1
|
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
|
2
|
Walter ERH, Leung PKK, Lee LCC, Lo KKW, Long NJ. Potent BODIPY-based photosensitisers for selective mitochondrial dysfunction and effective photodynamic therapy. J Mater Chem B 2024; 12:10409-10415. [PMID: 39297339 DOI: 10.1039/d4tb01609b] [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: 10/18/2024]
Abstract
The development of new and improved mitochondria-targeting photosensitisers (PSs) for photodynamic therapy (PDT) remains highly desirable, due to the critical role the mitochondria play in maintaining healthy cellular function. Here, we report the design, synthesis, photophysical properties and biological characterisation of a series of di-iodinated BODIPY-based PSs, BODIPY-Mito-I-n, for mitochondria-targeted PDT applications. Six BODIPY-Mito-I-n analogues were synthesised in good yields, with fast reaction times of between 30 and 60 min under mild conditions. The di-iodination of the BODIPY scaffold enabled highly efficient population of the triplet state, leading to high singlet oxygen (1O2) photosensitisation efficiencies (ΦΔ = 0.55-0.65). All BODIPY-Mito-I-n compounds exhibited very high photocytotoxic activity towards HeLa cells, with IC50,light values of between 1.30 and 6.93 nM, due to photoinduced 1O2 generation. Notably, the poly(ethylene glycol) (PEG)-modified BODIPY-Mito-I-6 showed remarkably lower dark cytotoxicity (IC50,dark = 6.68-7.25 μM) than the non-PEGylated analogues BODIPY-Mito-I-1 to BODIPY-Mito-I-5 (IC50,dark = 0.58-1.09 μM), resulting in photocytotoxicity indices up to 2120. Mechanistic studies revealed that BODIPY-Mito-I-6 induced reactive oxygen species overproduction and mitochondrial dysfunction in cells upon irradiation, leading to significant cell death through a combination of apoptosis and necrosis. It is anticipated that our design will contribute to the development of more effective mitochondria-targeting PSs for cancer therapy.
Collapse
Affiliation(s)
- Edward R H Walter
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, W12 0BZ, UK.
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17 W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Peter Kam-Keung Leung
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China.
- State Key Laboratory of Terahertz and Millimetre Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Lawrence Cho-Cheung Lee
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17 W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, 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 Millimetre Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Nicholas J Long
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, W12 0BZ, UK.
| |
Collapse
|
3
|
Zhou L, Li J, Chen J, Yao X, Zeng X, Liu Y, Wang Y, Wang X. Anticancer activity and mechanism studies of photoactivated iridium(III) complexes toward lung cancer A549 cells. Dalton Trans 2024; 53:15176-15189. [PMID: 39221457 DOI: 10.1039/d4dt01677g] [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: 09/04/2024]
Abstract
Cyclometalated iridium(III) compounds have been widely explored due to their outstanding photo-physical properties and multiple anticancer activities. In this paper, three cyclometalated iridium(III) compounds [Ir(ppy)2(DBDIP)]PF6 (5a), [Ir(bzq)2(DBDIP)]PF6 (5b), and [Ir(piq)2(DBDIP)]PF6 (5c) (ppy: 2-phenylpyridine; bzq: benzo[h]quinoline; piq: 1-phenylisoquinoline, and DBDIP: 2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-1H-imidazo[4,5-f][1,10]phenanthroline) were synthesized and the mechanism of antitumor activity was investigated. Compounds photoactivated by visible light show strong cytotoxicity against tumor cells, especially toward A549 cells. Biological experiments such as migration, cellular localization, mitochondrial membrane potential and permeability, reactive oxygen species (ROS) and calcium ion level detection were performed, and they demonstrated that the compounds induced the apoptosis of A549 cells through a mitochondrial pathway. At the same time, oxidative stress caused by ROS production increases the release of damage-related molecules and the expression of porogen gasdermin D (GSDMD), and the content of LDH released from damaged cell membranes also increased. Besides, the content of the lipid peroxidation product, malondialdehyde (MDA), increased and the expression of GPX4 decreased. These indicate that the compounds promote cell death by combining ferroptosis and pyroptosis. The results reveal that cyclometalated iridium(III) compounds 5a-5c may be a potential chemotherapeutic agent for photodynamic therapy of cancers.
Collapse
Affiliation(s)
- Lin Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Jiongbang Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Ju Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xin Yao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiandong Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Yi Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiuzhen Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
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
|
5
|
Sanz-Villafruela J, Bermejo-Casadesús C, Martínez-Alonso M, Moro A, Lima JC, Massaguer A, Espino G. Towards efficient Ir(III) anticancer photodynamic therapy agents by extending π-conjugation on N^N ligands. Dalton Trans 2024; 53:11393-11409. [PMID: 38899369 DOI: 10.1039/d4dt00390j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
In this work we disclose a new family of biscyclometallated Ir(III) complexes of the general formula [Ir(C^N)2(N^N)]Cl (IrL1-IrL5), where HC^N is 1-phenyl-β-carboline and N^N ligands (L1-L5) are different diimine ligands that differ from each other in the number of aromatic rings fused to the bipyridine scaffold. The photophysical properties of IrL1-IrL5 were thoroughly studied, and theoretical calculations were performed for a deeper comprehension of the respective variations along the series. All complexes exhibited high photostability under blue light irradiation. An increase in the number of aromatic rings led to a reduction in the HOMO-LUMO band gap causing a red-shift in the absorbance bands. Although all the complexes generated singlet oxygen (1O2) in aerated aqueous solutions through a photocatalytic process, IrL5 was by far the most efficient photosensitizer. Consequently, IrL5 was highly active in the photocatalytic oxidation of NADH. The formation of aggregates in DMSO at a high concentration (25 mM) was confirmed using different techniques, but was proved to be negligible in the concentration range of biological experiments. Moreover, ICP-MS studies proved that the cellular uptake of IrL2 and IrL3 is much better relative to that of IrL1, IrL4 and IrL5. The antiproliferative activity of IrL1-IrL5 was investigated in the dark and under blue light irradiation against different cancer cell lines. Complexes IrL1-IrL4 were found to be cytotoxic under dark conditions, while IrL5 turned out to be weakly cytotoxic. Despite the low cellular uptake of IrL5, this derivative exhibited a high increase of cytotoxicity upon blue light irradiation resulting in photocytotoxicity indexes (PI) up to 38. IrL1-IrL4 showed lower photocytotoxicity indexes ranging from 1.3 to 17.0. Haemolytic experiments corroborated the compatibility of our complexes with red blood cells. Confocal microscopy studies proved their accumulation in mitochondria, leading to mitochondrial membrane depolarization, and ruled out their localization in lysosomes. Overall, the mitochondria-targeted activity of IrL5, which inhibits considerably the viability of cancer cells upon blue light irradiation, allows us to outline this PS as a new alternative to traditional chemotherapeutic agents.
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-Casadesús
- 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.
| | - Artur Moro
- Universidade NOVA de Lisboa, LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, 2829-516 Caparica, Portugal
| | - João C Lima
- Universidade NOVA de Lisboa, LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, 2829-516 Caparica, Portugal
| | - Anna Massaguer
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003 Girona, Spain.
| | - Gustavo Espino
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, 09001, Burgos, Spain.
| |
Collapse
|
6
|
Chen L, Tang H, Chen W, Wang J, Zhang S, Gao J, Chen Y, Zhu X, Huang Z, Chen J. Mitochondria-targeted cyclometalated iridium (III) complexes: Dual induction of A549 cells apoptosis and autophagy. J Inorg Biochem 2023; 249:112397. [PMID: 37844533 DOI: 10.1016/j.jinorgbio.2023.112397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/01/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023]
Abstract
In this study, we synthesized 4 cyclometalated iridium complexes using N-(1,10-phenanthrolin-5-yl)picolinamide (PPA) as the main ligand, denoted as [Ir(ppy)2PPA]PF6 (ppy = 2-phenylpyridine, Ir1), [Ir(bzq)2PPA]PF6 (bzq = benzo[h]quinoline, Ir2), [Ir(dfppy)2PPA]PF6 (dfppy = 2-(3,5-difluorophenyl)pyridine, Ir3), and [Ir(thpy)2PPA]PF6 (thpy = 2-(thiophene-2-yl)pyridine, Ir4). Compared to cisplatin and oxaliplatin, all four complexes exhibited significant anti-tumor activity. Among them, Ir2 demonstrated higher cytotoxicity against A549 cells, with an IC50 value of 1.6 ± 0.2 μM. The experimental results indicated that Ir2 primarily localized in the mitochondria, inducing a large amount of reactive oxygen species (ROS) generation, that decreased in mitochondrial membrane potential (MMP), reduced ATP production, and further impaired mitochondrial function, leading to cytochrome c release. Additionally, Ir2 caused cell cycle arrest at the S phase and induced apoptosis through the AKT-mediated signaling pathway. Further investigations revealed that Ir2 could simultaneously induce both apoptosis and autophagy in A549 cells, with the latter acting as a non-protective mechanism that promoted cell death. More importantly, Ir2 exhibited low toxicity to both normal LO2 cells in vitro and zebrafish embryos in vivo. Consequently, these newly developed Ir(III) complexes show great potential in the development of novel and low-toxicity anticancer agents.
Collapse
Affiliation(s)
- Lanmei Chen
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Hong Tang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Weigang Chen
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China
| | - Jie Wang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China
| | - Shenting Zhang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Jie Gao
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China
| | - Yu Chen
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China
| | - Xufeng Zhu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China.
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China.
| | - Jincan Chen
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China.
| |
Collapse
|
7
|
Xu GX, Lee LCC, Leung PKK, Mak ECL, Shum J, Zhang KY, Zhao Q, Lo KKW. Bioorthogonal dissociative rhenium(i) photosensitisers for controlled immunogenic cell death induction. Chem Sci 2023; 14:13508-13517. [PMID: 38033895 PMCID: PMC10686031 DOI: 10.1039/d3sc04903e] [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: 09/16/2023] [Accepted: 10/21/2023] [Indexed: 12/02/2023] Open
Abstract
Photosensitisers for photoimmunotherapy with high spatiotemporal controllability are rare. In this work, we designed rhenium(i) polypyridine complexes modified with a tetrazine unit via a bioorthogonally activatable carbamate linker as bioorthogonally dissociative photosensitisers for the controlled induction of immunogenic cell death (ICD). The complexes displayed increased emission intensities and singlet oxygen (1O2) generation efficiencies upon reaction with trans-cyclooct-4-enol (TCO-OH) due to the separation of the quenching tetrazine unit from the rhenium(i) polypyridine core. One of the complexes containing a poly(ethylene glycol) (PEG) group exhibited negligible dark cytotoxicity but showed greatly enhanced (photo)cytotoxic activity towards TCO-OH-pretreated cells upon light irradiation. The reason is that TCO-OH allowed the synergistic release of the more cytotoxic rhenium(i) aminomethylpyridine complex and increased 1O2 generation. Importantly, the treatment induced a cascade of events, including lysosomal dysfunction, autophagy suppression and ICD. To the best of our knowledge, this is the very first example of using bioorthogonal dissociation reactions as a trigger to realise photoinduced ICD, opening up new avenues for the development of innovative photoimmunotherapeutic agents.
Collapse
Affiliation(s)
- Guang-Xi Xu
- Department of Chemistry, City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - 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 17 W, Hong Kong Science Park New Territories Hong Kong P. R. China
| | - Peter Kam-Keung Leung
- Department of Chemistry, City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong P. R. China
- State Key Laboratory of Terahertz and Millimetre Waves, City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Eunice Chiu-Lam Mak
- Department of Chemistry, City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Justin Shum
- Department of Chemistry, City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Kenneth Yin Zhang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications 9 Wenyuan Road Nanjing 210023 P. R. China
| | - Qiang Zhao
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications 9 Wenyuan Road Nanjing 210023 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 Millimetre Waves, City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong P. R. China
| |
Collapse
|
8
|
Das B, Gupta S, Mondal A, Kalita KJ, Mallick AI, Gupta P. Tuning the Organelle-Specific Imaging and Photodynamic Therapeutic Efficacy of Theranostic Mono- and Trinuclear Organometallic Iridium(III) Complexes. J Med Chem 2023; 66:15550-15563. [PMID: 37950696 DOI: 10.1021/acs.jmedchem.3c01875] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2023]
Abstract
The organelle-specific localization of mononuclear and trinuclear iridium(III) complexes and their photodynamic behavior within the cells are described herein, emphasizing their structure-activity relationship. Both the IrA2 and IrB2 complexes possess a pair of phenyl-benzothiazole derived from the -CHO moieties of mononuclear organometallic iridium(III) complexes IrA1 and IrB1, which chelates IrCp*Cl (Cp* = 1,2,3,4,5-pentamethylcyclopentadiene) to afford trinuclear complexes IrA3 and IrB3. Insights into the photophysical and electrochemical parameters of the complexes were obtained by a time-dependent density functional theory study. The synthesized complexes IrA2, IrA3, IrB2, and IrB3 were found to be nontoxic to human MCF7 breast carcinoma cells. However, the photoexcitation of complexes using LED light could effectively trigger intracellular reactive oxygen species (ROS) generation, leading to cell death. Furthermore, to check the organelle-specific localization of IrA2 and IrB2, we observed that both complexes could selectively localize in the endoplasmic reticulum. In contrast, trinuclear IrA3 and IrB3 accumulate in the nuclei. The photoexcitation of complexes using LED light could effectively trigger intracellular reactive oxygen species (ROS) generation, leading to cell death.
Collapse
Affiliation(s)
- Bishnu Das
- Department of Chemical Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Subhadeep Gupta
- Department of Biological Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Anushka Mondal
- Department of Biological Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Kalyan Jyoti Kalita
- Department of Chemical Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Parna Gupta
- Department of Chemical Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| |
Collapse
|
9
|
Borah ST, Das B, Biswas P, Mallick AI, Gupta P. Aqua-friendly organometallic Ir-Pt complexes: pH-responsive AIPE-guided imaging of bacterial cells. Dalton Trans 2023; 52:2282-2292. [PMID: 36723088 DOI: 10.1039/d2dt03390a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In this work, the aggregation-induced photoluminescence emission (AIPE) of three water-soluble heterobimetallic Ir-Pt complexes was reported with insight into their photophysical and electrochemical properties and imaging of bacterial cells. An alkyne appended Schiff's base L, bridges bis-cyclometalated iridium(III) and platinum(II) terpyridine centre. The Schiff's base (N-N fragment) serves as the ancillary ligand to the iridium(III) centre, while the alkynyl end is coordinated to platinum(II). The pH and ionic strength influence the aggregation kinetics of the alkynylplatinum(II) fragment, leading to metal-metal and π-π interactions with the emergence of a triplet metal-metal-to-ligand charge transfer (3MMLCT) emission. The excellent reversibility and photostability of aggregation-induced emission (AIE) of these aqua-friendly complexes were tested for their ability to sense and selectively image E. coli cells at various pH values.
Collapse
Affiliation(s)
- Sakira Tabassum Borah
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India.
| | - Bishnu Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India.
| | - Prakash Biswas
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Amirul I Mallick
- 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
|
10
|
Liu J, Wu Y, Yang G, Liu Z, Liu X. Mitochondrial targeting half-sandwich iridium(III) and ruthenium(II) dppf complexes and in vitro anticancer assay. J Inorg Biochem 2023; 239:112069. [PMID: 36423395 DOI: 10.1016/j.jinorgbio.2022.112069] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/10/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Considering the potential application of half-sandwich and ferrocenyl-containing organometallic complexes in the area of anticancer, four half-sandwich iridium(III) (IrIII) and ruthenium(II) (RuII) diphenylphosphino ferrocene (dppf) complexes were prepared in this study. Complexes showed favorable anti-proliferation activity towards A549 cell lines compared to cisplatin, meanwhile, which could effectively inhibit cell migration. These complexes followed an energy dependence uptake mechanism, effectively accumulated in mitochondria with a Pearson's Colocalization Coefficient (PCC) of 0.77, decreased the mitochondrial membrane potential, induced a surge of reactive oxygen species, disturbed cell cycle, and eventually led to apoptosis. Western blot assay further confirmed that these complexes induced apoptosis following a mitochondrial pathway. Above all, half-sandwich IrIII and RuII dppf complexes show the prospect of becoming a new multifunctional therapeutic platform for mitochondrial targeted imaging and anticancer drugs.
Collapse
Affiliation(s)
- Jinfeng Liu
- College of Life Sciences, Qufu Normal University, Qufu 273165, Shandong, China.
| | - Yuting Wu
- College of Life Sciences, Qufu Normal University, Qufu 273165, Shandong, China
| | - Ge Yang
- College of Life Sciences, Qufu Normal University, Qufu 273165, Shandong, China
| | - Zhe Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, China.
| | - Xicheng Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, China.
| |
Collapse
|
11
|
Qian Y, Wang J, Bu W, Zhu X, Zhang P, Zhu Y, Fan X, Wang C. Targeted implementation strategies of precise photodynamic therapy based on clinical and technical demands. Biomater Sci 2023; 11:704-718. [PMID: 36472233 DOI: 10.1039/d2bm01384c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
With the development of materials science, photodynamic-based treatments have gradually entered clinics. Photodynamic therapy is ideal for cancer treatment due to its non-invasive and spatiotemporal properties and is the first to be widely promoted in clinical practice. However, the shortcomings resulting from the gap between technical and clinical demands, such as phototoxicity, low tissue permeability, and tissue hypoxia, limit its wide applications. This article reviews the available data regarding the pharmacological and clinical factors affecting the efficacy of photodynamic therapy, such as photosensitizers and oxygen supply, disease diagnosis, and other aspects of photodynamic therapy. In addition, the synergistic treatment of photodynamic therapy with surgery and nanotechnology is also discussed, which is expected to provide inspiration for the design of photodynamic therapy strategies.
Collapse
Affiliation(s)
- Yun Qian
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Jialun Wang
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.
| | - Wenbo Bu
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Xiaoyan Zhu
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Ping Zhang
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Yun Zhu
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China. .,Department of Pharmacy, Nanjing Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.,Nanjing Medical Center for Clinical Pharmacy, Nanjing 210008, Jiangsu Province, China
| | - Xiaoli Fan
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China.
| |
Collapse
|
12
|
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]
|
13
|
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: 32] [Impact Index Per Article: 10.7] [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
|
14
|
Parshina YP, Kovylina TA, Konev AN, Belikov AA, Baber PO, Komarova AD, Romaeva EA, Bochkarev LN. Norbornene-Substituted Cationic Iridium(III) Complex and Water-Soluble Luminescent Polymers Based on It: Synthesis, Photophysical and Cytotoxic Properties. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222120167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
A norbornene-substituted cationic iridium(III) complex containing 1-phenylisoquinoline cyclometalating ligands and an additional phenylimidazophenanthroline ligand was synthesized. On the base of this complex, water-soluble polymers were obtained by ring-opening metathesis polymerization (ROMP). The resulting polymers showed oxygen-dependent phosphorescence in the orange spectral region and high cytotoxicity against HCT116 cancer cells.
Collapse
|
15
|
Mitochondria-targeted photosensitizer based nanoplatform loading glutathione inhibitor for enhanced breast cancer photodynamic therapy. Colloids Surf B Biointerfaces 2022. [DOI: 10.1016/j.colsurfb.2022.112956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
16
|
Estevão BM, Vilela RRC, Geremias IP, Zanoni KPS, de Camargo ASS, Zucolotto V. Mesoporous silica nanoparticles incorporated with Ir(III) complexes: From photophysics to photodynamic therapy. Photodiagnosis Photodyn Ther 2022; 40:103052. [PMID: 35934182 DOI: 10.1016/j.pdpdt.2022.103052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
Organically modified mesoporous silica nanoparticles (MSNs) containing Ir complexes (Ir1, Ir2 and Ir3) were successfully synthesized. These Ir-entrapped MCM41-COOH nanoparticles have shown relevant photophysical characteristics including high efficiency in the photoproduction and delivery of singlet oxygen (1O2), which is particularly promising for photodynamic therapy (PDT) applications. In vitro tests have evidenced that complex@MCM41-COOH are able to reduce cell proliferation after 10 min of blue-light irradiation in Hep-G2 liver cancer cells.
Collapse
Affiliation(s)
- Bianca M Estevão
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil.
| | - Raquel R C Vilela
- Laboratory of Spectroscopy of Functional Materials, São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil.
| | - Isabella P Geremias
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil
| | - Kassio P S Zanoni
- Laboratory of Spectroscopy of Functional Materials, São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil; Molecular Optoelectronic Devices, Instituto de Ciencia Molecular (ICMol), University of Valencia, Catedrático J. Beltrán 2, Paterna, Valencia 46980, Spain
| | - Andrea S S de Camargo
- Laboratory of Spectroscopy of Functional Materials, São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil
| |
Collapse
|
17
|
Naskar N, Liu W, Qi H, Stumper A, Fischer S, Diemant T, Behm RJ, Kaiser U, Rau S, Weil T, Chakrabortty S. A Carbon Nanodot Based Near-Infrared Photosensitizer with a Protein-Ruthenium Shell for Low-Power Photodynamic Applications. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48327-48340. [PMID: 36269223 DOI: 10.1021/acsami.2c08585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Near-infrared (NIR) light-activated photosensitization represents an encouraging therapeutic method in photodynamic therapy, especially for deep tissue penetration. In this context, two-photon activation, i.e., utilization of photons with relatively low energy but high photon flux for populating a virtual intermediate state leading to an excited state, is attractive. This concept would be highly advantageous in photodynamic therapy due to its minimal side effects. Herein, we propose that the combination of plasma protein serum albumin (HSA) containing several Ru complexes and NIR two-photon excitable carbon nanodots (Cdots), termed HSA-Ru-Cdots, provides several attractive features for enhancing singlet oxygen formation within the mitochondria of cancer cells stimulated by two-photon excitation in the NIR region. HSA-Ru-Cdot features biocompatibility, water solubility, and photostability as well as uptake into cancer cells with an endosomal release, which is an essential feature for subcellular targeting of mitochondria. The NIR two-photon excitation induced visible emission of the Cdots allows fluorescence resonance energy transfer (FRET) to excite the metal-to-ligand charge transfer of the Ru moiety, and fluorescence-lifetime imaging microscopy (FLIM) has been applied to demonstrate FRET within the cells. The NIR two-photon excitation is indirectly transferred to the Ru complexes, which leads to the production of singlet oxygen within the mitochondria of cancer cells. Consequently, we observe the destruction of filamentous mitochondrial structures into spheroid aggregates within various cancer cell lines. Cell death is induced by the long-wavelength NIR light irradiation at 810 nm with a low power density (7 mW/cm2), which could be attractive for phototherapy applications where deeper tissue penetration is crucial.
Collapse
Affiliation(s)
- Nilanjon Naskar
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Weina Liu
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Haoyuan Qi
- Central Facility for Materials Science Electron Microscopy, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
- Center for Advancing Electronics Dresden (cfaed) & Faculty of Chemistry and Food Chemistry, Dresden University of Technology, Mommsenstrasse 4, D-01062Dresden, Germany
| | - Anne Stumper
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Thomas Diemant
- Institute of Surface Chemistry and Catalysis, Ulm University, Albert-Einstein-Allee 47, D-89081Ulm, Germany
| | - R Jürgen Behm
- Institute of Surface Chemistry and Catalysis, Ulm University, Albert-Einstein-Allee 47, D-89081Ulm, Germany
| | - Ute Kaiser
- Central Facility for Materials Science Electron Microscopy, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Sven Rau
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Tanja Weil
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Sabyasachi Chakrabortty
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
- Department of Chemistry, SRM University AP Andhra Pradesh, Andhra Pradesh522502, India
| |
Collapse
|
18
|
Amarsy I, Papot S, Gasser G. Stimuli‐Responsive Metal Complexes for Biomedical Applications. Angew Chem Int Ed Engl 2022; 61:e202205900. [DOI: 10.1002/anie.202205900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Ivanna Amarsy
- Chimie ParisTech PSL University, CNRS Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France
| | - Sébastien Papot
- Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP) Université de Poitiers, CNRS Equipe Labellisée Ligue Contre le Cancer 4 rue Michel Brunet, TSA 51106 86073 Poitiers France
| | - Gilles Gasser
- Chimie ParisTech PSL University, CNRS Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France
| |
Collapse
|
19
|
Mitochondria-targeted cyclometalated iridium (III) complex for H 2S-responsive intracellular redox regulation as potent photo-oxidation anticancer agent. J Biol Inorg Chem 2022; 27:641-651. [PMID: 36058946 DOI: 10.1007/s00775-022-01957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
Abstract
Owing to the safety and low toxicity, photodynamic therapy (PDT) for cancer treatment has received extensive attention. However, the excess H2S in cancer cells reduces the PDT efficiency, because H2S indirectly depletes the reactive oxygen species (ROS). To improve anticancer efficiency, a mitochondria-targeted iridium(III) complex Ir-MMB has been developed as H2S consumer and photo-oxidation anticancer agent. On the one hand, complex Ir-MMB can consume H2S with sensitive phosphorescence turn-on, which has been successfully applied to exogenous and endogenous H2S response imaging in living cells. On the other hand, Ir-MMB can enhance its anticancer activity and cause photo-oxidation damage via catalyzing the oxidation of reduced form of nicotinamide-adenine dinucleotide (NADH) to NAD+ and producing H2O2 under light, and ultimately results in cell apoptosis through mitochondrial depolarization and ROS production.
Collapse
|
20
|
Lee CG, Lee C, Lee J, Nam JS, Kim B, Kwon T. Dual‐Modulated Release of a Cytotoxic Photosensitizer Using Photogenerated Reactive Oxygen Species and Glutathione. Angew Chem Int Ed Engl 2022; 61:e202210623. [DOI: 10.1002/anie.202210623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Chae Gyu Lee
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Chaiheon Lee
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Joonhee Lee
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Department of Chemistry Yonsei University Seoul 03722 Republic of Korea
| | - Jung Seung Nam
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Institute for Cancer Genetics Department of Genetics and Development Columbia University Irving Medical Center New York NY 10032 USA
- Herbert Irving Comprehensive Cancer Center Columbia University Irving Medical Center New York NY 10032 USA
| | - Byeong‐Su Kim
- Department of Chemistry Yonsei University Seoul 03722 Republic of Korea
| | - Tae‐Hyuk Kwon
- Department of Chemistry Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Center for Wave Energy Materials Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| |
Collapse
|
21
|
Lee CG, Lee C, Lee J, Nam JS, Kim BS, Kwon TH. Dual‐Modulated Release of a Cytotoxic Photosensitizer Using Photogenerated Reactive Oxygen Species and Glutathione. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202210623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Chae Gyu Lee
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Chaiheon Lee
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Joonhee Lee
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Jung Seung Nam
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| | - Byeong-Su Kim
- Yonsei University Department of Chemistry KOREA, REPUBLIC OF
| | - Tae-Hyuk Kwon
- Ulsan National Institute of Science and Technology Department of Chemistry KOREA, REPUBLIC OF
| |
Collapse
|
22
|
Abuduwaili W, Wang X, Huang AT, Sun JL, Xu RC, Zhang GC, Liu ZY, Wang F, Zhu CF, Liu TT, Dong L, Zhu JM, Weng SQ, Li Y, Shen XZ. Iridium Complex-Loaded Sorafenib Nanocomposites for Synergistic Chemo-photodynamic Therapy of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37356-37368. [PMID: 35951459 DOI: 10.1021/acsami.2c07247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Although sorafenib, a multi-kinase inhibitor, has provided noteworthy benefits in patients with hepatocellular carcinoma (HCC), the inevitable side effects, narrow therapeutic window, and low bioavailability seriously affect its clinical application. To be clinically distinctive, innovative drugs must meet the needs of reaching tumor tissues and cause limited side effects to normal organs and tissues. Recently, photodynamic therapy, utilizing a combination of a photosensitizer and light irradiation, was selectively accumulated at the tumor site and taken up effectively via inducing apoptosis or necrosis of cancer cells. In this study, a nano-chemo-phototherapy drug was fabricated to compose an iridium-based photosensitizer combined with sorafenib (IPS) via a self-assembly process. Compared to the free iridium photosensitizer or sorafenib, the IPS exhibited significantly improved therapeutic efficacy against tumor cells because of the increased cellular uptake and the subsequent simultaneous release of sorafenib and generation of reactive oxygen species production upon 532 nm laser irradiation. To evaluate the effect of synergistic treatment, cytotoxicity detection, live/dead staining, cell proliferative and apoptotic assay, and Western blot were performed. The IPS exhibited sufficient biocompatibility by hemolysis and serum biochemical tests. Also, the results suggested that IPS significantly inhibited HCC cell proliferation and promoted cell apoptosis. More importantly, marked anti-tumor growth effects via inhibiting cell proliferation and promoting tumor cell death were observed in an orthotopic xenograft HCC model. Therefore, our newly proposed nanotheranostic agent for combined chemotherapeutic and photodynamic therapy notably improves the therapeutic effect of sorafenib and has the potential to be a new alternative option for HCC treatment.
Collapse
Affiliation(s)
- Weinire Abuduwaili
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Xiang Wang
- Institute of Bismuth Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, 516 Jungong Rd., Shanghai 200093, China
| | - An-Tian Huang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Jia-Lei Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Ru-Chen Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Guang-Cong Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Zhi-Yong Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Fu Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Chang-Feng Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Tao-Tao Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Ji-Min Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Shu-Qiang Weng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
| | - Yuhao Li
- Institute of Bismuth Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, 516 Jungong Rd., Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, 516 Jungong Rd., Shanghai 200093, China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
- Shanghai Institute of Liver Disease, 180 Fenglin Rd., Shanghai 200032, China
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College of Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China
| |
Collapse
|
23
|
Phosphorescent Ir(III) Complexes for Biolabeling and Biosensing. Top Curr Chem (Cham) 2022; 380:35. [PMID: 35948820 DOI: 10.1007/s41061-022-00389-3] [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: 11/29/2021] [Accepted: 05/27/2022] [Indexed: 10/15/2022]
Abstract
Cyclometalated Ir(III) complexes exhibit strong phosphorescence emission with lifetime of submicroseconds to several microseconds at room temperature. Their synthetic versatility enables broad control of physical properties, such as charge and lipophilicity, as well as emission colors. These favorable properties have motivated the use of Ir(III) complexes in luminescent bioimaging applications. This review examines the recent progress in the development of phosphorescent biolabels and sensors based on Ir(III) complexes. It begins with a brief introduction about the basic principles of the syntheses and photophysical processes of cyclometalated Ir(III) complexes. Focus is placed on illustrating the broad imaging utility of Ir(III) complexes. Phosphorescent labels illuminating intracellular organelles, including mitochondria, lysosomes, and cell membranes, are summarized. Ir(III) complexes capable of visualization of tumor spheroids and parasites are also introduced. Facile chemical modification of the cyclometalating ligands endows the Ir(III) complexes with strong sensing ability. Sensors of temperature, pH, CO2, metal ions, anions, biosulfur species, reactive oxygen species, peptides, and viscosity have recently been added to the molecular imaging tools. This diverse utility demonstrates the potential of phosphorescent Ir(III) complexes toward bioimaging applications.
Collapse
|
24
|
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: 101] [Impact Index Per Article: 33.7] [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
|
25
|
Amarsy I, Papot S, Gasser G. Stimuli‐Responsive Metal Complexes for Biomedical Applications. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202205900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ivanna Amarsy
- Chimie ParisTech - PSL: Ecole nationale superieure de chimie de Paris PSL University FRANCE
| | - Sébastien Papot
- Université de Poitiers: Universite de Poitiers Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP) FRANCE
| | - Gilles Gasser
- Universite PSL Chimie ParisTech 11, rue Pierre et Marie Curie 75005 Paris FRANCE
| |
Collapse
|
26
|
Metal Peptide Conjugates in Cell and Tissue Imaging and Biosensing. Top Curr Chem (Cham) 2022; 380:30. [PMID: 35701677 PMCID: PMC9197911 DOI: 10.1007/s41061-022-00384-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 05/10/2022] [Indexed: 11/05/2022]
Abstract
Metal complex luminophores have seen dramatic expansion in application as imaging probes over the past decade. This has been enabled by growing understanding of methods to promote their cell permeation and intracellular targeting. Amongst the successful approaches that have been applied in this regard is peptide-facilitated delivery. Cell-permeating or signal peptides can be readily conjugated to metal complex luminophores and have shown excellent response in carrying such cargo through the cell membrane. In this article, we describe the rationale behind applying metal complexes as probes and sensors in cell imaging and outline the advantages to be gained by applying peptides as the carrier for complex luminophores. We describe some of the progress that has been made in applying peptides in metal complex peptide-driven conjugates as a strategy for cell permeation and targeting of transition metal luminophores. Finally, we provide key examples of their application and outline areas for future progress.
Collapse
|
27
|
Chen Q, Cuello-Garibo JA, Bretin L, Zhang L, Ramu V, Aydar Y, Batsiun Y, Bronkhorst S, Husiev Y, Beztsinna N, Chen L, Zhou XQ, Schmidt C, Ott I, Jager MJ, Brouwer AM, Snaar-Jagalska BE, Bonnet S. Photosubstitution in a trisheteroleptic ruthenium complex inhibits conjunctival melanoma growth in a zebrafish orthotopic xenograft model. Chem Sci 2022; 13:6899-6919. [PMID: 35774173 PMCID: PMC9200134 DOI: 10.1039/d2sc01646j] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/13/2022] [Indexed: 12/28/2022] Open
Abstract
In vivo data are rare but essential for establishing the clinical potential of ruthenium-based photoactivated chemotherapy (PACT) compounds, a new family of phototherapeutic drugs that are activated via ligand photosubstitution. Here a novel trisheteroleptic ruthenium complex [Ru(dpp)(bpy)(mtmp)](PF6)2 ([2](PF6)2, dpp = 4,7-diphenyl-1,10-phenanthroline, bpy = 2,2′-bipyridine, mtmp = 2-methylthiomethylpyridine) was synthesized and its light-activated anticancer properties were validated in cancer cell monolayers, 3D tumor spheroids, and in embryonic zebrafish cancer models. Upon green light irradiation, the non-toxic mtmp ligand is selectively cleaved off, thereby releasing a phototoxic ruthenium-based photoproduct capable notably of binding to nuclear DNA and triggering DNA damage and apoptosis within 24–48 h. In vitro, fifteen minutes of green light irradiation (21 mW cm−2, 19 J cm−2, 520 nm) were sufficient to generate high phototherapeutic indexes (PI) for this compound in a range of cancer cell lines including lung (A549), prostate (PC3Pro4), conjunctival melanoma (CRMM1, CRMM2, CM2005.1) and uveal melanoma (OMM1, OMM2.5, Mel270) cancer cell lines. The therapeutic potential of [2](PF6)2 was further evaluated in zebrafish embryo ectopic (PC3Pro4) or orthotopic (CRMM1, CRMM2) tumour models. The ectopic model consisted of red fluorescent PC3Pro4-mCherry cells injected intravenously (IV) into zebrafish, that formed perivascular metastatic lesions at the posterior ventral end of caudal hematopoietic tissue (CHT). By contrast, in the orthotopic model, CRMM1- and CRMM2-mCherry cells were injected behind the eye where they developed primary lesions. The maximally-tolerated dose (MTD) of [2](PF6)2 was first determined for three different modes of compound administration: (i) incubating the fish in prodrug-containing water (WA); (ii) injecting the prodrug intravenously (IV) into the fish; or (iii) injecting the prodrug retro-orbitally (RO) into the fish. To test the anticancer efficiency of [2](PF6)2, the embryos were treated 24 h after engraftment at the MTD. Optimally, four consecutive PACT treatments were performed on engrafted embryos using 60 min drug-to-light intervals and 90 min green light irradiation (21 mW cm−2, 114 J cm−2, 520 nm). Most importantly, this PACT protocol was not toxic to the zebrafish. In the ectopic prostate tumour models, where [2](PF6)2 showed the highest photoindex in vitro (PI > 31), the PACT treatment did not significantly diminish the growth of primary lesions, while in both conjunctival melanoma orthotopic tumour models, where [2](PF6)2 showed more modest photoindexes (PI ∼ 9), retro-orbitally administered PACT treatment significantly inhibited growth of the engrafted tumors. Overall, this study represents the first demonstration in zebrafish cancer models of the clinical potential of ruthenium-based PACT, here against conjunctival melanoma. A new tris-heteroleptic photoactivated chemotherapy ruthenium complex induces apoptosis upon green light activation in a zebrafish orthothopic conjunctival melanoma xenograft model.![]()
Collapse
Affiliation(s)
- Quanchi Chen
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing China.,Institute of Biology, Leiden University Leiden The Netherlands +31-71-527-4980
| | - Jordi-Amat Cuello-Garibo
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Ludovic Bretin
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Liyan Zhang
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Vadde Ramu
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Yasmin Aydar
- Institute of Biology, Leiden University Leiden The Netherlands +31-71-527-4980
| | - Yevhen Batsiun
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Sharon Bronkhorst
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Yurii Husiev
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Nataliia Beztsinna
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Lanpeng Chen
- Institute of Biology, Leiden University Leiden The Netherlands +31-71-527-4980
| | - Xue-Quan Zhou
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Claudia Schmidt
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstrasse 55 D-38106 Braunschweig Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstrasse 55 D-38106 Braunschweig Germany
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center Leiden The Netherlands
| | - Albert M Brouwer
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | | | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| |
Collapse
|
28
|
Rational design of mitochondria targeted thiabendazole-based Ir(III) biscyclometalated complexes for a multimodal photodynamic therapy of cancer. J Inorg Biochem 2022; 231:111790. [DOI: 10.1016/j.jinorgbio.2022.111790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022]
|
29
|
Liu B, Chen Z, Li Y, Du XF, Zhang W, Zhang W, Lai Y, Li Y. Brominated cyclometalated iridium(III) complexes for mitochondrial immobilization as potential anticancer agents. Dalton Trans 2022; 51:7650-7657. [PMID: 35510904 DOI: 10.1039/d2dt00587e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mitochondria-targeted iridium complexes for anticancer studies have received increasing attention in recent years. Herein, three cyclometalated iridium(III) complexes Ir1-Ir3 [Ir(N^C)2(N^N)](PF6) (N^N = 2,2'-bipyridine (bpy)) or 2-(5-bromopyridin-2-yl)benzo[d]thiazole (bpybt); [N^C = 2-phenylpyridine (ppy) or 2-phenylquinoline (pq) or 2-(4-bromophenyl)benzo[d]thiazole (bpbt)] had been explored as potential mitochondria-targeted anticancer agents. All of the complexes mainly localized in the mitochondria and could be fixed on the mitochondria through a nucleophilic reaction with reactive mitochondrial proteins. Further studies revealed that these complexes showed high anticancer activity, induced mitochondrial depolarization, elevated intracellular reactive oxygen species (ROS) levels, restrained thioredoxin reductase (TrxR) activity, and inhibited the formation of tumor cell colonies and angiogenesis. Further mechanistic studies showed that complex Ir2 could markedly stimulate the activation of caspase-3, regulate the expression of Bax and KI67, and trigger apoptosis.
Collapse
Affiliation(s)
- Ben Liu
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China.
| | - Zhiyin Chen
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China.
| | - Yu Li
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China.
| | - Xiang-Fu Du
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Wenjing Zhang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China. .,University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wei Zhang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China. .,University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China. .,University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yi Li
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China.
| |
Collapse
|
30
|
Tuning the emission maxima of iridium systems using benzimidazole-based cyclometallating framework. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
31
|
Wang X, Song K, Deng Y, Liu J, Peng Q, Lao X, Xu J, Wang D, Shi T, Li Y, Deng D, Miao Y. Benzothiazole-decorated iridium-based nanophotosensitizers for photodynamic therapy of cancer cells. Dalton Trans 2022; 51:3666-3675. [PMID: 35165680 DOI: 10.1039/d1dt04315c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Photodynamic therapy (PDT) is an effective non-invasive treatment for tumors. The structure of a photosensitizer has an important influence on light utilization and efficiency of singlet-oxygen generation. In this study, we synthesized three π-type iridium(III) complexes and modified the C^N and N^N ligands with benzothiazole (BTZ) to regulate their light-absorption capacity and efficiency of singlet-oxygen generation. We assembled the nano-photosensitizers by wrapping them with an amphiphilic polyethylene glycol polymer with folic acid-targeting function to improve their targeting ability and biocompatibility. Modification of the BTZ group on the C^N ligand enhanced the ability of the photosensitizer to generate singlet oxygen and improved the cell uptake and PDT efficacy of the corresponding nanophotosensitizer. We believe that this type of photosensitizer provides the basis for the design of new photosensitizers based on the structure of iridium(III) complexes.
Collapse
Affiliation(s)
- Xiang Wang
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Kang Song
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yong Deng
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Jie Liu
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Qin Peng
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Xiao Lao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Jiayu Xu
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Dong Wang
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Turong Shi
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yuhao Li
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Dan Deng
- Dermatology Department, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 200092, China.
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
32
|
|
33
|
Chen Z, Feng T, jinchao S, Karges J, Jin C, Zhao Y, Ji L, Chao H. A Mitochondria-Localized Iridium(III)-Chlorin E6 Conjugate for Synergistic Sonodynamic and Two-Photon Photodynamic Therapy Against Melanoma. Inorg Chem Front 2022. [DOI: 10.1039/d2qi00635a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
While melanoma in its early stages can be successfully treated, the prognosis strongly worsens with an increasing depth of the tumor. Capitalizing on this, there is an urgent need for...
Collapse
|
34
|
Zafon E, Echevarría I, Barrabés S, Manzano BR, Jalón FA, Rodríguez AM, Massaguer A, Espino G. Photodynamic therapy with mitochondria-targeted biscyclometallated Ir(III) complexes. Multi-action mechanism and strong influence of the cyclometallating ligand. Dalton Trans 2021; 51:111-128. [PMID: 34873601 DOI: 10.1039/d1dt03080a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Photodynamic therapy is an alternative to classical chemotherapy due to its potential to reduce side effects by a controlled activation of a photosensitizer through local irradiation with light. The photosensitizer then interacts with oxygen and generates reactive oxygen species. Iridium biscyclometallated complexes are very promising photosensitizers due to their exceptional photophysical properties and their ability to target mitochondria. Four Ir(III) biscyclometallated complexes of formula [Ir(C^N)2(N^N')]Cl, where N^N' is a ligand containing a benzimidazolyl fragment, have been synthesized and characterized. The C^N ligands were 2-phenylpyridinate (ppy) and 2-(2,4-difluorophenyl)pyridinate (dfppy). The complexes exhibited high photostability. The electrochemical and photophysical properties were modulated by both the cyclometallating and the ancillary ligands. The dfppy derivatives yielded the highest emission energy values, quantum yields of phosphorescence and excited state lifetimes. All complexes generated 1O2 in aerated solutions upon irradiation. Biological studies revealed that these complexes have a moderate cytotoxicity in the dark against different human cancer cell lines: prostate (PC-3), colon (CACO-2) and melanoma (SK-MEL-28), and against non-malignant fibroblasts (CCD-18Co). However, derivatives with ppy ligands ([1a]Cl, [2a]Cl) yielded a relevant photodynamic activity upon light irradiation (450 nm, 24.1 J cm-2), with phototoxicity indexes (EC50,dark/EC50,light) of 20.8 and 17.3, respectively, achieved in PC-3 cells. Mechanistic studies showed that these complexes are taken up by the cells through endocytosis and preferentially accumulate in mitochondria. Upon photoactivation, the complexes induced mitochondrial membrane depolarization and DNA damage, thus triggering cell death, mainly by apoptosis. Complex [1a]Cl is also able to oxidize NADH. This mitochondria-targeted photodynamic mechanism greatly inhibited the reproductive capacity of cancer cells and provides a valuable alternative to traditional chemotherapy for the controlled treatment of cancer.
Collapse
Affiliation(s)
- Elisenda Zafon
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003 Girona, Spain.
| | - Igor Echevarría
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, 09001, Burgos, Spain.
| | - Sílvia Barrabés
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003 Girona, Spain.
| | - Blanca R Manzano
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Químicas, Avda. Camilo J. Cela 10, 13071 Ciudad Real, Spain
| | - Félix A Jalón
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Químicas, Avda. Camilo J. Cela 10, 13071 Ciudad Real, Spain
| | - Ana M Rodríguez
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica. Escuela Técnica Superior de Ingenieros Industriales de Ciudad Real, Avda. Camilo J. Cela, 2, 13071 Ciudad Real, Spain
| | - Anna Massaguer
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003 Girona, Spain.
| | - Gustavo Espino
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, 09001, Burgos, Spain.
| |
Collapse
|
35
|
Zhu JH, Xu GX, Shum J, Lee LCC, Lo KKW. Tuning the organelle specificity and cytotoxicity of iridium(III) photosensitisers for enhanced phototheranostic applications. Chem Commun (Camb) 2021; 57:12008-12011. [PMID: 34709253 DOI: 10.1039/d1cc04982h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Luminescent cyclometallated iridium(III) complexes with a polyhedral oligomeric silsesquioxane (POSS) unit were designed as efficient theranostic agents that displayed tuneable organelle-targeting properties, minimal dark cytotoxicity and substantial photocytotoxicity even under hypoxic conditions.
Collapse
Affiliation(s)
- Jing-Hui Zhu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Guang-Xi Xu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Justin Shum
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
- State Key Laboratory of Terahertz and Millimetre Waves, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China
- Centre of Functional Photonics, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China
| |
Collapse
|
36
|
Pham TC, Nguyen VN, Choi Y, Lee S, Yoon J. Recent Strategies to Develop Innovative Photosensitizers for Enhanced Photodynamic Therapy. Chem Rev 2021; 121:13454-13619. [PMID: 34582186 DOI: 10.1021/acs.chemrev.1c00381] [Citation(s) in RCA: 792] [Impact Index Per Article: 198.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review presents a robust strategy to design photosensitizers (PSs) for various species. Photodynamic therapy (PDT) is a photochemical-based treatment approach that involves the use of light combined with a light-activated chemical, referred to as a PS. Attractively, PDT is one of the alternatives to conventional cancer treatment due to its noninvasive nature, high cure rates, and low side effects. PSs play an important factor in photoinduced reactive oxygen species (ROS) generation. Although the concept of photosensitizer-based photodynamic therapy has been widely adopted for clinical trials and bioimaging, until now, to our surprise, there has been no relevant review article on rational designs of organic PSs for PDT. Furthermore, most of published review articles in PDT focused on nanomaterials and nanotechnology based on traditional PSs. Therefore, this review aimed at reporting recent strategies to develop innovative organic photosensitizers for enhanced photodynamic therapy, with each example described in detail instead of providing only a general overview, as is typically done in previous reviews of PDT, to provide intuitive, vivid, and specific insights to the readers.
Collapse
Affiliation(s)
- Thanh Chung Pham
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Yeonghwan Choi
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Songyi Lee
- Department of Chemistry, Pukyong National University, Busan 48513, Korea.,Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
37
|
Malmberg R, Suter D, Blacque O, Venkatesan K. Monocyclometalated (C N) Gold(III) Metallacycles: Tunable Emission and Singlet Oxygen ( 1 O 2 ) Generation Properties. Chemistry 2021; 27:14410-14417. [PMID: 34406672 DOI: 10.1002/chem.202102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Indexed: 11/10/2022]
Abstract
The synthesis, characterization and photoluminescent properties of four cyclometalated (C N)-type gold(III) complexes bearing a bidentate diacetylide ligand, tolan-2,2'-diacetylide (tda), are reported. The complexes exhibit highly tunable excited state properties and show photoluminescence (PL) across the entire visible spectrum from sky-blue (λPL =493 nm) to red (λPL =675 nm) with absolute PL quantum yields (PLQY) of up to 75 % in solution, the highest PLQY found for any monocyclometalated Au(III) complex in solution. As a consequence of the use of the strongly rigidifying diacetylide bidentate ligand, a significant increase in the excited state lifetimes (τ0 =16-258 μs) was found in solution and in thin films. The complexes showed remarkable singlet oxygen generation in aerated solution with absolute singlet oxygen quantum yield (ϕ1Δ ) values reaching up to 7.5×10-5 and singlet oxygen lifetimes (τ0 1Δ ) in the range of 66-95 μs. Furthermore, the radiative and non-radiative rates of singlet oxygen were determined using the ϕ1Δ and τ0 1Δ values and correlations are drawn between the formation of singlet oxygen and its interaction with cyclometalated (C N) gold(III) complexes.
Collapse
Affiliation(s)
- Robert Malmberg
- Department of Molecular Sciences, MQ Photonics Research Centre and MQ Sustainable Energy Research Centre, Macquarie University, Sydney, NSW, 2109, Australia
| | - Dominik Suter
- Department of Molecular Sciences, MQ Photonics Research Centre and MQ Sustainable Energy Research Centre, Macquarie University, Sydney, NSW, 2109, Australia.,Department of Chemistry, University of Zurich, 8057, Zurich, Switzerland
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, 8057, Zurich, Switzerland
| | - Koushik Venkatesan
- Department of Molecular Sciences, MQ Photonics Research Centre and MQ Sustainable Energy Research Centre, Macquarie University, Sydney, NSW, 2109, Australia.,Department of Chemistry, University of Zurich, 8057, Zurich, Switzerland
| |
Collapse
|
38
|
Gkika KS, Kargaard A, Burke CS, Dolan C, Heise A, Keyes TE. Ru(ii)/BODIPY core co-encapsulated ratiometric nanotools for intracellular O 2 sensing in live cancer cells. RSC Chem Biol 2021; 2:1520-1533. [PMID: 34704057 PMCID: PMC8496004 DOI: 10.1039/d1cb00102g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022] Open
Abstract
Oxygen is a crucial reagent in many biochemical processes within living cells and its concentration can be an effective marker in disease, particularly in cancer where tissue hypoxia has been shown to indicate tumour growth. Probes that can reflect the oxygen concentration and distribution using ratiometric signals can be applied to a range of conventional methods without the need for specialised equipment and are particularly useful. The preparation and in cellulo study of luminescent ratiometric core–shell nanoparticles are presented. Here, a new lipophilic and oxygen-responsive Ru(ii) tris-heteroleptic polypyridyl complex is co-encapsulated with a reference BODIPY dye into the core of poly-l-lysine-coated polystyrene particles. The co-core encapsulation ensures oxygen response but reduces the impact of the environment on both probes. Single wavelength excitation of the particles, suspended in aqueous buffer, at 480 nm, triggers well-resolved dual emission from both dyes with peak maxima at 515 nm and 618 nm. A robust ratiometric oxygen response is observed from water, with a linear dynamic range of 3.6–262 μM which matches well with typical biological ranges. The uptake of RuBDP NPs was found to be cell-line dependent, but in cancerous cell lines, the particles were strongly permeable with late endosomal and partial lysosomal co-staining observed within 3 to 4 hours, eventually leading to extensive staining of the cytoplasm. The co-localisation of the ruthenium and BODIPY emission confirms that the particles remain intact in cellulo with no indication of dye leaching. The ratiometric O2 sensing response of the particles in cellulo was demonstrated using a plate-based assay and by confocal xyλ scanning of cells exposed to hypoxic conditions. Uptake and quantitative ratiometric oxygen sensing response of core–shell nanoparticles containing ruthenium probe and BODIPY reference is demonstrated using a plate reader-based assay and by confocal xyλ scanning of live cancer cells under hypoxic conditions.![]()
Collapse
Affiliation(s)
- Karmel Sofia Gkika
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Glasnevin Dublin 9 Ireland
| | | | - Christopher S Burke
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Glasnevin Dublin 9 Ireland .,Department of Chemistry, RCSI Dublin Ireland
| | - Ciaran Dolan
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Glasnevin Dublin 9 Ireland
| | - Andreas Heise
- Department of Chemistry, RCSI Dublin Ireland.,CÚRAM, SFI Research Centre for Medical Devices RCSI Dublin D02 Ireland.,AMBER, The SFI Advanced Materials and Bioengineering Research Centre RCSI Dublin D02 Ireland
| | - Tia E Keyes
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Glasnevin Dublin 9 Ireland
| |
Collapse
|
39
|
Guo X, Yang N, Ji W, Zhang H, Dong X, Zhou Z, Li L, Shen HM, Yao SQ, Huang W. Mito-Bomb: Targeting Mitochondria for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007778. [PMID: 34510563 DOI: 10.1002/adma.202007778] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 06/12/2021] [Indexed: 05/22/2023]
Abstract
Cancer has been one of the most common life-threatening diseases for a long time. Traditional cancer therapies such as surgery, chemotherapy (CT), and radiotherapy (RT) have limited effects due to drug resistance, unsatisfactory treatment efficiency, and side effects. In recent years, photodynamic therapy (PDT), photothermal therapy (PTT), and chemodynamic therapy (CDT) have been utilized for cancer treatment owing to their high selectivity, minor resistance, and minimal toxicity. Accumulating evidence has demonstrated that selective delivery of drugs to specific subcellular organelles can significantly enhance the efficiency of cancer therapy. Mitochondria-targeting therapeutic strategies are promising for cancer therapy, which is attributed to the essential role of mitochondria in the regulation of cancer cell apoptosis, metabolism, and more vulnerable to hyperthermia and oxidative damage. Herein, the rational design, functionalization, and applications of diverse mitochondria-targeting units, involving organic phosphine/sulfur salts, quaternary ammonium (QA) salts, peptides, transition-metal complexes, guanidinium or bisguanidinium, as well as mitochondria-targeting cancer therapies including PDT, PTT, CDT, and others are summarized. This review aims to furnish researchers with deep insights and hints in the design and applications of novel mitochondria-targeting agents for cancer therapy.
Collapse
Affiliation(s)
- Xiaolu Guo
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Naidi Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Wenhui Ji
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Hang Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Xiao Dong
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Zhiqiang Zhou
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
40
|
Holden L, Burke CS, Cullinane D, Keyes TE. Strategies to promote permeation and vectorization, and reduce cytotoxicity of metal complex luminophores for bioimaging and intracellular sensing. RSC Chem Biol 2021; 2:1021-1049. [PMID: 34458823 PMCID: PMC8341117 DOI: 10.1039/d1cb00049g] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/30/2021] [Indexed: 12/19/2022] Open
Abstract
Transition metal luminophores are emerging as important tools for intracellular imaging and sensing. Their putative suitability for such applications has long been recognised but poor membrane permeability and cytotoxicity were significant barriers that impeded early progress. In recent years, numerous effective routes to overcoming these issues have been reported, inspired in part, by advances and insights from the pharmaceutical and drug delivery domains. In particular, the conjugation of biomolecules but also other less natural synthetic species, from a repertoire of functional motifs have granted membrane permeability and cellular targeting. Such motifs can also reduce cytotoxicity of transition metal complexes and offer a valuable avenue to circumvent such problems leading to promising metal complex candidates for application in bioimaging, sensing and diagnostics. The advances in metal complex probes permeability/targeting are timely, as, in parallel, over the past two decades significant technological advances in luminescence imaging have occurred. In particular, super-resolution imaging is enormously powerful but makes substantial demands of its imaging contrast agents and metal complex luminophores frequently possess the photophysical characteristics to meet these demands. Here, we review some of the key vectors that have been conjugated to transition metal complex luminophores to promote their use in intra-cellular imaging applications. We evaluate some of the most effective strategies in terms of membrane permeability, intracellular targeting and what impact these approaches have on toxicity and phototoxicity which are important considerations in a luminescent contrast or sensing agent.
Collapse
Affiliation(s)
- Lorcan Holden
- School of Chemical Sciences, and National Centre for Sensor Research Dublin City University Dublin 9 Ireland
| | - Christopher S Burke
- School of Chemical Sciences, and National Centre for Sensor Research Dublin City University Dublin 9 Ireland
| | - David Cullinane
- School of Chemical Sciences, and National Centre for Sensor Research Dublin City University Dublin 9 Ireland
| | - Tia E Keyes
- School of Chemical Sciences, and National Centre for Sensor Research Dublin City University Dublin 9 Ireland
| |
Collapse
|
41
|
Liu B, Jiao J, Xu W, Zhang M, Cui P, Guo Z, Deng Y, Chen H, Sun W. Highly Efficient Far-Red/NIR-Absorbing Neutral Ir(III) Complex Micelles for Potent Photodynamic/Photothermal Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100795. [PMID: 34219286 DOI: 10.1002/adma.202100795] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/14/2021] [Indexed: 06/13/2023]
Abstract
A critical issue in photodynamic therapy (PDT) is inadequate reactive oxygen species (ROS) generation in tumors, causing inevitable survival of tumor cells that usually results in tumor recurrence and metastasis. Existing photosensitizers frequently suffer from relatively low light-to-ROS conversion efficiency with far-red/near-infrared (NIR) light excitation due to low-lying excited states that lead to rapid non-radiative decays. Here, a neutral Ir(III) complex bearing distyryl boron dipyrromethene (BODIPY-Ir) is reported to efficiently produce both ROS and hyperthermia upon far-red light activation for potentiating in vivo tumor suppression through micellization of BODIPY-Ir to form "Micelle-Ir". BODIPY-Ir absorbs strongly at 550-750 nm with a band maximum at 685 nm, and possesses a long-lived triplet excited state with sufficient non-radiative decays. Upon micellization, BODIPY-Ir forms J-type aggregates within Micelle-Ir, which boosts both singlet oxygen generation and the photothermal effect through the high molar extinction coefficient and amplification of light-to-ROS/heat conversion, causing severe cell apoptosis. Bifunctional Micelle-Ir that accumulates in tumors completely destroys orthotopic 4T1 breast tumors via synergistic PDT/photothermal therapy (PTT) damage under light irradiation, and enables remarkable suppression of metastatic nodules in the lungs, together without significant dark cytotoxicity. The present study offers an emerging approach to develop far-red/NIR photosensitizers toward potent cancer therapy.
Collapse
Affiliation(s)
- Bingqing Liu
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND, 58108, USA
| | - Jian Jiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Wan Xu
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND, 58108, USA
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, P. R. China
| | - Miya Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Peng Cui
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, 214122, P. R. China
| | - Zhengqing Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Huabing Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Wenfang Sun
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND, 58108, USA
| |
Collapse
|
42
|
Zhu JH, Yiu SM, Tang BZ, Lo KKW. Luminescent Neutral Cyclometalated Iridium(III) Complexes Featuring a Cubic Polyhedral Oligomeric Silsesquioxane for Lipid Droplet Imaging and Photocytotoxic Applications. Inorg Chem 2021; 60:11672-11683. [PMID: 34269564 DOI: 10.1021/acs.inorgchem.1c01728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New neutral iridium(III) complexes featuring a cubic polyhedral oligomeric silsesquioxane (POSS) unit, [Ir(N∧C)2(L1-POSS)] [HN∧C = 2-phenylpyridine (Hppy; 1), 2-phenylbenzothioazole (Hbt; 2), and 2-(1-naphthyl)benzothiazole (Hbsn; 3); L1-POSS = (E)-4-[(2-hydroxybenzylidene)amino]benzyl 3-heptakis(isobutyl)POSS-propyl carbamate], were designed and synthesized. Their POSS-free counterparts, [Ir(N∧C)2(L1)] [L1 = (E)-N-(4-hydroxymethylphenyl)-1-(2-hydroxyphenyl)methanimine; HN∧C = Hppy (1a), Hbt (2a), and Hbsn (3a)], and the poly(ethylene glycol) (PEG) derivatives [Ir(N∧C)2(L1-PEG)] [L1-PEG = (E)-4-[(2-hydroxybenzylidene)amino]benzyl 3-[2-[ω-methoxypoly(1-oxapropyl)]ethyl]carbamate; HN∧C = Hppy (1b), Hbt (2b), and Hbsn (3b)] were also prepared. The photophysical, photochemical, and biological properties of the POSS complexes were compared with those of their POSS-free and PEG-modified counterparts. Upon irradiation, all of these complexes displayed orange-to-red emission and long emission lifetimes under ambient conditions. The bsn complexes 3, 3a, and 3b exhibited the highest singlet oxygen (1O2) generation quantum yields (ΦΔ = 0.85-0.86) in aerated CH3CN. Laser-scanning confocal microscopy images revealed that complexes 1-3 and 1a-3a showed exclusive lipid-droplet staining upon cellular uptake, while the PEG derivatives 1b-3b displayed lysosomal localization. Complex 3 was utilized to study various lipid-droplet-related biological events including lipid-droplet accumulation under oleic acid stimulation, the movement of lipid droplets, and preadipocyte differentiation. Notably, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays indicated that the ppy complexes 1 and 1b and the bt complexes 2 and 2b were noncytotoxic both in the dark and upon irradiation at 450 nm for 5 min (IC50 > 200 μM), while the bsn complexes 3, 3a, and 3b showed low dark cytotoxicity (IC50 = 52.9 to >200 μM) and high photocytotoxicity (IC50 = 1.1-5.3 μM). The cellular uptake, internalization mechanisms, and cell death pathways of these complexes were also investigated. This work not only offers promising luminescent probes for lipid droplets through the structural modification of iridium(III) complexes but also paves the way to the construction of new reagents for theranostics.
Collapse
Affiliation(s)
- Jing-Hui Zhu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China
| | - Shek-Man Yiu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China
| | - Ben Zhong Tang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.,State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.,Center of Functional Photonics, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China
| |
Collapse
|
43
|
Zhu M, Zhang H, Ran G, Mangel DN, Yao Y, Zhang R, Tan J, Zhang W, Song J, Sessler JL, Zhang JL. Metal Modulation: An Easy-to-Implement Tactic for Tuning Lanthanide Phototheranostics. J Am Chem Soc 2021; 143:7541-7552. [PMID: 33973784 DOI: 10.1021/jacs.1c03041] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Phototheranostics constitute an emerging cancer treatment wherein the core diagnostic and therapeutic functions are integrated into a single photosensitizer (PS). Achieving the full potential of this modality requires being able to tune the photosensitizing properties of the PS in question. Structural modification of the organic framework represents a time-honored strategy for tuning the photophysical features of a given PS system. Here we report an easy-to-implement metal selection approach that allows for fine-tuning of excited-state energy dissipation and phototheranostics functions as exemplified by a set of lanthanide (Ln = Gd, Yb, Er) carbazole-containing porphyrinoid complexes. Femto- and nanosecond time-resolved spectroscopic studies, in conjunction with density functional theory calculations, revealed that the energy dissipation pathways for this set of PSs are highly dependent on the energy gap between the lowest triplet excited state of the ligand and the excited states of the coordinated Ln ions. The Yb complex displayed a balance of deactivation mechanisms that made it attractive as a potential combined photoacoustic imaging and photothermal/photodynamic therapy agent. It was encapsulated into mesoporous silica nanoparticles (MSN) to provide a biocompatible construct, YbL@MSN, which displays a high photothermal conversion efficiency (η = 45%) and a decent singlet oxygen quantum yield (ΦΔ = 31%). Mouse model studies revealed that YbL@MSN allows for both photoacoustic imaging and synergistic photothermal- and photodynamic-therapy-based tumor reduction in vivo. Our results lead us to suggest that metal selection represents a promising approach to fine-tuning the excited state properties and functional features of phototheranostics.
Collapse
Affiliation(s)
- Mengliang Zhu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hang Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Guangliu Ran
- Center for Advanced Quantum Studies, Department of Physics and Applied Optics Beijing Area Major Laboratory, Beijing Normal University, Beijing 100875, China
| | - Daniel N Mangel
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Yuhang Yao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ruijing Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jiao Tan
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Hunan Normal University, Changsha 410081, China
| | - Wenkai Zhang
- Center for Advanced Quantum Studies, Department of Physics and Applied Optics Beijing Area Major Laboratory, Beijing Normal University, Beijing 100875, China
| | - JianXin Song
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Hunan Normal University, Changsha 410081, China
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Jun-Long Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Chemistry and Chemical Engineering Guangdong Laboratory, Shantou 515031, China
| |
Collapse
|
44
|
Kritchenkov IS, Solomatina AI, Kozina DO, Porsev VV, Sokolov VV, Shirmanova MV, Lukina MM, Komarova AD, Shcheslavskiy VI, Belyaeva TN, Litvinov IK, Salova AV, Kornilova ES, Kachkin DV, Tunik SP. Biocompatible Ir(III) Complexes as Oxygen Sensors for Phosphorescence Lifetime Imaging. Molecules 2021; 26:2898. [PMID: 34068190 PMCID: PMC8153025 DOI: 10.3390/molecules26102898] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/04/2023] Open
Abstract
Synthesis of biocompatible near infrared phosphorescent complexes and their application in bioimaging as triplet oxygen sensors in live systems are still challenging areas of organometallic chemistry. We have designed and synthetized four novel iridium [Ir(N^C)2(N^N)]+ complexes (N^C-benzothienyl-phenanthridine based cyclometalated ligand; N^N-pyridin-phenanthroimidazol diimine chelate), decorated with oligo(ethylene glycol) groups to impart these emitters' solubility in aqueous media, biocompatibility, and to shield them from interaction with bio-environment. These substances were fully characterized using NMR spectroscopy and ESI mass-spectrometry. The complexes exhibited excitation close to the biological "window of transparency", NIR emission at 730 nm, and quantum yields up to 12% in water. The compounds with higher degree of the chromophore shielding possess low toxicity, bleaching stability, absence of sensitivity to variations of pH, serum, and complex concentrations. The properties of these probes as oxygen sensors for biological systems have been studied by using phosphorescence lifetime imaging experiments in different cell cultures. The results showed essential lifetime response onto variations in oxygen concentration (2.0-2.3 μs under normoxia and 2.8-3.0 μs under hypoxia conditions) in complete agreement with the calibration curves obtained "in cuvette". The data obtained indicate that these emitters can be used as semi-quantitative oxygen sensors in biological systems.
Collapse
Affiliation(s)
- Ilya S. Kritchenkov
- Institute of Chemistry, St. Petersburg State University, Universitetskii av., 26, 198504 St. Petersburg, Russia; (I.S.K.); (A.I.S.); (D.O.K.); (V.V.P.); (V.V.S.)
| | - Anastasia I. Solomatina
- Institute of Chemistry, St. Petersburg State University, Universitetskii av., 26, 198504 St. Petersburg, Russia; (I.S.K.); (A.I.S.); (D.O.K.); (V.V.P.); (V.V.S.)
| | - Daria O. Kozina
- Institute of Chemistry, St. Petersburg State University, Universitetskii av., 26, 198504 St. Petersburg, Russia; (I.S.K.); (A.I.S.); (D.O.K.); (V.V.P.); (V.V.S.)
| | - Vitaly V. Porsev
- Institute of Chemistry, St. Petersburg State University, Universitetskii av., 26, 198504 St. Petersburg, Russia; (I.S.K.); (A.I.S.); (D.O.K.); (V.V.P.); (V.V.S.)
| | - Victor V. Sokolov
- Institute of Chemistry, St. Petersburg State University, Universitetskii av., 26, 198504 St. Petersburg, Russia; (I.S.K.); (A.I.S.); (D.O.K.); (V.V.P.); (V.V.S.)
| | - Marina V. Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhskiy Research Medical University, Minin and Pozharsky sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.V.S.); (M.M.L.); (A.D.K.); (V.I.S.)
| | - Maria M. Lukina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhskiy Research Medical University, Minin and Pozharsky sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.V.S.); (M.M.L.); (A.D.K.); (V.I.S.)
| | - Anastasia D. Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhskiy Research Medical University, Minin and Pozharsky sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.V.S.); (M.M.L.); (A.D.K.); (V.I.S.)
| | - Vladislav I. Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhskiy Research Medical University, Minin and Pozharsky sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.V.S.); (M.M.L.); (A.D.K.); (V.I.S.)
- Becker&Hickl GmbH, Nunsdorfer Ring 7-9, 12277 Berlin, Germany
| | - Tatiana N. Belyaeva
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky av. 4, 194064 St. Petersburg, Russia; (T.N.B.); (I.K.L.); (A.V.S.); (E.S.K.)
| | - Ilia K. Litvinov
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky av. 4, 194064 St. Petersburg, Russia; (T.N.B.); (I.K.L.); (A.V.S.); (E.S.K.)
| | - Anna V. Salova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky av. 4, 194064 St. Petersburg, Russia; (T.N.B.); (I.K.L.); (A.V.S.); (E.S.K.)
| | - Elena S. Kornilova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky av. 4, 194064 St. Petersburg, Russia; (T.N.B.); (I.K.L.); (A.V.S.); (E.S.K.)
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnical University, Khlopina Str. 11, 194021 St. Petersburg, Russia
| | - Daniel V. Kachkin
- Faculty of Biology, St. Petersburg State University, Universitetskaya emb., 7/9, 199034 St. Petersburg, Russia;
| | - Sergey P. Tunik
- Institute of Chemistry, St. Petersburg State University, Universitetskii av., 26, 198504 St. Petersburg, Russia; (I.S.K.); (A.I.S.); (D.O.K.); (V.V.P.); (V.V.S.)
| |
Collapse
|
45
|
He G, Xu N, Ge H, Lu Y, Wang R, Wang H, Du J, Fan J, Sun W, Peng X. Red-Light-Responsive Ru Complex Photosensitizer for Lysosome Localization Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19572-19580. [PMID: 33900720 DOI: 10.1021/acsami.0c22551] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Photoresponsive ruthenium (Ru) complexes have been extensively studied in the photodynamic therapy (PDT) of cancer. The metal-to-ligand charge transfer (MLCT) absorption maximum of most Ru complexes is located in the short-wavelength visible region, which is well suited for superficial tumors but shows inefficient therapeutic effects for more deep-seated ones. Moreover, Ru complexes are primarily located in the mitochondria or nucleus, always resulting in high levels of dark toxicity and DNA mutation. Herein, we reported a new ruthenium complex (Ru-I) for red-light-triggered PDT. The activation wavelength of Ru-I is successfully extended to 660 nm. Importantly, the complex photosensitizer can be quickly taken up by cancer cells and selectively accumulated in the lysosome, an ideal localization for PDT purposes. Intratumoral injection of Ru-I into tumor-bearing mice achieved excellent therapeutic effects and thus holds great promise for applications in lysosome localization photodynamic therapy.
Collapse
Affiliation(s)
- Guangli He
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Ning Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Haoying Ge
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Yang Lu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Ran Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Hexiang Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, 26 Yucai Road, Jiangbei District, Ningbo 315016, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, 26 Yucai Road, Jiangbei District, Ningbo 315016, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, 26 Yucai Road, Jiangbei District, Ningbo 315016, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
46
|
Deng Y, Wang X, Liu Y, Xu Y, Zhang J, Huang F, Li B, Miao Y, Sun Y, Li Y. Dual-light triggered metabolizable nano-micelles for selective tumor-targeted photodynamic/hyperthermia therapy. Acta Biomater 2021; 119:323-336. [PMID: 33122146 DOI: 10.1016/j.actbio.2020.10.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 01/25/2023]
Abstract
Phototherapy, including photodynamic and photothermal therapies, is a non-invasive photo-triggered tumor treatment. Combination therapy and new synergistic therapeutic reagents may hold promise for improving these treatments. Herein, we report an amphiphilic iridium-based photosensitizer (C14-IP2000) loaded with a hydrophobic photo-thermal drug (ZnPc) to form nano-micelles (ZNPs) for dual-light triggered tumor phototherapy. The C14-IP2000 was contained within ZNPs consisting of an iridium complex core decorated with hydrophilic polyethylene glycol chains to extend the time in blood circulation, and two hydrophobic carbon chains to enhance the loading capacity and the hydrophobic interaction with the loaded reagent. The designed ZNPs showed effective blood circulation, passive tumor targeting ability, remarkable photodynamic conversion ability, and good photothermal conversion capability, and therefore may be used for combined tumor ablation. Our results demonstrated that the amphipathic bionic structure of ZNPs not only enables self-assembled reagent fabrication with prolonged circulation time and favorable metabolic characteristics for tumor combination therapy, but also provides a nanostructure strategy for the modification of functionalized reagents.
Collapse
Affiliation(s)
- Yong Deng
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xiang Wang
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yongtian Liu
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yao Xu
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jing Zhang
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Fei Huang
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Bing Li
- Department of Research and Development & Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai 201321, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai 201321, China
| | - Yuqing Miao
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yun Sun
- Department of Research and Development & Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai 201321, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai 201321, China.
| | - Yuhao Li
- Institute of Bismuth Science, College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
47
|
Gu Y, Wen H, Bai L, Zhou Y, Zhang H, Tian L, Zhang Y, Hao J, Liu Y. Exploring anticancer efficiency of mitochondria-targeted cyclometalated iridium(III) complexes. J Inorg Biochem 2020; 212:111215. [DOI: 10.1016/j.jinorgbio.2020.111215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022]
|
48
|
Lu C, Xu W, Shah H, Liu B, Xu W, Sun L, Qian SY, Sun W. In Vitro Photodynamic Therapy of Mononuclear and Dinuclear Iridium(III) Bis(terpyridine) Complexes. ACS APPLIED BIO MATERIALS 2020; 3:6865-6875. [PMID: 35019348 DOI: 10.1021/acsabm.0c00784] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Three mononuclear or dinuclear bis(terpyridine) (tpy) iridium(III) complexes bearing pyren-1-yl (pyr) group(s) were synthesized. Their photophysical properties in water and in vitro photodynamic therapy (PDT) effects toward the human lung epithelial cancer cell line A549 and the human epidermal skin cancer cell line A431 were investigated to evaluate the effects of dinuclear versus mononuclear complexes and the impact of the oligoether substituent at the ligand. All complexes possessed pyr-tpy ligand-associated charge transfer (1CT)/1π,π* absorption bands at 350-550 nm, with the dinuclear complex Ir3 showing the much enhanced absorptivity of this band. These complexes exhibited dual emission upon excitation at >430 nm in most cases, with the emitting states being ascribed to 1ILCT (intraligand charge transfer) and 3π,π*/3CT states, respectively. All complexes exhibited relatively weak to moderate cytotoxicity in the dark but high photocytotoxicity upon broadband visible light irradiation. Among them, the dinuclear complex Ir3 showed the highest intracellular reactive oxygen species (ROS) generation and PDT efficiency compared to its mononuclear counterpart Ir1. Introducing an oligoether substituent on one of the tpy ligands in Ir2 also improved its intracellular ROS generation and PDT efficacy compared to those induced by Ir1. Ir3 induced both mitochondrial dysfunction and lysosomal damage upon light activation toward both cell lines, whereas Ir1 and Ir2 caused both mitochondrial dysfunction and lysosomal damage in A431 cells but only lysosomal damage in A549 cells. The dominant cell death pathway induced by Ir1-Ir3 PDT is apoptosis.
Collapse
Affiliation(s)
- Cuifen Lu
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108-6050, United States.,Hubei Collaborative Innovation Center for Advanced Organochemical Materials and Ministry-of-Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, P. R. China
| | - Wan Xu
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Harshit Shah
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Bingqing Liu
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Wei Xu
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Liya Sun
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Steven Y Qian
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Wenfang Sun
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| |
Collapse
|
49
|
Li J, Chen T. Transition metal complexes as photosensitizers for integrated cancer theranostic applications. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213355] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
50
|
Shaikh S, Younis M, Rehman FU, Jiang H, Wang X. Specific Oxide Nanoclusters Enhance Intracellular Reactive Oxygen Species for Cancer-Targeted Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:9472-9480. [PMID: 32701296 DOI: 10.1021/acs.langmuir.0c01378] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Bio-nanotechnology based cancer therapeutics exponentially increase every year. A therapeutic strategy to induce intracellular reactive oxygen species (ROS) has received promising success in oncotherapy. In this study, the new strategy has been exploited by the treatment of iridium (Ir) and Fe2+ ions with cancer cells to biosynthesize the biocompatible fluorescent iridium oxide (IrO2) and iron oxide nanoclusters (NCs) under the specific redox heterogeneous microenvironment of these diseased cells and tumors. The hydroxyl radical produced by the presence of Fe2+ and H2O2 in cancer cells apparently increased the ROS level in cancer cells during the process of biosynthesized NCs and, hence, simultaneously instigated apoptosis of relevant cells. Therefore, intracellular ROS-mediated in situ biosynthesis of IrO2 and iron oxide NCs may also act as anticancer agents and provide a promising pathway for targeted cancer therapy.
Collapse
Affiliation(s)
- Sana Shaikh
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Muhammad Younis
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Fawad Ur Rehman
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| |
Collapse
|