1
|
Shi H, Marchi RC, Sadler PJ. Advances in the Design of Photoactivatable Metallodrugs: Excited State Metallomics. Angew Chem Int Ed Engl 2025; 64:e202423335. [PMID: 39806815 DOI: 10.1002/anie.202423335] [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: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Photoactivatable metal complexes offer the prospect of novel drugs with low side effects and new mechanisms of action to combat resistance to current therapy. We highlight recent progress in the design of platinum, ruthenium, iridium, gold and other transition metal complexes, especially for applications as anticancer and anti-infective agents. In particular, understanding excited state chemistry related to identification of the bioactive species (excited state metallomics/pharmacophores) is important. Photoactivatable metallodrugs are classified here as photocatalysts, photorelease agents and ligand-activated agents. Their activation wavelengths, cellular mechanisms of action, experimental and theoretical metallomics of excited states and photoproducts are discussed to explore new strategies for the design and investigation of photoactivatable metallodrugs. These photoactivatable metallodrugs have potential in clinical applications of Photodynamic Therapy (PDT), Photoactivated Chemotherapy (PACT) and Photothermal Therapy (PTT).
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Rafael C Marchi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Peter J Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
2
|
Liu X, Ji C, Tao R, Zheng W, Liu M, Bi S, Chang Q, Yuan XA, Yue M, Liu Z. Effects of structurally varied fluorescent half-sandwich iridium(III) Schiff base complexes on A549 cell line. J Inorg Biochem 2025; 263:112792. [PMID: 39615316 DOI: 10.1016/j.jinorgbio.2024.112792] [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: 09/21/2024] [Revised: 11/12/2024] [Accepted: 11/24/2024] [Indexed: 12/12/2024]
Abstract
Half-sandwich iridium(III) (IrIII) anticancer complexes, as promising alternatives to platinum-based drugs, especially for solving resistance to platinum drugs, have demonstrated excellent application prospect. The potency of these IrIII complexes as anticancer agents could be significantly enhanced through the strategic modification of their peripheral ligands. In this study, four structurally varied triphenylamine (TPA)-modified half-sandwich IrIII Schiff base complexes were designed and prepared. The incorporation of TPA unit has effectively endowed these complexes with suitable emission, which facilitates the evaluation of intracellular accumulation and cell morphology. These complexes demonstrated favorable in vitro anti-proliferative activity against A549 cell line (lung cancer cells, derived from alveolar basal epithelial cells), especially for pentamethylcyclopentadiene (Cp*)-based one (IrTS1 and IrTS3), and that is almost 2.5-fold more than cisplatin under the same conditions. Meanwhile, IrTS1 and IrTS3 possessed excellent activity against A549/DDP (cisplatin-resistant) cell line and the similar cytotoxicity to cisplatin against BEAS-2B cell line (derived from the bronchial epithelium of normal human lungs), then following a mitochondria apoptotic channel.
Collapse
Affiliation(s)
- Xicheng Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China.
| | - Changjian Ji
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Rui Tao
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Wenya Zheng
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Mengxian Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Shiqing Bi
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Qinghua Chang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Xiang-Ai Yuan
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Mingbo Yue
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China.
| |
Collapse
|
3
|
Park M, Nam JS, Kim T, Yoon G, Kim S, Lee C, Lee CG, Park S, Bejoymohandas KS, Yang J, Kwon YH, Lee YJ, Seo JK, Min D, Park T, Kwon T. Rational Design of Biocompatible Ir(III) Photosensitizer to Overcome Drug-Resistant Cancer via Oxidative Autophagy Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407236. [PMID: 39540573 PMCID: PMC11727131 DOI: 10.1002/advs.202407236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/25/2024] [Indexed: 11/16/2024]
Abstract
Autophagy is a crucial quality control mechanism that degrades damaged cellular components through lysosomal fusion with autophagosomes. However, elevated autophagy levels can promote drug resistance in cancer cells, enhancing their survival. Downregulation of autophagy through oxidative stress is a clinically promising strategy to counteract drug resistance, yet precise control of oxidative stress in autophagic proteins remains challenging. Here, a molecular design strategy of biocompatible neutral Ir(III) photosensitizers is demonstrated, B2 and B4, for precise reactive oxygen species (ROS) control at lysosomes to inhibit autophagy. The underlying molecular mechanisms for the biocompatibility and lysosome selectivity of Ir(III) complexes are explored by comparing B2 with the cationic or the non-lysosome-targeting analogs. Also, the biological mechanisms for autophagy inhibition via lysosomal oxidation are explored. Proteome analyses reveal significant oxidation of proteins essential for autophagy, including lysosomal and fusion-mediator proteins. These findings are verified in vitro, using mass spectrometry, live cell imaging, and a model SNARE complex. The anti-tumor efficacy of the precise lysosomal oxidation strategy is further validated in vivo with B4, engineered for red light absorbance. This study is expected to inspire the therapeutic use of spatiotemporal ROS control for sophisticated modulation of autophagy.
Collapse
Affiliation(s)
- Mingyu Park
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- X‐dynamic Research CenterUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Jung Seung Nam
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- X‐dynamic Research CenterUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Institute for Cancer GeneticsDepartment of Genetics and DevelopmentColumbia University Medical CenterNew YorkNY10032USA
- Herbert Irving Comprehensive Cancer CenterColumbia University Irving Medical CenterNew YorkNY10032USA
| | - Taehyun Kim
- Department of Chemical EngineeringPohang University of Science and Technology (POSTECH)77 Cheongam‐Ro, Nam‐GuPohangGyeongbuk37673Republic of Korea
| | - Gwangsu Yoon
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- X‐dynamic Research CenterUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Seoyoon Kim
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Chaiheon Lee
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- X‐dynamic Research CenterUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Chae Gyu Lee
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Sungjin Park
- Department of Chemical EngineeringPohang University of Science and Technology (POSTECH)77 Cheongam‐Ro, Nam‐GuPohangGyeongbuk37673Republic of Korea
| | - Kochan S. Bejoymohandas
- Department of Chemical EngineeringPohang University of Science and Technology (POSTECH)77 Cheongam‐Ro, Nam‐GuPohangGyeongbuk37673Republic of Korea
| | - Jihyeon Yang
- Research CenterO2MEDi inc.Ulsan44919Republic of Korea
| | - Yoon Hee Kwon
- Research CenterO2MEDi inc.Ulsan44919Republic of Korea
| | - Yoo Jin Lee
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Jeong Kon Seo
- Research CenterO2MEDi inc.Ulsan44919Republic of Korea
| | - Duyoung Min
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Taiho Park
- Department of Chemical EngineeringPohang University of Science and Technology (POSTECH)77 Cheongam‐Ro, Nam‐GuPohangGyeongbuk37673Republic of Korea
| | - Tae‐Hyuk Kwon
- Department of ChemistryUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- X‐dynamic Research CenterUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Research CenterO2MEDi inc.Ulsan44919Republic of Korea
| |
Collapse
|
4
|
Sil S, Hussain A, Das Sarma J, Gupta P. Cyclometalated Iridium(III) Complex with Substituted Benzimidazole: pH Directed Organelle-Specific Localization Within Lysosome. Chembiochem 2024; 25:e202400597. [PMID: 39285747 DOI: 10.1002/cbic.202400597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/12/2024] [Indexed: 11/06/2024]
Abstract
We report the synthesis and pH dependent emission spectral behaviour of four emissive iridium(III) complexes (Ir1-Ir4) with two isomeric pairs of bis-trifluoromethyl appended benzimidazole ligands. The imidazolyl hydrogen(N-H) has been replaced by phenyl groups (N-Ph) in two ligands to understand the impact of hydrogen bonding on the photophysical properties of the complexes and it indeed plays interesting role in the charge-transfer dynamics. The pH dependent electronic spectral change is observed for two of the complexes. The enhancement of emission intensity is observed at different wavelength for pH<7 and pH>7 for Ir1 and Ir3. The emission sensing of biogenic amines with pka values ranging from 5.80-9.74 is reported along with cellular imaging. The complex Ir1 specifically localizes within lysosome (pH=4.5-5) and thus image this organelle with great precision. The detail electronic spectra and electrochemical behaviour were reported here along with TDDFT results.
Collapse
Affiliation(s)
- Subhra Sil
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, 741246, India
| | - Afaq Hussain
- Bio-inspired Innovation Private Limited (RISE Foundation IISER, The Incubation Center of IISER Kolkata), Mohanpur, West Bengal, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, 741246, India
| | - Parna Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, 741246, India
| |
Collapse
|
5
|
Dixit T, Negi M, Venkatesh V. Mitochondria Localized Anticancer Iridium(III) Prodrugs for Targeted Delivery of Myeloid Cell Leukemia-1 (Mcl-1) Inhibitors and Cytotoxic Iridium(III) Complex. Inorg Chem 2024. [PMID: 39667040 DOI: 10.1021/acs.inorgchem.4c03950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Myeloid cell leukemia-1 (Mcl-1) is an antiapoptotic oncoprotein overexpressed in several malignancies and acts as one of the promising therapeutic targets for cancer. Even though there are several small molecule based Mcl-1 inhibitors reported, the delivery of Mcl-1 inhibitor at the target site is quite challenging. In this regard, we developed a series of mitochondria targeting luminescent cyclometalated iridium(III) prodrugs bearing Mcl-1 inhibitors via ester linkage due to the presence of Mcl-1 protein in the outer mitochondrial membrane. Among the synthesized prodrugs, IrThpy@L2 was found to exhibit the potent cytotoxicity (IC50 = 30.93 nM) against HCT116 cell line when compared with bare Mcl-1 inhibitors (IC50 > 100 μM). Mechanistic studies further revealed that IrThpy@L2 quickly gets internalized inside the mitochondria of HCT116 cells and undergoes activation in the presence of overexpressed esterase which leads to the release of two cytotoxic species i.e. Mcl-1 inhibitors (I-2) and cytotoxic iridium(III) complex (IrThpy@OH). The improved cytotoxicity of IrThpy@L2 is due to the mitochondria targeting ability of iridium(III) prodrug, subsequent esterase activated release of I-2 to inhibit Mcl-1 protein and IrThpy@OH to generate reactive oxygen species (ROS). After prodrug activation, the released cytotoxic species cause mitochondrial membrane depolarization, activate a cascade of mitochondria-mediated cell death events, and arrest the cell cycle in S-phase which leads to apoptosis. The potent anticancer activity of IrThpy@L2 was further evident from the drastic morphological changes, size reduction in the solid tumor mimicking 3D multicellular tumor spheroids (MCTS) of HCT116.
Collapse
Affiliation(s)
- Tejal Dixit
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Monika Negi
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - V Venkatesh
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| |
Collapse
|
6
|
Lee LC, Lo KK. Leveraging the Photofunctions of Transition Metal Complexes for the Design of Innovative Phototherapeutics. SMALL METHODS 2024; 8:e2400563. [PMID: 39319499 PMCID: PMC11579581 DOI: 10.1002/smtd.202400563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/03/2024] [Indexed: 09/26/2024]
Abstract
Despite the advent of various medical interventions for cancer treatment, the disease continues to pose a formidable global health challenge, necessitating the development of new therapeutic approaches for more effective treatment outcomes. Photodynamic therapy (PDT), which utilizes light to activate a photosensitizer to produce cytotoxic reactive oxygen species (ROS) for eradicating cancer cells, has emerged as a promising approach for cancer treatment due to its high spatiotemporal precision and minimal invasiveness. However, the widespread clinical use of PDT faces several challenges, including the inefficient production of ROS in the hypoxic tumor microenvironment, the limited penetration depth of light in biological tissues, and the inadequate accumulation of photosensitizers at the tumor site. Over the past decade, there has been increasing interest in the utilization of photofunctional transition metal complexes as photosensitizers for PDT applications due to their intriguing photophysical and photochemical properties. This review provides an overview of the current design strategies used in the development of transition metal complexes as innovative phototherapeutics, aiming to address the limitations associated with PDT and achieve more effective treatment outcomes. The current challenges and future perspectives on the clinical translation of transition metal complexes are also discussed.
Collapse
Affiliation(s)
- Lawrence Cho‐Cheung Lee
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| | - Kenneth Kam‐Wing Lo
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
- State Key Laboratory of Terahertz and Millimeter WavesCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| |
Collapse
|
7
|
Pattanayak PD, Banerjee A, Sahu G, Das S, Lima S, Akintola O, Buchholz A, Görls H, Plass W, Reuter H, Dinda R. Insights into the Theranostic Activity of Nonoxido V IV: Lysosome-Targeted Anticancer Metallodrugs. Inorg Chem 2024; 63:19418-19438. [PMID: 39340532 DOI: 10.1021/acs.inorgchem.4c03389] [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: 09/30/2024]
Abstract
Developing new anticancer agents can be useful, with the ability to diagnose and treat cancer worldwide. Previously, we focused on examining the effects of nonoxidovanadium(IV) complexes on insulin mimetic and cytotoxicity activity. In this study, in addition to the cytotoxic activity, we evaluated their bioimaging properties. This study investigates the synthesis of four stable nonoxido VIV complexes [VIV(L1-4)2] (1-4) using aroylhydrazone ligands (H2L1-4) and their full characterization in solid state and the solution phase stability using various physicochemical techniques. The biomolecular (DNA/HSA) interaction of the complexes was evaluated by using conventional methods. The in vitro cytotoxicity of 1-4 was studied against A549 and LN-229 cancer cell lines and found that drug 2 displayed the highest activity among the four. Since 1-4 are fluorescently active, live cell imaging was used to evaluate their cellular localization activity. Complexes specifically target the lysosome and damage lysosome integrity by producing an excessive amount (9.7-fold) of reactive oxygen species (ROS) compared to the control, which may cause cell apoptosis. Overall, this study indicates that 2 has the greatest potential for the development of multifunctional theranostic agents that combine imaging capabilities and anticancer properties of nonoxidovanadium(IV)-based metallodrugs.
Collapse
Affiliation(s)
| | - Atanu Banerjee
- Department of Chemistry, National Institute of Technology, 769008 Rourkela, Odisha, India
| | - Gurunath Sahu
- Department of Chemistry, National Institute of Technology, 769008 Rourkela, Odisha, India
| | - Sanchita Das
- Department of Chemistry, National Institute of Technology, 769008 Rourkela, Odisha, India
| | - Sudhir Lima
- Department of Chemistry, National Institute of Technology, 769008 Rourkela, Odisha, India
| | - Oluseun Akintola
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller-Universität Jena, Humboldtstr. 8, 07743 Jena, Germany
| | - Axel Buchholz
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller-Universität Jena, Humboldtstr. 8, 07743 Jena, Germany
| | - Helmar Görls
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller-Universität Jena, Humboldtstr. 8, 07743 Jena, Germany
| | - Winfried Plass
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller-Universität Jena, Humboldtstr. 8, 07743 Jena, Germany
| | - Hans Reuter
- Institute of Chemistry of New Materials, University of Osnabrück, Barbarastrasse 7, 49067 Osnabrück, Germany
| | - Rupam Dinda
- Department of Chemistry, National Institute of Technology, 769008 Rourkela, Odisha, India
| |
Collapse
|
8
|
Sanz-Villafruela J, Bermejo-Casadesús C, Riesco-Llach G, Iglesias M, Martínez-Alonso M, Planas M, Feliu L, Espino G, Massaguer A. Bombesin-Targeted Delivery of β-Carboline-Based Ir(III) and Ru(II) Photosensitizers for a Selective Photodynamic Therapy of Prostate Cancer. Inorg Chem 2024; 63:19140-19155. [PMID: 39361042 PMCID: PMC11483813 DOI: 10.1021/acs.inorgchem.4c02583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024]
Abstract
Despite advances in Ir(III) and Ru(II) photosensitizers (PSs), their lack of selectivity for cancer cells has hindered their use in photodynamic therapy (PDT). We disclose the synthesis and characterization of two pairs of Ir(III) and Ru(II) polypyridyl complexes bearing two β-carboline ligands (N^N') functionalized with -COOMe (L1) or -COOH (L2), resulting in PSs of formulas [Ir(C^N)2(N^N')]Cl (Ir-Me: C^N = ppy, N^N' = L1; Ir-H: C^N = ppy, N^N' = L2) and [Ru(N^N)2(N^N')](Cl)2 (Ru-Me: N^N = bpy, N^N' = L1; Ru-H: N^N = bpy, N^N' = L2). To enhance their selectivity toward cancer cells, Ir-H and Ru-H were coupled to a bombesin derivative (BN3), resulting in the metallopeptides Ir-BN and Ru-BN. Ir(III) complexes showed higher anticancer activity than their Ru(II) counterparts, particularly upon blue light irradiation, but lacked cancer cell selectivity. In contrast, Ir-BN and Ru-BN exhibited selective photocytoxicity against prostate cancer cells, with a lower effect against nonmalignant fibroblasts. All compounds generated ROS and induced severe mitochondrial toxicity upon photoactivation, leading to apoptosis. Additionally, the ability of Ir-Me to oxidize NADH was demonstrated, suggesting a mechanism for mitochondrial damage. Our findings indicated that the conjugation of metal PSs with BN3 creates efficient PDT agents, achieving selectivity through targeting bombesin receptors and local photoactivation.
Collapse
Affiliation(s)
- Juan Sanz-Villafruela
- Universidad
de Burgos, Departamento de
Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, Burgos 09001, Spain
| | - Cristina Bermejo-Casadesús
- Universitat
de Girona, Departament de
Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, Girona 17003, Spain
| | - Gerard Riesco-Llach
- LIPPSO,
Departament de Química, Facultat de Ciències, Universitat de Girona, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Mònica Iglesias
- Universitat
de Girona, Departament de Química,
Facultat de Ciències, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Marta Martínez-Alonso
- Universidad
de Burgos, Departamento de
Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, Burgos 09001, Spain
| | - Marta Planas
- LIPPSO,
Departament de Química, Facultat de Ciències, Universitat de Girona, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Lidia Feliu
- LIPPSO,
Departament de Química, Facultat de Ciències, Universitat de Girona, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Gustavo Espino
- Universidad
de Burgos, Departamento de
Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, Burgos 09001, Spain
| | - Anna Massaguer
- Universitat
de Girona, Departament de
Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, Girona 17003, Spain
| |
Collapse
|
9
|
Szymaszek P, Tyszka-Czochara M, Ortyl J. Iridium(III) complexes as novel theranostic small molecules for medical diagnostics, precise imaging at a single cell level and targeted anticancer therapy. Eur J Med Chem 2024; 276:116648. [PMID: 38968786 DOI: 10.1016/j.ejmech.2024.116648] [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: 05/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Medical applications of iridium (III) complexes include their use as state-of-the-art theranostic agents - molecules that combine therapeutic and diagnostic functions into a single entity. These complexes offer a promising avenue in medical diagnostics, precision imaging at single-cell resolution and targeted anticancer therapy due to their unique properties. In this review we report a short summary of their application in medical diagnostics, imaging at single-cell level and targeted anticancer therapy. The exceptional photophysical properties of Iridium (III) complexes, including their brightness and photostability, make them excellent candidates for bioimaging. They can be used to image cellular processes and the microenvironment within single cells with unprecedented clarity, aiding in the understanding of disease mechanisms at the molecular level. Moreover the iridium (III) complexes can be designed to selectively target cancer cells,. Upon targeting, these complexes can act as photosensitizers for photodynamic therapy (PDT), generating reactive oxygen species (ROS) upon light activation to induce cell death. The integration of diagnostic and therapeutic capabilities in Iridium (III) complexes offers the potential for a holistic approach to cancer treatment, enabling not only the precise eradication of cancer cells but also the real-time monitoring of treatment efficacy and disease progression. This aligns with the goals of personalized medicine, offering hope for more effective and less invasive cancer treatment strategies.
Collapse
Affiliation(s)
- Patryk Szymaszek
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155, Kraków, Poland
| | | | - Joanna Ortyl
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155, Kraków, Poland; Photo HiTech Ltd., Bobrzyńskiego 14, 30-348, Kraków, Poland; Photo4Chem ltd., Juliusza Lea 114/416A-B, 31-133, Kraków, Poland.
| |
Collapse
|
10
|
Sanz-Villafruela J, Bermejo-Casadesus C, Zafon E, Martínez-Alonso M, Durá G, Heras A, Soriano-Díaz I, Giussani A, Ortí E, Tebar F, Espino G, Massaguer A. Insights into the anticancer photodynamic activity of Ir(III) and Ru(II) polypyridyl complexes bearing β-carboline ligands. Eur J Med Chem 2024; 276:116618. [PMID: 38972079 DOI: 10.1016/j.ejmech.2024.116618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/31/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
Ir(III) and Ru(II) polypyridyl complexes are promising photosensitizers (PSs) for photodynamic therapy (PDT) due to their outstanding photophysical properties. Herein, one series of cyclometallated Ir(III) complexes and two series of Ru(II) polypyridyl derivatives bearing three different thiazolyl-β-carboline N^N' ligands have been synthesized, aiming to evaluate the impact of the different metal fragments ([Ir(C^N)2]+ or [Ru(N^N)2]2+) and N^N' ligands on the photophysical and biological properties. All the compounds exhibit remarkable photostability under blue-light irradiation and are emissive (605 < λem < 720 nm), with the Ru(II) derivatives displaying higher photoluminescence quantum yields and longer excited state lifetimes. The Ir PSs display pKa values between 5.9 and 7.9, whereas their Ru counterparts are less acidic (pKa > 9.3). The presence of the deprotonated form in the Ir-PSs favours the generation of reactive oxygen species (ROS) since, according to theoretical calculations, it features a low-lying ligand-centered triplet excited state (T1 = 3LC) with a long lifetime. All compounds have demonstrated anticancer activity. Ir(III) complexes 1-3 exhibit the highest cytotoxicity in dark conditions, comparable to cisplatin. Their activity is notably enhanced by blue-light irradiation, resulting in nanomolar IC50 values and phototoxicity indexes (PIs) between 70 and 201 in different cancer cell lines. The Ir(III) PSs are also activated by green (with PI between 16 and 19.2) and red light in the case of complex 3 (PI = 8.5). Their antitumor efficacy is confirmed by clonogenic assays and using spheroid models. The Ir(III) complexes rapidly enter cells, accumulating in mitochondria and lysosomes. Upon photoactivation, they generate ROS, leading to mitochondrial dysfunction and lysosomal damage and ultimately cell apoptosis. Additionally, they inhibit cancer cell migration, a crucial step in metastasis. In contrast, Ru(II) complex 6 exhibits moderate mitochondrial activity. Overall, Ir(III) complexes 1-3 show potential for selective light-controlled cancer treatment, providing an alternative mechanism to chemotherapy and the ability to inhibit lethal cancer cell dissemination.
Collapse
Affiliation(s)
- Juan Sanz-Villafruela
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Cristina Bermejo-Casadesus
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain
| | - Elisenda Zafon
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain
| | - Marta Martínez-Alonso
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Gema Durá
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica. Facultad de Químicas, Avda. Camilo J. Cela 10, 13071, Ciudad Real, Spain
| | - Aranzazu Heras
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Iván Soriano-Díaz
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain
| | - Angelo Giussani
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain
| | - Enrique Ortí
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain.
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain.
| | - Gustavo Espino
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain.
| | - Anna Massaguer
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain.
| |
Collapse
|
11
|
Guo L, Li P, Jing Z, Gong Y, Lai K, Fu H, Dong H, Yang Z, Liu Z. Iminoamido chelated iridium(III) and ruthenium(II) anticancer complexes with mitochondria-targeting ability and potential to overcome cisplatin resistance. J Inorg Biochem 2024; 258:112631. [PMID: 38843774 DOI: 10.1016/j.jinorgbio.2024.112631] [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: 03/03/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 07/01/2024]
Abstract
A diverse set of neutral half-sandwich iminoamido iridium and ruthenium organometallic complexes is synthesized through the utilization of Schiff base pro-ligands with N˄N donors. Notably, these metal complexes with varying leaving groups (Cl- or OAc-) are formed by employing different quantities of the deprotonating agent NaOAc, and exhibit promising cytotoxicity against various cancer cell lines such as A549 and cisplatin-resistant A549/DDP lung cancer cells, as well as HeLa cells, with IC50 values spanning from 9.26 to 15.98 μM. Cytotoxicity and anticancer selectivity (SI: 1.9-2.4) of these metal complexes remain unaffected by variations in the metal center, leaving group, and ligand substitution. Further investigations reveal that these metal complexes specifically target mitochondria, leading to the depolarization of the mitochondrial membrane and instigating the production of intracellular reactive oxygen species. Furthermore, the metal complexes are found to induce late apoptosis and disrupt the cell cycle, leading to G2/M cell cycle arrest specifically in A549 cancer cells. In light of these findings, it is evident that the primary mechanism contributing to the anticancer effectiveness of these metal complexes is the redox pathway.
Collapse
Affiliation(s)
- Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China.
| | - Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Zhihong Jing
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Kangning Lai
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Hanxiu Fu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Heqian Dong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Zhihao Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China.
| |
Collapse
|
12
|
Lee LCC, Lo KKW. Shining New Light on Biological Systems: Luminescent Transition Metal Complexes for Bioimaging and Biosensing Applications. Chem Rev 2024; 124:8825-9014. [PMID: 39052606 PMCID: PMC11328004 DOI: 10.1021/acs.chemrev.3c00629] [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: 07/27/2024]
Abstract
Luminescence imaging is a powerful and versatile technique for investigating cell physiology and pathology in living systems, making significant contributions to life science research and clinical diagnosis. In recent years, luminescent transition metal complexes have gained significant attention for diagnostic and therapeutic applications due to their unique photophysical and photochemical properties. In this Review, we provide a comprehensive overview of the recent development of luminescent transition metal complexes for bioimaging and biosensing applications, with a focus on transition metal centers with a d6, d8, and d10 electronic configuration. We elucidate the structure-property relationships of luminescent transition metal complexes, exploring how their structural characteristics can be manipulated to control their biological behavior such as cellular uptake, localization, biocompatibility, pharmacokinetics, and biodistribution. Furthermore, we introduce the various design strategies that leverage the interesting photophysical properties of luminescent transition metal complexes for a wide variety of biological applications, including autofluorescence-free imaging, multimodal imaging, organelle imaging, biological sensing, microenvironment monitoring, bioorthogonal labeling, bacterial imaging, and cell viability assessment. Finally, we provide insights into the challenges and perspectives of luminescent transition metal complexes for bioimaging and biosensing applications, as well as their use in disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
13
|
Tan Z, Lin M, Liu J, Wu H, Chao H. Cyclometalated iridium(III) tetrazine complexes for mitochondria-targeted two-photon photodynamic therapy. Dalton Trans 2024; 53:12917-12926. [PMID: 39028267 DOI: 10.1039/d4dt01665c] [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: 07/20/2024]
Abstract
The fast-moving field of photodynamic therapy (PDT) has provided fresh opportunities to expand the potential of metallodrugs to combat cancers in a light-controlled manner. As such, in the present study, a series of cyclometalated Ir(III) complexes modified with a tetrazine functional group (namely, Ir-ppy-Tz, Ir-pbt-Tz, and Ir-dfppy-Tz) are developed as potential two-photon photodynamic anticancer agents. These complexes target mitochondria but exhibit low toxicity towards HLF primary lung fibroblast normal cells in the dark. When receiving a low-dose one- or two-photon PDT, they become highly potent towards A549 lung cancer cells (with IC50 values ranging from 24.0 nM to 96.0 nM) through the generation of reactive oxygen species (ROS) to induce mitochondrial damage and subsequent apoptosis. Our results indicated that the incorporation of tetrazine with cyclometalated Ir(III) matrices would increase the singlet oxygen (1O2) quantum yield (ΦΔ) and, meanwhile, enable a type I PDT mechanism. Ir-pbt-Tz, with the largest two-photon absorption (TPA) cross-section (σ2 = 102 GM), shows great promise in serving as a two-photon PDT agent for phototherapy.
Collapse
Affiliation(s)
- Zanru Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Mingwei Lin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Jiangping Liu
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361102, P. R. China.
| | - Huihui Wu
- Department of Dermatology, The East Division of the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510700, P. R. China.
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 400201, P. R. China
| |
Collapse
|
14
|
Wang WJ, Ling YY, Shi Y, Wu XW, Su X, Li ZQ, Mao ZW, Tan CP. Identification of mitochondrial ATP synthase as the cellular target of Ru-polypyridyl- β-carboline complexes by affinity-based protein profiling. Natl Sci Rev 2024; 11:nwae234. [PMID: 39114378 PMCID: PMC11304990 DOI: 10.1093/nsr/nwae234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Ruthenium polypyridyl complexes are promising anticancer candidates, while their cellular targets have rarely been identified, which limits their clinical application. Herein, we design a series of Ru(II) polypyridyl complexes containing bioactive β-carboline derivatives as ligands for anticancer evaluation, among which Ru5 shows suitable lipophilicity, high aqueous solubility, relatively high anticancer activity and cancer cell selectivity. The subsequent utilization of a photo-clickable probe, Ru5a, serves to validate the significance of ATP synthase as a crucial target for Ru5 through photoaffinity-based protein profiling. Ru5 accumulates in mitochondria, impairs mitochondrial functions and induces mitophagy and ferroptosis. Combined analysis of mitochondrial proteomics and RNA-sequencing shows that Ru5 significantly downregulates the expression of the chloride channel protein, and influences genes related to ferroptosis and epithelial-to-mesenchymal transition. Finally, we prove that Ru5 exhibits higher anticancer efficacy than cisplatin in vivo. We firstly identify the molecular targets of ruthenium polypyridyl complexes using a photo-click proteomic method coupled with a multiomics approach, which provides an innovative strategy to elucidate the anticancer mechanisms of metallo-anticancer candidates.
Collapse
Affiliation(s)
- Wen-Jin Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yu-Yi Ling
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yin Shi
- School of Pharmacy, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Xiao-Wen Wu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xuxian Su
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zheng-Qiu Li
- School of Pharmacy, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou 510006, China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
15
|
Chen SQ, Lu XY, Zhu LY, Zhu H, Li RT, Ye RR. Design, synthesis, and antitumor mechanism investigation of iridium(III) complexes conjugated with ibuprofen. J Inorg Biochem 2024; 257:112596. [PMID: 38759264 DOI: 10.1016/j.jinorgbio.2024.112596] [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: 02/26/2024] [Revised: 04/18/2024] [Accepted: 05/05/2024] [Indexed: 05/19/2024]
Abstract
The design and synthesis of a series of metal complexes formed by non-steroidal anti-inflammatory drugs (NSAIDs) ibuprofen (IBP) and iridium(III), with the molecular formula [Ir(C^N)2bpy(4-CH2OIBP-4'-CH2OIBP)](PF6) (Ir-IBP-1, Ir-IBP-2) (C^N = 2-phenylpyridine (ppy, Ir-IBP-1), 2-(2-thienyl)pyridine (thpy, Ir-IBP-2)) was introduced in this article. Firstly, it was found that the anti-proliferative activity of these complexes was more effective than that of cisplatin. Further research showed that Ir-IBP-1 and Ir-IBP-2 can accumulate in intracellular mitochondria, thereby disrupting mitochondrial membrane potential (MMP), increasing intracellular reactive oxygen species (ROS), blocking the G2/M phase of the cell cycle, and inducing cell apoptosis. In terms of protein expression, the expression of COX-2, MMP-9, NLRP3 and Caspase-1 proteins can be downregulated, indicating their ability to anti-inflammatory and overcome immune evasion. Furthermore, Ir-IBP-1 and Ir-IBP-2 can induce immunogenic cell death (ICD) by triggering the release of cell surface calreticulin (CRT), high mobility group box 1 (HMGB1) and adenosine triphosphate (ATP). Overall, iridium(III)-IBP conjugates exhibit various anti-tumor mechanisms, including mitochondrial damage, cell cycle arrest, inflammatory suppression, and induction of ICD.
Collapse
Affiliation(s)
- Si-Qin Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Xing-Yun Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Lin-Yuan Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Hou Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| |
Collapse
|
16
|
Chen Y, Liang C, Kou M, Tang X, Ru J. Lysosome-targeted cyclometalated Ir(III) complexes as photosensitizers/photoredox catalysts for cancer therapy. Dalton Trans 2024; 53:11836-11849. [PMID: 38949269 DOI: 10.1039/d4dt01345j] [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: 07/02/2024]
Abstract
A novel lysosome-targeted photosensitizer/photoredox catalyst based on cyclometalated Ir(III) complex IrL has been designed and synthesized, which exhibited excellent phosphorescence properties and the ability to generate single oxygen (1O2) and photocatalytically oxidize 1,4-dihydronicotinamide adenine dinucleotide (NADH) under light irradiation. Most importantly, the aforementioned activities are significantly enhanced due to protonation under acidic conditions, which makes them highly attractive in light-activated tumor therapy, especially for acidic lysosomes and tumor microenvironments. The photocytotoxicity of IrL and the mechanism of cell death have been investigated. Additionally, the tumor-killing ability of IrL under light irradiation was evaluated using a 4T1 tumor-bearing mouse model. This work provides a strategy for the development of lysosome-targeted photosensitizers/photoredox catalysts to overcome hypoxic tumors.
Collapse
Affiliation(s)
- Yu Chen
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China.
| | - Chao Liang
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China.
| | - Manchang Kou
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Xiaoliang Tang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Jiaxi Ru
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China.
| |
Collapse
|
17
|
Meng T, Shi X, Chen H, Xu Z, Qin W, Wei K, Yang X, Huang J, Liao C. Mitochondrial-targeted cyclometalated Ir(III)-5,7-dibromo/dichloro-2-methyl-8-hydroxyquinoline complexes and their anticancer efficacy evaluation in Hep-G2 cells. Metallomics 2024; 16:mfae032. [PMID: 38955388 DOI: 10.1093/mtomcs/mfae032] [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: 04/02/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Both 8-hydroxyquinoline compounds and iridium (Ir) complexes have emerged as potential novel agents for tumor therapy. In this study, we synthesized and characterized two new Ir(III) complexes, [Ir(L1)(bppy)2] (Br-Ir) and [Ir(L2)(bppy)2] (Cl-Ir), with 5,7-dibromo-2-methyl-8-hydroxyquinoline (HL-1) or 5,7-dichloro-2-methyl-8-hydroxyquinoline as the primary ligand. Complexes Br-Ir and Cl-Ir successfully inhibited antitumor activity in Hep-G2 cells. In addition, complexes Br-Ir and Cl-Ir were localized in the mitochondrial membrane and caused mitochondrial damage, autophagy, and cellular immunity in Hep-G2 cells. We tested the proteins related to mitochondrial and mitophagy by western blot analysis, which showed that they triggered mitophagy-mediated apoptotic cell death. Remarkably, complex Br-Ir showed high in vivo antitumor activity, and the tumor growth inhibition rate was 63.0% (P < 0.05). In summary, our study on complex Br-Ir revealed promising results in in vitro and in vivo antitumor activity assays.
Collapse
Affiliation(s)
- Ting Meng
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| | - Xiongzhi Shi
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, China
| | - Hongfen Chen
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| | - Zhong Xu
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| | - Weirong Qin
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| | - Kehua Wei
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| | - Xin Yang
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing Jiangsu, China
| | - Jin Huang
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| | - Chuanan Liao
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Pharmaceutical College, Guangxi Medical University, Nanning Guangxi, China
| |
Collapse
|
18
|
Hashemzadeh T, Christofferson AJ, White KF, Barnard PJ. Experimental and theoretical studies of pH-responsive iridium(III) complexes of azole and N-heterocyclic carbene ligands. Dalton Trans 2024; 53:8478-8493. [PMID: 38687288 DOI: 10.1039/d3dt03766e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
A series of nine luminescent iridium(III) complexes with pH-responsive imidazole and benzimidazole ligands have been prepared and characterized. The first series of complexes were of the form [Ir(ppy)2(N^N)]+ or [Ir(ppy)2(C^N)]+ (where ppy is 2-phenylpyridine and N^N is 2-(2-pyridyl)imidazole or 2-(2-pyridyl)benzimidazole and C^N represents a pyridyl-triazolylidene-based N-heterocyclic carbene ligand). For these complexes, the benzimidazole group was either unsubstituted or substituted with electron-withdrawing (Cl) or electron-donating (Me) groups. The second series of complexes were of the form [Ir(phbim)2(N^N)]+ or [Ir(phbim)2(C^N)]+ (where phbim is 2-phenylbenzimidazole and N^N is either 2,2'-bipyridine or 1,10-phenanthroline and C^N is either a pyridyl-imidazolylidene or pyridyl-triazolylidene N-heterocyclic carbene ligand). UV-visible and photoluminescence pH titration studies showed that changing the protonation state of these complexes results in significant changes in the photoluminescence emission properties. The pKa values of prepared complexes were estimated from the spectroscopic pH titration data and these values show that the nature of the pH-sensitive ligands (either main or ancillary ligands) resulted in a significant capacity to modulate the pKa values for these compounds with values ranging from 5.19-11.22. Theoretical investigations into the nature of the electronic transitions for the different protonation states of compounds were performed and the results were consistent with the experimental results.
Collapse
Affiliation(s)
- Tahmineh Hashemzadeh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia.
| | - Andrew J Christofferson
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
- ARC Centre of Excellence in Exciton Science, School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | - Keith F White
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia.
| | - Peter J Barnard
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia.
| |
Collapse
|
19
|
Luo Z, Zhou Z, Pan Y, Zhu Z, Yuan H, Li Y, Feng S, Hong Y, Xu L. Cell-penetrating peptides noncovalently modified red phosphorescent nanoparticles for high-efficiency imaging. RSC Adv 2024; 14:11891-11899. [PMID: 38623284 PMCID: PMC11017195 DOI: 10.1039/d4ra01531b] [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: 02/28/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
The application of long-lived phosphorescence probes in time-resolved luminescence imaging is limited by their low quantum yield in aqueous solutions. However, sensitization of thermally activated delayed fluorescence (TADF) materials can compensate for this limitation while addressing the issue of insufficient proportion of their own long lifetime. In this study, we utilized the characteristics of phosphorescence and TADF materials simultaneously by doping the receptor iridium complex PMD-Ir into the donor TADF polymer PCzDP-20 through donor-receptor doping method, and successfully prepared highly efficient red phosphorescent nanoparticles. The quantum yield of the nanoparticles obtained by this method reaches up to 30%, and the luminescence lifetime can reach several thousand nanoseconds. Additionally, due to the low concentration doping of PMD-Ir, the risk of transition metal toxicity is greatly reduced. Furthermore, we used non-covalent modification with amphiphilic cell-penetrating peptides (CPPs) to increase the cell membrane permeability of the nanoparticles. The CPPs modified nanoparticles achieve in vivo confocal imaging of zebrafish and intracellular time-resolved imaging by its significantly improved bioimaging capabilities. The functional nanoparticles designing method fully utilizes the characteristics of PMD-Ir, PCzDP-20, and CPPs, solving the problems of low quantum yield and poor membrane permeability of Ir-complex nanoparticles. This will greatly promote the development of time-resolved luminescence imaging.
Collapse
Affiliation(s)
- Zihan Luo
- Department of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| | - Zhuofan Zhou
- Department of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| | - Yiwen Pan
- Department of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| | - Zece Zhu
- School of Bioengineering and Health, State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University Wuhan 430200 P. R. China
| | - Huanxiang Yuan
- Department of Chemistry, College of Chemistry and Materials Engineering, Beijing Technology and Business University Beijing 100048 P. R. China
| | - Yutao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| | - Shumin Feng
- Department of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| | - Yi Hong
- Department of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| | - Li Xu
- Department of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 P. R. China
| |
Collapse
|
20
|
Das U, Paira P. Exploring the phototoxicity of GSH-resistant 2-(5,6-dichloro-1 H-benzo[ d]imidazol-2-yl)quinoline-based Ir(III)-PTA complexes in MDA-MB-231 cancer cells. Dalton Trans 2024; 53:6459-6471. [PMID: 38512047 DOI: 10.1039/d3dt04361d] [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: 03/22/2024]
Abstract
Metal complexes play a crucial role in photo-activated chemotherapy (PACT), which has recently been used to treat specific disorders. Triple-negative breast cancer has an enormously high rate of relapse due to the existence and survival of cancer stem cells (CSCs) characterized by increased amounts of glutathione (GSH). Hence, designing a phototoxic molecule is an enticing area of research to combat triple-negative breast cancer (TNBC) via GSH depletion and DNA photocleavage. Herein, we focus on the application of PTA and non-PTA Ir(III) complexes for phototoxicity in the absence and presence of GSH against MDA-MB-231 TNBC cells. Between these two complexes, [Cp*IrIII(DD)PTA]·2Cl (DDIRP) exhibited better phototoxicity (IC50 ∼ 2.80 ± 0.52 μM) compared to the non-PTA complex [Cp*IrIII(DD)Cl]·Cl (DDIR) against TNBC cells because of the high GSH resistance power of the complex DDIRP. The significant potency of the complex DDIRP under photo irradiation in both normoxia and hypoxia conditions can be attributed to selective transportation, high cellular permeability and uptake towards the nucleus, GSH depletion by GSH-GSSG conversion, the ability of strong DNA binding including intercalation, and oxidative stress. The strong affinity to serum albumin, which serves as a carrier protein, aids in the transport of the complex to its target site while preventing glutathione (GSH) deactivation. Consequently, the complex DDIRP was developed as a suitable phototoxic complex in selective cancer therapy, ruling over the usual chemotherapeutic drug cisplatin and the PDT drug Photofrin. The ability of ROS generation under hypoxic conditions delivers this complex as a hypoxia-efficient selective metallodrug for the treatment of TNBC.
Collapse
Affiliation(s)
- Utpal Das
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore-632014, Tamil Nadu, India.
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore-632014, Tamil Nadu, India.
| |
Collapse
|
21
|
Martínez-Alonso M, Jones CG, Shipp JD, Chekulaev D, Bryant HE, Weinstein JA. Phototoxicity of cyclometallated Ir(III) complexes bearing a thio-bis-benzimidazole ligand, and its monodentate analogue, as potential PDT photosensitisers in cancer cell killing. J Biol Inorg Chem 2024; 29:113-125. [PMID: 38183420 PMCID: PMC11001735 DOI: 10.1007/s00775-023-02031-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/18/2023] [Indexed: 01/08/2024]
Abstract
Two novel cyclometallated iridium(III) complexes have been prepared with one bidentate or two monodentate imidazole-based ligands, 1 and 2, respectively. The complexes showed intense emission with long lifetimes of the excited state. Femtosecond transient absorption experiments established the nature of the lowest excited state as 3IL state. Singlet oxygen generation with good yields (40% for 1 and 82% for 2) was established by detecting 1O2 directly, through its emission at 1270 nm. Photostability studies were also performed to assess the viability of the complexes as photosensitizers (PS) for photodynamic therapy (PDT). Complex 1 was selected as a good candidate to investigate light-activated killing of cells, whilst complex 2 was found to be toxic in the dark and unstable under light. Complex 1 demonstrated high phototoxicity indexes (PI) in the visible region, PI > 250 after irradiation at 405 nm and PI > 150 at 455 nm, in EJ bladder cancer cells.
Collapse
Affiliation(s)
- Marta Martínez-Alonso
- Department of Chemistry, University of Sheffield, Sheffield, S3 7HF, UK
- Department of Oncology and Metabolism, Medical School, The University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Callum G Jones
- Department of Chemistry, University of Sheffield, Sheffield, S3 7HF, UK
- Department of Oncology and Metabolism, Medical School, The University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - James D Shipp
- Department of Chemistry, University of Sheffield, Sheffield, S3 7HF, UK
| | - Dimitri Chekulaev
- Department of Chemistry, University of Sheffield, Sheffield, S3 7HF, UK
| | - Helen E Bryant
- Department of Oncology and Metabolism, Medical School, The University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | - Julia A Weinstein
- Department of Chemistry, University of Sheffield, Sheffield, S3 7HF, UK.
| |
Collapse
|
22
|
Li P, Guo L, Li J, Yang Z, Fu H, Lai K, Dong H, Fan C, Liu Z. Mitochondria-targeted neutral and cationic iridium(III) anticancer complexes chelating simple hybrid sp 2-N/sp 3-N donor ligands. Dalton Trans 2024; 53:1977-1988. [PMID: 38205595 DOI: 10.1039/d3dt03700b] [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: 01/12/2024]
Abstract
Most platinum group-based cyclometalated neutral and cationic anticancer complexes with the general formula [(C^N)2Ir(XY)]0/+ (neutral complex: XY = bidentate anionic ligand; cationic complex: XY = bidentate neutral ligand) are notable owing to their intrinsic luminescence properties, good cell permeability, interaction with some biomolecular targets and unique mechanisms of action (MoAs). We herein synthesized a series of neutral and cationic amine-imine cyclometalated iridium(III) complexes using Schiff base ligands with sp2-N/sp3-N N^NH2 chelating donors. The cyclometalated iridium(III) complexes were identified by various techniques. They were stable in aqueous media, displayed moderate fluorescence and exhibited affinity toward bovine serum albumin (BSA). The complexes demonstrated promising cytotoxicity against lung cancer A549 cells, cisplatin-resistant lung cancer A549/DDP cells, cervical carcinoma HeLa cells and human liver carcinoma HepG2 cells, with IC50 values ranging from 9.98 to 19.63 μM. Unfortunately, these complexes had a low selectivity (selectivity index: 1.62-1.98) towards A549 cells and BEAS-2B normal cells. The charge pattern of the metal center (neutral or cationic) and ligand substituents showed little influence on the cytotoxicity and selectivity of these complexes. The study revealed that these complexes could target mitochondria, cause depolarization of the mitochondrial membrane, and trigger the production of intracellular ROS. Additionally, the complexes were observed to induce late apoptosis and perturb the cell cycle in the G2/M or S phase in A549 cells. Based on these results, it appears that the anticancer efficacy of these complexes was predominantly attributed to the redox mechanism.
Collapse
Affiliation(s)
- Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Jiaxing Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Zhihao Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Hanxiu Fu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Kangning Lai
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Heqian Dong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Chunyan Fan
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, Institute of Anticancer Agents Development and Theranostic Application, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China.
| |
Collapse
|
23
|
Kasparkova J, Hernández-García A, Kostrhunova H, Goicuría M, Novohradsky V, Bautista D, Markova L, Santana MD, Brabec V, Ruiz J. Novel 2-(5-Arylthiophen-2-yl)-benzoazole Cyclometalated Iridium(III) dppz Complexes Exhibit Selective Phototoxicity in Cancer Cells by Lysosomal Damage and Oncosis. J Med Chem 2024; 67:691-708. [PMID: 38141031 PMCID: PMC10788912 DOI: 10.1021/acs.jmedchem.3c01978] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
A second-generation series of biscyclometalated 2-(5-aryl-thienyl)-benzimidazole and -benzothiazole Ir(III) dppz complexes [Ir(C^N)2(dppz)]+, Ir1-Ir4, were rationally designed and synthesized, where the aryl group attached to the thienyl ring was p-CF3C6H4 or p-Me2NC6H4. These new Ir(III) complexes were assessed as photosensitizers to explore the structure-activity correlations for their potential use in biocompatible anticancer photodynamic therapy. When irradiated with blue light, the complexes exhibited high selective potency across several cancer cell lines predisposed to photodynamic therapy; the benzothiazole derivatives (Ir1 and Ir2) were the best performers, Ir2 being also activatable with green or red light. Notably, when irradiated, the complexes induced leakage of lysosomal content into the cytoplasm of HeLa cancer cells and induced oncosis-like cell death. The capability of the new Ir complexes to photoinduce cell death in 3D HeLa spheroids has also been demonstrated. The investigated Ir complexes can also catalytically photo-oxidate NADH and photogenerate 1O2 and/or •OH in cell-free media.
Collapse
Affiliation(s)
- Jana Kasparkova
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Alba Hernández-García
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| | - Hana Kostrhunova
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Marta Goicuría
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| | - Vojtěch Novohradsky
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | | | - Lenka Markova
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - María Dolores Santana
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| | - Viktor Brabec
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - José Ruiz
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| |
Collapse
|
24
|
Bai Y, Aodeng G, Ga L, Hai W, Ai J. Research Progress of Metal Anticancer Drugs. Pharmaceutics 2023; 15:2750. [PMID: 38140091 PMCID: PMC10747151 DOI: 10.3390/pharmaceutics15122750] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer treatments, including traditional chemotherapy, have failed to cure human malignancies. The main reasons for the failure of these treatments are the inevitable drug resistance and serious side effects. In clinical treatment, only 5 percent of the 50 percent of cancer patients who are able to receive conventional chemotherapy survive. Because of these factors, being able to develop a drug and treatment that can target only cancer cells without affecting normal cells remains a big challenge. Since the special properties of cisplatin in the treatment of malignant tumors were accidentally discovered in the last century, metal anticancer drugs have become a research hotspot. Metal anticancer drugs have unique pharmaceutical properties, such as ruthenium metal drugs with their high selectivity, low toxicity, easy absorption by tumor tissue, excretion, and so on. In recent years, efficient and low-toxicity metal antitumor complexes have been synthesized. In this paper, the scientific literature on platinum (Pt), ruthenium (Ru), iridium (Ir), gold (Au), and other anticancer complexes was reviewed by referring to a large amount of relevant literature at home and abroad.
Collapse
Affiliation(s)
- Yun Bai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Gerile Aodeng
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Lu Ga
- College of Pharmacy, Inner Mongolia Medical University, Jinchuankaifaqu, Hohhot 010110, China;
| | - Wenfeng Hai
- Inner Mongolia Key Laboratory of Carbon Nanomaterials, Nano Innovation Institute (NII), College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Jun Ai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| |
Collapse
|
25
|
Joshi B, Shivashankar M. Recent Advancement in the Synthesis of Ir-Based Complexes. ACS OMEGA 2023; 8:43408-43432. [PMID: 38027378 PMCID: PMC10666285 DOI: 10.1021/acsomega.3c04867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/20/2023] [Indexed: 12/01/2023]
Abstract
Cancer is a devastating disease with over 100 types, including lung and breast cancer. Cisplatin and metal-based drugs are limited due to their drug resistance and side effects. Iridium-based compounds have emerged as promising candidates due to their unique chemical properties and resemblance to platinum compounds. The objective of this study is to investigate the synthesis and categorization of iridium complexes, with a particular emphasis on their potential use as anticancer agents. The major focus of this research is to examine the synthesis of these complexes and their relevance to the field of cancer treatment. The negligible side effects and flexibility of cyclometalated iridium(III) complexes have garnered significant interest. Organometallic half-sandwich Ir(III) complexes have notable benefits in cancer research and treatment. The review places significant emphasis on categorizing iridium complexes according to their ligand environment, afterward considering the ligand density and coordination number. This study primarily focuses on several methods for synthesizing cyclometalated and half-sandwich Ir complexes, divided into subgroups based on ligand denticity. The coordination number of iridium complexes determines the number of ligands coordinated to the central iridium atom, which impacts their stability and reactivity. Understanding these complexes is crucial for designing compounds with desired properties and investigating their potential as anticancer agents. Cyclometalated iridium(III) complexes, which contain a meta-cycle with the E-M-C order σ bond, were synthesized in 1999. These complexes have high quantum yields, significant stock shifts, luminescence qualities, cell permeability, and strong photostability. They have been promising in biosensing, bioimaging, and phosphorescence of heavy metal complexes.
Collapse
Affiliation(s)
- Bhumika Joshi
- Department of Chemistry,
School of Advance Science, VIT University, Vellore 632014, India
| | - Murugesh Shivashankar
- Department of Chemistry,
School of Advance Science, VIT University, Vellore 632014, India
| |
Collapse
|
26
|
Wang Y, Luo YZ, Liu ZJ, Yao ZJ. Cationic N,S-chelate half-sandwich iridium complexes: synthesis, characterization, anticancer and antiplasmodial activity. Biomater Sci 2023; 11:7090-7098. [PMID: 37667825 DOI: 10.1039/d3bm01027a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
A series of pyrazole-based ligands and their corresponding cationic N,S-chelate half-sandwich iridium complexes were successfully synthesized. All iridium complexes exhibited good anticancer activity against the MCF-7 and MDA-MB-231 human breast cancer cells. The cytotoxic activity of unsubstituted iridium complex 1 is greater than that of cisplatin against MCF-7 cells. In addition, the cationic half-sandwich iridium complexes are also efficient in antiplasmodial study and complex 1 displayed the best activity as its IC50 was observed to be approximately 0.11 μM against the CQS-NF54 strain. These iridium complexes generally exhibited enhanced activity against the CQS-NF54 strain in comparison with that against the CQR-K1 strain. An "IC50 speed assay" investigation against the CQS-NF54 strain indicated complexes 1-3 to be fast-acting complexes that reach their lowest IC50 values within 16 hours. All complexes were fully characterized by IR spectroscopy, NMR spectroscopy, and elemental analysis, and the structure of the iridium complex was confirmed by single-crystal X-ray diffraction.
Collapse
Affiliation(s)
- Yang Wang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China.
| | - Yu-Zhou Luo
- Scientific Research Office, Guangzhou College of Commerce, Guangzhou, 511363, China.
| | - Zhen-Jiang Liu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China.
| | - Zi-Jian Yao
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China.
| |
Collapse
|
27
|
Zhu J, Liu Y, Zhang Z, Yang X, Qiu F. Cyclometalated Ir(III) Complexes as Lysosome-Targeted Photodynamic Anticancer Agents. ACS OMEGA 2023; 8:34557-34563. [PMID: 37779987 PMCID: PMC10536871 DOI: 10.1021/acsomega.3c03234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023]
Abstract
We have designed and synthesized two Ir(III) complexes (Ir1 and Ir2) coordinated with an 8-sulfonamidoquinoline derivative ligand as photosensitizers, which exhibit strong red phosphorescence emission and a long phosphorescence lifetime. The Ir(III) complexes exhibit a high population of triplet states, which enable red phosphorescence and efficient singlet oxygen generation. Ir1 and Ir2 rapidly enter the cancer cells and accumulate in lysosomes, producing large amounts of intracellular singlet oxygen when exposed to light irradiation, eventually leading to cancer cell death, and the phototoxic indexes of complexes Ir1 and Ir2 against cancer cells are in the range of 76-228. Overall, our studies indicate that the synthesized Ir(III) complexes with quinoline ligands exhibit photosensitizing properties, effectively inducing cancer cell death when exposed to light. These promising results suggest their potential application in photodynamic therapy.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department
of Cardiology, The First People’s
Hospital of Foshan, No. 81 Lingnan RD, Foshan 528000, China
| | - Yan Liu
- Guangdong
Provincial Key Laboratory of Sensor Technology and Biomedical Instrument,
School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhao Zhang
- Department
of Biochemistry and Molecular Biology, Guangdong
Medical University, Zhanjiang 524023, China
| | - Xili Yang
- Department
of Cardiology, The First People’s
Hospital of Foshan, No. 81 Lingnan RD, Foshan 528000, China
| | - Feng Qiu
- Department
of Laboratory Medicine, The Seventh Affiliated
Hospital of Southern Medical University, Foshan 516006, China
| |
Collapse
|
28
|
Das U, Paira P. Synthesis, characterization, photophysical and electrochemical properties, and biomolecular interaction of 2,2'-biquinoline based phototoxic Ru(II)/Ir(II) complexes. Dalton Trans 2023; 52:12608-12617. [PMID: 37314097 DOI: 10.1039/d3dt01348k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The phototoxic nature of drugs has been seen to convey immense importance in photo activated chemotherapy (PACT) for the selective treatment of disease. Rationally, in order to eradicate the vehemence of cancer in a living body, the design of phototoxic molecules has been of growing interest in research to establish a selective strategy for cancer therapy. Therefore, the present work portrays the synthesis of a phototoxic anticancer agent by incorporating ruthenium(II) and iridium(III) metals into a biologically active 2,2'-biquinoline moiety, BQ. The complexes, RuBQ and IrBQ, have been revealed as effective anticancer agents with remarkable toxicity in the presence of light compared to the dark towards HeLa and MCF-7 cancer cell lines due to the production of a profuse amount of singlet oxygen (1O2) upon irradiation by visible light (400-700 nm). Complex IrBQ exhibited the best toxicity (IC50 = 8.75 μM in MCF-7 and 7.23 μM in HeLa) in comparison to the RuBQ complex under visible light. RuBQ and IrBQ displayed considerable quantum yields (Φf) along with a good lipophilic property, indicating the cellular imaging capability of both complexes upon significant accumulation in cancer cells. Also, the complexes have shown significant binding propensity with biomolecules, viz. deoxyribonucleic acid (DNA) as well as serum albumin (BSA, HSA).
Collapse
Affiliation(s)
- Utpal Das
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology Vellore-632014, Tamilnadu, India.
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology Vellore-632014, Tamilnadu, India.
| |
Collapse
|
29
|
Wang H, Li Q, Alam P, Bai H, Bhalla V, Bryce MR, Cao M, Chen C, Chen S, Chen X, Chen Y, Chen Z, Dang D, Ding D, Ding S, Duo Y, Gao M, He W, He X, Hong X, Hong Y, Hu JJ, Hu R, Huang X, James TD, Jiang X, Konishi GI, Kwok RTK, Lam JWY, Li C, Li H, Li K, Li N, Li WJ, Li Y, Liang XJ, Liang Y, Liu B, Liu G, Liu X, Lou X, Lou XY, Luo L, McGonigal PR, Mao ZW, Niu G, Owyong TC, Pucci A, Qian J, Qin A, Qiu Z, Rogach AL, Situ B, Tanaka K, Tang Y, Wang B, Wang D, Wang J, Wang W, Wang WX, Wang WJ, Wang X, Wang YF, Wu S, Wu Y, Xiong Y, Xu R, Yan C, Yan S, Yang HB, Yang LL, Yang M, Yang YW, Yoon J, Zang SQ, Zhang J, Zhang P, Zhang T, Zhang X, Zhang X, Zhao N, Zhao Z, Zheng J, Zheng L, Zheng Z, Zhu MQ, Zhu WH, Zou H, Tang BZ. Aggregation-Induced Emission (AIE), Life and Health. ACS NANO 2023; 17:14347-14405. [PMID: 37486125 PMCID: PMC10416578 DOI: 10.1021/acsnano.3c03925] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Light has profoundly impacted modern medicine and healthcare, with numerous luminescent agents and imaging techniques currently being used to assess health and treat diseases. As an emerging concept in luminescence, aggregation-induced emission (AIE) has shown great potential in biological applications due to its advantages in terms of brightness, biocompatibility, photostability, and positive correlation with concentration. This review provides a comprehensive summary of AIE luminogens applied in imaging of biological structure and dynamic physiological processes, disease diagnosis and treatment, and detection and monitoring of specific analytes, followed by representative works. Discussions on critical issues and perspectives on future directions are also included. This review aims to stimulate the interest of researchers from different fields, including chemistry, biology, materials science, medicine, etc., thus promoting the development of AIE in the fields of life and health.
Collapse
Affiliation(s)
- Haoran Wang
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Qiyao Li
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
- State
Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial
Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| | - Parvej Alam
- Clinical
Translational Research Center of Aggregation-Induced Emission, School
of Medicine, The Second Affiliated Hospital, School of Science and
Engineering, The Chinese University of Hong
Kong, Shenzhen (CUHK- Shenzhen), Guangdong 518172, China
| | - Haotian Bai
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Organic
Solids, Institute of Chemistry, Chinese
Academy of Sciences, Beijing 100190, China
| | - Vandana Bhalla
- Department
of Chemistry, Guru Nanak Dev University, Amritsar 143005, India
| | - Martin R. Bryce
- Department
of Chemistry, Durham University, South Road, Durham DH1 3LE, United Kingdom
| | - Mingyue Cao
- State
Key Laboratory of Crystal Materials, Shandong
University, Jinan 250100, China
| | - Chao Chen
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Sijie Chen
- Ming
Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sha Tin, Hong Kong SAR 999077, China
| | - Xirui Chen
- State Key
Laboratory of Food Science and Resources, School of Food Science and
Technology, Nanchang University, Nanchang 330047, China
| | - Yuncong Chen
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), Department of Cardiothoracic Surgery, Nanjing Drum Tower
Hospital, Medical School, Nanjing University, Nanjing 210023, China
| | - Zhijun Chen
- Engineering
Research Center of Advanced Wooden Materials and Key Laboratory of
Bio-based Material Science and Technology of Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Dongfeng Dang
- School
of Chemistry, Xi’an Jiaotong University, Xi’an 710049 China
| | - Dan Ding
- State
Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive
Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Siyang Ding
- Department
of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Yanhong Duo
- Department
of Radiation Oncology, Shenzhen People’s Hospital (The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China
| | - Meng Gao
- National
Engineering Research Center for Tissue Restoration and Reconstruction,
Key Laboratory of Biomedical Engineering of Guangdong Province, Key
Laboratory of Biomedical Materials and Engineering of the Ministry
of Education, Innovation Center for Tissue Restoration and Reconstruction,
School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Wei He
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Xuewen He
- The
Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College
of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren’ai Road, Suzhou 215123, China
| | - Xuechuan Hong
- State
Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital
of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuning Hong
- Department
of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Jing-Jing Hu
- State
Key Laboratory of Biogeology and Environmental Geology, Engineering
Research Center of Nano-Geomaterials of Ministry of Education, Faculty
of Materials Science and Chemistry, China
University of Geosciences, Wuhan 430074, China
| | - Rong Hu
- School
of Chemistry and Chemical Engineering, University
of South China, Hengyang 421001, China
| | - Xiaolin Huang
- State Key
Laboratory of Food Science and Resources, School of Food Science and
Technology, Nanchang University, Nanchang 330047, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Xingyu Jiang
- Guangdong
Provincial Key Laboratory of Advanced Biomaterials, Shenzhen Key Laboratory
of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
| | - Gen-ichi Konishi
- Department
of Chemical Science and Engineering, Tokyo
Institute of Technology, O-okayama, Meguro-ku, Tokyo 152-8552, Japan
| | - Ryan T. K. Kwok
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Jacky W. Y. Lam
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Chunbin Li
- College
of Chemistry and Chemical Engineering, Inner Mongolia Key Laboratory
of Fine Organic Synthesis, Inner Mongolia
University, Hohhot 010021, China
| | - Haidong Li
- State
Key Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Kai Li
- College
of Chemistry, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Nan Li
- Key
Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory
of Applied Surface and Colloid Chemistry of Ministry of Education,
School of Chemistry & Chemical Engineering, Shaanxi Normal University, Xi’an 710119, China
| | - Wei-Jian Li
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes & Chang-Kung
Chuang Institute, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200062, China
| | - Ying Li
- Innovation
Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal
and Guangdong Provincial Key Laboratory of Molecular Target &
Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory
Disease, School of Pharmaceutical Sciences and the Fifth Affiliated
Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xing-Jie Liang
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- School
of Biomedical Engineering, Guangzhou Medical
University, Guangzhou 511436, China
| | - Yongye Liang
- Department
of Materials Science and Engineering, Shenzhen Key Laboratory of Printed
Organic Electronics, Southern University
of Science and Technology, Shenzhen 518055, China
| | - Bin Liu
- Department
of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Guozhen Liu
- Ciechanover
Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK- Shenzhen), Guangdong 518172, China
| | - Xingang Liu
- Department
of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Xiaoding Lou
- State
Key Laboratory of Biogeology and Environmental Geology, Engineering
Research Center of Nano-Geomaterials of Ministry of Education, Faculty
of Materials Science and Chemistry, China
University of Geosciences, Wuhan 430074, China
| | - Xin-Yue Lou
- International
Joint Research Laboratory of Nano-Micro Architecture Chemistry, College
of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Liang Luo
- National
Engineering Research Center for Nanomedicine, College of Life Science
and Technology, Huazhong University of Science
and Technology, Wuhan 430074, China
| | - Paul R. McGonigal
- Department
of Chemistry, University of York, Heslington, York YO10 5DD, United
Kingdom
| | - Zong-Wan Mao
- MOE
Key Laboratory of Bioinorganic and Synthetic Chemistry, School of
Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Guangle Niu
- State
Key Laboratory of Crystal Materials, Shandong
University, Jinan 250100, China
| | - Tze Cin Owyong
- Department
of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Andrea Pucci
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via Moruzzi 13, Pisa 56124, Italy
| | - Jun Qian
- State
Key Laboratory of Modern Optical Instrumentations, Centre for Optical
and Electromagnetic Research, College of Optical Science and Engineering,
International Research Center for Advanced Photonics, Zhejiang University, Hangzhou 310058, China
| | - Anjun Qin
- State
Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial
Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| | - Zijie Qiu
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Andrey L. Rogach
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Bo Situ
- Department
of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kazuo Tanaka
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura,
Nishikyo-ku, Kyoto 615-8510, Japan
| | - Youhong Tang
- Institute
for NanoScale Science and Technology, College of Science and Engineering, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Bingnan Wang
- State
Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial
Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| | - Dong Wang
- Center
for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jianguo Wang
- College
of Chemistry and Chemical Engineering, Inner Mongolia Key Laboratory
of Fine Organic Synthesis, Inner Mongolia
University, Hohhot 010021, China
| | - Wei Wang
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes & Chang-Kung
Chuang Institute, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200062, China
| | - Wen-Xiong Wang
- School
of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Wen-Jin Wang
- MOE
Key Laboratory of Bioinorganic and Synthetic Chemistry, School of
Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Central
Laboratory of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-
Shenzhen), & Longgang District People’s Hospital of Shenzhen, Guangdong 518172, China
| | - Xinyuan Wang
- Department
of Materials Science and Engineering, Shenzhen Key Laboratory of Printed
Organic Electronics, Southern University
of Science and Technology, Shenzhen 518055, China
| | - Yi-Feng Wang
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- School
of Biomedical Engineering, Guangzhou Medical
University, Guangzhou 511436, China
| | - Shuizhu Wu
- State
Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial
Key Laboratory of Luminescence from Molecular Aggregates, College
of Materials Science and Engineering, South
China University of Technology, Wushan Road 381, Guangzhou 510640, China
| | - Yifan Wu
- Innovation
Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal
and Guangdong Provincial Key Laboratory of Molecular Target &
Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory
Disease, School of Pharmaceutical Sciences and the Fifth Affiliated
Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Yonghua Xiong
- State Key
Laboratory of Food Science and Resources, School of Food Science and
Technology, Nanchang University, Nanchang 330047, China
| | - Ruohan Xu
- School
of Chemistry, Xi’an Jiaotong University, Xi’an 710049 China
| | - Chenxu Yan
- Key
Laboratory for Advanced Materials and Joint International Research,
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals,
Frontiers Science Center for Materiobiology and Dynamic Chemistry,
School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Saisai Yan
- Center
for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Hai-Bo Yang
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes & Chang-Kung
Chuang Institute, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200062, China
| | - Lin-Lin Yang
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Mingwang Yang
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Ying-Wei Yang
- International
Joint Research Laboratory of Nano-Micro Architecture Chemistry, College
of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Juyoung Yoon
- Department
of Chemistry and Nanoscience, Ewha Womans
University, Seoul 03760, Korea
| | - Shuang-Quan Zang
- College
of Chemistry, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Jiangjiang Zhang
- Guangdong
Provincial Key Laboratory of Advanced Biomaterials, Shenzhen Key Laboratory
of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- Key
Laboratory of Molecular Medicine and Biotherapy, the Ministry of Industry
and Information Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Pengfei Zhang
- Guangdong
Key Laboratory of Nanomedicine, Shenzhen, Engineering Laboratory of
Nanomedicine and Nanoformulations, CAS Key Lab for Health Informatics,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, University Town of Shenzhen, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Tianfu Zhang
- School
of Biomedical Engineering, Guangzhou Medical
University, Guangzhou 511436, China
| | - Xin Zhang
- Department
of Chemistry, Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang Province 310030, China
- Westlake
Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, Zhejiang Province 310024, China
| | - Xin Zhang
- Ciechanover
Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK- Shenzhen), Guangdong 518172, China
| | - Na Zhao
- Key
Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory
of Applied Surface and Colloid Chemistry of Ministry of Education,
School of Chemistry & Chemical Engineering, Shaanxi Normal University, Xi’an 710119, China
| | - Zheng Zhao
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Jie Zheng
- Department
of Chemical, Biomolecular, and Corrosion Engineering The University of Akron, Akron, Ohio 44325, United States
| | - Lei Zheng
- Department
of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zheng Zheng
- School of
Chemistry and Chemical Engineering, Hefei
University of Technology, Hefei 230009, China
| | - Ming-Qiang Zhu
- Wuhan
National
Laboratory for Optoelectronics, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wei-Hong Zhu
- Key
Laboratory for Advanced Materials and Joint International Research,
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals,
Frontiers Science Center for Materiobiology and Dynamic Chemistry,
School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hang Zou
- Department
of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ben Zhong Tang
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| |
Collapse
|
30
|
Ortega-Forte E, Rovira A, López-Corrales M, Hernández-García A, Ballester FJ, Izquierdo-García E, Jordà-Redondo M, Bosch M, Nonell S, Santana MD, Ruiz J, Marchán V, Gasser G. A near-infrared light-activatable Ru(ii)-coumarin photosensitizer active under hypoxic conditions. Chem Sci 2023; 14:7170-7184. [PMID: 37416722 PMCID: PMC10321499 DOI: 10.1039/d3sc01844j] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Photodynamic therapy (PDT) represents a promising approach for cancer treatment. However, the oxygen dependency of PDT to generate reactive oxygen species (ROS) hampers its therapeutic efficacy, especially against hypoxic solid tumors. In addition, some photosensitizers (PSs) have dark toxicity and are only activatable with short wavelengths such as blue or UV-light, which suffer from poor tissue penetration. Herein, we developed a novel hypoxia-active PS with operability in the near-infrared (NIR) region based on the conjugation of a cyclometalated Ru(ii) polypyridyl complex of the type [Ru(C^N)(N^N)2] to a NIR-emitting COUPY dye. The novel Ru(ii)-coumarin conjugate exhibits water-solubility, dark stability in biological media and high photostability along with advantageous luminescent properties that facilitate both bioimaging and phototherapy. Spectroscopic and photobiological studies revealed that this conjugate efficiently generates singlet oxygen and superoxide radical anions, thereby achieving high photoactivity toward cancer cells upon highly-penetrating 740 nm light irradiation even under hypoxic environments (2% O2). The induction of ROS-mediated cancer cell death upon low-energy wavelength irradiation along with the low dark toxicity exerted by this Ru(ii)-coumarin conjugate could circumvent tissue penetration issues while alleviating the hypoxia limitation of PDT. As such, this strategy could pave the way to the development of novel NIR- and hypoxia-active Ru(ii)-based theragnostic PSs fuelled by the conjugation of tunable, low molecular-weight COUPY fluorophores.
Collapse
Affiliation(s)
- Enrique Ortega-Forte
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Anna Rovira
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
| | - Marta López-Corrales
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
| | - Alba Hernández-García
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Francisco José Ballester
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Eduardo Izquierdo-García
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology F-75005 Paris France
| | - Mireia Jordà-Redondo
- Institut Químic de Sarrià, Universitat Ramon Llull Vía Augusta 390 E-08017 Barcelona Spain
| | - Manel Bosch
- Unitat de Microscòpia Òptica Avançada, Centres Científics i Tecnològics, Universitat de Barcelona Av. Diagonal 643 E-08028 Barcelona Spain
| | - Santi Nonell
- Institut Químic de Sarrià, Universitat Ramon Llull Vía Augusta 390 E-08017 Barcelona Spain
| | - María Dolores Santana
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - José Ruiz
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Vicente Marchán
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology F-75005 Paris France
| |
Collapse
|
31
|
Aggregation-Induced emission photosensitizer with lysosomal response for photodynamic therapy against cancer. Bioorg Chem 2023; 132:106349. [PMID: 36716644 DOI: 10.1016/j.bioorg.2023.106349] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/26/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Photosensitizers play a key role in bioimaging and photodynamic therapy (PDT) of cancer. However, conventional photosensitizers usually do not achieve the desired efficacy in PDT due to their poor photostability, targeting ability, and responsiveness. Herein, we designed a series of photosensitizers with aggregation-induced emission (AIE) effect using benzothiazole- triphenylamine (BZT-triphenylamine) as the parent nucleus. The synthesized compound SIN ((E)-2-(4-(diphenylamino)styryl)-3-(4-iodobutyl)benzo[d]thiazol-3-ium) exhibits good biocompatibility, photostability, and bright emission in the near-infrared range (600-800 nm). The fluorescence emission intensity is responsive to viscosity, with significant fluorescence enhancement (48 times) and high fluorescence quantum yield (4.45 %) at high viscosity. Moreover, SIN has particular lysosome targeting properties with a Pearson correlation coefficient (PCC) of 0.97 and has good 1O2 generation ability under white light irradiation, especially in a weak acidic environment. Thus, SIN can realize good bioimaging ability and photodynamic therapeutic efficacy under the highly viscous and weakly acidic environment of lysosomes in the tumor cells. This study indicates that SIN has potential as a multifunctional organic photosensitizer for bioimaging and PDT of tumor.
Collapse
|
32
|
Das U, Shanavas S, Nagendra AH, Kar B, Roy N, Vardhan S, Sahoo SK, Panda D, Bose B, Paira P. Luminescent 11-{Naphthalen-1-yl}dipyrido[3,2-a:2',3'-c]phenazine-Based Ru(II)/Ir(III)/Re(I) Complexes for HCT-116 Colorectal Cancer Stem Cell Therapy. ACS APPLIED BIO MATERIALS 2023; 6:410-424. [PMID: 36638050 DOI: 10.1021/acsabm.2c00556] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Due to a number of unpleasant considerations, marketed drugs have steadily lost their importance in the treatment of cancer. In order to find a viable cancer cell diagnostic agent, we therefore focused on metal complexes that displayed target adequacy, permeability to cancer cells, high standard water solubility, cytoselectivity, and luminescent behavior. In this aspect, luminescent 11-{naphthalen-1-yl} dipyrido [3,2-a:2',3'-c] phenazine based Ru(II)/Ir(III)/Re(I) complexes have been prepared for HCT-116 colorectal cancer stem cell therapy. Our study successfully established the possible cytotoxicity of IrL complex at different doses on HCT-116 colorectal cancer stem cells (CRCSCs). Additionally, an immunochemistry analysis of the complex IrL showed that the molecule was subcellularly localized in the nucleus and other regions of the cytoplasm, where it caused nuclear DNA damage and mitochondrial dysfunction. The level of BAX and Bcl-2 was further quantified by qRT-PCR. The expression of proapoptotic BAX showed increased expression in the complex IrL-treated cell compared to the control, indicating the potential of complex IrL for apoptotic induction. Upon further validation, complex IrL was developed as an inhibitor of autophagy for the eradication of cancer stem cells.
Collapse
Affiliation(s)
- Utpal Das
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - Shanooja Shanavas
- Department Stem Cells and Regenerative Medicine Centre, Institution Yenepoya Research Centre, Yenepoya University, University Road, Derlakatte, Mangalore 575018, Karnataka, India
| | - Apoorva H Nagendra
- Department Stem Cells and Regenerative Medicine Centre, Institution Yenepoya Research Centre, Yenepoya University, University Road, Derlakatte, Mangalore 575018, Karnataka, India
| | - Binoy Kar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - Nilmadhab Roy
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - Seshu Vardhan
- Department of Applied Chemistry, S. V. National Institute of Technology (SVNIT) Ichchanath, Surat 395007, Gujrat, India
| | - Suban K Sahoo
- Department of Applied Chemistry, S. V. National Institute of Technology (SVNIT) Ichchanath, Surat 395007, Gujrat, India
| | - Debashis Panda
- Department of Chemistry, Rajiv Gandhi Institute of Petroleum Technology (Institute of National Importance (GOI Act. 54/2007), Jais 229304, Uttar Pradesh, India
| | - Bipasha Bose
- Department Stem Cells and Regenerative Medicine Centre, Institution Yenepoya Research Centre, Yenepoya University, University Road, Derlakatte, Mangalore 575018, Karnataka, India
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| |
Collapse
|
33
|
Zhong M, He J, Zhang B, Liu Q, Fang J. Mitochondria-targeted iridium-based photosensitizers enhancing photodynamic therapy effect by disturbing cellular redox balance. Free Radic Biol Med 2023; 195:121-131. [PMID: 36581057 DOI: 10.1016/j.freeradbiomed.2022.12.091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 12/27/2022]
Abstract
Photodynamic therapy (PDT) is a non-invasive, light-activated treatment approach that has been broadly employed in cancer. Cyclometallic iridium (Ш) complexes are candidates for ideal photosensitizers due to their unique photophysical and photochemical features, such as high quantum yield, large Stokes shift, strong resistance to photobleaching, and high cellular permeability. We evaluated a panel of iridium complexes and identified PC9 as a powerful photosensitizer to kill cancer cells. PC9 shows an 8-fold increase of cytotoxicity to HeLa cells under light irradiation. Further investigation discloses that PC9 has a strong mitochondrial-targeting ability and can inhibit the antioxidant enzyme thioredoxin reductase, which contributes to improving PDT efficacy. Our data indicate that iridium complexes are efficient photosensitizers with distinct physicochemical properties and cellular actions, and deserve further development as promising agents for PDT.
Collapse
Affiliation(s)
- Miao Zhong
- The State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jian He
- The State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; Sichuan Key Laboratory of Medical Imaging, School of Medical Imaging, North Sichuan Medical College, Nanchong, 637000, China
| | - Baoxin Zhang
- The State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Qiang Liu
- The State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China.
| | - Jianguo Fang
- The State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu, 210094, China.
| |
Collapse
|
34
|
Olelewe C, Awuah SG. Mitochondria as a target of third row transition metal-based anticancer complexes. Curr Opin Chem Biol 2023; 72:102235. [PMID: 36516614 PMCID: PMC9870944 DOI: 10.1016/j.cbpa.2022.102235] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
In pursuit of better treatment options for malignant tumors, metal-based complexes continue to show promise as attractive chemotherapeutics due to tunability, novel mechanisms, and potency exemplified by platinum agents. The metabolic character of tumors renders the mitochondria and other metabolism pathways fruitful targets for medicinal inorganic chemistry. Cumulative understanding of the role of mitochondria in tumorigenesis has ignited research in mitochondrial targeting metal-based complexes to overcome resistance and inhibit tumor growth with high potency and selectivity. Here, we discuss recent progress made in third row transition metal-based mitochondrial targeting agents with the goal of stimulating an active field of research toward new clinical anticancer agents and the elucidation of novel mechanisms of action.
Collapse
Affiliation(s)
- Chibuzor Olelewe
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, 40506, USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, 40506, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, United States; University of Kentucky Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, United States.
| |
Collapse
|
35
|
Pan ZY, Liang BF, Zhi YS, Yao DH, Li CY, Wu HQ, He L. Near-infrared AIE-active phosphorescent iridium(III) complex for mitochondria-targeted photodynamic therapy. Dalton Trans 2023; 52:1291-1300. [PMID: 36625001 DOI: 10.1039/d2dt03861g] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mitochondria-targeted photodynamic therapy (PDT) has recently been recognized as a promising strategy for effective cancer treatment. In this work, a mitochondria-targeted near-infrared (NIR) aggregation-induced emission (AIE)-active phosphorescent Ir(III) complex (Ir1) is reported with highly favourable mitochondria-targeted bioimaging and cancer PDT properties. Complex Ir1 has strong absorption in the visible light region (∼500 nm) and can effectively produce singlet oxygen (1O2) under green light (525 nm) irradiation. It preferentially accumulates in the mitochondria of human breast cancer MDA-MB-231 cells as revealed by colocalization analysis. Complex Ir1 displays high phototoxicity toward human breast cancer MDA-MB-231 cells and mouse breast cancer 4T1 cells. Complex Ir1 induces reactive oxygen species (ROS) production, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress in MDA-MB-231 cells upon photoirradiation, leading to apoptotic cell death. The favorable PDT performance of Ir1in vivo has been further demonstrated in tumour-bearing mice. Together, the results suggest that Ir1 is a promising photosensitizer for mitochondria-targeted imaging and cancer phototherapy.
Collapse
Affiliation(s)
- Zheng-Yin Pan
- College of Applied Sciences, Shenzhen University, Shenzhen 518060, China.,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China.,College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Bin-Fa Liang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China.
| | - Yun-Shi Zhi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China.
| | - Da-Hong Yao
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Chen-Yang Li
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China.
| | - Hai-Qiang Wu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China.
| | - Liang He
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
36
|
Wang X, Zhang C, Madji R, Voros C, Mazères S, Bijani C, Deraeve C, Cuvillier O, Gornitzka H, Maddelein ML, Hemmert C. N-Heterocyclic Carbene-Iridium Complexes as Photosensitizers for In Vitro Photodynamic Therapy to Trigger Non-Apoptotic Cell Death in Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020691. [PMID: 36677751 PMCID: PMC9861386 DOI: 10.3390/molecules28020691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/13/2023]
Abstract
A series of seven novel iridium complexes were synthetized and characterized as potential photosensitizers for photodynamic therapy (PDT) applications. Among them, four complexes were evaluated in vitro for their anti-proliferative activity with and without irradiation on a panel of five cancer cell lines, namely PC-3 (prostate cancer), T24 (bladder cancer), MCF7 (breast cancer), A549 (lung cancer) and HeLa (cervix cancer), and two non-cancerous cell models (NIH-3T3 fibroblasts and MC3T3 osteoblasts). After irradiation at 458 nm, all tested complexes showed a strong selectivity against cancer cells, with a selectivity index (SI) ranging from 8 to 34 compared with non-cancerous cells. The cytotoxic effect of all these complexes was found to be independent of the anti-apoptotic protein Bcl-xL. The compound exhibiting the best selectivity, complex 4a, was selected for further investigations. Complex 4a was mainly localized in the mitochondria. We found that the loss of cell viability and the decrease in ATP and GSH content induced by complex 4a were independent of both Bcl-xL and caspase activation, leading to a non-apoptotic cell death. By counteracting the intrinsic or acquired resistance to apoptosis associated with cancer, complex 4a could be an interesting therapeutic alternative to be studied in preclinical models.
Collapse
Affiliation(s)
- Xing Wang
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Chen Zhang
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Ryma Madji
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Camille Voros
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Christian Bijani
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Céline Deraeve
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Olivier Cuvillier
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| | - Heinz Gornitzka
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| | - Marie-Lise Maddelein
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| | - Catherine Hemmert
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| |
Collapse
|
37
|
Chao X, Yao D, Chen C. Desirable photo-physicochemical properties of dicyanomethylene-4H-pyran probe enables long-term fluorescent tracking of lysosomal dynamics in live cells. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2023.114571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
38
|
Recent advances on organelle specific Ru(II)/Ir(III)/Re(I) based complexes for photodynamic therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Ling YY, Wang WJ, Hao L, Wu XW, Liang JH, Zhang H, Mao ZW, Tan CP. Self-Amplifying Iridium(III) Photosensitizer for Ferroptosis-Mediated Immunotherapy Against Transferrin Receptor-Overexpressing Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203659. [PMID: 36310137 DOI: 10.1002/smll.202203659] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Photoimmunotherapy is attractive for cancer treatment due to its spatial controllability and sustained responses. This work presents a ferrocene-containing Ir(III) photosensitizer (IrFc1) that can bind with transferrin and be transported into triple-negative breast cancer (TNBC) cells via a transferrin receptor-mediated pathway. When the ferrocene in IrFc1 is oxidized by reactive oxygen species, its capability to photosensitize both type I (electron transfer) and type II (energy transfer) pathways is activated through a self-amplifying process. Upon irradiation, IrFc1 induces the generation of lipid oxidation to cause ferroptosis in TNBC cells, which promotes immunogenic cell death (ICD) under both normoxia and hypoxia. In vivo, IrFc1 treatment elicits a CD8+ T-cell response, which activates ICD in TNBC resulting in enhanced anticancer immunity. In summary, this work reports a small molecule-based photosensitizer with enhanced cancer immunotherapeutic properties by eliciting ferroptosis through a self-amplifying process.
Collapse
Affiliation(s)
- Yu-Yi Ling
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Wen-Jin Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Liang Hao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Xiao-Wen Wu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Jing-Hao Liang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Hang Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| |
Collapse
|
40
|
Liu J, Chen W, Zheng C, Hu F, Zhai J, Bai Q, Sun N, Qian G, Zhang Y, Dong K, Lu T. Recent molecular design strategies for efficient photodynamic therapy and its synergistic therapy based on AIE photosensitizers. Eur J Med Chem 2022; 244:114843. [DOI: 10.1016/j.ejmech.2022.114843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/14/2022] [Accepted: 10/08/2022] [Indexed: 11/04/2022]
|
41
|
Yang J, Griffin A, Qiang Z, Ren J. Organelle-targeted therapies: a comprehensive review on system design for enabling precision oncology. Signal Transduct Target Ther 2022; 7:379. [PMID: 36402753 PMCID: PMC9675787 DOI: 10.1038/s41392-022-01243-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer is a major threat to human health. Among various treatment methods, precision therapy has received significant attention since the inception, due to its ability to efficiently inhibit tumor growth, while curtailing common shortcomings from conventional cancer treatment, leading towards enhanced survival rates. Particularly, organelle-targeted strategies enable precise accumulation of therapeutic agents in organelles, locally triggering organelle-mediated cell death signals which can greatly reduce the therapeutic threshold dosage and minimize side-effects. In this review, we comprehensively discuss history and recent advances in targeted therapies on organelles, specifically including nucleus, mitochondria, lysosomes and endoplasmic reticulum, while focusing on organelle structures, organelle-mediated cell death signal pathways, and design guidelines of organelle-targeted nanomedicines based on intervention mechanisms. Furthermore, a perspective on future research and clinical opportunities and potential challenges in precision oncology is presented. Through demonstrating recent developments in organelle-targeted therapies, we believe this article can further stimulate broader interests in multidisciplinary research and technology development for enabling advanced organelle-targeted nanomedicines and their corresponding clinic translations.
Collapse
Affiliation(s)
- Jingjing Yang
- grid.24516.340000000123704535Institute of Nano and Biopolymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China
| | - Anthony Griffin
- grid.267193.80000 0001 2295 628XSchool of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406 USA
| | - Zhe Qiang
- grid.267193.80000 0001 2295 628XSchool of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406 USA
| | - Jie Ren
- grid.24516.340000000123704535Institute of Nano and Biopolymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China
| |
Collapse
|
42
|
Lu Y, Zhu D, Le Q, Wang Y, Wang W. Ruthenium-based antitumor drugs and delivery systems from monotherapy to combination therapy. NANOSCALE 2022; 14:16339-16375. [PMID: 36341705 DOI: 10.1039/d2nr02994d] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Ruthenium complex is an important compound group for antitumor drug research and development. NAMI-A, KP1019, TLD1433 and other ruthenium complexes have entered clinical research. In recent years, the research on ruthenium antitumor drugs has not been limited to single chemotherapy drugs; other applications of ruthenium complexes have emerged such as in combination therapy. During the development of ruthenium complexes, drug delivery forms of ruthenium antitumor drugs have also evolved from single-molecule drugs to nanodrug delivery systems. The review summarizes the following aspects: (1) ruthenium complexes from monotherapy to combination therapy, including the development of single-molecule compounds, carrier nanomedicine, and self-assembly of carrier-free nanomedicine; (2) ruthenium complexes in the process of ADME in terms of absorption, distribution, metabolism and excretion; (3) the applications of ruthenium complexes in combination therapy, including photodynamic therapy (PDT), photothermal therapy (PTT), photoactivated chemotherapy (PACT), immunotherapy, and their combined application; (4) the future prospects of ruthenium-based antitumor drugs.
Collapse
Affiliation(s)
- Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
- Department of Chemistry, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway
| | - Di Zhu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
| | - Quynh Le
- Center for Pharmacy, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway.
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
| | - Wei Wang
- Center for Pharmacy, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway.
- Department of Chemistry, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway
| |
Collapse
|
43
|
Joksimović N, Petronijević J, Radisavljević S, Petrović B, Mihajlović K, Janković N, Milović E, Milivojević D, Ilić B, Djurić A. Synthesis, characterization, antitumor potential, and investigation of mechanism of action of copper(ii) complexes with acylpyruvates as ligands: interactions with biomolecules and kinetic study. RSC Adv 2022; 12:30501-30513. [PMID: 36337968 PMCID: PMC9597287 DOI: 10.1039/d2ra05797b] [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: 09/14/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Considering the urgency of finding a cure for vicious diseases such as tumors, we have synthesized and characterized a small series of new copper(ii) complexes with biologically important ligands such as acylpyruvate. In addition to this, we used another four copper(ii) complexes, with ligands of the same type to examine the antitumor potential. The antitumor potential of the copper(ii) complexes was examined on three tumor cell lines and one normal human cell line using the MTT assay. All seven tested complexes showed very good cytotoxic effects. Two copper complexes that showed the best antitumor potential were selected for further testing that showed the best potential for potential application in the future. The mechanism of activity of these complexes was examined in detail using tests such as cell cycle, ROS level, oxidative DNA damage, and proteins related to hypoxia analysis. In addition, we examined the binding abilities of these complexes with biomolecules (Guo, Ino, 5'-GMP, BSA, and DNA). The results showed that the tested compounds bind strongly to DNA molecules through intercalation. Also, it has been shown that the tested compounds adequately bind to the BSA molecule, which indicates an even greater potential for some future application of these compounds in clinical practice.
Collapse
Affiliation(s)
- Nenad Joksimović
- University of Kragujevac, Faculty of Science, Department of Chemistry Radoja Domanovića 12 34000 Kragujevac Serbia
| | - Jelena Petronijević
- University of Kragujevac, Faculty of Science, Department of Chemistry Radoja Domanovića 12 34000 Kragujevac Serbia
| | - Snežana Radisavljević
- University of Kragujevac, Faculty of Science, Department of Chemistry Radoja Domanovića 12 34000 Kragujevac Serbia
| | - Biljana Petrović
- University of Kragujevac, Faculty of Science, Department of Chemistry Radoja Domanovića 12 34000 Kragujevac Serbia
| | - Kristina Mihajlović
- University of Kragujevac, Faculty of Science, Department of Chemistry Radoja Domanovića 12 34000 Kragujevac Serbia
| | - Nenad Janković
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Sciences Jovana Cvijića bb 34000 Kragujevac Serbia
| | - Emilija Milović
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Sciences Jovana Cvijića bb 34000 Kragujevac Serbia
| | - Dušan Milivojević
- Vinča Institute of Nuclear Science University of Belgrade P.O. Box 522 11001 Belgrade Serbia
| | - Bojana Ilić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia Belgrade Serbia
| | - Ana Djurić
- Institute of Oncology and Radiology of Serbia Pasterova 14 11000 Belgrade Serbia
| |
Collapse
|
44
|
Zhang Q, Shao J, Wang J, Gong XJ, Liu WX, Wang S, Zhang Y, Yang S, Zhang QS, Wei JX, Tian JL. Antitumor effects of new glycoconjugated Pt II agents dual-targeting GLUT1 and Pgp proteins. Dalton Trans 2022; 51:16082-16092. [PMID: 36178270 DOI: 10.1039/d2dt02455a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel and highly efficient dual-targeting PtII system was designed to improve the drug delivery capacity and selectivity in cancer treatment. The dual-targeting monofunctional PtII complexes (1-8) having glycosylated pendants as tridentated ligand were achieved by introducing glycosylation modification in the thioaminocarbazone compounds with potential lysosomal targeting ability. The structures and stability of 1-8 were further established by various techniques. Molecular docking studies showed that 2 was efficiently docked into glucose transporters protein 1 (GLUT1) and P-glycoprotein (Pgp) proteins with the optimal CDocker-interaction-energy of -64.84 and -48.85 kcal mol-1. Complex 2 with higher protein binding capacity demonstrated significant and broad-spectrum antitumor efficacy in vitro, even exhibiting a half maximal inhibitory concentration (IC50) value (∼10 μM) than cisplatin (∼17 μM) against human lung adenocarcinoma cells (A549). The inhibitor experiment revealed GLUT-mediated uptake of 2, and the subcellular localization experiment in A549 also proved that 2 could be localized in the lysosome, thereby causing cell apoptosis. Moreover, cellular thermal shift assay (CETSA) confirmed the binding of 2 with the target proteins of GLUT1 and Pgp. The above results indicated that 2 represents a potential anticancer candidate with dual-targeting functions.
Collapse
Affiliation(s)
- Qiang Zhang
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Jia Shao
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, PR China. .,National Health Commission's Key Laboratory of Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Jin Wang
- Outpatient Office, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Xian-Jin Gong
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Wei-Xing Liu
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Shan Wang
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, PR China.
| | - Yi Zhang
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, PR China. .,National Health Commission's Key Laboratory of Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Shuang Yang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, Tianjin 300071, PR China
| | - Quan-Sheng Zhang
- Tianjin Key Laboratory of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Jin-Xia Wei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Jin-Lei Tian
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
45
|
Redrado M, Miñana M, Coogan MP, Concepción Gimeno M, Fernández‐Moreira V. Tunable Emissive Ir(III) Benzimidazole-quinoline Hybrids as Promising Theranostic Lead Compounds. ChemMedChem 2022; 17:e202200244. [PMID: 35767349 PMCID: PMC9796238 DOI: 10.1002/cmdc.202200244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/28/2022] [Indexed: 01/01/2023]
Abstract
Bioactive and luminescent cyclometallated Ir(III) complexes [Ir(ppy)2 L1]Cl (1) and [Ir(ppy)2 L2]Cl (2) containing a benzimidazole derivative (L1/L2) as auxiliary mimic of a nucleotide have been synthesised. The emissive properties of both complexes are conditioned by the nature of L1 and L2, rendering an orange and a green emitter respectively. Both are highly emissive with quantum yield increasing in absence of oxygen up to 0.26 (1) and 0.36 (2), suggesting their phosphorescent character. Antiproliferative activity against lung cancer A549 cells increased up to 15 times upon irradiation conditions, reaching IC50 values in the nanomolar range (0.3±0.09 μM (1) and 0.26±0.14 μM (2)) and pointing them as good PSs candidates for photodynamic therapy via 1 O2 generation. Cellular biodistribution analysis by fluorescence microscopy suggest the lysosomes as the preferential accumulation organelle. Time-resolved studies showed a greatly increased cellular emission lifetime compared to the solution values, indicating binding to macromolecules or cellular structures and restriction of collision and vibrational quenching.
Collapse
Affiliation(s)
- Marta Redrado
- Departamento de Química InorgánicaInstituto de Síntesis Química y Catálisis Homogénea (ISQCH)CSIC-Universidad de ZaragozaPedro Cerbuna 1250009ZaragozaSpain
| | - Miriam Miñana
- Departamento de Química InorgánicaInstituto de Síntesis Química y Catálisis Homogénea (ISQCH)CSIC-Universidad de ZaragozaPedro Cerbuna 1250009ZaragozaSpain
| | | | - M. Concepción Gimeno
- Departamento de Química InorgánicaInstituto de Síntesis Química y Catálisis Homogénea (ISQCH)CSIC-Universidad de ZaragozaPedro Cerbuna 1250009ZaragozaSpain
| | - Vanesa Fernández‐Moreira
- Departamento de Química InorgánicaInstituto de Síntesis Química y Catálisis Homogénea (ISQCH)CSIC-Universidad de ZaragozaPedro Cerbuna 1250009ZaragozaSpain
| |
Collapse
|
46
|
Lu JJ, Ma XR, Xie K, Chen MR, Huang B, Li RT, Ye RR. Lysosome-targeted cyclometalated iridium(III) complexes: JMJD inhibition, dual induction of apoptosis and autophagy. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6694002. [PMID: 36073756 DOI: 10.1093/mtomcs/mfac068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/28/2022] [Indexed: 11/14/2022]
Abstract
A series of cyclometalated iridium(III) complexes with the formula [Ir(C^N)2 L](PF6) (C^N = 2-phenylpyridine (ppy, in Ir-1), 2-(2-thienyl)pyridine (thpy, in Ir-2), 2-(2,4-difluorophenyl)pyridine (dfppy, in Ir-3), L = 2-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)quinolin-8-ol) were designed and synthesized, which utilize 8-hydroxyquinoline derivative as N^N ligands to chelate the cofactor Fe2+ of the Jumonji domain-containing protein (JMJD) histone demethylase. As expected, the results of UV/Vis titration analysis confirm the chelating capabilities of Ir-1-3 for Fe2+, and molecular docking studies also show that Ir-1-3 can interact with the active pocket of JMJD protein, and treatment of cells with Ir-1-3 results in significant upregulation of trimethylated histone 3 lysine 9 (H3K9Me3), indicating the inhibition of JMJD activity. Meanwhile, Ir-1-3 exhibit much higher cytotoxicity against the tested tumor cell lines compared with the clinical chemotherapeutic agent cisplatin. And Ir-1-3 can block the cell cycle at G2/M phase and inhibit cell migration and colony formation. Further studies show that Ir-1-3 can specifically accumulate in lysosomes, damage the integrity of lysosomes, and induce apoptosis and autophagy. Reduction of mitochondrial membrane potential (MMP) and elevation of reactive oxygen species (ROS) also contribute to the antitumor effects of Ir-1-3. Finally, Ir-1 can inhibit tumor growth effectively in vivo and increase the expression of H3K9Me3 in tumor tissues. Our study demonstrates that these iridium(III) complexes are promising anticancer agents with multiple functions, including the inhibition of JMJD and induction of apoptosis and autophagy.
Collapse
Affiliation(s)
- Jun-Jian Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Xiu-Rong Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Kai Xie
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Mei-Ru Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Bo Huang
- Faculty of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming 650500, P. R. China
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| |
Collapse
|
47
|
Lee LCC, Lo KKW. Luminescent and Photofunctional Transition Metal Complexes: From Molecular Design to Diagnostic and Therapeutic Applications. J Am Chem Soc 2022; 144:14420-14440. [PMID: 35925792 DOI: 10.1021/jacs.2c03437] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been emerging interest in the exploitation of the photophysical and photochemical properties of transition metal complexes for diagnostic and therapeutic applications. In this Perspective, we highlight the major recent advances in the development of luminescent and photofunctional transition metal complexes, in particular, those of rhenium(I), ruthenium(II), osmium(II), iridium(III), and platinum(II), as bioimaging reagents and phototherapeutic agents, with a focus on the molecular design strategies that harness and modulate the interesting photophysical and photochemical behavior of the complexes. We also discuss the current challenges and future outlook of transition metal complexes for both fundamental research and clinical applications.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R. China.,Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P.R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R. China.,State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R. China
| |
Collapse
|
48
|
Cyclometalated iridium(III) dithioformic acid complexes as mitochondria-targeted imaging and anticancer agents. J Inorg Biochem 2022; 233:111855. [DOI: 10.1016/j.jinorgbio.2022.111855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022]
|
49
|
Novel 4-/5-bromo-8-hydroxyquinoline cyclometalated iridium(III) complexes as highly potent anticancer and bioimaging agents. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.109609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Liu L, Chen J, Wang MM, Huang Y, Qian Y, Xue X, Su Z, Liu HK. The cyclometalated iridium (III) complex based on 9-Anthracenecarboxylic acid as a lysosomal-targeted anticancer agent. J Inorg Biochem 2022; 235:111913. [PMID: 35905643 DOI: 10.1016/j.jinorgbio.2022.111913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/18/2022] [Accepted: 07/03/2022] [Indexed: 12/26/2022]
Abstract
9-Anthracenecarboxylic acid (9-Ac) was reported early as a chloride channel inhibitor and was found to exhibit significant anti-proliferative activity on leukemic cells, but has not been researched in solid tumor cells. Herein, a 9-anthraceneic acid derivative was introduced into the cyclometalated Iridium (III) species to construct a novel Iridium (Ir) complex Ir-9-Ac, [Ir(ppy)2(9-Ac-L)]PF6 (ppy = 2-phenylpyridine, 9-Ac-L = N-((4'-methyl-[2,2'-bipyridin]-4-yl)methyl)anthracene-9-carboxamide), which could accumulated in lysosomes. Ir-9-Ac showed good cytotoxic activity against several tumor cell lines, notably on A549 cells. Besides Ir-9-Ac could inhibit the cell colony formation and growth of the 3D cell spheroids, demonstrating the potential to suppress tumors in vivo. This design provided a platform for the design of cyclometalated Iridium (III) anticancer complexes.
Collapse
Affiliation(s)
- Lu Liu
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jun Chen
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Meng-Meng Wang
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yuanlei Huang
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yong Qian
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xuling Xue
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Zhi Su
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Hong-Ke Liu
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|