1
|
Chen M, Wang J, Cai F, Guo J, Qin X, Zhang H, Chen T, Ma L. Chirality-driven strong thioredoxin reductase inhibition. Biomaterials 2024; 311:122705. [PMID: 39047537 DOI: 10.1016/j.biomaterials.2024.122705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
Overexpression of thioredoxin reductase (TXNRD) plays crucial role in tumorigenesis. Therefore, designing TXNRD inhibitors is a promising strategy for targeted anticancer drug development. However, poor selectivity has always been a challenge, resulting in unavoidable toxicity in clinic. Herein we demonstrate a strategy to develop highly selective chiral metal complexes-based TXNRD inhibitors. By manipulating the conformation of two distinct weakly interacting groups, we optimize the compatibility between the drug and the electrophilic group within the active site of TXNRD to enhance their non-covalent interaction, thus effectively avoids the poor selectivity deriving from covalent drug interaction, on the basis of ensuring the strong inhibition. Detailed experimental and computational results demonstrate that the chiral isomeric drugs bind to the active site of TXNRD, and the interaction strength is well modulated by chirality. Especially, the meso-configuration, in which the two large sterically hindered active groups are positioned on opposite sides of the drug, exhibits the highest number of non-covalent interactions and most effective inhibition on TXNRD. Taken together, this work not only provides a novel approach for developing highly selective proteinase inhibitors, but also sheds light on possible underlying mechanisms for future application.
Collapse
Affiliation(s)
- Mingkai Chen
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Junping Wang
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Fei Cai
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Junxian Guo
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Xiaoyu Qin
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Huajie Zhang
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China.
| | - Li Ma
- Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
2
|
Gong N, Alameh MG, El-Mayta R, Xue L, Weissman D, Mitchell MJ. Enhancing in situ cancer vaccines using delivery technologies. Nat Rev Drug Discov 2024; 23:607-625. [PMID: 38951662 DOI: 10.1038/s41573-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/03/2024]
Abstract
In situ cancer vaccination refers to any approach that exploits tumour antigens available at a tumour site to induce tumour-specific adaptive immune responses. These approaches hold great promise for the treatment of many solid tumours, with numerous candidate drugs under preclinical or clinical evaluation and several products already approved. However, there are challenges in the development of effective in situ cancer vaccines. For example, inadequate release of tumour antigens from tumour cells limits antigen uptake by immune cells; insufficient antigen processing by antigen-presenting cells restricts the generation of antigen-specific T cell responses; and the suppressive immune microenvironment of the tumour leads to exhaustion and death of effector cells. Rationally designed delivery technologies such as lipid nanoparticles, hydrogels, scaffolds and polymeric nanoparticles are uniquely suited to overcome these challenges through the targeted delivery of therapeutics to tumour cells, immune cells or the extracellular matrix. Here, we discuss delivery technologies that have the potential to reduce various clinical barriers for in situ cancer vaccines. We also provide our perspective on this emerging field that lies at the interface of cancer vaccine biology and delivery technologies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Rakan El-Mayta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Zafar H, Zhang J, Raza F, Pan X, Hu Z, Feng H, Shen Q. Biomimetic gold nanocages incorporating copper-human serum albumin for tumor immunotherapy via cuproptosis-lactate regulation. J Control Release 2024; 372:446-466. [PMID: 38917953 DOI: 10.1016/j.jconrel.2024.06.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Cancer immunotherapy remains a significant challenge due to insufficient proliferation of immune cells and the sturdy immunosuppressive tumor microenvironment. Herein, we proposed the hypothesis of cuproptosis-lactate regulation to provoke cuproptosis and enhance anti-tumor immunity. For this purpose, copper-human serum albumin nanocomplex loaded gold nanocages with bacterial membrane coating (BAu-CuNCs) were developed. The targeted delivery and disassembly of BAu-CuNCs in tumor cells initiated a cascade of reactions. Under near infrared (NIR) laser irradiation, the release of copper-human serum albumin (Cu-HSA) was enhanced that reacted with intratumoral glutathione (GSH) via a disulfide exchange reaction to liberate Cu2+ ions and exert cuproptosis. Subsequently, the cuproptosis effect triggered immunogenic cell death (ICD) in tumor by the release of damage associated molecular patterns (DAMPs) to realize anti-tumor immunity via robust production of cytotoxic T cells (CD8+) and helper T cells (CD4+). Meanwhile, under NIR irradiation, gold nanocages (AuNCs) promoted excessive reactive oxygen species (ROS) generation that played a primary role in inhibiting glycolysis, reducing the lactate and ATP level. The combine action of lower lactate level, ATP reduction and GSH depletion further sensitized the tumor cells to cuproptosis. Also, the lower lactate production led to the significant blockage of immunosuppressive T regulatory cells (Tregs) and boosted the anti-tumor immunity. Additionally, the effective inhibition of breast cancer metastasis to the lungs enhanced the anti-tumor therapeutic impact of BAu-CuNCs + NIR treatment. Hence, BAu-CuNCs + NIR concurrently induced cuproptosis, ICD and hindered lactate production, leading to the inhibition of tumor growth, remodeling of the immunosuppressive tumor microenvironment and suppression of lung metastasis. Therefore, leveraging cuproptosis-lactate regulation, this approach presents a novel strategy for enhanced tumor immunotherapy.
Collapse
Affiliation(s)
- Hajra Zafar
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jun Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Faisal Raza
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiuhua Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zongwei Hu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hanxiao Feng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China..
| |
Collapse
|
4
|
Xie K, Lu XY, Zhu H, Zhu LY, Li RT, Ye RR. Iridium(III) complexes conjugated with naproxen exhibit potent anti-tumor activities by inducing mitochondrial damage, modulating inflammation, and enhancing immunity. Dalton Trans 2024; 53:8772-8780. [PMID: 38712840 DOI: 10.1039/d4dt00575a] [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: 05/08/2024]
Abstract
A series of Ir(III)-naproxen (NPX) conjugates with the molecular formula [Ir(C^N)2bpy(4-CH2ONPX-4'-CH2ONPX)](PF6) (Ir-NPX-1-3) were designed and synthesized, including C^N = 2-phenylpyridine (ppy, Ir-NPX-1), 2-(2-thienyl)pyridine (thpy, Ir-NPX-2) and 2-(2,4-difluorophenyl)pyridine (dfppy, Ir-NPX-3). Cytotoxicity tests showed that Ir-NPX-1-3 exhibited excellent antitumor activity, especially in A549R cells. The cellular uptake experiment showed that the complexes were mainly localized in mitochondria, and induced apoptosis in A549R cells by damaging the structure and function of mitochondria. The main manifestations are a decrease in the mitochondrial membrane potential (MMP), an increase in reactive oxygen species (ROS) levels, and cell cycle arrest. Furthermore, Ir-NPX-1-3 could inhibit the migration and colony formation of cancer cells, demonstrating potential anti-metastatic ability. Finally, the anti-inflammatory and immunological applications of Ir-NPX-1-3 were verified. The downregulation of cyclooxygenase-2 (COX-2) and programmed death-ligand 1 (PD-L1) expression levels and the release of immunogenic cell death (ICD) related signaling molecules such as damage-associated molecular patterns (DAMPs) (cell surface calreticulin (CRT), high mobility group box 1 (HMGB1), and adenosine triphosphate (ATP)) indicate that these Ir(III) -NPX conjugates are novel ICD inducers with synergistic effects in multiple anti-tumor pathways.
Collapse
Affiliation(s)
- Kai Xie
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Xing-Yun Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Hou Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Lin-Yuan Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| |
Collapse
|
5
|
Liao J, Zhang Y, Huang M, Liang Z, Gong Y, Liu B, Li Y, Chen J, Wu W, Huang Z, Sun J. Cyclometalated iridium(III) complexes induce immunogenic cell death in HepG2 cells via paraptosis. Bioorg Chem 2023; 140:106837. [PMID: 37683535 DOI: 10.1016/j.bioorg.2023.106837] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/25/2023] [Accepted: 09/03/2023] [Indexed: 09/10/2023]
Abstract
Immunotherapy has been shown to provide superior antitumor efficacy by activating the innate immune system to recognize, attack and eliminate tumor cells without seriously harming normal cells. Herein, we designed and synthesized three new cyclometalated iridium(III) complexes (Ir1, Ir2, Ir3) then evaluated their antitumor activity. When co-incubated with HepG2 cells, the complex Ir1 localized in the lysosome, where it induced paraptosis and endoplasmic reticulum stress (ER stress). Notably, Ir1 also induced immunogenic cell death (ICD), promoted dendritic cell maturation that enhanced effector T cell chemotaxis to tumor tissues, down-regulated proportions of immunosuppressive regulatory T cells within tumor tissues and triggered activation of antitumor immunity throughout the body. To date, Ir1 is the first reported iridium(III) complex-based paraptosis inducer to successfully induce tumor cell ICD. Furthermore, Ir1 induced ICD of HepG2 cells without affecting cell cycle or reactive oxygen species levels.
Collapse
Affiliation(s)
- Jiaxin Liao
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yuqing Zhang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Minying Huang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhijun Liang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yao Gong
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Ben Liu
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yuling Li
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaxi Chen
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Wei Wu
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| | - Jing Sun
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
6
|
Amiri M, Molavi O, Sabetkam S, Jafari S, Montazersaheb S. Stimulators of immunogenic cell death for cancer therapy: focusing on natural compounds. Cancer Cell Int 2023; 23:200. [PMID: 37705051 PMCID: PMC10500939 DOI: 10.1186/s12935-023-03058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
A growing body of evidence indicates that the anticancer effect of the immune system can be activated by the immunogenic modulation of dying cancer cells. Cancer cell death, as a result of the activation of an immunomodulatory response, is called immunogenic cell death (ICD). This regulated cell death occurs because of increased immunogenicity of cancer cells undergoing ICD. ICD plays a crucial role in stimulating immune system activity in cancer therapy. ICD can therefore be an innovative route to improve anticancer immune responses associated with releasing damage-associated molecular patterns (DAMPs). Several conventional and chemotherapeutics, as well as preclinically investigated compounds from natural sources, possess immunostimulatory properties by ICD induction. Natural compounds have gained much interest in cancer therapy owing to their low toxicity, low cost, and inhibiting cancer cells by interfering with different mechanisms, which are critical in cancer progression. Therefore, identifying natural compounds with ICD-inducing potency presents agents with promising potential in cancer immunotherapy. Naturally derived compounds are believed to act as immunoadjuvants because they elicit cancer stress responses and DAMPs. Acute exposure to DAMP molecules can activate antigen-presenting cells (APCs), such as dendritic cells (DCs), which leads to downstream events by cytotoxic T lymphocytes (CTLs) and natural killer cells (NKs). Natural compounds as inducers of ICD may be an interesting approach to ICD induction; however, parameters that determine whether a compound can be used as an ICD inducer should be elucidated. Here, we aimed to discuss the impact of multiple ICD inducers, mainly focusing on natural agents, including plant-derived, marine molecules, and bacterial-based compounds, on the release of DAMP molecules and the activation of the corresponding signaling cascades triggering immune responses. In addition, the potential of synthetic agents for triggering ICD is also discussed.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahnaz Sabetkam
- Department of Anatomy, Faculty of Medicine, university of Kyrenia, Kyrenia, Northern Cyprus
- Department of Anatomy and histopathology, Faculty of medicine, Tabriz medical sciences, Islamic Azad University, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Liu SY, Huang DJ, En-yu Tang, Zhang RX, Zhang ZM, Gao T, Xu GQ. Construction of a non-negative matrix factorization model of immunogenic cell death-related genes in lung adenocarcinoma and analysis of survival prognosis. Heliyon 2023; 9:e14820. [PMID: 37025770 PMCID: PMC10070601 DOI: 10.1016/j.heliyon.2023.e14820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Purpose To explore the effectiveness of the model based on non-negative matrix factorization (NMF), analyze the tumor microenvironment and immune microenvironment for evaluating the prognosis of lung adenocarcinoma, establish a risk model, and screen independent prognostic factors. Methods Downloading the transcription data files and clinical information files of lung adenocarcinoma from TCGA database and GO database, the R software was used to establish the NMF cluster model, and then the survival analysis between groups, tumor microenvironment analysis, and immune microenvironment analysis was performed according to the NMF cluster result. R software was used to construct prognostic models and calculate risk scores. Survival analysis was used to compare survival differences between different risk score groups. Results Two ICD subgroups were established according to the NMF model. The survival of the ICD low-expression subgroup was better than that of the ICD high-expression subgroup. Univariate COX analysis screened out HSP90AA1, IL1, and NT5E as prognostic genes, and the prognostic model established on this basis has clinical guiding significance. Conclusion The model based on NMF has the prognostic ability for lung adenocarcinoma, and the prognostic model of ICD-related genes has a certain guiding significance for survival.
Collapse
|
8
|
Xiong X, Wang Y, Zou T. Towards Understanding the Molecular Mechanisms of Immunogenic Cell Death. Chembiochem 2023; 24:e202200621. [PMID: 36445798 DOI: 10.1002/cbic.202200621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 11/30/2022]
Abstract
The discovery of immunogenic cell death (ICD) by small molecules (e. g., chemotherapeutic drugs) intrigued medicinal chemists and led them to exploit anticancer agents with such a trait because ICD agents provoke anticancer immune responses in addition to their cytotoxicity. However, the unclear molecular mechanism of ICD hampers further achievements in drug development. Fortunately, increasing efforts have been made in this area in recent years by using either chemical or biological approaches. Here, we review the current achievements towards understanding the mechanisms of small molecule-induced ICD effects. Based on the established role of the unfolded protein response (UPR) in ICD, we classify the mechanisms of different inducers by their dependency on UPR. Key proteins and pathways with important implications are discussed in depth. We also give our perspectives on the research strategies for future investigation in this field.
Collapse
Affiliation(s)
- Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yuan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
9
|
Lou X, Wang H, Liu Y, Huang Y, Liu Z, Zhang W, Wang T. Perylene-Based Reactive Oxygen Species Supergenerator for Immunogenic Photochemotherapy against Hypoxic Tumors. Angew Chem Int Ed Engl 2023; 62:e202214586. [PMID: 36597125 DOI: 10.1002/anie.202214586] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/03/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
Reactive oxygen species (ROS) can act as cytotoxic radicals to directly kill tumor cells and concurrently trigger immunogenic cell death (ICD) to efficiently achieve tumor therapy. Thus motivated, we herein present one perylene monoamide-based ROS supergenerator (PMIC-NC) that not only induces hypoxia-enhanced Type-I ROS burst aided by proton transients but also triggers Type-I/II ROS production by electron or energy transfer under near-infrared (NIR) light irradiation and also elicits a strong ICD effect. More interesting, the mitochondria- and lung-specific distribution of PMIC-NC also boosts the tumor therapeutic efficiency. As a result, PMIC-NC was employed for NIR-triggered photodynamic therapy, hypoxia-enhanced chemotherapy and also displayed robust immunogenicity for systemic tumor eradication. This work thus contributes one proof-of-concept demonstration of perylene as an integrated therapeutic platform for efficient immunogenic photochemotherapy against hypoxic tumors.
Collapse
Affiliation(s)
- Xue Lou
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Kaifeng, 475004, P. R. China
| | - Hui Wang
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Kaifeng, 475004, P. R. China
| | - Yu Liu
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Kaifeng, 475004, P. R. China
| | - Yongwei Huang
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Kaifeng, 475004, P. R. China
| | - Zhonghua Liu
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Kaifeng, 475004, P. R. China
| | - Wei Zhang
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, Life and Health Intelligent Research Institute, Tianjin University of Technology, Tianjin, 300384, P. R. China
| | - Tie Wang
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, Life and Health Intelligent Research Institute, Tianjin University of Technology, Tianjin, 300384, P. R. China
| |
Collapse
|
10
|
Zhang L, Montesdeoca N, Karges J, Xiao H. Immunogenic Cell Death Inducing Metal Complexes for Cancer Therapy. Angew Chem Int Ed Engl 2023; 62:e202300662. [PMID: 36807420 DOI: 10.1002/anie.202300662] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 02/23/2023]
Abstract
Cancer is one of the deadliest diseases worldwide. Recent statistics have shown that metastases and tumor relapse are the leading causes of cancer-associated deaths. While traditional treatments are able to efficiently remove the primary tumor, secondary tumors remain poorly accessible. Capitalizing on this there is an urgent need for novel treatment modalities. Among the most promising approaches, increasing research interest has been devoted to immunogenic cell death inducing agents that are able to trigger localized cell death of the cancer cells as well as induce an immune response inside the whole organism. Preliminary studies have shown that immunogenic cell death inducing compounds could be able to overcome metastatic and relapsing tumors. Herein, the application of metal complexes as immunogenic cell death inducing compounds is systematically reviewed.
Collapse
Affiliation(s)
- Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
| | - Nicolás Montesdeoca
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
| |
Collapse
|
11
|
Highlights of New Strategies to Increase the Efficacy of Transition Metal Complexes for Cancer Treatments. Molecules 2022; 28:molecules28010273. [PMID: 36615466 PMCID: PMC9822110 DOI: 10.3390/molecules28010273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Although important progress has been made, cancer still remains a complex disease to treat. Serious side effects, the insurgence of resistance and poor selectivity are some of the problems associated with the classical metal-based anti-cancer therapies currently in clinical use. New treatment approaches are still needed to increase cancer patient survival without cancer recurrence. Herein, we reviewed two promising-at least in our opinion-new strategies to increase the efficacy of transition metal-based complexes. First, we considered the possibility of assembling two biologically active fragments containing different metal centres into the same molecule, thus obtaining a heterobimetallic complex. A critical comparison with the monometallic counterparts was done. The reviewed literature has been divided into two groups: the case of platinum; the case of gold. Secondly, the conjugation of metal-based complexes to a targeting moiety was discussed. Particularly, we highlighted some interesting examples of compounds targeting cancer cell organelles according to a third-order targeting approach, and complexes targeting the whole cancer cell, according to a second-order targeting strategy.
Collapse
|
12
|
Sun J, Zhang X, Wang X, Peng J, Song G, Di Y, Feng F, Wang S. Dithiol-Activated Bioorthogonal Chemistry for Endoplasmic Reticulum-Targeted Synergistic Chemophototherapy. Angew Chem Int Ed Engl 2022; 61:e202213765. [PMID: 36342403 DOI: 10.1002/anie.202213765] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Indexed: 11/09/2022]
Abstract
The controlled intracellular release of nitrite is still an unmet challenge due to the lack of bio-friendly donors, and the antitumor effect of nitrite is limited by its physiologically inert activity. Herein, we designed benzothiadiazole-based organic nitrite donors that are stable against bio-relevant species but selectively respond to dithiol species through SN Ar/intramolecular cyclization tandem reactions in the aqueous media. The bioorthogonal system was established to target the endoplasmic reticulum (ER) of liver cancer HepG2 cells. The nitrite and nonivamide were coupled to induce elevation of intracellular levels of calcium ions as well as reactive oxygen/nitrogen species, which resulted in ER stress and mitochondrial dysfunction. We demonstrated that a combination of photoactivation and "click to release" strategy could enhance antitumor effect in cellular level and show good potential for cancer precision therapy.
Collapse
Affiliation(s)
- Jian Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Xiaoran Zhang
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Xia Wang
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Jinlei Peng
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Gang Song
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,College of Chemistry, University of Chinese Academy of Sciences, 100190, Beijing, P. R. China
| | - Yufei Di
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,College of Chemistry, University of Chinese Academy of Sciences, 100190, Beijing, P. R. China
| | - Fude Feng
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Jiangsu, Nanjing, P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.,College of Chemistry, University of Chinese Academy of Sciences, 100190, Beijing, P. R. China
| |
Collapse
|
13
|
Bera A, Gautam S, Sahoo S, Pal AK, Kondaiah P, Chakravarty AR. Red light active Pt(iv)-BODIPY prodrug as a mitochondria and endoplasmic reticulum targeted chemo-PDT agent. RSC Med Chem 2022; 13:1526-1539. [PMID: 36561074 PMCID: PMC9749958 DOI: 10.1039/d2md00225f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/22/2022] [Indexed: 12/25/2022] Open
Abstract
A cisplatin-based platinum(iv) prodrug, [Pt(NH3)2Cl2(OH)(L 1 )], having L 1 as a red-light active boron-dipyrromethene (BODIPY) pendant, was synthesized and characterized and its application as a chemo-cum-photodynamic therapy agent was studied. Me-L 1 as the ligand precursor is structurally characterized. The complex displayed an intense absorption band near 650 nm (ε ∼ 8.8 × 104 dm3 mol-1 cm-1) in 1 : 1 (v/v) DMSO/DPBS. It showed an emission band at 674 nm (λ ex = 630 nm) with a fluorescence quantum yield (Φ F) value of 0.37. In red light (600-720 nm), it generated singlet oxygen as evidenced from the 1,3-diphenylisobenzofuran (DPBF) titration experiment giving a singlet oxygen quantum yield (Φ Δ) value of 0.28 in DMSO. The mechanistic pUC19 DNA photocleavage study and singlet oxygen sensor green (SOSG) assay ascertained its ability to generate singlet oxygen in both extracellular and intracellular media by a type-II photo-process. The complex exhibited high stability in the dark, but on red-light irradiation, it displayed rapid activation in the presence of a reducing environment. It displayed remarkable apoptotic photocytotoxicity with half-maximal inhibitory concentration (IC50) ranging from 0.58 to 0.76 μM in human cervical cancer (HeLa) and breast cancer (MCF-7) cells with a respective photo-cytotoxicity index value of >172 and >131. The photodynamic activity was significantly less in non-cancerous human peripheral lung epithelial (HPL1D) cells. The emissive complex showed localization in the mitochondria and endoplasmic reticulum (ER) with a similar Pearson's correlation coefficient value, making it a dual organelle-targeted therapeutic agent. JC-1, fluo-4-AM and annexin V-FITC/propidium iodide assays in HeLa cells showed cellular apoptosis by arresting cells in the sub-G1 phase via mitochondrial dysfunction and ER stress.
Collapse
Affiliation(s)
- Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| | - Srishti Gautam
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560012 India +91 80 22932688
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| | - Apurba Kumar Pal
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560012 India +91 80 22932688
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| |
Collapse
|
14
|
Ling Y, Xia X, Hao L, Wang W, Zhang H, Liu L, Liu W, Li Z, Tan C, Mao Z. Simultaneous Photoactivation of cGAS‐STING Pathway and Pyroptosis by Platinum (II) Triphenylamine Complexes for Cancer Immunotherapy. Angew Chem Int Ed Engl 2022; 61:e202210988. [DOI: 10.1002/anie.202210988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Yu‐Yi Ling
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Xiao‐Yu Xia
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Liang Hao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Wen‐Jin Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Hang Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Liu‐Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Wenting Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Zhi‐Yuan Li
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Cai‐Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Zong‐Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University Guangzhou 510006 P. R. China
| |
Collapse
|
15
|
Ling YY, Xia XY, Hao L, Wang WJ, Zhang H, Liu LY, Liu W, Li ZY, Tan CP, Mao ZW. Simultaneous Photoactivation of cGAS‐STING Pathway and Pyroptosis by Pt(II)‐Triphenylamine Complexes for Cancer Immunotherapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202210988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yu-Yi Ling
- Sun Yat-sen University School of Chemistry Sun Yat-sen University School of Chemistry CHINA
| | - Xiao-Yu Xia
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Liang Hao
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Wen-Jin Wang
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Hang Zhang
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Liu-Yi Liu
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Wenting Liu
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Zhi-Yuan Li
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Cai-Ping Tan
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Zong-Wan Mao
- Sun Yat-sen University School of Chemistry School of Chemistry No. 135 Xingang Xi Road 510006 Guangzhou CHINA
| |
Collapse
|
16
|
Wang MM, Xu FJ, Su Y, Geng Y, Qian XT, Xue XL, Kong YQ, Yu ZH, Liu HK, Su Z. A New Strategy to Fight Metallodrug Resistance: Mitochondria-Relevant Treatment through Mitophagy to Inhibit Metabolic Adaptations of Cancer Cells. Angew Chem Int Ed Engl 2022; 61:e202203843. [PMID: 35384194 DOI: 10.1002/anie.202203843] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Indexed: 12/12/2022]
Abstract
Metabolic adaptations can help cancer cells to escape from chemotherapeutics, mainly involving autophagy and ATP production. Herein, we report a new rhein-based cyclometalated IrIII complex, Ir-Rhein, that can accurately target mitochondria and effectively inhibit metabolic adaptations. The complex Ir-Rhein induces severe mitochondrial damage and initiates mitophagy to reduce the number of mitochondria and subsequently inhibit both mitochondrial and glycolytic bioenergetics, which eventually leads to ATP starvation death. Moreover, Ir-Rhein can overcome cisplatin resistance. Co-incubation experiment, 3D tumor spheroids experiment and transcriptome analysis reveal that Ir-Rhein shows promising antiproliferation performance for cisplatin-resistant cancer cells with the regulation of platinum resistance-related transporters. To our knowledge, this is a new strategy to overcome metallodrug resistance with a mitochondria-relevant treatment.
Collapse
Affiliation(s)
- Meng-Meng Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Fu-Jie Xu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Yan Su
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.,Department of Rheumatology and Immunology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Yun Geng
- Institute of Functional Material Chemistry, Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Xiao-Ting Qian
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Xu-Ling Xue
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Ya-Qiong Kong
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Zheng-Hong Yu
- Department of Rheumatology and Immunology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Hong-Ke Liu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Zhi Su
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
17
|
Gu S, Yang X, jiang Q, Luo Y, Wang D, Shi P. Insights into the crystal structure and optical property for complexes of iminodiacetic‐terpyridine. Z Anorg Allg Chem 2022. [DOI: 10.1002/zaac.202200052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Shunxin Gu
- Jiangsu Ocean University School of Environmental and Chemical Engineering CHINA
| | - Xinda Yang
- Tongji University School of Chemical Science and Engineering CHINA
| | - qin jiang
- Jiangsu Ocean University School of Enviromental and Chemical Engineering 59 Cangwu Road 222005 Lianyungang CHINA
| | - Yuhui Luo
- Jiangsu Ocean University School of Environmental and Chemical Engineering CHINA
| | - Daqi Wang
- Liaocheng University School of Chemistry CHINA
| | - Pengfei Shi
- Jiangsu Ocean University School of Environmental and Chemical Engineering CHINA
| |
Collapse
|
18
|
Wang M, Xu F, Su Y, Geng Y, Qian X, Xue X, Kong Y, Yu Z, Liu H, Su Z. A New Strategy to Fight Metallodrug Resistance: Mitochondria‐Relevant Treatment through Mitophagy to Inhibit Metabolic Adaptations of Cancer Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202203843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Meng‐Meng Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Fu‐Jie Xu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Yan Su
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
- Department of Rheumatology and Immunology Jinling Hospital Medical School of Nanjing University Nanjing 210002 China
| | - Yun Geng
- Institute of Functional Material Chemistry Faculty of Chemistry Northeast Normal University Changchun 130024 China
| | - Xiao‐Ting Qian
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Xu‐Ling Xue
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Ya‐Qiong Kong
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Zheng‐Hong Yu
- Department of Rheumatology and Immunology Jinling Hospital Medical School of Nanjing University Nanjing 210002 China
| | - Hong‐Ke Liu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Zhi Su
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials College of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| |
Collapse
|
19
|
Li Q, Chen C, Kong J, Li L, Li J, Huang Y. Stimuli-responsive nano vehicle enhances cancer immunotherapy by coordinating mitochondria-targeted immunogenic cell death and PD-L1 blockade. Acta Pharm Sin B 2022; 12:2533-2549. [PMID: 35646521 PMCID: PMC9136536 DOI: 10.1016/j.apsb.2021.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/03/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022] Open
Abstract
Induction of immunogenic cell death promotes antitumor immunity against cancer. However, majority of clinically-approved drugs are unable to elicit sufficient ICD. Here, our study revealed that mitochondria-targeted delivery of doxorubicin (DOX) massively amplified ICD via substantial generation of reactive oxygen species (ROS) after mitochondrial damage. The underlying mechanism behind increased ICD was further demonstrated to be ascribed to two pathways: (1) ROS elevated endoplasmic reticulum (ER) stress, leading to surface exposure of calreticulin; (2) ROS promoted release of various mitochondria-associated damage molecules including mitochondrial transcription factor A. Nevertheless, adaptive upregulation of PD-L1 was found after such ICD-inducing treatment. To overcome such immunosuppressive feedback, we developed a tumor stimuli-responsive nano vehicle to simultaneously exert mitochondrial targeted ICD induction and PD-L1 blockade. The nano vehicle was self-assembled from ICD-inducing copolymer and PD-L1 blocking copolymer, and possessed long-circulating property which contributed to better tumor accumulation and mitochondrial targeting. As a result, the nano vehicle remarkably activated antitumor immune responses and exhibited robust antitumor efficacy in both immunogenic and non-immunogenic tumor mouse models.
Collapse
|
20
|
Sen S, Won M, Levine MS, Noh Y, Sedgwick AC, Kim JS, Sessler JL, Arambula JF. Metal-based anticancer agents as immunogenic cell death inducers: the past, present, and future. Chem Soc Rev 2022; 51:1212-1233. [PMID: 35099487 PMCID: PMC9398513 DOI: 10.1039/d1cs00417d] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer is the deadliest disease in the world behind heart disease. Sadly, this remains true even as we suffer the ravages of the Covid-19 pandemic. Whilst current chemo- and radiotherapeutic treatment strategies have significantly improved the patient survival rate, disease reoccurrence continues to pose a deadly risk for all too many patients. Incomplete removal of tumour cells from the body increases the chances of metastasis and developing resistance against current treatments. Immunotherapy represents a therapeutic modality that has helped to overcome these limitations in recent decades. However, further progress is needed. So-called immunogenic cell death (ICD) is a recently discovered and unique mode of cell death that could trigger this necessary further progress. ICD involves stimulation of a tumour-specific immune response as a downstream effect. Facilitated by certain treatment modalities, cells undergoing ICD can trigger the IFN-γ mediated immune response involving cytotoxic T cells (CTLs) and γδ T cells that eradicate residual tumour cells. In recent years, there has been a significant increase in the number of small-molecules being tested as potential ICD inducers. A large number of these ICD inducers are metal-based complexes. In fact, anticancer metal drugs based on Pt, Ru, Ir, Cu, and Au are now known to give rise to an immune response against tumour cells as the result of ICD. Advances have also been made in terms of exploiting combinatorial and delivery strategies. In favourable cases, these approaches have been shown to increase the efficacy of otherwise ICD "silent" metal complexes. Taken in concert, rationally designed novel anticancer metal complexes that can act as ICD inducers show promise as potential new immunotherapies for neoplastic disease. This Tutorial Review will allow the readers to assess the progress in this fast-evolving field thus setting the stage for future advances.
Collapse
Affiliation(s)
- Sajal Sen
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA.
| | - Miae Won
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Matthew S Levine
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA.
| | - Yuvin Noh
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Adam C Sedgwick
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA.
| | - Jonathan F Arambula
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA.
- OncoTEX, Inc. 3800 North Lamar Blvd., Austin, Texas 78756, USA
| |
Collapse
|
21
|
Wang WJ, Ling YY, Zhong YM, Li ZY, Tan CP, Mao ZW. Ferroptosis‐Enhanced Cancer Immunity by a Ferrocene‐Appended Iridium(III) Diphosphine Complex. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202115247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Wen-Jin Wang
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Yu-Yi Ling
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Yan-Mei Zhong
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Zhi-Yuan Li
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Cai-Ping Tan
- Sun Yat-sen University School of Chemistry School of Chemistry CHINA
| | - Zong-Wan Mao
- Sun Yat-sen University School of Chemistry School of Chemistry No. 135 Xingang Xi Road 510006 Guangzhou CHINA
| |
Collapse
|
22
|
Wang WJ, Ling YY, Zhong YM, Li ZY, Tan CP, Mao ZW. Ferroptosis-Enhanced Cancer Immunity by a Ferrocene-Appended Iridium(III) Diphosphine Complex. Angew Chem Int Ed Engl 2021; 61:e202115247. [PMID: 34965011 DOI: 10.1002/anie.202115247] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Indexed: 11/10/2022]
Abstract
Ferroptosis is a programmed cell death pathway discovered in recent years, and ferroptosis-inducing agents have great potential as new antitumor candidates. Here, we report a Ir(III) complex (Ir1) containing a ferrocene-modified diphosphine ligand that localizes in lysosomes. Under the acidic environments of lysosomes, Ir1 can effectively catalyze Fenton reaction, produce hydroxyl radicals, induce lipid peroxidation, down-regulate glutathione peroxidase 4, and induce ferroptosis. RNA sequencing analysis shows that Ir1 can significantly affect pathways related to ferroptosis and cancer immunity. Accordingly, Ir1 can induce immunogenic cells death and suppress tumor growth in vitro, regulate T cell activity and immune microenvironments in vivo. In conclusion, we show the potential of small molecules with ferroptosis-inducing capabilities for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Wen-Jin Wang
- Sun Yat-sen University School of Chemistry, School of Chemistry, CHINA
| | - Yu-Yi Ling
- Sun Yat-sen University School of Chemistry, School of Chemistry, CHINA
| | - Yan-Mei Zhong
- Sun Yat-sen University School of Chemistry, School of Chemistry, CHINA
| | - Zhi-Yuan Li
- Sun Yat-sen University School of Chemistry, School of Chemistry, CHINA
| | - Cai-Ping Tan
- Sun Yat-sen University School of Chemistry, School of Chemistry, CHINA
| | - Zong-Wan Mao
- Sun Yat-sen University School of Chemistry, School of Chemistry, No. 135 Xingang Xi Road, 510006, Guangzhou, CHINA
| |
Collapse
|
23
|
Qiao L, Shao X, Gao S, Ming Z, Fu X, Wei Q. Research on endoplasmic reticulum-targeting fluorescent probes and endoplasmic reticulum stress-mediated nanoanticancer strategies: A review. Colloids Surf B Biointerfaces 2021; 208:112046. [PMID: 34419809 DOI: 10.1016/j.colsurfb.2021.112046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 07/12/2021] [Accepted: 08/14/2021] [Indexed: 01/18/2023]
Abstract
Subcellular localization of organelles can achieve accurate drug delivery and maximize drug efficacy. As the largest organelle in eukaryotic cells, the endoplasmic reticulum (ER) plays an important role in protein synthesis, folding, and posttranslational modification; lipid biosynthesis; and calcium homeostasis. Observing the changes in various metal ions, active substances, and the microenvironment in the ER is crucial for diagnosing and treating many diseases, including cancer. Excessive endoplasmic reticulum stress (ERS) can have a killing effect on malignant cells and can mediate cell apoptosis, proper modulation of ERS can provide new perspectives for the treatment of many diseases, including cancer. Therefore, the ER is used as a new anticancer target in cancer treatment. This review discusses ER-targeting fluorescent probes and ERS-mediated nanoanticancer strategies.
Collapse
Affiliation(s)
- Li Qiao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Xinxin Shao
- Laboratory of Traditional Chinese Medicine Network Pharmacology, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Shijie Gao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Zheng Ming
- International Office, Shandong University of Traditional Chinese Medicine, PR China
| | - Xianjun Fu
- Laboratory of Traditional Chinese Medicine Network Pharmacology, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Qingcong Wei
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, PR China.
| |
Collapse
|
24
|
Wang P, Wang JW, Zhang WH, Bai H, Tang G, Young DJ. In Vitro Anticancer Activity of Nanoformulated Mono- and Di-nuclear Pt Compounds. Chem Asian J 2021; 16:2993-3000. [PMID: 34387027 DOI: 10.1002/asia.202100901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Indexed: 12/14/2022]
Abstract
Nanoformulations of mononuclear Pt complexes cis-PtCl2 (PPh3 )2 (1), [Pt(PPh3 )2 (L-Cys)] ⋅ H2 O (3, L-Cys=L-cysteinate), trans-PtCl2 (PPh2 PhNMe2 )2 (4; PPh2 PhNMe2 =4-(dimethylamine)triphenylphosphine), trans-PtI2 (PPh2 PhNMe2 )2 (5) and dinuclear Pt cluster Pt2 (μ-S)2 (PPh3 )4 (2) have comparable cytotoxicity to cisplatin against murine melanoma cell line B16F10. Masking of these discrete molecular entities within the hydrophobic core of Pluronic® F-127 significantly boosted their solubility and stability, ensuring efficient cellular uptake, giving in vitro IC50 values in the range of 0.87-11.23 μM. These results highlight the potential therapeutic value of Pt complexes featuring stable Pt-P bonds in nanocomposite formulations with biocompatible amphiphilic polymers.
Collapse
Affiliation(s)
- Pan Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Jian-Wei Wang
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, P. R. China
| | - Wen-Hua Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Hongzhen Bai
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, P. R. China
| | - Guping Tang
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, P. R. China
| | - David J Young
- College of Engineering Information Technology & Environment, Charles Darwin University, Darwin, Northern Territory, 0909, Australia
| |
Collapse
|