1
|
Sasanipoor F, Zhang Z. Molybdenum Disulfide Nanocomposites for Cancer Diagnosis and Therapeutics: Biosensors, Bioimaging, and Phototherapy. Adv Healthc Mater 2025; 14:e2500655. [PMID: 40289409 DOI: 10.1002/adhm.202500655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/10/2025] [Indexed: 04/30/2025]
Abstract
Molybdenum disulfide (MoS₂) nanomaterials have attracted significant interest in cancer diagnosis and therapy due to their unique physicochemical properties. Due to its extensive surface area and adaptable structure, MoS₂ may engage with pharmaceuticals and biomolecules via covalent and non-covalent interactions. This versatility enhances the sensitivity of identifying specific biomarkers, colloidal stability, and tumor-targeting capabilities. In the near-infrared (NIR) spectrum, MoS₂ exhibits strong optical absorption and efficient photothermal conversion, making it suitable for NIR-driven phototherapy and regulated medication release. Functionalized MoS₂ nanocomposites react differently to the tumor microenvironment, which improves treatment effectiveness by increasing drug accumulation at cancer sites and decreasing off-target effects on healthy tissues. Recent developments in MoS₂-based nanocomposites for cancer detection and treatment are reviewed in this study, with particular attention paid to their uses in photothermal therapy, photodynamic therapy, biosensing, and bioimaging. Additionally, it looks at the difficulties and potential applications of MoS₂ nanocomposites in cancer.
Collapse
Affiliation(s)
- Fatemeh Sasanipoor
- Department of Chemistry, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| | - Zhiqing Zhang
- Department of Chemistry, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| |
Collapse
|
2
|
Sahoo SS, Manna D. Nanomaterial-Triggered Ferroptosis and Cuproptosis in Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412462. [PMID: 40018870 DOI: 10.1002/smll.202412462] [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: 12/20/2024] [Revised: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains one of the leading causes of the death of individuals globally. Conventional treatment techniques like chemotherapy and radiation often suffer various drawbacks like toxicity and drug resistance. The study of cell death has been predominantly focused on classical forms like apoptosis, but the role of metal ions in governing controlled cell death is a fascinating and less explored area. Metal-mediated controlled cell death is a process where metal triggers cell death via a unique mechanism. Nanomaterial-based strategies have gained attention for their ability to deliver precise therapeutic agents while also triggering Regulated Cell Death (RCD) mechanisms in cancer cells. The recently discovered metal-mediated controlled cell death techniques like cuproptosis and ferroptosis can be used in cancer treatment as they can be used selectively for the treatment of drug-resistant cancer. Nano material-based delivery system can also be used for the precise delivery of the drug to the targeted sites. In this review, we have given some idea about the mechanism of metal-mediated controlled cell death techniques (ferroptosis and cuproptosis) and how we can initiate controlled cell deaths using nanomaterials for cancer treatment.
Collapse
Affiliation(s)
- Suman Sekhar Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| | - Debasish Manna
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
3
|
Xu W, Guan G, Yue R, Dong Z, Lei L, Kang H, Song G. Chemical Design of Magnetic Nanomaterials for Imaging and Ferroptosis-Based Cancer Therapy. Chem Rev 2025; 125:1897-1961. [PMID: 39951340 DOI: 10.1021/acs.chemrev.4c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of regulatory cell death, has garnered significant interest as a therapeutic target in cancer treatment due to its distinct characteristics, including lipid peroxide generation and redox imbalance. However, its clinical application in oncology is currently limited by issues such as suboptimal efficacy and potential off-target effects. The advent of nanotechnology has provided a new way for overcoming these challenges through the development of activatable magnetic nanoparticles (MNPs). These innovative MNPs are designed to improve the specificity and efficacy of ferroptosis induction. This Review delves into the chemical and biological principles guiding the design of MNPs for ferroptosis-based cancer therapies and imaging-guided therapies. It discusses the regulatory mechanisms and biological attributes of ferroptosis, the chemical composition of MNPs, their mechanism of action as ferroptosis inducers, and their integration with advanced imaging techniques for therapeutic monitoring. Additionally, we examine the convergence of ferroptosis with other therapeutic strategies, including chemodynamic therapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, within the context of nanomedicine strategies utilizing MNPs. This Review highlights the potential of these multifunctional MNPs to surpass the limitations of conventional treatments, envisioning a future of drug-resistance-free, precision diagnostics and ferroptosis-based therapies for treating recalcitrant cancers.
Collapse
Affiliation(s)
- Wei Xu
- School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Guoqiang Guan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, PR China
| | - Zhe Dong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Lingling Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, 12 Seoul 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| |
Collapse
|
4
|
Liu Y, Liu Y, Li X, Li S, Zhang X, Si L, Jiang S, Hu J, Chen J. Versatile Nanomaterials That Interfere with Ferroptosis in the Tumor Microenvironment. Int J Nanomedicine 2025; 20:2461-2473. [PMID: 40027870 PMCID: PMC11871933 DOI: 10.2147/ijn.s508767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a type of iron-dependent programmed cell death characterized by a depletion of glutathione. Although generally less harmful to normal cells, in tumor cells, the high demand for iron ions provides conditions conducive to ferroptosis. In this review, we provide an overview of recent progress in research on the regulation of ferroptosis in tumor cells, summarizing and assessing the current state, trends, and applications of nanomaterials in the regulation of ferroptosis in tumor cells. Given the advantages of nanomaterials in terms of targeting, safety, improved drug efficacy, and reduced side effects, these materials are considered to have potential therapeutic value in modulating ferroptosis in tumor cells via different mechanisms. In this respect, we describe methods for modifying the regulation of iron ions and interfering with glutathione activity and lipid peroxidation. The development of nanomaterials that can be applied to induce or inhibit ferroptosis is anticipated to provide new therapeutic options for the treatment of a diverse range of diseases.
Collapse
Affiliation(s)
- Yurong Liu
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Yunheng Liu
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Xinting Li
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Song Li
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Xiaokang Zhang
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Longqing Si
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Shaojing Jiang
- Yantai Engineering Research Center for Digital Technology of Stomatology, Characteristic Laboratories of Colleges and Universities in Shandong Province for Digital Stomatology, Institute of Stomatology, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Jinghui Hu
- Yantai Engineering Research Center for Digital Technology of Stomatology, Characteristic Laboratories of Colleges and Universities in Shandong Province for Digital Stomatology, Institute of Stomatology, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| | - Jing Chen
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People’s Republic of China
| |
Collapse
|
5
|
Xie B, Xiao Z, Ling J, Peng Y, Chen T. Exploring the application of metal-based photothermal agents in photothermal therapy combined with immune checkpoint therapy. Front Pharmacol 2025; 16:1553158. [PMID: 40017598 PMCID: PMC11865196 DOI: 10.3389/fphar.2025.1553158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Photothermal therapy (PTT), a popular local treatment that uses heat to ablate tumors, has limited efficacy in addressing metastatic and deeply located tumors when used alone. Integrating PTT with immunotherapy not only yields a synergistic effect but also promotes cancer regression and confers the benefit of immune memory, which can surmount the challenges faced by PTT when used in isolation. Metal-based nanomaterials, renowned for their superior photothermal conversion efficiency and distinctive photochemical properties, have been extensively researched and applied in the field of PTT. This review summarizes the latest developments in combination therapies, with a specific focus on the combination of PTT and immune checkpoint therapy (ICT) for cancer treatment, including a comprehensive overview of the recent advancements in noble metal-based and 2D transition metal chalcogenides (TMDCs)-based photothermal agents, and their anticancer effect when combining PTT with immune checkpoint blockades (anti-CTLA-4 and anti-PD-L1) therapy. The goal of this review is to present an overview of the application, current challenges and future prospects of metal-based photothermal agents in PTT combined with ICT for cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Yichao Peng
- Department of Pharmacy and General Surgery of Puning People’s Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, China
| | - Tianfeng Chen
- Department of Pharmacy and General Surgery of Puning People’s Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, China
| |
Collapse
|
6
|
Du Y, Luo Y, Gu Z. Molecular dynamics simulations reveal concentration-dependent blockage of graphene quantum dots to water channel protein openings. Sci Rep 2024; 14:26485. [PMID: 39489799 PMCID: PMC11532551 DOI: 10.1038/s41598-024-77592-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Graphene quantum dots (GQDs) have attracted significant attention across various scientific research areas due to their exceptional properties. However, studies on the potential toxicity of GQDs have yielded conflicting results. Therefore, a comprehensive evaluation of the toxicity profile of GQDs is essential for a thorough understanding of their biosafety. In this work, employing a molecular dynamics (MD) simulation approach, we investigate the interactions between GQDs and graphene oxide quantum dots (GOQDs) with the AQP1 water channel protein, aiming to explore the potential biological influence of GQDs/GOQDs. Our MD simulation results reveal that GQDs can adsorb to the loop region around the openings of AQP1 water channels, resulting in the blockage of these channels and potential toxicity. Interestingly, this blockage is concentration-dependent, with higher GQD concentrations leading to a greater likelihood of blockage. Additionally, GOQDs show a lower probability of blocking the openings of AQP1 water channels compared to GQDs, due to the hydrophobicity of the loop regions around the openings, which ultimately leads to lower interaction energy. Therefore, these findings provide new insights into the potential adverse impact of GQDs on AQP1 water channels through the blockage of their openings, offering valuable molecular insights into the toxicity profile of GQD nanomaterials.
Collapse
Affiliation(s)
- Yunbo Du
- Department of Critical Care Medicine, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Road, Longhua District, Shenzhen, 518110, Guangdong Province, China
| | - Yuqi Luo
- Department of Gastrointestinal and Hepatobiliary Surgery, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Road, Longhua District, Shenzhen, 518110, Guangdong Province, China.
| | - Zonglin Gu
- College of Physical Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, China
| |
Collapse
|
7
|
Li M, Liu Z, Tang J, Cheng L, Xue Y, Liu Y, Liu J. Facile Synthesis of a Multifunctional Porous Organic Polymer Nanosonosensitizer (mHM@HMME) for Enhanced Cancer Sonodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28104-28117. [PMID: 38769350 DOI: 10.1021/acsami.4c02651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Sonodynamic therapy (SDT), which involves the activation of sonosensitizers to generate cytotoxic reactive oxygen species under ultrasound irradiation, is a promising noninvasive modality for cancer treatment. However, the clinical translational application of SDT is impeded by the lack of efficient sonosensitizers, the inefficient accumulation of sonosensitizers at tumor sites, and the complicated immunosuppressive tumor microenvironment. Herein, we developed a facilely synthesized multifunctional porous organic polymer nanosonosensitizer (mHM@HMME) for enhanced SDT. Specifically, mHM@HMME nanosonosensitizers were prepared by incorporating chemotherapeutic mitoxantrone into the one-step synthesis process of disulfide bond containing porous organic polymers, followed by loading with organic sonosensitizer (HMME) and camouflaging with a cancer cell membrane. Due to the cancer cell membrane camouflage, this multifunctional mHM@HMME nanosonosensitizer showed prolonged blood circulation and tumor targeting aggregation. Under ultrasound irradiation, the mHM@HMME nanosonosensitizer exhibited a satisfactory SDT performance both in vitro and in vivo. Moreover, the potent SDT combined with glutathione-responsive drug release in tumor cells induced robust immunogenic cell death to enhance the antitumor effect of SDT in turn. Overall, this facilely synthesized multifunctional mHM@HMME nanosonosensitizer shows great potential application in enhanced SDT.
Collapse
Affiliation(s)
- Meiting Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Zhuoyin Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Junjie Tang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - LiLi Cheng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Yifan Xue
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Yadong Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| |
Collapse
|
8
|
Li X, Lin H, Hu J, Fang J, Liu H, Fu C, Zhao K. A redox homeostasis disruptor based on a biodegradable nanoplatform for ultrasound (US) imaging-guided high-performance ferroptosis therapy of tumors. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2351354. [PMID: 38800054 PMCID: PMC11123443 DOI: 10.1080/14686996.2024.2351354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
The synergistic disruption of intracellular redox homeostasis through the combination of ferroptosis/gas therapy shows promise in enhancing the antitumor efficacy. However, the development of an optimal delivery system encounters significant challenges, including effective storage, precise delivery, and controlled release of therapeutic gas. In this study, we propose the utilization of a redox homeostasis disruptor that is selectively activated by the tumor microenvironment (TME), in conjunction with our newly developed nanoplatforms (MC@HMOS@Au@RGD), for highly efficient ferroptosis therapy of tumors. The TME-triggered degradation of HMOS initiates the release of MC and AuNPs from the MC@HMOS@Au@RGD nanoplatform. The released MC subsequently reacts with endogenous hydrogen peroxide (H2O2) and H+ to enable the on-demand release of CO gas, leading to mitochondrial damage. Simultaneously, the released AuNPs exhibit GOx-like activity, catalyzing glucose to generate gluconic acid and H2O2. This process not only promotes the decomposition of MnCO to enhance CO production but also enhances the Fenton-like reaction between Mn2+ and H2O2, generating ROS through the modulation of the H+ and H2O2-enriched TME. Moreover, the generation of CO bubbles enables the monitoring of the ferroptosis treatment process through ultrasound (US) imaging. The efficacy of our prepared MC@HMOS@Au@RGD disruptors in ferroptosis therapy is validated through both in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Xia Li
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huijian Lin
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianbo Hu
- Medical Imaging Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiajin Fang
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Science Experiment Center, Guangdong Huayan Biomedical Technology Centre, Guangzhou, China
| | - Hongsheng Liu
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Science Experiment Center, Guangdong Huayan Biomedical Technology Centre, Guangzhou, China
| | - Can Fu
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kewei Zhao
- Laboratory Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Ghosh S, Lai JY. An insight into the dual role of MoS2-based nanocarriers in anticancer drug delivery and therapy. Acta Biomater 2024; 179:36-60. [PMID: 38552760 DOI: 10.1016/j.actbio.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
Over the years, nanomaterials have been exploited as drug delivery systems and therapeutic agents in cancer treatment. Special emphasis has been placed on structure and shape-mediated drug loading and release. Functional materials, including molybdenum disulfide (MoS2), have shown promising results because of their tunable structure and unmatched physicochemical properties. Specifically, easy surface functionalization and high drug adsorption ability make them ideal candidates. Although the large surface area of nanosheets/nanoflakes may result in high drug loading, the encapsulation efficiency is better for MoS2 nanoflower structures. Due to its high targeting abilities, the loading of chemotherapeutic drugs onto MoS2 may minimize nonspecific cellular death and undesired side effects. Furthermore, due to their strong light-absorption ability, MoS2 nanostructures have been widely exploited as photothermal and photodynamic therapeutic agents. The unexplored dimensions of cancer therapy, including chemodynamic (Fenton-like reaction) and piezo-catalytic (ultrasound-mediated reactive oxygen generation), have been recently unlocked, in which the catalytic properties of MoS2 are utilized to generate toxic free radicals to eliminate cancer. Intriguingly, combining these therapeutic modalities often results in high therapeutic efficacy at low doses and minimizes side effects. With a plethora of recent studies, a thorough analysis of current findings is crucial. Therefore, this review discusses the major advances in this field of research. A brief commentary on the limitations/future outlook/ethical issues of the clinical translation of MoS2-mediated cancer treatments is also deliberated. Overall, in our observations, the MoS2-based nanoformulations hold great potential for future cancer therapy applications. STATEMENT OF SIGNIFICANCE: Development of nanomedicines based on MoS2 has opened new avenues in cancer treatment. The MoS2 with different morphologies (nanosheet/nanoflower/QDs) has shown promising results in controlled and targeted drug delivery, leading to minimized side effects and increased therapeutic efficacy. While existing reviews have primarily focused on the optical/thermal properties utilized in photodynamic/photothermal therapy, the outstanding catalytic properties of MoS2 utilized in cancer therapies (chemodynamic/piezo-catalytic) are often overlooked. This review critically highlights and praises/criticizes individual articles reporting the MoS2-based nanoplatforms for cancer therapy applications. Additionally, MoS2-based combined therapies for synergistic effects are discussed. Furthermore, a brief commentary on the future prospects for clinical translations is also deliberated, which is appealing to various research communities engaged in cancer theranostics and biomedical sciences research.
Collapse
Affiliation(s)
- Sandip Ghosh
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Jui-Yang Lai
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; Center for Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
10
|
Shao L, Wang X, Du X, Yin S, Qian Y, Yao Y, Yang L. Application of Multifunctional Nanozymes in Tumor Therapy. ACS OMEGA 2024; 9:15753-15767. [PMID: 38617672 PMCID: PMC11007812 DOI: 10.1021/acsomega.4c00258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/25/2024] [Accepted: 03/13/2024] [Indexed: 04/16/2024]
Abstract
Tumors are one of the main diseases threatening human life and health. The emergence of nanotechnology in recent years has introduced a novel therapeutic avenue for addressing tumors. Through the amalgamation of nanotechnology's inherent attributes with those of natural enzymes, nanozymes have demonstrated the ability to initiate catalytic reactions, modulate the biological microenvironment, and facilitate the adoption of multifaceted therapeutic approaches, thereby exhibiting considerable promise in the realm of cancer treatment. In this Review, the application of nanozymes in chemodynamic therapy, radiotherapy, photodynamic therapy, photothermal therapy, and starvation therapy are summarized. Moreover, a detailed discussion regarding the mechanism of conferring physiotherapeutic functionality upon catalytic nanosystems is provided. It is posited that this innovative catalytic treatment holds significant potential to play a crucial role within the domain of nanomedicine.
Collapse
Affiliation(s)
- Lihua Shao
- Department
of Colorectal Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Xueyuan Wang
- College of
Life Science, Nanjing Normal University, Nanjing, Jiangsu 210046, China
| | - Xiao Du
- Department
of Pharmacy, Nanjing Medical Center for Clinical Pharmacy, Nanjing
Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Shaoping Yin
- School of
Pharmacy, Nanjing University of Chinese
Medicine, Nanjing, Jiangsu 210023, China
| | - Yun Qian
- Dermatologic
Surgery Department, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing, Jiangsu 210042, China
| | - Yawen Yao
- Department
of Pharmaceutics, School of Pharmacy, China
Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Lin Yang
- College of
Science, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| |
Collapse
|
11
|
Wei R, Fu G, Li Z, Liu Y, Xue M. Engineering Iron-Based Nanomaterials for Breast Cancer Therapy Associated with Ferroptosis. Nanomedicine (Lond) 2024; 19:537-555. [PMID: 38293902 DOI: 10.2217/nnm-2023-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/08/2023] [Indexed: 02/01/2024] Open
Abstract
Ferroptosis has received increasing attention as a novel nonapoptotic programmed death. Recently, iron-based nanomaterials have been extensively exploited for efficient tumor ferroptosis therapy, as they directly release high concentrations of iron and increase intracellular reactive oxygen species levels. Breast cancer is one of the commonest malignant tumors in women; inhibiting breast cancer cell proliferation through activating the ferroptosis pathway could be a potential new target for patient treatment. Here, we briefly introduce the background of ferroptosis and systematically review the current cancer therapeutic strategies based on iron-based ferroptosis inducers. Finally, we summarize the advantages of these various ferroptosis inducers and shed light on future perspectives. This review aims to provide better guidance for the development of iron-based nanomaterial ferroptosis inducers.
Collapse
Affiliation(s)
- Ruixue Wei
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Gaoliang Fu
- Henan Provincial Key Laboratory of Nanocomposites & Applications, Institute of Nanostructured Functional Materials, Huanghe Science & Technology College, Zhengzhou, 450006, Henan, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
12
|
Xiao P, Li C, Liu Y, Gao Y, Liang X, Liu C, Yang W. The role of metal ions in the occurrence, progression, drug resistance, and biological characteristics of gastric cancer. Front Pharmacol 2024; 15:1333543. [PMID: 38370477 PMCID: PMC10869614 DOI: 10.3389/fphar.2024.1333543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Metal ions exert pivotal functions within the human body, encompassing essential roles in upholding cell structure, gene expression regulation, and catalytic enzyme activity. Additionally, they significantly influence various pathways implicated in divergent mechanisms of cell death. Among the prevailing malignant tumors of the digestive tract worldwide, gastric cancer stands prominent, exhibiting persistent high mortality rates. A compelling body of evidence reveals conspicuous ion irregularities in tumor tissues, encompassing gastric cancer. Notably, metal ions have been observed to elicit distinct contributions to the progression, drug resistance, and biological attributes of gastric cancer. This review consolidates pertinent literature on the involvement of metal ions in the etiology and advancement of gastric cancer. Particular attention is directed towards metal ions, namely, Na, K, Mg, Ca, Fe, Cu, Zn, and Mn, elucidating their roles in the initiation and progression of gastric cancer, cellular demise processes, drug resistance phenomena, and therapeutic approaches.
Collapse
Affiliation(s)
- Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Gao
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaojing Liang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
13
|
Farzipour S, Zefrei FJ, Bahadorikhalili S, Alvandi M, Salari A, Shaghaghi Z. Nanotechnology Utilizing Ferroptosis Inducers in Cancer Treatment. Anticancer Agents Med Chem 2024; 24:571-589. [PMID: 38275050 DOI: 10.2174/0118715206278427231215111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
Current cancer treatment options have presented numerous challenges in terms of reaching high efficacy. As a result, an immediate step must be taken to create novel therapies that can achieve more than satisfying outcomes in the fight against tumors. Ferroptosis, an emerging form of regulated cell death (RCD) that is reliant on iron and reactive oxygen species, has garnered significant attention in the field of cancer therapy. Ferroptosis has been reported to be induced by a variety of small molecule compounds known as ferroptosis inducers (FINs), as well as several licensed chemotherapy medicines. These compounds' low solubility, systemic toxicity, and limited capacity to target tumors are some of the significant limitations that have hindered their clinical effectiveness. A novel cancer therapy paradigm has been created by the hypothesis that ferroptosis induced by nanoparticles has superior preclinical properties to that induced by small drugs and can overcome apoptosis resistance. Knowing the different ideas behind the preparation of nanomaterials that target ferroptosis can be very helpful in generating new ideas. Simultaneously, more improvement in nanomaterial design is needed to make them appropriate for therapeutic treatment. This paper first discusses the fundamentals of nanomedicine-based ferroptosis to highlight the potential and characteristics of ferroptosis in the context of cancer treatment. The latest study on nanomedicine applications for ferroptosis-based anticancer therapy is then highlighted.
Collapse
Affiliation(s)
- Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali Zefrei
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahadorikhalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, 43007, Tarragona, Spain
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Shaghaghi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
14
|
Self A, Farell M, Samineni L, Kumar M, Gomez EW. 2D Materials for Combination Therapy to Address Challenges in the Treatment of Cancer. ADVANCED NANOBIOMED RESEARCH 2023; 3. [DOI: 10.1002/anbr.202300070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
2D materials exhibit a variety of characteristics that make them appealing platforms for cancer treatment such as high drug loading capacity and photothermal and photodynamic properties. A key advantage of 2D material platforms for oncological applications is the ability to harness multiple modalities including drug delivery, photothermal therapy, photodynamic therapy, chemodynamic therapy, gene delivery, and immunotherapy approaches for improved efficacy. In this review, a comparison of the unique properties of different classes of 2D materials that enable their usage as platforms for multimodal therapy is provided. Further, the benefits and drawbacks of different platforms are also highlighted. Finally, current challenges and emerging opportunities for future development of 2D materials to further enable combination therapy and translation from the bench to clinical oncology applications are discussed.
Collapse
Affiliation(s)
- Ava Self
- Department of Chemical Engineering The Pennsylvania State University University Park PA 16802 USA
| | - Megan Farell
- Department of Chemical Engineering The Pennsylvania State University University Park PA 16802 USA
| | - Laximicharan Samineni
- Department of Civil, Architectural, and Environmental Engineering The University of Texas at Austin Austin TX 78712 USA
- McKetta Department of Chemical Engineering The University of Texas at Austin Austin TX 78712 USA
| | - Manish Kumar
- Department of Civil, Architectural, and Environmental Engineering The University of Texas at Austin Austin TX 78712 USA
- McKetta Department of Chemical Engineering The University of Texas at Austin Austin TX 78712 USA
| | - Esther W. Gomez
- Department of Chemical Engineering The Pennsylvania State University University Park PA 16802 USA
- Department of Biomedical Engineering The Pennsylvania State University University Park PA 16802 USA
| |
Collapse
|
15
|
Yadav VK, Choudhary N, Gacem A, Verma RK, Abul Hasan M, Tarique Imam M, Almalki ZS, Yadav KK, Park HK, Ghosh T, Kumar P, Patel A, Kalasariya H, Jeon BH, Ali AlMubarak H. Deeper insight into ferroptosis: association with Alzheimer's, Parkinson's disease, and brain tumors and their possible treatment by nanomaterials induced ferroptosis. Redox Rep 2023; 28:2269331. [PMID: 38010378 PMCID: PMC11001282 DOI: 10.1080/13510002.2023.2269331] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Ferroptosis is an emerging and novel type of iron-dependent programmed cell death which is mainly caused by the excessive deposition of free intracellular iron in the brain cells. This deposited free iron exerts a ferroptosis pathway, resulting in lipid peroxidation (LiPr). There are mainly three ferroptosis pathways viz. iron metabolism-mediated cysteine/glutamate, and LiPr-mediated. Iron is required by the brain as a redox metal for several physiological activities. Due to the iron homeostasis balance disruption, the brain gets adversely affected which further causes neurodegenerative diseases (NDDs) like Parkinson's and Alzheimer's disease, strokes, and brain tumors like glioblastoma (GBS), and glioma. Nanotechnology has played an important role in the prevention and treatment of these NDDs. A synergistic effect of nanomaterials and ferroptosis could prove to be an effective and efficient approach in the field of nanomedicine. In the current review, the authors have highlighted all the latest research in the field of ferroptosis, specifically emphasizing on the role of major molecular key players and various mechanisms involved in the ferroptosis pathway. Moreover, here the authors have also addressed the correlation of ferroptosis with the pathophysiology of NDDs and theragnostic effect of ferroptosis and nanomaterials for the prevention and treatment of NDDs.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Nisha Choudhary
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Amel Gacem
- Department of Physics, Faculty of Sciences, University 20 Août 1955, Skikda, Algeria
| | - Rakesh Kumar Verma
- Department of Biosciences, School of Liberal Arts & Sciences, Mody University of Science and Technology, Sikar, India
| | - Mohd Abul Hasan
- Civil Engineering Department, College of Engineering, King Khalid University, Abha, Kingdom of Saudi Arabia (KSA)
| | - Mohammad Tarique Imam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Ziyad Saeed Almalki
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Krishna Kumar Yadav
- Faculty of Science and Technology, Madhyanchal Professional University, Bhopal, India
- Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Nasiriyah, Iraq
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Tathagata Ghosh
- Department of Arts, School of Liberal Arts & Sciences, Mody University of Science and Technology, Sikar, India
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Haresh Kalasariya
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hassan Ali AlMubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University (KKU), Abha, Kingdom of Saudi Arabia
| |
Collapse
|
16
|
Sun S, Shen J, Jiang J, Wang F, Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther 2023; 8:372. [PMID: 37735472 PMCID: PMC10514338 DOI: 10.1038/s41392-023-01606-1] [Citation(s) in RCA: 204] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death with distinct characteristics, including altered iron homeostasis, reduced defense against oxidative stress, and abnormal lipid peroxidation. Recent studies have provided compelling evidence supporting the notion that ferroptosis plays a key pathogenic role in many diseases such as various cancer types, neurodegenerative disease, diseases involving tissue and/or organ injury, and inflammatory and infectious diseases. Although the precise regulatory networks that underlie ferroptosis are largely unknown, particularly with respect to the initiation and progression of various diseases, ferroptosis is recognized as a bona fide target for the further development of treatment and prevention strategies. Over the past decade, considerable progress has been made in developing pharmacological agonists and antagonists for the treatment of these ferroptosis-related conditions. Here, we provide a detailed overview of our current knowledge regarding ferroptosis, its pathological roles, and its regulation during disease progression. Focusing on the use of chemical tools that target ferroptosis in preclinical studies, we also summarize recent advances in targeting ferroptosis across the growing spectrum of ferroptosis-associated pathogenic conditions. Finally, we discuss new challenges and opportunities for targeting ferroptosis as a potential strategy for treating ferroptosis-related diseases.
Collapse
Affiliation(s)
- Shumin Sun
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Shen
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Jiang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
17
|
Liu B, Jiang W, Ye Y, Liu L, Wei X, Zhang Q, Xing B. 2D MoS 2 Nanosheets Induce Ferroptosis by Promoting NCOA4-Dependent Ferritinophagy and Inhibiting Ferroportin. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208063. [PMID: 36908089 DOI: 10.1002/smll.202208063] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/22/2023] [Indexed: 06/15/2023]
Abstract
The exposure of MoS2 nanosheets can cause cytotoxicity, which causes health risks and affects its medical applications. However, knowledge of the underlying molecular mechanisms remains limited. This study reports that MoS2 nanosheets induces ferroptosis in vivo and in vitro, which is caused by the nanosheet themselves rather than by the dissolved ions. MoS2 nanosheets induce ferroptosis in epithelial (BEAS-2B) and macrophage (RAW264.7) cells due to nuclear receptor coactivator 4 (NCOA4)-dependent excusive ferritinophagy and the inhibition of ferroportin-1 (FPN). In this process, most of the MoS2 nanosheets enter the cells via macropinocytosis and are localized to the lysosome, contributing to an increase in the lysosomal membrane permeability. At the same time, NCOA4-dependent ferritinophagy is activated, and ferritin is degraded in the lysosome, which generates Fe2+ .Fe2+ leaks into the cytoplasm, leading to ferroptosis. Furthermore, the inhibition of FPN further aggravates the overload of Fe2+ in the cell. It has also been observed that ferroptosis is increased in lung tissue in mouse models exposed to MoS2 nanosheets. This work highlights a novel mechanism by which MoS2 nanosheets induce ferroptosis by promoting NCOA4-dependent ferritinophagy and inhibiting FPN, which could be of importance to elucidate the toxicity and identify the medical applications of 2D nanoparticles.
Collapse
Affiliation(s)
- Bingyan Liu
- Environment Research Institute, Shandong University, Qingdao, 266237, P. R. China
| | - Wei Jiang
- Environment Research Institute, Shandong University, Qingdao, 266237, P. R. China
| | - Yiyuan Ye
- Environment Research Institute, Shandong University, Qingdao, 266237, P. R. China
| | - Ling Liu
- Environment Research Institute, Shandong University, Qingdao, 266237, P. R. China
- Marine College, Shandong University, Weihai, 264209, P. R. China
| | - Xiaoran Wei
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266071, P. R. China
| | - Qiu Zhang
- School of Environmental Science and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Baoshan Xing
- Stockbridge School of Agriculture, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
18
|
Venkatesan J, Hur W, Gupta PK, Son SE, Lee HB, Lee SJ, Ha CH, Hwa CS, Kim DH, Seong GH. Gum Arabic-mediated liquid exfoliation of transition metal dichalcogenides as photothermic anti-breast cancer candidates. Int J Biol Macromol 2023:124982. [PMID: 37244326 DOI: 10.1016/j.ijbiomac.2023.124982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
Transition metal dichalcogenides (TMDs) have gained considerable attention for a broad range of applications, including cancer therapy. Production of TMD nanosheets using liquid exfoliation provides an inexpensive and facile route to achieve high yields. In this study, we developed TMD nanosheets using gum arabic as an exfoliating and stabilizing agent. Different types of TMDs, including MoS2, WS2, MoSe2, and WSe2 nanosheets, were produced using gum arabic and were characterized physicochemically. The developed gum arabic TMD nanosheets exhibited a remarkable photothermal absorption capacity in the near-infrared (NIR) region (808 nm and 1 W⋅cm-2). The drug doxorubicin was loaded on the gum arabic-MoSe2 nanosheets (Dox-G-MoSe2), and the anticancer activity was evaluated using MDA-MB-231 cells and a water-soluble tetrazolium salt (WST-1) assay, live and dead cell assays, and flow cytometry. Dox-G-MoSe2 significantly inhibited MDA-MB-231 cancer cell proliferation under the illumination ofan NIR laser at 808 nm. These results indicate that Dox-G-MoSe2 is a potentially valuable biomaterial for breast cancer therapy.
Collapse
Affiliation(s)
- Jayachandran Venkatesan
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea; Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya Deemed to be University, Deralakatte, Mangaluru 575018, India
| | - Won Hur
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Pramod K Gupta
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Seong Eun Son
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Han Been Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Su Jeong Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Chang Hyeon Ha
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Cheon Se Hwa
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Do Hyeon Kim
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Gi Hun Seong
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea.
| |
Collapse
|
19
|
Xie Z, Zhou Q, Qiu C, Zhu D, Li K, Huang H. Inaugurating a novel adjuvant therapy in urological cancers: Ferroptosis. CANCER PATHOGENESIS AND THERAPY 2023; 1:127-140. [PMID: 38328400 PMCID: PMC10846326 DOI: 10.1016/j.cpt.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 02/09/2024]
Abstract
Ferroptosis, a distinctive form of programmed cell death, is involved in numerous diseases with specific characteristics, including certain cell morphology, functions, biochemistry, and genetics, that differ from other forms of programmed cell death, such as apoptosis. Many studies have explored ferroptosis and its associated mechanisms, drugs, and clinical applications in diseases such as kidney injury, stroke, ischemia-reperfusion injury, and prostate cancer. In this review, we summarize the regulatory mechanisms of some ferroptosis inducers, such as enzalutamide and erastin. These are current research focuses and have already been studied extensively. In summary, this review focuses on the use of ferroptosis induction as a therapeutic strategy for treating tumors of the urinary system.
Collapse
Affiliation(s)
- Zhaoxiang Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Cheng Qiu
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Dingjun Zhu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Kaiwen Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China
| |
Collapse
|
20
|
He Z, Guo Y, Chen J, Luo H, Liu X, Zhang X, Sun Y, Ge D, Ye S, Shi W. Unsaturated phospholipid modified FeOCl nanosheets for enhancing tumor ferroptosis. J Mater Chem B 2023; 11:1891-1903. [PMID: 36744515 DOI: 10.1039/d2tb01854c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Iron-dependent accumulation of reactive oxygen species (ROS) and lipid peroxidation play key roles in ferroptosis, which has been an attractive strategy to kill tumor cells. However, the rapid annihilation of hydroxyl radicals (˙OH) produced from the Fenton reaction has become a major obstacle in inducing lipid peroxidation in cells. In this study, we develop a nano-delivery system of unsaturated phospholipid (Lip) and polyacrylic acid (PAA) functionalized FeOCl nanosheets (FeOCl@PAA-Lip). In this system, the ˙OH radicals produced from the Fenton reaction between FeOCl nanosheets and endogenous H2O2 of tumor cells attack Lip on the nanosheets in situ to initiate the lipid peroxidation chain reaction, which not only realizes free radical conversion but also leads to the amplification of ROS and lipid peroxides, thus enhancing tumor ferroptosis. The in vitro and in vivo results confirmed that FeOCl@PAA-Lip nanosheets exhibited specific tumor cell-killing effects, good biocompatibility, long circulation time, low side effects, high tumor targeting and an excellent tumor inhibition rate (73%). The Lip functionalization strategy offers a paradigm of enhancing ferroptosis treatment by conversion of ˙OH/phospholipid radicals/lipid peroxyl radicals and strengthening lipid peroxidation.
Collapse
Affiliation(s)
- Zi He
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Yijun Guo
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Jinzhu Chen
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Huiling Luo
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Xinxin Liu
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Xiuming Zhang
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Yanan Sun
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Dongtao Ge
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Shefang Ye
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| | - Wei Shi
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province/Research Center of Biomedical Engineering of Xiamen, Fujian Key Laboratory of Surface and Interface Engineering for High Performance Materials, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
21
|
Yang F, Xiao Y, Ding JH, Jin X, Ma D, Li DQ, Shi JX, Huang W, Wang YP, Jiang YZ, Shao ZM. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab 2023; 35:84-100.e8. [PMID: 36257316 DOI: 10.1016/j.cmet.2022.09.021] [Citation(s) in RCA: 268] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/24/2022] [Accepted: 09/21/2022] [Indexed: 01/20/2023]
Abstract
Treatment of triple-negative breast cancer (TNBC) remains challenging. Deciphering the orchestration of metabolic pathways in regulating ferroptosis will provide new insights into TNBC therapeutic strategies. Here, we integrated the multiomics data of our large TNBC cohort (n = 465) to develop the ferroptosis atlas. We discovered that TNBCs had heterogeneous phenotypes in ferroptosis-related metabolites and metabolic pathways. The luminal androgen receptor (LAR) subtype of TNBC was characterized by the upregulation of oxidized phosphatidylethanolamines and glutathione metabolism (especially GPX4), which allowed the utilization of GPX4 inhibitors to induce ferroptosis. Furthermore, we verified that GPX4 inhibition not only induced tumor ferroptosis but also enhanced antitumor immunity. The combination of GPX4 inhibitors and anti-PD1 possessed greater therapeutic efficacy than monotherapy. Clinically, higher GPX4 expression correlated with lower cytolytic scores and worse prognosis in immunotherapy cohorts. Collectively, this study demonstrated the ferroptosis landscape of TNBC and revealed an innovative immunotherapy combination strategy for refractory LAR tumors.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Jia-Han Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xi Jin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Da-Qiang Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jin-Xiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai 201203, China
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai 201203, China
| | - Yi-Ping Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
22
|
Wang Z, Zhang H, Wang L, Ma Z, Cui Y, Fu H, Yu C. Bibliometric analysis of ferroptosis: a comprehensive evaluation of its contribution to cancer immunity and immunotherapy. Front Oncol 2023; 13:1183405. [PMID: 37182170 PMCID: PMC10174302 DOI: 10.3389/fonc.2023.1183405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/06/2023] [Indexed: 05/16/2023] Open
Abstract
Background In the past 5 years, ferroptosis-associated cancer immunity has been attracted significant research interest. Objective This study was performed to identify and analyze the global output trend for ferroptosis in cancer immunity. Methods Relevant studies were retrieved from the Web of Science Core Collection on Feb 10th, 2023. The VOSviewer and Histcite softwares were utilized to perform the visual bibliometric and deep mining analyses. Results A total of 694 studies (530 articles (76.4%) and 164 (23.6%) review articles) were retrieved from the Web of Science Core Collection for visualization analyses. The top 3 key keywords were ferroptosis, prognosis and immunotherapy. The top 30 local citation score (LCS) authors were all collaborators of Zou Weiping. Deep mining of 51 nanoparticle-related articles showed that BIOMATERIALS was the most popular journal. The primary goal of gene signatures related to ferroptosis and cancer immunity was to establish prognostic predictions. Conclusion There has been a significant increase in ferroptosis-associated immune publications in the recent 3 years. The key research hotspots include mechanisms, prediction and therapeutic outcomes. The most influential article was from the Zou Weiping's group, which proposed that system xc-mediated ferroptosis is induced by CD8(+) T cell-secreted IFNγ after PD-L1 blockage for immunotherapy. The frontier of research in the field of ferroptosis-associated immune is the study on nanoparticle and gene signature The limitation of this bibliometric study is that publications on this topic are few.
Collapse
Affiliation(s)
- Zhen Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hui Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Li Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhen Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yu’ang Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Haitian Fu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
- *Correspondence: Chunjing Yu,
| |
Collapse
|
23
|
Biomaterial-assisted photoimmunotherapy for synergistic suppression of cancer progression. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
24
|
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X, Shi S. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol 2022; 15:174. [PMID: 36482419 PMCID: PMC9733270 DOI: 10.1186/s13045-022-01392-3] [Citation(s) in RCA: 435] [Impact Index Per Article: 145.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Many types of human cells self-destruct to maintain biological homeostasis and defend the body against pathogenic substances. This process, called regulated cell death (RCD), is important for various biological activities, including the clearance of aberrant cells. Thus, RCD pathways represented by apoptosis have increased in importance as a target for the development of cancer medications in recent years. However, because tumor cells show avoidance to apoptosis, which causes treatment resistance and recurrence, numerous studies have been devoted to alternative cancer cell mortality processes, namely necroptosis, pyroptosis, ferroptosis, and cuproptosis; these RCD modalities have been extensively studied and shown to be crucial to cancer therapy effectiveness. Furthermore, evidence suggests that tumor cells undergoing regulated death may alter the immunogenicity of the tumor microenvironment (TME) to some extent, rendering it more suitable for inhibiting cancer progression and metastasis. In addition, other types of cells and components in the TME undergo the abovementioned forms of death and induce immune attacks on tumor cells, resulting in enhanced antitumor responses. Hence, this review discusses the molecular processes and features of necroptosis, pyroptosis, ferroptosis, and cuproptosis and the effects of these novel RCD modalities on tumor cell proliferation and cancer metastasis. Importantly, it introduces the complex effects of novel forms of tumor cell death on the TME and the regulated death of other cells in the TME that affect tumor biology. It also summarizes the potential agents and nanoparticles that induce or inhibit novel RCD pathways and their therapeutic effects on cancer based on evidence from in vivo and in vitro studies and reports clinical trials in which RCD inducers have been evaluated as treatments for cancer patients. Lastly, we also summarized the impact of modulating the RCD processes on cancer drug resistance and the advantages of adding RCD modulators to cancer treatment over conventional treatments.
Collapse
Affiliation(s)
- Xuhui Tong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Tang
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Mingming Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiang Liu
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Recent progress in two-dimensional nanomaterials for cancer theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
27
|
Song C, Sun Q, Qin L, Chen M, Li Y, Niu D. Confined Construction of Ultrasmall Molybdenum Disulfide-Loaded Porous Silica Particles for Efficient Tumor Therapy. ACS Biomater Sci Eng 2022; 8:3377-3386. [PMID: 35861149 DOI: 10.1021/acsbiomaterials.2c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recently, molybdenum sulfide (MoS2) has shown great application potential in tumor treatment because of its good photothermal properties. Unfortunately, most of the current molybdenum disulfide-based nanotherapeutic agents suffer from complex preparation processes, low photothermal conversion efficiencies, and poor structural/compositional regulation. To address these issues, in this paper, a facile "confined solvothermal" method is proposed to construct an MoS2-loaded porous silica nanosystem (designated as MoS2@P-hSiO2). The maximum photothermal efficiency of 79.5% of molybdenum-based materials reported in the literature at present was obtained due to the ultrasmall MoS2 nanoclusters and the rich porous channels. Furthermore, both in vitro and in vivo experiments showed that the cascade hybrid system (MoS2/GOD@P-hSiO2) after efficient loading of glucose oxidase (GOD) displayed a significant tumor-suppressive effect and good biosafety through the combined effects of photothermal and enzyme-mediated cascade catalytic therapy. Consequently, this hybrid porous network system combining the in situ solvothermal strategy of inorganic functional components and the efficient encapsulation of organic enzyme macromolecules can provide a new pathway to construct synergistic agents for the efficient and safe treatment of tumors.
Collapse
Affiliation(s)
- Chunya Song
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, Shanghai Engineering Research Center of Hierarchical Nanomaterials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Qiqi Sun
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, Shanghai Engineering Research Center of Hierarchical Nanomaterials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Limei Qin
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, Shanghai Engineering Research Center of Hierarchical Nanomaterials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Meiwan Chen
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, Shanghai Engineering Research Center of Hierarchical Nanomaterials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Yongsheng Li
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, Shanghai Engineering Research Center of Hierarchical Nanomaterials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China.,Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, School of Chemistry and Chemical Engineering, Shihezi University, Shihezi 832003, People's Republic of China
| | - Dechao Niu
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, Shanghai Engineering Research Center of Hierarchical Nanomaterials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| |
Collapse
|
28
|
Liu X, Zhang Y, Wu X, Xu F, Ma H, Wu M, Xia Y. Targeting Ferroptosis Pathway to Combat Therapy Resistance and Metastasis of Cancer. Front Pharmacol 2022; 13:909821. [PMID: 35847022 PMCID: PMC9280276 DOI: 10.3389/fphar.2022.909821] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis is an iron-dependent regulated form of cell death caused by excessive lipid peroxidation. This form of cell death differed from known forms of cell death in morphological and biochemical features such as apoptosis, necrosis, and autophagy. Cancer cells require higher levels of iron to survive, which makes them highly susceptible to ferroptosis. Therefore, it was found to be closely related to the progression, treatment response, and metastasis of various cancer types. Numerous studies have found that the ferroptosis pathway is closely related to drug resistance and metastasis of cancer. Some cancer cells reduce their susceptibility to ferroptosis by downregulating the ferroptosis pathway, resulting in resistance to anticancer therapy. Induction of ferroptosis restores the sensitivity of drug-resistant cancer cells to standard treatments. Cancer cells that are resistant to conventional therapies or have a high propensity to metastasize might be particularly susceptible to ferroptosis. Some biological processes and cellular components, such as epithelial–mesenchymal transition (EMT) and noncoding RNAs, can influence cancer metastasis by regulating ferroptosis. Therefore, targeting ferroptosis may help suppress cancer metastasis. Those progresses revealed the importance of ferroptosis in cancer, In order to provide the detailed molecular mechanisms of ferroptosis in regulating therapy resistance and metastasis and strategies to overcome these barriers are not fully understood, we described the key molecular mechanisms of ferroptosis and its interaction with signaling pathways related to therapy resistance and metastasis. Furthermore, we summarized strategies for reversing resistance to targeted therapy, chemotherapy, radiotherapy, and immunotherapy and inhibiting cancer metastasis by modulating ferroptosis. Understanding the comprehensive regulatory mechanisms and signaling pathways of ferroptosis in cancer can provide new insights to enhance the efficacy of anticancer drugs, overcome drug resistance, and inhibit cancer metastasis.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yiqian Zhang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| | - Fuyan Xu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Ma
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengling Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
- *Correspondence: Yong Xia,
| |
Collapse
|
29
|
Zhang D, Meng Y, Song Y, Cui P, Hu Z, Zheng X. Precision therapy through breaking the intracellular redox balance with an MOF-based hydrogel intelligent nanobot for enhancing ferroptosis and activating immunotherapy. NANOSCALE 2022; 14:8441-8453. [PMID: 35647731 DOI: 10.1039/d2nr00950a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
With the advancement and development of nanomedicine, tumor precision therapy provides technical support for effective accumulation and targeted drug delivery, and reduces toxic side effects. In cancer cells, breaking the redox balance could induce cancer cell death. Herein, a novel iron-containing intelligent hydrogel nanobot (FeSe2-Ce6/MOF@HA/PEI/CpG@HHPA NPs, abbreviated as FSMH) is proposed to break the intracellular redox balance and trigger the immune response. The as-fabricated multifunctional FSMH could not only exert Fenton reactions in the acidic tumor microenvironment, converting hydrogen peroxide (H2O2) into highly toxic hydroxyl radicals (˙OH), but also effectively consume GSH to attenuate the intracellular oxidative stress. The negative charge of the FSMH nanohydrogel system guarantees its superexcellent stabilization in blood circulation and optimal tumor collection. Subsequently, the surface charge of the endocytosed FSMH was transformed to a positive charge after exposure to the acidic tumor environment, further improving its tumor collection and locally releasing Fe ions and immune adjuvants. Furthermore, Ce6 was released in a pH-responsive manner in the acidic microenvironment. In the presence of near-infrared light, singlet oxygen was produced by the FSMH nanohydrogel system, to ablate tumors and promote the maturation of dendritic cells, achieving the precision-combined strategies effect of CDT, PDT, and immunotherapy.
Collapse
Affiliation(s)
- Dongsheng Zhang
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P.R. China.
| | - Yanfei Meng
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P.R. China.
- College of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P.R. China
| | - Yingzi Song
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P.R. China.
- College of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P.R. China
| | - Ping Cui
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P.R. China.
- College of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P.R. China
| | - Zunfu Hu
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P.R. China.
- School of Material Science and Engineering, Linyi University, Linyi 276000, Shandong, P.R. China
| | - Xiuwen Zheng
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P.R. China.
- College of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P.R. China
| |
Collapse
|
30
|
Dual key co-activated nanoplatform for switchable MRI monitoring accurate ferroptosis-based synergistic therapy. Chem 2022. [DOI: 10.1016/j.chempr.2022.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
31
|
He X, Chen S, Mao X. Utilization of metal or non-metal-based functional materials as efficient composites in cancer therapies. RSC Adv 2022; 12:6540-6551. [PMID: 35424648 PMCID: PMC8982229 DOI: 10.1039/d1ra08335j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/30/2022] [Indexed: 12/03/2022] Open
Abstract
There has been great progress in cancer treatment through traditional approaches, even though some of them are still trapped in relative complications such as certain side effects and prospective chances of full recovery. As a conventional method, the immunotherapy approach is regarded as an effective approach to cure cancer. It is mainly promoted by immune checkpoint blocking and adoptive cell therapy, which can utilize the human immune system to attack tumor cells and make them necrose completely or stop proliferating cancer cells. Currently however, immunotherapy shows limited success due to the limitation of real applicable cases of targeted tumor environments and immune systems. Considering the urgent need to construct suitable strategies towards cancer therapy, metallic materials can be used as delivery systems for immunotherapeutic agents in the human body. Metallic materials exhibit a high degree of specificity, effectiveness, diagnostic ability, imaging ability and therapeutic effects with different biomolecules or polymers, which is an effective option for cancer treatment. In addition, these modified metallic materials contain immune-modulators, which can activate immune cells to regulate tumor microenvironments and enhance anti-cancer immunity. Additionally, they can be used as adjuvants with immunomodulatory activities, or as carriers for molecular transport to specific targets, which results in the loading of specific ligands to facilitate specific uptake. Here, we provide an overview of the different types of metallic materials used as efficient composites in cancer immunotherapy. We elaborate on the advancements using metallic materials with functional agents as effective composites in synergistic cancer treatment. Some nonmetallic functional composites also appear as a common phenomenon. Ascribed to the design of the composites themselves, the materials' surface structural characteristics are introduced as the drug-loading substrate. The physical and chemical properties of the functional materials emphasize that further research is required to fully characterize their mechanism, showing appropriate relevance for material toxicology and biomedical applications.
Collapse
Affiliation(s)
- Xiaoxiao He
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Shiyue Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Xiang Mao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| |
Collapse
|
32
|
Chang ZX, Li CH, Chang YC, Huang CYF, Chan MH, Hsiao M. Novel monodisperse FePt nanocomposites for T2-weighted magnetic resonance imaging: biomedical theranostics applications. NANOSCALE ADVANCES 2022; 4:377-386. [PMID: 36132698 PMCID: PMC9419603 DOI: 10.1039/d1na00613d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/19/2021] [Indexed: 06/07/2023]
Abstract
Given the high incidence and mortality of cancer, current research is focused on designing efficient diagnostic methods. At present, clinical diagnoses are made based on X-ray, computed tomography, magnetic resonance imaging (MRI), ultrasound, and fiber optic endoscopy. MRI is a powerful diagnostic tool because it is non-invasive, has a high spatial resolution, uses non-ionizing radiation, and has good soft-tissue contrast. However, the long relaxation time of water protons may result in the inability to distinguish different tissues. To overcome this drawback of MRI, magnetic resonance contrast agents can enhance the contrast, improve the sensitivity of MRI-based diagnoses, increase the success rate of surgery, and reduce tumor recurrence. This review focuses on using iron-platinum (FePt) nanoparticles (NPs) in T2-weighted MRI to detect tumor location based on dark-field changes. In addition, existing methods for optimizing and improving FePt NPs are reviewed, and the MRI applications of FePt NPs are discussed. FePT NPs are expected to strengthen MRI resolution, thereby helping to inhibit tumor development.
Collapse
Affiliation(s)
- Zhi-Xuan Chang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 112 Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University Taipei 112 Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 112 Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University Taipei 112 Taiwan
| | | | - Michael Hsiao
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
- Department of Biochemistry College of Medicine, Kaohsiung Medical University Kaohsiung 807 Taiwan
| |
Collapse
|
33
|
Nie Q, Hu Y, Yu X, Li X, Fang X. Induction and application of ferroptosis in cancer therapy. Cancer Cell Int 2022; 22:12. [PMID: 34996454 PMCID: PMC8742449 DOI: 10.1186/s12935-021-02366-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
At present, more than one cell death pathways have been found, one of which is ferroptosis. Ferroptosis was discovered in 2012 and described as an iron-dependent and lipid peroxidation-driven regulated cell death pathway. In the past few years, ferroptosis has been shown to induce tumor cell death, providing new ideas for tumor treatment. In this article, we summarize the latest advances in ferroptosis-induced tumor therapy at the intersection of tumor biology, molecular biology, redox biology, and materials chemistry. First, we state the characteristics of ferroptosis in cells, then introduce the key molecular mechanism of ferroptosis, and describes the relationship between ferroptosis and oxidative stress signaling pathways. Finally, we focused on several types of ferroptosis inducers discovered by scholars, and the application of ferroptosis in systemic chemotherapy, radiotherapy, immunotherapy and nanomedicine, in the hope that ferroptosis can exert its potential in the treatment of tumors.
Collapse
Affiliation(s)
- Qing Nie
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yue Hu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiao Yu
- First Affiliated Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiao Li
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xuedong Fang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
34
|
Hu Z, Wei Q, Zhang H, Tang W, Kou Y, Sun Y, Dai Z, Zheng X. Advances in FePt-involved nano-system design and application for bioeffect and biosafety. J Mater Chem B 2021; 10:339-357. [PMID: 34951441 DOI: 10.1039/d1tb02221k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The rapid development and wide application of nanomaterial-involved theranostic agents have drawn surging attention for improving the living standard of humankind and healthcare conditions. In this review, recent developments in the design, synthesis, biocompatibility evaluation and potential nanomedicine applications of FePt-involved nano-systems are summarized, especially for cancer theranostic and biological molecule detection. The in vivo multi-model imaging capability is discussed in detail, including magnetic resonance imaging and computed tomography. Furthermore, we highlight the significant achievements of various FePt-involved nanotherapeutics for cancer treatment, such as drug delivery, chemodynamic therapy, photodynamic therapy, radiotherapy and immunotherapy. In addition, a series of FePt-involved nanocomposites are also applied for biological molecule detection, such as H2O2, glucose and naked-eye detection of cancer cells. Ultimately, we also summarize the challenges and prospects of FePt-involved nano-systems in nanocatalytic medicine. This review is expected to give a general pattern for the development of FePt-involved nano-systems in the field of nanocatalytic medicine and analytical determination.
Collapse
Affiliation(s)
- Zunfu Hu
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China. .,School of Materials Science and Engineering, Linyi University, Linyi 276000, P. R. China
| | - Qiulian Wei
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China. .,School of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao 266510, P. R. China
| | - Huimin Zhang
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China.
| | - Weina Tang
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China.
| | - Yunkai Kou
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China.
| | - Yunqiang Sun
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China.
| | - Zhichao Dai
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China.
| | - Xiuwen Zheng
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi, China.
| |
Collapse
|
35
|
Liu Y, Zhu S, Gu Z, Zhao Y. A bibliometric analysis: Research progress and prospects on transition metal dichalcogenides in the biomedical field. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.04.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
NIR-Laser Triggered Drug Release from Molybdenum Disulfide Nanosheets Modified with Thermosensitive Polymer for Prostate Cancer Treatment. J Inorg Organomet Polym Mater 2021. [DOI: 10.1007/s10904-021-02075-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
37
|
Research progress on the occurrence and therapeutic mechanism of ferroptosis in NSCLC. Naunyn Schmiedebergs Arch Pharmacol 2021; 395:1-12. [PMID: 34779876 DOI: 10.1007/s00210-021-02178-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/06/2021] [Indexed: 10/19/2022]
Abstract
Ferroptosis refers to a novel way of cell death, inconsistent with the conventional concept of apoptosis and necrosis. It shows a close association with iron metabolism and oxidative damage, as marked by the significant increase of reactive oxygen species, the decreases of mitochondrial volume, and the thickening of membrane density. Recent studies confirmed that ferroptosis is closely associated with the occurrence, development, and therapy of the tumors. As impacted by the high levels of reactive oxygen species and lipid peroxides in lung cancer tissues, it is suggested that ferroptosis is more likely to occur in lung cancer tissues, which may act as a novel approach for non-small cell lung cancer (NSCLC) therapy. In the present study, the research achievements in recent years on the regulating mechanism of ferroptosis and its effect on the occurrence and the therapy of lung cancer are reviewed.
Collapse
|
38
|
Tang Z, Xiao Y, Kong N, Liu C, Chen W, Huang X, Xu D, Ouyang J, Feng C, Wang C, Wang J, Zhang H, Tao W. Nano-bio interfaces effect of two-dimensional nanomaterials and their applications in cancer immunotherapy. Acta Pharm Sin B 2021; 11:3447-3464. [PMID: 34900529 PMCID: PMC8642437 DOI: 10.1016/j.apsb.2021.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
The field of two-dimensional (2D) nanomaterial-based cancer immunotherapy combines research from multiple subdisciplines of material science, nano-chemistry, in particular nano-biological interactions, immunology, and medicinal chemistry. Most importantly, the "biological identity" of nanomaterials governed by bio-molecular corona in terms of bimolecular types, relative abundance, and conformation at the nanomaterial surface is now believed to influence blood circulation time, bio-distribution, immune response, cellular uptake, and intracellular trafficking. A better understanding of nano-bio interactions can improve utilization of 2D nano-architectures for cancer immunotherapy and immunotheranostics, allowing them to be adapted or modified to treat other immune dysregulation syndromes including autoimmune diseases or inflammation, infection, tissue regeneration, and transplantation. The manuscript reviews the biological interactions and immunotherapeutic applications of 2D nanomaterials, including understanding their interactions with biological molecules of the immune system, summarizes and prospects the applications of 2D nanomaterials in cancer immunotherapy.
Collapse
Affiliation(s)
- Zhongmin Tang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yufen Xiao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chuang Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiangang Huang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daiyun Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Jiang Ouyang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chan Feng
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Cong Wang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China
| | - Junqing Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Han Zhang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
39
|
Ma B, Bianco A. Recent Advances in 2D Material-Mediated Immuno-Combined Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102557. [PMID: 34510729 DOI: 10.1002/smll.202102557] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/28/2021] [Indexed: 06/13/2023]
Abstract
In the last years, cancer immunotherapy has started to attract a lot of attention, becoming one of the alternatives in the clinical treatment of cancer. Indeed, one of the advantages of immunotherapy is that both primary and distant tumors can be efficiently eradicated through a triggered immune response. Due to their large specific surface area and unique physicochemical properties, 2D materials have become popular in cancer immunotherapy, especially as efficient drug carriers. They have been also exploited as photothermal platforms, chemodynamic agents, and photosensitizers to further enhance the efficacy of the therapy. In this review, the focus is on the recent development of 2D materials as new tools to combine immunotherapy with chemotherapy, photothermal therapy, photodynamic therapy, chemodynamic therapy, radiotherapy, and radiodynamic therapy. These innovative synergistic approaches intend to go beyond the classical strategies based on a simple delivery function of immune modulators by nanomaterials. Furthermore, the effects of the 2D materials themselves and their surface properties (e.g., chemical modification and protein corona formation) on the induction of an immune response will be also discussed.
Collapse
Affiliation(s)
- Baojin Ma
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| |
Collapse
|
40
|
Wang J, Sui L, Huang J, Miao L, Nie Y, Wang K, Yang Z, Huang Q, Gong X, Nan Y, Ai K. MoS 2-based nanocomposites for cancer diagnosis and therapy. Bioact Mater 2021; 6:4209-4242. [PMID: 33997503 PMCID: PMC8102209 DOI: 10.1016/j.bioactmat.2021.04.021] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 12/24/2022] Open
Abstract
Molybdenum is a trace dietary element necessary for the survival of humans. Some molybdenum-bearing enzymes are involved in key metabolic activities in the human body (such as xanthine oxidase, aldehyde oxidase and sulfite oxidase). Many molybdenum-based compounds have been widely used in biomedical research. Especially, MoS2-nanomaterials have attracted more attention in cancer diagnosis and treatment recently because of their unique physical and chemical properties. MoS2 can adsorb various biomolecules and drug molecules via covalent or non-covalent interactions because it is easy to modify and possess a high specific surface area, improving its tumor targeting and colloidal stability, as well as accuracy and sensitivity for detecting specific biomarkers. At the same time, in the near-infrared (NIR) window, MoS2 has excellent optical absorption and prominent photothermal conversion efficiency, which can achieve NIR-based phototherapy and NIR-responsive controlled drug-release. Significantly, the modified MoS2-nanocomposite can specifically respond to the tumor microenvironment, leading to drug accumulation in the tumor site increased, reducing its side effects on non-cancerous tissues, and improved therapeutic effect. In this review, we introduced the latest developments of MoS2-nanocomposites in cancer diagnosis and therapy, mainly focusing on biosensors, bioimaging, chemotherapy, phototherapy, microwave hyperthermia, and combination therapy. Furthermore, we also discuss the current challenges and prospects of MoS2-nanocomposites in cancer treatment.
Collapse
Affiliation(s)
- Jianling Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Lihua Sui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jia Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Lu Miao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yubing Nie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Zhichun Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Qiong Huang
- Department of Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xue Gong
- Department of Radiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yayun Nan
- Geriatric Medical Center, Ningxia People's Hospital, Yinchuan, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
41
|
Luo L, Wang H, Tian W, Li X, Zhu Z, Huang R, Luo H. Targeting ferroptosis-based cancer therapy using nanomaterials: strategies and applications. Theranostics 2021; 11:9937-9952. [PMID: 34815796 PMCID: PMC8581438 DOI: 10.7150/thno.65480] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/10/2021] [Indexed: 12/14/2022] Open
Abstract
As an iron-dependent mode of programmed cell death induced by lipid peroxidation, ferroptosis plays an important role in cancer therapy. The metabolic reprogramming in tumor microenvironment allows the possibility of targeting ferroptosis in cancer treatment. Recent studies reveal that nanomaterials targeting ferroptosis have prospects for the development of new cancer treatments. However, the design ideas of nanomaterials targeting ferroptosis sometimes vary. Therefore, in addition to the need for a systematic summary of these ideas, new ideas and insights are needed to make possible the construction of nanomaterials for effectively targeting this cell death pathway. At the same time, further optimization of nanomaterials design is required to make them appropriate for clinical treatment. In this context, we summarize this cross-cutting research area covering from the known mechanism of ferroptosis to providing feasible ideas for nanomaterials design as well as their clinical application. We aim to provide new insights and enlightenment for the next step in developing new nanomaterials for cancer treatment.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, 524023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, 524023, China
| | - Han Wang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Wen Tian
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Xiaoling Li
- Experimental Animal Center, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Zheng Zhu
- Affiliations Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, 524023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
42
|
Wu Y, Chen F, Huang N, Li J, Wu C, Tan B, Liu Y, Li L, Yang C, Shao D, Liao J. Near-infrared light-responsive hybrid hydrogels for the synergistic chemo-photothermal therapy of oral cancer. NANOSCALE 2021; 13:17168-17182. [PMID: 34636386 DOI: 10.1039/d1nr04625j] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Light-stimulus-responsive therapies have been recognized as a promising strategy for the efficient and safe treatment of oral squamous cell carcinoma (OSCC). Hydrogels have emerged as a promising multifunctional platform combining localized drug delivery and sustained drug release with multimodal properties for combined OSCC therapy. However, inaccurate drug release and limited light-absorption efficiency have hindered their on-demand chemo-photothermal applications. To tackle these problems, an injectable and near-infrared (NIR) light-responsive hybrid system was developed by incorporating light-responsive mesoporous silica nanoparticles (MSNs) as doxorubicin (DOX) carriers into the IR820/methylcellulose hydrogel networks for chemophotothermal therapy. Under NIR radiation, the incorporated IR820, a new green cyanine dye, was excited to induce photothermal effects against tumor cells. Meanwhile, MSNs achieved self-degradation-controlled DOX release via the cleavage of diselenide bonds induced by reactive oxygen species. Through the combination of chemotherapy and phototherapy, a long-lasting synergistic anti-tumor effect was achieved in vitro and in vivo with less toxicity. These findings demonstrate the potential of light-responsive hydrogels as a multifunctional platform for accurate synergistic chemophotothermal treatment of OSCC.
Collapse
Affiliation(s)
- Yongzhi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Fangman Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China.
| | - Nengwen Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Jinjin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Bowen Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yunkun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Chao Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China.
| | - Dan Shao
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
43
|
Hou H, Wang Z, Ma Y, Yu K, Zhao J, Lin H, Qu F. NIR-driven intracellular photocatalytic oxygen-supply on metallic molybdenum carbide@N-carbon for hypoxic tumor therapy. J Colloid Interface Sci 2021; 607:1-15. [PMID: 34500412 DOI: 10.1016/j.jcis.2021.08.177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
The intracellular O2-supply not only can relieve tumor hypoxia but also enhance the effects of photodynamic therapy (PDT). In this work, metallic Mo2C@N-carbon@PEG nanoparticles were constructed to reveal the near infrared (NIR)-photocatalytic O2 generation and promote photodynamic therapy (PDT). Here, (NH4)6Mo7O24·4H2O nanorods and urea were adopted as resources that were calcined to obtain Mo2C@N-carbon nanoparticles (20 nm). All samples displayed high NIR absorption as well as photothermal conversion efficiency of up to 52.7 % (Mo2C@N-Carbon-3@PEG). The density functional theory calculations demonstrated the metallic characteristic of Mo2C and that the consecutive interband/intraband charge-transition was responsible for the high NIR harvest and redox ability of electron-hole pairs, making the NIR-photocatalytic O2 and reactive oxygen species (ROS) generation. In comparison with the pure Mo2C, the heterostructure displayed twice the performance due to the enhanced charge-segregation between Mo2C and N-carbon. Given the high X-ray absorption coefficient and photothermal ability, the nanocomposite could be used in novel computer tomography and photothermal imaging contrast. Furthermore, the novel biodegradation and metabolism behaviors of nanocomposites were investigated, which were reflected as elimination from the body (mouse) via feces and urine within 14 days. The as-synthesized Mo2C@N-Carbon@PEG nanocomposites integrated the dual-model imaging, intracellular O2-supply, and phototherapy into one nanoplatform, revealing its potential for anti-cancer therapy.
Collapse
Affiliation(s)
- Huaying Hou
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Zhongxu Wang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Yajie Ma
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Kai Yu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Jingxiang Zhao
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China.
| | - Huiming Lin
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China; Laboratory for Photon and Electronic Bandgap Materials, Ministry of Education, Harbin Normal University, Harbin 150025, China.
| | - Fengyu Qu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China.
| |
Collapse
|
44
|
Xu C, Chen H. A Ferroptosis-Related Gene Model Predicts Prognosis and Immune Microenvironment for Cutaneous Melanoma. Front Genet 2021; 12:697043. [PMID: 34447410 PMCID: PMC8384470 DOI: 10.3389/fgene.2021.697043] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Background Cutaneous melanoma is a common but aggressive tumor. Ferroptosis is a recently discovered cell death with important roles in tumor biology. Nevertheless, the prognostic power of ferroptosis-linked genes remained unclear in cutaneous melanoma. Methods Cutaneous melanoma patients of TCGA (The Cancer Genome Atlas) were taken as the training cohort while GSE65904 and GSE22153 as the validation cohorts. Multifactor Cox regression model was used to build a prognostic model, and the performance of the model was assessed. Functional enrichment and immune infiltration analysis were used to clarify the mechanisms. Results A five ferroptosis-linked gene predictive model was developed. ALOX5 and GCH1 were illustrated as independent predictive factors. Functional assessment showed enriched immune-linked cascades. Immune infiltrating analysis exhibited the distinct immune microenvironment. Conclusion Herein, a novel ferroptosis-related gene prognostic model was built in cutaneous melanoma. This model could be used for prognostic prediction, and maybe helpful for the targeted and immunotherapies.
Collapse
Affiliation(s)
- Congcong Xu
- Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Hao Chen
- Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
45
|
Wang Z, Chen L, Ma Y, Li X, Hu A, Wang H, Wang W, Li X, Tian B, Dong J. Peptide vaccine-conjugated mesoporous carriers synergize with immunogenic cell death and PD-L1 blockade for amplified immunotherapy of metastatic spinal. J Nanobiotechnology 2021; 19:243. [PMID: 34384429 PMCID: PMC8362242 DOI: 10.1186/s12951-021-00975-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/27/2021] [Indexed: 12/30/2022] Open
Abstract
The clinical treatment of metastatic spinal tumor remains a huge challenge owing to the intrinsic limitations of the existing methods. Programmed cell death protein 1 (PD1)/programmed cell death ligand 1 (PD-L1) pathway blockade has been explored as a promising immunotherapeutic strategy; however, their inhibition has a low response rate, leading to the minimal cytotoxic T cell infiltration. To ameliorate the immunosuppressive microenvironment of intractable tumor and further boost the efficacy of immunotherapy, we report an all-round mesoporous nanocarrier composed of an upconverting nanoparticle core and a large-pore mesoporous silica shell (UCMS) that is simultaneously loaded with photosensitizer molecules, the IDO-derived peptide vaccine AL-9, and PD-L1 inhibitor. The IDO-derived peptide can be recognized by the dendritic cells and presented to CD8+ cytotoxic T cells, thereby enhancing the immune response and promoting the killing of the IDO-expressed tumor cells. Meanwhile, the near-infrared (NIR) activated photodynamic therapy (PDT) could induce immunogenic cell death (ICD), which promotes the effector T-cell infiltration. By combining the PDT-elicited ICD, peptide vaccine and immune checkpoint blockade, the designed UCMS@Pep-aPDL1 successfully potentiated local and systemic antitumor immunity and reduced the progression of metastatic foci, demonstrating a synergistic strategy for cancer immunotherapy. ![]()
Collapse
Affiliation(s)
- Zhenqing Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Yiqun Ma
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xilei Li
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Annan Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Huiren Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wenxing Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Xiaomin Li
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, People's Republic of China.
| | - Bo Tian
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Department of Orthopaedic Surgery, Shanghai Baoshan District Wusong Center Hospital, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 200940, People's Republic of China.
| |
Collapse
|
46
|
Liu Y, Chen Q, Zhu Y, Wang T, Ye L, Han L, Yao Z, Yang Z. Non-coding RNAs in necroptosis, pyroptosis and ferroptosis in cancer metastasis. Cell Death Discov 2021; 7:210. [PMID: 34381023 PMCID: PMC8358062 DOI: 10.1038/s41420-021-00596-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Distant metastasis is the main cause of death for cancer patients. Recently, the newly discovered programmed cell death includes necroptosis, pyroptosis, and ferroptosis, which possesses an important role in the process of tumor metastasis. At the same time, it is widely reported that non-coding RNA precisely regulates programmed death and tumor metastasis. In the present review, we summarize the function and role of necroptosis, pyrolysis, and ferroptosis involving in cancer metastasis, as well as the regulatory factors, including non-coding RNAs, of necroptosis, pyroptosis, and ferroptosis in the process of tumor metastasis.
Collapse
Affiliation(s)
- Yan Liu
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Qiuyun Chen
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Yanan Zhu
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Tiying Wang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Lijuan Ye
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Lei Han
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Zhihong Yao
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Cancer Hospital of Yunnan Province), Kunming, Yunnan, China.
| |
Collapse
|
47
|
Zhang Y, He Z, Yang F, Ye C, Xu X, Wang S, Zhang L, Zou D. Novel PVA-Based Microspheres Co-Loaded with Photothermal Transforming Agent and Chemotherapeutic for Colorectal Cancer Treatment. Pharmaceutics 2021; 13:984. [PMID: 34209684 PMCID: PMC8309159 DOI: 10.3390/pharmaceutics13070984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/06/2021] [Accepted: 06/17/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND We previously designed an electrospinning chitosan (CS) nanofiber-based carrier, using polyvinyl alcohol (PVA) as an adjuvant to deliver doxorubicin (DOX) and MoS2 nanosheets for postoperative tumor re-occurrence inhibition. However, owing to that the nanofibrous mat is un-injectable, this composite nanofiber is far from being clinically applicable. MATERIALS AND METHODS Via modulating the electrospray parameters, polyvinyl alcohol (PVA) beads string doped with DOX and MoS2 (PVA/MoS2/DOX microspheres) were prepared, which were further crosslinked with glutaraldehyde to obtain the water-stability. RESULTS Under the 808-nm laser irradiation, MoS2 nanosheets rendered the prepared PVA/MoS2/DOX microspheres an excellent light-to-heat conversion performance with η of 23.2%. Besides, the heat generated by near-infrared laser irradiation can improve the effect of chemotherapy by promoting the release rate of DOX. HT29 cell and tumor-bearing nude mice were used to systematically study the combined tumor treatment efficiency of composite nanospheres. CONCLUSION PVA/MoS2/DOX nanospheres have excellent photothermal effect and chemotherapy effect, which can completely suppress the tumor recurrence. Therefore, the PVA/MoS2/DOX nanospheres are anticipated to find potential applications in the treatment of local colorectal cancer.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai 200025, China;
| | - Zirui He
- Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai 200025, China;
| | - Fan Yang
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai 200093, China; (F.Y.); (C.Y.); (X.X.); (S.W.)
| | - Changqing Ye
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai 200093, China; (F.Y.); (C.Y.); (X.X.); (S.W.)
| | - Xia Xu
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai 200093, China; (F.Y.); (C.Y.); (X.X.); (S.W.)
| | - Shige Wang
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai 200093, China; (F.Y.); (C.Y.); (X.X.); (S.W.)
| | - Ling Zhang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai 200025, China;
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai 200025, China;
| |
Collapse
|
48
|
Zhao J, Zhang L, Qi Y, Liao K, Wang Z, Wen M, Zhou D. NIR Laser Responsive Nanoparticles for Ovarian Cancer Targeted Combination Therapy with Dual-Modal Imaging Guidance. Int J Nanomedicine 2021; 16:4351-4369. [PMID: 34234430 PMCID: PMC8254569 DOI: 10.2147/ijn.s299376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/01/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose Multifunctional nanoparticles with targeted therapeutic function and diagnostic-imaging are of great interest in the domain of precision therapy. NIR laser responsive nanoparticles (PLGA-PEG-FA encapsulating Bi2S3, PFP, and Dox (designed as FBPD NPs)) are synthesized for ovarian cancer targeted combination therapy with CT/PA dual-modal imaging guidance (PA: photoacoustic; CT: X-ray computed tomography). Methods and Results The FBPD NPS prepared by the double emulsification method revealed excellent dispersity, great stability, outstanding optical properties. The temperature of FBPD NPs increased rapidly after laser irradiation, inducing liquid-to-gas conversion of perfluoropentane (PFP), and promoting the release of Dox up to 86.7%. These FBPD NPs demonstrated their outstanding imaging capability for both PA and CT imaging both in vitro and in vivo, providing the potential for therapeutic guidance and monitoring. Assisted by folic acid, these nanoparticles could highly enrich in ovarian tumor tissue and the accumulation peaked at 3 h after intravenous administration. The desirable photothermal-conversion efficiency of the nanoparticles combined with chemotherapy achieved highly efficient therapy, which was demonstrated both in vitro and in vivo. Conclusion We successfully constructed multifunctional theranostic FBPD NPs for highly efficient PTT/chemotherapy combined therapy with dual CT/PA imaging guidance/monitoring. The unique nanoparticles with multiple abilities pave an emerging way toward precise treatment of ovarian cancer.
Collapse
Affiliation(s)
- Jiawen Zhao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yingjie Qi
- Department of Intensive Care Unit (ICU), Dianjiang People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Kui Liao
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ming Wen
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Di Zhou
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
49
|
Pu XQ, Ju XJ, Zhang L, Cai QW, Liu YQ, Peng HY, Xie R, Wang W, Liu Z, Chu LY. Novel Multifunctional Stimuli-Responsive Nanoparticles for Synergetic Chemo-Photothermal Therapy of Tumors. ACS APPLIED MATERIALS & INTERFACES 2021; 13:28802-28817. [PMID: 34109788 DOI: 10.1021/acsami.1c05330] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
In this study, a novel class of multifunctional responsive nanoparticles is designed and fabricated as drug nanocarriers for synergetic chemo-photothermal therapy of tumors. The proposed nanoparticles are composed of a thermo-/pH-responsive poly(N-isopropylacrylamide-co-acrylic acid) (PNA) nanogel core, a polydopamine (PDA) layer for photothermal conversion, and an outer folic acid (FA) layer as a targeting agent for the folate receptors on tumor cells. The fabricated nanoparticles show good biocompatibility and outstanding photothermal conversion efficiency. The proposed nanoparticles loaded with doxorubicin (DOX) drug molecules are stable under physiological conditions with low leakage of drugs, while rapidly release drugs in environments with low pH conditions and at high temperature. The experimental results show that the drug release process is mainly governed by Fickian diffusion. In vitro cell experimental results demonstrate that the PNA-DOX@PDA-FA nanoparticles can be phagocytized by 4T1 tumor cells and release drugs in tumor cell acidic environments, and confirm that the combined chemo and photothermal therapeutic efficacy of PNA-DOX@PDA-FA nanoparticles is higher than the photothermal therapeutic efficacy or the chemotherapeutic efficacy alone. The proposed multifunctional responsive nanoparticles in this study provide a novel class of drug nanocarriers as a promising tool for synergetic chemo-photothermal therapy of tumors.
Collapse
Affiliation(s)
- Xing-Qun Pu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Xiao-Jie Ju
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Lei Zhang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Quan-Wei Cai
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yu-Qiong Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Han-Yu Peng
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Rui Xie
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Wei Wang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Zhuang Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Liang-Yin Chu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| |
Collapse
|
50
|
Zafar H, Raza F, Ma S, Wei Y, Zhang J, Shen Q. Recent progress on nanomedicine-induced ferroptosis for cancer therapy. Biomater Sci 2021; 9:5092-5115. [PMID: 34160488 DOI: 10.1039/d1bm00721a] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current treatment strategies for cancer therapy have posed many problems in achieving high efficacy. Therefore, an urgent step is needed to develop innovative therapies that can win beyond satisfactory results against tumor. Ferroptosis that is a kind of non-apoptotic based programmed cell death has played a crucial role in eradicating tumors by reactive oxygen species and iron-dependent pathways. Research shows a remarkable potential of ferroptosis in eliminating aggressive malignancies resistant to traditional therapies. The combination of nanomedicine and ferroptosis has revealed a close relationship for the treatment of various cancer types with high efficacy. This review introduces the basics of nanomedicine-based ferroptosis first to emphasize the feasibility and properties of ferroptosis in cancer therapy. Then, the current research on the applications of nanomedicine for the ferroptosis-based anticancer therapy is highlighted. Finally, conclusions and future research directions in perspective of various challenges in developing nanomedicine-based ferroptosis into clinical therapeutics are discussed.
Collapse
Affiliation(s)
- Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan, Road, Shanghai, 200240, China.
| | | | | | | | | | | |
Collapse
|