1
|
Nazari S, Mosaffa F, Poustforoosh A, Saso L, Firuzi O, Moosavi F. c-MET tyrosine kinase inhibitors reverse multidrug resistance in breast cancer cells by targeting ABCG2 transporter. J Pharm Pharmacol 2025; 77:685-697. [PMID: 40053482 DOI: 10.1093/jpp/rgaf008] [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: 07/25/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Overcoming multidrug resistance (MDR), which is often caused by the overexpression of ATP binding cassette (ABC) transporters in cancer cells remains a major challenge for cancer treatment. Receptor tyrosine kinase inhibitors have demonstrated potential in reversing MDR. This study aimed to investigate the effects of c-MET RTKIs on the reversal of MDR induced by ABCG2 in breast cancer cells. METHODS MTT assay was employed to assess antiproliferative activity of c-MET inhibitors, including cabozantinib, crizotinib, and PHA665752. The accumulation of the fluorescent probe mitoxantrone was evaluated by flow cytometry. The drug-drug interaction in combination treatments was analyzed using CalcuSyn software. RESULTS The combination of cabozantinib, crizotinib, and PHA665752 with mitoxantrone resulted in synergistic effects in MDR cells. This was demonstrated by the mean CI values of 0.32 ± 0.07, 0.53 ± 0.05, and 0.59 ± 0.03, respectively. In the same cells, c-MET inhibitors enhanced the accumulation of mitoxantrone, with accumulation ratios ranging from 1.6 to 3.8, while no change was found in parental MCF-7 cells. Computational analysis revealed that the drug-binding region of ABCG2 transporters could be a viable target for these compounds. CONCLUSION c-MET inhibitors hold potential as effective agents for reversing MDR in ABCG2-medicated drug-resistant cancer cells.
Collapse
Affiliation(s)
- Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Wattanasuntorn P, Poapolathep S, Phuektes P, Alassane-Kpembi I, Fink-Gremmels J, Oswald IP, Poapolathep A. Apoptotic Effect of Combinations of T-2, HT-2, and Diacetoxyscirpenol on Human Jurkat T Cells. Toxins (Basel) 2025; 17:203. [PMID: 40278701 PMCID: PMC12030997 DOI: 10.3390/toxins17040203] [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: 03/06/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Trichothecene type A mycotoxins, such as T-2, HT-2, and diacetoxyscirpenol (DAS), are known to induce cytotoxicity and apoptosis in different cell types. As all three Fusarium toxins may occur concomitantly in a given food or feed commodity, there is growing interest in the effect of such mycotoxin mixtures. This study aimed to identify the toxic interactions among T-2, HT-2, and DAS in a human Jurkat cell model. As a first step, an MTT assay was used to assess cytotoxicity after 24 h of cell exposure to individual mycotoxins and their mixtures. The results were used to calculate the combination index (CI), which indicates the nature of the mycotoxin interactions. In Jurkat T cells, the toxicity ranking for the individual mycotoxins was T-2 > HT-2 > DAS. The CI values of the dual and triple mycotoxin combinations calculated from the results of the MTT and reactive oxygen species assays showed synergistic effects at low concentrations and an apparent antagonism at very high concentrations for all combinations. The additional cytometric analyses confirmed the synergistic effects, as expected, following co-exposure to the three tested trichothecenes. As the lower toxin concentrations investigated reflect natural contamination levels in food and feeds, the synergistic effects identified should be considered in risk characterization for trichothecene exposure in humans and animals.
Collapse
Affiliation(s)
- Phattarawadee Wattanasuntorn
- Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok 10900, Thailand;
| | - Saranya Poapolathep
- Department of Pharmacology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand;
| | - Patchara Phuektes
- Department of Pathobiology, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Imourana Alassane-Kpembi
- Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2R 0A8, Canada;
| | - Johanna Fink-Gremmels
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 Utrecht, The Netherlands;
| | - Isabelle P. Oswald
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP-Purpon, UPS, 31000 Toulouse, France;
| | - Amnart Poapolathep
- Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok 10900, Thailand;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand;
| |
Collapse
|
3
|
Fuentes C, Zingales V, Barat JM, Ruiz MJ. Combined Cytotoxic Effects of the Fungicide Azoxystrobin and Common Food-Contaminating Mycotoxins. Foods 2025; 14:1226. [PMID: 40238467 PMCID: PMC11988556 DOI: 10.3390/foods14071226] [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: 02/27/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
This study assessed the cytotoxicity of the individual and combined exposure to the fungicide azoxystrobin (AZX) and the three common mycotoxins found in food: ochratoxin A (OTA), deoxynivalenol (DON), and T-2 toxin. Cytotoxic effects were evaluated using the resazurin and MTT assays in human hepatocarcinoma (HepG2) cells after 24 h of exposure, and the type of interaction between the compounds was determined using the isobologram method. Results showed that T-2 was the most cytotoxic compound, followed by DON, OTA, and AZX. The compound ratios in the mixture were calculated using three sublethal concentrations (IC50/2, IC50/4, and IC50/8) to achieve equal toxicity for each compound. Interaction analysis revealed that the nature of the interaction varied across components and concentrations. The AZX and DON mixture produced an antagonistic effect at all the analyzed effect levels. AZX and OTA or T2 mixtures, and tertiary combinations displayed antagonism at low effect values but additivity at high effect levels. Importantly, the quaternary mixture demonstrated synergism at all the effect levels. These findings highlight that the co-occurrence of fungicides and mycotoxins in food commodities can lead to complex exposure scenarios that may result in combined toxic effects on the organism.
Collapse
Affiliation(s)
- Cristina Fuentes
- University Institute of Food Engineering–FoodUPV, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain;
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (V.Z.); (M.-J.R.)
| | - Veronica Zingales
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (V.Z.); (M.-J.R.)
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain
| | - José Manuel Barat
- University Institute of Food Engineering–FoodUPV, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain;
| | - María-José Ruiz
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (V.Z.); (M.-J.R.)
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain
| |
Collapse
|
4
|
Nichols Doyle R, Yang V, Kayode YI, Damoiseaux R, Taylor HE, Fregoso OI. NSC95397 Is a Novel HIV-1 Latency-Reversing Agent. Viruses 2024; 16:1783. [PMID: 39599897 PMCID: PMC11599149 DOI: 10.3390/v16111783] [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: 10/15/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
The latent viral reservoir represents one of the major barriers to curing HIV-1. Focus on the "kick and kill" (also called "shock and kill") approach, in which virus expression is reactivated, and then cells producing virus are selectively depleted, has led to the discovery of many latency-reversing agents (LRAs) that have furthered our understanding of the mechanisms driving HIV-1 latency and latency reversal. Thus far, individual compounds have yet to be robust enough to work as a therapy, highlighting the importance of identifying new compounds that target novel pathways and synergize with known LRAs. In this study, we identified a promising LRA, NSC95397, from a screen of ~4250 compounds. We validated that NSC95397 reactivates latent viral transcription and protein expression from cells with unique integration events and across different latency models. Co-treating cells with NSC95397 and known LRAs demonstrated that NSC95397 synergizes with different drugs under both standard normoxic and physiological hypoxic conditions. NSC95397 does not globally increase open chromatin, and bulk RNA sequencing revealed that NSC95397 does not greatly increase cellular transcription. Instead, NSC95397 downregulates pathways key to metabolism, cell growth, and DNA repair-highlighting the potential of these pathways in regulating HIV-1 latency. Overall, we identified NSC95397 as a novel LRA that does not largely alter global transcription, shows potential for synergy with known LRAs, and may act through novel pathways not previously recognized for their ability to modulate HIV-1 latency.
Collapse
Affiliation(s)
- Randilea Nichols Doyle
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.N.D.); (V.Y.)
| | - Vivian Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.N.D.); (V.Y.)
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Yetunde I. Kayode
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA; (Y.I.K.); (H.E.T.)
| | - Robert Damoiseaux
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA;
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Harry E. Taylor
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA; (Y.I.K.); (H.E.T.)
| | - Oliver I. Fregoso
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.N.D.); (V.Y.)
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
He L, Tian Y, Liu Q, Bao J, Ding RB. Antidepressant Sertraline Synergistically Enhances Paclitaxel Efficacy by Inducing Autophagy in Colorectal Cancer Cells. Molecules 2024; 29:3733. [PMID: 39202813 PMCID: PMC11357241 DOI: 10.3390/molecules29163733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. It is important to discover new therapeutic regimens for treating CRC. Depression is known to be an important complication of cancer diseases. Repurposing antidepressants into anticancer drugs and exploring the combinational efficacy of antidepressants and chemotherapy are potentially good options for developing CRC treatment regimens. In this study, sertraline, an antidepressant drug, and paclitaxel, an anticancer drug, were chosen to study their antitumor effects in the treatment of colorectal cancer, alone or in combination, and to explore their underlying mechanisms. The data showed that sertraline exerted a dose-dependent cytotoxic effect on MC38 and CT26 colorectal cancer cell lines with IC50 values of 10.53 μM and 7.47 μM, respectively. Furthermore, sertraline synergistically sensitized chemotherapeutic agent paclitaxel efficacy in CRC cells with combination index (CI) values at various concentrations consistently lower than 1. Sertraline remarkably augmented paclitaxel-induced autophagy by increasing autophagosome formation indicated by elevated LC3-II/I ratio and promoting autophagic flux by degrading autophagy cargo receptor SQSTM1/p62, which may explain the synergistically cytotoxic effect of sertraline and paclitaxel combination therapy on CRC cells. This study provides important evidence to support repurposing sertraline as an anticancer agent and suggests a novel combinational regimen for effectively treating CRC as well as in the simultaneous treatment of CRC and depression.
Collapse
Affiliation(s)
- Leping He
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; (L.H.); (Y.T.); (Q.L.); (J.B.)
| | - Yuxi Tian
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; (L.H.); (Y.T.); (Q.L.); (J.B.)
| | - Qingqing Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; (L.H.); (Y.T.); (Q.L.); (J.B.)
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; (L.H.); (Y.T.); (Q.L.); (J.B.)
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; (L.H.); (Y.T.); (Q.L.); (J.B.)
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| |
Collapse
|
6
|
Markova L, Maji M, Kostrhunova H, Novohradsky V, Kasparkova J, Gibson D, Brabec V. Multiaction Pt(IV) Prodrugs Releasing Cisplatin and Dasatinib Are Potent Anticancer and Anti-Invasive Agents Displaying Synergism between the Two Drugs. J Med Chem 2024; 67:9745-9758. [PMID: 38819023 DOI: 10.1021/acs.jmedchem.4c00888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Herein, we describe the general design, synthesis, characterization, and biological activity of new multitargeting Pt(IV) prodrugs that combine antitumor cisplatin and dasatinib, a potent inhibitor of Src kinase. These prodrugs exhibit impressive antiproliferative and anti-invasive activities in tumor cell lines in both two-dimensional (2D) monolayers of cell cultures and three-dimensional (3D) spheroids. We show that the cisplatin moiety and dasatinib in the investigated Pt(IV) complexes are both involved in the mechanism of action in MCF7 breast cancer cells and act synergistically. Thus, combining dasatinib and cisplatin into one molecule, compared to using individual components in a mix, may bring several advantages, such as significantly higher activity in cancer cell lines and higher selectivity for tumor cells. Most importantly, Pt(IV)-dasatinib complexes hold significant promise for potential anticancer therapies by targeting epithelial-mesenchymal transition, thus preventing the spread and metastasis of tumors, a value unachievable by a simple combination of both individual components.
Collapse
Affiliation(s)
- Lenka Markova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| | - Moumita Maji
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Hana Kostrhunova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| | - Vojtech Novohradsky
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| | - Jana Kasparkova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Viktor Brabec
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| |
Collapse
|
7
|
Gökşen Tosun N. Enhancing therapeutic efficacy in breast cancer: a study on the combined cytotoxic effects of doxorubicin and MPC-3100. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3249-3259. [PMID: 37917369 DOI: 10.1007/s00210-023-02807-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
PURPOSE Combination therapy is a strategy aimed at the combined use of agents targeting different mechanisms in cancer treatment. This study aimed to examine the cytotoxic and apoptotic effects of the traditional chemotherapeutic agent doxorubicin (DOX) and the next-generation HSP90 inhibitor MPC-3100 on breast cancer cell lines. METHODS Firstly, molecular docking analyses were performed, and then the MTT test was conducted to evaluate the individual and combined cytotoxic effects of DOX and MPC-3100 on MCF-7 and MDA-MB-231 breast cancer cell lines. The effect of two drugs combination was assessed by the Chou and Talalay approach. To further investigate the underlying molecular mechanism responsible for this synergistic effect, the gene expression levels of apoptotic and heat shock proteins (HSP), as well as the protein expression levels, were examined using quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) and Western Blotting, respectively. RESULTS Based on the molecular docking results, it was observed that MPC-3100 specifically binds to the ATP binding pocket of Hsp90, exhibiting an estimated free binding energy of -7.9 kcal/mol. MTT results indicated that both DOX and MPC-3100, as well as their combination, exhibited dose-dependent cytotoxicity. The drug combination showed a synergistic effect on both MCF-7 and MDA-MB-231 cell lines. Finally, the investigated molecular mechanism demonstrated that the combination of DOX and MPC-3100 induced apoptosis in breast cancer cells more efficiently than either drug alone. CONCLUSIONS This study showed a possible coordinated mechanism of action between DOX and MPC-3100, pointing to the possibility of a more effective therapeutic strategy for breast cancer therapy.
Collapse
Affiliation(s)
- Nazan Gökşen Tosun
- Tokat Vocational School of Health Services, Department of Medical Services and Techniques, Tokat Gaziosmanpaşa University, Tokat, Turkey.
| |
Collapse
|
8
|
Mathes D, Macedo LB, Pieta TB, Maia BC, Rodrigues OED, Leal JG, Wendt M, Rolim CMB, Mitjans M, Nogueira-Librelotto DR. Co-Delivery of an Innovative Organoselenium Compound and Paclitaxel by pH-Responsive PCL Nanoparticles to Synergistically Overcome Multidrug Resistance in Cancer. Pharmaceutics 2024; 16:590. [PMID: 38794252 PMCID: PMC11124783 DOI: 10.3390/pharmaceutics16050590] [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: 03/19/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
In this study, we designed the association of the organoselenium compound 5'-Seleno-(phenyl)-3'-(ferulic-amido)-thymidine (AFAT-Se), a promising innovative nucleoside analogue, with the antitumor drug paclitaxel, in poly(ε-caprolactone) (PCL)-based nanoparticles (NPs). The nanoprecipitation method was used, adding the lysine-based surfactant, 77KS, as a pH-responsive adjuvant. The physicochemical properties presented by the proposed NPs were consistent with expectations. The co-nanoencapsulation of the bioactive compounds maintained the antioxidant activity of the association and evidenced greater antiproliferative activity in the resistant/MDR tumor cell line NCI/ADR-RES, both in the monolayer/two-dimensional (2D) and in the spheroid/three-dimensional (3D) assays. Hemocompatibility studies indicated the safety of the nanoformulation, corroborating the ability to spare non-tumor 3T3 cells and human mononuclear cells of peripheral blood (PBMCs) from cytotoxic effects, indicating its selectivity for the cancerous cells. Furthermore, the synergistic antiproliferative effect was found for both the association of free compounds and the co-encapsulated formulation. These findings highlight the antitumor potential of combining these bioactives, and the proposed nanoformulation as a potentially safe and effective strategy to overcome multidrug resistance in cancer therapy.
Collapse
Affiliation(s)
- Daniela Mathes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Letícia Bueno Macedo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Engenharia e Processos Químicos, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil
| | - Taís Baldissera Pieta
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Bianca Costa Maia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Oscar Endrigo Dorneles Rodrigues
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (O.E.D.R.); (J.G.L.); (M.W.)
| | - Julliano Guerin Leal
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (O.E.D.R.); (J.G.L.); (M.W.)
| | - Marcelo Wendt
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (O.E.D.R.); (J.G.L.); (M.W.)
| | - Clarice Madalena Bueno Rolim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Montserrat Mitjans
- Departament de Bioquimica i Fisiologia, Facultat de Farmacia i Ciències de l’Alimentaciò, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Daniele Rubert Nogueira-Librelotto
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| |
Collapse
|
9
|
Sarkar A, Novohradsky V, Maji M, Babu T, Markova L, Kostrhunova H, Kasparkova J, Gandin V, Brabec V, Gibson D. Multitargeting Prodrugs that Release Oxaliplatin, Doxorubicin and Gemcitabine are Potent Inhibitors of Tumor Growth and Effective Inducers of Immunogenic Cell Death. Angew Chem Int Ed Engl 2023; 62:e202310774. [PMID: 37646232 DOI: 10.1002/anie.202310774] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/01/2023]
Abstract
A multitargeting prodrug (2) that releases gemcitabine, oxaliplatin, and doxorubicin in their active form in cancer cells is a potent cytotoxic agent with nM IC50s ; it is highly selective to cancer cells with mean selectivity indices to human (136) and murine (320) cancer cells. It effectively induces release of DAMPs (CALR, ATP & HMGB1) in CT26 cells facilitating more efficient phagocytosis by J774 macrophages than the FDA drugs or their co-administration. The viability of CT26 cells co-cultured with J774 macrophages and treated with 2 was reduced by 32 % compared to the non-treated cells, suggesting a synergistic antiproliferative effect between the chemical and immune reactions. 2 inhibited in vivo tumor growth in two murine models (LLC and CT26) better than the FDA drugs or their co-administration with significantly lower body weight loss. Mice inoculated with CT26 cells treated with 2 showed slightly better tumor free survival than doxorubicin.
Collapse
Affiliation(s)
- Amrita Sarkar
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61200, Brno, Czech Republic
| | - Moumita Maji
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Tomer Babu
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Lenka Markova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61200, Brno, Czech Republic
| | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61200, Brno, Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61200, Brno, Czech Republic
- Department of Biophysics, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic
| | - Valentina Gandin
- Dipartimento di Scienze del Farmaco, Universita di Padova, 35131, Padova, Italy
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61200, Brno, Czech Republic
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| |
Collapse
|
10
|
Kaplan Ö. Synergistic induction of apoptosis in liver cancer cells: exploring the combined potential of doxorubicin and XL-888. Med Oncol 2023; 40:318. [PMID: 37794195 DOI: 10.1007/s12032-023-02181-9] [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: 08/14/2023] [Accepted: 09/02/2023] [Indexed: 10/06/2023]
Abstract
Combination therapy has been frequently preferred in treating various types of cancer in recent years. Targeted cancer therapy has also become one of the remarkable treatment modalities. Therefore, the aim of the study to investigate its cytotoxic and apoptotic effects on liver cancer cell lines by combining the classical chemotherapeutic drug doxorubicin (DOX) and a targeted agent, the new generation HSP90 inhibitor XL-888. The molecular docking method was used to predict the binding conformation of XL-888 on the human Hsp90. The single and combined cytotoxic effects of DOX and XL-888 on liver cancer cell lines HepG2 and HUH-7 were determined by MTT assay. The effect of the combined use of two drugs was evaluated using Chou and Talalay method. The levels of apoptotic genes and heat shock proteins gene and protein expression levels were investigated by quantitative real-time polymerase chain reaction and western blotting, respectively. Molecular docking results showed that XL-888 selectively binds to the ATP binding pocket of HSP90 with an estimated free binding energy of - 7.8 kcal/mol. DOX and XL-888 and their combination showed dose-dependent cytotoxic effect. The combination of drugs showed a synergistic effect on both cell lines. The results revealed that the combination of DOX and XL-888 potently induced apoptosis in liver cancer cell lines rather than using drugs alone. The combined treatment of DOX and XL-888 demonstrated synergistic cytotoxic and apoptotic effects on liver cancer cell lines, presenting a promising approach for combination therapy in liver cancer.
Collapse
Affiliation(s)
- Özlem Kaplan
- Department of Genetics and Bioengineering, Rafet Kayış Faculty of Engineering, Alanya Alaaddin Keykubat University, Antalya, Turkey.
| |
Collapse
|
11
|
Coverdale JPC, Kostrhunova H, Markova L, Song H, Postings M, Bridgewater HE, Brabec V, Rogers NJ, Scott P. Triplex metallohelices have enantiomer-dependent mechanisms of action in colon cancer cells. Dalton Trans 2023; 52:6656-6667. [PMID: 37114730 DOI: 10.1039/d3dt00948c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Self-assembled enantiomers of an asymmetric di-iron metallohelix differ in their antiproliferative activities against HCT116 colon cancer cells such that the compound with Λ-helicity at the metals becomes more potent than the Δ compound with increasing exposure time. From concentration- and temperature-dependent 57Fe isotopic labelling studies of cellular accumulation we postulate that while the more potent Λ enantiomer undergoes carrier-mediated efflux, for Δ the process is principally equilibrative. Cell fractionation studies demonstrate that both enantiomers localise in a similar fashion; compound is observed mostly within the cytoskeleton and/or genomic DNA, with significant amounts also found in the nucleus and membrane, but with negligible concentration in the cytosol. Cell cycle analyses using flow cytometry reveal that the Δ enantiomer induces mild arrest in the G1 phase, while Λ causes a very large dose-dependent increase in the G2/M population at a concentration significantly below the relevant IC50. Correspondingly, G2-M checkpoint failure as a result of Λ-metallohelix binding to DNA is shown to be feasible by linear dichroism studies, which indicate, in contrast to the Δ compound, a quite specific mode of binding, probably in the major groove. Further, spindle assembly checkpoint (SAC) failure, which could also be responsible for the observed G2/M arrest, is established as a feasible mechanism for the Λ helix via drug combination (synergy) studies and the discovery of tubulin and actin inhibition. Here, while the Λ compound stabilizes F-actin and induces a distinct change in tubulin architecture of HCT116 cells, Δ promotes depolymerization and more subtle changes in microtubule and actin networks.
Collapse
Affiliation(s)
- J P C Coverdale
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - H Kostrhunova
- The Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - L Markova
- The Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - H Song
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - M Postings
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - H E Bridgewater
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- Centre of Exercise, Sport and Life Science, Faculty of Health and Life Sciences, Coventry University, Coventry, CV1 5FB, UK
| | - V Brabec
- The Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - N J Rogers
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - P Scott
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
12
|
Jin CE, Yoon MS, Jo MJ, Kim SY, Lee JM, Kang SJ, Park CW, Kim JS, Shin DH. Synergistic Encapsulation of Paclitaxel and Sorafenib by Methoxy Poly(Ethylene Glycol)- b-Poly(Caprolactone) Polymeric Micelles for Ovarian Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15041206. [PMID: 37111691 PMCID: PMC10146360 DOI: 10.3390/pharmaceutics15041206] [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: 02/27/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Ovarian cancer has a high mortality rate due to difficult detection at an early stage. It is necessary to develop a novel anticancer treatment that demonstrates improved efficacy while reducing toxicity. Here, using the freeze-drying method, micelles encapsulating paclitaxel (PTX) and sorafenib (SRF) with various polymers were prepared, and the optimal polymer (mPEG-b-PCL) was selected by measuring drug loading (%), encapsulation efficiency (%), particle size, polydispersity index, and zeta potential. The final formulation was selected based on a molar ratio (PTX:SRF = 1:2.3) with synergistic effects on two ovarian cancer cell lines (SKOV3-red-fluc, HeyA8). In the in vitro release assay, PTX/SRF micelles showed a slower release than PTX and SRF single micelles. In pharmacokinetic evaluation, PTX/SRF micelles showed improved bioavailability compared to PTX/SRF solution. In in vivo toxicity assays, no significant differences were observed in body weight between the micellar formulation and the control group. The anticancer effect of PTX/SRF combination therapy was improved compared to the use of a single drug. In the xenografted BALB/c mouse model, the tumor growth inhibition rate of PTX/SRF micelles was 90.44%. Accordingly, PTX/SRF micelles showed improved anticancer effects compared to single-drug therapy in ovarian cancer (SKOV3-red-fluc).
Collapse
Affiliation(s)
- Chae Eun Jin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Moon Sup Yoon
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Min Jeong Jo
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Seo Yeon Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jae Min Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Su Jeong Kang
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Chun-Woong Park
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jin-Seok Kim
- Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul 04310, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| |
Collapse
|
13
|
Mabrouk AA, El-Mezayen NS, Awaad AK, Tadros MI, El-Gazayerly ON, El-Refaie WM. Novel celecoxib-loaded chitosan-fucoidan nanoparticles as potential immunotherapy for oral squamous cell carcinoma: Mechanistic insights. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
14
|
Dong L, Wang S, Zhang L, Liu D, You H. DBDPE and ZnO NPs synergistically induce neurotoxicity of SK-N-SH cells and activate mitochondrial apoptosis signaling pathway and Nrf2-mediated antioxidant pathway. JOURNAL OF HAZARDOUS MATERIALS 2023; 441:129872. [PMID: 36084461 DOI: 10.1016/j.jhazmat.2022.129872] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/07/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
Decabromodiphenyl ethane (DBDPE), a new brominated flame retardant, could negatively affect neurobehavior and pose health risks to humans. Humans are also exposed to widely used nanomaterials. This study investigated the combined toxic effects and action types of DBDPE and Zinc oxide nanoparticles (ZnO NPs) on human neuroblastoma SK-N-SH cells and the toxicity mechanisms. DBDPE inhibited the viability of SK-N-SH cells by 21.87% at 25 mg/L. ZnO NPs synergistically exacerbated the toxic effects of DBDPE. DBDPE and ZnO NPs caused excessive ROS production and inhibition of antioxidant enzyme (SOD and GSH) activity in cells, thus causing oxidative cellular damage. Moreover, DBDPE and ZnO NPs caused apoptosis by disrupting mitochondrial kinetic homeostasis, reducing mitochondrial membrane potential (MMP), increasing cytochrome C release and regulating Bax/Bcl-2 and Caspase-3 mRNA and protein expression. DBDPE and ZnO NPs increased the mRNA expression of nuclear factor erythroid 2- related factor (Nrf2) and its downstream genes. The molecular mechanisms revealed that oxidative stress, apoptosis and mitochondrial dysfunction were the critical factors in combined cytotoxicity. The bioinformatics analysis further indicated that co-exposure affected Nrf2 activation, apoptotic factors expression and mitochondrial fusion. The findings enrich the risk perception of neurotoxicity caused by DBDPE and ZnO NPs.
Collapse
Affiliation(s)
- Liying Dong
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin150090, China.
| | - Shutao Wang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin150090, China.
| | - Lin Zhang
- Queen Mary University of London Engineering School, Northwestern Polytechnical University, Xi'an 710129, China.
| | - Dongmei Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin150090, China.
| | - Hong You
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin150090, China.
| |
Collapse
|
15
|
Discovery of potential natural dihydroorotate dehydrogenase inhibitors and their synergism with brequinar via integrated molecular docking, dynamic simulations and in vitro approach. Sci Rep 2022; 12:19037. [PMID: 36351991 PMCID: PMC9646789 DOI: 10.1038/s41598-022-23006-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/21/2022] [Indexed: 11/10/2022] Open
Abstract
The critical function of dihydroorotate dehydrogenase (DHODH) in pyrimidine synthesis attracted a great interest throughout beyond decades. Inhibitors of human DHODH (hDHODH) have validated efficacy for remedy of many immunological diseases. Brequinar and leflunomide are examples of such compounds. However, most of such immunosuppressive medications suffer from a lot of side effects and accompanied by adverse metabolic disturbances and toxicities. So that, immunomodulation utilizing natural products received the attention of many researchers. In this study, computer-aided molecular docking, molecular dynamic (MD) simulations and biochemical testing were utilized to find new pharmacologically active chemical entities from natural sources to combat immunosuppressive diseases. More specifically, Glide docking was used for a structure-based virtual screening of in-house 3D database of compounds retrieved from some traditionally known immunomodulatory plants surveyed from literature. The top five scored plants were found to be Zingiber officinale, Curcuma longa, Glycyrrhiza glabra, Allium sativum and Olea europaea. In vitro hDHODH inhibitory assays illustrated the ability of Allium sativum and silymarin standard hits; specifically, silibinin, to significantly inhibit the hDHODH enzyme. Molecular docking and MD simulations revealed a strong binding of the discovered hits within the active site. Following that, the most promising hits were tested separately with brequinar in a fixed-ratio combination setting to assess their combined effects on hDHODH catalytic inhibition. The binary combination of silibinin and brequinar revealed that in this combination, brequinar could be utilized at a dose 9.33-fold less when compared to its single-use to produce 99% inhibition for hDHODH enzyme. These findings confirmed that this binary mixture is an excellent combination providing better therapeutic effects and lower side effects.
Collapse
|
16
|
Doki T, Takahashi K, Hasegawa N, Takano T. In vitro antiviral effects of GS-441524 and itraconazole combination against feline infectious peritonitis virus. Res Vet Sci 2022; 144:27-33. [PMID: 35033848 PMCID: PMC8739810 DOI: 10.1016/j.rvsc.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/30/2022]
Abstract
Feline infectious peritonitis virus (FIPV: virulent feline coronavirus) causes a fatal disease called feline infectious peritonitis (FIP) in wild and domestic cat species. Recent studies identified several antiviral drugs that are effective against FIPV. Drug combination is one of the important strategies in the development of novel treatments for viral infections. GS-441524, a nucleoside analog, and itraconazole, a triazole antifungal drug, have been reported that have antiviral effect against FIPV. This study aims to investigate whether the combination of GS-441524 and itraconazole has synergic antiviral effect against FIPV. The antiviral effect was measured by plaque reduction assay using felis catus whole fatus-4 cell. The plaque reduction of GS-441524 against type I FIPVs increased as the concentration of itraconazole increased. The similar result was obtained for type II FIPV. In addition, the calculated combination index (CI) demonstrated that there was a strong synergy between GS-441524 and itraconazole. It is concluded that the combination of GS-441524 and itraconazole may enhance the individual effect of each drug against replication of type I FIPVs and may contribute to development more effective treatment strategy for FIP.
Collapse
Affiliation(s)
- Tomoyoshi Doki
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| | - Ken Takahashi
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | - Nobuhisa Hasegawa
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| | - Tomomi Takano
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| |
Collapse
|
17
|
Macedo LB, Nogueira-Librelotto DR, Mathes D, de Vargas JM, da Rosa RM, Rodrigues OED, Vinardell MP, Mitjans M, Rolim CMB. Overcoming MDR by Associating Doxorubicin and pH-Sensitive PLGA Nanoparticles Containing a Novel Organoselenium Compound-An In Vitro Study. Pharmaceutics 2021; 14:80. [PMID: 35056975 PMCID: PMC8779681 DOI: 10.3390/pharmaceutics14010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, we developed PLGA nanoparticles (NPs) as an effective carrier for 5'-Se-(phenyl)-3-(amino)-thymidine (ACAT-Se), an organoselenium compound, nucleoside analogue that showed promising antitumor activity in vitro. The PLGA NPs were prepared by the nanoprecipitation method and modified with a pH-responsive lysine-based surfactant (77KL). The ACAT-Se-PLGA-77KL-NPs presented nanometric size (around 120 nm), polydispersity index values < 0.20 and negative zeta potential values. The nanoencapsulation of ACAT-Se increased its antioxidant (DPPH and ABTS assays) and antitumor activity in MCF-7 tumor cells. Hemolysis study indicated that ACAT-Se-PLGA-77KL-NPs are hemocompatible and that 77KL provided a pH-sensitive membranolytic behavior to the NPs. The NPs did not induce cytotoxic effects on the nontumor cell line 3T3, suggesting its selectivity for the tumor cells. Moreover, the in vitro antiproliferative activity of NPs was evaluated in association with the antitumor drug doxorubicin. This combination result in synergistic effect in sensitive (MCF-7) and resistant (NCI/ADR-RES) tumor cells, being especially able to successfully sensitize the MDR cells. The obtained results suggested that the proposed ACAT-Se-loaded NPs are a promising delivery system for cancer therapy, especially associated with doxorubicin.
Collapse
Affiliation(s)
- Letícia Bueno Macedo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil; (L.B.M.); (D.M.); (C.M.B.R.)
- Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil;
| | - Daniele Rubert Nogueira-Librelotto
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil; (L.B.M.); (D.M.); (C.M.B.R.)
- Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil;
| | - Daniela Mathes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil; (L.B.M.); (D.M.); (C.M.B.R.)
- Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil;
| | - Josiele Melo de Vargas
- Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil;
| | - Raquel Mello da Rosa
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (O.E.D.R.)
| | - Oscar Endrigo Dorneles Rodrigues
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (O.E.D.R.)
| | - Maria Pilar Vinardell
- Departament de Bioquimica i Fisiologia, Facultat de Farmacia i Ciències de l’Alimentaciò, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
- Institute of Nanocience and Nanotechnology (IN2UB), Universitat de Barcelona, Av. Diagonal 465, 08028 Barcelona, Spain
| | - Montserrat Mitjans
- Departament de Bioquimica i Fisiologia, Facultat de Farmacia i Ciències de l’Alimentaciò, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
- Institute of Nanocience and Nanotechnology (IN2UB), Universitat de Barcelona, Av. Diagonal 465, 08028 Barcelona, Spain
| | - Clarice Madalena Bueno Rolim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil; (L.B.M.); (D.M.); (C.M.B.R.)
- Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, RS, Brazil;
| |
Collapse
|
18
|
Zhou LL, Guan Q, Li WY, Zhang Z, Li YA, Dong YB. A Ferrocene-Functionalized Covalent Organic Framework for Enhancing Chemodynamic Therapy via Redox Dyshomeostasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101368. [PMID: 34216420 DOI: 10.1002/smll.202101368] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/05/2021] [Indexed: 06/13/2023]
Abstract
Chemodynamic therapy (CDT), which induces cell death by decomposing high levels of H2 O2 in tumor cells into highly toxic ·OH, is recognized as a promising antineoplastic approach. However, current CDT approaches are often restricted by the highly controlled and upregulated cellular antioxidant defense. To enhance ·OH-induced cellular damage by CDT, a covalent organic framework (COF)-based, ferrocene (Fc)- and glutathione peroxidase 4 (GPX4) inhibitor-loaded nanodrug, RSL3@COF-Fc (2b), is fabricated. The obtained 2b not only promotes in situ Fenton-like reactions to trigger ·OH production in cells, but also attenuates the repair mechanisms under oxidative stress via irreversible covalent GPX4 inhibition. As a result, these two approaches synergistically result in massive lipid peroxide accumulation, subsequent cell damage, and ultimately ferroptosis, while not being limited by intracellular glutathione. It is believed that this research provides a paradigm for enhancing reactive oxygen species-mediated oncotherapy through redox dyshomeostasis and may provide new insights for developing COF-based nanomedicine.
Collapse
Affiliation(s)
- Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, P. R. China
| | - Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, P. R. China
| | - Wen-Yan Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhiyong Zhang
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, P. R. China
| | - Yan-An Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, P. R. China
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, P. R. China
| |
Collapse
|
19
|
Shaheer K, Lakshmanan MD. Effect of Piperine in Combination with Gamma Radiation on A549 Cells. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2021. [DOI: 10.1055/s-0040-1722808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract
Background Lung cancer is a major constrain that increases mortality globally. Radiotherapy is one of the treatment modalities against lung cancer. A high dose of targeted radiation is required to achieve the treatment efficacy of cell killing. After radiotherapy, eventual tumor progression and therapy resistance are still a consequence of patient who undertakes nonsurgical radiation therapy. Piperine, a plant alkaloid, has been known to enhance the action of the anticancer drugs in various drug-resistant cancer cells. The aim of the current in vitro study was to study the effect of piperine on radiosensitizing property against A549 cells.
Methods In vitro radiosensitizing activity of piperine was elucidated on A549 cells using MTT (3-(4, 5-dimethylthiazol-2-yl)-25-diphenyltetrazolium bromide) assay. CompuSyn analysis was used to compute the combination index values to analyze the combinatory effect of piperine and radiation
Results and Conclusion We observed that piperine increased tumor cell killing in combination with the γ-radiation in vitro. However, further studies are warranted to understand the molecular mechanism of the radiosensitizing action of piperine.
Collapse
Affiliation(s)
- Koniyan Shaheer
- Molecular Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| | - M. Divya Lakshmanan
- Molecular Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| |
Collapse
|
20
|
Assessing the combined toxicity effects of three neonicotinoid pesticide mixtures on human neuroblastoma SK-N-SH and lepidopteran Sf-9 cells. Food Chem Toxicol 2020; 145:111632. [DOI: 10.1016/j.fct.2020.111632] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/02/2020] [Accepted: 07/16/2020] [Indexed: 12/28/2022]
|
21
|
Poly-(Lactic-co-Glycolic) Acid Nanoparticles for Synergistic Delivery of Epirubicin and Paclitaxel to Human Lung Cancer Cells. Molecules 2020; 25:molecules25184243. [PMID: 32947799 PMCID: PMC7570462 DOI: 10.3390/molecules25184243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Combination therapy using chemically distinct drugs has appeared as one of the promising strategies to improve anticancer treatment efficiency. In the present investigation, poly-(lactic-co-glycolic) acid (PLGA) nanoparticles electrostatically conjugated with polyethylenimine (PEI)-based co-delivery system for epirubicin and paclitaxel (PLGA-PEI-EPI-PTX NPs) has been developed. The PLGA-PEI-EPI-PTX NPs exhibited a monodispersed size distribution with an average size of 240.93 ± 12.70 nm as measured through DLS and 70.8-145 nm using AFM. The zeta potential of 41.95 ± 0.65 mV from -17.45 ± 2.15 mV further confirmed the colloidal stability and PEI modification on PLGA nanoparticles. Encapsulation and loading efficiency along with in vitro release of drug for nanoparticles were done spectrophotometrically. The FTIR analysis of PLGA-PEI-EPI-PTX NPs revealed the involvement of amide moiety between polymer PLGA and PEI. The effect of nanoparticles on the cell migration was also corroborated through wound healing assay. The MTT assay demonstrated that PLGA-PEI-EPI-PTX NPs exhibited considerable anticancer potential as compared to the naïve drugs. Further, p53 protein expression analysed through western blot showed enhanced expression. This study suggests that combination therapy using PLGA-PEI-EPI-PTX NPs represent a potential approach and could offer clinical benefits in the future for lung cancer patients.
Collapse
|
22
|
Ling A, Sun L, Guo W, Sun S, Yang J, Zhao Z. Individual and combined cytotoxic effects of T-2 toxin and its four metabolites on porcine Leydig cells. Food Chem Toxicol 2020; 139:111277. [DOI: 10.1016/j.fct.2020.111277] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/17/2020] [Accepted: 03/15/2020] [Indexed: 01/29/2023]
|
23
|
Lasch A, Lichtenstein D, Marx-Stoelting P, Braeuning A, Alarcan J. Mixture effects of chemicals: The difficulty to choose appropriate mathematical models for appropriate conclusions. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 260:113953. [PMID: 31962267 DOI: 10.1016/j.envpol.2020.113953] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/16/2019] [Accepted: 01/08/2020] [Indexed: 06/10/2023]
Abstract
Many different approaches have been proposed to evaluate and predict mixture effects. From a regulatory perspective, several guidance documents have been recently published and provide a strategy for mixture risk assessment based on valuable frameworks to investigate potential synergistic effects. However, some methodological aspects, e.g. for considering mathematical models, are not sufficiently defined. Therefore, the aim of this study was to examine the usefulness of five main mathematical models for mixture effect interpretation: theoretical additivity (TA), concentration addition (CA), independent action (IA), Chou-Talalay (CT), and a benchmark dose approach (BMD) were tested using a fictional data set depicting scenarios of additivity, synergism and antagonism. The synergism and antagonism scenarios were split in x-axis and y-axis synergism/antagonism, meaning a shift of the curve on x-axis or y-axis. The BMD approach was the only model which showed a perfect correspondence for dose addition. Regarding synergism and antagonism, all approaches correspond well for the x-axis synergism and antagonism with only few exceptions. In contrast, some limitations were observed in the particular scenarios of y-axis synergism and antagonism. Therefore our results show that each model has advantages and disadvantages, and that therefore no single model appears the best one for all kinds of application. We would recommend instead the parallel use of different models to increase confidence in the result of mixture effect evaluation.
Collapse
Affiliation(s)
- Alexandra Lasch
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment, Department Pesticides Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Jimmy Alarcan
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| |
Collapse
|
24
|
Chou TC, Shapiro TA, Fu J, Chou JH, Ulrich-Merzenich GS. Computerized quantification of drugs synergism in animal studies or in clinical trials using only ten data points. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.synres.2019.100049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
25
|
Synergistic lethal mutagenesis of hepatitis C virus. Antimicrob Agents Chemother 2019:AAC.01653-19. [PMID: 31570400 DOI: 10.1128/aac.01653-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Lethal mutagenesis is an antiviral approach that consists in extinguishing a virus by an excess of mutations acquired during replication in the presence of a mutagenic agent, often a nucleotide analogue. One of its advantages is its broad spectrum nature that renders the strategy potentially effective against emergent RNA viral infections. Here we describe synergistic lethal mutagenesis of hepatitis C virus (HCV) by a combination of favipiravir (T-705) and ribavirin. Synergy has been documented over a broad range of analogue concentrations using the Chou-Talalay method as implemented in the CompuSyn graphics, with average dose reduction index (DRI) above 1 (68.02±101.6 for favipiravir, and 5.83±6.07 for ribavirin), and average combination indices (CI) below 1 (0.52±0.28). Furthermore, analogue concentrations that individually did not extinguish high fitness HCV in ten serial infections, when used in combination they extinguished high fitness HCV in one to two passages. Although both analogues display a preference for G→A and C→U transitions, deep sequencing analysis of mutant spectra indicated a different preference of the two analogues for the mutation sites, thus unveiling a new possible synergy mechanism in lethal mutagenesis. Prospects of synergy among mutagenic nucleotides as a strategy to confront emerging viral infections are discussed.
Collapse
|
26
|
Nixon SA, Saez NJ, Herzig V, King GF, Kotze AC. The antitrypanosomal diarylamidines, diminazene and pentamidine, show anthelmintic activity against Haemonchus contortus in vitro. Vet Parasitol 2019; 270:40-46. [PMID: 31213240 DOI: 10.1016/j.vetpar.2019.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 12/13/2022]
Abstract
Parasitic nematodes pose a major threat to livestock production worldwide. The blood-feeding parasite Haemonchus contortus is a key small-ruminant pathogen that causes anaemia, and thereby seriously impacts animal health and production. Control of this parasite relies largely upon broad-spectrum anthelmintics, but new drugs are urgently needed to combat the threat of widespread multidrug resistance. Repurposing drugs can accelerate the development pipeline by reducing costs and risks, and can be an effective way of quickly bringing new antiparasitic drugs to market. Diarylamidine compounds such as pentamidine and diminazene have been employed in the treatment of trypanosomiasis and leishmaniasis in both human and veterinary settings, but their activity against parasitic worms has not yet been reported. We screened a small panel of diarylamidine compounds against H. contortus to assess their potential to be repurposed as anthelmintic drugs. Pentamidine and diminazene inhibited H. contortus larval development at low micromolar concentrations (IC50 4.9 μM and 16.1 μM, respectively, in a drug-susceptible isolate) with no existing cross-resistance in two multidrug resistant isolates and a monepantel-resistant isolate. Combinations of pentamidine with commercial anthelmintics showed additive activity, with no significant synergism detected. Pentamidine and diminazene showed different life-stage patterns of activity; both were active against early stage larvae in development assays, but only diminazene was active against the infective L3 stage in migration assays. This suggests some differences in uptake of the two drugs across the nematode cuticle, or differences in the nature and expression patterns of their molecular targets. As pentamidine and diminazene have been reported to be potent inhibitors of mammalian acid-sensing ion channels (ASIC), we tested the activity of known ASIC inhibitors against H. contortus to probe whether these channels may represent potential anthelmintic targets in nematodes. Remarkably, the spider-venom peptide Hi1a, a potent inhibitor of ASIC1a, inhibited H. contortus larval development with an IC50 of 22.9 ± 1.9 μM. This study highlights the potential use of diarylamidines as anthelmintics, although their activity needs to be confirmed in vivo. In addition, our demonstration that ASIC inhibitors have anthelmintic activity raises the possibility that this family of ion channels may represent a novel anthelmintic target.
Collapse
Affiliation(s)
- Samantha A Nixon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; CSIRO Agriculture and Food, Queensland Bioscience Precinct, St. Lucia, QLD 4067, Australia
| | - Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Andrew C Kotze
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, St. Lucia, QLD 4067, Australia.
| |
Collapse
|
27
|
Regulation of monocyte redox balance by 1,8-cineole (eucalyptol) controls oxidative stress and pro-inflammatory responses in vitro: A new option to increase the antioxidant effects of combined respiratory therapy with budesonide and formoterol? ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.synres.2018.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Chou TC. The combination index (CI < 1) as the definition of synergism and of synergy claims. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.synres.2018.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
29
|
Cancer combination therapies with artemisinin-type drugs. Biochem Pharmacol 2017; 139:56-70. [DOI: 10.1016/j.bcp.2017.03.019] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/28/2017] [Indexed: 01/28/2023]
|
30
|
Gao Y, Li J, Qiao N, Meng Q, Zhang M, Wang X, Jia J, Yang S, Qu C, Li W, Wang D. Adrenomedullin blockade suppresses sunitinib-resistant renal cell carcinoma growth by targeting the ERK/MAPK pathway. Oncotarget 2016; 7:63374-63387. [PMID: 27556517 PMCID: PMC5325371 DOI: 10.18632/oncotarget.11463] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/13/2016] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To evaluate the mechanisms underlying sunitinib resistance in RCC and to identify targets that may be used to overcome this resistance. RESULTS Reanalysis of transcriptome microarray datasets (GSE64052 and GSE76068) showed that adrenomedullin expression was increased in sunitinib-resistant tumors. And adrenomedullin expression was increased in sunitinib-resistant tumor xenografts, accompanied by upregulation of phospho-ERK levels. However, blocking adrenomedullin inhibited sunitinib-resistant tumor growth. Treatment of RCC cells with sunitinib and ADM22-52 was superior to monotherapy with either agent. Additionally, adrenomedullin upregulated cAMP and activated the ERK/MAPK pathway, promoting cell proliferation, while knockdown of adrenomedullin inhibited RCC cell growth and invasion in vitro. MATERIALS AND METHODS We searched the Gene Expression Omnibus (GEO) database to find data regarding sunitinib-resistant RCC. These data were subsequently reanalyzed to identify targets that contribute to sunitinib resistance, and adrenomedullin upregulation was found to mediate sunitinib resistance in RCC. Then, we created an RCC mouse xenograft model. Mice were treated with sunitinib, an adrenomedullin receptor antagonist (ADM22-52), a MEK inhibitor (PD98059) and different combinations of these three drugs to investigate their effects on tumor growth. RCC cells (786-0) were cultured in vitro and treated with an ADM22-52 or PD98059 to determine whether adrenomedullin activates the ERK/MAPK pathway. Adrenomedullin was knocked down in 786-0 cells via siRNA, and the effects of this knockdown on cell were subsequently investigated. CONCLUSIONS Adrenomedullin plays an important role in RCC resistance to sunitinib treatment. The combination of sunitinib and an adrenomedullin receptor antagonist may result in better outcomes in advanced RCC patients.
Collapse
Affiliation(s)
- Yongqian Gao
- Department of Interventional Radiology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan 063000, P.R. China
| | - Jinyi Li
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Na Qiao
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Qingsong Meng
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ming Zhang
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Xin Wang
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jianghua Jia
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Shuwen Yang
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Changbao Qu
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Wei Li
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Dongbin Wang
- Department of Urologic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| |
Collapse
|
31
|
Fu J, Zhang N, Chou JH, Dong HJ, Lin SF, Ulrich-Merzenich GS, Chou TC. Drug combination in vivo using combination index method: Taxotere and T607 against colon carcinoma HCT-116 xenograft tumor in nude mice. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.synres.2016.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Onionin A inhibits ovarian cancer progression by suppressing cancer cell proliferation and the protumour function of macrophages. Sci Rep 2016; 6:29588. [PMID: 27404320 PMCID: PMC4941721 DOI: 10.1038/srep29588] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 06/22/2016] [Indexed: 12/23/2022] Open
Abstract
It is well known that tumour-associated macrophages (TAMs) play an important role in tumour development by modulating the tumour microenvironment, and targeting of protumour activation or the M2 polarization of TAMs is expected to be an effective therapy for cancer patients. We previously demonstrated that onionin A (ONA), a natural low molecular weight compound isolated from onions, has an inhibitory effect on M2 macrophage polarization. In the present study, we investigated whether ONA had a therapeutic anti-ovarian cancer effect using in vitro and in vivo studies. We found that ONA reduced the extent of ovarian cancer cell proliferation induced by co-culture with human macrophages. In addition, we also found that ONA directly suppressed cancer cell proliferation. A combinatorial effect with ONA and anti-cancer drugs was also observed. The activation of signal transducer and activator of transcription 3 (STAT3), which is involved in cell proliferation and chemo-resistance, was significantly abrogated by ONA in ovarian cancer cells. Furthermore, the administration of ONA suppressed cancer progression and prolonged the survival time in a murine ovarian cancer model under single and combined treatment conditions. Thus, ONA is considered useful for the additional treatment of patients with ovarian cancer owing to its suppression of the protumour activation of TAMs and direct cytotoxicity against cancer cells.
Collapse
|
33
|
García-Vilas JA, Quesada AR, Medina MÁ. Screening of synergistic interactions of epigallocatechin-3-gallate with antiangiogenic and antitumor compounds. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.synres.2016.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
Ferron PJ, Dumazeau K, Beaulieu JF, Le Hégarat L, Fessard V. Combined Effects of Lipophilic Phycotoxins (Okadaic Acid, Azapsiracid-1 and Yessotoxin) on Human Intestinal Cells Models. Toxins (Basel) 2016; 8:50. [PMID: 26907345 PMCID: PMC4773803 DOI: 10.3390/toxins8020050] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 12/02/2022] Open
Abstract
Phycotoxins are monitored in seafood because they can cause food poisonings in humans. Phycotoxins do not only occur singly but also as mixtures in shellfish. The aim of this study was to evaluate the in vitro toxic interactions of binary combinations of three lipophilic phycotoxins commonly found in Europe (okadaic acid (OA), yessotoxin (YTX) and azaspiracid-1 (AZA-1)) using the neutral red uptake assay on two human intestinal cell models, Caco-2 and the human intestinal epithelial crypt-like cells (HIEC). Based on the cytotoxicity of individual toxins, we studied the interactions between toxins in binary mixtures using the combination index-isobologram equation, a method widely used in pharmacology to study drug interactions. This method quantitatively classifies interactions between toxins in mixtures as synergistic, additive or antagonistic. AZA-1/OA, and YTX/OA mixtures showed increasing antagonism with increasing toxin concentrations. In contrast, the AZA-1/YTX mixture showed increasing synergism with increasing concentrations, especially for mixtures with high YTX concentrations. These results highlight the hazard potency of AZA-1/YTX mixtures with regard to seafood intoxication.
Collapse
Affiliation(s)
- Pierre-Jean Ferron
- Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères 35300, France.
| | - Kevin Dumazeau
- Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères 35300, France.
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, University of Sherbrooke, Sherbrooke, QC J1G 0A2, Canada.
| | - Ludovic Le Hégarat
- Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères 35300, France.
| | - Valérie Fessard
- Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères 35300, France.
| |
Collapse
|
35
|
Kim CS, Mout R, Zhao Y, Yeh YC, Tang R, Jeong Y, Duncan B, Hardy JA, Rotello VM. Co-delivery of protein and small molecule therapeutics using nanoparticle-stabilized nanocapsules. Bioconjug Chem 2015; 26:950-4. [PMID: 25894332 DOI: 10.1021/acs.bioconjchem.5b00146] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Combination therapy employing proteins and small molecules provides access to synergistic treatment strategies. Co-delivery of these two payloads is challenging due to the divergent physicochemical properties of small molecule and protein cargos. Nanoparticle-stabilized nanocapsules (NPSCs) are promising for combination treatment strategies since they have the potential to deliver small molecule drugs and proteins simultaneously into the cytosol. In this study, we loaded paclitaxel into the hydrophobic core of the NPSC and self-assembled caspase-3 and nanoparticles on the capsule surface. The resulting combination NPSCs showed higher cytotoxicity than either of the single agent NPSCs, with synergistic action established using combination index values.
Collapse
Affiliation(s)
- Chang Soo Kim
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Rubul Mout
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Yunlong Zhao
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Yi-Cheun Yeh
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Rui Tang
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Youngdo Jeong
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Bradley Duncan
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Jeanne A Hardy
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|