1
|
Brisdelli F, Bognanni N, Piccirilli A, Perilli M, Bellotti D, Remelli M, Vecchio G. Polyimidazole ligands: Copper(II) complexes and antiproliferative activity in cancer cells. J Inorg Biochem 2024; 260:112685. [PMID: 39142054 DOI: 10.1016/j.jinorgbio.2024.112685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024]
Abstract
The design of novel chelators for therapeutic applications has been the subject of extensive research to address various diseases. Many chelators can manipulate the levels of metal ions within cells and effectively modulate the metal excess. In some cases, chelators show significant toxicity to cells. We investigated polyimidazole ligands by potentiometry and UV-Vis spectroscopy for their ability to form copper(II) complexes. We also compared the antiproliferative activity of the polyimidazole ligands and their copper(II) complexes with polypyridine ligands in CaCo-2 (colorectal adenocarcinoma), SH-SY5Y (neuroblastoma) and K562 (chronic myelogenous leukemia) cells and normal HaCaT (keratinocyte) cells. Polyimidazole ligands are less cytotoxic than their analogous polypyridine ligands. All polyimidazole ligands, except the tetraimidazole ligand for K562 cells, did not show any significant effect on the viability of cancer and normal cells. In contrast, the cytotoxic activity of polypiridine ligands was also observed in normal cells with IC50 values similar to those of cancer cells. Tetraimidazole ligand, the only ligand active on the leukemic K562 cell line, induced caspase-dependent apoptosis and increased intracellular reactive oxygen species production with mitochondrial damage. The low cytotoxicity of the polyimidazole ligands, even if it limits their use as anticancer agents, could make them useful in other medical applications, such as in the treatment of metal overload, microbial infections, inflammation or neurodegenerative disorders.
Collapse
Affiliation(s)
- Fabrizia Brisdelli
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, Università degli Studi dell'Aquila, via Vetoio, Coppito 67100, L'Aquila, Italy
| | - Noemi Bognanni
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Alessandra Piccirilli
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, Università degli Studi dell'Aquila, via Vetoio, Coppito 67100, L'Aquila, Italy
| | - Mariagrazia Perilli
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, Università degli Studi dell'Aquila, via Vetoio, Coppito 67100, L'Aquila, Italy
| | - Denise Bellotti
- Dipartimento di Scienze Chimiche, Farmaceutiche ed Agrarie, Università degli Studi di Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| | - Maurizio Remelli
- Dipartimento di Scienze Chimiche, Farmaceutiche ed Agrarie, Università degli Studi di Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, V.le A. Doria 6, 95125 Catania, Italy.
| |
Collapse
|
2
|
Liao H, He B. Predictive value of cuproptosis and disulfidptosis-related lncRNA in head and neck squamous cell carcinoma prognosis and treatment. Heliyon 2024; 10:e37996. [PMID: 39323825 PMCID: PMC11422553 DOI: 10.1016/j.heliyon.2024.e37996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024] Open
Abstract
Objective Head and neck squamous cell carcinoma (HNSCC) is a highly lethal and prevalent malignant tumor with a poor prognosis due to its high recurrence rate, This study aims to develop a prognostic index for HNSCC patients based on Cuproptosis and Disulfidptosis-related long noncoding RNA. Methods Gene expression and clinical data for HNSCC were obtained from The Cancer Genome Atlas (TCGA). Using Lasso regression and multivariate Cox regression, we established a risk scoring model. The predictive ability of the nomogram, based on clinical features and risk scores, was verified using receiver operating characteristics and calibration curves. We compared independent prognostic parameters, risk score distribution, and survival between high-risk and low-risk groups, followed by preliminary validity evaluations of the model. Results Our systematic evaluation of prognostic risk provides a new direction for improving the survival prognosis of HNSCC patients in clinical practice, The model effectively categorized patients into high- and low-risk groups with distinct outcomes, identifying numerous gene mutations in these groups, A low-risk score was associated with a better prognosis and higher survival rates. Conclusion The risk score prognostic prediction system developed in this study shows potential efficacy in predicting the prognosis of HNSCC patients and has practical applications in clinical settings.
Collapse
Affiliation(s)
- Hongming Liao
- Department of Otolaryngology Head and neck surgery, Tianmen first people's Hospital, Tianmen, Hubei, 431700, China
| | - Benchao He
- Department of Otolaryngology Head and neck surgery, Tianmen first people's Hospital, Tianmen, Hubei, 431700, China
| |
Collapse
|
3
|
Bhat AA, Afzal M, Moglad E, Thapa R, Ali H, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Subramaniyan V. lncRNAs as prognostic markers and therapeutic targets in cuproptosis-mediated cancer. Clin Exp Med 2024; 24:226. [PMID: 39325172 PMCID: PMC11427524 DOI: 10.1007/s10238-024-01491-0] [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: 08/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in various cellular processes, including cancer progression and stress response. Recent studies have demonstrated that copper accumulation induces a unique form of cell death known as cuproptosis, with lncRNAs playing a key role in regulating cuproptosis-associated pathways. These lncRNAs may trigger cell-specific responses to copper stress, presenting new opportunities as prognostic markers and therapeutic targets. This paper delves into the role of lncRNAs in cuproptosis-mediated cancer, underscoring their potential as biomarkers and targets for innovative therapeutic strategies. A thorough review of scientific literature was conducted, utilizing databases such as PubMed, Google Scholar, and ScienceDirect, with search terms like 'lncRNAs,' 'cuproptosis,' and 'cancer.' Studies were selected based on their relevance to lncRNA regulation of cuproptosis pathways and their implications for cancer prognosis and treatment. The review highlights the significant contribution of lncRNAs in regulating cuproptosis-related genes and pathways, impacting copper metabolism, mitochondrial stress responses, and apoptotic signaling. Specific lncRNAs are potential prognostic markers in breast, lung, liver, ovarian, pancreatic, and gastric cancers. The objective of this article is to explore the role of lncRNAs as potential prognostic markers and therapeutic targets in cancers mediated by cuproptosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
4
|
Njenga LW, Mbugua SN, Odhiambo RA, Onani MO. Correction: Addressing the gaps in homeostatic mechanisms of copper and copper dithiocarbamate complexes in cancer therapy: a shift from classical platinum-drug mechanisms. Dalton Trans 2024; 53:12391-12394. [PMID: 38989691 DOI: 10.1039/d3dt90193a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Correction for 'Addressing the gaps in homeostatic mechanisms of copper and copper dithiocarbamate complexes in cancer therapy: a shift from classical platinum-drug mechanisms' by Lydia W. Njenga et al., Dalton Trans., 2023, 52, 5823-5847, https://doi.org/10.1039/D3DT00366C.
Collapse
Affiliation(s)
- Lydia W Njenga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya.
| | - Simon N Mbugua
- Department of Chemistry, Kisii University, P.O. Box 408-40200, Kisii, Kenya
| | - Ruth A Odhiambo
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya.
| | - Martin O Onani
- Department of Chemical Sciences, University of the Western Cape, Private Bag X17, Belville, 7535, South Africa
| |
Collapse
|
5
|
Massoud SS, Mautner FA, Louka FR, Salem NMH, Fischer RC, Torvisco A, Vančo J, Belza J, Dvořák Z, Trávníček Z. Structurally diverse zinc(II) complexes containing tripodal tetradentate phenoxido-amines with promising antiproliferative effects. Dalton Trans 2024; 53:12261-12280. [PMID: 38980002 DOI: 10.1039/d4dt00942h] [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: 07/10/2024]
Abstract
Structurally diverse zinc(II) complexes with tripodal tetradentate phenolic-amines of variable substituents in the phenol and amine moieties were synthesized and thoroughly characterized. The two dinuclear [Zn2(L1)2](ClO4)2·MeOH (1), [Zn2(L2)2](ClO4)2 (2), and four mononuclear [Zn(L3)(H2O)]·MeOH (3), [Zn(L4)] (4), [Zn(L5)] (5) and [Zn(L6)] (6) complexes revealed distorted octahedral, trigonal-bipyramidal or tetrahedral geometries. The free HL1 and H2L3-6 ligands, and complexes 1-6 were evaluated for in vitro cytotoxicity against human cancer cell lines (A2780, A2780R, PC-3 and 22Rv1) and normal healthy MRC-5 cells. Overall results revealed high-to-moderate cytotoxicity (with the best IC50 values for complex 6 ranging from 2.4 to 4.5 μM), which is however, significantly higher than that of the reference drug cisplatin. The moderately active complexes 1-4 showed considerable selectivity on A2780 cells (IC50 ≈ 16.3-19.5 μM) over MRC-5 ones (with IC50 >50 μM for 1, 2 and 4, and with IC50 >25 μM for 3). The complexes 1, 2, and 6 and the ligand H2L6 were chosen for subsequent deeper biological evaluations. Their time-resolved cellular uptake and other cellular effects in A2780 cells were studied, such as cell cycle profile, intracellular ROS production, induction of apoptosis and activation of caspases 3/7. Complexes 1 and 2 caused significant G0/G1 cell cycle arrest in A2780 cells and antioxidant effects at normal conditions. They showed only limited effects on cellular processes connected with cytotoxicity, i.e. induction of apoptosis, depletion of mitochondrial membrane potential, and autophagy. These findings can be at least partly attributed to the low ability of the complexes to enter the A2780 cells and the depression of metabolic activity of the target cancer cells.
Collapse
Affiliation(s)
- Salah S Massoud
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 43700, Lafayette, LA 70504, USA.
- Department of Chemistry, Faculty of Science, Alexandria University, Moharam Bey 21511, Alexandria, Egypt
| | - Franz A Mautner
- Institut für Physikalische and Theoretische Chemie, Technische Universität Graz, Stremayrgasse 9/II, A-8010, Graz, Austria.
| | - Febee R Louka
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 43700, Lafayette, LA 70504, USA.
| | - Nahed M H Salem
- Department of Chemistry, Faculty of Science, Alexandria University, Moharam Bey 21511, Alexandria, Egypt
| | - Roland C Fischer
- Institut für Anorganische Chemische, Technische Universität Graz, Stremayrgasse 9/V, A-8010 Graz, Austria
| | - Ana Torvisco
- Institut für Anorganische Chemische, Technische Universität Graz, Stremayrgasse 9/V, A-8010 Graz, Austria
| | - Ján Vančo
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic.
| | - Jan Belza
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic.
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-783 71 Olomouc, Czech Republic
| | - Zdeněk Trávníček
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic.
| |
Collapse
|
6
|
Khalil MNA, Afifi SM, Eltanany BM, Pont L, Benavente F, El-Sonbaty SM, Sedeek MS. Assessment of the effect of drying on Brassica greens via a multiplex approach based on LC-QTOF-MS/MS, molecular networking, and chemometrics along with their antioxidant and anticancer activities. Food Res Int 2024; 180:114053. [PMID: 38395547 DOI: 10.1016/j.foodres.2024.114053] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/13/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024]
Abstract
Turnip (Brassica rapa var rapa L.) leaves are a rich source of versatile bioactive phytochemicals with great potential in the food and herbal industries. However, the effect of drying on its constituents has never been studied before. Hereto, three drying techniques were compared, namely, lyophilization (LY), vacuum oven (VO), and shade drying (SD). Chemical profiling utilizing liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (LC-QTOF-MS/MS) combined with chemometrics showed the different impacts of the drying methods on the phytochemical composition of the alcoholic leaf extracts. Unsupervised principal component analysis (PCA) and supervised partial least squares-discriminant analysis (PLS-DA) of the LC-QTOF-MS/MS data showed distinct distant clustering across the three drying techniques. Loading plots and VIP scores demonstrated that sinapic acid, isorhamnetin glycosides, and sinapoyl malate were key markers for LY samples. Meanwhile, oxygenated and polyunsaturated fatty acids were characteristic for SD samples and oxygenated polyunsaturated fatty acids and verbascoside were characteristic for VO samples. LY resulted in the highest total phenolics (TP) and total flavonoid (TF) contents followed by SD and VO. LY and SD samples had much higher antioxidant activity than VO measured by 2,2-diphenyl-1-picrylhydrazyl (DPPH), oxygen radical absorbance capacity (ORAC), and iron metal chelation assays. According to the anticancer activity, the drying methods were ranked in descending order as SD > LY ≫ VO when tested against colon, breast, liver, and lung cancer cell lines. Among the identified compounds, flavonoids and omega-3 fatty acids were key metabolites responsible for the anticancer activity as revealed by partial least squares (PLS) regression and correlation analyses. In conclusion, compared to LY, SD projected out as a cost-effective drying method without compromising the phytochemical and biological activities of Brassica greens. The current findings lay the foundation for further studies concerned with the valorization of Brassica greens.
Collapse
Affiliation(s)
- Mohammed N A Khalil
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Sherif M Afifi
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt
| | - Basma M Eltanany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Laura Pont
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona 08028, Spain; Serra Húnter Program, Generalitat de Catalunya, Barcelona 08007, Spain
| | - Fernando Benavente
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona 08028, Spain.
| | - Sawsan M El-Sonbaty
- Department of Radiation Microbiology, The National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Nasr City 11787, Egypt
| | - Mohamed S Sedeek
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
7
|
Ran XM, Xiao H, Tang YX, Jin X, Tang X, Zhang J, Li H, Li YK, Tang ZZ. The effect of cuproptosis-relevant genes on the immune infiltration and metabolism of gynecological oncology by multiply analysis and experiments validation. Sci Rep 2023; 13:19474. [PMID: 37945610 PMCID: PMC10636103 DOI: 10.1038/s41598-023-45076-5] [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: 07/20/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
Gynecological cancers are a leading cause of mortality for women, including ovarian cancer (OC), cervical squamous cell carcinoma (CESC), and uterine corpus endometrial carcinoma (UCEC). Nevertheless, these gynecological cancer types have not elucidated the role of cuproptosis and the correlated tumor microenvironment (TME) infiltration features. CRGs had important potential molecular functions and prognostic significance in gynecological cancers, especially in UCEC. Hub CRG, FDX1, was correlated with the CD8+ T cell immune infiltration in UCEC and CESC. FDX1 OE could significantly repress the proliferation ability in UCEC cells by MTT, EdU, and clone formation. High levels of FDX1 could repress ATP and lactic acid but enhance ROS and glucose levels by metabolism assay. The xenograft tumor model indicated that FDX1 OE significantly inhibited the growth of UCEC and attenuated the PCNA, HK2, PKM2, and Ki-67 expression. These CRGs are significant roles that could be potential markers and treatment targets to optimize the TME immune cell infiltration features for gynecological cancer types. FDX1 is a hub CRGs in UCEC to promote immune infiltration and attenuate proliferation and metabolism.
Collapse
Affiliation(s)
- Xiao-Min Ran
- Department of Gynecologic Oncology Ward5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Xiao
- Department of Gynecologic Oncology Ward1, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Xiang Tang
- Department of Gynecologic Oncology Ward1, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xia Jin
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xing Tang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital, Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital, Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yu-Kun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital, Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Zhen-Zi Tang
- Department of Gynecologic Oncology Ward1, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Zhou C, Yang J, Liu T, Jia R, Yang L, Sun P, Zhao W. Copper metabolism and hepatocellular carcinoma: current insights. Front Oncol 2023; 13:1186659. [PMID: 37476384 PMCID: PMC10355993 DOI: 10.3389/fonc.2023.1186659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Copper is an essential trace element that acts as a cofactor in various enzyme active sites in the human body. It participates in numerous life activities, including lipid metabolism, energy metabolism, and neurotransmitter synthesis. The proposal of "Cuproptosis" has made copper metabolism-related pathways a research hotspot in the field of tumor therapy, which has attracted great attention. This review discusses the biological processes of copper uptake, transport, and storage in human cells. It highlights the mechanisms by which copper metabolism affects hepatocellular carcinogenesis and metastasis, including autophagy, apoptosis, vascular invasion, cuproptosis, and ferroptosis. Additionally, it summarizes the current clinical applications of copper metabolism-related drugs in antitumor therapy.
Collapse
Affiliation(s)
- Cheng Zhou
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinqiu Yang
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ran Jia
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital Affiliated to Shaanxi University of Chinese Medicine, Xianyang, China
| | - Pengfei Sun
- Department of Orthopaedics, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenxia Zhao
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
9
|
Njenga LW, Mbugua SN, Odhiambo RA, Onani MO. Addressing the gaps in homeostatic mechanisms of copper and copper dithiocarbamate complexes in cancer therapy: a shift from classical platinum-drug mechanisms. Dalton Trans 2023; 52:5823-5847. [PMID: 37021641 DOI: 10.1039/d3dt00366c] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
The platinum drug, cisplatin, is considered as among the most successful medications in cancer treatment. However, due to its inherent toxicity and resistance limitations, research into other metal-based non-platinum anticancer medications with diverse mechanisms of action remains an active field. In this regard, copper complexes feature among non-platinum compounds which have shown promising potential as effective anticancer drugs. Moreover, the interesting discovery that cancer cells can alter their copper homeostatic processes to develop resistance to platinum-based treatments leads to suggestions that some copper compounds can indeed re-sensitize cancer cells to these drugs. In this work, we review copper and copper complexes bearing dithiocarbamate ligands which have shown promising results as anticancer agents. Dithiocarbamate ligands act as effective ionophores to convey the complexes of interest into cells thereby influencing the metal homeostatic balance and inducing apoptosis through various mechanisms. We focus on copper homeostasis in mammalian cells and on our current understanding of copper dysregulation in cancer and recent therapeutic breakthroughs using copper coordination complexes as anticancer drugs. We also discuss the molecular foundation of the mechanisms underlying their anticancer action. The opportunities that exist in research for these compounds and their potential as anticancer agents, especially when coupled with ligands such as dithiocarbamates, are also reviewed.
Collapse
Affiliation(s)
- Lydia W Njenga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya.
| | - Simon N Mbugua
- Department of Chemistry, Kisii University, P.O. Box 408-40200, Kisii, Kenya
| | - Ruth A Odhiambo
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya.
| | - Martin O Onani
- Department of Chemical Sciences, University of the Western Cape, Private Bag X17, Belville, 7535, South Africa
| |
Collapse
|
10
|
Ji HB, Kim CR, Min CH, Han JH, Kim S, Lee C, Choy YB. Fe-containing metal-organic framework with D-penicillamine for cancer-specific hydrogen peroxide generation and enhanced chemodynamic therapy. Bioeng Transl Med 2023; 8:e10477. [PMID: 37206221 PMCID: PMC10189484 DOI: 10.1002/btm2.10477] [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: 09/13/2022] [Revised: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 02/04/2023] Open
Abstract
Chemodynamic therapy (CDT) is based on the production of cytotoxic reactive oxygen species, such as hydroxyl radicals (•OH). Thus, CDT can be advantageous when it is cancer-specific, in terms of efficacy and safety. Therefore, we propose NH2-MIL-101(Fe), a Fe-containing metal-organic framework (MOF), as a carrier of Cu (copper)-chelating agent, d-penicillamine (d-pen; i.e., the NH2-MIL-101(Fe)/d-pen), as well as a catalyst with Fe-metal clusters for Fenton reaction. NH2-MIL-101(Fe)/d-pen in the form of nanoparticles was efficiently taken into cancer cells and released d-pen in a sustained manner. The released d-pen chelated Cu that is highly expressed in cancer environments and this produces extra H2O2, which is then decomposed by Fe in NH2-MIL-101(Fe) to generate •OH. Therefore, the cytotoxicity of NH2-MIL-101(Fe)/d-pen was observed in cancer cells, not in normal cells. We also suggest a formulation of NH2-MIL-101(Fe)/d-pen combined with NH2-MIL-101(Fe) loaded with the chemotherapeutic drug, irinotecan (CPT-11; NH2-MIL-101(Fe)/CPT-11). When intratumorally injected into tumor-bearing mice in vivo, this combined formulation exhibited the most prominent anticancer effects among all tested formulations, owing to the synergistic effect of CDT and chemotherapy.
Collapse
Affiliation(s)
- Han Bi Ji
- Interdisciplinary Program in BioengineeringCollege of Engineering, Seoul National UniversitySeoulRepublic of Korea
| | - Cho Rim Kim
- Interdisciplinary Program in BioengineeringCollege of Engineering, Seoul National UniversitySeoulRepublic of Korea
| | - Chang Hee Min
- Interdisciplinary Program in BioengineeringCollege of Engineering, Seoul National UniversitySeoulRepublic of Korea
| | - Jae Hoon Han
- Interdisciplinary Program in BioengineeringCollege of Engineering, Seoul National UniversitySeoulRepublic of Korea
| | - Se‐Na Kim
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National UniversitySeoulRepublic of Korea
| | - Cheol Lee
- Department of PathologySeoul National University College of MedicineSeoulRepublic of Korea
| | - Young Bin Choy
- Interdisciplinary Program in BioengineeringCollege of Engineering, Seoul National UniversitySeoulRepublic of Korea
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National UniversitySeoulRepublic of Korea
- Department of Biomedical EngineeringSeoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
11
|
Pastuch-Gawołek G, Szreder J, Domińska M, Pielok M, Cichy P, Grymel M. A Small Sugar Molecule with Huge Potential in Targeted Cancer Therapy. Pharmaceutics 2023; 15:913. [PMID: 36986774 PMCID: PMC10056414 DOI: 10.3390/pharmaceutics15030913] [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: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The number of cancer-related diseases is still growing. Despite the availability of a large number of anticancer drugs, the ideal drug is still being sought that would be effective, selective, and overcome the effect of multidrug resistance. Therefore, researchers are still looking for ways to improve the properties of already-used chemotherapeutics. One of the possibilities is the development of targeted therapies. The use of prodrugs that release the bioactive substance only under the influence of factors characteristic of the tumor microenvironment makes it possible to deliver the drug precisely to the cancer cells. Obtaining such compounds is possible by coupling a therapeutic agent with a ligand targeting receptors, to which the attached ligand shows affinity and is overexpressed in cancer cells. Another way is to encapsulate the drug in a carrier that is stable in physiological conditions and sensitive to conditions of the tumor microenvironment. Such a carrier can be directed by attaching to it a ligand recognized by receptors typical of tumor cells. Sugars seem to be ideal ligands for obtaining prodrugs targeted at receptors overexpressed in cancer cells. They can also be ligands modifying polymers' drug carriers. Furthermore, polysaccharides can act as selective nanocarriers for numerous chemotherapeutics. The proof of this thesis is the huge number of papers devoted to their use for modification or targeted transport of anticancer compounds. In this work, selected examples of broad-defined sugars application for improving the properties of both already-used drugs and substances exhibiting anticancer activity are presented.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Julia Szreder
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Monika Domińska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mateusz Pielok
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Piotr Cichy
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mirosława Grymel
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| |
Collapse
|
12
|
Kontoghiorghes GJ. Deferiprone and Iron-Maltol: Forty Years since Their Discovery and Insights into Their Drug Design, Development, Clinical Use and Future Prospects. Int J Mol Sci 2023; 24:ijms24054970. [PMID: 36902402 PMCID: PMC10002863 DOI: 10.3390/ijms24054970] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
The historical insights and background of the discovery, development and clinical use of deferiprone (L1) and the maltol-iron complex, which were discovered over 40 years ago, highlight the difficulties, complexities and efforts in general orphan drug development programs originating from academic centers. Deferiprone is widely used for the removal of excess iron in the treatment of iron overload diseases, but also in many other diseases associated with iron toxicity, as well as the modulation of iron metabolism pathways. The maltol-iron complex is a recently approved drug used for increasing iron intake in the treatment of iron deficiency anemia, a condition affecting one-third to one-quarter of the world's population. Detailed insights into different aspects of drug development associated with L1 and the maltol-iron complex are revealed, including theoretical concepts of invention; drug discovery; new chemical synthesis; in vitro, in vivo and clinical screening; toxicology; pharmacology; and the optimization of dose protocols. The prospects of the application of these two drugs in many other diseases are discussed under the light of competing drugs from other academic and commercial centers and also different regulatory authorities. The underlying scientific and other strategies, as well as the many limitations in the present global scene of pharmaceuticals, are also highlighted, with an emphasis on the priorities for orphan drug and emergency medicine development, including the roles of the academic scientific community, pharmaceutical companies and patient organizations.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
13
|
Alkis ME, Buldurun K, Alan Y, Turan N, Altun A. Electroporation Enhances the Anticancer Effects of Novel Cu(II) and Fe(II) Complexes in Chemotherapy-Resistant Glioblastoma Cancer Cells. Chem Biodivers 2023; 20:e202200710. [PMID: 36601965 DOI: 10.1002/cbdv.202200710] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/26/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
Schiff base ligand (L) was obtained by condensation reaction between 4-aminopyrimidin-2(1H)-one (cytosine) with 2-hydroxybenzaldehyde. The synthesized Schiff base was used for complexation with Cu(II) and Fe(II) ions used by a molar (2 : 1 mmol ration) in methanol solvent. The structural features of ligand, Cu(II), and Fe(II) metal complexes were determined by standard spectroscopic methods (FT-IR, elemental analysis, proton and carbon NMR spectra, UV/VIS, and mass spectroscopy, magnetic susceptibility, thermal analysis, and powder X-ray diffraction). The synthesized compounds (Schiff base and its metal complexes) were screened in terms of their anti-proliferative activities in U118 and T98G human glioblastoma cell lines alone or in combination with electroporation (EP). Moreover, the human HDF (human dermal fibroblast) cell lines was used to check the bio-compatibility of the compounds. Anti-proliferative activities of all compounds were ascertained using an MTT assay. The complexes exhibited a good anti-proliferative effect on U118 and T98G glioblastoma cell lines. In addition, these compounds had a negligible cytotoxic effect on the fibroblast HDF cell lines. The use of compounds in combination with EP significantly decreased the IC50 values compared to the use of compounds alone (p<0.05). These results show that newly synthesized Cu(II) and Fe(II) complexes can be developed for use in the treatment of chemotherapy-resistant U118 and T98G glioblastoma cells and that treatment with lower doses can be provided when used in combination with EP.
Collapse
Affiliation(s)
- Mehmet Esref Alkis
- Department of Occupational Health and Safety, Faculty of Health Sciences, Muş Alparslan University, 49250, Muş, Turkey
| | - Kenan Buldurun
- Department of Food Processing, Technical Science Vocational School, Muş Alparslan University, 49250, Muş, Turkey
| | - Yusuf Alan
- Department of Molecular Biology, Faculty of Arts and Sciences, Muş Alparslan University, 49250, Muş, Turkey
| | - Nevin Turan
- Department of Chemistry, Faculty of Arts and Sciences, Muş Alparslan University, 49250, Muş, Turkey
| | - Ayhan Altun
- Department of Chemistry, Gebze Technical University, 41400, Kocaeli, Turkey
| |
Collapse
|
14
|
Massoud SS, Louka FR, Salem NMH, Fischer RC, Torvisco A, Mautner FA, Vančo J, Belza J, Dvořák Z, Trávníček Z. Dinuclear doubly bridged phenoxido copper(II) complexes as efficient anticancer agents. Eur J Med Chem 2023; 246:114992. [PMID: 36525695 DOI: 10.1016/j.ejmech.2022.114992] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/12/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Two cationic [Cu2(L1-2)2](ClO4)2 (1, 2), and four neutral doubly bridged-phenoxido-copper(II) complexes [Cu2(L3-4)2] (3, 4) and [Cu2(L5-6)2(H2O)]‧2H2O (5, 6) as well as 1D polymeric catena-[Cu(L7)] (7), where HL1-2 and H2L3-7 represent tripodal tetradentate pyridyl or aliphatic-amino groups based 2,4-disubstituted phenolates, were synthesized and thoroughly characterized by various spectroscopic methods and single crystal X-ray analysis. The molecular structures of the complexes exhibited diverse geometrical environments around the central Cu(II) atoms. The in vitro antiproliferative activity of the isolated complexes and selected parent free ligands were screened against some human cancer cell lines (A2780, A2780R, PC-3, 22Rv1, MCF-7). The most promising cytotoxicity against cancer cells were obtained for 1-6, while complex 6 was found as the best performing as compared to the reference drug cisplatin. The cytotoxicity study of complex 6 was therefore extended to wider variety of cancer cell lines (HOS, A549, PANC-1, CaCo2, HeLa) and results revealed its significant cytotoxicity on all investigated human cancer cells. The cell uptake study showed that cytotoxicity of 6 (3 μM concentration and 24 h of incubation) against A2780 cells was almost independent from the intracellular levels of copper. The effect of complexes 4, 6 and 7 on cell cycle of A2780 cells indicates that the mechanism of action in these complexes is not only different from that of cisplatin but also different among them. Complex 7 was able to induce apoptosis in A2780 cells, while complexes 4 and 6 did not and on the other hand, they showed considerable effect on autophagy induction and there are some clues that these complexes were able to induce cuproptosis in A2780 cells.
Collapse
Affiliation(s)
- Salah S Massoud
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 43700, Lafayette, LA, 70504, USA; Department of Chemistry, Faculty of Science, Alexandria University, Moharam Bey, 21511, Alexandria, Egypt.
| | - Febee R Louka
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 43700, Lafayette, LA, 70504, USA
| | - Nahed M H Salem
- Department of Chemistry, Faculty of Science, Alexandria University, Moharam Bey, 21511, Alexandria, Egypt
| | - Roland C Fischer
- Institut für Anorganische Chemische, Technische Universität Graz, Stremayrgasse 9/V, A-8010, Graz, Austria
| | - Ana Torvisco
- Institut für Anorganische Chemische, Technische Universität Graz, Stremayrgasse 9/V, A-8010, Graz, Austria
| | - Franz A Mautner
- Institut für Physikalische and Theoretische Chemie, Technische Universität Graz, Stremayrgasse 9/II, A-8010, Graz, Austria.
| | - Ján Vančo
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Křížkovského 511/8, CZ-779 00, Olomouc, Czech Republic
| | - Jan Belza
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Křížkovského 511/8, CZ-779 00, Olomouc, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic
| | - Zdeněk Trávníček
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Křížkovského 511/8, CZ-779 00, Olomouc, Czech Republic.
| |
Collapse
|
15
|
Fathy A, Ibrahim AB, Elkhalik SA, Villinger A, Abbas S. New iron(III) complexes with 2-formylpyridine thiosemicarbazones: Synthetic aspects, structural and spectral analyses and cytotoxicity screening against MCF-7 human cancer cells. Heliyon 2023; 9:e13008. [PMID: 36711299 PMCID: PMC9880397 DOI: 10.1016/j.heliyon.2023.e13008] [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: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
2-Formylpyridine thiosemicarbazone - iron (III) chelates [F e L 2 ] C l • 2 H 2 O {L = L1 (C1) [HL 1 = 4-(4-Nitrophenyl)-1-((pyridin-2-yl)methylene)thiosemicarbazide] and L = L2 (C2) [HL 2 = 4-(2,5-Dimethoxyphenyl)-1-((pyridin-2-yl)methylene)thiosemicarbazide]} were prepared. The two ligand anions in each complex resulted in saturation of the iron coordination number and consequently the existence of these complexes as 1:1 electrolytes. As well, the iron in these complexes exhibits low-spin electronic configuration. X-ray crystallography of complex C1 indicated its triclinic crystal system and P1 ‾ space group. In addition, it proved the ligation through a thiol sulfur atom and two nitrogen atoms of pyridine and azomethine groups. This is while the presence of two water molecules of crystallization in the complex structure was also indicated. The ligand HL 1 was selected for cytotoxicity screening against human MCF-7, A-549, HEPG-2 and HCT-116 cancer cells and the most enhanced activities were detected against the breast cells. Against these cells, the compounds HL 1 , HL 2 , C1 and C2 induced cytotoxicity, respectively, with IC50 values of 52.4, 145.4, 34.3 and 62.0 μM. However, against the healthy BHK cells, HL 1 and HL 2 caused cytotoxicity, respectively, with IC50 values of 54.8 and 110.6 μM and cytotoxicity with percent viabilities of 56.7 and 55.4% of the BHK cells by the complexes (137.4 μM of C1 and 131.9 μM of C2) was determined. These activities against MCF-7 cells are less significant compared with the measured value for doxorubicin. But this standard is more toxic to normal cells than the thiosemicarbazones (IC50 (doxorubicin) = 9.66 μM against MCF-7 cells and 36.42 μM against BHK cells).
Collapse
Affiliation(s)
- Amany Fathy
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Ahmed B.M. Ibrahim
- Department of Chemistry, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - S. Abd Elkhalik
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Alexander Villinger
- Institut für Chemie, Universität Rostock, Albert-Einstein-Str. 3a, 18059 Rostock, Germany
| | - S.M. Abbas
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| |
Collapse
|
16
|
New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int J Mol Sci 2022; 23:ijms232213990. [PMID: 36430469 PMCID: PMC9696688 DOI: 10.3390/ijms232213990] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
There is new and increasing evidence from in vitro, in vivo and clinical studies implicating the pivotal role of iron and associated metabolic pathways in the initiation, progression and development of cancer and in cancer metastasis. New metabolic and toxicity mechanisms and pathways, as well as genomic, transcription and other factors, have been linked to cancer and many are related to iron. Accordingly, a number of new targets for iron chelators have been identified and characterized in new anticancer strategies, in addition to the classical restriction of/reduction in iron supply, the inhibition of transferrin iron delivery, the inhibition of ribonucleotide reductase in DNA synthesis and high antioxidant potential. The new targets include the removal of excess iron from iron-laden macrophages, which affects anticancer activity; the modulation of ferroptosis; ferritin iron removal and the control of hyperferritinemia; the inhibition of hypoxia related to the role of hypoxia-inducible factor (HIF); modulation of the function of new molecular species such as STEAP4 metalloreductase and the metastasis suppressor N-MYC downstream-regulated gene-1 (NDRG1); modulation of the metabolic pathways of oxidative stress damage affecting mitochondrial function, etc. Many of these new, but also previously known associated iron metabolic pathways appear to affect all stages of cancer, as well as metastasis and drug resistance. Iron-chelating drugs and especially deferiprone (L1), has been shown in many recent studies to fulfill the role of multi-target anticancer drug linked to the above and also other iron targets, and has been proposed for phase II trials in cancer patients. In contrast, lipophilic chelators and their iron complexes are proposed for the induction of ferroptosis in some refractory or recurring tumors in drug resistance and metastasis where effective treatments are absent. There is a need to readdress cancer therapy and include therapeutic strategies targeting multifactorial processes, including the application of multi-targeting drugs involving iron chelators and iron-chelator complexes. New therapeutic protocols including drug combinations with L1 and other chelating drugs could increase anticancer activity, decrease drug resistance and metastasis, improve treatments, reduce toxicity and increase overall survival in cancer patients.
Collapse
|
17
|
Heuberger DM, Wolint P, Jang JH, Itani S, Jungraithmayr W, Waschkies CF, Meier-Bürgisser G, Andreoli S, Spanaus K, Schuepbach RA, Calcagni M, Fahrni CJ, Buschmann J. High-Affinity Cu(I)-Chelator with Potential Anti-Tumorigenic Action-A Proof-of-Principle Experimental Study of Human H460 Tumors in the CAM Assay. Cancers (Basel) 2022; 14:cancers14205122. [PMID: 36291910 PMCID: PMC9600560 DOI: 10.3390/cancers14205122] [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: 09/14/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022] Open
Abstract
Human lung cancer ranks among the most frequently treated cancers worldwide. As copper appears critical to angiogenesis and tumor growth, selective removal of copper represents a promising strategy to restrict tumor growth. To this end, we explored the activity of the novel high-affinity membrane-permeant Cu(I) chelator PSP-2 featuring a low-zeptomolar dissociation constant. Using H460 human lung cancer cells, we generated small tumors on the chorioallantoic membrane of the chicken embryo (CAM assay) and studied the effects of topical PSP-2 application on their weight and vessel density after one week. We observed a significant angiosuppression along with a marked decrease in tumor weight under PSP-2 application compared to controls. Moreover, PSP-2 exposure resulted in lower ki67+ cell numbers at a low dose but increased cell count under a high dose. Moreover, HIF-1α+ cells were significantly reduced with low-dose PSP-2 exposure compared to high-dose and control. The total copper content was considerably lower in PSP-2 treated tumors, although statistically not significant. Altogether, PSP-2 shows promising potential as an anti-cancer drug. Nevertheless, further animal experiments and application to different tumor types are mandatory to support these initial findings, paving the way toward clinical trials.
Collapse
Affiliation(s)
- Dorothea M. Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Petra Wolint
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Jae-Hwi Jang
- Division of Thoracic Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Saria Itani
- Division of Thoracic Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Wolfgang Jungraithmayr
- Division of Thoracic Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
- Department of Thoracic Surgery, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Conny F. Waschkies
- Division of Radiation Protection, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Gabriella Meier-Bürgisser
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Stefano Andreoli
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Katharina Spanaus
- Clinical Chemistry, University Hospital Zurich, 8001 Zurich, Switzerland
| | - Reto A. Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Maurizio Calcagni
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
| | - Christoph J. Fahrni
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332-0400, USA
| | - Johanna Buschmann
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland
- Correspondence: ; Tel.: +41-442559895
| |
Collapse
|
18
|
Glycoconjugation of Quinoline Derivatives Using the C-6 Position in Sugars as a Strategy for Improving the Selectivity and Cytotoxicity of Functionalized Compounds. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27206918. [PMID: 36296513 PMCID: PMC9607644 DOI: 10.3390/molecules27206918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022]
Abstract
Based on the Warburg effect and the increased demand for glucose by tumor cells, a targeted drug delivery strategy was developed. A series of new glycoconjugates with increased ability to interact with GLUT transporters, responsible for the transport of sugars to cancer cells, were synthesized. Glycoconjugation was performed using the C-6 position in the sugar unit, as the least involved in the formation of hydrogen bonds with various aminoacids residues of the transporter. The carbohydrate moiety was connected with the 8-hydroxyquinoline scaffold via a 1,2,3-triazole linker. For the obtained compounds, several in vitro biological tests were performed using HCT-116 and MCF-7 cancer cells as well as NHDF-Neo healthy cells. The highest cytotoxicity of both cancer cell lines in the MTT test was noted for glycoconjugates in which the triazole-quinoline was attached through the triazole nitrogen atom to the d-glucose unit directly to the carbon at the C-6 position. These compounds were more selective than the analogous glycoconjugates formed by the C-1 anomeric position of d-glucose. Experiments with an EDG inhibitor have shown that GLUTs can be involved in the transport of glycoconjugates. The results of apoptosis and cell cycle analyses by flow cytometry confirmed that the new type of glycoconjugates shows pro-apoptotic properties, without significantly affecting changes in the distribution of the cell cycle. Moreover, glycoconjugates were able to decrease the clonogenic potential of cancer cells, inhibit the migration capacity of cells and intercalate with DNA.
Collapse
|
19
|
Sajjad A, Bhatti SH, Zia M. Photo excitation of silver ions during the synthesis of silver nanoparticles modify physiological, chemical, and biological properties. PARTICULATE SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1080/02726351.2022.2126340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Anila Sajjad
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | | | - Muhammad Zia
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
20
|
Yang T, Zhu M, Jiang M, Yang F, Zhang Z. Current status of iridium-based complexes against lung cancer. Front Pharmacol 2022; 13:1025544. [PMID: 36210835 PMCID: PMC9538862 DOI: 10.3389/fphar.2022.1025544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022] Open
Abstract
Lung cancer is one of the most common malignant tumors, with the highest mortality rate in the world, and its incidence is second only to breast cancer. It has posed a serious threat to human health. Cisplatin, a metal-based drug, is one of the most widely used chemotherapeutic agents for the treatment of various cancers. However, its clinical efficacy is seriously limited by numerous side effects and drug resistance. This has led to the exploration and development of other transition metal complexes for the treatment of malignant tumors. In recent years, iridium-based complexes have attracted extensive attention due to their potent anticancer activities, limited side effects, unique antitumor mechanisms, and rich optical properties, and are expected to be potential antitumor drugs. In this review, we summarize the recent progress of iridium complexes against lung cancer and introduce their anti-tumor mechanisms, including apoptosis, cycle arrest, inhibition of lung cancer cell migration, induction of immunogenic cell death, etc.
Collapse
Affiliation(s)
- Tongfu Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, China
| | - Ming Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, China
- School of food and biochemical engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, China
- *Correspondence: Zhenlei Zhang,
| |
Collapse
|
21
|
Brogyányi T, Kaplánek R, Kejík Z, Hosnedlová B, Antonyová V, Abramenko N, Veselá K, Martásek P, Vokurka M, Richardson DR, Jakubek M. Azulene hydrazide-hydrazones for selective targeting of pancreatic cancer cells. Biomed Pharmacother 2022; 155:113736. [PMID: 36156366 DOI: 10.1016/j.biopha.2022.113736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/02/2022] Open
Abstract
Dysregulation of iron homeostasis is one of the important processes in the development of many oncological diseases, such as pancreatic cancer. Targeting it with specific agents, such as an iron chelator, are promising therapeutic methods. In this study, we tested the cytotoxicity of novel azulene hydrazide-hydrazone-based chelators against pancreatic cancer cell lines (MIA PaCa-2, PANC-1, AsPC-1). All prepared chelators (compounds 4-6) showed strong cytotoxicity against pancreatic cancer cell lines and high selectivity for cancer cell lines compared to the healthy line. Their cytotoxicity is lower than thiosemicarbazone-based chelators Dp44mT and DpC, but significantly higher than hydroxamic acid-based chelator DFO. The chelator tested showed mitochondrial and lysosomal co-localization and its mechanism of action was based on the induction of hypoxia-inducible factor-1-alpha (HIF-1α), N-myc downstream-regulated gene-1 (NDRG1) and transferrin receptor 1 (TfR1). This strongly implies that the cytotoxic effect of tested chelators could be associated with mitophagy induction. Lipinski's rule of five analyses was performed to determine whether the prepared compounds had properties ensuring their bioavailability. In addition, the drug-likeness and drug-score were calculated and discussed.
Collapse
Affiliation(s)
- Tereza Brogyányi
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 5, 128 53 Prague, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic
| | - Božena Hosnedlová
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic
| | - Veronika Antonyová
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic
| | - Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 5, 128 53 Prague, Czech Republic
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD, Australia
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00 Prague, Czech Republic.
| |
Collapse
|
22
|
Murillo MI, Gaiddon C, Le Lagadec R. Targeting of the intracellular redox balance by metal complexes towards anticancer therapy. Front Chem 2022; 10:967337. [PMID: 36034648 PMCID: PMC9405673 DOI: 10.3389/fchem.2022.967337] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The development of cancers is often linked to the alteration of essential redox processes, and therefore, oxidoreductases involved in such mechanisms can be considered as attractive molecular targets for the development of new therapeutic strategies. On the other hand, for more than two decades, transition metals derivatives have been leading the research on drugs as alternatives to platinum-based treatments. The success of such compounds is particularly due to their attractive redox kinetics properties, favorable oxidation states, as well as routes of action different to interactions with DNA, in which redox interactions are crucial. For instance, the activity of oxidoreductases such as PHD2 (prolyl hydroxylase domain-containing protein) which can regulate angiogenesis in tumors, LDH (lactate dehydrogenase) related to glycolysis, and enzymes, such as catalases, SOD (superoxide dismutase), TRX (thioredoxin) or GSH (glutathione) involved in controlling oxidative stress, can be altered by metal effectors. In this review, we wish to discuss recent results on how transition metal complexes have been rationally designed to impact on redox processes, in search for effective and more specific cancer treatments.
Collapse
Affiliation(s)
- María Isabel Murillo
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Christian Gaiddon
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
- *Correspondence: Ronan Le Lagadec,
| |
Collapse
|
23
|
Pandey P, Khan F, Qari HA, Upadhyay TK, Alkhateeb AF, Oves M. Evidence of Metallic and Polyether Ionophores as Potent Therapeutic Drug Candidate in Cancer Management. Molecules 2022; 27:4708. [PMID: 35897885 PMCID: PMC9329979 DOI: 10.3390/molecules27154708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer remains one of the most crucial human malignancies with a higher mortality rate globally, and is predicted to escalate soon. Dysregulated ion homeostasis in cancerous cells prompted the researchers to investigate further ion homeostasis impeding agents as potent anticancerous agents. Reutilization of FDA-approved non-cancerous drugs has emerged as a practical approach to developing potent, cost-effective drugs for cancer treatment. Across the globe, most nations are incapable of fulfilling the medical demands of cancer patients due to costlier cancerous drugs. Therefore, we have inclined our review towards emphasizing recent advancements in cancer therapies involving ionophores utilization in exploring potent anticancer drugs. Numerous research reports have established the significant anticancerous potential of ionophores in several pre-clinical reports via modulating aberrant cell signaling pathways and enhancing antitumor immunity in immune cells. This review has mainly summarized the most significant ion homeostasis impeding agents, including copper, zinc, calcium, and polyether, that presented remarkable potential in cancer therapeutics via enhanced antitumor immunity and apoptosis induction. Altogether, this study could provide a robust future perspective for developing cost-effective anticancerous drugs rapidly and cost-effectively, thereby combating the limitations of currently available drugs used in cancer treatment.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida 201306, India;
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida 201306, India;
| | - Huda A. Qari
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India;
| | - Abdulhameed F. Alkhateeb
- Department of Electrical & Computer Engineering, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad Oves
- Center of Excellence in Environmental Studies, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Sendai 980-8577, Japan
| |
Collapse
|
24
|
Rahimifard M, Jalalimanesh N, Movahed MA, Hadjighassem M, Pourahmad Jaktaji R, Bagheri Z, Pourahmad J, Zarghi A. Antiproliferative activity of new derivatives of pyrazino[1,2‐
a
]benzimidazole: Integrated cell‐based assay and computational studies with divalent magnesium, iron, and copper ions. J Biochem Mol Toxicol 2022; 36:e23155. [DOI: 10.1002/jbt.23155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 05/31/2022] [Accepted: 06/27/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Mahban Rahimifard
- Pharmaceutical Sciences Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | | | - Mahsa A. Movahed
- Department of Medicinal and Pharmaceutical Chemistry Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute Tehran University of Medical Sciences Tehran Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| | | | - Zeinab Bagheri
- Faculty of Life Sciences and Biotechnology Shahid Beheshti University G.C. Tehran Iran
| | - Jalal Pourahmad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Afshin Zarghi
- Department of Medicinal and Pharmaceutical Chemistry Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
25
|
Synthesis and Preliminary Evaluation of the Cytotoxicity of Potential Metabolites of Quinoline Glycoconjugates. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27031040. [PMID: 35164304 PMCID: PMC8838273 DOI: 10.3390/molecules27031040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
Abstract
The design of prodrugs is one of the important strategies for selective anti-cancer therapies. When designing prodrugs, attention is paid to the possibility of their targeting tumor-specific markers such as proteins responsible for glucose uptake. That is why glycoconjugation of biologically active compounds is a frequently used strategy. Glycoconjugates consisting of three basic building blocks: a sugar unit, a linker containing a 1,2,3-triazole ring, and an 8-hydroxyquinoline fragment was described earlier. It is not known whether their cytotoxicity is due to whole glycoconjugates action or their metabolites. To check the biological activity of products that can be released from glycoconjugates under the action of hydrolytic enzymes, the synthetically obtained potential metabolites were tested in vitro for the inhibition of proliferation of HCT-116, MCF-7, and NHDF-Neo cell lines using the MTT assay. Research shows that for the full activity of glycoconjugates, the presence of all three building blocks in the structure of a potential drug is necessary. For selected derivatives, additional tests of targeted drug delivery to tumor cells were carried out using polymer nanocarriers in which they are encapsulated. This approach significantly lowered the determined IC50 values of the tested compounds and improved their selectivity and effectiveness.
Collapse
|
26
|
Domiński A, Domińska M, Skonieczna M, Pastuch-Gawołek G, Kurcok P. Shell-Sheddable Micelles Based on Poly(ethylene glycol)-hydrazone-poly[R,S]-3-hydroxybutyrate Copolymer Loaded with 8-Hydroxyquinoline Glycoconjugates as a Dual Tumor-Targeting Drug Delivery System. Pharmaceutics 2022; 14:290. [PMID: 35214023 PMCID: PMC8877687 DOI: 10.3390/pharmaceutics14020290] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 02/05/2023] Open
Abstract
The development of selective delivery of anticancer drugs into tumor tissues to avoid systemic toxicity is a crucial challenge in cancer therapy. In this context, we evaluated the efficacy of a combination of nanocarrier pH-sensitivity and glycoconjugation of encapsulated drugs, since both vectors take advantage of the tumor-specific Warburg effect. Herein, we synthesized biodegradable diblock copolymer, a poly(ethylene glycol)-hydrazone linkage-poly[R,S]-3-hydroxybutyrate, which could further self-assemble into micelles with a diameter of ~55 nm. The hydrazone bond was incorporated between two copolymer blocks under an acidic pH, causing the shell-shedding of micelles which results in the drug's release. The micelles were stable at pH 7.4, but decompose in acidic pH, as stated by DLS studies. The copolymer was used as a nanocarrier for 8-hydroxyquinoline glucose and galactose conjugates as well as doxorubicin, and exhibited pH-dependent drug release behavior. In vitro cytotoxicity, apoptosis, and life cycle assays studies of blank and drug-loaded micelles were performed on Normal Human Dermal Fibroblasts-Neonatal (NHDF-Neo), colon carcinoma (HCT-116), and breast cancer (MCF-7) for 24, 48, and 72 h. A lack of toxicity of blank micelles was demonstrated, whereas the glycoconjugates-loaded micelles revealed enhanced selectivity to inhibit the proliferation of cancer cells. The strategy of combining pH-responsive nanocarriers with glycoconjugation of the drug molecule provides an alternative to the modus operandi of designing multi-stimuli nanocarriers to increase the selectivity of anticancer therapy.
Collapse
Affiliation(s)
- Adrian Domiński
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowskiej St., 41-819 Zabrze, Poland
| | - Monika Domińska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland; (M.D.); (G.P.-G.)
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland;
| | - Magdalena Skonieczna
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland;
- Department of Systems Biology and Engineering, Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland; (M.D.); (G.P.-G.)
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland;
| | - Piotr Kurcok
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowskiej St., 41-819 Zabrze, Poland
| |
Collapse
|
27
|
Lee D, Ha J, Kang M, Yang Z, Jiang W, Kim BYS. Strategies of Perturbing Ion Homeostasis for Cancer Therapy. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - JongHoon Ha
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Minjeong Kang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Zhaogang Yang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Wen Jiang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Betty Y. S. Kim
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
28
|
Xi Y, Yan X, Bigdeli F, Zhang Q, Esrafili L, Hanifehpour Y, Zhang W, Hu M, Morsali A. Two new Cu (II) complexes based on 5‐fluorouracil‐1‐yl acetic acid and N‐donor ligands: Investigation of their interaction with DNA and anticancer activity. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Yun‐Hong Xi
- College of Chemistry and Materials Engineering Wenzhou University Wenzhou China
| | - Xin Yan
- College of Chemistry and Materials Engineering Wenzhou University Wenzhou China
| | - Fahime Bigdeli
- Department of Chemistry, Faculty of Sciences Tarbiat Modares University Tehran Iran
| | - Qianwen Zhang
- Department of Dermatology The First Affiliated Hospital of Wenzhou Medical University Wenzhou People's Republic of China
| | - Leili Esrafili
- Department of Chemistry, Faculty of Sciences Tarbiat Modares University Tehran Iran
| | - Younes Hanifehpour
- Faculty of Science, Department of Chemistry Sayyed Jamaleddin Asadabadi University Asadabad Iran
| | - Wei‐Bing Zhang
- College of Chemistry and Materials Engineering Wenzhou University Wenzhou China
| | - Mao‐Lin Hu
- College of Chemistry and Materials Engineering Wenzhou University Wenzhou China
| | - Ali Morsali
- Department of Chemistry, Faculty of Sciences Tarbiat Modares University Tehran Iran
| |
Collapse
|
29
|
O'Sullivan JJ, Medici V, Heffern MC. A caged imidazopyrazinone for selective bioluminescence detection of labile extracellular copper( ii). Chem Sci 2022; 13:4352-4363. [PMID: 35509459 PMCID: PMC9006956 DOI: 10.1039/d1sc07177g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
Copper is an essential redox-active metal that plays integral roles in biology ranging from enzymatic catalysis to mitochondrial respiration. However, if not adequately regulated, this redox activity has the potential to cause oxidative stress through the production of reactive oxygen species. Indeed, the dysregulation of copper has been associated with a variety of disease states including diabetes, neurodegenerative disorders, and multiple cancers. While increasing tools are being developed for illuminating labile intracellular copper pools and the trafficking pathways in which they are involved, significantly less attention has been given to the analogous extracellular labile pool. To address this gap, we have developed a bioluminescence-based imaging probe, picolinic ester caged-diphenylterazine (pic-DTZ) for monitoring labile, extracellular copper using a coelenterazine-like imidazopyrazinone and the genetically-engineered, marine-based luciferase, nanoluciferase. Unlike the more commonly-used firefly luciferase, nanoluciferase does not require ATP, allowing its application to the extracellular milieu. pic-DTZ demonstrates high metal and oxidation state selectivity for Cu(ii) in aqueous buffer as well as selectivity for labile pools over coordinatively inaccessible protein-bound Cu(ii). We demonstrate the potential of pic-DTZ as a diagnostic tool in human serum and plasma for copper-associated diseases. Additionally, we apply pic-DTZ to lend insight into the extracellular copper dynamic in anticancer treatments. A caged imidazopyrazinone can detect reactive extracellular copper in mammalian systems using bioluminescence generated with the ATP-independent nanoluciferase enzyme.![]()
Collapse
Affiliation(s)
- Justin J. O'Sullivan
- Department of Chemistry, University of California Davis, One Shields Drive, Davis, CA 95616, USA
| | - Valentina Medici
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, PSSB Suite 3500, Sacramento, CA 95817, USA
| | - Marie C. Heffern
- Department of Chemistry, University of California Davis, One Shields Drive, Davis, CA 95616, USA
| |
Collapse
|
30
|
The ion channel TRPM7 regulates zinc-depletion-induced MDMX degradation. J Biol Chem 2021; 297:101292. [PMID: 34627839 PMCID: PMC8561006 DOI: 10.1016/j.jbc.2021.101292] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/22/2022] Open
Abstract
Zinc deficiency has been linked to human diseases, including cancer. MDMX, a crucial zinc-containing negative regulator of p53, has been found to be amplified or overexpressed in various cancers and implicated in the cancer initiation and progression. We report here that zinc depletion by the ion chelator TPEN or Chelex resin results in MDMX protein degradation in a ubiquitination-independent and 20S proteasome-dependent manner. Restoration of zinc led to recovery of cellular levels of MDMX. Further, TPEN treatment inhibits growth of the MCF-7 breast cancer cell line, which is partially rescued by overexpression of MDMX. Moreover, in a mass-spectrometry-based proteomics analysis, we identified TRPM7, a zinc-permeable ion channel, as a novel MDMX-interacting protein. TRPM7 stabilizes and induces the appearance of faster migrating species of MDMX on SDS-PAGE. Depletion of TRPM7 attenuates, while TRPM7 overexpression facilitates, the recovery of MDMX levels upon adding back zinc to TPEN-treated cells. Importantly, we found that TRPM7 inhibition, like TPEN treatment, decreases breast cancer cell MCF-7 proliferation and migration. The inhibitory effect on cell migration upon TRPM7 inhibition is also partially rescued by overexpression of MDMX. Together, our data indicate that TRPM7 regulates cellular levels of MDMX in part by modulating the intracellular Zn2+ concentration to promote tumorigenesis.
Collapse
|
31
|
Massoud SS, Louka FR, Dial MT, Malek AJ, Fischer RC, Mautner FA, Vančo J, Malina T, Dvořák Z, Trávníček Z. Identification of potent anticancer copper(ii) complexes containing tripodal bis[2-ethyl-di(3,5-dialkyl-1H-pyrazol-1-yl)]amine moiety. Dalton Trans 2021; 50:11521-11534. [PMID: 34346447 DOI: 10.1039/d1dt01724a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of heteroleptic copper(ii) complexes of the composition [Cu(L1-5)Cl]X, where X = ClO4 and/or PF6 and [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl))-(6-methyl-(2-pyridylmethyl))]amine (L1), [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl))-(3,4-dimethoxy-(2-pyridylmethyl))]amine (L2), [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl)-(2-quinolymethyl)]amine (L3), [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazolyl)-(di(3,5-dimethyl-1H-pyrazol-1-yl-methyl))]amine (L4) and [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl)-(5-methyl-3-phenyl-1H-pyrazol-1-yl-methyl)]amine (L5), were prepared and thoroughly characterized including single-crystal X-ray diffraction technique. The in vitro cytotoxicity of complexes against A2780, A2780R, HOS and MCF-7 human cancer cell lines was evaluated using the MTT test. The results revealed that complexes [Cu(L1)Cl]PF6 (1-PF6), [Cu(L2)Cl]ClO4 (2-ClO4) and [Cu(L3)Cl]PF6 (3-PF6) are the most effective, with IC50 values ranging from 1.4 to 6.3 μM, thus exceeding the cytotoxic potential of metallodrug cisplatin (IC50 values ranging from 29.9 to 82.0 μM). The complexes [Cu(L4)Cl]PF6 (4-PF6) and [Cu(L5)Cl]PF6 (5-PF6) showed only moderate cytotoxicity against A2780, with IC50 = 53.6 μM, and 33.8 μM, respectively. The cell cycle profile, time-resolved cellular uptake, interactions with small sulfur-containing biomolecules (cysteine and glutathione), intracellular ROS production, induction of apoptosis and activation of caspases 3/7 were also evaluated in the case of the selected complexes. It has been found that the best performing complexes 1 and 2 cause cell arrest in the G2/M phase and induce apoptosis via the increase in production of ROS, dominantly due to the overproduction of superoxide.
Collapse
Affiliation(s)
- Salah S Massoud
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 43700, Lafayette, LA 70504, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Harismah K, Hajali N, Zandi H. 6-Thioguanine bimolecular formation for dual chelation of iron: DFT study. COMPUT THEOR CHEM 2021. [DOI: 10.1016/j.comptc.2021.113308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Gaur K, Pérez Otero SC, Benjamín-Rivera JA, Rodríguez I, Loza-Rosas SA, Vázquez Salgado AM, Akam EA, Hernández-Matias L, Sharma RK, Alicea N, Kowaleff M, Washington AV, Astashkin AV, Tomat E, Tinoco AD. Iron Chelator Transmetalative Approach to Inhibit Human Ribonucleotide Reductase. JACS AU 2021; 1:865-878. [PMID: 34240081 PMCID: PMC8243325 DOI: 10.1021/jacsau.1c00078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Indexed: 05/04/2023]
Abstract
Efforts directed at curtailing the bioavailability of intracellular iron could lead to the development of broad-spectrum anticancer drugs given the metal's role in cancer proliferation and metastasis. Human ribonucleotide reductase (RNR), the key enzyme responsible for synthesizing the building blocks of DNA replication and repair, depends on Fe binding at its R2 subunit to activate the catalytic R1 subunit. This work explores an intracellular iron chelator transmetalative approach to inhibit RNR using the titanium(IV) chemical transferrin mimetic (cTfm) compounds Ti(HBED) and Ti(Deferasirox)2. Whole-cell EPR studies reveal that the compounds can effectively attenuate RNR activity though seemingly causing different changes to the labile iron pool that may account for differences in their potency against cells. Studies of Ti(IV) interactions with the adenosine nucleotide family at pH 7.4 reveal strong metal binding and extensive phosphate hydrolysis, which suggest the capacity of the metal to disturb the nucleotide substrate pool of the RNR enzyme. By decreasing intracellular Fe bioavailability and altering the nucleotide substrate pool, the Ti cTfm compounds could inhibit the activity of the R1 and R2 subunits of RNR. The compounds arrest the cell cycle in the S phase, indicating suppressed DNA replication, and induce apoptotic cell death. Cotreatment cell viability studies with cisplatin and Ti(Deferasirox)2 reveal a promising synergism between the compounds that is likely owed to their distinct but complementary effect on DNA replication.
Collapse
Affiliation(s)
- Kavita Gaur
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Sofia C. Pérez Otero
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Josué A. Benjamín-Rivera
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Israel Rodríguez
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Sergio A. Loza-Rosas
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | | | - Eman A. Akam
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| | - Liz Hernández-Matias
- Department
of Biology, University of Puerto Rico Río
Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Rohit K. Sharma
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Nahiara Alicea
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Martin Kowaleff
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Anthony V. Washington
- Department
of Biology, University of Puerto Rico Río
Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Andrei V. Astashkin
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| | - Elisa Tomat
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| | - Arthur D. Tinoco
- Department
of Chemistry, University of Puerto Rico
Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| |
Collapse
|
34
|
Steinbrueck A, Sedgwick AC, Han HH, Zhao MY, Sen S, Huang DY, Zang Y, Li J, He XP, Sessler JL. In vitro studies of deferasirox derivatives as potential organelle-targeting traceable anti-cancer therapeutics. Chem Commun (Camb) 2021; 57:5678-5681. [PMID: 33977921 PMCID: PMC8456774 DOI: 10.1039/d0cc08156f] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report here strategic functionalization of the FDA approved chelator deferasirox (1) in an effort to produce organelle-targeting iron chelators with enhanced activity against A549 lung cancer cells. Derivative 8 was found to have improved antiproliferative activity relative to 1. Fluorescent cell imaging revealed that compound 8 preferentially localises within the lysosome.
Collapse
Affiliation(s)
- Axel Steinbrueck
- Department of Chemistry, University of Texas at Austin, 105 E 24th street A5300, Austin, TX 78712-1224, USA.
| | - Adam C Sedgwick
- Department of Chemistry, University of Texas at Austin, 105 E 24th street A5300, Austin, TX 78712-1224, USA.
| | - Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China.
| | - Michael Y Zhao
- Department of Chemistry, University of Texas at Austin, 105 E 24th street A5300, Austin, TX 78712-1224, USA.
| | - Sajal Sen
- Department of Chemistry, University of Texas at Austin, 105 E 24th street A5300, Austin, TX 78712-1224, USA.
| | - Dan-Ying Huang
- Department of Chemistry, University of Texas at Austin, 105 E 24th street A5300, Austin, TX 78712-1224, USA.
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China.
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China.
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China.
| | - Jonathan L Sessler
- Department of Chemistry, University of Texas at Austin, 105 E 24th street A5300, Austin, TX 78712-1224, USA.
| |
Collapse
|
35
|
Effect of the complex-formation ability of thiosemicarbazones containing (aza)benzene or 3-nitro-1,8-naphthalimide unit towards Cu(II) and Fe(III) ions on their anticancer activity. J Photochem Photobiol A Chem 2021. [DOI: 10.1016/j.jphotochem.2021.113314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
36
|
Nahari G, Hoffman RE, Tshuva EY. From medium to endoplasmic reticulum: Tracing anticancer phenolato titanium(IV) complex by 19F NMR detection. J Inorg Biochem 2021; 221:111492. [PMID: 34051630 DOI: 10.1016/j.jinorgbio.2021.111492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/22/2021] [Accepted: 05/12/2021] [Indexed: 01/12/2023]
Abstract
Titanium(IV) complexes of diaminobis(phenolato)-bis(alkoxo) ligands are promising anticancer drugs, showing marked in-vivo efficacy with no toxic side-effects in mice, hence, it is of interest to elucidate their mechanism of action. Herein, we employed a fluoro-substituted derivative, FenolaTi, for mechanistic analysis of the active species and its cellular target by quantitative 19F NMR detection to reveal its biodistribution and reactivity in extracellular and intracellular matrices. Upon administration to the serum-containing medium, FenolaTi interacted with bovine serum albumin. 20 h post administration, the cellular accumulation of FenolaTi derivatives was estimated as 37% of the administered compound, in a concentration three orders-of-magnitude higher than the administered dose, implying that active membrane transportation facilitates cellular penetration. An additional 19% of the administered dose that was detected in the extracellular environment had originated from post-apoptotic cells. In the cell, interaction with cellular proteins was detected. Although some intact Ti(IV) complex localized in the nucleus, no signals for isolated DNA fractions were detected and no reactivity with nuclear proteins was observed. Interestingly, higher accumulation of FenolaTi-derived compounds in the endoplasmic reticulum (ER) and interaction with proteins therein were detected, supporting the role of the ER as a possible target for cytotoxic bis(phenolato)-bis(alkoxo) Ti(IV) complexes.
Collapse
Affiliation(s)
- Gilad Nahari
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Roy E Hoffman
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Edit Y Tshuva
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| |
Collapse
|
37
|
Liang P, Ballou B, Lv X, Si W, Bruchez MP, Huang W, Dong X. Monotherapy and Combination Therapy Using Anti-Angiogenic Nanoagents to Fight Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005155. [PMID: 33684242 DOI: 10.1002/adma.202005155] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/10/2020] [Indexed: 06/12/2023]
Abstract
Anti-angiogenic therapy, targeting vascular endothelial cells (ECs) to prevent tumor growth, has been attracting increasing attention in recent years, beginning with bevacizumab (Avastin) through its Phase II/III clinical trials on solid tumors. However, these trials showed only modest clinical efficiency; moreover, anti-angiogenic therapy may induce acquired resistance to the drugs employed. Combining advanced drug delivery techniques (e.g., nanotechnology) or other therapeutic strategies (e.g., chemotherapy, radiotherapy, phototherapy, and immunotherapy) with anti-angiogenic therapy results in significantly synergistic effects and has opened a new horizon in fighting cancer. Herein, clinical difficulties in using traditional anti-angiogenic therapy are discussed. Then, several promising applications of anti-angiogenic nanoagents in monotherapies and combination therapies are highlighted. Finally, the challenges and perspectives of anti-angiogenic cancer therapy are summarized. A useful introduction to anti-angiogenic strategies, which may significantly improve therapeutic outcomes, is thus provided.
Collapse
Affiliation(s)
- Pingping Liang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Byron Ballou
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA, 15213, United States
| | - Xinyi Lv
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Weili Si
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Marcel P Bruchez
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA, 15213, United States
| | - Wei Huang
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
- School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| |
Collapse
|
38
|
Jargalsaikhan G, Wu JY, Chen YC, Yang LL, Wu MS. Comparison of the Phytochemical Properties, Antioxidant Activity and Cytotoxic Effect on HepG2 Cells in Mongolian and Taiwanese Rhubarb Species. Molecules 2021; 26:1217. [PMID: 33668690 PMCID: PMC7956657 DOI: 10.3390/molecules26051217] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 11/30/2022] Open
Abstract
The Mongolian rhubarb-Rheum undulatum L. (RU)-and Rumex crispus L. (RC)-a Taiwanese local rhubarb belonging to the family of Polygonaceae-are principal therapeutic materials in integrative medicine due to their rich quantities of bioactive compounds; however, their phytochemical and antioxidant properties, and anti-cancer activity is poorly investigated. Furthermore, the phytochemical characteristics of both species may be affected by their different geographical distribution and climatic variance. The current study aimed to compare RU with RC extracts in different polarity solvents (n-hexane, ethyl acetate, acetone, ethanol, and water) for their phytochemical contents including the total phenolic content (TPC), total anthraquinone content (TAC), total flavonoid content (TFC), antioxidant and free radical scavenging capacities, and anticancer ability on the HepG2 cell. Except for the n-hexane extract, all of the RU extracts had considerably higher TPCs than RC extracts, ranging from 8.39 to 11.16 mg gallic acid equivalent (GAE) per gram of dry weight, and the TPCs of each extract were also significantly correlated with their antioxidant capacities by ABTS, DPPH, and FRAP assays (p < 0.05). Moreover, there was no remarkable association between the antioxidant capacities and either TACs or TFCs in both the RU and RC extracts. Besides, high-performance liquid chromatography (HPLC) analysis revealed that both the RU and RC extracts contained chrysophanol, emodin, and physcion, and those bioactive compounds were relatively higher in the n-hexane solvent extracts. Additionally, we observed different levels of dose-dependent cytotoxic effects in all the extracts by cell viability assay. Notably, the ethanol extract of RU had a compelling cytotoxic effect with the lowest half-maximum inhibition concentration (IC50-171.94 ± 6.56 µg/mL at 48 h) among the RU extracts than the ethanol extract of RC. Interestingly, the ethanol extract of RU but not RC significantly induced apoptosis in the human liver cancer cell line, HepG2, with a distinct pattern in caspase-3 activation, resulting in increased PARP cleavage and DNA damage. In summary, Mongolian Rhubarb, RU, showed more phytochemical contents, as well as a higher antioxidant capacity and apoptotic effect to HepG2 than RC; thus, it can be exploited for the proper source of natural antioxidants and liver cancer treatment in further investigation.
Collapse
Affiliation(s)
- Ganbolor Jargalsaikhan
- International MS/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (G.J.); (Y.-C.C.)
- Liver Center, Ulaanbaatar 14230, Mongolia
| | - Jin-Yi Wu
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi 60004, Taiwan;
| | - Yen-Chou Chen
- International MS/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (G.J.); (Y.-C.C.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cancer Research Center and Orthopedics Research Center, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Ling-Ling Yang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan;
- American College of Acupuncture and Oriental Medicine, Houston, TX 77063, USA
| | - Ming-Shun Wu
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Integrative Therapy Center for Gastroenterological Cancers, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
39
|
Rodríguez I, Gautam R, Tinoco AD. Using X-ray Diffraction Techniques for Biomimetic Drug Development, Formulation, and Polymorphic Characterization. Biomimetics (Basel) 2020; 6:1. [PMID: 33396786 PMCID: PMC7838816 DOI: 10.3390/biomimetics6010001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 12/31/2022] Open
Abstract
Drug development is a decades-long, multibillion dollar investment that often limits itself. To decrease the time to drug approval, efforts are focused on drug targets and drug formulation for optimal biocompatibility and efficacy. X-ray structural characterization approaches have catalyzed the drug discovery and design process. Single crystal X-ray diffraction (SCXRD) reveals important structural details and molecular interactions for the manifestation of a disease or for therapeutic effect. Powder X-ray diffraction (PXRD) has provided a method to determine the different phases, purity, and stability of biological drug compounds that possess crystallinity. Recently, synchrotron sources have enabled wider access to the study of noncrystalline or amorphous solids. One valuable technique employed to determine atomic arrangements and local atom ordering of amorphous materials is the pair distribution function (PDF). PDF has been used in the study of amorphous solid dispersions (ASDs). ASDs are made up of an active pharmaceutical ingredient (API) within a drug dispersed at the molecular level in an amorphous polymeric carrier. This information is vital for appropriate formulation of a drug for stability, administration, and efficacy purposes. Natural or biomimetic products are often used as the API or the formulation agent. This review profiles the deep insights that X-ray structural techniques and associated analytical methods can offer in the development of a drug.
Collapse
Affiliation(s)
- Israel Rodríguez
- Department of Chemistry, University of Puerto Rico Río Piedras, San Juan, PR 00925, USA
| | - Ritika Gautam
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico Río Piedras, San Juan, PR 00925, USA
| |
Collapse
|
40
|
Planeta K, Setkowicz Z, Janik-Olchawa N, Matusiak K, Ryszawy D, Drozdz A, Janeczko K, Ostachowicz B, Chwiej J. Comparison of Elemental Anomalies Following Implantation of Different Cell Lines of Glioblastoma Multiforme in the Rat Brain: A Total Reflection X-ray Fluorescence Spectroscopy Study. ACS Chem Neurosci 2020; 11:4447-4459. [PMID: 33205959 PMCID: PMC7747222 DOI: 10.1021/acschemneuro.0c00648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor with a very high degree of malignancy and is classified by WHO as a glioma IV. At present, the treatment of patients suffering from GBM is based on surgical resection of the tumor with maximal protection of surrounding tissues followed by radio- and pharmacological therapy using temozolomide as the most frequently recommended drug. This strategy, however, does not guarantee success and has devastating consequences. Testing of new substances or therapies having potential in the treatment of GBM as well as detection of their side effects cannot be done on humans. Animal models of the disease are usually used for these purposes, and one possibility is the implantation of human tumor cells into rodent brains. Such a solution was used in the present study the purpose of which was comparison of elemental anomalies appearing in the brain as a result of implantation of different glioblastoma cell lines. These were two commercially available cell lines (U87MG and T98G), as well as tumor cells taken directly from a patient diagnosed with GBM. Using total reflection X-ray fluorescence we determined the contents of P, S, K, Ca, Fe, Cu, Zn, and Se in implanted-left and intact-right brain hemispheres. The number of elemental anomalies registered for both hemispheres was positively correlated with the invasiveness of GBM cells and was the highest for animals subjected to U87MG cell implantation, which presented significant decrease of P, K, and Cu levels and an increase of Se concentration within the left hemisphere. The abnormality common for all three groups of animals subjected to glioma cell implantation was increased Fe level in the brain, which may result from higher blood supply or the presence of hemorrhaging regions. In the case of the intact hemisphere, elevated Fe concentration may also indicate higher neuronal activity caused by taking over some functions of the left hemisphere impaired as a result of tumor growth.
Collapse
Affiliation(s)
- Karolina Planeta
- AGH
University of Science and Technology, Faculty of Physics and Applied Computer Science, Krakow 30-059, Poland
| | - Zuzanna Setkowicz
- Jagiellonian
University, Institute of Zoology
and Biomedical Research, Krakow 31-007, Poland
| | - Natalia Janik-Olchawa
- AGH
University of Science and Technology, Faculty of Physics and Applied Computer Science, Krakow 30-059, Poland
| | - Katarzyna Matusiak
- AGH
University of Science and Technology, Faculty of Physics and Applied Computer Science, Krakow 30-059, Poland
| | - Damian Ryszawy
- Jagiellonian
University, Faculty of Biochemistry,
Biophysics, and Biotechnology, Krakow 31-007, Poland
| | - Agnieszka Drozdz
- AGH
University of Science and Technology, Faculty of Physics and Applied Computer Science, Krakow 30-059, Poland
| | - Krzysztof Janeczko
- Jagiellonian
University, Institute of Zoology
and Biomedical Research, Krakow 31-007, Poland
| | - Beata Ostachowicz
- AGH
University of Science and Technology, Faculty of Physics and Applied Computer Science, Krakow 30-059, Poland
| | - Joanna Chwiej
- AGH
University of Science and Technology, Faculty of Physics and Applied Computer Science, Krakow 30-059, Poland
| |
Collapse
|
41
|
Quiles JL, Sánchez-González C, Vera-Ramírez L, Giampieri F, Navarro-Hortal MD, Xiao J, Llopis J, Battino M, Varela-López A. Reductive Stress, Bioactive Compounds, Redox-Active Metals, and Dormant Tumor Cell Biology to Develop Redox-Based Tools for the Treatment of Cancer. Antioxid Redox Signal 2020; 33:860-881. [PMID: 32064905 DOI: 10.1089/ars.2020.8051] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Significance: Cancer is related to redox biology from many points of view, such as initiation and promotion, metabolism and growth, invasion and metastasis, vascularization, or through the interaction with the immune system. In addition, this extremely complex relationship depends on the redox homeostasis of each cellular compartment, which might be used to fight cancer. Recent Advances: New ways of modulating specific and little explored aspects of redox biology have been revealed, as well as new delivery methods or uses of previously known treatments against cancer. Here, we review the latest experimental evidence regarding redox biology in cancer treatment and analyze its potential impact in the development of improved and more effective antineoplastic therapies. Critical Issues: A critical issue that deserves particular attention is the understanding that both extremes of redox biology (i.e., oxidative stress [OS] and reductive stress) might be useful or harmful in relation to cancer prevention and treatment. Future Directions: Additional research is needed to understand how to selectively induce reductive or OS adequately to avoid cancer proliferation or to induce cancer cell death.
Collapse
Affiliation(s)
- José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.,College of Food Science and Technology, Northwest University, Xi'an, China
| | - Cristina Sánchez-González
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Laura Vera-Ramírez
- Department of Genomic Medicine, GENYO: Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), Granada, Spain
| | - Francesca Giampieri
- College of Food Science and Technology, Northwest University, Xi'an, China.,Department of Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - M Dolores Navarro-Hortal
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
| | - Juan Llopis
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Maurizio Battino
- Department of Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy.,International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China.,Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, Vigo, Spain
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| |
Collapse
|
42
|
Krawczyk M, Pastuch-Gawołek G, Hadasik A, Erfurt K. 8-Hydroxyquinoline Glycoconjugates Containing Sulfur at the Sugar Anomeric Position-Synthesis and Preliminary Evaluation of Their Cytotoxicity. Molecules 2020; 25:E4174. [PMID: 32933091 PMCID: PMC7570910 DOI: 10.3390/molecules25184174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
One of the main factors limiting the effectiveness of many drugs is the difficulty of their delivery to their target site in the cell and achieving the desired therapeutic dose. Moreover, the accumulation of the drug in healthy tissue can lead to serious side effects. The way to improve the selectivity of a drug to the cancer cells seems to be its conjugation with a sugar molecule, which should facilitate its selective transport through GLUT transporters (glucose transporters), whose overexpression is seen in some types of cancer. This was the idea behind the synthesis of 8-hydroxyquinoline (8-HQ) derivative glycoconjugates, for which 1-thiosugar derivatives were used as sugar moiety donors. It was expected that the introduction of a sulfur atom instead of an oxygen atom into the anomeric position of the sugar would increase the stability of the obtained glycoconjugates against untimely hydrolytic cleavage. The anticancer activity of new compounds was determined based on the results of the MTT cytotoxicity tests. Because of the assumption that the activity of this type of compounds was based on metal ion chelation, the effect of the addition of copper ions on cell proliferation was tested for some of them. It turned out that cancer cells treated with glycoconjugates in the presence of Cu2+ had a much slower growth rate compared to cells treated with free glycoconjugates in the absence of copper. The highest cytotoxic activity of the compounds was observed against the MCF-7 cell line.
Collapse
Affiliation(s)
- Monika Krawczyk
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; (G.P.-G.); (A.H.)
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; (G.P.-G.); (A.H.)
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Agnieszka Hadasik
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; (G.P.-G.); (A.H.)
| | - Karol Erfurt
- Department of Chemical Organic Technology and Petrochemistry, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland;
| |
Collapse
|
43
|
Benjamín-Rivera JA, Cardona-Rivera AE, Vázquez-Maldonado ÁL, Dones-Lassalle CY, Pabón-Colon HL, Rodríguez-Rivera HM, Rodríguez I, González-Espiet JC, Pazol J, Pérez-Ríos JD, Catala-Torres JF, Carrasquillo Rivera M, De Jesus-Soto MG, Cordero-Virella NA, Cruz-Maldonado PM, González-Pagan P, Hernández-Ríos R, Gaur K, Loza-Rosas SA, Tinoco AD. Exploring Serum Transferrin Regulation of Nonferric Metal Therapeutic Function and Toxicity. INORGANICS 2020; 8:48. [PMID: 36844373 PMCID: PMC9957567 DOI: 10.3390/inorganics8090048] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Serum transferrin (sTf) plays a pivotal role in regulating iron biodistribution and homeostasis within the body. The molecular details of sTf Fe(III) binding blood transport, and cellular delivery through transferrin receptor-mediated endocytosis are generally well-understood. Emerging interest exists in exploring sTf complexation of nonferric metals as it facilitates the therapeutic potential and toxicity of several of them. This review explores recent X-ray structural and physiologically relevant metal speciation studies to understand how sTf partakes in the bioactivity of key non-redox active hard Lewis acidic metals. It challenges preconceived notions of sTf structure function correlations that were based exclusively on the Fe(III) model by revealing distinct coordination modalities that nonferric metal ions can adopt and different modes of binding to metal-free and Fe(III)-bound sTf that can directly influence how they enter into cells and, ultimately, how they may impact human health. This knowledge informs on biomedical strategies to engineer sTf as a delivery vehicle for metal-based diagnostic and therapeutic agents in the cancer field. It is the intention of this work to open new avenues for characterizing the functionality and medical utility of nonferric-bound sTf and to expand the significance of this protein in the context of bioinorganic chemistry.
Collapse
Affiliation(s)
- Josué A. Benjamín-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Andrés E. Cardona-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | | | - Héctor L. Pabón-Colon
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | - Israel Rodríguez
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Jean C. González-Espiet
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Jessika Pazol
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Jobaniel D. Pérez-Ríos
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - José F. Catala-Torres
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | - Michael G. De Jesus-Soto
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | - Paola M. Cruz-Maldonado
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Patricia González-Pagan
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Raul Hernández-Ríos
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Kavita Gaur
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Sergio A. Loza-Rosas
- Departamento de Química y Bioquímica, Facultad de Ciencias e Ingeniería, Universidad de Boyacá, Tunja 150003, Colombia
| | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
- Correspondence: ; Tel.: +1-939-319-9701
| |
Collapse
|
44
|
Hecel A, Ostrowska M, Stokowa-Sołtys K, Wątły J, Dudek D, Miller A, Potocki S, Matera-Witkiewicz A, Dominguez-Martin A, Kozłowski H, Rowińska-Żyrek M. Zinc(II)-The Overlooked Éminence Grise of Chloroquine's Fight against COVID-19? Pharmaceuticals (Basel) 2020; 13:E228. [PMID: 32882888 PMCID: PMC7558363 DOI: 10.3390/ph13090228] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/24/2022] Open
Abstract
Zn(II) is an inhibitor of SARS-CoV-2's RNA-dependent RNA polymerase, and chloroquine and hydroxychloroquine are Zn(II) ionophores-this statement gives a curious mind a lot to think about. We show results of the first clinical trials on chloroquine (CQ) and hydroxychloroquine (HCQ) in the treatment of COVID-19, as well as earlier reports on the anticoronaviral properties of these two compounds and of Zn(II) itself. Other FDA-approved Zn(II) ionophores are given a decent amount of attention and are thought of as possible COVID-19 therapeutics.
Collapse
Affiliation(s)
- Aleksandra Hecel
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Małgorzata Ostrowska
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Kamila Stokowa-Sołtys
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Joanna Wątły
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Dorota Dudek
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Adriana Miller
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Sławomir Potocki
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| | - Agnieszka Matera-Witkiewicz
- Screening Laboratory of Biological Activity Tests and Collection of Biological Material, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Alicia Dominguez-Martin
- Department of Inorganic Chemistry, Faculty of Pharmacy, University of Granada, E-18071 Granada, Spain;
| | - Henryk Kozłowski
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
- Department of Physiotherapy, Opole Medical School, Katowicka 68, 40-060 Opole, Poland
| | - Magdalena Rowińska-Żyrek
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (A.H.); (M.O.); (K.S.-S.); (J.W.); (D.D.); (A.M.); (S.P.); (H.K.)
| |
Collapse
|
45
|
Das D, Raza MK, Goswami TK. Evaluation of photochemotherapeutic potential of a few oxo-bridged dimeric Fe(III) compounds having Salen-type ligands. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Nahari G, Braitbard O, Larush L, Hochman J, Tshuva EY. Effective Oral Administration of an Antitumorigenic Nanoformulated Titanium Complex. ChemMedChem 2020; 16:108-112. [PMID: 32657024 DOI: 10.1002/cmdc.202000384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Indexed: 12/13/2022]
Abstract
Orally administered anticancer drugs facilitate treatment, but the acidic conditions in the stomach often challenge their availability. PhenolaTi is a TiIV -based nontoxic anticancer drug with marked in-vivo efficacy. We report that nanoformulation protects phenolaTi from decomposition in stomach-like conditions. This is evidenced by similar NMR characteristics and similar in-vitro cytotoxicity toward murine (CT-26) and human (HT-29) colon cancer cells before and after incubation of nanoformulated phenolaTi (phenolaTi-F) at pH 2, unlike results with the unformulated form of the complex. Furthermore, administration of phenolaTi-F in animal drinking water revealed a notable inhibition of tumor growth in Balb/c and immune-deficient (Nude) mice inoculated with CT-26 and HT-29 cells, respectively. In-vivo efficacy was at least similar to that of the corresponding intraperitoneal treatment with phenolaTi-F and the clinically employed oral drug, capecitabine. No body weight loss or clinical signs of toxicity were evident in the phenolaTi-F-treated animals. These findings demonstrate a new convenient mode of cancer treatment through oral administration by safe titanium-based drugs.
Collapse
Affiliation(s)
- Gilad Nahari
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Ori Braitbard
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Liraz Larush
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Jacob Hochman
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Edit Y Tshuva
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| |
Collapse
|
47
|
Taviano MF, Miceli N, Acquaviva R, Malfa GA, Ragusa S, Giordano D, Cásedas G, Les F, López V. Cytotoxic, Antioxidant, and Enzyme Inhibitory Properties of the Traditional Medicinal Plant Matthiola incana (L.) R. Br. BIOLOGY 2020; 9:E163. [PMID: 32668697 PMCID: PMC7407578 DOI: 10.3390/biology9070163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Matthiola incana (L.) R. Br. (Brassicaceae) is widely cultivated for ornamental purposes and utilized as a medicinal plant. In the present work, the hydroalcoholic extract from the aerial parts of this species has been evaluated in different bioassays in order to detect potential pharmacological applications. The cytotoxic capacity against the human colorectal adenocarcinoma (CaCo-2) and breast cancer (MCF-7) cell lines was tested using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. The extract was investigated as a neuroprotective inhibitor of central nervous system (CNS) enzymes such as monoamine oxidase A, tyrosinase, acetylcholinesterase, and as a natural enzyme inhibitor of α-glucosidase and lipase involved in some metabolic disorders such as obesity or type 2 diabetes. The antioxidant ability was also evaluated in an enzymatic system (xanthine/xanthine oxidase assay). Results showed that the M. incana extract displayed moderate to low cytotoxicity vs. CaCo-2 cells. The extract acted as a superoxide radical scavenger and enzymatic inhibitor of monoamine oxidase A, tyrosinase, α-glucosidase, and lipase. The best results were found in the α-glucosidase assay, as M. incana hydroalcoholic extract was able to inhibit the enzyme α-glucosidase up to 100% without significant differences, compared to the antidiabetic drug acarbose. Matthiola incana has been demonstrated to exert different biological properties. These are important in order to consider this species as a source of bioactive compounds.
Collapse
Affiliation(s)
- Maria Fernanda Taviano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Polo Annunziata, University of Messina, 98168 Messina, Italy; (M.F.T.); (N.M.); (D.G.)
| | - Natalizia Miceli
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Polo Annunziata, University of Messina, 98168 Messina, Italy; (M.F.T.); (N.M.); (D.G.)
| | - Rosaria Acquaviva
- Department of Drug Science, Biochemistry Section, University of Catania, 95123 Catania, Italy; (R.A.); (G.A.M.)
| | - Giuseppe Antonio Malfa
- Department of Drug Science, Biochemistry Section, University of Catania, 95123 Catania, Italy; (R.A.); (G.A.M.)
| | - Salvatore Ragusa
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Deborah Giordano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Polo Annunziata, University of Messina, 98168 Messina, Italy; (M.F.T.); (N.M.); (D.G.)
| | - Guillermo Cásedas
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain; (G.C.); (F.L.)
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain; (G.C.); (F.L.)
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Víctor López
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain; (G.C.); (F.L.)
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, 50013 Zaragoza, Spain
| |
Collapse
|
48
|
Kontoghiorghes GJ, Kontoghiorghe CN. Iron and Chelation in Biochemistry and Medicine: New Approaches to Controlling Iron Metabolism and Treating Related Diseases. Cells 2020; 9:E1456. [PMID: 32545424 PMCID: PMC7349684 DOI: 10.3390/cells9061456] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Iron is essential for all living organisms. Many iron-containing proteins and metabolic pathways play a key role in almost all cellular and physiological functions. The diversity of the activity and function of iron and its associated pathologies is based on bond formation with adjacent ligands and the overall structure of the iron complex in proteins or with other biomolecules. The control of the metabolic pathways of iron absorption, utilization, recycling and excretion by iron-containing proteins ensures normal biologic and physiological activity. Abnormalities in iron-containing proteins, iron metabolic pathways and also other associated processes can lead to an array of diseases. These include iron deficiency, which affects more than a quarter of the world's population; hemoglobinopathies, which are the most common of the genetic disorders and idiopathic hemochromatosis. Iron is the most common catalyst of free radical production and oxidative stress which are implicated in tissue damage in most pathologic conditions, cancer initiation and progression, neurodegeneration and many other diseases. The interaction of iron and iron-containing proteins with dietary and xenobiotic molecules, including drugs, may affect iron metabolic and disease processes. Deferiprone, deferoxamine, deferasirox and other chelating drugs can offer therapeutic solutions for most diseases associated with iron metabolism including iron overload and deficiency, neurodegeneration and cancer, the detoxification of xenobiotic metals and most diseases associated with free radical pathology.
Collapse
Affiliation(s)
- George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, CY-3021 Limassol, Cyprus;
| | | |
Collapse
|
49
|
Fik-Jaskółka MA, Mkrtchyan AF, Saghyan AS, Palumbo R, Belter A, Hayriyan LA, Simonyan H, Roviello V, Roviello GN. Biological macromolecule binding and anticancer activity of synthetic alkyne-containing L-phenylalanine derivatives. Amino Acids 2020; 52:755-769. [PMID: 32430874 DOI: 10.1007/s00726-020-02849-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/29/2020] [Indexed: 01/17/2023]
Abstract
Herein, we described the synthesis of two L-phenylalanines α-derivatized with a terminal alkyne moiety whose structures differed by phenyl ring halogen substitution (two o-Cl in 1 vs. one p-Br in 2) and investigated their effect on biological macromolecules and living cells. We explored their interaction with quadruplex DNA (G4 DNA), using tel26 and c-myc as models, and bovine serum albumin (BSA). By CD spectroscopy, we found that 1 caused minor tel26 secondary structure changes, leading also to a slight thermal stabilization of this hybrid antiparallel/parallel G4 structure, while the c-myc parallel topology remained essentially unchanged upon 1 binding. Other CD evidences showed the ability of 1 to bind BSA, while molecular docking studies suggested that the same molecule could be housed into the hydrophobic cavity between sub-domains IIA, IIB, and IIIA of the protein. Furthermore, preliminary aggregation studies, based on concentration-dependent spectroscopic experiments, suggested the ability of 1 to aggregate forming noncovalent polymeric systems in aqueous solution. Differently from 1, the bromine-modified compound was able to bind Cu(II) ion, likely with the formation of a CuL2 complex, as found by UV spectroscopy. Finally, cell tests excluded any cytotoxic effect of both compounds toward normal cells, but showed slight antiproliferative effects of 2 on PC3 cancerous cells at 24 h, and of 1 on both T98G and MDA-MB-231 cancer cells at 48 h.
Collapse
Affiliation(s)
- Marta A Fik-Jaskółka
- Department of Bioinorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego Str. 8, 61-614, Poznan, Poland.,Centre for Advanced Technology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego Str. 10, 61-614, Poznan, Poland.,Istituto di Biostrutture e Bioimmagini, IBB-CNR, Via Mezzocannone 16, 80134, Naples, Italy
| | - Anna F Mkrtchyan
- Scientific and Production Center, Armbiotechnology" of NAS RA, 14 Gyurjyan Str., 0056, Yerevan, Armenia.,Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., 0025, Yerevan, Armenia
| | - Ashot S Saghyan
- Scientific and Production Center, Armbiotechnology" of NAS RA, 14 Gyurjyan Str., 0056, Yerevan, Armenia.,Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., 0025, Yerevan, Armenia
| | - Rosanna Palumbo
- Istituto di Biostrutture e Bioimmagini, IBB-CNR, Via Mezzocannone 16, 80134, Naples, Italy
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland
| | - Liana A Hayriyan
- Scientific and Production Center, Armbiotechnology" of NAS RA, 14 Gyurjyan Str., 0056, Yerevan, Armenia.,Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., 0025, Yerevan, Armenia
| | - Hayarpi Simonyan
- Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., 0025, Yerevan, Armenia
| | - Valentina Roviello
- Department of Chemical, Materials and Industrial Production Engineering (DICMaPI), University of Naples Federico II, Piazzale V. Tecchio 80, 80125, Naples, Italy
| | - Giovanni N Roviello
- Istituto di Biostrutture e Bioimmagini, IBB-CNR, Via Mezzocannone 16, 80134, Naples, Italy.
| |
Collapse
|
50
|
Rada JP, Forté J, Gontard G, Corcé V, Salmain M, Rey NA. Isoxazole-Derived Aroylhydrazones and Their Dinuclear Copper(II) Complexes Show Antiproliferative Activity on Breast Cancer Cells with a Potentially Alternative Mechanism Of Action. Chembiochem 2020; 21:2474-2486. [PMID: 32282111 DOI: 10.1002/cbic.202000122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Indexed: 12/15/2022]
Abstract
This paper reports the design, synthesis and cytotoxicity studies of two new isoxazole-derived aroylhydrazone ligands and their dinuclear copper(II) complexes. Compounds were fully characterized by various spectroscopic and analytical techniques. The molecular structures of four derivatives were confirmed by X-ray crystallography. The stability of the ligands and the complexes in aqueous medium was monitored spectroscopically. Both the ligands and the complexes were shown to interact with calf thymus DNA (ct-DNA). Additionally, structures containing a phenol pendant arm were significantly more cytotoxic than those carrying a pendant pyridine substituent, reaching sub-micromolar IC50 values on the triple-negative human breast cancer cell line MDA-MB-231. The metal chelation and transchelation ability of the compounds towards FeII , FeIII and ZnII ions was explored as a possible mechanism of action of these compounds.
Collapse
Affiliation(s)
- Jesica Paola Rada
- LABSO-Bio Laboratory, Department of Chemistry, Pontifical Catholic University of Rio de Janeiro, 225 Rua Marquês de, São Vicente, Brazil
| | - Jéremy Forté
- Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, CNRS, 4 place Jussieu, 75005, Paris, France
| | - Geoffrey Gontard
- Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, CNRS, 4 place Jussieu, 75005, Paris, France
| | - Vincent Corcé
- Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, CNRS, 4 place Jussieu, 75005, Paris, France
| | - Michèle Salmain
- Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, CNRS, 4 place Jussieu, 75005, Paris, France
| | - Nicolás A Rey
- LABSO-Bio Laboratory, Department of Chemistry, Pontifical Catholic University of Rio de Janeiro, 225 Rua Marquês de, São Vicente, Brazil
| |
Collapse
|