fays E, Grandemange S, Flament S, Boisbrun M. New desulfured troglitazone derivatives: Improved synthesis and biological evaluation. Eur J Med Chem 2020; 187:111939. [PMID: 31838327 DOI: 10.1016/j.ejmech.2019.111939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 02/07/2023]
Abstract
Breast cancer is a major medical threat which cannot be sufficiently addressed by current therapies because of spontaneous or acquired treatment resistance. Besides, triple-negative breast cancer (TNBC) tumors do not respond to targeted therapies, thus new therapeutic strategies are needed. In this context, we designed and prepared new desulfured troglitazone (TGZ)-derived molecules and evaluated them in vitro for their anti-proliferative activity, with a special focus on triple-negative breast cancer cell lines. Optimization of the synthetic strategies and deracemization of the lead compound were performed to give highly active compound 10 with low-micromolar potency. Further studies revealed that this compound triggers apoptosis rather than cell cycle arrest as observed with TGZ.
Collapse
Affiliation(s)
| | - Claire Muller
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France
| | - Corinne Comoy
- Université de Lorraine, CNRS, L2CM, F-54000, Nancy, France
| | | | | | - Eline Piquard
- Université de Lorraine, CNRS, L2CM, F-54000, Nancy, France
| | - Manon Pawlak
- Université de Lorraine, CNRS, L2CM, F-54000, Nancy, France
| | | | - Hélène Martin
- PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - Elia De Fays
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France
| | | | | | | |
Collapse
|
16
|
Kolluri PK, Gurrapu N, Edigi PK, N. J. P. S, Putta S, Singh SS. Design, synthesis of (
Z
)‐3‐benzyl‐5‐((1‐phenyl‐3‐(3‐((1‐ substituted phenyl‐1
H
‐1,2,3‐triazol‐4‐yl)methoxy)phenyl)‐1
H
‐pyrazol‐4‐yl)methylene)thiazolidine‐2,4‐dione analogues as potential cytotoxic agents. J Heterocycl Chem 2019. [DOI: 10.1002/jhet.3720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Prashanth Kumar Kolluri
- Department of Chemistry, University College of ScienceOsmania University Hyderabad Telangana 500 007 India
| | - Nirmala Gurrapu
- Department of Chemistry, University College of ScienceOsmania University Hyderabad Telangana 500 007 India
| | - Praveen Kumar Edigi
- Department of Chemistry, University College of ScienceOsmania University Hyderabad Telangana 500 007 India
| | - Subhashini N. J. P.
- Department of Chemistry, University College of ScienceOsmania University Hyderabad Telangana 500 007 India
| | - Shravani Putta
- Department of Biochemistry, University College of ScienceOsmania University Hyderabad Telangana 500 007 India
| | - Surya Satyanarayana Singh
- Department of Biochemistry, University College of ScienceOsmania University Hyderabad Telangana 500 007 India
| |
Collapse
|
17
|
Rodriguez-Duarte J, Dapueto R, Galliussi G, Turell L, Kamaid A, Khoo NKH, Schopfer FJ, Freeman BA, Escande C, Batthyány C, Ferrer-Sueta G, López GV. Electrophilic nitroalkene-tocopherol derivatives: synthesis, physicochemical characterization and evaluation of anti-inflammatory signaling responses. Sci Rep 2018; 8:12784. [PMID: 30143727 PMCID: PMC6109136 DOI: 10.1038/s41598-018-31218-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Inflammation plays a major role in the onset and development of chronic non-communicable diseases like obesity, cardiovascular diseases and cancer. Combined, these diseases represent the most common causes of death worldwide, thus development of novel pharmacological approaches is crucial. Electrophilic nitroalkenes derived from fatty acids are formed endogenously and exert anti-inflammatory actions by the modification of proteins involved in inflammation signaling cascades. We have developed novel nitroalkenes derived from α-tocopherol aiming to increase its salutary actions by adding anti-inflammatory properties to a well-known nutraceutical. We synthesized and characterized an α-tocopherol-nitroalkene (NATOH) and two hydrosoluble analogues derived from Trolox (NATxME and NATx0). We analyzed the kinetics of the Michael addition reaction of these compounds with thiols in micellar systems aiming to understand the effect of hydrophobic partition on the reactivity of nitroalkenes. We studied NATxME in vitro showing it exerts non-conventional anti-inflammatory responses by inducing Nrf2-Keap1-dependent gene expression and inhibiting the secretion of NF-κB dependent pro-inflammatory cytokines. NATxME was also effective in vivo, inhibiting neutrophil recruitment in a zebrafish model of inflammation. This work lays the foundation for the rational design of a new therapeutic strategy for the prevention and treatment of metabolic and inflammation-related diseases.
Collapse
Affiliation(s)
- Jorge Rodriguez-Duarte
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay.,INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Rosina Dapueto
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay.,INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Germán Galliussi
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, Montevideo, Uruguay.,INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Lucía Turell
- Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Andrés Kamaid
- INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Analytical Biochemistry and Proteomics Unit, Montevideo, Uruguay
| | - Nicholas K H Khoo
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carlos Escande
- INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, Montevideo, Uruguay. .,INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay. .,Analytical Biochemistry and Proteomics Unit, Montevideo, Uruguay.
| | - Gerardo Ferrer-Sueta
- Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay.
| | - Gloria V López
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, Montevideo, Uruguay. .,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay. .,INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.
| |
Collapse
|
18
|
Parmentier C, Hendriks DFG, Heyd B, Bachellier P, Ingelman-Sundberg M, Richert L. Inter-individual differences in the susceptibility of primary human hepatocytes towards drug-induced cholestasis are compound and time dependent. Toxicol Lett 2018; 295:187-194. [PMID: 29913214 DOI: 10.1016/j.toxlet.2018.06.1069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/07/2018] [Accepted: 06/14/2018] [Indexed: 01/01/2023]
Abstract
Cholestasis represents a major subtype of drug-induced liver injury and novel preclinical models for its prediction are needed. Here we used primary human hepatocytes (PHH) from different donors in 2D-sandwich (2D-sw) and/or 3D-spheroid cultures to study inter-individual differences in the response towards cholestatic hepatotoxins after short-term (48-72 hours) and long-term repeated exposures (14 days). The cholestatic liabilities of drugs were determined by comparing cell viability upon exposure to the highest non-cytotoxic drug concentration in the presence and absence of a non-cytotoxic concentrated bile acid mixture. In 2D-sw culture, cyclosporine A and amiodarone presented clear cholestatic liabilities in all four PHH donors tested, whereas differences in the susceptibility of the various PHH donors towards the cholestatic toxicity of bosentan, chlorpromazine and troglitazone were observed. In PHH from one donor, the cholestatic liabilities of chlorpromazine and troglitazone could only be detected after long-term repeated exposures when maintained in 3D-spheroid culture, but not after short-term exposures in either 2D-sw or 3D-spheroid culture, suggesting that cholestatic hepatotoxicity may require time to develop. In conclusion, inter-individual susceptibility exists towards drug-induced cholestasis, which depends on the compound as well as the exposure time.
Collapse
Affiliation(s)
| | - Delilah F G Hendriks
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Bruno Heyd
- Hôpital Jean Minjoz, 3 Boulevard Alexandre Fleming 25000 Besançon, France; Université de Bourgogne Franche-Comté, EA 4267 PEPITE, France
| | | | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Lysiane Richert
- KaLy-Cell, 20A rue du Général Leclerc, 67115 Plobsheim, France; Université de Bourgogne Franche-Comté, EA 4267 PEPITE, France.
| |
Collapse
|
19
|
Ingold M, Dapueto R, Victoria S, Galliusi G, Batthyàny C, Bollati-Fogolín M, Tejedor D, García-Tellado F, Padrón JM, Porcal W, López GV. A green multicomponent synthesis of tocopherol analogues with antiproliferative activities. Eur J Med Chem 2018; 143:1888-1902. [DOI: 10.1016/j.ejmech.2017.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 11/16/2022]
|
20
|
den Braver-Sewradj SP, den Braver MW, Baze A, Decorde J, Fonsi M, Bachellier P, Vermeulen NPE, Commandeur JNM, Richert L, Vos JC. Direct comparison of UDP-glucuronosyltransferase and cytochrome P450 activities in human liver microsomes, plated and suspended primary human hepatocytes from five liver donors. Eur J Pharm Sci 2017; 109:96-110. [PMID: 28778465 DOI: 10.1016/j.ejps.2017.07.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 11/26/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) and cytochrome P450s (CYPs) are the major enzymes involved in hepatic metabolism of drugs. Hepatic drug metabolism is commonly investigated using human liver microsomes (HLM) or primary human hepatocytes (PHH). We describe the development of a sensitive assay to phenotype activities of six major hepatic UGT isoforms (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9 and UGT2B7) in intact PHH by analysis of glucuronidation of selective probe substrates. The non-selective, general substrate 7-hydroxycoumarin was included for comparison. For each liver donor preparation (five donors) UGT activities in cryopreserved suspended and plated PHH were compared to HLM prepared from the same donors. Standard CYP reaction phenotyping of seven major isoforms was performed in parallel. For all donors, CYP- and UGT-isoforms activity profiles were comparable in PHH and HLM, indicating that reaction phenotyping with selective probe substrates in intact cells primarily reflects respective CYP or UGT activity. System-dependent effects on UGT and CYP isoform activity were still found. While UGT activity of UGT1A1 was equivalent in plated and suspended PHH, UGT1A3, UGT1A6 and UGT2B7 activity was higher in suspended PHH and UGT1A9 and UGT1A4 activity was higher in plated PHH. The well-known decrease in activity of most CYP isoforms in plated compared to suspended PHH was confirmed. Importantly, we found a significant loss in CYP2C19 and CYP2B6 in HLM, activity being lower than in intact cells. Taken together, these findings implicate that, dependent on the UGT or CYP isoforms involved in the metabolism of a given compound, the outcome of metabolic assays is strongly dependent on the choice of the in vitro system. The currently described UGT- and CYP- activity profiling method can be used as a standard assay in intact cells and can especially aid in reaction phenotyping of in vitro systems for which a limited number of cells are available.
Collapse
Affiliation(s)
- Shalenie P den Braver-Sewradj
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Michiel W den Braver
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Audrey Baze
- Kaly-Cell, 20A Rue du Général Leclerc, Plobsheim, France; UNISTRA, 4 Rue Blaise Pascal, Strasbourg, France
| | | | | | - Philippe Bachellier
- UNISTRA, 4 Rue Blaise Pascal, Strasbourg, France; Centre de Chirurgie Viscérale et de Transplantation, Hôpital de Hautepierre, 67098 Strasbourg, France
| | - Nico P E Vermeulen
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jan N M Commandeur
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Lysiane Richert
- Kaly-Cell, 20A Rue du Général Leclerc, Plobsheim, France; PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - J Chris Vos
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Dixit VA, Deshpande S. Advances in Computational Prediction of Regioselective and Isoform-Specific Drug Metabolism Catalyzed by CYP450s. ChemistrySelect 2016. [DOI: 10.1002/slct.201601051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Vaibhav A. Dixit
- Department of Pharmaceutical Chemistry; School of Pharmacy and Technology Management (SPTM), Shri Vile Parle Kelavani Mandal's (SVKM's) Narsee Monjee Institute of Management Studies (NMIMS), Mukesh Patel Technology Park, Babulde, Bank of Tapi River; Mumbai-Agra Road Shirpur, Dist. Dhule−425405 India
| | - Shirish Deshpande
- Department of Pharmaceutical Chemistry; School of Pharmacy and Technology Management (SPTM), Shri Vile Parle Kelavani Mandal's (SVKM's) Narsee Monjee Institute of Management Studies (NMIMS), Mukesh Patel Technology Park, Babulde, Bank of Tapi River; Mumbai-Agra Road Shirpur, Dist. Dhule−425405 India
| |
Collapse
|
22
|
Richert L, Baze A, Parmentier C, Gerets HHJ, Sison-Young R, Dorau M, Lovatt C, Czich A, Goldring C, Park BK, Juhila S, Foster AJ, Williams DP. Cytotoxicity evaluation using cryopreserved primary human hepatocytes in various culture formats. Toxicol Lett 2016; 258:207-215. [PMID: 27363785 DOI: 10.1016/j.toxlet.2016.06.1127] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 11/25/2022]
Abstract
Sixteen training compounds selected in the IMI MIP-DILI consortium, 12 drug-induced liver injury (DILI) positive compounds and 4 non-DILI compounds, were assessed in cryopreserved primary human hepatocytes. When a ten-fold safety margin threshold was applied, the non-DILI-compounds were correctly identified 2h following a single exposure to pooled human hepatocytes (n=13 donors) in suspension and 14-days following repeat dose exposure (3 treatments) to an established 3D-microtissue co-culture (3D-MT co-culture, n=1 donor) consisting of human hepatocytes co-cultured with non-parenchymal cells (NPC). In contrast, only 5/12 DILI-compounds were correctly identified 2h following a single exposure to pooled human hepatocytes in suspension. Exposure of the 2D-sandwich culture human hepatocyte monocultures (2D-sw) for 3days resulted in the correct identification of 11/12 DILI-positive compounds, whereas exposure of the human 3D-MT co-cultures for 14days resulted in identification of 9/12 DILI-compounds; in addition to ximelagatran (also not identified by 2D-sw monocultures, Sison-Young et al., 2016), the 3D-MT co-cultures failed to detect amiodarone and bosentan. The sensitivity of the 2D human hepatocytes co-cultured with NPC to ximelagatran was increased in the presence of lipopolysaccharide (LPS), but only at high concentrations, therefore preventing its classification as a DILI positive compound. In conclusion (1) despite suspension human hepatocytes having the greatest metabolic capacity in the short term, they are the least predictive of clinical DILI across the MIP-DILI test compounds, (2) longer exposure periods than 72h of human hepatocytes do not allow to increase DILI-prediction rate, (3) co-cultures of human hepatocytes with NPC, in the presence of LPS during the 72h exposure period allow the assessment of innate immune system involvement of a given drug.
Collapse
Affiliation(s)
- Lysiane Richert
- KaLy-Cell, 20A rue du Général Leclerc, 67115 Plobsheim, France; Université de Franche-Comté, EA 4267 Besançon, France.
| | - Audrey Baze
- KaLy-Cell, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| | | | - Helga H J Gerets
- UCB BioPharma SPRL, Non-Clinical Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium.
| | - Rowena Sison-Young
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, University of Liverpool, Liverpool L69 3GE, UK.
| | - Martina Dorau
- Sanofi-Aventis Deutschland GmbH, R&D DSAR Preclinical Safety, Industriepark Hoechst, D-65926 Frankfurt, Germany.
| | - Cerys Lovatt
- GlaxoSmithKline, Safety Assessment, Stevenage, Hertfordshire, UK.
| | - Andreas Czich
- Sanofi-Aventis Deutschland GmbH, R&D DSAR Preclinical Safety, Industriepark Hoechst, D-65926 Frankfurt, Germany.
| | - Christopher Goldring
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, University of Liverpool, Liverpool L69 3GE, UK.
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, University of Liverpool, Liverpool L69 3GE, UK.
| | - Satu Juhila
- Orion Corporation, R&D, In Vitro Biology, Orionintie 1A, P.O. Box 65, FI-02101 Espoo, Finland.
| | - Alison J Foster
- Translational Safety, Drug Safety & Metabolism, AstraZeneca, Cambridge Science Park, Cambridge, UK.
| | - Dominic P Williams
- Translational Safety, Drug Safety & Metabolism, AstraZeneca, Cambridge Science Park, Cambridge, UK.
| |
Collapse
|
23
|
Bisikirska B, Bansal M, Shen Y, Teruya-Feldstein J, Chaganti R, Califano A. Elucidation and Pharmacological Targeting of Novel Molecular Drivers of Follicular Lymphoma Progression. Cancer Res 2016; 76:664-74. [PMID: 26589882 PMCID: PMC4738055 DOI: 10.1158/0008-5472.can-15-0828] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022]
Abstract
Follicular lymphoma, the most common indolent subtype of non-Hodgkin lymphoma, is associated with a relatively long overall survival rate ranging from 6 to 10 years from the time of diagnosis. However, in 20% to 60% of follicular lymphoma patients, transformation to aggressive diffuse large B-cell lymphoma (DLBCL) reduces median survival to only 1.2 years. The specific functional and genetic determinants of follicular lymphoma transformation remain elusive, and genomic alterations underlying disease advancement have only been identified for a subset of cases. Therefore, to identify candidate drivers of follicular lymphoma transformation, we performed systematic analysis of a B-cell-specific regulatory model exhibiting follicular lymphoma transformation signatures using the Master Regulator Inference algorithm (MARINa). This analysis revealed FOXM1, TFDP1, ATF5, HMGA1, and NFYB to be candidate master regulators (MR) contributing to disease progression. Accordingly, validation was achieved through synthetic lethality assays in which RNAi-mediated silencing of MRs individually or in combination reduced the viability of (14;18)-positive DLBCL (t-DLBCL) cells. Furthermore, specific combinations of small-molecule compounds targeting synergistic MR pairs induced loss of viability in t-DLBCL cells. Collectively, our findings indicate that MR analysis is a valuable method for identifying bona fide contributors to follicular lymphoma transformation and may therefore guide the selection of compounds to be used in combinatorial treatment strategies.
Collapse
Affiliation(s)
| | - Mukesh Bansal
- Department of Systems Biology, Columbia University, New York, New York
| | - Yao Shen
- Department of Systems Biology, Columbia University, New York, New York
| | - Julie Teruya-Feldstein
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York. Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Raju Chaganti
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, New York.
| |
Collapse
|
24
|
|
25
|
Kuntz S, Mazerbourg S, Boisbrun M, Cerella C, Diederich M, Grillier-Vuissoz I, Flament S. Energy restriction mimetic agents to target cancer cells: comparison between 2-deoxyglucose and thiazolidinediones. Biochem Pharmacol 2014; 92:102-11. [PMID: 25083915 DOI: 10.1016/j.bcp.2014.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 01/02/2023]
Abstract
The use of energy restriction mimetic agents (ERMAs) to selectively target cancer cells addicted to glycolysis could be a promising therapeutic approach. Thiazolidinediones (TZDs) are synthetic agonists of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)γ that were developed to treat type II diabetes. These compounds also display anticancer effects which appear mainly to be independent of their PPARγ agonist activity but the molecular mechanisms involved in the anticancer action are not yet well understood. Results obtained on ciglitazone derivatives, mainly in prostate cancer cell models, suggest that these compounds could act as ERMAs. In the present paper, we introduce how compounds like 2-deoxyglucose target the Warburg effect and then we discuss the possibility that the PPARγ-independent effects of various TZD could result from their action as ERMAs.
Collapse
Affiliation(s)
- Sandra Kuntz
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France
| | - Sabine Mazerbourg
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France
| | - Michel Boisbrun
- Université de Lorraine, SRSMC, UMR 7565, Vandœuvre-lès-Nancy, F-54506, France; CNRS, SRSMC, UMR 7565, Vandœuvre-lès-Nancy, F-54506, France
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer. Hôpital Kirchberg, L-2540, Luxembourg
| | - Marc Diederich
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer. Hôpital Kirchberg, L-2540, Luxembourg; Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | - Isabelle Grillier-Vuissoz
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France
| | - Stephane Flament
- Université de Lorraine, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France; CNRS, CRAN, UMR 7039, Vandœuvre-lès-Nancy, F-54506, France.
| |
Collapse
|