1
|
Mao X, Fei X, Cai T, Xu S, Zhang D, Pu S, Li Z. A turn-on mitochondria-targeted iridium (Ⅲ) Complex-Based probe for glutathione detection and photodynamic therapy of cancer cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125579. [PMID: 39689545 DOI: 10.1016/j.saa.2024.125579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024]
Abstract
As one of the most abundant biothiols in cells, glutathione (GSH) usually exists in a dynamic equilibrium of oxidized glutathione (GSSG) and reduces glutathione redox, and plays an essential reducing substance to maintain the REDOX balance of the microenvironment. So, the development of a reliable GSH sensor will be important for living cells and organisms. We fabricated a mitochondria targeted "turn-on" fluorescent sensor based on Ir (III) complex and successfully detected endogenous and exogenous GSH in living cells and zebrafish. For the probe Ir-DINI, a robust electron-withdrawing group 2,4-dinitrobenzoyl was introduced to quench the fluorescence, which could be broken through electrostatic interaction with GSH, following exposing a strong fluorescent Ir (Ⅲ) complex Ir-OH. On the other hand, photodynamic therapy (PDT) has attracted much attention in recent years due to its minimally invasive treatment. We found that singlet oxygen yields of probe Ir-DINI displayed an enhancement before and after the detection of GSH. Additionally, photodynamic studies in living cells illustrated that after reacting with GSH, probe Ir-DINI exhibited more obvious phototoxicity than before the detection of GSH. So the probe Ir-DINI could be served as a GSH sensor and potential GSH-activated photosensitizer for photodynamic therapy.
Collapse
Affiliation(s)
- Xueting Mao
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Xiao Fei
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Tangxuan Cai
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Sha Xu
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Daobin Zhang
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| | - Shouzhi Pu
- Institute of Carbon Neutral New Energy Research, Yuzhang Normal University, Nanchang 330031, China.
| | - Zhijian Li
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
2
|
Yang P, Wang T, Zhang L, Wang L. Fluorescent Microorganism-Based Composite: Enhancing Curcumin Delivery Efficiency and its Antitumor Application in Prostate Cancer. J Fluoresc 2025:10.1007/s10895-025-04185-2. [PMID: 39954179 DOI: 10.1007/s10895-025-04185-2] [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: 01/03/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Prostate cancer is a major global health concern, ranking as the second most common malignancy in men and the fifth leading cause of cancer-related deaths. Although curcumin exhibits potent antioxidant, anti-inflammatory, and antitumor properties, its clinical application is limited by poor solubility, low bioavailability, and rapid metabolism. In this study, we developed a microorganism-metal-organic framework (MOF)-based carrier (1-CP1) by combining a novel Zn(II) coordination polymer, [Zn(Hbcb)(PYTPY)] (1), with CP1. The carrier, loaded with curcumin to form 1-CP1@Curcumin, significantly enhanced the solubility, bioavailability, and stability of curcumin. Fluorescence assays revealed that the composite demonstrated a fluorescence emission peak at 511 nm, with a strong response to Fe³⁺ ions, showing a quenching efficiency of over 95%. In vitro experiments on LNCaP prostate cancer cells showed that 1-CP1@Curcumin significantly inhibited cell viability, with a reduction of approximately 50% at 20 µM curcumin concentration after 48 h of treatment. Additionally, quantitative PCR analysis of apoptosis-related gene expression revealed a significant decrease in Bcl-2 mRNA levels, indicating that the composite induced apoptosis in prostate cancer cells. These results highlight that 1-CP1@Curcumin effectively overcomes curcumin's delivery limitations and offers strong antitumor efficacy, providing an innovative platform for potential clinical applications in prostate cancer therapy.
Collapse
Affiliation(s)
- Ping Yang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai, China
| | - Tian Wang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai, China
| | - Lian Zhang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai, China
| | - Li Wang
- Department of Urology, Liyang People's Hospital, Changzhou, Jiangsu, China.
| |
Collapse
|
3
|
Nsubuga A, Fayad N, Pini F, Natile MM, Hildebrandt N. Small upconversion-ruthenium nanohybrids for cancer theranostics. NANOSCALE 2025; 17:3809-3821. [PMID: 39761019 DOI: 10.1039/d4nr04210g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Photoresponsive drug delivery systems have great potential for improved cancer therapy. However, most of the currently available drug-delivery nanosystems are relatively large and require light excitation with low tissue penetration. Here, we designed a near infrared responsive drug delivery system by loading [Ru(terpyridine)(dipyridophenazine)(H2O)]2+ (Ru(tpy)DPPZ) in azobenzene-modified mesoporous silica coated NaGdF4:Nd0.01/Yb0.2/Tm0.01 upconversion nanoparticles (azo-mSiO2-UCNPs). Upon 808 nm excitation, the generated ultraviolet and blue upconversion luminescence induced a reversible cis-trans isomerization of azobenzene for on-demand release of Ru(tpy)DPPZ. Imaging of both the UCNPs and Ru(tpy)DPPZ revealed targeted drug delivery to the nucleus of MCF-7 breast cancer cells, inducing DNA damage and concomitant cell destruction. Considering that cell nuclei are the core of cellular transcription and the main site of action for multiple chemotherapeutic drugs, our NIR-excitable and small (10 nm diameter) nanohybrids can potentially become highly versatile tools for targeted cancer theranostics.
Collapse
Affiliation(s)
- Anne Nsubuga
- Univ Rouen, CNRS, INSA Rouen, Normandie Université, Laboratoire COBRA, 76000 Rouen, France.
| | - Nour Fayad
- Univ Rouen, CNRS, INSA Rouen, Normandie Université, Laboratoire COBRA, 76000 Rouen, France.
| | - Federico Pini
- Univ Rouen, CNRS, INSA Rouen, Normandie Université, Laboratoire COBRA, 76000 Rouen, France.
- Istituto di Chimica della Materia Condensata e Tecnologie per l'Energia (ICMATE), Consiglio Nazionale delle Ricerche (CNR), Dipartimento di Scienze Chimiche, Università di Padova, 35131 Padova, Italy
| | - Marta M Natile
- Istituto di Chimica della Materia Condensata e Tecnologie per l'Energia (ICMATE), Consiglio Nazionale delle Ricerche (CNR), Dipartimento di Scienze Chimiche, Università di Padova, 35131 Padova, Italy
| | - Niko Hildebrandt
- McMaster University, Department of Engineering Physics, Hamilton, ON M8S 4K1, Canada.
| |
Collapse
|
4
|
Wang C, Xiu Y, Zhang Y, Wang Y, Xu J, Yu W, Xing D. Recent advances in biotin-based therapeutic agents for cancer therapy. NANOSCALE 2025; 17:1812-1873. [PMID: 39676680 DOI: 10.1039/d4nr03729d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Biotin receptors, as biomarkers for cancer cells, are overexpressed in various tumor types. Compared to other vitamin receptors, such as folate receptors and vitamin B12 receptors, biotin receptor-based targeting strategies exhibit superior specificity and broader potential in treating aggressive cancers, including ovarian cancer, leukemia, colon cancer, breast cancer, kidney cancer, and lung cancer. These strategies promote biotin transport via receptor-mediated endocytosis, which is triggered upon ligand binding. Biotin, as the ligand of the biotin receptor, can be conjugated to anti-cancer drugs to form targeted therapies that bind to receptors overexpressed on tumor cells, thus increasing drug uptake. Despite these advantages, many candidate drugs have progressed slowly and remain in the preclinical stage, impeding clinical translation. This is mainly due to the effects of various conjugation methods and drug formulations on their functionality and efficacy. Therefore, developing novel biotin-based therapeutics is crucial. The innovation of this strategy lies in its multifunctionality-researchers can use different conjugation methods to design and synthesize these drugs, enabling precise targeting of various tumor types while minimizing toxicity to normal cells. These drugs include small-molecule-biotin conjugates (SMBCs) and nano-biotin conjugates (NBCs). This dual-platform approach represents a significant advancement in targeted therapy, offering unprecedented flexibility in drug design and delivery. Compared to chemotherapy drugs and traditional delivery systems, biotin-based drugs with tumor-specific targeting demonstrate enhanced targeting, improved efficacy, and reduced toxicity. This review examines strategies and applications for enhancing the delivery of chemotherapy drugs to cancer cells, highlighting the need for high-quality conjugates and strategies. It not only summarizes the latest progress but also provides key insights into how this emerging field could revolutionize personalized cancer treatment, especially in the context of precision medicine. Additionally, it offers perspectives on future research directions in this field.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Yutao Xiu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Jiazhen Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, 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
|
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
|
Mariano S, Carata E, Calcagnile L, Panzarini E. Recent Advances in Photodynamic Therapy: Metal-Based Nanoparticles as Tools to Improve Cancer Therapy. Pharmaceutics 2024; 16:932. [PMID: 39065629 PMCID: PMC11280090 DOI: 10.3390/pharmaceutics16070932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer remains a significant global health challenge, with traditional therapies like surgery, chemotherapy, and radiation often accompanied by systemic toxicity and damage to healthy tissues. Despite progress in treatment, these approaches have limitations such as non-specific targeting, systemic toxicity, and resistance development in cancer cells. In recent years, nanotechnology has emerged as a revolutionary frontier in cancer therapy, offering potential solutions to these challenges. Nanoparticles, due to their unique physical and chemical properties, can carry therapeutic payloads, navigate biological barriers, and selectively target cancer cells. Metal-based nanoparticles, in particular, offer unique properties suitable for various therapeutic applications. Recent advancements have focused on the integration of metal-based nanoparticles to enhance the efficacy and precision of photodynamic therapy. Integrating nanotechnology into cancer therapy represents a paradigm shift, enabling the development of strategies with enhanced specificity and reduced off-target effects. This review aims to provide a comprehensive understanding of the pivotal role of metal-based nanoparticles in photodynamic therapy. We explore the mechanisms, biocompatibility, and applications of metal-based nanoparticles in photodynamic therapy, highlighting the challenges and the limitations in their use, as well as the combining of metal-based nanoparticles/photodynamic therapy with other strategies as a synergistic therapeutic approach for cancer treatment.
Collapse
Affiliation(s)
- Stefania Mariano
- Department of Mathematics and Physics, University of Salento, 73100 Lecce, Italy; (S.M.); (L.C.)
| | - Elisabetta Carata
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Lucio Calcagnile
- Department of Mathematics and Physics, University of Salento, 73100 Lecce, Italy; (S.M.); (L.C.)
- CEDAD (CEntre of Applied Physics, DAtation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 73100 Lecce, Italy
| | - Elisa Panzarini
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| |
Collapse
|
8
|
Zhou M, Wang Y, Xia Y, Li Y, Bao J, Zhang Y, Cheng J, Shi Y. MRI-guided cell membrane-camouflaged bimetallic coordination nanoplatform for combined tumor phototherapy. Mater Today Bio 2024; 26:101019. [PMID: 38516170 PMCID: PMC10950690 DOI: 10.1016/j.mtbio.2024.101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 03/23/2024] Open
Abstract
Nanotechnology for tumor diagnosis and optical therapy has attracted widespread interest due to its low toxicity and convenience but is severely limited due to uncontrollable tumor targeting. In this work, homologous cancer cell membrane-camouflaged multifunctional hybrid metal coordination nanoparticles (DRu/Gd@CM) were prepared for MRI-guided photodynamic therapy (PDT) and photothermal therapy (PTT) of tumors. Bimetallic coordination nanoparticles are composed of three functional modules: dopamine, Ru(dcbpy)3Cl2 and GdCl3, which are connected through 1,4-Bis[(1H-imidazole-1-yl)methyl]benzene (BIX). Their morphology can be easily controlled by adjusting the ratio of precursors. Optimistically, the intrinsic properties of the precursors, including the photothermal properties of polydopamine (PDA), the magnetic resonance (MR) response of Gd3+, and the singlet oxygen generation of Ru(dcbpy)3Cl2, are well preserved in the hybrid metal nanoparticles. Furthermore, the targeting of homologous cancer cell membranes enables these coordinated nanoparticles to precisely target tumor cells. The MR imaging capabilities and the combination of PDT and PTT were demonstrated in in vitro experiments. In addition, in vivo experiments indicated that the nanoplatform showed excellent tumor accumulation and therapeutic effects on mice with subcutaneous tumors, and could effectively eliminate tumors within 14 days. Therefore, it expanded the new horizon for the preparation of modular nanoplatform and imaging-guided optical therapy of tumors.
Collapse
Affiliation(s)
| | | | - Yaning Xia
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Yinhua Li
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Jianfeng Bao
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Yong Zhang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Yupeng Shi
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| |
Collapse
|
9
|
Wang H, Lai Y, Li D, Karges J, Zhang P, Huang H. Self-Assembly of Erlotinib-Platinum(II) Complexes for Epidermal Growth Factor Receptor-Targeted Photodynamic Therapy. J Med Chem 2024; 67:1336-1346. [PMID: 38183413 DOI: 10.1021/acs.jmedchem.3c01889] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2024]
Abstract
Due to cell mutation and self-adaptation, the application of clinical drugs with early epidermal growth factor receptor (EGFR)-targeted inhibitors is severely limited. To overcome this limitation, herein, the synthesis and in-depth biological evaluation of an erlotinib-platinum(II) complex as an EGFR-targeted anticancer agent is reported. The metal complex is able to self-assemble inside an aqueous solution and readily form nanostructures with strong photophysical properties. While being poorly toxic toward healthy cells and upon treatment in the dark, the compound was able to induce a cytotoxic effect in the very low micromolar range upon irradiation against EGFR overexpressing (drug resistant) human lung cancer cells as well as multicellular tumor spheroids. Mechanistic insights revealed that the compound was able to selectively degrade the EGFR using the lysosomal degradation pathway upon generation of singlet oxygen at the EGFR. We are confident that this work will open new avenues for the treatment of EGFR-overexpressing tumors.
Collapse
Affiliation(s)
- Haobing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yidan Lai
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Science, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Dan Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Bochum 44780, Germany
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Huaiyi Huang
- School of Pharmaceutical Science, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
10
|
Chen Q, Yuan L, Chou WC, Cheng YH, He C, Monteiro-Riviere NA, Riviere JE, Lin Z. Meta-Analysis of Nanoparticle Distribution in Tumors and Major Organs in Tumor-Bearing Mice. ACS NANO 2023; 17:19810-19831. [PMID: 37812732 PMCID: PMC10604101 DOI: 10.1021/acsnano.3c04037] [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: 05/05/2023] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
Low tumor delivery efficiency is a critical barrier in cancer nanomedicine. This study reports an updated version of "Nano-Tumor Database", which increases the number of time-dependent concentration data sets for different nanoparticles (NPs) in tumors from the previous version of 376 data sets with 1732 data points from 200 studies to the current version of 534 data sets with 2345 data points from 297 studies published from 2005 to 2021. Additionally, the current database includes 1972 data sets for five major organs (i.e., liver, spleen, lung, heart, and kidney) with a total of 8461 concentration data points. Tumor delivery and organ distribution are calculated using three pharmacokinetic parameters, including delivery efficiency, maximum concentration, and distribution coefficient. The median tumor delivery efficiency is 0.67% injected dose (ID), which is low but is consistent with previous studies. Employing the best regression model for tumor delivery efficiency, we generate hypothetical scenarios with different combinations of NP factors that may lead to a higher delivery efficiency of >3%ID, which requires further experimentation to confirm. In healthy organs, the highest NP accumulation is in the liver (10.69%ID/g), followed by the spleen 6.93%ID/g and the kidney 3.22%ID/g. Our perspective on how to facilitate NP design and clinical translation is presented. This study reports a substantially expanded "Nano-Tumor Database" and several statistical models that may help nanomedicine design in the future.
Collapse
Affiliation(s)
- Qiran Chen
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Long Yuan
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Wei-Chun Chou
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Yi-Hsien Cheng
- Department
of Anatomy and Physiology, Kansas State
University, Manhattan, Kansas 66506, United States
- Institute
of Computational Comparative Medicine, Kansas
State University, Manhattan, Kansas 66506, United States
| | - Chunla He
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Department
of Biostatistics College of Public Health and Health Professions, University of Florida, Gainesville, Florida 32608, United States
| | - Nancy A. Monteiro-Riviere
- Nanotechnology
Innovation Center of Kansas State, Kansas
State University, Manhattan, Kansas 66506, United States
- Center
for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Jim E. Riviere
- Center
for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, North Carolina 27606, United States
- 1
Data Consortium, Kansas State University, Olathe, Kansas 66061, United States
| | - Zhoumeng Lin
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
11
|
Bera A, Nepalia A, Upadhyay A, Kumar Saini D, Chakravarty AR. Biotin and boron-dipyrromethene-tagged platinum(IV) prodrug for cellular imaging and mito-targeted photocytotoxicity in red light. Dalton Trans 2023; 52:13339-13350. [PMID: 37671587 DOI: 10.1039/d3dt01796f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
A platinum(IV) prodrug, cis,cis,trans-[Pt(NH3)2Cl2(biotin)(L)] (1), derived from cisplatin, where HL is the PEGylated red-light active boron-dipyrromethene (BODIPY) ligand, was synthesized, characterized and its photocytotoxicity evaluated. The complex showed a near-IR absorption band at 653 nm (ε ∼9.19 × 104 M-1 cm-1) in dimethyl sulfoxide and Dulbecco's phosphate-buffered saline (1 : 1 v/v) at pH 7.2. When excited at 630 nm, it showed an emission band at 677 nm in DMSO with a fluorescence quantum yield of 0.13. The 1,3-diphenylisobenzofuran titration experiment gave a singlet oxygen quantum yield (ΦΔ) of ∼0.32. A mechanistic DNA photocleavage study revealed singlet oxygen as the reactive oxygen species (ROS). The complex with biotin and PEGylated-distyryl-BODIPY showed significantly higher cellular uptake in A549 cancer cells as compared to non-cancerous Beas-2B cells from flow cytometry, indicating selectivity towards cancer cells. A dichlorodihydrofluorescein diacetate assay showed cellular ROS generation. Confocal images revealed predominant internalization in the mitochondria. The prodrug showed remarkable photodynamic therapy (PDT) activity in cancerous A549 and multidrug-resistant MDA-MB-231 cells with a high photocytotoxicity index value (half-maximal inhibitory concentration (IC50): 0.61-1.54 μM in red light), while being non-toxic in the dark. The chemo-PDT activity was significantly less in non-tumorigenic lung epithelial cells (Beas-2B). The prodrug effectively triggered cellular apoptosis, which was confirmed by the Annexin V-FITC/propidium iodide assay, and the alteration of the mitochondrial membrane potential was substantiated by the JC-1 dye assay. The β-tubulin immunofluorescence assay confirmed that incubating the cells with a light-treated complex resulted in the rapture of the cytoskeletal structure and the formation of apoptotic bodies. The results demonstrate that the prodrug triggered apoptosis via DNA damage, a reduction in mitochondrial function and disruption of the cytoskeletal framework.
Collapse
Affiliation(s)
- Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Amrita Nepalia
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India.
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Deepak Kumar Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India.
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
12
|
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
|
13
|
Zhou H, Tang D, Yu Y, Zhang L, Wang B, Karges J, Xiao H. Theranostic imaging and multimodal photodynamic therapy and immunotherapy using the mTOR signaling pathway. Nat Commun 2023; 14:5350. [PMID: 37660174 PMCID: PMC10475087 DOI: 10.1038/s41467-023-40826-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/11/2023] [Indexed: 09/04/2023] Open
Abstract
Tumor metastases are considered the leading cause of cancer-associated deaths. While clinically applied drugs have demonstrated to efficiently remove the primary tumor, metastases remain poorly accessible. To overcome this limitation, herein, the development of a theranostic nanomaterial by incorporating a chromophore for imaging and a photosensitizer for treatment of metastatic tumor sites is presented. The mechanism of action reveals that the nanoparticles are able to intervene by local generation of cellular damage through photodynamic therapy as well as by systemic induction of an immune response by immunotherapy upon inhibition of the mTOR signaling pathway which is of crucial importance for tumor onset, progression and metastatic spreading. The nanomaterial is able to strongly reduce the volume of the primary tumor as well as eradicates tumor metastases in a metastatic breast cancer and a multi-drug resistant patient-derived hepatocellular carcinoma models in female mice.
Collapse
Affiliation(s)
- Huiling Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, P.R. China
| | - Lingpu Zhang
- College of Life Science and Technology and State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Bin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, Bochum, 44780, Germany.
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China.
| |
Collapse
|
14
|
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: 6.5] [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
|
15
|
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: 48] [Impact Index Per Article: 24.0] [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
|
16
|
Wei F, Chen Z, Shen XC, Ji L, Chao H. Recent progress in metal complexes functionalized nanomaterials for photodynamic therapy. Chem Commun (Camb) 2023. [PMID: 37184685 DOI: 10.1039/d3cc01355c] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Metal complexes have shown promise as photosensitizers for cancer diagnosis and therapeutics. However, the vast majority of metal photosensitizers are not ideal and associated with several limitations including pharmacokinetic limitations, off-target toxicity, fast systemic clearance, poor membrane permeability, and hypoxic tumour microenvironments. Metal complex functionalized nanomaterials have the potential to construct multifunctional systems, which not only overcome the above defects of metal complexes but are also conducive to modulating the tumour microenvironment (TME) and employing combination therapies to boost photodynamic therapy (PDT) efficacy. In this review, we first introduce the current challenges of photodynamic therapy and summarize the recent research strategies (such as metal coordination bonds, self-assembly, π-π stacking, physisorption, and so on) used for preparing metal complexes functionalized nanomaterials in the application of PDT.
Collapse
Affiliation(s)
- Fangmian Wei
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China.
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, MOE Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China.
| | - Zhuoli Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China.
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, MOE Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China.
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China.
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China.
| |
Collapse
|
17
|
Zhang L, Zhu L, Tang L, Xie J, Gao Y, Yu C, Shang K, Han H, Liu C, Lu Y. Glutathione-Responsive Nanoparticles of Camptothecin Prodrug for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205246. [PMID: 36442854 PMCID: PMC9875659 DOI: 10.1002/advs.202205246] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/17/2022] [Indexed: 05/28/2023]
Abstract
Camptothecin (CPT) is a potent chemotherapeutic agent for various cancers, but the broader application of CPT is still hindered by its poor bioavailability and systemic toxicity. Here, a prodrug that releases CPT in response to glutathione (GSH), which is commonly overexpressed by cancer cells is reported. Through assembling with PEGylated lipids, the prodrug is incorporated within as-assembled nanoparticles, affording CPT with a prolonged half-life in blood circulation, enhanced tumor targetingability, and improved therapeutic efficacy. Furthermore, such prodrug nanoparticles can also promote dendritic cell maturation and tumor infiltration of CD8+ T cells, providing a novel strategy to improve the therapeutic efficacy of CPT.
Collapse
Affiliation(s)
- Lingpu Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lin Zhu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lin Tang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Jiayi Xie
- Department of AutomaticTsinghua UniversityPeking University Third HospitalBeijing Key Laboratory of Magnetic Resonance Imaging Devices and TechnologyBeijing100191P. R. China
| | - Yajuan Gao
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Changyuan Yu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Kun Shang
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Hongbin Han
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Yunfeng Lu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| |
Collapse
|
18
|
Recent advances on organelle specific Ru(II)/Ir(III)/Re(I) based complexes for photodynamic therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214860] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Lu Y, Zhu D, Le Q, Wang Y, Wang W. Ruthenium-based antitumor drugs and delivery systems from monotherapy to combination therapy. NANOSCALE 2022; 14:16339-16375. [PMID: 36341705 DOI: 10.1039/d2nr02994d] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Ruthenium complex is an important compound group for antitumor drug research and development. NAMI-A, KP1019, TLD1433 and other ruthenium complexes have entered clinical research. In recent years, the research on ruthenium antitumor drugs has not been limited to single chemotherapy drugs; other applications of ruthenium complexes have emerged such as in combination therapy. During the development of ruthenium complexes, drug delivery forms of ruthenium antitumor drugs have also evolved from single-molecule drugs to nanodrug delivery systems. The review summarizes the following aspects: (1) ruthenium complexes from monotherapy to combination therapy, including the development of single-molecule compounds, carrier nanomedicine, and self-assembly of carrier-free nanomedicine; (2) ruthenium complexes in the process of ADME in terms of absorption, distribution, metabolism and excretion; (3) the applications of ruthenium complexes in combination therapy, including photodynamic therapy (PDT), photothermal therapy (PTT), photoactivated chemotherapy (PACT), immunotherapy, and their combined application; (4) the future prospects of ruthenium-based antitumor drugs.
Collapse
Affiliation(s)
- Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
- Department of Chemistry, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway
| | - Di Zhu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
| | - Quynh Le
- Center for Pharmacy, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway.
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
| | - Wei Wang
- Center for Pharmacy, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway.
- Department of Chemistry, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway
| |
Collapse
|
20
|
Gandioso A, Vidal A, Burckel P, Gasser G, Alessio E. Ruthenium(II) Polypyridyl Complexes Containing Simple Dioxo Ligands: a Structure-Activity Relationship Study Shows the Importance of the Charge. Chembiochem 2022; 23:e202200398. [PMID: 35924883 DOI: 10.1002/cbic.202200398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/04/2022] [Indexed: 01/07/2023]
Abstract
Cancer is one of the main causes of death worldwide. Platinum complexes (i. e., cisplatin, carboplatin, and others) are currently heavily used for the treatment of different types of cancer, but unwanted effects occur. Ruthenium complexes have been shown to be potential promising alternatives to these metal-based drugs. In this work, we performed a structure-activity relationship (SAR) study on two small series of Ru(II) polypyridyl complexes of the type [Ru(L1)2 (O^O)]Cln (3-8), where L1 is 4,7-diphenyl-1,10-phenantroline (DIP) or 1,10-phenantroline (phen), and O^O is a symmetrical anionic dioxo ligand: oxalate (ox, n=0), malonate (mal, n=0), or acetylacetonate (acac, n=1). These two self-consistent series of compounds allowed us to perform a systematic investigation for establishing how the nature of the ligands and the charge affect the anticancer properties of the complexes. Cytotoxicity tests on different cell lines demonstrated that some of the six compounds 3-8 have a promising anticancer activity. More specifically, the cationic complex [Ru(DIP)2 (η2 -acac)]Cl (4) has IC50 values in the mid-nanomolar concentration range, lower than those of cisplatin on the same cell lines. Interestingly, [Ru(DIP)2 (η2 -acac)]Cl was found to localize mainly in the mitochondria, whereas a smaller fraction was detected in the nucleus. Overall, our SAR investigation demonstrates the importance of combining the positive charge of the complex with the highly lipophilic diimine ligand DIP.
Collapse
Affiliation(s)
- Albert Gandioso
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Alessio Vidal
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127, Trieste, Italy
| | - Pierre Burckel
- Université de Paris, Institut de physique du globe de Paris, CNRS, 75005, Paris, France.,Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-, 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
| | - Enzo Alessio
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127, Trieste, Italy
| |
Collapse
|
21
|
Kar B, Paira P. One pot three component synthesis of DNA targeting phototoxic Ru(II)- p-cymene dipyrido[3,2- a:2',3'- c]phenazine analogues. Dalton Trans 2022; 51:15686-15695. [PMID: 36173180 DOI: 10.1039/d2dt01659a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have developed a one pot three component synthetic protocol for half-sandwich Ru(II)-p-cymene dipyrido[3,2-a:2',3'-c]phenazine analogues for selective cancer therapy under light irradiation. On average, the cytotoxicity of all the complexes is indeed doubled upon light irradiation and also exhibited significant photo and dark selectivity against cancer cells with respect to normal cells. Out of five Ru(II) complexes (RuL1-RuL5), [(η6-p-cymene)RuIICl(K2-N,N-11-nitrodipyrido[3,2-a:2',3'-c]phenazine]PF6 (RuL4) exhibited the best phototoxicity (lowest IC50 under light irradiation). Intracellular ROS generation was studied by the 2',7'-dichlorofluorescein diacetate (DCFH-DA) assay. Moreover, these complexes exhibited a strong serum albumin and DNA binding capacity. These complexes also exhibited good stability in 10% DMSO-buffer and under 1 mM GSH conditions. Overall, the remarkable photocytotoxic efficacy of new Ru(II)-p-cymene dipyrido[3,2-a:2',3'-c]phenazine analogues (RuL1-RuL5) makes them potential photochemotherapeutics as an alternative of current PDT agents.
Collapse
Affiliation(s)
- Binoy Kar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| |
Collapse
|
22
|
Vinck R, Gandioso A, Burckel P, Saubaméa B, Cariou K, Gasser G. Red-Absorbing Ru(II) Polypyridyl Complexes with Biotin Targeting Spontaneously Assemble into Nanoparticles in Biological Media. Inorg Chem 2022; 61:13576-13585. [PMID: 35960605 DOI: 10.1021/acs.inorgchem.2c02214] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Four new ruthenium(II) polypyridyl complexes were synthesized to study the effect of poly(ethylene glycol) and/or biotin conjugation on their physical and biological properties, including their hydrophilicity, their cellular uptake, and their phototoxicity. Unexpectedly, these complexes self-assembled into nanoparticles upon dilution in biological media. This behavior leads to their accumulation in lysosomes following their internalization by cells. While a significant increase in cellular uptake was observed for the biotin-conjugated complexes, it did not result in an increase in their phototoxicity. However, their high phototoxicity upon irradiation at long wavelengths (645-670 nm) and their self-assembling behavior make them a promising backbone for the development of new lysosome-targeted photosensitizers for photodynamic therapy.
Collapse
Affiliation(s)
- Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Albert Gandioso
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Pierre Burckel
- Institut de Physique du Globe de Paris, Biogéochimie à l'Anthropocène des Eléments et Contaminants Emergents, 75005 Paris, France
| | - Bruno Saubaméa
- Cellular and Molecular Imaging platform, US 25 Inserm, UMS 3612 CNRS, Faculté de Pharmacie de Paris, Université Paris Cité, 75006 Paris, France
| | - 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
|
23
|
Polymeric Nanosystems Applied for Metal-Based Drugs and Photosensitizers Delivery: The State of the Art and Recent Advancements. Pharmaceutics 2022; 14:pharmaceutics14071506. [PMID: 35890401 PMCID: PMC9320085 DOI: 10.3390/pharmaceutics14071506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/03/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Nanotechnology-based approaches for targeting the delivery and controlled release of metal-based therapeutic agents have revealed significant potential as tools for enhancing the therapeutic effect of metal-based agents and minimizing their systemic toxicities. In this context, a series of polymer-based nanosized systems designed to physically load or covalently conjugate metal-based therapeutic agents have been remarkably improving their bioavailability and anticancer efficacy. Initially, the polymeric nanocarriers were applied for platinum-based chemotherapeutic agents resulting in some nanoformulations currently in clinical tests and even in medical applications. At present, these nanoassemblies have been slowly expanding for nonplatinum-containing metal-based chemotherapeutic agents. Interestingly, for metal-based photosensitizers (PS) applied in photodynamic therapy (PDT), especially for cancer treatment, strategies employing polymeric nanocarriers have been investigated for almost 30 years. In this review, we address the polymeric nanocarrier-assisted metal-based therapeutics agent delivery systems with a specific focus on non-platinum systems; we explore some biological and physicochemical aspects of the polymer–metallodrug assembly. Finally, we summarize some recent advances in polymeric nanosystems coupled with metal-based compounds that present potential for successful clinical applications as chemotherapeutic or photosensitizing agents. We hope this review can provide a fertile ground for the innovative design of polymeric nanosystems for targeting the delivery and controlled release of metal-containing therapeutic agents.
Collapse
|
24
|
Côrte-Real L, Brás AR, Pilon A, Mendes N, Ribeiro AS, Martins TD, Farinha JPS, Oliveira MC, Gärtner F, Garcia MH, Preto A, Valente A. Biotinylated Polymer-Ruthenium Conjugates: In Vitro and In Vivo Studies in a Triple-Negative Breast Cancer Model. Pharmaceutics 2022; 14:1388. [PMID: 35890283 PMCID: PMC9315599 DOI: 10.3390/pharmaceutics14071388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/15/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022] Open
Abstract
The need for new therapeutic approaches for triple-negative breast cancer is a clinically relevant problem that needs to be solved. Using a multi-targeting approach to enhance cancer cell uptake, we synthesized a new family of ruthenium(II) organometallic complexes envisaging simultaneous active and passive targeting, using biotin and polylactide (PLA), respectively. All compounds with the general formula, [Ru(η5-CpR)(P)(2,2'-bipy-4,4'-PLA-biotin)][CF3SO3], where R is -H or -CH3 and P is P(C6H5)3, P(C6H4F)3 or P(C6H4OCH3)3, were tested against triple-negative breast cancer cells MDA-MB-231 showing IC50 values between 2.3-14.6 µM, much better than cisplatin, a classical chemotherapeutic drug, in the same experimental conditions. We selected compound 1 (where R is H and P is P(C6H5)3), for further studies as it was the one showing the best biological effect. In a competitive assay with biotin, we showed that cell uptake via SMVT receptors seems to be the main transport route into the cells for this compound, validating the strategy of including biotin in the design of the compound. The effects of the compound on the hallmarks of cancer show that the compound leads to apoptosis, interferes with proliferation by affecting the formation of cell colonies in a dose-dependent manner and disrupts the cell cytoskeleton. Preliminary in vivo assays in N: NIH(S)II-nu/nu mice show that the concentrations of compound 1 used in this experiment (maximum 4 mg/kg) are safe to use in vivo, although some signs of liver toxicity are already found. In addition, the new compound shows a tendency to control tumor growth, although not significantly. In sum, we showed that compound 1 shows promising anti-cancer effects, bringing a new avenue for triple-negative breast cancer therapy.
Collapse
Affiliation(s)
- Leonor Côrte-Real
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| | - Ana Rita Brás
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Edifício 18, 4710-057 Braga, Portugal
| | - Adhan Pilon
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| | - Nuno Mendes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (N.M.); (A.S.R.); (F.G.)
| | - Ana Sofia Ribeiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (N.M.); (A.S.R.); (F.G.)
| | - Tiago D. Martins
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.D.M.); (J.P.S.F.); (M.C.O.)
| | - José Paulo S. Farinha
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.D.M.); (J.P.S.F.); (M.C.O.)
| | - M. Conceição Oliveira
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.D.M.); (J.P.S.F.); (M.C.O.)
| | - Fátima Gärtner
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (N.M.); (A.S.R.); (F.G.)
| | - M. Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| | - Ana Preto
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Edifício 18, 4710-057 Braga, Portugal
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| |
Collapse
|
25
|
An J, Tang S, Hong G, Chen W, Chen M, Song J, Li Z, Peng X, Song F, Zheng WH. An unexpected strategy to alleviate hypoxia limitation of photodynamic therapy by biotinylation of photosensitizers. Nat Commun 2022; 13:2225. [PMID: 35469028 PMCID: PMC9038921 DOI: 10.1038/s41467-022-29862-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/04/2022] [Indexed: 01/25/2023] Open
Abstract
The most common working mechanism of photodynamic therapy is based on high-toxicity singlet oxygen, which is called Type II photodynamic therapy. But it is highly dependent on oxygen consumption. Recently, Type I photodynamic therapy has been found to have better hypoxia tolerance to ease this restriction. However, few strategies are available on the design of Type I photosensitizers. We herein report an unexpected strategy to alleviate the limitation of traditional photodynamic therapy by biotinylation of three photosensitizers (two fluorescein-based photosensitizers and the commercially available Protoporphyrin). The three biotiylated photosensitizers named as compound 1, 2 and 3, exhibit impressive ability in generating both superoxide anion radicals and singlet oxygen. Moreover, compound 1 can be activated upon low-power white light irradiation with stronger ability of anion radicals generation than the other two. The excellent combinational Type I / Type II photodynamic therapy performance has been demonstrated with the photosensitizers 1. This work presents a universal protocol to provide tumor-targeting ability and enhance or trigger the generation of anion radicals by biotinylation of Type II photosensitizers against tumor hypoxia. Type I photodynamic therapy (PDT) sensitizers show good hypoxia tolerance but only few strategies are available for the design of purely organic Type I photosensitizers (PS). Here, the authors use biotinylation as design strategy to obtain PS-Biotin sensitizers with high efficiency for the generation of superoxide anion radicals and singlet oxygen.
Collapse
Affiliation(s)
- Jing An
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Shanliang Tang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Gaobo Hong
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Wenlong Chen
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Miaomiao Chen
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Jitao Song
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, 266237, Qingdao, China
| | - Zhiliang Li
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, 266237, Qingdao, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Fengling Song
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China. .,Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, 266237, Qingdao, China.
| | - Wen-Heng Zheng
- Department of Interventional Therapy, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, 110042, Shenyang, China.
| |
Collapse
|
26
|
Feng T, Karges J, Liao X, Ji L, Chao H. Engineered exosomes as a natural nanoplatform for cancer targeted delivery of metal-based drugs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
Kuang S, Wei F, Karges J, Ke L, Xiong K, Liao X, Gasser G, Ji L, Chao H. Photodecaging of a Mitochondria-Localized Iridium(III) Endoperoxide Complex for Two-Photon Photoactivated Therapy under Hypoxia. J Am Chem Soc 2022; 144:4091-4101. [PMID: 35171598 DOI: 10.1021/jacs.1c13137] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite the clinical success of photodynamic therapy (PDT), the application of this medical technique is intrinsically limited by the low oxygen concentrations found in cancer tumors, hampering the production of therapeutically necessary singlet oxygen (1O2). To overcome this limitation, we report on a novel mitochondria-localized iridium(III) endoperoxide prodrug (2-O-IrAn), which, upon two-photon irradiation in NIR, synergistically releases a highly cytotoxic iridium(III) complex (2-IrAn), singlet oxygen, and an alkoxy radical. 2-O-IrAn was found to be highly (photo-)toxic in hypoxic tumor cells and multicellular tumor spheroids (MCTS) in the nanomolar range. To provide cancer selectivity and improve the pharmacological properties of 2-O-IrAn, it was encapsulated into a biotin-functionalized polymer. The generated nanoparticles were found to nearly fully eradicate the tumor inside a mouse model within a single treatment. This study presents, to the best of our knowledge, the first example of an iridium(III)-based endoperoxide prodrug for synergistic photodynamic therapy/photoactivated chemotherapy, opening up new avenues for the treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Shi Kuang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Fangmian Wei
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Johannes Karges
- Department of Chemistry and Biochemistry, University of California, 9500 Gilman Drive, La Jolla, San Diego, California 92093, United States
| | - Libing Ke
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Xinxing Liao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China.,MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 400201, P. R. China
| |
Collapse
|
28
|
Karges J. Clinical Development of Metal Complexes as Photosensitizers for Photodynamic Therapy of Cancer. Angew Chem Int Ed Engl 2022; 61:e202112236. [PMID: 34748690 DOI: 10.1002/anie.202112236] [Citation(s) in RCA: 199] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 12/12/2022]
Abstract
Cancer has emerged over the last decades as one of the deadliest diseases in the world. Among the most commonly used techniques (i.e. surgery, immunotherapy, radiotherapy or chemotherapy), increasing attention has been devoted towards photodynamic therapy. However, the vast majority of clinically applied photosensitizers are not ideal and associated with several limitations including poor aqueous solubility, poor photostability and slow clearance from the body, causing photosensitivity. In an effort to overcome these drawbacks, much attention has been devoted towards the incorporation of a metal ion. Herein, the clinical development of metal-containing compounds including Purlytin® , Lutrin® /Antrin® , Photosens® , TOOKAD® soluble or TLD-1433 is critically reviewed.
Collapse
Affiliation(s)
- Johannes Karges
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| |
Collapse
|
29
|
Karges J. Klinische Entwicklung von Metallkomplexen als Photosensibilisatoren für die photodynamische Therapie von Krebs. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202112236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Johannes Karges
- Department of Chemistry and Biochemistry University of California, San Diego 9500 Gilman Drive La Jolla CA 92093 USA
| |
Collapse
|
30
|
McKenzie LK, Flamme M, Felder PS, Karges J, Bonhomme F, Gandioso A, Malosse C, Gasser G, Hollenstein M. A ruthenium-oligonucleotide bioconjugated photosensitizing aptamer for cancer cell specific photodynamic therapy. RSC Chem Biol 2022; 3:85-95. [PMID: 35128412 PMCID: PMC8729177 DOI: 10.1039/d1cb00146a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/31/2021] [Indexed: 12/15/2022] Open
Abstract
Ruthenium complexes have emerged as a promising class of compounds for use as photosensitizers (PSs) in photodynamic therapy (PDT) due to their attractive photophysical properties and relative ease of chemical alteration. While promising, they generally are not inherently targeting to disease sites and may therefore be prone to side effects and require higher doses. Aptamers are short oligonucleotides that bind specific targets with high affinity. One such aptamer is AS1411, a nucleolin targeting, G-quadruplex forming, DNA aptamer. Here we present the first example of direct conjugation of a Ru(ii) polypyridyl complex-based PS to an aptamer and an assessment of its in vitro cancer cell specific photosensitization including discussion of the challenges faced.
Collapse
Affiliation(s)
- Luke K McKenzie
- Institut Pasteur, Department of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, CNRS UMR3523 28 rue du Docteur Roux 75724 Paris Cedex 15 France https://research.pasteur.fr/en/team/bioorganic-chemistry-of-nucleic-acids/ +33 1 44 38 94 66
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France www.gassergroup.com +33 1 85 78 41 51
| | - Marie Flamme
- Institut Pasteur, Department of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, CNRS UMR3523 28 rue du Docteur Roux 75724 Paris Cedex 15 France https://research.pasteur.fr/en/team/bioorganic-chemistry-of-nucleic-acids/ +33 1 44 38 94 66
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France www.gassergroup.com +33 1 85 78 41 51
- Université de Paris 12 rue de l'École de Médecine 75006 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 www.gassergroup.com +33 1 85 78 41 51
| | - Johannes Karges
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France www.gassergroup.com +33 1 85 78 41 51
| | - Frederic Bonhomme
- Institut Pasteur, Department of Structural Biology and Chemistry, Unité de Chimie Biologique Epigénétique, UMR CNRS 3523 28 rue du Docteur Roux 75724 Paris Cedex 15 France
| | - Albert Gandioso
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France www.gassergroup.com +33 1 85 78 41 51
| | - Christian Malosse
- Institut Pasteur, Mass Spectrometry for Biology Unit 28 rue du Docteur Roux 75724 Paris Cedex 15 France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France www.gassergroup.com +33 1 85 78 41 51
| | - Marcel Hollenstein
- Institut Pasteur, Department of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, CNRS UMR3523 28 rue du Docteur Roux 75724 Paris Cedex 15 France https://research.pasteur.fr/en/team/bioorganic-chemistry-of-nucleic-acids/ +33 1 44 38 94 66
| |
Collapse
|
31
|
Chen Z, Feng T, jinchao S, Karges J, Jin C, Zhao Y, Ji L, Chao H. A Mitochondria-Localized Iridium(III)-Chlorin E6 Conjugate for Synergistic Sonodynamic and Two-Photon Photodynamic Therapy Against Melanoma. Inorg Chem Front 2022. [DOI: 10.1039/d2qi00635a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
While melanoma in its early stages can be successfully treated, the prognosis strongly worsens with an increasing depth of the tumor. Capitalizing on this, there is an urgent need for...
Collapse
|
32
|
Bonelli J, Ortega-Forte E, Vigueras G, Bosch M, Cutillas N, Rocas J, Ruiz J, Marchan V. Polyurethane-polyurea hybrid nanocapsules as efficient delivery systems of anticancer Ir(III) metallodrugs. Inorg Chem Front 2022. [DOI: 10.1039/d1qi01542g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cyclometalated Ir(III) complexes hold great promise as an alternative to platinum metallodrugs for therapy and diagnosis of cancer. However, low aqueous solubility and poor cell membrane permeability difficult in vivo...
Collapse
|
33
|
Photoactivable Ruthenium-Based Coordination Polymer Nanoparticles for Light-Induced Chemotherapy. NANOMATERIALS 2021; 11:nano11113089. [PMID: 34835853 PMCID: PMC8617783 DOI: 10.3390/nano11113089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022]
Abstract
Green light photoactive Ru-based coordination polymer nanoparticles (CPNs), with chemical formula [[Ru(biqbpy)]1.5(bis)](PF6)3 (biqbpy = 6,6'-bis[N-(isoquinolyl)-1-amino]-2,2'-bipyridine; bis = bis(imidazol-1-yl)-hexane), were obtained through polymerization of the trans-[Ru(biqbpy)(dmso)Cl]Cl complex (Complex 1) and bis bridging ligands. The as-synthesized CPNs (50 ± 12 nm diameter) showed high colloidal and chemical stability in physiological solutions. The axial bis(imidazole) ligands coordinated to the ruthenium center were photosubstituted by water upon light irradiation in aqueous medium to generate the aqueous substituted and active ruthenium complexes. The UV-Vis spectral variations observed for the suspension upon irradiation corroborated the photoactivation of the CPNs, while High Performance Liquid Chromatography (HPLC) of irradiated particles in physiological media allowed for the first time precisely quantifying the amount of photoreleased complex from the polymeric material. In vitro studies with A431 and A549 cancer cell lines revealed an 11-fold increased uptake for the nanoparticles compared to the monomeric complex [Ru(biqbpy)(N-methylimidazole)2](PF6)2 (Complex 2). After irradiation (520 nm, 39.3 J/cm2), the CPNs yielded up to a two-fold increase in cytotoxicity compared to the same CPNs kept in the dark, indicating a selective effect by light irradiation. Meanwhile, the absence of 1O2 production from both nanostructured and monomeric prodrugs concluded that light-induced cell death is not caused by a photodynamic effect but rather by photoactivated chemotherapy.
Collapse
|
34
|
Pham TC, Nguyen VN, Choi Y, Lee S, Yoon J. Recent Strategies to Develop Innovative Photosensitizers for Enhanced Photodynamic Therapy. Chem Rev 2021; 121:13454-13619. [PMID: 34582186 DOI: 10.1021/acs.chemrev.1c00381] [Citation(s) in RCA: 803] [Impact Index Per Article: 200.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review presents a robust strategy to design photosensitizers (PSs) for various species. Photodynamic therapy (PDT) is a photochemical-based treatment approach that involves the use of light combined with a light-activated chemical, referred to as a PS. Attractively, PDT is one of the alternatives to conventional cancer treatment due to its noninvasive nature, high cure rates, and low side effects. PSs play an important factor in photoinduced reactive oxygen species (ROS) generation. Although the concept of photosensitizer-based photodynamic therapy has been widely adopted for clinical trials and bioimaging, until now, to our surprise, there has been no relevant review article on rational designs of organic PSs for PDT. Furthermore, most of published review articles in PDT focused on nanomaterials and nanotechnology based on traditional PSs. Therefore, this review aimed at reporting recent strategies to develop innovative organic photosensitizers for enhanced photodynamic therapy, with each example described in detail instead of providing only a general overview, as is typically done in previous reviews of PDT, to provide intuitive, vivid, and specific insights to the readers.
Collapse
Affiliation(s)
- Thanh Chung Pham
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Yeonghwan Choi
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Songyi Lee
- Department of Chemistry, Pukyong National University, Busan 48513, Korea.,Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
35
|
Bjelosevic A, Sakoff J, Gilbert J, Zhang Y, McGhie B, Gordon C, Aldrich-Wright JR. Synthesis, characterisation and biological activity of the ruthenium(II) complexes of the N 4-tetradentate (N 4-T L), 1,6-di(2'-pyridyl)-2,5-dibenzyl-2,5-diazahexane (picenBz 2). J Inorg Biochem 2021; 226:111629. [PMID: 34740037 DOI: 10.1016/j.jinorgbio.2021.111629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/04/2021] [Accepted: 10/10/2021] [Indexed: 10/20/2022]
Abstract
A series of complexes of the type rac-cis-β-[Ru(N4-TL)(N2-bidentates)]2+ (where N4-TL = 1,6-di(2'-pyridyl)-2,5-dibenzyl-2,5-diazahexane (picenBz2, N4-TL-2) and N2-bidentates = 1,10-phenanthroline (phen, Ru-2), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq, Ru-3), 7,8-dimethyl-dipyrido[3,2-a:2',3'-c] phenazine (dppzMe2,Ru-4), 2-phenyl-1H-imidazo[4,5-f][1,10]phenanthroline (phenpyrBz, Ru-5), 2-(p-tolyl)-1H-imidazo[4,5-f][1,10]phenanthroline (phenpyrBzMe, Ru-6), 2-(4-nitrophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline (phenpyrBzNO2,Ru-7), were synthesised and characterised and X-ray crystallography of Ru-5 obtained. The in vitro cytotoxicity assays revealed that Ru-6 was 5, 2 and 19-fold more potent than oxaliplatin, cisplatin, and carboplatin, respectively displaying an average GI50 value of ≈ 0.76 μM against a panel of 11 cancer cell lines.
Collapse
Affiliation(s)
- Aleksandra Bjelosevic
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South DC, 2751, NSW, Australia
| | - Jennette Sakoff
- Calvary Mater Newcastle Hospital, Waratah, NSW 2298, Australia
| | - Jayne Gilbert
- Calvary Mater Newcastle Hospital, Waratah, NSW 2298, Australia
| | - Yingjie Zhang
- Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Brondwyn McGhie
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South DC, 2751, NSW, Australia
| | - Christopher Gordon
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South DC, 2751, NSW, Australia
| | - Janice R Aldrich-Wright
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South DC, 2751, NSW, Australia; School of Medicine, Western Sydney University, Locked Bag 1797, Penrith South DC, 2751, NSW, Australia.
| |
Collapse
|
36
|
Paul S, Kundu P, Kondaiah P, Chakravarty AR. BODIPY-Ruthenium(II) Bis-Terpyridine Complexes for Cellular Imaging and Type-I/-II Photodynamic Therapy. Inorg Chem 2021; 60:16178-16193. [PMID: 34672556 DOI: 10.1021/acs.inorgchem.1c01850] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of multichromophoric ruthenium(II) complexes with the formulation [Ru(tpy-BODIPY)(tpy-R)]Cl2 (1-4), having a heteroleptic Ru(II)-bis-tpy (tpy = 4'-phenyl-2,2':6',2″-terpyridine) moiety covalently linked to a boron-dipyrromethene (BODIPY) pendant, have been prepared and characterized and their application as a phototherapeutic and photodetection agent in cancer therapy has been explored. Ligand L1 with a terpyridine-BODIPY moiety and complex 1 as its PF6 salt (1a) have been structurally characterized by a single-crystal X-ray diffraction study. Complex 1a has a distorted-octahedral RuN6 core with a Ru(II)-bis-terpyridine unit that is covalently linked to one photoactive BODIPY unit. The complexes exhibit strong absorbance near 502 nm (ε ≈ (3.7-7.8) × 104 M-1 cm-1) and high singlet oxygen sensitization ability, giving singlet oxygen quantum yield (ΦΔ) values ranging from 0.57 to 0.75 in DMSO. An emission-based study using complex 4 and Singlet Oxygen Sensor Green (SOSG) displays the formation of singlet oxygen inside the cells and also in the buffer medium upon light irradiation. DNA (pUC19) photocleavage experiments using ROS scavengers/stabilizers reveal photoinduced generation of singlet oxygen by a type-II process and of the superoxide anion radical by a type-I process. Complex 4 having a pendant biotin moiety as a cancer cell targeting group shows high photocytotoxicity with a remarkable phototherapeutic index (PI) value of >1400 in HeLa cancer cells with a low light dose activation (400-700 nm, 2.2 J cm-2). The complexes display reduced activity in noncancerous HPL1D cells. The emission property of the complexes is used for cellular imaging, thus making them suitable as next-generation theranostic PDT agents.
Collapse
|
37
|
Gandioso A, Purkait K, Gasser G. Recent Approaches towards the Development of Ru(II) Polypyridyl Complexes for Anticancer Photodynamic Therapy. Chimia (Aarau) 2021; 75:845-855. [PMID: 34728011 DOI: 10.2533/chimia.2021.845] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Photodynamic therapy (PDT) is a remarkable alternative or complementary technique to chemotherapy, radiotherapy or immunotherapy to treat certain forms of cancer. The synergistic effect of light, photosensitizer (PS) and oxygen allows for the treatment of tumours with an extremely high spatio-tumoral control, therefore minimizing the severe side effects usually observed in chemotherapy. The currently employed PDT PSs based on porphyrins have, in some cases, some limitations, which include a low absorbance in the therapeutic window, a low body clearance, photobleaching, among others. In this context, Ru(ii) polypyridyl complexes are interesting alternatives. They have low lying excited energy states and the presence of a heavy metal increases the possibility of spin-orbit coupling. Moreover, their photophysical properties are relatively easy to tune and they have very low photobleaching rates. All of these make them attractive candidates for further development as therapeutically suitable PDT PSs. In this review, after having presented this field of research, we discuss the developments made by our group in this field of research since 2017. We notably describe how we tuned the photophysical properties of our complexes from the visible region to the therapeutically suitable red region. This was accompanied by the preparation of PSs with enhanced phototoxicity and high phototoxicity index. We also discuss the use of two-photon excitation to eradicate tumours in nude mice. Furthermore, we describe our approach for the selective delivery of our complexes using targeting agents. Lastly, we report on our very recent synergistic approach to treat cancer using bimetallic Ru(ii)-Pt(iv) prodrug candidates.
Collapse
Affiliation(s)
- Albert Gandioso
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| | - Kallol Purkait
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France;,
| |
Collapse
|
38
|
Vinck R, Karges J, Tharaud M, Cariou K, Gasser G. Physical, spectroscopic, and biological properties of ruthenium and osmium photosensitizers bearing diversely substituted 4,4'-di(styryl)-2,2'-bipyridine ligands. Dalton Trans 2021; 50:14629-14639. [PMID: 34581373 DOI: 10.1039/d1dt02083h] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Capitalising on the previous identification of a distyryl coordinated Ru(II) polypyridine complex as a promising photosensitizer for photodynamic therapy, eight new complexes were synthesized by modifications of the ligands or by changing the metal coordinated. We report in this work the effects of these modifications on the physical, spectroscopic, and biological properties of the synthesized complexes. Subtle structural modifications of the distyryl ligand only had a moderate effect on the corresponding complexes' visible light absorption and singlet oxygen quantum yield. These modifications however had a significant effect on the lipophilicity, the cellular uptake and the phototoxicity of the complexes. Although the lipophilicity of the complexes had a somewhat expected effect on their cellular uptake, this last parameter could not be directly correlated to their phototoxicity, revealing other underlying phenomena. Overall, this work allowed identification of two promising ruthenium complexes as photosensitisers for photodynamic therapy and provides some guidance on how to design better photosensitizers.
Collapse
Affiliation(s)
- Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Johannes Karges
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Mickaël Tharaud
- Institut de Physique du Globe de Paris, Biogéochimie à l'Anthropocène des Eléments et Contaminants Emergents, 75005 Paris, France
| | - 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
|
39
|
Yang M, Zhao H, Zhang Z, Yuan Q, Feng Q, Duan X, Wang S, Tang Y. CO/light dual-activatable Ru(ii)-conjugated oligomer agent for lysosome-targeted multimodal cancer therapeutics. Chem Sci 2021; 12:11515-11524. [PMID: 34667555 PMCID: PMC8447874 DOI: 10.1039/d1sc01317c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/21/2021] [Indexed: 01/10/2023] Open
Abstract
Stimuli-activatable and subcellular organelle-targeted agents with multimodal therapeutics are urgently desired for highly precise and effective cancer treatment. Herein, a CO/light dual-activatable Ru(ii)-oligo-(thiophene ethynylene) (Ru-OTE) for lysosome-targeted cancer therapy is reported. Ru-OTE is prepared via the coordination-driven self-assembly of a cationic conjugated oligomer (OTE-BN) ligand and a Ru(ii) center. Upon the dual-triggering of internal gaseous signaling molecular CO and external light, Ru-OTE undergoes ligand substitution and releases OTE-BN followed by dramatic fluorescence recovery, which could be used for monitoring drug delivery and imaging guided anticancer treatments. The released OTE-BN selectively accumulates in lysosomes, physically breaking their integrity. Then, the generated cytotoxic singlet oxygen (1O2) causes severe lysosome damage, thus leading to cancer cell death via photodynamic therapy (PDT). Meanwhile, the release of the Ru(ii) core also suppresses cancer cell growth as an anticancer metal drug. Its significant anticancer effect is realized via the multimodal therapeutics of physical disruption/PDT/chemotherapy. Importantly, Ru-OTE can be directly photo-activated using a two-photon laser (800 nm) for efficient drug release and near-infrared PDT. Furthermore, Ru-OTE with light irradiation inhibits tumor growth in an MDA-MB-231 breast tumor model with negligible side effects. This study demonstrates that the development of an activatable Ru(ii)-conjugated oligomer potential drug provides a new strategy for effective subcellular organelle-targeted multimodal cancer therapeutics. The anticancer therapeutics of lysosome disruption/PDT/chemotherapy based on Ru-OTE complex was achieved, which provides a new strategy for developing multimodal and effective stimuli-activatable subcellular organelle-targeted cancer therapeutics.![]()
Collapse
Affiliation(s)
- Min Yang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Hao Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences P. R. China
| | - Ziqi Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Qiong Yuan
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Qian Feng
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Xinrui Duan
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences P. R. China
| | - Yanli Tang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| |
Collapse
|
40
|
Lin M, Zou S, Liao X, Chen Y, Luo D, Ji L, Chao H. Ruthenium(II) complexes as bioorthogonal two-photon photosensitizers for tumour-specific photodynamic therapy against triple-negative breast cancer cells. Chem Commun (Camb) 2021; 57:4408-4411. [PMID: 33949487 DOI: 10.1039/d1cc00661d] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Herein, we developed the first Ru(ii) complex-based bioorthogonal two-photon photosensitizers. Through bioorthogonal labelling, they realize effective tumour-specific photodynamic therapy against triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Mingwei Lin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Shanshan Zou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Xinxing Liao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Diqing Luo
- Department of Dermatology, The Eastern Division of the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China.
| |
Collapse
|
41
|
Shen J, Karges J, Xiong K, Chen Y, Ji L, Chao H. Cancer cell membrane camouflaged iridium complexes functionalized black-titanium nanoparticles for hierarchical-targeted synergistic NIR-II photothermal and sonodynamic therapy. Biomaterials 2021; 275:120979. [PMID: 34166910 DOI: 10.1016/j.biomaterials.2021.120979] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
The diagnosis and treatment of cancer is one of the biggest medical challenges of the century. Despite significant improvements, there remains an urgent need for novel anticancer procedures. Among the most promising approaches, increasing attention has been devoted towards photothermal and sonodynamic therapy in which sensitizers are activated upon light/ultrasound radiation to generate a cytotoxic effect. While these methods have undoubtedly shown a high therapeutic success, these techniques are intrinsically limited. Herein, the functionalization of black-titanium nanoparticle with iridium complexes and cancer cell membranes in a nanoplatform for hierarchical targeted synergistic photothermal and sonodynamic cancer imaging and therapy is proposed. The particles showed to generate efficiently heat upon irradiation and catalytically form reactive oxygen species upon ultrasound radiation. The nanoparticle formulation demonstrated to selectively localize in the mitochondria as well as to preferentially accumulate in cancerous over non-cancerous cells as well as in the tumor inside a mouse model, presenting a hierarchical targeting strategy. Upon synergistic irradiation at 1064 nm and ultrasound radiation, the nanoparticles were able to act as an imaging agent and identify the tumor site with high spatial resolution as well as act as a therapeutic agent and completely eradicate a tumor inside a mouse model.
Collapse
Affiliation(s)
- Jinchao Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Johannes Karges
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, United States
| | - Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China; MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 400201, PR China.
| |
Collapse
|
42
|
Karges J, Díaz-García D, Prashar S, Gómez-Ruiz S, Gasser G. Ru(II) Polypyridine Complex-Functionalized Mesoporous Silica Nanoparticles as Photosensitizers for Cancer Targeted Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2021; 4:4394-4405. [PMID: 35006851 DOI: 10.1021/acsabm.1c00151] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer is the leading cause of death in the developed world. In the last few decades, photodynamic therapy (PDT) has augmented the number of medical techniques to treat this disease in the clinics. As the pharmacological active species to kill cancer cells are only generated upon light irradiation, PDT is associated with an intrinsic first level of selectivity. However, since PDT agents also accumulate in the surrounding, healthy tissue and since it is practically very challenging to only expose the tumor site to light, some side effects can be observed. Consequently, there is a need for a selective drug delivery system, which would give a second level of selectivity. In this work, a dual tumor targeting approach is presented based on mesoporous silica nanoparticles, which act by the enhanced permeability and retention effect, and the conjugation to folic acid, which acts as a targeting moiety for folate receptor-overexpressed cancer cells. The conjugates were found to be nontoxic in noncancerous human normal lung fibroblast cells while showing a phototoxic effect upon irradiation at 480 or 540 nm in the low nanomolar range in folate receptor overexpressing cancerous human ovarian carcinoma cells, demonstrating their potential for cancer targeted treatment.
Collapse
Affiliation(s)
- Johannes Karges
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University, Paris 75005, France
| | - Diana Díaz-García
- COMET-NANO Group, Departamento de Biología y Geología, Física y Química Inorgánica, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, Móstoles, Madrid E-28933, Spain
| | - Sanjiv Prashar
- COMET-NANO Group, Departamento de Biología y Geología, Física y Química Inorgánica, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, Móstoles, Madrid E-28933, Spain
| | - Santiago Gómez-Ruiz
- COMET-NANO Group, Departamento de Biología y Geología, Física y Química Inorgánica, ESCET, Universidad Rey Juan Carlos, Calle Tulipán s/n, Móstoles, Madrid E-28933, Spain
| | - Gilles Gasser
- Chimie ParisTech, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, PSL University, Paris 75005, France
| |
Collapse
|
43
|
Karges J, Tharaud M, Gasser G. Polymeric Encapsulation of a Ru(II)-Based Photosensitizer for Folate-Targeted Photodynamic Therapy of Drug Resistant Cancers. J Med Chem 2021; 64:4612-4622. [PMID: 33818111 DOI: 10.1021/acs.jmedchem.0c02006] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The currently used photodynamic therapy (PDT) photosensitizers (PSs) are generally associated with a poor cancer cell selectivity, which is responsible for some undesirable side effects. To overcome these problems, there is an urgent need for a selective drug delivery system for PDT PSs. Herein, the encapsulation of a promising Ru(II) polypyridine complex in a polymer with terminal folate groups to form nanoparticles is presented. While the Ru(II) complex itself has a cytotoxic effect in the dark, the encapsulation is able to overcome this drawback. Upon light exposure, the nanoparticles were found to be highly phototoxic in 2D monolayer cells as well as 3D multicellular tumor spheroids upon 480 or 595 nm irradiation. Importantly, the nanoparticles demonstrated a high selectivity for cancerous cells over noncancerous cells and were found to be active in drug resistant cancer cells lines, indicating that they are able to overcome drug resistances.
Collapse
Affiliation(s)
- Johannes Karges
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France
| | - Mickaël Tharaud
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, F-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
|