1
|
Hao L, Ling YY, Wang J, Shen QH, Li ZY, Tan CP. Theranostic Rhenium(I)-Based ER-Phagy Retardant Promotes Immunogenic Cell Death. J Med Chem 2024. [PMID: 39720929 DOI: 10.1021/acs.jmedchem.4c01948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
ER-phagy is a double-edged sword in the occurrence, development, and treatment of cancer; especially, its functions in immunotherapy are still unknown. In this work, we designed a theranostic Re complex (Re1) containing a BODIPY-derived ligand and a β-carboline ligand to target the endoplasmic reticulum (ER) and block ER-phagy at the late stages. Interestingly, as validated both in vitro and in vivo, ER-phagy blockage greatly enhances the capability of Re1 to induce immunogenic cell death (ICD). In summary, we dexterously fused two molecular modules for ER targeting and ER-phagy blockage into a coordination complex to afford a highly effective ICD inducer, which provides clues for designing new cancer immunotherapeutics.
Collapse
Affiliation(s)
- Liang Hao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Yu-Yi Ling
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Jie Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Qing-Hua Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Zhi-Yuan Li
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, P. R. China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Guangzhou 510006, P. R. China
| |
Collapse
|
2
|
Montesdeoca N, Mohr JM, Kruss S, Karges J. Shift of cell-death mechanisms in primary human neutrophils with a ruthenium photosensitizer. J Biol Inorg Chem 2024:10.1007/s00775-024-02088-4. [PMID: 39673631 DOI: 10.1007/s00775-024-02088-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 11/30/2024] [Indexed: 12/16/2024]
Abstract
Primary human neutrophils are the most abundant human white blood cells and are central for innate immunity. They act as early responders at inflammation sites, guided by chemotactic gradients to find infection or inflammation sites. Neutrophils can undergo both apoptosis as well as NETosis. NETosis is a form of neutrophil cell death that releases chromatin-based extracellular traps (NETs) to capture and neutralize pathogens. Understanding or controlling the balance between these cell-death mechanisms is crucial. In this study, the chemical synthesis and biologic assessment of a ruthenium complex as a light-activated photosensitizer that creates reactive oxygen species (ROS) in primary human neutrophils is reported. The ruthenium complex remains non-toxic in the dark. However, upon exposure to blue light at 450 nm, it exhibits potent cytotoxic effects in both cancerous and non-cancerous cell lines. Interestingly, the metal complex shifts the cell-death mechanism of primary human neutrophils from NETosis to apoptosis. Cells irradiated directly by the light source immediately undergo apoptosis, whereas those further away from the light source perform NETosis at a slower rate. This indicates that high ROS levels trigger apoptosis and lower ROS levels NETosis. The ability to control the type of cell death undergone in primary human neutrophils could have implications in managing acute and chronic infectious diseases.
Collapse
Affiliation(s)
- Nicolás Montesdeoca
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Jennifer M Mohr
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Sebastian Kruss
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
- Fraunhofer Institute for Microelectronic Circuits and Systems, Duisburg, Germany.
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
3
|
Marco A, Kasparkova J, Bautista D, Kostrhunova H, Cutillas N, Markova L, Novohradsky V, Ruiz J, Brabec V. A Novel Substituted Benzo[ g]quinoxaline-Based Cyclometalated Ru(II) Complex as a Biocompatible Membrane-Targeted PDT Colon Cancer Stem Cell Agent. J Med Chem 2024; 67:21470-21485. [PMID: 39620973 DOI: 10.1021/acs.jmedchem.4c02357] [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: 12/13/2024]
Abstract
Herein, we describe and investigate biological activity of three octahedral ruthenium(II) complexes of the type [Ru(C∧N)(phen)2]+, RuL1-RuL3, containing a π-expansive cyclometalating substituted benzo[g]quinoxaline ligand (C∧N ligand) (phen = 1,10-phenanthroline). Compounds RuL1-RuL3 in cervical, melanoma, and colon human cancer cells exhibit high phototoxicity after irradiation with light (particularly blue), with the phototoxicity index reaching 100 for the complex RuL2 in most sensitive HCT116 cells. RuL2 accumulates in the cellular membranes. If irradiated, it induces lipid peroxidation, likely connected with photoinduced ROS generation. Oxidative damage to the fatty acids leads to the attenuation of the membranes, the activation of caspase 3, and the triggering of the apoptotic pathway, thus implementing membrane-localized photodynamic therapy. RuL2 is the first photoactive ruthenium-based complex capable of killing the hardly treatable colon cancer stem cells, a highly resilient subpopulation within a heterogeneous tumor mass, responsible for tumor recurrence and the metastatic progression of cancer.
Collapse
Affiliation(s)
- Alicia Marco
- Departamento de Química Inorgánica, Universidad de Murcia and Murcia BioHealth Research Institute (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61 200 Brno, Czech Republic
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, CZ-783 71 Olomouc, Czech Republic
| | | | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61 200 Brno, Czech Republic
| | - Natalia Cutillas
- Departamento de Química Inorgánica, Universidad de Murcia and Murcia BioHealth Research Institute (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Lenka Markova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61 200 Brno, Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61 200 Brno, Czech Republic
| | - José Ruiz
- Departamento de Química Inorgánica, Universidad de Murcia and Murcia BioHealth Research Institute (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, CZ-61 200 Brno, Czech Republic
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, CZ-783 71 Olomouc, Czech Republic
| |
Collapse
|
4
|
Yu J, Xu W, Chen H, Yuan H, Wang Y, Qian X, Zhang J, Ji Y, Zhao Q, Li S. Charge Engineering of Star-Shaped Organic Photosensitizers Enables Efficient Type-I Radicals for Photodynamic Therapy of Multidrug-Resistant Bacterial Infection. Adv Healthc Mater 2024:e2402615. [PMID: 39648533 DOI: 10.1002/adhm.202402615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/18/2024] [Indexed: 12/10/2024]
Abstract
Infection induced by multidrug-resistant bacteria is now the second most common cause of accidental death worldwide. However, identifying a high-performance strategy with good efficiency and low toxicity is still urgently needed. Antibacterial photodynamic therapy (PDT) is considered a non-invasive and efficient approach with minimal drug resistance. Whereas, the precise molecular design for highly efficient oxygen-independent type-I photosensitizers is still undefined. In this work, the regulation of the positive charge of star-shaped NIR-emissive organic photosensitizers can boost radical generation for the efficient treatment of wounds infected with multidrug-resistant bacteria. With positive charge engineering, TPAT-DNN, which has six positive charges, mainly produces hydroxyl radicals via the type-I pathway, while TPAT-DN, which has three positive charges, tends to generate singlet oxygen and superoxide radicals. For multidrug-resistant bacteria, TPAT-DNN exhibited specific killing effects on multidrug-resistant gram-positive bacteria at low concentrations, while TPAT-DN is similar antibacterial effects on both multidrug-resistant gram-negative and gram-positive bacteria. Furthermore, the efficiency and safety of TPAT-DNN for eradicating multidrug-resistant bacteria methicillin-resistant S. aureus (MRSA) infection and accelerating wound healing in an MRSA-infected mouse model are demonstrated. This work offers a new approach toward manipulating efficient type-I photosensitizers for MRSA treatment.
Collapse
Affiliation(s)
- Jie Yu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Wenchang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Huan Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
| | - Haitao Yuan
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Yu Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Xiandie Qian
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Jie Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yu Ji
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Qi Zhao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
5
|
Zheng H, Wang K, Ji D, Liu X, Wang C, Jiang Y, Jia Z, Xiong B, Ling Y, Miao J. Novel tris-bipyridine based Ru(II) complexes as type-I/-II photosensitizers for antitumor photodynamic therapy through ferroptosis and immunogenic cell death. Eur J Med Chem 2024; 279:116909. [PMID: 39357314 DOI: 10.1016/j.ejmech.2024.116909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Ru(II) complexes have attracted attention as photosensitizers for their promising photodynamic properties. Herein, novel tris-bipyridine based Ru(II) complexes (6a-e) were synthesized by introducing saturated heterocycles to improve photodynamic properties and lipid-water partition coefficients. Among them, 6d demonstrated significant phototoxicity towards three cancer cells, with IC50 values of 5.66-7.17 μM, exceeding values in dark (IC50s > 100 μM). Under hypoxic conditions, 6d maintained excellent photodynamic activity in A549 cells, with PI values exceeding 24, highlighting its potential for highly effective type-I/-II photodynamic therapy by inducing ROS generation, oxidative stress, and mitochondrial damage. Additionally, it induced ferroptosis and immunogenic cell death of A549 cells by regulating the expression of relevant markers. Finally, 6d remarkably inhibited the growth of A549 transplanted tumor growth by 95.4 %. This Ru(II) complex shows great potential for cancer treatment with its potent photodynamic activity and diverse mechanisms of tumor cell death.
Collapse
Affiliation(s)
- Hongwei Zheng
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Kai Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Dongliang Ji
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Xiao Liu
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Chen Wang
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Yangyang Jiang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Zihan Jia
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China
| | - Biao Xiong
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China.
| | - Yong Ling
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China.
| | - Jiefei Miao
- Department of Oncology, Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, Nantong 226001, China.
| |
Collapse
|
6
|
Yin CW, Zhuo LT, Chen JY, Lin YH, Lin YT, Chen HY, Tsai MK, Chen YJ. Intrinsic 77 K Phosphorescence Characteristics and Computational Modeling of Ru(II)-(Bidentate Cyclometalated-Aromatic Ligand) Chromophores: Their Relatively Low Nonradiative Rate Constants Originating from Low Spin-Orbit Coupling Driven Vibronic Coupling Amplitudes between Emitting and Ground States. Inorg Chem 2024; 63:21981-21993. [PMID: 39509593 DOI: 10.1021/acs.inorgchem.4c03390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
We investigated the photoinduced relaxation of Kasha-type emitting ruthenium-(bidentate cyclometalated aromatic ligand), Ru-CM, chromophores of [Ru(pzpy)2(CM)]+ ions (CM = 1-phenylisoquinoline, 2,3-diphenylpyrazine, and 1,4-diazatriphenylene and pzpy = 2-pyrazol-1-yl-pyridine). This is the first report of the phosphorescence behavior of pure Ru-(bidentate CM) chromophores. The 77 K photoinduced relaxation characteristics of phosphorescence chromophores showed emission quantum yields higher than those of reference Ru-bpy (bpy = 2,2'-bipyridine) chromophores in the emission region of 670-900 nm. This phenomenon of the Ru-CM chromophores could be attributed to their unusually low magnitudes for 77 K nonradiative rate constants (kNRD), although their radiative rate-constants (kRAD) are not remarkable. In order to examine the 77 K photoinduced behavioral relaxation difference between Ru-CM and Ru-bpy chromophores, we used computational simulation, applying the fundamental formalism of kRAD and temperature-independent kNRD equations, which included calculated spin-orbit coupling values.
Collapse
Affiliation(s)
- Chi-Wei Yin
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Li-Ting Zhuo
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Jie Ying Chen
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Yu-Hui Lin
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Yu-Ting Lin
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Hsing-Yin Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Ming-Kang Tsai
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan, R.O.C
| | - Yuan Jang Chen
- Department of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| |
Collapse
|
7
|
Cui X, Fan J, Gao Y, Zhou X, Zhang C, Meng Q. Designing Quasi-Intrinsic Photosensitizers with Dual Function of Fluorescence Imaging and Photodynamic Therapy. J Med Chem 2024; 67:19826-19836. [PMID: 39485727 DOI: 10.1021/acs.jmedchem.4c02213] [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: 11/03/2024]
Abstract
Photosensitizers (PSs) with effective two-photon absorption in the therapeutic window are the key to two-photon photodynamic therapy. However, the traditional exogenous PSs usually lead to rejection in the body. Besides, the precise visualization of treatments proposes new demands and challenges for the design of PSs. Accordingly, in this work, a series of quasi-intrinsic PSs are obtained based on the artificial base 2-amino-8-(1'-β-d-2'-deoxyribofuranosyl)-imidazo[1,2-α]-1,3,5-triazin-4(8H)-one (P). The calculations show that the structural modification could enhance the two-photon absorption and fluorescence emission, which is beneficial for tumor localization. Furthermore, the reduced singlet-triplet energy gaps and enhanced spin-orbit coupling contribute to the rapid intersystem crossing process, which results in a triplet state with high quantum yields. To ensure the phototherapeutic performance of the newly designed PSs, we also examined the vertical electron affinity and vertical ionization potential for generation of superoxide anions, as well as the T1 energy required to produce singlet oxygen.
Collapse
Affiliation(s)
- Xixi Cui
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Jianzhong Fan
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Yang Gao
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Xucong Zhou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, P. R. China
| | - Changzhe Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Qingtian Meng
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| |
Collapse
|
8
|
Park M, Nam JS, Kim T, Yoon G, Kim S, Lee C, Lee CG, Park S, Bejoymohandas KS, Yang J, Kwon YH, Lee YJ, Seo JK, Min D, Park T, Kwon TH. Rational Design of Biocompatible Ir(III) Photosensitizer to Overcome Drug-Resistant Cancer via Oxidative Autophagy Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407236. [PMID: 39540573 DOI: 10.1002/advs.202407236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/25/2024] [Indexed: 11/16/2024]
Abstract
Autophagy is a crucial quality control mechanism that degrades damaged cellular components through lysosomal fusion with autophagosomes. However, elevated autophagy levels can promote drug resistance in cancer cells, enhancing their survival. Downregulation of autophagy through oxidative stress is a clinically promising strategy to counteract drug resistance, yet precise control of oxidative stress in autophagic proteins remains challenging. Here, a molecular design strategy of biocompatible neutral Ir(III) photosensitizers is demonstrated, B2 and B4, for precise reactive oxygen species (ROS) control at lysosomes to inhibit autophagy. The underlying molecular mechanisms for the biocompatibility and lysosome selectivity of Ir(III) complexes are explored by comparing B2 with the cationic or the non-lysosome-targeting analogs. Also, the biological mechanisms for autophagy inhibition via lysosomal oxidation are explored. Proteome analyses reveal significant oxidation of proteins essential for autophagy, including lysosomal and fusion-mediator proteins. These findings are verified in vitro, using mass spectrometry, live cell imaging, and a model SNARE complex. The anti-tumor efficacy of the precise lysosomal oxidation strategy is further validated in vivo with B4, engineered for red light absorbance. This study is expected to inspire the therapeutic use of spatiotemporal ROS control for sophisticated modulation of autophagy.
Collapse
Affiliation(s)
- Mingyu Park
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- X-dynamic Research Center, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jung Seung Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- X-dynamic Research Center, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Taehyun Kim
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Gwangsu Yoon
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- X-dynamic Research Center, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seoyoon Kim
- Department of Chemistry, 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
- X-dynamic Research Center, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Chae Gyu Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Sungjin Park
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Kochan S Bejoymohandas
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Jihyeon Yang
- Research Center, O2MEDi inc., Ulsan, 44919, Republic of Korea
| | - Yoon Hee Kwon
- Research Center, O2MEDi inc., Ulsan, 44919, Republic of Korea
| | - Yoo Jin Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jeong Kon Seo
- Research Center, O2MEDi inc., Ulsan, 44919, Republic of Korea
| | - Duyoung Min
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Taiho Park
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Tae-Hyuk Kwon
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- X-dynamic Research Center, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Research Center, O2MEDi inc., Ulsan, 44919, Republic of Korea
| |
Collapse
|
9
|
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
|
10
|
Qiao W, Ma T, Xie G, Xu J, Yang ZR, Zhong C, Jiang H, Xia J, Zhang L, Zhu J, Li Z. Supramolecular H-Aggregates of Squaraines with Enhanced Type I Photosensitization for Combined Photodynamic and Photothermal Therapy. ACS NANO 2024; 18:25671-25684. [PMID: 39223995 DOI: 10.1021/acsnano.4c07764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Combined photodynamic and photothermal therapy (PDT and PTT) can achieve more superior therapeutic effects than the sole mode by maximizing the photon utilization, but there remains a significant challenge in the development of related single-molecule photosensitizers (PSs), particularly those with type I photosensitization. In this study, self-assembly of squaraine dyes (SQs) is shown to be a promising strategy for designing PSs for combined type I PDT and PTT, and a supramolecular PS (TPE-SQ7) has been successfully developed through subtle molecular design of an indolenine SQ, which can self-assemble into highly ordered H-aggregates in aqueous solution as well as nanoparticles (NPs). In contrast to the typical quenching effect of H-aggregates on reactive oxygen species (ROS) generation, our results encouragingly manifest that H-aggregates can enhance type I ROS (•OH) generation by facilitating the intersystem crossing process while maintaining a high PTT performance. Consequently, TPE-SQ7 NPs with ordered H-aggregates not only exhibit superior combined therapeutic efficacy than the well-known PS (Ce6) under both normoxic and hypoxic conditions but also have excellent biosafety, making them have important application prospects in tumor phototherapy and antibacterial fields. This study not only proves that the supramolecular self-assembly of SQs is an effective strategy toward high-performance PSs for combined type I PDT and PTT but also provides a different understanding of the effect of H-aggregates on the PDT performance.
Collapse
Affiliation(s)
- Weiguo Qiao
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Teng Ma
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ge Xie
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jingwen Xu
- School of Chemistry, Chemical Engineering and Life Science, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Zhuo-Ran Yang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cheng Zhong
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Hao Jiang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jianlong Xia
- School of Chemistry, Chemical Engineering and Life Science, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Lianbin Zhang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhong'an Li
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
11
|
Chauhan N, Koli M, Ghosh R, Majumdar AG, Ghosh A, Ghanty TK, Mula S, Patro BS. A BODIPY-Naphtholimine-BF 2 Dyad for Precision Photodynamic Therapy, Targeting, and Dual Imaging of Endoplasmic Reticulum and Lipid Droplets in Cancer. JACS AU 2024; 4:2838-2852. [PMID: 39211629 PMCID: PMC11350743 DOI: 10.1021/jacsau.3c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 05/11/2024] [Accepted: 05/22/2024] [Indexed: 09/04/2024]
Abstract
Currently, effective therapeutic modalities for pancreatic ductal adenocarcinoma (PDAC) are quite limited, leading to gloomy prognosis and ∼6-months median patient survival. Recent advances showed the promise of photodynamic therapy (PDT) for PDAC patients. Next generation photosensitizers (PS) are based on "organelle-targeted-PDT" and provide new paradigm in the field of precision medicines to address the current challenge for treating PDAC. In this investigation, we have constructed a novel PS, named as N b B, for precise and simultaneous targeting of endoplasmic reticulum (ER) and lipid droplets (LDs) in PDAC, based on the fact that malignant PDAC cells are heavily relying on ER for hormone synthesis. Our live cell imaging and fluorescence recovery after photobleaching (FRAP) experiments revealed that N b B is quickly targeted to ER and subsequently to LDs and shows simultaneous dual fluorescence color due to polar and nonpolar milieu of ER and LDs. Interestingly, the same molecule generates triplet state and singlet oxygen efficiently and causes robust ER stress and cellular lipid peroxidation, leading to apoptosis in two different PDAC cells in the presence of light. Together, we present, for the first time, a potential next generation precision medicine for ER-LD organelle specific imaging and PDT of pancreatic cancer.
Collapse
Affiliation(s)
- Nitish Chauhan
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Mrunesh Koli
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Rajib Ghosh
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Ananda Guha Majumdar
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Ayan Ghosh
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
| | - Tapan K. Ghanty
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Soumyaditya Mula
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| | - Birija Sankar Patro
- Bio-Organic
Division, Radiation
and Photochemistry Division, Laser and Plasma Technology
Division, Bio-Science
Group, Bhabha Atomic Research Centre, Mod. Lab, Trombay, Mumbai-400085, India
- Homi Bhabha
National Institute, Anushaktinagar, Mumbai-400094, India
| |
Collapse
|
12
|
Bonelli J, Ortega-Forte E, Vigueras G, Follana-Berná J, Ashoo P, Abad-Montero D, Isidro N, López-Corrales M, Hernández A, Ortiz J, Izquierdo-García E, Bosch M, Rocas J, Sastre-Santos Á, Ruiz J, Marchán V. A Nanoencapsulated Ir(III)-Phthalocyanine Conjugate as a Promising Photodynamic Therapy Anticancer Agent. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38916-38930. [PMID: 39041453 PMCID: PMC11299137 DOI: 10.1021/acsami.4c05181] [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: 03/29/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Despite the potential of photodynamic therapy (PDT) in cancer treatment, the development of efficient and photostable photosensitizing molecules that operate at long wavelengths of light has become a major hurdle. Here, we report for the first time an Ir(III)-phthalocyanine conjugate (Ir-ZnPc) as a novel photosensitizer for high-efficiency synergistic PDT treatment that takes advantage of the long-wavelength excitation and near infrared (NIR) emission of the phthalocyanine scaffold and the known photostability and high phototoxicity of cyclometalated Ir(III) complexes. In order to increase water solubility and cell membrane permeability, the conjugate and parent zinc phthalocyanine (ZnPc) were encapsulated in amphoteric redox-responsive polyurethane-polyurea hybrid nanocapsules (Ir-ZnPc-NCs and ZnPc-NCs, respectively). Photobiological evaluations revealed that the encapsulated Ir-ZnPc conjugate achieved high photocytotoxicity in both normoxic and hypoxic conditions under 630 nm light irradiation, which can be attributed to dual Type I and Type II reactive oxygen species (ROS) photogeneration. Interestingly, PDT treatments with Ir-ZnPc-NCs and ZnPc-NCs significantly inhibited the growth of three-dimensional (3D) multicellular tumor spheroids. Overall, the nanoencapsulation of Zn phthalocyanines conjugated to cyclometalated Ir(III) complexes provides a new strategy for obtaining photostable and biocompatible red-light-activated nano-PDT agents with efficient performance under challenging hypoxic environments, thus offering new therapeutic opportunities for cancer treatment.
Collapse
Affiliation(s)
- Joaquín Bonelli
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat
de Barcelona (UB), and Institut de Biomedicina de la Universitat de
Barcelona (IBUB), Martí
i Franquès 1-11, E-08028 Barcelona, Spain
- Ecopol
Tech S.L., Nanobiotechnological Polymers
Division, R&D Department, El Foix Business Park, Indústria 7, E-43720 L’Arboç del Penedès, Tarragona, Spain
| | - Enrique Ortega-Forte
- Departamento
de Química Inorgánica, Universidad
de Murcia, and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Gloria Vigueras
- Departamento
de Química Inorgánica, Universidad
de Murcia, and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Jorge Follana-Berná
- Área
de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández, Avda. de la Universidad s/n, E-03203 Elche, Spain
| | - Pezhman Ashoo
- Departamento
de Química Inorgánica, Universidad
de Murcia, and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Diego Abad-Montero
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat
de Barcelona (UB), and Institut de Biomedicina de la Universitat de
Barcelona (IBUB), Martí
i Franquès 1-11, E-08028 Barcelona, Spain
| | - Neus Isidro
- Ecopol
Tech S.L., Nanobiotechnological Polymers
Division, R&D Department, El Foix Business Park, Indústria 7, E-43720 L’Arboç del Penedès, Tarragona, Spain
| | - Marta López-Corrales
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat
de Barcelona (UB), and Institut de Biomedicina de la Universitat de
Barcelona (IBUB), Martí
i Franquès 1-11, E-08028 Barcelona, Spain
| | - Adrián Hernández
- Área
de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández, Avda. de la Universidad s/n, E-03203 Elche, Spain
| | - Javier Ortiz
- Área
de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández, Avda. de la Universidad s/n, E-03203 Elche, Spain
| | - Eduardo Izquierdo-García
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat
de Barcelona (UB), and Institut de Biomedicina de la Universitat de
Barcelona (IBUB), Martí
i Franquès 1-11, E-08028 Barcelona, Spain
| | - Manel Bosch
- Unitat
de Microscòpia Òptica Avançada, Centres Científics
i Tecnològics, Universitat de Barcelona, Av. Diagonal 643, E-08028 Barcelona, Spain
| | - Josep Rocas
- Ecopol
Tech S.L., Nanobiotechnological Polymers
Division, R&D Department, El Foix Business Park, Indústria 7, E-43720 L’Arboç del Penedès, Tarragona, Spain
| | - Ángela Sastre-Santos
- Área
de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández, Avda. de la Universidad s/n, E-03203 Elche, Spain
| | - José Ruiz
- Departamento
de Química Inorgánica, Universidad
de Murcia, and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Vicente Marchán
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat
de Barcelona (UB), and Institut de Biomedicina de la Universitat de
Barcelona (IBUB), Martí
i Franquès 1-11, E-08028 Barcelona, Spain
| |
Collapse
|
13
|
Wei L, Kushwaha R, Sadhukhan T, Wu H, Dao A, Zhang Z, Zhu H, Gong Q, Ru J, Liang C, Zhang P, Banerjee S, Huang H. Dinuclear Tridentate Ru(II) Complex with Strong Near-Infrared Light-Triggered Anticancer Activity. J Med Chem 2024; 67:11125-11137. [PMID: 38905437 DOI: 10.1021/acs.jmedchem.4c00624] [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: 06/23/2024]
Abstract
The design of the dinuclear Ru(II) complex (Ru2) with strong near-infrared (NIR) absorption properties has been reported for efficient anticancer phototherapy. Under 700 nm LED light excitation, Ru2 exhibited remarkable synergistic type I/II photosensitization ability and photocatalytic activity toward intracellular biomolecules. Ru2 showed impressive 700 nm light-triggered anticancer activity under normoxia and hypoxia compared with the clinically used photosensitizer Chlorin e6. The mechanistic studies showed that Ru2 induced intracellular redox imbalance and perturbed the energy metabolism and biosynthesis in A549 cancer cells. Overall, this work provides a new strategy for developing efficient metal-based complexes for anticancer phototherapy under NIR light.
Collapse
Affiliation(s)
- Li Wei
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Tumpa Sadhukhan
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Haorui Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Anyi Dao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Zhishang Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Haotu Zhu
- Department of Oncology, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Qiufang Gong
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejinag 325035, P.R. China
| | - Jiaxi Ru
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejinag 325035, P.R. China
| | - Chao Liang
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejinag 325035, P.R. China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Huaiyi Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| |
Collapse
|
14
|
Welsh A, Matshitse R, Khan SF, Nyokong T, Prince S, Smith GS. Trinuclear ruthenium(II) polypyridyl complexes: Evaluation as photosensitizers for enhanced cervical cancer treatment. J Inorg Biochem 2024; 256:112545. [PMID: 38581803 DOI: 10.1016/j.jinorgbio.2024.112545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/23/2024] [Accepted: 03/24/2024] [Indexed: 04/08/2024]
Abstract
Trinuclear ruthenium(II) polypyridyl complexes anchored to benzimidazole-triazine / trisamine scaffolds were investigated as photosensitizers for photodynamic therapy. The trinuclear complexes were noted to produce a significant amount of singlet oxygen in both DMF and aqueous media, are photostable and show appreciable emission quantum yields (ɸem). In our experimental setting, despite the moderate phototoxic activity in the HeLa cervical cancer cell line, the phototoxic indices (PI) of the trinuclear complexes are superior relative to the PIs of a clinically approved photosensitizer, Photofrin®, and the pro-drug 5-aminolevulinic acid (PI: >7 relative to PI: >1 and PI: 4.4 for 5-aminolevulinic acid and Photofrin®, respectively). Furthermore, the ruthenium complexes were noted to show appreciable long-term cytotoxicity upon light irradiation in HeLa cells in a concentration-dependent manner. Consequently, this long-term activity of the ruthenium(II) polypyridyl complexes embodies their ability to reduce the probability of the recurrence of cervical cancer. Taken together, this presents a strong motivation for the development of polymetallic complexes as anticancer agents.
Collapse
Affiliation(s)
- Athi Welsh
- Department of Chemistry, University of Cape Town, Rondebosch 7700, ,South Africa
| | - Refilwe Matshitse
- Institute for Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Saif F Khan
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Faculty of Health Science, Observatory, 7925, South Africa
| | - Tebello Nyokong
- Institute for Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Faculty of Health Science, Observatory, 7925, South Africa
| | - Gregory S Smith
- Department of Chemistry, University of Cape Town, Rondebosch 7700, ,South Africa.
| |
Collapse
|
15
|
Chen D, Shao J, Zhang T, Xu K, Liang C, Cai Y, Guo Y, Chen P, Mou XZ, Dong X. Aromaticity Tuning of Heavy-Atom-Free Photosensitizers for Singlet Fission-Enhanced Immunogenic Photodynamic Oncotherapy. NANO LETTERS 2024. [PMID: 38857313 DOI: 10.1021/acs.nanolett.4c01862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The quantum yield of reactive oxygen species is of central importance for the development of organic photosensitizers and photodynamic therapy (PDT). A common molecular design approach for optimizing organic photosensitizers involves the incorporation of heavy atoms into their backbones. However, this raises concerns regarding heightened dark cytotoxicity and a shortened triplet-state lifetime. Herein, we demonstrate a heavy-atom-free (HAF) photosensitizer design strategy founded on the singlet fission (SF) mechanism for cancer PDT. Through the "single-atom surgery" approach to deleting oxygen atoms in pyrazino[2,3-g]quinoxaline skeleton photosensitizers, photosensitizers PhPQ and TriPhPQ are produced with Huckel's aromaticity and Baird's aromaticity in the ground state and triplet state, respectively, enabling the generation of two triplet excitons through SF. The SF process endows photosensitizer PhPQ with an ultrahigh triplet-state quantum yield (186%) and an outstanding 1O2 quantum yield (177%). Notably, HAF photosensitizers PhPQ and TriPhPQ enhanced PDT efficacy and potentiated αPD-L1 immune check blockade therapy in vivo, which show their promise for translational oncology treatment.
Collapse
Affiliation(s)
- Dapeng Chen
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211816, China
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, Illinois 60637, United States
| | - Jinjun Shao
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211816, China
| | - Tian Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211816, China
| | - Kang Xu
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211816, China
| | - Chen Liang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yu Cai
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yuxin Guo
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, China
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459
| | - Xiao-Zhou Mou
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211816, China
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
16
|
Mandal AA, Upadhyay A, Mandal A, Nayak M, K MS, Mukherjee S, Banerjee S. Visible-Light-Responsive Novel Ru(II)-Metallo-Antibiotics with Potential Antibiofilm and Antibacterial Activity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28118-28133. [PMID: 38783713 DOI: 10.1021/acsami.4c02979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Growing challenges with antibiotic resistance pose immense challenges in combating microbial infections and biofilm prevention on medical devices. Lately, antibacterial photodynamic therapy (aPDT) is now emerging as an alternative therapy to overcome this problem. Herein, we synthesized and characterized four Ru(II)-complexes, viz., [Ru(ph-tpy)(bpy)Cl]PF6 (Ru1), [Ru(ph-tpy)(dpq)Cl]PF6 (Ru2), [Ru(ph-tpy)(dppz)Cl]PF6 (Ru3), and [Ru(ph-tpy)(dppn)Cl]PF6 (Ru4) (where 4'-phenyl-2,2':6',2″-terpyridine = ph-tpy; 2,2'-bipyridine = bpy; dipyrido[3,2-f:2',3'-h]quinoxaline = dpq; dipyrido[3,2-a:2',3'-c]phenazine = dppz; and Benzo[I]dipyrido[3,2-a:2',3'-c]phenazine = dppn), among which Ru2-Ru4 are novel. Octahedral geometry of the complexes with a RuN5Cl core was evident from the crystal structure of Ru2. Ru1-Ru4 showed an MLCT absorption band in the 450-600 nm region, useful for aPDT performances. Further, optimum triplet excited state energy and excellent photostability of Ru1-Ru4 made them good photosensitizers for aPDT. Ru1-Ru4 demonstrated enhanced antimicrobial activity on visible-light exposure (400-700 nm, 10 J cm-2), confirmed using different antibacterial assays. Mechanistic studies revealed that inhibition of bacterial growth was due to the generation of oxidative stress (via NADH oxidation and ROS generation) upon treatment with Ru2-Ru4, resulting in destruction of the bacterial wall. Ru2 performed best killing performance against both Gram-negative (Escherichia coli) and Gram-positive (Bacillus subtilis) bacteria when exposed to light. Ru2-Ru4, when coated on a polydimethylsiloxane (PDMS) disk, showed long-term reusability and durable antibiofilm properties. Molecular docking confirmed the efficient interaction of Ru2-Ru4 with FabH (regulates fatty acid biosynthesis of E. coli) and PgaB (gives structural stability and helps biofilm formation of E. coli), resulting in probable downregulation. In vivo studies with healthy Wistar rats confirmed the biocompatibility of Ru2. This study shows that these lead complexes (Ru2-Ru4) can be used as potent alternative antimicrobial agents in low concentrations toward bacterial eradication with photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Arif Ali Mandal
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Anjali Upadhyay
- School of Biomedical Engineering, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Apurba Mandal
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Malay Nayak
- School of Biomedical Engineering, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Mohammad Sabeel K
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
17
|
Gul A, Ahmad M, Ullah R, Ullah R, Kang Y, Liao W. Systematic review on antibacterial photodynamic therapeutic effects of transition metals ruthenium and iridium complexes. J Inorg Biochem 2024; 255:112523. [PMID: 38489864 DOI: 10.1016/j.jinorgbio.2024.112523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 03/17/2024]
Abstract
The prevalence of antibiotic-resistant pathogenic bacteria poses a significant threat to public health and ranks among the principal causes of morbidity and mortality worldwide. Antimicrobial photodynamic therapy is an emerging therapeutic technique that has excellent potential to embark upon antibiotic resistance problems. The efficacy of this therapy hinges on the careful selection of suitable photosensitizers (PSs). Transition metal complexes, such as Ruthenium (Ru) and Iridium (Ir), are highly suitable for use as PSs because of their surface plasmonic resonance, crystal structure, optical characteristics, and photonics. These metals belong to the platinum family and exhibit similar chemical behavior due to their partially filled d-shells. Ruthenium and Iridium-based complexes generate reactive oxygen species (ROS), which interact with proteins and DNA to induce cell death. As photodynamic therapeutic agents, these complexes have been widely studied for their efficacy against cancer cells, but their potential for antibacterial activity remains largely unexplored. Our study focuses on exploring the antibacterial photodynamic effect of Ruthenium and Iridium-based complexes against both Gram-positive and Gram-negative bacteria. We aim to provide a comprehensive overview of various types of research in this area, including the structures, synthesis methods, and antibacterial photodynamic applications of these complexes. Our findings will provide valuable insights into the design, development, and modification of PSs to enhance their photodynamic therapeutic effect on bacteria, along with a clear understanding of their mechanism of action.
Collapse
Affiliation(s)
- Anadil Gul
- College of Applied Sciences, Shenzhen University, Shenzhen 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China; College of Health Science and Environmental Engineering, Shenzhen Technology University, Pingshan District, Shenzhen 518118, China
| | - Munir Ahmad
- Shenzhen Key Laboratory of Advanced Thin Films and Applications, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Raza Ullah
- College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Rizwan Ullah
- School of Physics, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yan Kang
- College of Applied Sciences, Shenzhen University, Shenzhen 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China; College of Health Science and Environmental Engineering, Shenzhen Technology University, Pingshan District, Shenzhen 518118, China.
| | - Wenchao Liao
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Pingshan District, Shenzhen 518118, China.
| |
Collapse
|
18
|
Noakes F, Smitten KL, Maple LEC, Bernardino de la Serna J, Robertson CC, Pritchard D, Fairbanks SD, Weinstein JA, Smythe CGW, Thomas JA. Phenazine Cations as Anticancer Theranostics †. J Am Chem Soc 2024; 146:12836-12849. [PMID: 38683943 PMCID: PMC11082890 DOI: 10.1021/jacs.4c03491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
The biological properties of two water-soluble organic cations based on polypyridyl structures commonly used as ligands for photoactive transition metal complexes designed to interact with biomolecules are investigated. A cytotoxicity screen employing a small panel of cell lines reveals that both cations show cytotoxicity toward cancer cells but show reduced cytotoxicity to noncancerous HEK293 cells with the more extended system being notably more active. Although it is not a singlet oxygen sensitizer, the more active cation also displayed enhanced potency on irradiation with visible light, making it active at nanomolar concentrations. Using the intrinsic luminescence of the cations, their cellular uptake was investigated in more detail, revealing that the active compound is more readily internalized than its less lipophilic analogue. Colocalization studies with established cell probes reveal that the active cation predominantly localizes within lysosomes and that irradiation leads to the disruption of mitochondrial structure and function. Stimulated emission depletion (STED) nanoscopy and transmission electron microscopy (TEM) imaging reveal that treatment results in distinct lysosomal swelling and extensive cellular vacuolization. Further imaging-based studies confirm that treatment with the active cation induces lysosomal membrane permeabilization, which triggers lysosome-dependent cell-death due to both necrosis and caspase-dependent apoptosis. A preliminary toxicity screen in the Galleria melonella animal model was carried out on both cations and revealed no detectable toxicity up to concentrations of 80 mg/kg. Taken together, these studies indicate that this class of synthetically easy-to-access photoactive compounds offers potential as novel therapeutic leads.
Collapse
Affiliation(s)
- Felicity
F. Noakes
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
- Department
of Biomedical Science, The University of
Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Kirsty L. Smitten
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
- Department
of Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Laura E. C. Maple
- Department
of Biomedical Science, The University of
Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Jorge Bernardino de la Serna
- National
Heart and Lung Institute, Imperial College
London, London SW7 2AZ, U.K.
- Central
Laser
Facility, Rutherford Appleton Laboratory, Research Complex at Harwell, Science and Technology Facilities Council, Harwell-Oxford, Didcot OX11 0QX, U.K.
| | - Craig C. Robertson
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Dylan Pritchard
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Simon D. Fairbanks
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Julia A. Weinstein
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Carl G. W. Smythe
- Department
of Biomedical Science, The University of
Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Jim A. Thomas
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| |
Collapse
|
19
|
Robinette FN, Valentine NP, Sehler KM, Medeck AM, Reynolds KE, Lane SN, Price AN, Cavanaugh IG, Shell SM, Ashford DL. Modulating Excited State Properties and Ligand Ejection Kinetics in Ruthenium Polypyridyl Complexes Designed to Mimic Photochemotherapeutics. Inorg Chem 2024; 63:8426-8439. [PMID: 38662617 DOI: 10.1021/acs.inorgchem.4c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Ruthenium(II) polypyridyl complexes have gained significant interest as photochemotherapeutics (PCTs) due to their synthetic viability, strong light absorption, well understood excited state properties, and high phototoxicity indexes. Herein, we report the synthesis, characterization, electrochemical, spectrochemical, and preliminary cytotoxicity analyses of three series of ruthenium(II) polypyridyl complexes designed to mimic PCTs. The three series have the general structure of [Ru(bpy)2(N-N)]2+ (Series 1), [Ru(bpy)(dmb)(N-N)]2+ (Series 2), and [Ru(dmb)2(N-N)]2+ (Series 3, where N-N is a bidentate polypyridyl ligand, bpy = 2,2'-bipyridine, and dmb = 6,6'-dimethyl-2,2'-bipyridine). In the three series, the N-N ligand was systematically modified to incorporate increased conjugation and/or electronegative heteroatoms to increase dπ-π* backbonding, red-shifting the lowest energy metal-to-ligand charge transfer (MLCT) absorptions from λmax = 454 to λmax = 580 nm, nearing the therapeutic window for PCTs (600-1100 nm). In addition, steric bulk was systematically introduced through the series, distorting the Ru(II) octahedra, making the dissociative 3dd* state thermally accessible at room and body temperatures. This resulted in a 4 orders of magnitude increase in photoinduced ligand ejection kinetics, and demonstrates the ability to modulate both the MLCT* and dd* manifolds in the complexes, which is critical in PCT drug design. Preliminary cell viability assays suggest that the increased steric bulk to lower the 3dd* states may interfere with the cytotoxicity mechanism, limiting photoinitiated toxicity of the complexes. This work demonstrates the importance of understanding both the MLCT* and dd* manifolds and how they impact the ability of a complex to act as a PCT agent.
Collapse
Affiliation(s)
- Faith N Robinette
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Nathaniel P Valentine
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Konrad M Sehler
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Andrew M Medeck
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Keylon E Reynolds
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Skylar N Lane
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Averie N Price
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Ireland G Cavanaugh
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Steven M Shell
- Department of Natural Sciences, University of Virginia College at Wise, Wise, Virginia 24293, United States
| | - Dennis L Ashford
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| |
Collapse
|
20
|
Mandal AA, Singh V, Saha S, Peters S, Sadhukhan T, Kushwaha R, Yadav AK, Mandal A, Upadhyay A, Bera A, Dutta A, Koch B, Banerjee S. Green Light-Triggered Photocatalytic Anticancer Activity of Terpyridine-Based Ru(II) Photocatalysts. Inorg Chem 2024; 63:7493-7503. [PMID: 38578920 DOI: 10.1021/acs.inorgchem.4c00650] [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: 04/07/2024]
Abstract
The relentless increase in drug resistance of platinum-based chemotherapeutics has opened the scope for other new cancer therapies with novel mechanisms of action (MoA). Recently, photocatalytic cancer therapy, an intrusive catalytic treatment, is receiving significant interest due to its multitargeting cell death mechanism with high selectivity. Here, we report the synthesis and characterization of three photoresponsive Ru(II) complexes, viz., [Ru(ph-tpy)(bpy)Cl]PF6 (Ru1), [Ru(ph-tpy)(phen)Cl]PF6 (Ru2), and [Ru(ph-tpy)(aip)Cl]PF6 (Ru3), where, ph-tpy = 4'-phenyl-2,2':6',2″-terpyridine, bpy = 2,2'-bipyridine, phen = 1,10-phenanthroline, and aip = 2-(anthracen-9-yl)-1H-imidazo[4,5-f][1,10] phenanthroline, showing photocatalytic anticancer activity. The X-ray crystal structures of Ru1 and Ru2 revealed a distorted octahedral geometry with a RuN5Cl core. The complexes showed an intense absorption band in the 440-600 nm range corresponding to the metal-to-ligand charge transfer (MLCT) that was further used to achieve the green light-induced photocatalytic anticancer effect. The mitochondria-targeting photostable complex Ru3 induced phototoxicity with IC50 and PI values of ca. 0.7 μM and 88, respectively, under white light irradiation and ca. 1.9 μM and 35 under green light irradiation against HeLa cells. The complexes (Ru1-Ru3) showed negligible dark cytotoxicity toward normal splenocytes (IC50s > 50 μM). The cell death mechanistic study revealed that Ru3 induced ROS-mediated apoptosis in HeLa cells via mitochondrial depolarization under white or green light exposure. Interestingly, Ru3 also acted as a highly potent catalyst for NADH photo-oxidation under green light. This NADH photo-oxidation process also contributed to the photocytotoxicity of the complexes. Overall, Ru3 presented multitargeting synergistic type I and type II photochemotherapeutic effects.
Collapse
Affiliation(s)
- Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Sukanta Saha
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Silda Peters
- Departmentof Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Tumpa Sadhukhan
- Departmentof Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Apurba Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Arnab Dutta
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
21
|
Naganawa R, Zhao H, Takano Y, Maeki M, Tokeshi M, Harashima H, Yamada Y. Investigation of the Nanoparticulation Method and Cell-Killing Effect following the Mitochondrial Delivery of Hydrophobic Porphyrin-Based Photosensitizers. Int J Mol Sci 2024; 25:4294. [PMID: 38673875 PMCID: PMC11050504 DOI: 10.3390/ijms25084294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Photodynamic therapy is expected to be a less invasive treatment, and strategies for targeting mitochondria, the main sources of singlet oxygen, are attracting attention to increase the efficacy of photodynamic therapy and reduce its side effects. To date, we have succeeded in encapsulating the photosensitizer rTPA into MITO-Porter (MP), a mitochondria-targeted Drug Delivery System (DDS), aimed at mitochondrial delivery of the photosensitizer while maintaining its activity. In this study, we report the results of our studies to alleviate rTPA aggregation in an effort to improve drug efficacy and assess the usefulness of modifying the rTPA side chain to improve the mitochondrial retention of MITO-Porter, which exhibits high therapeutic efficacy. Conventional rTPA with anionic side chains and two rTPA analogs with side chains that were converted to neutral or cationic side chains were encapsulated into MITO-Porter. Low-MP (MITO-Porter with Low Drug/Lipid) exhibited high drug efficacy for all three types of rTPA, and in Low-MP, charged rTPA-encapsulated MP exhibited high drug efficacy. The cellular uptake and mitochondrial translocation capacities were similar for all particles, suggesting that differences in aggregation rates during the incorporation of rTPA into MITO-Porter resulted in differences in drug efficacy.
Collapse
Grants
- 23H00541 Ministry of Education, Culture, Sports, Science and Technology
- 21H01753 Ministry of Education, Culture, Sports, Science and Technology
- 21K19928 Ministry of Education, Culture, Sports, Science and Technology
- Special Education and Research Expenses Ministry of Education, Culture, Sports, Science and Technology
- JPMJFR203X Japan Science and Technology Agency
Collapse
Affiliation(s)
- Rina Naganawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hanjun Zhao
- Graduate School of Environmental Science, Hokkaido University, Sapporo 060-0810, Japan (Y.T.)
| | - Yuta Takano
- Graduate School of Environmental Science, Hokkaido University, Sapporo 060-0810, Japan (Y.T.)
- Research Institute for Electronic Science, Hokkaido University, Sapporo 010-0020, Japan
| | - Masatoshi Maeki
- Graduate School of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Manabu Tokeshi
- Graduate School of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Fusion Oriented Research for Disruptive Science and Technology (FOREST) Program, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| |
Collapse
|
22
|
Ballester F, Hernández-García A, Santana MD, Bautista D, Ashoo P, Ortega-Forte E, Barone G, Ruiz J. Photoactivatable Ruthenium Complexes Containing Minimal Straining Benzothiazolyl-1,2,3-triazole Chelators for Cancer Treatment. Inorg Chem 2024; 63:6202-6216. [PMID: 38385171 PMCID: PMC11005040 DOI: 10.1021/acs.inorgchem.3c04432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/19/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
Ruthenium(II) complexes containing diimine ligands have contributed to the development of agents for photoactivated chemotherapy. Several approaches have been used to obtain photolabile Ru(II) complexes. The two most explored have been the use of monodentate ligands and the incorporation of steric effects between the bidentate ligands and the Ru(II). However, the introduction of electronic effects in the ligands has been less explored. Herein, we report a systematic experimental, theoretical, and photocytotoxicity study of a novel series of Ru(II) complexes Ru1-Ru5 of general formula [Ru(phen)2(N∧N')]2+, where N∧N' are different minimal strained ligands based on the 1-aryl-4-benzothiazolyl-1,2,3-triazole (BTAT) scaffold, being CH3 (Ru1), F (Ru2), CF3 (Ru3), NO2 (Ru4), and N(CH3)2 (Ru5) substituents in the R4 of the phenyl ring. The complexes are stable in solution in the dark, but upon irradiation in water with blue light (λex = 465 nm, 4 mW/cm2) photoejection of the ligand BTAT was observed by HPLC-MS spectrometry and UV-vis spectroscopy, with t1/2 ranging from 4.5 to 14.15 min depending of the electronic properties of the corresponding BTAT, being Ru4 the less photolabile (the one containing the more electron withdrawing substituent, NO2). The properties of the ground state singlet and excited state triplet of Ru1-Ru5 have been explored using density functional theory (DFT) and time-dependent DFT (TD-DFT) calculations. A mechanism for the photoejection of the BTAT ligand from the Ru complexes, in H2O, is proposed. Phototoxicity studies in A375 and HeLa human cancer cell lines showed that the new Ru BTAT complexes were strongly phototoxic. An enhancement of the emission intensity of HeLa cells treated with Ru5 was observed in response to increasing doses of light due to the photoejection of the BTAT ligand. These studies suggest that BTAT could serve as a photocleavable protecting group for the cytotoxic bis-aqua ruthenium warhead [Ru(phen)2(OH2)2]2+.
Collapse
Affiliation(s)
- Francisco
J. Ballester
- Departamento
de Química Inorgánica, Universidad
de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Alba Hernández-García
- Departamento
de Química Inorgánica, Universidad
de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - M. Dolores Santana
- Departamento
de Química Inorgánica, Universidad
de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | | | - Pezhman Ashoo
- Departamento
de Química Inorgánica, Universidad
de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Enrique Ortega-Forte
- Departamento
de Química Inorgánica, Universidad
de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| | - Giampaolo Barone
- Dipartimento
di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (SteBiCeF), Università degli Studi di Palermo, I-90128 Palermo, Italy
| | - José Ruiz
- Departamento
de Química Inorgánica, Universidad
de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca), E-30100 Murcia, Spain
| |
Collapse
|
23
|
Zhang H, Cui M, Tang D, Wang B, Liang G, Xu C, Xiao H. Localization of Cancer Cells for Subsequent Robust Photodynamic Therapy by ROS Responsive Polymeric Nanoparticles With Anti-Metastasis Complexes NAMI-A. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310298. [PMID: 38145801 DOI: 10.1002/adma.202310298] [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: 10/05/2023] [Revised: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Photodynamic therapy (PDT), as a new type of light-mediated reactive oxygen species (ROS) cancer therapy, has the advantages of high therapeutic efficiency, non-resistance, and less trauma than traditional cancer therapy such as surgery, radiotherapy, and chemotherapy. However, oxygen-dependent PDT further exacerbates tumor metastasis. To this end, a strategy that circumvents tumor metastasis to improve the therapeutic efficacy of PDT is proposed. Herein, a near-infrared light-activated photosensitive polymer is synthesized and branched the anti-metastatic ruthenium complex NAMI-A on the side, which is further assembled to form nanoparticles (NP2) for breast cancer therapy. NP2 can kill tumor cells by generating ROS under 808 nm radiation (NP2 + L), reduce the expression of matrix metalloproteinases (MMP2/9) in cancer cells, decrease the invasive and migration capacity of cancer cells, and eliminate cancer cells. Further animal experiments show that NP2 + L can inhibit tumor growth and reduce liver and lung metastases. In addition, NP2 + L can activate the immune system in mice to avoid tumor recurrence. In conclusion, a PDT capable of both preventing tumor metastasis and precisely hitting the primary tumor to achieve effective treatment of highly metastatic cancers is developed.
Collapse
Affiliation(s)
- Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Minhui Cui
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bin Wang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
24
|
Tai Y, Chen Z, Luo T, Luo B, Deng C, Lu Z, Wen S, Wang J. MOF@COF Nanocapsules Enhance Soft Tissue Sarcoma Treatment: Synergistic Effects of Photodynamic Therapy and PARP Inhibition on Tumor Growth Suppression and Immune Response Activation. Adv Healthc Mater 2024; 13:e2303911. [PMID: 38215731 DOI: 10.1002/adhm.202303911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/07/2024] [Indexed: 01/14/2024]
Abstract
Soft tissue sarcomas (STS) are highly malignant tumors with limited treatment options owing to their heterogeneity and resistance to conventional therapies. Photodynamic therapy (PDT) and poly-ADP-ribose polymerase (PARP) inhibitors (PARPi) have shown potential for STS treatment, with PDT being effective for sarcomas located on the extremities and body surface and PARPi targeting defects in homologous recombination repair. To address the limitations of PDT and harness the potential of PARPi, herein, a novel therapeutic approach for STS treatment combining nanocapsules bearing integrated metal-organic frameworks (MOFs) and covalent organic frameworks (COFs), i.e., MOF@COF, with PDT and PARPi is proposed. Nanocapsules are designed, referred to as ZTN@COF@poloxamer, which contain a Zr-based MOF and tetrakis (4-carbethoxyphenyl) porphyrin as a photosensitizer, are coated with a COF to improve the sensitizing properties, and are loaded with niraparib to inhibit DNA repair. Experiments demonstrate that this new nanocapsules treatment significantly inhibits STS growth, promotes tumor cell apoptosis, exhibits high antitumor activity with minimal side effects, activates the immune response of the tumor, and inhibits lung metastasis in vivo. Therefore, MOF@COF nanocapsules combined with PARPi offer a promising approach for STS treatment, with the potential to enhance the efficacy of PDT and prevent tumor recurrence.
Collapse
Affiliation(s)
- Yi Tai
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Zhihao Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Tianqi Luo
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Bingling Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Chuangzhong Deng
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Zhenhai Lu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Jin Wang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| |
Collapse
|
25
|
Prieto Otoya TD, McQuaid KT, Hennessy J, Menounou G, Gibney A, Paterson NG, Cardin DJ, Kellett A, Cardin CJ. Probing a Major DNA Weakness: Resolving the Groove and Sequence Selectivity of the Diimine Complex Λ-[Ru(phen) 2 phi] 2. Angew Chem Int Ed Engl 2024; 63:e202318863. [PMID: 38271265 DOI: 10.1002/anie.202318863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
The grooves of DNA provide recognition sites for many nucleic acid binding proteins and anticancer drugs such as the covalently binding cisplatin. Here we report a crystal structure showing, for the first time, groove selectivity by an intercalating ruthenium complex. The complex Λ-[Ru(phen)2 phi]2+ , where phi=9,10-phenanthrenediimine, is bound to the DNA decamer duplex d(CCGGTACCGG)2 . The structure shows that the metal complex is symmetrically bound in the major groove at the central TA/TA step, and asymmetrically bound in the minor groove at the adjacent GG/CC steps. A third type of binding links the strands, in which each terminal cytosine base stacks with one phen ligand. The overall binding stoichiometry is four Ru complexes per duplex. Complementary biophysical measurements confirm the binding preference for the Λ-enantiomer and show a high affinity for TA/TA steps and, more generally, TA-rich sequences. A striking enantiospecific elevation of melting temperatures is found for oligonucleotides which include the TATA box sequence.
Collapse
Affiliation(s)
| | - Kane T McQuaid
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6AD, UK
| | - Joseph Hennessy
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin, 9, Ireland Email
| | - Georgia Menounou
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin, 9, Ireland Email
| | - Alex Gibney
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin, 9, Ireland Email
| | - Neil G Paterson
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, UK
| | - David J Cardin
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6AD, UK
| | - Andrew Kellett
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin, 9, Ireland Email
| | - Christine J Cardin
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6AD, UK
| |
Collapse
|
26
|
Li Z, Zhang Z, Ma L, Wen H, Kang M, Li D, Zhang W, Luo S, Wang W, Zhang M, Wang D, Li H, Li X, Wang H. Combining Multiple Photosensitizer Modules into One Supramolecular System for Synergetic Enhanced Photodynamic Therapy. Angew Chem Int Ed Engl 2024; 63:e202400049. [PMID: 38193338 DOI: 10.1002/anie.202400049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/10/2024]
Abstract
Photodynamic therapy (PDT), as an emerging cancer treatment, requires the development of highly desirable photosensitizers (PSs) with integrated functional groups to achieve enhanced therapeutic efficacy. Coordination-driven self-assembly (CDSA) would provide an alternative approach for combining multiple PSs synergistically. Here, we demonstrate a simple yet powerful strategy of combining conventional chromophores (tetraphenylethylene, porphyrin, or Zn-porphyrin) with pyridinium salt PSs together through condensation reactions, followed by CDSA to construct a series of novel metallo-supramolecular PSs (S1-S3). The generation of reactive oxygen species (ROS) is dramatically enhanced by the direct combination of two different PSs, and further reinforced in the subsequent ensembles. Among all the ensembles, S2 with two porphyrin cores shows the highest ROS generation efficiency, specific interactions with lysosome, and strong emission for probing cells. Moreover, the cellular and living experiments confirm that S2 has excellent PDT efficacy, biocompatibility, and biosafety. As such, this study will enable the development of more efficient PSs with potential clinical applications.
Collapse
Affiliation(s)
- Zhikai Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Zhijun Zhang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Lingzhi Ma
- State Key Laboratory for Mechanical Behavior of Materials, Shaanxi International Research Center for Soft Matter, School of Materials Science and Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Haifei Wen
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Miaomiao Kang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Danxia Li
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Wenjing Zhang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Siqi Luo
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Weiguo Wang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, 116023, China
| | - Mingming Zhang
- State Key Laboratory for Mechanical Behavior of Materials, Shaanxi International Research Center for Soft Matter, School of Materials Science and Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Haiyang Li
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, 116023, China
| | - Xiaopeng Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, Guangdong, 518055, China
| | - Heng Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
27
|
Peng Y, Da X, Zhou W, Xu Y, Liu X, Wang X, Zhou Q. A photo-degradable BODIPY-modified Ru(II) photosensitizer for safe and efficient PDT under both normoxic and hypoxic conditions. Dalton Trans 2024; 53:3579-3588. [PMID: 38314620 DOI: 10.1039/d3dt04063a] [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: 02/06/2024]
Abstract
Photodynamic therapy (PDT) is promising for cancer treatment but still suffers from some limitations. For instance, PDT based on 1O2 generation (in a type-II mechanism) is heavily dependent on high oxygen concentrations and will be significantly depressed in hypoxic tumors. In addition, the residual photosensitizers after PDT treatment may cause severe side-effects under light irradiation. To solve these problems, herein a BODIPY (boron dipyrromethene)-modified Ru(II) complex [Ru(dip)2(tpy-BODIPY)]2+ (complex 1, dip = 4,7-diphenyl-1,10-phenanthroline, tpy = 2,2':6',2''-terpyridine) was designed and synthesized. Complex 1 exhibited both high singlet oxygen quantum yield (Φ = 0.7 in CH3CN) and excellent superoxide radical (O2˙-) generation, and thus demonstrated efficient PDT activity under both normoxic and hypoxic conditions. Moreover, complex 1 is photo-degradable in water, and greatly loses its ROS generation ability after PDT treatment. These novel properties of complex 1 make it promising for efficient PDT under both normoxic and hypoxic conditions with reduced side-effects.
Collapse
Affiliation(s)
- Yatong Peng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xuwen Da
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Wanpeng Zhou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yunli Xu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiulian Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xuesong Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Qianxiong Zhou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| |
Collapse
|
28
|
Wu R, Yuen J, Cheung E, Huang Z, Chu E. Review of three-dimensional spheroid culture models of gynecological cancers for photodynamic therapy research. Photodiagnosis Photodyn Ther 2024; 45:103975. [PMID: 38237651 DOI: 10.1016/j.pdpdt.2024.103975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Photodynamic therapy (PDT) is a specific cancer treatment with minimal side effects. However, it remains challenging to apply PDT clinically, partially due to the difficulty of translating research findings to clinical settings as the conventional 2D cell models used for in vitro research are accepted as less physiologically relevant to a solid tumour. 3D spheroids offer a better model for testing PDT mechanisms and efficacy, particularly on photosensitizer uptake, cellular and subcellular distribution and interaction with cellular oxygen consumption. 3D spheroids are usually generated by scaffold-free and scaffold-based methods and are accepted as physiologically relevant models for PDT anticancer research. Scaffold-free methods offer researchers advantages including high efficiency, reproducible, and controlled microenvironment. While the scaffold-based methods offer an extracellular matrix-like 3D scaffold with the necessary architecture and chemical mediators to support the spheroid formation, the natural scaffold used may limit its usage because of low reproducibility due to patch-to-patch variation. Many studies show that the 3D spheroids do offer advantages to gynceologcial cancer PDT investigation. This article will provide a review of the applications of 3D spheroid culture models for the PDT research of gynaecological cancers.
Collapse
Affiliation(s)
- Rwk Wu
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, Scotland, UK.
| | - Jwm Yuen
- School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region of China
| | - Eyw Cheung
- School of Medical and Health Sciences, Tung Wah College, Hong Kong Special Administrative Region of China
| | - Z Huang
- MOE Key Laboratory of Photonics Science and Technology for Medicine, Fujian Normal University, Fuzhou, China
| | - Esm Chu
- School of Medical and Health Sciences, Tung Wah College, Hong Kong Special Administrative Region of China.
| |
Collapse
|
29
|
Kasparkova J, Hernández-García A, Kostrhunova H, Goicuría M, Novohradsky V, Bautista D, Markova L, Santana MD, Brabec V, Ruiz J. Novel 2-(5-Arylthiophen-2-yl)-benzoazole Cyclometalated Iridium(III) dppz Complexes Exhibit Selective Phototoxicity in Cancer Cells by Lysosomal Damage and Oncosis. J Med Chem 2024; 67:691-708. [PMID: 38141031 PMCID: PMC10788912 DOI: 10.1021/acs.jmedchem.3c01978] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
A second-generation series of biscyclometalated 2-(5-aryl-thienyl)-benzimidazole and -benzothiazole Ir(III) dppz complexes [Ir(C^N)2(dppz)]+, Ir1-Ir4, were rationally designed and synthesized, where the aryl group attached to the thienyl ring was p-CF3C6H4 or p-Me2NC6H4. These new Ir(III) complexes were assessed as photosensitizers to explore the structure-activity correlations for their potential use in biocompatible anticancer photodynamic therapy. When irradiated with blue light, the complexes exhibited high selective potency across several cancer cell lines predisposed to photodynamic therapy; the benzothiazole derivatives (Ir1 and Ir2) were the best performers, Ir2 being also activatable with green or red light. Notably, when irradiated, the complexes induced leakage of lysosomal content into the cytoplasm of HeLa cancer cells and induced oncosis-like cell death. The capability of the new Ir complexes to photoinduce cell death in 3D HeLa spheroids has also been demonstrated. The investigated Ir complexes can also catalytically photo-oxidate NADH and photogenerate 1O2 and/or •OH in cell-free media.
Collapse
Affiliation(s)
- Jana Kasparkova
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Alba Hernández-García
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| | - Hana Kostrhunova
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Marta Goicuría
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| | - Vojtěch Novohradsky
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | | | - Lenka Markova
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - María Dolores Santana
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| | - Viktor Brabec
- Czech
Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - José Ruiz
- Departamento
de Química Inorgánica, Universidad de Murcia, and Biomedical
Research Institute of Murcia (IMIB-Arrixaca), Murcia E-30100, Spain
| |
Collapse
|
30
|
Huang R, Huang CH, Chen J, Yan ZY, Tang M, Shao J, Cai K, Zhu BZ. Unprecedented enantio-selective live-cell mitochondrial DNA super-resolution imaging and photo-sensitizing by the chiral ruthenium polypyridyl DNA "light-switch". Nucleic Acids Res 2023; 51:11981-11998. [PMID: 37933856 PMCID: PMC10711558 DOI: 10.1093/nar/gkad799] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 07/25/2023] [Accepted: 09/19/2023] [Indexed: 11/08/2023] Open
Abstract
Mitochondrial DNA (mtDNA) is known to play a critical role in cellular functions. However, the fluorescent probe enantio-selectively targeting live-cell mtDNA is rare. We recently found that the well-known DNA 'light-switch' [Ru(phen)2dppz]Cl2 can image nuclear DNA in live-cells with chlorophenolic counter-anions via forming lipophilic ion-pairing complex. Interestingly, after washing with fresh-medium, [Ru(phen)2dppz]Cl2 was found to re-localize from nucleus to mitochondria via ABC transporter proteins. Intriguingly, the two enantiomers of [Ru(phen)2dppz]Cl2 were found to bind enantio-selectively with mtDNA in live-cells not only by super-resolution optical microscopy techniques (SIM, STED), but also by biochemical methods (mitochondrial membrane staining with Tomo20-dronpa). Using [Ru(phen)2dppz]Cl2 as the new mtDNA probe, we further found that each mitochondrion containing 1-8 mtDNA molecules are distributed throughout the entire mitochondrial matrix, and there are more nucleoids near nucleus. More interestingly, we found enantio-selective apoptotic cell death was induced by the two enantiomers by prolonged visible light irradiation, and in-situ self-monitoring apoptosis process can be achieved by using the unique 'photo-triggered nuclear translocation' property of the Ru complex. This is the first report on enantio-selective targeting and super-resolution imaging of live-cell mtDNA by a chiral Ru complex via formation and dissociation of ion-pairing complex with suitable counter-anions.
Collapse
Affiliation(s)
- Rong Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Chun-Hua Huang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jing Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Zhu-Ying Yan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Miao Tang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jie Shao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ben-Zhan Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Environmental Science and Technology, Shandong University, Qingdao, Shandong 266237, PR China
| |
Collapse
|
31
|
Bai Y, Aodeng G, Ga L, Hai W, Ai J. Research Progress of Metal Anticancer Drugs. Pharmaceutics 2023; 15:2750. [PMID: 38140091 PMCID: PMC10747151 DOI: 10.3390/pharmaceutics15122750] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer treatments, including traditional chemotherapy, have failed to cure human malignancies. The main reasons for the failure of these treatments are the inevitable drug resistance and serious side effects. In clinical treatment, only 5 percent of the 50 percent of cancer patients who are able to receive conventional chemotherapy survive. Because of these factors, being able to develop a drug and treatment that can target only cancer cells without affecting normal cells remains a big challenge. Since the special properties of cisplatin in the treatment of malignant tumors were accidentally discovered in the last century, metal anticancer drugs have become a research hotspot. Metal anticancer drugs have unique pharmaceutical properties, such as ruthenium metal drugs with their high selectivity, low toxicity, easy absorption by tumor tissue, excretion, and so on. In recent years, efficient and low-toxicity metal antitumor complexes have been synthesized. In this paper, the scientific literature on platinum (Pt), ruthenium (Ru), iridium (Ir), gold (Au), and other anticancer complexes was reviewed by referring to a large amount of relevant literature at home and abroad.
Collapse
Affiliation(s)
- Yun Bai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Gerile Aodeng
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Lu Ga
- College of Pharmacy, Inner Mongolia Medical University, Jinchuankaifaqu, Hohhot 010110, China;
| | - Wenfeng Hai
- Inner Mongolia Key Laboratory of Carbon Nanomaterials, Nano Innovation Institute (NII), College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Jun Ai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| |
Collapse
|
32
|
Sarkar T, Sahoo S, Neekhra S, Paul M, Biswas S, Babu BN, Srivastava R, Hussain A. A dipyridophenazine Ni(II) dithiolene complex as a dual-acting cancer phototherapy agent activatable within the phototherapeutic window. Eur J Med Chem 2023; 261:115816. [PMID: 37717381 DOI: 10.1016/j.ejmech.2023.115816] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/03/2023] [Accepted: 09/10/2023] [Indexed: 09/19/2023]
Abstract
A combination of photodynamic therapy (PDT) and photothermal therapy (PTT) within the phototherapeutic window (600-900 nm) can lead to significantly enhanced therapeutic outcomes, surpassing the efficacy observed with PDT or PTT alone in cancer phototherapy. Herein, we report a novel small-molecule mixed-ligand Ni(II)-dithiolene complex (Ni-TDD) with a dipyridophenazine ligand, demonstrating potent red-light PDT and significant near-infrared (NIR) light mild-temperature PTT activity against cancer cells and 3D multicellular tumour spheroids (MCTSs). The four-coordinate square planar complex exhibited a moderately intense absorption band (ε ∼ 3700 M-1cm-1) centered around 900 nm and demonstrated excellent dark and photostability in an aqueous phase. Ni-TDD induced a potent red-light (600-720 nm) PDT effect on HeLa cancer cells (IC50 = 1.8 μM, photo irritation factor = 44), triggering apoptotic cell death through efficient singlet oxygen generation. Ni-TDD showed a significant intercalative binding affinity towards double-helical calf thymus DNA, resulting in a binding constant (Kb) ∼ 106 M-1. The complex induced mild hyperthermia and exerted a significant mild-temperature PTT effect on MDA-MB-231 cancer cells upon irradiation with 808 nm NIR light. Simultaneous irradiation of Ni-TDD-treated HeLa MCTSs with red and NIR light led to a remarkable synergistic inhibition of growth, exceeding the effects of individual irradiation, through the generation of singlet oxygen and mild hyperthermia. Ni-TDD displayed minimal toxicity towards non-cancerous HPL1D and L929 cells, even at high micromolar concentrations. This is the first report of a Ni(II) complex demonstrating red-light PDT activity and the first example of a first-row transition metal complex exhibiting combined PDT and PTT effects within the clinically relevant phototherapeutic window. Our findings pave the way for designing and developing metal-dithiolene complexes as dual-acting cancer phototherapy agents using long wavelength light for treating solid tumors.
Collapse
Affiliation(s)
- Tukki Sarkar
- Department of Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Suditi Neekhra
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India
| | - Milan Paul
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, 500078, Telangana, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, 500078, Telangana, India.
| | - Bathini Nagendra Babu
- Department of Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India.
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India.
| | - Akhtar Hussain
- Department of Chemistry, Handique Girls' College, Guwahati, 781001, Assam, India.
| |
Collapse
|
33
|
Puttaswamy NY, Mahanta P, Sarma P, Medhi C, Kaid SMA, Kullaiah B, Basumatary D, Manjasetty BA. Structure-based biological investigations on ruthenium complexes containing 2,2'-bipyridine ligands and their applications in photodynamic therapy as a potential photosensitizer. Chem Biol Drug Des 2023; 102:1506-1520. [PMID: 37722881 DOI: 10.1111/cbdd.14341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 09/20/2023]
Abstract
Ruthenium complexes have been investigated for various biological applications by virtue of their radical scavenging, DNA binding, receptor binding, and cytotoxic abilities; especially the possible potential application of these complexes in photodynamic therapy (PDT). This study focuses on the synthesis, structural characterization and biological application (pertaining to its cytotoxicity and radical generation) of ruthenium complexed with salicylaldehyde fumaryl-dihydrazone (slfhH4 ), salicylaldehyde glutaryl-di-hydrazone (slfgH4 ) and 2,2'-bipyridine (bpy). During the synthesis, the anticipated complex was precipitated out but as serendipity, Ruthenium(II) tris (2,2'-bipyridyl) monochloride nonahydrate {[Ru(bpy)3 ]2+ .Cl.9H2 O} (RBMN) and Ruthenium(II) tris (2,2'-bipyridyl) monochloride septahydrate {[Ru(bpy)3 ]2+ .Cl.7H2 O}(RBMS) were crystallized from the filtrate. The crystal structure of complexes RBMN and RBMS were determined by a single-crystal X-ray diffraction methods and it showed that chlorine anion lies at the crystallographic axis and forms a halogen hydrogen-bonded organic framework (XHOF) to provide the stability. In comparison with similar structures in Cambridge Crystallographic Data Center (CCDC) revealed that the nature of the XHOF framework and the layered packing are conserved. The compounds showed excellent cytotoxic ability (against L6 cells) and the nitro blue tetrazolium (NBT) assay upon irradiation to light revealed its ability to produce reactive oxygen species (ROS). The presence of partially occupied water molecules in the layered organization within the crystal packing mimics the release of ROS resulting in cytotoxicity. The structural results together with the biological data make these complexes interesting candidates for potential photosensitizers for PDT applications.
Collapse
Affiliation(s)
- Naveen Y Puttaswamy
- Department of Studies and Research in Physics, Department of Biochemistry, Adichunchanagiri School of Natural Sciences, Centre for Research and Innovation, Adichunchanagiri University, Karnataka, BG Nagara, India
| | - Pranami Mahanta
- Department of Chemistry, Department of Applied Sciences, Gauhati University, Guwahati, Assam, India
| | - Pranjit Sarma
- Department of Chemistry, Department of Applied Sciences, Gauhati University, Guwahati, Assam, India
| | - Chitrani Medhi
- Department of Chemistry, Department of Applied Sciences, Gauhati University, Guwahati, Assam, India
| | - Sanaa Mohammed Abdu Kaid
- Department of Studies and Research in Physics, Department of Biochemistry, Adichunchanagiri School of Natural Sciences, Centre for Research and Innovation, Adichunchanagiri University, Karnataka, BG Nagara, India
| | - Byrappa Kullaiah
- Department of Studies and Research in Physics, Department of Biochemistry, Adichunchanagiri School of Natural Sciences, Centre for Research and Innovation, Adichunchanagiri University, Karnataka, BG Nagara, India
| | - Debajani Basumatary
- Department of Chemistry, Department of Applied Sciences, Gauhati University, Guwahati, Assam, India
| | - Babu A Manjasetty
- Department of Studies and Research in Physics, Department of Biochemistry, Adichunchanagiri School of Natural Sciences, Centre for Research and Innovation, Adichunchanagiri University, Karnataka, BG Nagara, India
| |
Collapse
|
34
|
Mishra R, Saha A, Chatterjee P, Bhattacharyya A, Patra AK. Ruthenium(II) Polypyridyl-Based Photocages for an Anticancer Phytochemical Diallyl Sulfide: Comparative Dark and Photoreactivity Studies of Caged and Precursor Uncaged Complexes. Inorg Chem 2023; 62:18839-18855. [PMID: 37930798 DOI: 10.1021/acs.inorgchem.3c02038] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The spatiotemporal control over the drug's action offered by ruthenium(II) polypyridyl complexes by the selective activation of the prodrug inside the tumor has beaconed toward much-desired selectivity issues in cancer chemotherapy. The photocaging of anticancer bioactive ligands attached synergistically with cytotoxic Ru(II) polypyridyl cores and selective release thereof in cancer cells are a promising modality for more effective drug action. Diallyl sulfide (DAS) naturally found in garlic has anticancer, antioxidant, and anti-inflammatory activities. Herein, we designed two Ru(II) polypyridyl complexes to cage DAS having a thioether-based donor site. For in-depth photocaging studies, we compared the reactivity of the DAS-caged compounds with the uncaged Ru(II)-complexes with the general formula [Ru(ttp)(NN)(L)]+/2+. Here, in the first series, ttp = p-tolyl terpyridine, NN = phen (1,10-phenanthroline), and L = Cl- (1-Cl) and H2O (1-H2O), while for the second series, NN = dpq (pyrazino[2,3-f][1,10]phenanthroline), and L = Cl- (2-Cl) and H2O (2-H2O). The reaction of DAS with 1-H2O and 2-H2O yielded the caged complexes [Ru(ttp)(NN)(DAS)](PF6)2, i.e., 1-DAS and 2-DAS, respectively. The complexes were structurally characterized by X-ray crystallography, and the solution-state characterization was done by 1H NMR and ESI-MS studies. Photoinduced release of DAS from the Ru(II) core was monitored by 1H NMR and UV-vis spectroscopy. When irradiated with a 470 nm blue LED in DMSO, the photosubstitution quantum yields (Φ) of 0.035 and 0.057 were observed for 1-DAS and 2-DAS, respectively. Intriguing solution-state speciation and kinetic behaviors of the uncaged and caged Ru(II)-complexes emerged from 1H NMR studies in the dark, and they are depicted in this work. The caged 1-DAS and 2-DAS complexes remained mostly structurally intact for a reasonably long period in DMSO. The uncaged 1-Cl and 2-Cl complexes, although did not undergo substitution in only DMSO but in the 10% DMSO/H2O mixture, completely converted to the corresponding DMSO-adduct within 16 h. Toward gaining insights into the reactivity with the biological targets, we observed that 1-Cl upon hydrolysis formed an adduct with 5'-GMP, while a small amount of GSSG-adduct was observed when 1-Cl was reacted with GSH in H2O at 323 K. 1-Cl after hydrolysis reacted with l-methionine, although the rate was slightly slower compared with that with DMSO, suggesting varying reaction kinetics with different sulfur-based linkages. Although 1-H2O reacted with sulfoxide and thioether ligands at room temperature, the rate was much faster at higher temperatures obviously, and thiol-based systems needed higher thermal energy for conjugation. Overall, these studies provide insight for thoughtful design of new generation Ru(II) polypyridyl complexes for caging suitable bioactive organic molecules.
Collapse
Affiliation(s)
- Ramranjan Mishra
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Abhijit Saha
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Pritha Chatterjee
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Atish Bhattacharyya
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Ashis K Patra
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
35
|
Pozza MD, Mesdom P, Abdullrahman A, Prieto Otoya TD, Arnoux P, Frochot C, Niogret G, Saubaméa B, Burckel P, Hall JP, Hollenstein M, Cardin CJ, Gasser G. Increasing the π-Expansive Ligands in Ruthenium(II) Polypyridyl Complexes: Synthesis, Characterization, and Biological Evaluation for Photodynamic Therapy Applications. Inorg Chem 2023; 62:18510-18523. [PMID: 37913550 DOI: 10.1021/acs.inorgchem.3c02606] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Lack of selectivity is one of the main issues with currently used chemotherapies, causing damage not only to altered cells but also to healthy cells. Over the last decades, photodynamic therapy (PDT) has increased as a promising therapeutic tool due to its potential to treat diseases like cancer or bacterial infections with a high spatiotemporal control. Ruthenium(II) polypyridyl compounds are gaining attention for their application as photosensitizers (PSs) since they are generally nontoxic in dark conditions, while they show remarkable toxicity after light irradiation. In this work, four Ru(II) polypyridyl compounds with sterically expansive ligands were studied as PDT agents. The Ru(II) complexes were synthesized using an alternative route to those described in the literature, which resulted in an improvement of the synthesis yields. Solid-state structures of compounds [Ru(DIP)2phen]Cl2 and [Ru(dppz)2phen](PF6)2 have also been obtained. It is well-known that compound [Ru(dppz)(phen)2]Cl2 binds to DNA by intercalation. Therefore, we used [Ru(dppz)2phen]Cl2 as a model for DNA interaction studies, showing that it stabilized two different sequences of duplex DNA. Most of the synthesized Ru(II) derivatives showed very promising singlet oxygen quantum yields, together with noteworthy photocytotoxic properties against two different cancer cell lines, with IC50 in the micro- or even nanomolar range (0.06-7 μM). Confocal microscopy studies showed that [Ru(DIP)2phen]Cl2 and [Ru(DIP)2TAP]Cl2 accumulate preferentially in mitochondria, while no mitochondrial internalization was observed for the other compounds. Although [Ru(dppn)2phen](PF6)2 did not accumulate in mitochondria, it interestingly triggered an impairment in mitochondrial respiration after light irradiation. Among others, [Ru(dppn)2phen](PF6)2 stands out for its very good IC50 values, correlated with a very high singlet oxygen quantum yield and mitochondrial respiration disruption.
Collapse
Affiliation(s)
- Maria Dalla Pozza
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, Paris 75005, France
| | - Pierre Mesdom
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, Paris 75005, France
| | - Ahmad Abdullrahman
- Department of Pharmacy, Chemistry and Pharmacy Building, University of Reading, Whiteknights Campus, Reading, Berkshire RG6 6AD, U.K
| | | | | | - Céline Frochot
- Université de Lorraine, CNRS, LRGP, Nancy F-54000, France
| | - Germain Niogret
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Departement of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, Paris 75015, France
| | - Bruno Saubaméa
- Université Paris Cité, INSERM, CNRS, P-MIM, Plateforme d'Imagerie Cellulaire et Moléculaire (PICMO), Paris F-75006, France
| | - Pierre Burckel
- Université de Paris, Institut de physique du globe de Paris, CNRS, Paris F-75005, France
| | - James P Hall
- Department of Pharmacy, Chemistry and Pharmacy Building, University of Reading, Whiteknights Campus, Reading, Berkshire RG6 6AD, U.K
| | - Marcel Hollenstein
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Departement of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, Paris 75015, France
| | - Christine J Cardin
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, Paris 75005, France
| |
Collapse
|
36
|
Wang Y, Felder PS, Mesdom P, Blacque O, Mindt TL, Cariou K, Gasser G. Towards Ruthenium(II)-Rhenium(I) Binuclear Complexes as Photosensitizers for Photodynamic Therapy. Chembiochem 2023; 24:e202300467. [PMID: 37526951 DOI: 10.1002/cbic.202300467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/02/2023]
Abstract
The search for new metal-based photosensitizers (PSs) for anticancer photodynamic therapy (PDT) is a fast-developing field of research. Knowing that polymetallic complexes bear a high potential as PDT PSs, in this study, we aimed at combining the known photophysical properties of a rhenium(I) tricarbonyl complex and a ruthenium(II) polypyridyl complex to prepare a ruthenium-rhenium binuclear complex that could act as a PS for anticancer PDT. Herein, we present the synthesis and characterization of such a system and discuss its stability in aqueous solution. In addition, one of our complexes prepared, which localized in mitochondria, was found to have some degree of selectivity towards two types of cancerous cells: human lung carcinoma A549 and human colon colorectal adenocarcinoma HT29, with interesting photo-index (PI) values of 135.1 and 256.4, respectively, compared to noncancerous retinal pigment epithelium RPE1 cells (22.4).
Collapse
Affiliation(s)
- Youchao Wang
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Patrick S Felder
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Pierre Mesdom
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Olivier Blacque
- University of Zurich, Department of Chemistry, CH-8057, Zurich, Switzerland
| | - Thomas L Mindt
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währingerstraße 42, 1090, Vienna, Austria
- Joint Applied Medicinal Radiochemistry Facility, University of Vienna, Währingerstraße 42, 1090, Vienna, Austria
- Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, 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
|
37
|
Vinck R, Dömötör O, Karges J, Jakubaszek M, Seguin J, Tharaud M, Guérineau V, Cariou K, Mignet N, Enyedy ÉA, Gasser G. In Situ Bioconjugation of a Maleimide-Functionalized Ruthenium-Based Photosensitizer to Albumin for Photodynamic Therapy. Inorg Chem 2023; 62:15510-15526. [PMID: 37708255 DOI: 10.1021/acs.inorgchem.3c01984] [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: 09/16/2023]
Abstract
Maleimide-containing prodrugs can quickly and selectively react with circulating serum albumin following their injection in the bloodstream. The drug-albumin complex then benefits from longer blood circulation times and better tumor accumulation. Herein, we have applied this strategy to a previously reported highly phototoxic Ru polypyridyl complex-based photosensitizer to increase its accumulation at the tumor, reduce off-target cytotoxicity, and therefore improve its pharmacological profile. Specifically, two complexes were synthesized bearing a maleimide group: one complex with the maleimide directly incorporated into the bipyridyl ligand, and the other has a hydrophilic linker between the ligand and the maleimide group. Their interaction with albumin was studied in-depth, revealing their ability to efficiently bind both covalently and noncovalently to the plasma protein. A crucial finding is that the maleimide-functionalized complexes exhibited significantly lower cytotoxicity in noncancerous cells under dark conditions compared to the nonfunctionalized complex, which is a highly desirable property for a photosensitizer. The binding to albumin also led to a decrease in the phototoxicity of the Ru bioconjugates in comparison to the nonfunctionalized complex, probably due to a decreased cellular uptake. Unfortunately, this decrease in phototoxicity was not compensated by a dramatic increase in tumor accumulation, as was demonstrated in a tumor-bearing mouse model using inductively coupled plasma mass spectrometry (ICP-MS) studies. Consequently, this study provides valuable insight into the future design of in situ albumin-binding complexes for photodynamic therapy in order to maximize their effectiveness and realize their full potential.
Collapse
Affiliation(s)
- Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Orsolya Dömötör
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7. H-6720 Szeged, Hungary
| | - Johannes Karges
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Marta Jakubaszek
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Johanne Seguin
- Université Paris Cité, UTCBS, INSERM, CNRS, 75006 Paris, France
| | - Mickaël Tharaud
- Biogéochimie à l'Anthropocène des Eléments et Contaminants Emergents, Institut de Physique du Globe de Paris, 75005 Paris, France
| | - Vincent Guérineau
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Sud, Université Paris-Saclay, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Nathalie Mignet
- Université Paris Cité, UTCBS, INSERM, CNRS, 75006 Paris, France
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7. H-6720 Szeged, Hungary
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| |
Collapse
|
38
|
Zhang Y, Doan BT, Gasser G. Metal-Based Photosensitizers as Inducers of Regulated Cell Death Mechanisms. Chem Rev 2023; 123:10135-10155. [PMID: 37534710 DOI: 10.1021/acs.chemrev.3c00161] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Over the last few decades, various forms of regulated cell death (RCD) have been discovered and were found to improve cancer treatment. Although there are several reviews on RCD induced by photodynamic therapy (PDT), a comprehensive summary covering metal-based photosensitizers (PSs) as RCD inducers has not yet been presented. In this review, we systematically summarize the works on metal-based PSs that induce different types of RCD, including ferroptosis, immunogenic cell death (ICD), and pyroptosis. The characteristics and mechanisms of each RCD are explained. At the end of each section, a summary of the reported commonalities between different metal-based PSs inducing the same RCD is emphasized, and future perspectives on metal-based PSs inducing novel forms of RCD are discussed at the end of the review. Considering the essential roles of metal-based PSs and RCD in cancer therapy, we hope that this review will provide the stage for future advances in metal-based PSs as RCD inducers.
Collapse
Affiliation(s)
- Yiyi Zhang
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemistry, 75005 Paris, France
| | - Bich-Thuy Doan
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory of Synthesis, Electrochemistry, Imaging and Analytical Systems for Diagnosis, 75005 Paris, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemistry, 75005 Paris, France
| |
Collapse
|
39
|
Curley R, Burke CS, Gkika KS, Noorani S, Walsh N, Keyes TE. Phototoxicity of Tridentate Ru(II) Polypyridyl Complex with Expanded Bite Angles toward Mammalian Cells and Multicellular Tumor Spheroids. Inorg Chem 2023; 62:13089-13102. [PMID: 37535942 PMCID: PMC10428208 DOI: 10.1021/acs.inorgchem.3c01982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 08/05/2023]
Abstract
Tridentate ligand-coordinated ruthenium (II) polypyridyl complexes with large N-Ru-N bite angles have been shown to promote ligand field splitting and reduce singlet-triplet state mixing leading to dramatically extended emission quantum yields and lifetimes under ambient conditions. These effects are anticipated to enhance their photoinduced singlet oxygen production, promoting prospects for such complexes as type II phototherapeutics. In this contribution, we examined this putative effect for [Ru(bqp)(bqpCOOEt)]2+, Ru-bqp-ester, a heteroleptic complex containing bqp = [2,6-bi(quinolin-8-yl)pyridine], a well-established large bite angle tridentate ligand, as well as its peptide conjugates [Ru(bqp)(bqpCONH-ahx-FrFKFrFK(Ac)-CONH2)]5+ (Ru-bqp-MPP) and [Ru(bqp) (bqp)(CONH-ahx-RRRRRRRR-CONH2)]10+ (Ru-bqp-R8) that were prepared in an effort to promote live cell/tissue permeability and targeting of the parent. Membrane permeability of both parent and peptide conjugates were compared across 2D cell monolayers; A549, Chinese hamster ovary, human pancreatic cancer (HPAC), and 3D HPAC multicellular tumor spheroids (MCTS) using confocal microscopy. Both the parent complex and peptide conjugates showed exceptional permeability with rapid uptake in both 2D and 3D cell models but with little distinction in permeability or distribution in cells between the parent or peptide conjugates. Unexpectedly, the uptake was temperature independent and so attributed to passive permeation. Both dark and photo-toxicity of the Ru(II) complexes were assessed across cell types, and the parent showed notably low dark toxicity. In contrast, the parent and conjugates were found to be highly phototoxic, with impressive phototoxic indices (PIs) toward HPAC cell monolayers in particular, with PI values ranging from ∼580 to 760. Overall, our data indicate that the Ru(II) parent complex and its peptide conjugates show promise at both cell monolayers and 3D MCTS as photosensitizers for photodynamic therapy.
Collapse
Affiliation(s)
- Rhianne
C. Curley
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Christopher S. Burke
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Karmel S. Gkika
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Sara Noorani
- National
Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Naomi Walsh
- National
Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Tia E. Keyes
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| |
Collapse
|
40
|
Karges J. Encapsulation of Ru(II) Polypyridine Complexes for Tumor-Targeted Anticancer Therapy. BME FRONTIERS 2023; 4:0024. [PMID: 37849670 PMCID: PMC10392611 DOI: 10.34133/bmef.0024] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/02/2023] [Indexed: 10/19/2023] Open
Abstract
Ru(II) polypyridine complexes have attracted much attention as anticancer agents because of their unique photophysical, photochemical, and biological properties. Despite their promising therapeutic profile, the vast majority of compounds are associated with poor water solubility and poor cancer selectivity. Among the different strategies employed to overcome these pharmacological limitations, many research efforts have been devoted to the physical or covalent encapsulation of the Ru(II) polypyridine complexes into nanoparticles. This article highlights recent developments in the design, preparation, and physicochemical properties of Ru(II) polypyridine complex-loaded nanoparticles for their potential application in anticancer therapy.
Collapse
Affiliation(s)
- Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| |
Collapse
|
41
|
Aboudzadeh MA, Rodríguez-Fanjul V, Terenzi A, Gónzalez de San Román E, Miranda JI, Pizarro AM, Salassa L, Barroso-Bujans F. Synthesis and Characterization of a Luminescent Cyclic Poly(ethylene oxide)-Polypyridyl Ruthenium Complex. ACS Macro Lett 2023:999-1004. [PMID: 37406348 DOI: 10.1021/acsmacrolett.3c00225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
We report the synthesis of a macrocyclic poly(ethylene oxide) (PEO) connected by one [Ru(bpy)3]2+ unit (where bpy = 2,2'-bipyridine), a photoactive metal complex that provides photosensitivity and potential biomedical applications to this polymer structure. The PEO chain provides biocompatibility, water solubility, and topological play. The macrocycles were successfully synthesized by copper-free click cycloaddition between a bifunctional dibenzocyclooctyne (DBCO)-PEO precursor and 4,4'-diazido-2,2'-bipyridine, followed by complexation with [Ru(bpy)2Cl2]. The cyclic product accumulated efficiently in MCF7 cancer cells and exhibited a longer fluorescence lifetime than its linear analogue, likely due to differences in the accessibility of the ligand-centered/intraligand states of Ru polypyridyls in both topologies.
Collapse
Affiliation(s)
- M Ali Aboudzadeh
- Donostia International Physics Center (DIPC), Paseo Manuel Lardizabal 4, 20018 Donostia-San Sebastian, Spain
- Materials Physics Center, CSIC-UPV/EHU, Paseo Manuel Lardizabal 5, 20018 Donostia-San Sebastian, Spain
| | | | - Alessio Terenzi
- Donostia International Physics Center (DIPC), Paseo Manuel Lardizabal 4, 20018 Donostia-San Sebastian, Spain
| | - Estibaliz Gónzalez de San Román
- POLYMAT, University of the Basque Country UPV/EHU, Joxe Mari Korta Center, Avda. Tolosa 72, 20018 Donostia-San Sebastian, Spain
| | - José I Miranda
- SGIKer, NMR Service, University of the Basque Country UPV/EHU, Joxe Mari Korta R&D Ctr, Avda. Tolosa-72, Donostia-San Sebastian 20018, Spain
| | - Ana M Pizarro
- IMDEA Nanociencia, Faraday 9, 28049 Madrid, Spain
- Unidad Asociada de Nanobiotecnología CNB-CSIC-IMDEA, 28049 Madrid, Spain
| | - Luca Salassa
- Donostia International Physics Center (DIPC), Paseo Manuel Lardizabal 4, 20018 Donostia-San Sebastian, Spain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastian, 20018, Spain
- IKERBASQUE - Basque Foundation for Science, María Díaz de Haro 3, E-48013 Bilbao, Spain
| | - Fabienne Barroso-Bujans
- Donostia International Physics Center (DIPC), Paseo Manuel Lardizabal 4, 20018 Donostia-San Sebastian, Spain
- Materials Physics Center, CSIC-UPV/EHU, Paseo Manuel Lardizabal 5, 20018 Donostia-San Sebastian, Spain
- IKERBASQUE - Basque Foundation for Science, María Díaz de Haro 3, E-48013 Bilbao, Spain
| |
Collapse
|
42
|
Zhou XQ, Wang P, Ramu V, Zhang L, Jiang S, Li X, Abyar S, Papadopoulou P, Shao Y, Bretin L, Siegler MA, Buda F, Kros A, Fan J, Peng X, Sun W, Bonnet S. In vivo metallophilic self-assembly of a light-activated anticancer drug. Nat Chem 2023; 15:980-987. [PMID: 37169984 PMCID: PMC10322715 DOI: 10.1038/s41557-023-01199-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/05/2023] [Indexed: 05/13/2023]
Abstract
Self-assembling molecular drugs combine the easy preparation typical of small-molecule chemotherapy and the tumour-targeting properties of drug-nanoparticle conjugates. However, they require a supramolecular interaction that survives the complex environment of a living animal. Here we report that the metallophilic interaction between cyclometalated palladium complexes generates supramolecular nanostructures in living mice that have a long circulation time (over 12 h) and efficient tumour accumulation rate (up to 10.2% of the injected dose per gram) in a skin melanoma tumour model. Green light activation leads to efficient tumour destruction due to the type I photodynamic effect generated by the self-assembled palladium complexes, as demonstrated in vitro by an up to 96-fold cytotoxicity increase upon irradiation. This work demonstrates that metallophilic interactions are well suited to generating stable supramolecular nanotherapeutics in vivo with exceptional tumour-targeting properties.
Collapse
Affiliation(s)
- Xue-Quan Zhou
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Peiyuan Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, People's Republic of China
| | - Vadde Ramu
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | - Liyan Zhang
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | - Suhua Jiang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, People's Republic of China
| | - Xuezhao Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China
| | - Selda Abyar
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | | | - Yang Shao
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | - Ludovic Bretin
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | - Maxime A Siegler
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Francesco Buda
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | - Alexander Kros
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, People's Republic of China.
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Universiteit Leiden, Leiden, the Netherlands.
| |
Collapse
|
43
|
Li C, Pang Y, Xu Y, Lu M, Tu L, Li Q, Sharma A, Guo Z, Li X, Sun Y. Near-infrared metal agents assisting precision medicine: from strategic design to bioimaging and therapeutic applications. Chem Soc Rev 2023. [PMID: 37334831 DOI: 10.1039/d3cs00227f] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Metal agents have made incredible strides in preclinical research and clinical applications in recent years, but their short emission/absorption wavelengths continue to be a barrier to their distribution, therapeutic action, visual tracking, and efficacy evaluation. Nowadays, the near-infrared window (NIR, 650-1700 nm) provides a more accurate imaging and treatment option. Thus, there has been ongoing research focusing on developing multifunctional NIR metal agents for imaging and therapy that have deeper tissue penetration. The design, characteristics, bioimaging, and therapy of NIR metal agents are covered in this overview of papers and reports published to date. To start with, we focus on describing the structure, design strategies, and photophysical properties of metal agents from the NIR-I (650-1000 nm) to NIR-II (1000-1700 nm) region, in order of molecular metal complexes (MMCs), metal-organic complexes (MOCs), and metal-organic frameworks (MOFs). Next, the biomedical applications brought by these superior photophysical and chemical properties for more accurate imaging and therapy are discussed. Finally, we explore the challenges and prospects of each type of NIR metal agent for future biomedical research and clinical translation.
Collapse
Affiliation(s)
- Chonglu Li
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, School of Public Health, Wuhan University of Science and Technology, Wuhan 430065, China.
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Yida Pang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Yuling Xu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Mengjiao Lu
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China.
| | - Le Tu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Qian Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Amit Sharma
- CSIR-Central Scientific Instruments Organisation, Sector-30C, Chandigarh 160030, India
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, School of Public Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Xiangyang Li
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China.
| | - Yao Sun
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
44
|
Kessel D. Critical PDT Theory VII: The Saga of Ruthenium. Photodiagnosis Photodyn Ther 2023:103615. [PMID: 37201769 DOI: 10.1016/j.pdpdt.2023.103615] [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: 03/28/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 05/20/2023]
Abstract
Recent studies involving photosensitizing agents containing ruthenium (Ru) are interpreted as an indication that this approach might be useful for treatment of bladder cancer. The absorbance spectrum of such agents tends to be limited to wavelengths < 600 nm. While this can spare underlying tissues from photodamage, this will limit applications to instances where only a thin layer of malignant cells is present. Among the more potentially interesting results is a protocol that uses only Ru nanoparticles. Other issues in Ru-based photodynamic therapy are discussed including the limited absorbance spectrum, questions relating to methodology and a general lack of details concerning localization and death pathways.
Collapse
Affiliation(s)
- David Kessel
- Department of Pharmacology, School of Medicine, Wayne State University; Detroit, MI 48201 USA.
| |
Collapse
|
45
|
Zhu JH, Mei LP, Wang AJ, Song YY, Feng JJ. Integration of phosphate functionalized Pt/TiO 2 and Ru(bpy) 32+ sensitization for ultrasensitive assay of adenosine deaminase activity on a novel split-typed PEC aptasensor. Biosens Bioelectron 2023; 226:115141. [PMID: 36796307 DOI: 10.1016/j.bios.2023.115141] [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: 10/25/2022] [Revised: 01/16/2023] [Accepted: 02/07/2023] [Indexed: 02/10/2023]
Abstract
To date, it is still a challenge for high-performance photoelectrochemical (PEC) assay of low-abundance adenosine deaminase (ADA) in fundamental research and clinical diagnosis. Herein, phosphate-functionalized Pt/TiO2 (termed PO43-/Pt/TiO2) was prepared as ideal photoactive material to develop a split-typed PEC aptasensor for detection of ADA activity, coupled by a Ru(bpy)32+ sensitization strategy. We critically studied the effects of the PO43- and Ru(bpy)32+ on the detection signals, and discussed the signal-amplified mechanism. Specifically, hairpin-structured adenosine (AD) aptamer was splited into single chain via ADA-induced catalytic reaction, and subsequently hybridized with complementary DNA (cDNA, initially coating on magnetic beads). The in-situ formed double-stranded DNA (dsDNA) was further intercalated by more Ru(bpy)32+ to amplify the photocurrents. The resultant PEC biosensor showed a broader linear range of 0.05-100 U L-1 and a lower limit of detection (0.019 U L-1), which can fill the blank for analysis of ADA activity. This research would provide some valuable insights for building advanced PEC aptasensors in ADA-related research and clinical diagnosis.
Collapse
Affiliation(s)
- Jian-Hong Zhu
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Sciences, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China; College of Sciences, Northeastern University, Box 332, Shenyang, 110004, China
| | - Li-Ping Mei
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Sciences, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
| | - Ai-Jun Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Sciences, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
| | - Yan-Yan Song
- College of Sciences, Northeastern University, Box 332, Shenyang, 110004, China
| | - Jiu-Ju Feng
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Sciences, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China.
| |
Collapse
|
46
|
Gao Y, Wang Z, Wu J, Lu L. A cellular NO sensor based on aggregation-induced electrochemiluminescence and photoelectron transfer of a novel ruthenium(II) complex. Microchem J 2023. [DOI: 10.1016/j.microc.2023.108714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
47
|
Yin CW, Tsai MK, Chen YJ. Low-Temperature Observation of the Excited-State Decay of Ruthenium-(Mono-2,2':6',2″-Terpyridine) Ions with Innocent Ligands: DFT Modeling of an 3MLCT- 3MC Intersystem Crossing Pathway. ACS OMEGA 2023; 8:11623-11633. [PMID: 37008138 PMCID: PMC10061511 DOI: 10.1021/acsomega.3c01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 06/19/2023]
Abstract
The synthesis, electrochemistry, and photophysical characterization of five 2,2':6',2″-terpyridine ruthenium complexes (Ru-tpy complexes) is reported. The electrochemical and photophysical behavior varied depending on the ligands, i.e., amine (NH3), acetonitrile (AN), and bis(pyrazolyl)methane (bpm), for this series of Ru-tpy complexes. The target [Ru(tpy)(AN)3]2+ and [Ru(tpy)(bpm)(AN)]2+ complexes were found to have low-emission quantum yields in low-temperature observations. To better understand this phenomenon, density functional theory (DFT) calculations were performed to simulate the singlet ground state (S0), Te, and metal-centered excited states (3MC) of these complexes. The calculated energy barriers between Te and the low-lying 3MC state for [Ru(tpy)(AN)3]2+ and [Ru(tpy)(bpm)(AN)]2+ provided clear evidence in support of their emitting state decay behavior. Developing a knowledge of the underlying photophysics of these Ru-tpy complexes will allow new complexes to be designed for use in photophysical and photochemical applications in the future.
Collapse
Affiliation(s)
- Chi-Wei Yin
- Department
of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, ROC
| | - Ming-Kang Tsai
- Department
of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, ROC
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan, ROC
| | - Yuan Jang Chen
- Department
of Chemistry, Fu-Jen Catholic University, New Taipei City 24205, Taiwan, ROC
| |
Collapse
|
48
|
Cao F, Wang H, Lu N, Zhang P, Huang H. A Photoisomerizable Zinc (II) Complex Inhibits Microtubule Polymerization for Photoactive Therapy. Angew Chem Int Ed Engl 2023; 62:e202301344. [PMID: 36749111 DOI: 10.1002/anie.202301344] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/08/2023]
Abstract
The photoisomerization-induced cytotoxicity in photopharmacology provides a unique pathway for phototherapy because it is independent of endogenous oxygen. In this study, we developed a biosafe photoisomerizable zinc(II) complex (Zn1), which releases its trans ligand (trans-L1) after being irradiated with blue light. This causes the complex to undergo photoisomerization and produce the toxic cis product (cis-L1) and generate singlet oxygen (1 O2 ). The resulting series of events caused impressive phototoxicity in hypoxic A431 skin cancer cells, as well as in a tumor model in vivo. Interestingly, Zn1 was able to inhibit tumor microtubule polymerization, while still showing good biocompatibility and biosafety in vivo. This photoisomerizable zinc(II) complex provides a novel strategy for addressing the oxygen-dependent limitation of traditional photodynamic therapy.
Collapse
Affiliation(s)
- Fengshu Cao
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
- School of Pharmaceutical Science (Shenzhen), Shenzhen campus of Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Haobing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Nong Lu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Huaiyi Huang
- School of Pharmaceutical Science (Shenzhen), Shenzhen campus of Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| |
Collapse
|
49
|
Wei X, Cui WB, Qin GY, Zhang XE, Sun FY, Li H, Guo JF, Ren AM. Theoretical Investigation of Ru(II) Complexes with Long Lifetime and a Large Two-Photon Absorption Cross-Section in Photodynamic Therapy. J Med Chem 2023; 66:4167-4178. [PMID: 36884221 DOI: 10.1021/acs.jmedchem.3c00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Two-photon photodynamic therapy (TP-PDT), as a new method for cancer, has shown unique advantages in tumors. A low two-photon absorption cross-section (δ) in the biologic spectral window and a short triplet state lifetime are the important issues faced by the current photosensitizers (PSs) in TP-PDT. In this paper, the photophysical properties of a series of Ru(II) complexes were studied by density functional theory and time-dependent density functional theory methods. The electronic structure, one- and two-photon absorption properties, type I/II mechanisms, triplet state lifetime, and solvation free energy were calculated. The results showed that the substitution of methoxyls by pyrene groups greatly improved the lifetime of the complex. Furthermore, the addition of acetylenyl groups subtly enhanced δ. Overall, complex 3b possess a large δ(1376 GM), a long lifetime (136 μs), and better solvation free energy. It is hoped that it can provide valuable theoretical guidance for the design and synthesis of efficient two-photon PSs in the experiment.
Collapse
Affiliation(s)
- Xue Wei
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Liutiao Road #2, Changchun 130061, P. R. China
| | - Wei-Bo Cui
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Liutiao Road #2, Changchun 130061, P. R. China
| | - Gui-Ya Qin
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Liutiao Road #2, Changchun 130061, P. R. China
| | - Xiu-E Zhang
- School of Physics, Northeast Normal University, Changchun 130024, P. R. China
| | - Feng-Yi Sun
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Liutiao Road #2, Changchun 130061, P. R. China
| | - Hui Li
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Liutiao Road #2, Changchun 130061, P. R. China
| | - Jing-Fu Guo
- School of Physics, Northeast Normal University, Changchun 130024, P. R. China
| | - Ai-Min Ren
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Liutiao Road #2, Changchun 130061, P. R. China
| |
Collapse
|
50
|
Quílez-Alburquerque J, Saad MA, Descalzo AB, Orellana G, Hasan T. Hyaluronic acid-poly(lactic-co-glycolic acid) nanoparticles with a ruthenium photosensitizer cargo for photokilling of oral cancer cells. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2022.114349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|