1
|
Nkune NW, Abrahamse H. The Combination of Active-Targeted Photodynamic Therapy and Photoactivated Chemotherapy for Enhanced Cancer Treatment. JOURNAL OF BIOPHOTONICS 2025:e70005. [PMID: 40083278 DOI: 10.1002/jbio.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
Scientists have been actively investigating novel therapies that can effectively eradicate cancer cells with negligible side effects in normal tissues when used alone or in a combinatorial approach. Photodynamic therapy has emerged as a promising non-invasive therapy that integrates photosensitizer, oxygen, and a specific wavelength of light for the treatment of cancer. Despite encouraging outcomes yielded by PDT, conventional PSs are faced with longstanding challenges such as poor water solubility, a short half-life, and off-target toxicity. Development of nanotherapeutics has shown great potential in overcoming this issue. The tumor microenvironment is inherently hypoxic, and this promotes tumor resistance to PDT, as it is oxygen-dependent. Photoactivated chemotherapy, an oxygen-independent light-based therapy, utilizes chemotherapeutic regimens that remain inert until exposed to light, allowing target-specific activation while minimizing off-target toxicity. Integration of these techniques can improve selectivity and yield synergistic cytotoxic effects that could improve cancer treatment.
Collapse
Affiliation(s)
- Nkune Williams Nkune
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| |
Collapse
|
2
|
Chang MR, Matnurov EM, Wu C, Arakelyan J, Choe HJ, Kushnarev V, Yap JY, Soo XX, Chow MJ, Berger W, Ang WH, Babak MV. Leveraging Immunogenic Cell Death to Enhance the Immune Response against Malignant Pleural Mesothelioma Tumors. J Am Chem Soc 2025; 147:7908-7920. [PMID: 39992709 PMCID: PMC11887451 DOI: 10.1021/jacs.4c17966] [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: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
Although various metal-based compounds have exhibited excellent immunogenic cell death (ICD)-inducing properties both in vitro and in vivo, the majority of these compounds have been discovered serendipitously. In this work, we have successfully synthesized and characterized 35 cyclometalated Au(III) complexes containing dithiocarbamate ligands, with 25 of these complexes being previously unreported. Their ability to induce phagocytosis in vitro against immunologically "cold" malignant pleural mesothelioma (MPM) cells was strongly dependent on the cyclometalated scaffold and the overall lipophilicity of the complexes. We elucidated the role of cell death mechanisms in the observed ICD effects and identified correlations between the ability of the complexes to induce necrotic cell death and ICD, both in vitro and in vivo. Complex 2G, with its high phagocytosis rates and low necrosis rates, was recognized as a bona fide ICD inducer, demonstrating a remarkably long-lasting immune response in vaccinated mice. In contrast, complex 1C, characterized by high phagocytosis rates and high necrosis rates, failed to elicit a sustained immune response upon following vaccination; however, it triggered selective activation of calreticulin in tumors upon direct in vivo administration. Overall, this study offers a framework for predicting ICD effects in vivo for structurally similar Au(III) complexes, with the potential for extension to other series of metal complexes.
Collapse
Affiliation(s)
- Meng Rui Chang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Egor M. Matnurov
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Chengnan Wu
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jemma Arakelyan
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Ho-Jung Choe
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Vladimir Kushnarev
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jian Yu Yap
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Xiu Xuan Soo
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Mun Juinn Chow
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Walter Berger
- Center for
Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, Vienna 1090, Austria
| | - Wee Han Ang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Maria V. Babak
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| |
Collapse
|
3
|
Shi H, Marchi RC, Sadler PJ. Advances in the Design of Photoactivatable Metallodrugs: Excited State Metallomics. Angew Chem Int Ed Engl 2025; 64:e202423335. [PMID: 39806815 DOI: 10.1002/anie.202423335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Photoactivatable metal complexes offer the prospect of novel drugs with low side effects and new mechanisms of action to combat resistance to current therapy. We highlight recent progress in the design of platinum, ruthenium, iridium, gold and other transition metal complexes, especially for applications as anticancer and anti-infective agents. In particular, understanding excited state chemistry related to identification of the bioactive species (excited state metallomics/pharmacophores) is important. Photoactivatable metallodrugs are classified here as photocatalysts, photorelease agents and ligand-activated agents. Their activation wavelengths, cellular mechanisms of action, experimental and theoretical metallomics of excited states and photoproducts are discussed to explore new strategies for the design and investigation of photoactivatable metallodrugs. These photoactivatable metallodrugs have potential in clinical applications of Photodynamic Therapy (PDT), Photoactivated Chemotherapy (PACT) and Photothermal Therapy (PTT).
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Rafael C Marchi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Peter J Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
4
|
Li Y, Liu H, Fang R, Jin J, Yang F, Chen J, Zhang J. Designing novel Au(III) complexes based on the structure of diazepam: Achieving a multiaction mechanism against glioma. Eur J Med Chem 2025; 283:117171. [PMID: 39705733 DOI: 10.1016/j.ejmech.2024.117171] [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: 10/09/2024] [Revised: 12/01/2024] [Accepted: 12/12/2024] [Indexed: 12/22/2024]
Abstract
Metal-based drugs have been used in the clinical treatment of tumors for over 30 years. However, no metal-based drugs have been clinically approved to treat glioma. Although metal complexes have excellent cytotoxicity, their most critical problem is crossing the blood-brain barrier. Therefore, to enable metal complexes to cross blood-brain barrier and target glioma therapy, herein, we propose to rationally used the basic structure of diazepam (5-chlorobenzophenone) and thiosemicarbazide to synthesize gold (Au) complexes C1, C2 and C3 with antiglioma activity. The C3 complex with two methyl groups attached to the N3 of thiosemicarbazone exhibited excellent cytotoxicity to glioma cells through its multiaction mechanism against glioma, inducing apoptosis, autophagy death, and deoxyribonucleic acid damage. In addition, the synthesized C3 complex can effectively cross the blood-brain barrier and accumulate in glioma, considerably decreasing the untoward reaction in vivo. Our findings provide a novel strategy for designing metal-based complexes for the treatment of glioma.
Collapse
Affiliation(s)
- Yanping Li
- Mental Health Education Center of College Student, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Haoran Liu
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, 541004, PR China
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| | - Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| |
Collapse
|
5
|
Chen F, Huang H, Zhang F, Wang R, Wang L, Chang Z, Cao L, Zhang W, Li L, Chen M, Shao D, Yang C, Dong WF, Sun W. Biomimetic Chlorosomes: Oxygen-Independent Photocatalytic Nanoreactors for Efficient Combination Photoimmunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413385. [PMID: 39499050 DOI: 10.1002/adma.202413385] [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: 09/06/2024] [Indexed: 11/07/2024]
Abstract
Photocatalytic therapy for hypoxic tumors often suffers from inefficiencies due to its dependence on oxygen and the risk of uncontrolled activation. Inspired by the oxygen-independent and precisely regulated photocatalytic functions of natural light-harvesting chlorosomes, chlorosome-mimetic nanoreactors, termed Ru-Chlos, are engineered by confining the aggregation of photosensitive ruthenium-polypyridyl-silane monomers. These Ru-Chlos exhibit markedly enhanced photocatalytic performance compared to their monomeric counterparts under acidic conditions, while notably bypassing the consumption of oxygen or hydrogen peroxide. The photocatalytic activity of Ru-Chlos is finely tunable through light-responsive disassembly of the Ru-bridged matrix, with tunability governed by pre-irradiation duration. Utilization of Ru-Chlos loading prodrug [2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)] (ABTS) for phototherapy facilitates the generation of toxic radicals (oxABTS) and the photocatalytic conversion of endogenous NADH to NAD+, inducing oxidative stress in hypoxic cancer cells. Simultaneously, the light-responsive degradation of Ru-Chlos produces Ru-based toxins that further contribute to the therapeutic effect. This dual-action mechanism elicits potent immunogenic cell death effects and significantly enhances antitumor efficacy with the aid of a PD-l blockade. These biomimetic chlorosomes highlight their potential to advance oxygen-independent photocatalytic nanoreactors with controlled activity for novel cancer photoimmunotherapy strategies.
Collapse
Affiliation(s)
- Fangman Chen
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hanyao Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fan Zhang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, 215163, China
| | - Ran Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Lei Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Zhimin Chang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, 215163, China
| | - Lei Cao
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Wensheng Zhang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Li Li
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, 215163, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Dan Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Wen-Fei Dong
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, 215163, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
6
|
Lee LC, Lo KK. Leveraging the Photofunctions of Transition Metal Complexes for the Design of Innovative Phototherapeutics. SMALL METHODS 2024; 8:e2400563. [PMID: 39319499 PMCID: PMC11579581 DOI: 10.1002/smtd.202400563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/03/2024] [Indexed: 09/26/2024]
Abstract
Despite the advent of various medical interventions for cancer treatment, the disease continues to pose a formidable global health challenge, necessitating the development of new therapeutic approaches for more effective treatment outcomes. Photodynamic therapy (PDT), which utilizes light to activate a photosensitizer to produce cytotoxic reactive oxygen species (ROS) for eradicating cancer cells, has emerged as a promising approach for cancer treatment due to its high spatiotemporal precision and minimal invasiveness. However, the widespread clinical use of PDT faces several challenges, including the inefficient production of ROS in the hypoxic tumor microenvironment, the limited penetration depth of light in biological tissues, and the inadequate accumulation of photosensitizers at the tumor site. Over the past decade, there has been increasing interest in the utilization of photofunctional transition metal complexes as photosensitizers for PDT applications due to their intriguing photophysical and photochemical properties. This review provides an overview of the current design strategies used in the development of transition metal complexes as innovative phototherapeutics, aiming to address the limitations associated with PDT and achieve more effective treatment outcomes. The current challenges and future perspectives on the clinical translation of transition metal complexes are also discussed.
Collapse
Affiliation(s)
- Lawrence Cho‐Cheung Lee
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| | - Kenneth Kam‐Wing Lo
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
- State Key Laboratory of Terahertz and Millimeter WavesCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| |
Collapse
|
7
|
Kuznetsov KM, Cariou K, Gasser G. Two in one: merging photoactivated chemotherapy and photodynamic therapy to fight cancer. Chem Sci 2024:d4sc04608k. [PMID: 39464604 PMCID: PMC11499979 DOI: 10.1039/d4sc04608k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
The growing number of cancer cases requires the development of new approaches for treatment. A therapy that has attracted the special attention of scientists is photodynamic therapy (PDT) due to its spatial and temporal resolution. However, it is accepted that this treatment methodology has limited application in cases of low cellular oxygenation, which is typical of cancerous tissues. Therefore, a strategy to overcome this drawback has been to combine this therapy with photoactivated chemotherapy (PACT), which works independently of the presence of oxygen. In this perspective, we examine compounds that act as both PDT and PACT agents and summarize their photophysical and biological characteristics.
Collapse
Affiliation(s)
- Kirill M Kuznetsov
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France http://www.gassergroup.com/ +33 1 85 78 41 51
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France http://www.gassergroup.com/ +33 1 85 78 41 51
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France http://www.gassergroup.com/ +33 1 85 78 41 51
| |
Collapse
|
8
|
Barretta P, Ponte F, Escudero D, Mazzone G. Computational Exploration of the Mechanism of Action of a Sorafenib-Containing Ruthenium Complex as an Anticancer Agent for Photoactivated Chemotherapy. Molecules 2024; 29:4298. [PMID: 39339293 PMCID: PMC11433670 DOI: 10.3390/molecules29184298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Ruthenium(II) polypyridyl complexes are being tested as potential anticancer agents in different therapies, which include conventional chemotherapy and light-activated approaches. A mechanistic study on a recently synthesized dual-action Ru(II) complex [Ru(bpy)2(sora)Cl]+ is described here. It is characterized by two mono-dentate leaving ligands, namely, chloride and sorafenib ligands, which make it possible to form a di-aquo complex able to bind DNA. At the same time, while the released sorafenib can induce ferroptosis, the complex is also able to act as a photosensitizer according to type II photodynamic therapy processes, thus generating one of the most harmful cytotoxic species, 1O2. In order to clarify the mechanism of action of the drug, computational strategies based on density functional theory are exploited. The photophysical properties of the complex, which include the absorption spectrum, the kinetics of ISC, and the character of all the excited states potentially involved in 1O2 generation, as well as the pathway providing the di-aquo complex, are fully explored. Interestingly, the outcomes show that light is needed to form the mono-aquo complex, after releasing both chloride and sorafenib ligands, while the second solvent molecule enters the coordination sphere of the metal once the system has come back to the ground-state potential energy surface. In order to simulate the interaction with canonical DNA, the di-aquo complex interaction with a guanine nucleobase as a model has also been studied. The whole study aims to elucidate the intricate details of the photodissociation process, which could help with designing tailored metal complexes as potential anticancer agents.
Collapse
Affiliation(s)
- Pierraffaele Barretta
- Department of Chemistry and Chemical Technologies, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy
| | - Daniel Escudero
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Heverlee, Belgium
| | - Gloria Mazzone
- Department of Chemistry and Chemical Technologies, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy
| |
Collapse
|
9
|
Chen M, Zhu Q, Zhang Z, Chen Q, Yang H. Recent Advances in Photosensitizer Materials for Light-Mediated Tumor Therapy. Chem Asian J 2024; 19:e202400268. [PMID: 38578217 DOI: 10.1002/asia.202400268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/06/2024]
Abstract
Photodynamic therapy (PDT) as an emerging therapeutic method has drawn much attention in the treatment field for cancer. Photosensitizer, which can convert photon energy into cytotoxic species under light irradiation, is the core component in PDT. The design of photosensitizers still faces problems of light absorption, targeting, penetration and oxygen dependence. With the rapid progress of material science, various photosensitizers have been developed to produce cytotoxic species for treatment of tumor with high selectivity, safety, and noninvasiveness. Besides, the applications of photosensitizers have been expanded to diverse cancer treatments such as drug release, optogenetics and immune checkpoint blockade. In this review, we summarize the recent advances of photosensitizers in various therapeutic methods for cancer. Prevailing challenges and further prospects associated with photosensitizers are also discussed.
Collapse
Affiliation(s)
- Minle Chen
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350002, People's Republic of China
| | - Qianru Zhu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350002, People's Republic of China
| | - Zhenzhen Zhang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350002, People's Republic of China
| | - Qiushui Chen
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350002, People's Republic of China
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350002, People's Republic of China
| |
Collapse
|
10
|
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
|
11
|
Xie Z, Cao B, Zhao J, Liu M, Lao Y, Luo H, Zhong Z, Xiong X, Wei W, Zou T. Ion Pairing Enables Targeted Prodrug Activation via Red Light Photocatalysis: A Proof-of-Concept Study with Anticancer Gold Complexes. J Am Chem Soc 2024; 146:8547-8556. [PMID: 38498689 DOI: 10.1021/jacs.4c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Photocatalysis has found increasing applications in biological systems, for example, in localized prodrug activation; however, high-energy light is usually required without giving sufficient efficiency and target selectivity. In this work, we report that ion pairing between photocatalysts and prodrugs can significantly improve the photoactivation efficiency and enable tumor-targeted activation by red light. This is exemplified by a gold-based prodrug (1d) functionalized with a morpholine moiety. Such a modification causes 1d to hydrolyze in aqueous solution, forming a cationic species that tightly interacts with anionic photosensitizers including Eosin Y (EY) and Rose Bengal (RB), along with a significant bathochromic shift of absorption tailing to the far-red region. As a result, a high photoactivation efficiency of 1d by EY or RB under low-energy light was found, leading to an effective release of active gold species in living cells, as monitored by a gold-specific biosensor (GolS-mCherry). Importantly, the morpholine moiety, with pKa ∼6.9, in 1d brings in a highly pH-sensitive and preferential ionic interaction under a slightly acidic condition over the normal physiological pH, enabling tumor-targeted prodrug activation by red light irradiation in vitro and in vivo. Since a similar absorption change was found in other morpholine/amine-containing clinic drugs, photocages, and precursors of reactive labeling intermediates, it is believed that the ion-pairing strategy could be extended for targeted activation of different prodrugs and for mapping of an acidic microenvironment by low-energy light.
Collapse
Affiliation(s)
- Zhiying Xie
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Bei Cao
- Warshel Institute for Computational Biology, and General Education Division, The Chinese University of Hong Kong, Shenzhen 518172, China
- School of Education Sciences, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, China
| | - Jing Zhao
- State Key Laboratory of Coordination Chemistry, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Moyi Liu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yuhan Lao
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hejiang Luo
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhi Zhong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaolin Xiong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Wei Wei
- State Key Laboratory of Coordination Chemistry, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Taotao Zou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, China
| |
Collapse
|
12
|
Yuan X, Xie Z, Zou T. Recent advances in hypoxia-activated compounds for cancer diagnosis and treatment. Bioorg Chem 2024; 144:107161. [PMID: 38306826 DOI: 10.1016/j.bioorg.2024.107161] [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: 11/01/2023] [Revised: 12/28/2023] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
Hypoxia, as a prevalent feature of solid tumors, is correlated with tumorigenesis, proliferation, and invasion, playing an important role in mediating the drug resistance and affecting the cancer treatment outcomes. Due to the distinct oxygen levels between tumor and normal tissues, hypoxia-targeted therapy has attracted significant attention. The hypoxia-activated compounds mainly depend on reducible organic groups including azo, nitro, N-oxides, quinones and azide as well as some redox-active metal complex that are selectively converted into active species by the increased reduction potential under tumor hypoxia. In this review, we briefly summarized our current understanding on hypoxia-activated compounds with a particular highlight on the recently developed prodrugs and fluorescent probes for tumor treatment and diagnosis. We have also discussed the challenges and perspectives of small molecule-based hypoxia-activatable prodrug for future development.
Collapse
Affiliation(s)
- Xiaoyu Yuan
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhiying Xie
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Taotao Zou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
13
|
Wang L, Qian Y. A type I and II compatible vinyl-pyridine modified BODIPY dimer photosensitizer for photodynamic therapy in A-549 cells. Org Biomol Chem 2023; 21:7339-7350. [PMID: 37642553 DOI: 10.1039/d3ob01130e] [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: 08/31/2023]
Abstract
In this paper, the vinyl-pyridine group was used to modify the BODIPY dimer photosensitizer (T-BDP2) to obtain a VP-BDP2 photosensitizer. Compared with the T-BDP2 photosensitizer, the VP-BDP2 photosensitizer could work under pure water conditions, the singlet oxygen yield was increased from 9.38% to 22.2%, the charge transfer rate was increased from about 30 ps to about 10 ps, and the red emission was enhanced in fluorescence imaging. In addition, the VP-BDP2 photosensitizer could also generate the superoxide radical (O2˙-) under pure water conditions. The ROS generation mechanism of the VP-BDP2 photosensitizer was considered to be the spin-orbit charge-transfer intersystem crossing (SOCT-ISC) mechanism, which was verified by fs-transient absorption spectra and theoretical calculation. In the photodynamic therapy of A-549 cells, the VP-BDP2 photosensitizers could generate singlet oxygen and superoxide radicals (O2˙-) under biological conditions, and showed high phototoxicity with the IC50 value at 12.1 μM under light at 525 nm. Additionally, the multiple dipolar configuration meant that the VP-BDP2 photosensitizer could be used in two-photon fluorescence zebrafish imaging under 800 nm excitation, which sets the stage for future two-photon photodynamic therapy.
Collapse
Affiliation(s)
- Lingfeng Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Ying Qian
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
14
|
Yang Z, Bian M, Lv L, Chang X, Wen Z, Li F, Lu Y, Liu W. Tumor-Targeting NHC-Au(I) Complex Induces Immunogenic Cell Death in Hepatocellular Carcinoma. J Med Chem 2023; 66:3934-3952. [PMID: 36827091 DOI: 10.1021/acs.jmedchem.2c01798] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Immunogenic cell death (ICD) is a promising direction of cancer immunotherapy in hepatocellular carcinoma (HCC). A series of novel NHC-Au(I) complexes derived from 4,5-diarylimidazole, containing glycyrrhetinic acid (GA) as an efficient targeting ligand for HCC, were herein designed and synthesized. Among these, complex 4C exhibited excellent effectiveness for tumor targeting and antitumor activity, which induced the occurrence of ICD in HCC cells. Additionally, 4C can effectively inhibit TrxR enzyme activity, increase reactive oxygen species (ROS) expression, lead to redox homeostasis disorder, mediate mitochondrial dysfunction and endoplasmic reticulum stress (ERS), and cause the characteristic discharge of damage-associated molecular patterns (DAMPs) in HCC cells. More importantly, 4C showed a great ICD-inducing effect in a vaccination mouse model and activated antitumor immunity in a tumor-bearing C57BL/6 mouse model, which is consistent with the in vitro results. In conclusion, we found the potential of Au(I) complex with HCC-targeted capability for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Zhibin Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali 671000, P. R. China
| | - Mianli Bian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Lin Lv
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Xingyu Chang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Zhenfan Wen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Fuwei Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
15
|
Au(III) Cyclometallated Compounds with 2-Arylpyridines and Their Derivatives or Analogues: 34 Years (1989–2022) of NMR and Single Crystal X-ray Studies. INORGANICS 2023. [DOI: 10.3390/inorganics11030100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
A review paper on Au(III) cyclometallated compounds with 2-arylpyridines (2-phenylpyridine, 2-benzylpyridine, 2-benzoylpyridine, 2-phenoxypyridine, 2-phenylsulfanylpyridine, 2-anilinopyridine, 2-(naphth-2-yl)pyridine, 2-(9,9-dialkylfluoren-2-yl)pyridines, 2-(dibenzofuran-4-yl)pyridine, and their derivatives) and their analogues (2-arylquinolines, 1- and 3-arylisoquinolines, 7,8-benzoquinoline), with 113 references. A total of 554 species, containing κ2-N(1),C(6′)*-Au(III), or analogous moiety (i.e., chelated by nitrogen of the pyridine-like ring and the deprotonated ortho- carbon of the phenyl-like ring) and, thus, possessing a character intermediate between metal complexes and organometallics, studied in the years 1989–2022 by NMR spectroscopy and/or single crystal X-ray diffraction (207 X-ray structures), are described. The compounds for which biological or catalytic activity and the luminescence properties were studied are also quoted.
Collapse
|