1
|
Orooji N, Babaei S, Fadaee M, Abbasi-Kenarsari H, Eslami M, Kazemi T, Yousefi B. Novel therapeutic approaches for non-small cell lung cancer: an updated view. J Drug Target 2025:1-16. [PMID: 40186594 DOI: 10.1080/1061186x.2025.2489986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-small cell lung cancer (NSCLC) continues to be one of the leading causes of cancer-related mortality globally. Most patients who undergo surgical procedures may encounter distant metastasis or local recurrence, necessitating supplementary treatments such as radiation therapy, chemotherapy, or targeted therapy as adjuvant alternatives. Recent advancements in molecular biology and immunotherapy have paved the way for innovative therapeutic approaches that target specific genetic mutations and promote the immune response against tumour cells. This review explores emerging therapies, including targeted therapies such as tyrosine kinase inhibitors (TKIs) for actionable mutations (e.g., EGFR, ALK, ROS1), as well as the role of immune checkpoint inhibitors (ICIs) that employ the body's immune system to combat cancer. Additionally, we discuss the potential of exosome therapies, as well as promising nanotherapeutic options for the treatment of NSCLC. This study attempts to provide a thorough overview of the changing landscape of NSCLC treatment and its implications for enhancing patient outcomes by presenting these innovative techniques.
Collapse
Affiliation(s)
- Niloufar Orooji
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Shabnam Babaei
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Abbasi-Kenarsari
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
2
|
Ow V, Lin Q, Wong JHM, Sim B, Tan YL, Leow Y, Goh R, Loh XJ. Understanding the interplay between pH and charges for theranostic nanomaterials. NANOSCALE 2025; 17:6960-6980. [PMID: 40008569 DOI: 10.1039/d4nr03706e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Nanotechnology has emerged as a highly promising platform for theranostics, offering dual capabilities in targeted imaging and therapy. Interactions between the nanomaterial and biological components determine the in vivo fate of these materials which makes the control of their surface properties of utmost importance. Nanoparticles with neutral or negative surface charge have a longer circulation time while positively charged nanoparticles have higher affinity to cells and better cellular uptake. This trade-off presents a key challenge in optimizing surface charge for theranostic applications. A sophisticated solution is an on-demand switch of surface charge, enabled by leveraging the distinct pH conditions at the target site. In this review, we explore the intricate relationship between pH and charge modulation, summarizing recent advances in pH-induced charge-switchable nanomaterials for theranostics over the past five years. Additionally, we discuss how these innovations enhance targeted drug delivery and imaging contrast and provide perspectives on future directions for this transformative field.
Collapse
Affiliation(s)
- Valerie Ow
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
- Department of Biomedical Engineering, National University of Singapore (NUS), Singapore
| | - Qianyu Lin
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Joey Hui Min Wong
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Belynn Sim
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore
| | - Yee Lin Tan
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Yihao Leow
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore
| | - Rubayn Goh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
3
|
Malla P, Wang YM, Su CH. New horizons for the therapeutic application of nanozymes in cancer treatment. J Nanobiotechnology 2025; 23:130. [PMID: 39979897 PMCID: PMC11844087 DOI: 10.1186/s12951-025-03185-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/01/2025] [Indexed: 02/22/2025] Open
Abstract
The advent of nanozymes has revolutionized approaches to cancer diagnosis and therapy, introducing innovative strategies that address the limitations of conventional treatments. Nanozyme nanostructures with enzyme-mimicking catalytic abilities exhibit exceptional stability, biocompatibility, and customizable functions, positioning them as promising tools for cancer theranostics. By emulating natural enzyme reactions, nanozymes can selectively target and eradicate cancer cells, minimizing harm to adjacent healthy tissues. Nanozymes can also be functionalized with specific targeting ligands, allowing for the precise delivery and regulated release of therapeutic agents, improving treatment effectiveness and reducing adverse effects. However, issues such as biocompatibility, selectivity, and regulatory compliance remain critical challenges for the clinical application of nanozymes. This review provides an overview of nanozymes, highlighting their unique properties, various classifications, catalytic activities, and diverse applications in cancer treatments. The strategic oncological deployment of nanozymes could profoundly impact future advancements in personalized medicine, highlighting recent progress and prospective directions in enzyme-mimetic approaches for cancer treatment. This review summarizes an overview of nanozymes, highlighting their unique properties, various classifications, catalytic activities, and diverse applications in cancer treatments.
Collapse
Affiliation(s)
- Pravanjan Malla
- Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yu-Ming Wang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.
| | - Chia-Hao Su
- Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
4
|
Cao Y, Zhao X, Miao Y, Wang X, Deng D. How the Versatile Self-Assembly in Drug Delivery System to Afford Multimodal Cancer Therapy? Adv Healthc Mater 2025; 14:e2403715. [PMID: 39587000 DOI: 10.1002/adhm.202403715] [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: 09/27/2024] [Revised: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The rapid development of self-assembly technology during the past few decades has effectively addressed plenty of the issues associated with carrier-based drug delivery systems, such as low loading efficiency, complex fabrication processes, and inherent toxicity of carriers. The integration of nanoscale delivery systems with self-assembly techniques has enabled efficient and targeted self-administration of drugs, enhanced bioavailability, prolonged circulation time, and controllable drug release. Concurrently, the limitations of single-mode cancer treatment, including low bioavailability, poor therapeutic outcomes, and significant side effects, have highlighted the urgent need for multimodal combined antitumor therapies. Set against the backdrop of multimodal cancer therapy, this review summarizes the research progress and applications of a large number of self-assembled drug delivery platforms, including natural small molecule self-assembled, carrier-free self-assembled, amphiphilic polymer-based self-assembled, peptide-based self-assembled, and metal-based self-assembled nano drug delivery systems. This review particularly analyzes the latest advances in the application of self-assembled nano drug delivery platforms in combined antitumor therapies mediated by chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, and immunotherapy, providing innovative research insights for further optimization and expansion of self-assembled nano drug delivery systems in the clinical translation and development of antitumor combined therapy.
Collapse
Affiliation(s)
- Yuqi Cao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaomin Zhao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
5
|
Hou DY, You Q, Zhang P, Li XP, Wu JC, Wang Y, You HH, Lv MY, Wu G, Liu X, Guo P, Cheng DB, Chen X, Xu W. Cascade-Activatable Nanoprodrug System Augments Sonochemotherapy of Bladder Cancer. ACS NANO 2024; 18:35507-35519. [PMID: 39686741 DOI: 10.1021/acsnano.4c12967] [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: 12/18/2024]
Abstract
Sonochemotherapy (SCT) has emerged as a powerful modality for cancer treatment by triggering excessive production of reactive oxygen species (ROS) and controlled release of chemotherapeutic agents under ultrasound. However, achieving spatiotemporally controlled release of chemotherapeutic agents during ROS generation is still an enormous challenge. In this work, we developed a cascade-activated nanoprodrug (CAN) system that utilizes a reversible covalent Schiff base mixed with a hypoxia-activatable camptothecin (CPT) prodrug. Briefly, the designed fluorinated CAN system is self-assembled into nanoparticles under aqueous conditions, which could penetrate deep tumors to offer sufficient oxygen for ultrasound-triggered ROS production. Consequently, the nanoparticles substantially exacerbated the hypoxia of the tumor microenvironment (TME) by elevating oxygen consumption. The aggravated hypoxia in turn served as a positive amplifier to boost the tumor-specific CPT release of Azo-CPT prodrug, which made up for the insufficient treatment efficacy of sonodynamic therapy (SDT). On this basis, we observed a substantial reduction, approximately 3.5-fold, in the half-maximal inhibitory concentration (IC50) of the CAN system compared to that of free CPT in bladder cancer cell lines (T24). Furthermore, the CAN system demonstrated potent antitumor efficacy with reduced side effects, resulting in regression and eradication of T24 tumors in various mouse models. In summary, the CAN system can be easily extended by incorporating different chemotherapeutic agents, showing great potential to revolutionize the clinical management paradigm of bladder cancer.
Collapse
Affiliation(s)
- Da-Yong Hou
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Peng Zhang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xiang-Peng Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Jiong-Cheng Wu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Yueze Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Hui-Hui You
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Mei-Yu Lv
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiao Liu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Pengyu Guo
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| |
Collapse
|
6
|
Zhao X, Qi X, Liu D, Che X, Wu G. A Novel Approach for Bladder Cancer Treatment: Nanoparticles as a Drug Delivery System. Int J Nanomedicine 2024; 19:13461-13483. [PMID: 39713223 PMCID: PMC11662911 DOI: 10.2147/ijn.s498729] [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: 10/01/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Bladder cancer represents one of the most prevalent malignant neoplasms of the urinary tract. In the Asian context, it represents the eighth most common cancer in males. In 2022, there were approximately 613,791 individuals diagnosed with bladder cancer worldwide. Despite the availability of efficacious treatments for the two principal forms of bladder cancer, namely non-invasive and invasive bladder cancer, the high incidence of recurrence following treatment and the suboptimal outcomes observed in patients with high-grade and advanced disease represent significant concerns in the management of bladder cancer at this juncture. Nanoparticles have gained attention for their excellent properties, including stable physical properties, a porous structure that can be loaded with a variety of substances, and so on. The in-depth research on nanoparticles has led to their emergence as a new class of nanoparticles for combination therapy, due to their advantageous properties. These include the extension of the drug release window, the enhancement of drug bioavailability, the improvement of drug targeting ability, the reduction of local and systemic toxicity, and the simultaneous delivery of multiple drugs for combination therapy. As a result, nanoparticles have become a novel agent of the drug delivery system. The advent of nanoparticles has provided a new impetus for the development of non-surgical treatments for bladder cancer, including chemotherapy, immunotherapy, gene therapy and phototherapy. The unique properties of nanoparticles have facilitated the combination of diverse non-surgical therapeutic modalities, enhancing their overall efficacy. This review examines the recent advancements in the use of nanoparticles in non-surgical bladder cancer treatments, encompassing aspects such as delivery, therapeutic efficacy, and the associated toxicity of nanoparticles, as well as the challenges encountered in clinical applications.
Collapse
Affiliation(s)
- Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| |
Collapse
|
7
|
Gong Z, Mao Y, Liu Y, Hu X, Zhang Y, Zhu L, Guo S, Ding Z, Zhang L. Sono-promoted piezocatalysis and low-dose drug penetration for personalized therapy via tumor organoids. J Colloid Interface Sci 2024; 675:192-206. [PMID: 38968636 DOI: 10.1016/j.jcis.2024.07.010] [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: 04/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Chemotherapy is a widely used cancer treatment, however, it can have notable side effects owing to the high-doses of drugs administered. Sonodynamic therapy (SDT) induced by sonosensitizers has emerged as a promising approach to treat cancer, however, there is limited research evaluating its therapeutic effects on human tumors. In this study, we introduced a dual therapy that combines low-dose chemotherapeutic drugs with enhanced sonodynamic therapy, utilizing barium titanate (BaTiO3, BTO) nanoparticles (NPs) as sonosensitizers to treat tumor organoids. We demonstrated that ultrasound could improve the cellular uptake of chemotherapy drugs, while the chemotherapeutic effect of the drugs made it easier for BTO NPs to enter tumor cells, and the dual therapy synergistically inhibited tumor cell viability. Moreover, different patient-derived tumor organoids exhibited different sensitivities to this therapy, highlighting the potential to evaluate individual responses to combination therapies prior to clinical intervention. Furthermore, this dual therapy exhibited therapeutic effects equivalent to those of high-dose chemotherapy drugs on drug-resistant tumor organoids and showed the potential to enhance the efficacy of killing drug-resistant tumors. In addition, the biosafety of the BTO NPs was successfully verified in live mice via oral administration. This evidence confirms the reliable and safe nature of the dual therapy approach, making it a feasible option for precise and personalized therapy in clinical applications.
Collapse
Affiliation(s)
- Zhiyi Gong
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Jiufengshan Laboratory, Wuhan 430206, China; Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Microelectronics, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Yiqian Mao
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Microelectronics, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Yichao Liu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiao Hu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yusen Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lili Zhu
- Hubei Jiufengshan Laboratory, Wuhan 430206, China
| | - Shishang Guo
- Hubei Yangtze Memory Laboratories, Wuhan 430205, China; Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Microelectronics, School of Physics and Technology, Wuhan University, Wuhan 430072, China.
| | - Zhao Ding
- Department of Colorectal and Anal Surgery, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Lingling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
8
|
Ji L, Huang J, Yu L, Jin H, Hu X, Sun Y, Yin F, Cai Y. Recent advances in nanoagents delivery system-based phototherapy for osteosarcoma treatment. Int J Pharm 2024; 665:124633. [PMID: 39187032 DOI: 10.1016/j.ijpharm.2024.124633] [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: 04/02/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
Osteosarcoma (OS) is a prevalent and highly malignant bone tumor, characterized by its aggressive nature, invasiveness, and rapid progression, contributing to a high mortality rate, particularly among adolescents. Traditional treatment modalities, including surgical resection, radiotherapy, and chemotherapy, face significant challenges, especially in addressing chemotherapy resistance and managing postoperative recurrence and metastasis. Phototherapy (PT), encompassing photodynamic therapy (PDT) and photothermal therapy (PTT), offers unique advantages such as low toxicity, minimal drug resistance, selective destruction, and temporal control, making it a promising approach for the clinical treatment of various malignant tumors. Constructing multifunctional delivery systems presents an opportunity to effectively combine tumor PDT, PTT, and chemotherapy, creating a synergistic anti-tumor effect. This review aims to consolidate the progress in the application of novel delivery system-mediated phototherapy in osteosarcoma. By summarizing advancements in this field, the objective is to propose a rational combination therapy involving targeted delivery systems and phototherapy for tumors, thereby expanding treatment options and enhancing the prognosis for osteosarcoma patients. In conclusion, the integration of innovative delivery systems with phototherapy represents a promising avenue in osteosarcoma treatment, offering a comprehensive approach to overcome challenges associated with conventional treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Lichen Ji
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jiaqing Huang
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Department of Hematology, Hangzhou First People's Hospital, Hangzhou 310003, China
| | - Liting Yu
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huihui Jin
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Xuanhan Hu
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yuan Sun
- College of Chemistry Engineering, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Feng Yin
- Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Yu Cai
- Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
9
|
Guo J, Zhao W, Xiao X, Liu S, Liu L, Zhang L, Li L, Li Z, Li Z, Xu M, Peng Q, Wang J, Wei Y, Jiang N. Reprogramming exosomes for immunity-remodeled photodynamic therapy against non-small cell lung cancer. Bioact Mater 2024; 39:206-223. [PMID: 38827172 PMCID: PMC11141154 DOI: 10.1016/j.bioactmat.2024.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/11/2024] [Accepted: 05/16/2024] [Indexed: 06/04/2024] Open
Abstract
Traditional treatments against advanced non-small cell lung cancer (NSCLC) with high morbidity and mortality continue to be dissatisfactory. Given this situation, there is an urgent requirement for alternative modalities that provide lower invasiveness, superior clinical effectiveness, and minimal adverse effects. The combination of photodynamic therapy (PDT) and immunotherapy gradually become a promising approach for high-grade malignant NSCLC. Nevertheless, owing to the absence of precise drug delivery techniques as well as the hypoxic and immunosuppressive characteristics of the tumor microenvironment (TME), the efficacy of this combination therapy approach is less than ideal. In this study, we construct a novel nanoplatform that indocyanine green (ICG), a photosensitizer, loads into hollow manganese dioxide (MnO2) nanospheres (NPs) (ICG@MnO2), and then encapsulated in PD-L1 monoclonal antibodies (anti-PD-L1) reprogrammed exosomes (named ICG@MnO2@Exo-anti-PD-L1), to effectively modulate the TME to oppose NSCLC by the synergy of PDT and immunotherapy modalities. The ICG@MnO2@Exo-anti-PD-L1 NPs are precisely delivered to the tumor sites by targeting specially PD-L1 highly expressed cancer cells to controllably release anti-PD-L1 in the acidic TME, thereby activating T cell response. Subsequently, upon endocytic uptake by cancer cells, MnO2 catalyzes the conversion of H2O2 to O2, thereby alleviating tumor hypoxia. Meanwhile, ICG further utilizes O2 to produce singlet oxygen (1O2) to kill tumor cells under 808 nm near-infrared (NIR) irradiation. Furthermore, a high level of intratumoral H2O2 reduces MnO2 to Mn2+, which remodels the immune microenvironment by polarizing macrophages from M2 to M1, further driving T cells. Taken together, the current study suggests that the ICG@MnO2@Exo-anti-PD-L1 NPs could act as a novel drug delivery platform for achieving multimodal therapy in treating NSCLC.
Collapse
Affiliation(s)
- Jiao Guo
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Zhao
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Xiao
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Shanshan Liu
- Department of Plastic and Maxillofacial Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liang Liu
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - La Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lu Li
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Zhenghang Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhi Li
- Traditional Chinese Medicine Hospital of Bijie City, Guizhou province, 551700, China
| | - Mengxia Xu
- Traditional Chinese Medicine Hospital of Bijie City, Guizhou province, 551700, China
| | - Qiling Peng
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
- Bijie Municipal Health Bureau, Guizhou province, 551700, China
- Health Management Center, the Affiliated Hospital of Guizhou Medical University
| | - Jianwei Wang
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ning Jiang
- Department of Pathology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
10
|
Fattahi MR, Dehghani M, Paknahad S, Rahiminia S, Zareie D, Hoseini B, Oroomi TR, Motedayyen H, Arefnezhad R. Clinical insights into nanomedicine and biosafety: advanced therapeutic approaches for common urological cancers. Front Oncol 2024; 14:1438297. [PMID: 39193389 PMCID: PMC11347329 DOI: 10.3389/fonc.2024.1438297] [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: 05/25/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Urological cancers including those of the prostate, bladder, and kidney, are prevalent and often lethal malignancies besides other less common ones like testicular and penile cancers. Current treatments have major limitations like side effects, recurrence, resistance, high costs, and poor quality of life. Nanotechnology offers promising solutions through enhanced diagnostic accuracy, targeted drug delivery, controlled release, and multimodal imaging. This review reflects clinical challenges and nanomedical advances across major urological cancers. In prostate cancer, nanoparticles improve delineation and radiosensitization in radiation therapy, enable fluorescent guidance in surgery, and enhance chemotherapy penetration in metastatic disease. Nanoparticles also overcome bladder permeability barriers to increase the residence time of intravesical therapy and chemotherapy agents. In renal cancer, nanocarriers potentiate tyrosine kinase inhibitors and immunotherapy while gene vectors and zinc oxide nanoparticles demonstrate antiproliferative effects. Across modalities, urological applications of nanomedicine include polymeric, liposomal, and metal nanoparticles for targeted therapy, prodrug delivery, photodynamic therapy, and thermal ablation. Biosafety assessments reveal favorable profiles but clinical translation remains limited, necessitating further trials. In conclusion, nanotechnology holds significant potential for earlier detection, precise intervention, and tailored treatment of urological malignancies, warranting expanded research to transform patient outcomes.
Collapse
Affiliation(s)
- Mohammad Reza Fattahi
- Student Research Committee, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Shafa Rahiminia
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Deniz Zareie
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Behzad Hoseini
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Reza Arefnezhad
- Coenzyme R Research Institute, Tehran, Iran
- Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Jin N, Wang Z, Yin C, Bu W, Jin N, Ou L, Xie W, He J, Lai X, Shao L. Novel Carbon Quantum Dots Precisely Trigger Ferroptosis in Cancer Cells through Antioxidant Inhibition Synergistic Nanocatalytic Activity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37456-37467. [PMID: 39007694 DOI: 10.1021/acsami.4c04307] [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: 07/16/2024]
Abstract
High levels of glutathione (GSH) are an important characteristic of malignant tumors and a significant cause of ineffective treatment and multidrug resistance. Although reactive oxygen species (ROS) therapy has been shown to induce tumor cell death, the strong clearance effect of GSH on ROS significantly reduces its therapeutic efficacy. Therefore, there is a need to develop new strategies for targeting GSH. In this study, novel carbon quantum dots derived from gentamycin (GM-CQDs) were designed and synthesized. On the basis of the results obtained, GM-CQDs contain sp2 and sp3 carbon atoms as well as nitrogen oxygen groups, which decrease the intracellular levels of GSH by downregulating SLC7A11, thereby disrupting redox balance, mediating lipid peroxidation, and inducing ferroptosis. Transcriptome analysis demonstrated that GM-CQDs downregulated the expression of molecules related to GSH metabolism while significantly increasing the expression of molecules related to ferroptosis. The in vivo results showed that the GM-CQDs exhibited excellent antitumor activity and immune activation ability. Furthermore, because of their ideal biological safety, GM-CQDs are highly promising for application as drugs targeting GSH in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Nianqiang Jin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, People's Republic of China
- School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning 110002, People's Republic of China
| | - Zilin Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Chengcheng Yin
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning 110002, People's Republic of China
- School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130000, People's Republic of China
| | - Wenhuan Bu
- School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130000, People's Republic of China
| | - Nuo Jin
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Lingling Ou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, People's Republic of China
| | - Wenqiang Xie
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, People's Republic of China
| | - Jiankang He
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, People's Republic of China
| | - Xuan Lai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, People's Republic of China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, People's Republic of China
| |
Collapse
|
12
|
Yao L, Zhu X, Shan Y, Zhang L, Yao J, Xiong H. Recent Progress in Anti-Tumor Nanodrugs Based on Tumor Microenvironment Redox Regulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310018. [PMID: 38269480 DOI: 10.1002/smll.202310018] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/30/2023] [Indexed: 01/26/2024]
Abstract
The growth state of tumor cells is strictly affected by the specific abnormal redox status of the tumor microenvironment (TME). Moreover, redox reactions at the biological level are also central and fundamental to essential energy metabolism reactions in tumors. Accordingly, anti-tumor nanodrugs targeting the disruption of this abnormal redox homeostasis have become one of the hot spots in the field of nanodrugs research due to the effectiveness of TME modulation and anti-tumor efficiency mediated by redox interference. This review discusses the latest research results of nanodrugs in anti-tumor therapy, which regulate the levels of oxidants or reductants in TME through a variety of therapeutic strategies, ultimately breaking the original "stable" redox state of the TME and promoting tumor cell death. With the gradual deepening of study on the redox state of TME and the vigorous development of nanomaterials, it is expected that more anti-tumor nano drugs based on tumor redox microenvironment regulation will be designed and even applied clinically.
Collapse
Affiliation(s)
- Lan Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Xiang Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Yunyi Shan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Liang Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Hui Xiong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| |
Collapse
|
13
|
Ren X, Yang Y, Kong X, Liu Z. Integrin α vβ 3-targeted self-assembled polypeptide nanomicelles for efficacious sonodynamic therapy against breast cancer. NANOSCALE 2024; 16:9953-9965. [PMID: 38693876 DOI: 10.1039/d4nr00794h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Sonodynamic therapy (SDT) is an advanced non-invasive cancer treatment strategy with moderate tissue penetration, less invasiveness and a reliable curative effect. However, due to the low stability, potential bio-toxicity and lack of tumor targeting capability of most sonosensitizers, the vast clinical application of SDT has been challenging and limited. Therefore, it is desirable to develop a novel approach to implement sonosensitizers to SDT for cancer treatments. In this study, an amphiphilic polypeptide was designed to effectively encapsulate rose bengal (RB) as a model sonosensitizer to form peptido-nanomicelles (REPNs). The as-fabricated REPNs demonstrated satisfactory tumor targeting and fluorescence performances, which made them superb imaging tracers in vivo. In the meantime, they generated considerable amounts of reactive oxygen species (ROS) to promote tumor cell apoptosis under ultrasound irradiation and showed excellent anti-tumor performance without obvious side effects. These engineered nanomicelles in combination with medical ultrasound may be used to achieve integrin αvβ3-targeted sonodynamic therapy against breast cancer, and it is also a promising non-invasive cancer treatment strategy for clinical translations.
Collapse
Affiliation(s)
- Xueli Ren
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| | - Yanxi Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| | - Xinru Kong
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| | - Zhe Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| |
Collapse
|
14
|
Geng P, Li Y, Macharia DK, Ren X, Meng R, Wang W, Lan H, Xiao S. One Stone, Three Birds: Design and Synthesis of "All-in-One" Nanoscale Mn-Porphyrin Coordination Polymers for Magnetic Resonance Imaging-Guided Synergistic Photodynamic-Sonodynamic Therapy. J Colloid Interface Sci 2024; 660:1021-1029. [PMID: 38295540 DOI: 10.1016/j.jcis.2024.01.157] [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: 10/25/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/02/2024]
Abstract
Multifunctional nanomaterials with potential applications in both bioimaging and photodynamic-sonodynamic therapy have great advantages in cancer theranostic, but the design and preparation of "all-in-one" type of multifunctional nanomaterials with single component remains challenging. Herein the "all-in-one" type of Mn-PpIX (Protoporphyrin IX) coordination polymers (MnPPs) was reported as efficient nano-photo/sonosensitizers. The MnPPs had an average size of ∼ 110 nm. Upon light/US (ultrasound) irradiation for 5 min, 61.8 % (light) and 32.4 % (US) of DPBF (1.3-diphenyl isobenzofuran) was found to be oxidized by MnPPs, which showed effective ROS (reactive oxygen species) generation for photodynamic/sonodynamic therapy (PDT/SDT). In addition, MnPPs revealed excellent biosafety and could be engulfed by cells to produce intracellular ROS under light/US excitation for efficient killing tumor cells. When MnPPs was injected into mice, the tumor could be monitored via MRI (magnetic resonance imaging). In addition, tumor growth could be significantly inhibited by the synergistic PDT-SDT. Therefore, the present study not only represents MnPPs as an "all-in-one" type of multifunctional nanomaterials for MRI-guided PDT-SDT therapy, but also provides some insights for designing other PpIX-related molecules with the similar structure for bioapplication.
Collapse
Affiliation(s)
- Peng Geng
- College of Materials and Chemical Engineering, China Three Gorges University, Yichang 443002, China
| | - Yan Li
- College of Materials and Chemical Engineering, China Three Gorges University, Yichang 443002, China
| | - Daniel K Macharia
- College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Xiaoling Ren
- College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Ruru Meng
- College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Wei Wang
- College of Materials and Chemical Engineering, China Three Gorges University, Yichang 443002, China
| | - Haichuang Lan
- College of Materials and Chemical Engineering, China Three Gorges University, Yichang 443002, China.
| | - Shuzhang Xiao
- College of Materials and Chemical Engineering, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
15
|
Huang Y, Lv Y, Yang B, Zhang S, Bixia liu, Zhang C, Hu W, Jiang L, Chen C, Ji D, Xiong C, Liang Y, Liu M, Ying X, Ji W. Enhancing m 6A modification of lncRNA through METTL3 and RBM15 to promote malignant progression in bladder cancer. Heliyon 2024; 10:e28165. [PMID: 38560117 PMCID: PMC10979072 DOI: 10.1016/j.heliyon.2024.e28165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Objective Bladder cancer is one of the most prominent malignancies affecting the urinary tract, characterized by a poor prognosis. Our previous research has underscored the pivotal role of m6A methylation in the progression of bladder cancer. Nevertheless, the precise relationship between N6-methyladenosine (m6A) regulation of long non-coding RNA (lncRNA) and bladder cancer remains elusive. Methods This study harnessed sequencing data and clinical records from 408 bladder cancer patients in the TCGA database. Employing R software, we conducted bioinformatics analysis to establish an m6A-lncRNA co-expression network. Analyzing the differences between high and low-risk groups, particularly at the immunological level, and subsequently investigating the primary regulatory factors of these lncRNA, validating the findings through experiments, and exploring their specific cellular functions. Results We identified 50 m6A-related lncRNA with prognostic significance through univariate Cox regression analysis. In parallel, we employed a LASSO-Cox regression model to pinpoint 11 lncRNA and calculate risk scores for bladder cancer patients. Based on the median risk score, patients were categorized into low-risk and high-risk groups. The high-risk cohort exhibited notably lower survival rates than their low-risk counterparts. Further analysis pointed to RBM15 and METTL3 as potential master regulators of these m6A-lncRNA. Experimental findings also shed light on the upregulated expression of METTlL3 and RBM15 in bladder cancer, where they contributed to the malignant progression of tumors. The experimental findings demonstrated a significant upregulation of METTL3 and RBM15 in bladder cancer specimens, implicating their contributory role in the oncogenic progression. Knockdown of METTL3 and RBM15 resulted in a marked attenuation of tumor cell proliferation, invasion, and migration, which was concomitant with a downregulation in the cellular m6A methylation status. Moreover, these results revealed that RBM15 and METTL3 function in a synergistic capacity, positing their involvement in cancer promotion via the upregulation of m6A modifications in long non-coding RNAs. Additionally, this study successfully developed an N-methyl-N-nitrosourea (MNU)-induced rat model of in situ bladder carcinoma, confirming the elevated expression of RBM15 and METTL3, which paralleled the overexpression of m6A-related- lncRNAs observed in bladder cancer cell lines. This congruence underscores the potential utility of these molecular markers in in vivo models that mirror human malignancies. Conclusion This study not only offers novel molecular targets,but also enriches the research on m6A modification in bladder cancer, thereby facilitating its clinical translation.
Collapse
Affiliation(s)
- Yapeng Huang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yifan Lv
- Guangdong Provincial Key Laboratory of Urology, Guangzhou, 510230, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Baotong Yang
- Guangdong Provincial Key Laboratory of Urology, Guangzhou, 510230, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shike Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bixia liu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chengcheng Zhang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenyu Hu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | | | - Cong Chen
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ding Ji
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chang Xiong
- Guangdong Provincial People's Hospital, China
| | - Yaoming Liang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mingrui Liu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoling Ying
- Guangdong Provincial Key Laboratory of Urology, Guangzhou, 510230, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510220, China
| | - Weidong Ji
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Nasrollahian S, Moradi F, Hadi N, Ranjbar S, Ranjbar R. An update on alternative therapy for Escherichia coli causing urinary tract infections; a narrative review. Photodiagnosis Photodyn Ther 2024; 46:104075. [PMID: 38574879 DOI: 10.1016/j.pdpdt.2024.104075] [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: 10/27/2023] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Urinary tract infections (UTIs) are the most common type of nosocomial infection and severe health issues because of the difficulties and frequent recurrence. Today, alternative methods such as sonodynamic therapy (SDT), photodynamic therapy (PDT) and herbal materials use for treating infections like UTI in many countries. METHOD We conducted searches of the biomedical databases (Google Scholar, Scopus, PubMed, and Web of sciences) to identify related studies from 2008 to 2023. RESULT SDT aims to use ultrasound to activate a sonosensitizer, which causes a biological effect by raising reactive oxygen species (ROS). When bacteria are exposed to ROS, several important effects occur: oxidative damage, DNA damage, protein dysfunction etc. SDT with herbal medicine significantly reduced the number of colony-forming units and bactericidal activity for Klebsiella pneumonia and E. coli. PDT is a promising treatment for cancer and microbial infections, combining a photosensitiser, light and tissue molecular oxygen. It involves a photosensitizer, light source, and oxygen, with variations affecting microbial binding and bactericidal activity. Factors affecting antibacterial properties include plant type, growing conditions, harvesting, and processing. This review highlights the recent advancements in sonodynamic, photodynamic, herbal, and bio-material-based approaches in the treatment of E. coli infections. CONCLUSIONS These alternative therapies offer exciting prospects for addressing UTIs, especially in cases where traditional antibiotic treatments may be less effective. Further research and clinical studies are warranted to fully explore the potential of these innovative treatment modalities in combating UTIs and improving patient outcomes.
Collapse
Affiliation(s)
- Sina Nasrollahian
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Moradi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nahal Hadi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Ranjbar
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Zhou Z, Wang H, Li J, Jiang X, Li Z, Shen J. Recent progress, perspectives, and issues of engineered PD-L1 regulation nano-system to better cure tumor: A review. Int J Biol Macromol 2024; 254:127911. [PMID: 37939766 DOI: 10.1016/j.ijbiomac.2023.127911] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Currently, immune checkpoint blockade (ICB) therapies that target the programmed cell death ligand-1 (PD-L1) have been used as revolutionary cancer treatments in the clinic. Apart from restoring the antitumor response of cytotoxic T cells by blocking the interaction between PD-L1 on tumor cells and programmed cell death-1 (PD-1) on T cells, PD-L1 proteins were also newly revealed to possess the capacity to accelerate DNA damage repair (DDR) and enhance tumor growth through multiple mechanisms, leading to the impaired efficacy of tumor therapies. Nevertheless, current free anti-PD-1/PD-L1 therapy still suffered from poor therapeutic outcomes in most solid tumors due to the non-selective tumor accumulation, ineludible severe cytotoxic effects, as well as the common occurrence of immune resistance. Recently, nanoparticles with efficient tumor-targeting capacity, tumor-responsive prosperity, and versatility for combination therapy were identified as new avenues for PD-L1 targeting cancer immunotherapies. In this review, we first summarized the multiple functions of PD-L1 protein in promoting tumor growth, accelerating DDR, as well as depressing immunotherapy efficacy. Following this, the effects and mechanisms of current clinically widespread tumor therapies on tumor PD-L1 expression were discussed. Then, we reviewed the recent advances in nanoparticles for anti-PD-L1 therapy via using PD-L1 antibodies, small interfering RNA (siRNA), microRNA (miRNA), clustered, regularly interspaced, short palindromic repeats (CRISPR), peptide, and small molecular drugs. At last, we discussed the challenges and perspectives to promote the clinical application of nanoparticles-based PD-L1-targeting therapy.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou 325000, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhangping Li
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
18
|
Tian H, Shang H, Chen Y, Wu B, Wang C, Wang X, Cheng W. Sonosensitizer Nanoplatforms Augmented Sonodynamic Therapy-Sensitizing Shikonin-Induced Necroptosis Against Hepatocellular Carcinoma. Int J Nanomedicine 2023; 18:7079-7092. [PMID: 38050474 PMCID: PMC10693983 DOI: 10.2147/ijn.s435104] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/16/2023] [Indexed: 12/06/2023] Open
Abstract
Background Apoptosis resistance of hepatocellular carcinoma (HCC) often leads to treatment failure. Nonetheless, overcoming the resistance of HCC to apoptosis by inducing necroptosis of tumor cells to bypass the apoptotic pathway may be a promising treatment strategy. Sonodynamic therapy (SDT) has broad prospects in disease treatment because of its noninvasive characteristic and spatiotemporal control. The combination of SDT and shikonin in the treatment of HCC is expected to be a new tumor treatment method that can overcome apoptosis resistance. Methods In this study, the antitumor effect was evaluated using normal liver cell line WRL68, HCC cell line HepG2 and HepG2 xenograft mouse models. Indocyanine green (ICG) was loaded on nanobubbles (NBs) to construct ICG-loaded nanobubbles (ICG-NBs). Combined sonosensitizer nanoplatforms with ultrasound (US) to achieve efficient SDT, the combination of SDT and shikonin in treating HCC can activate shikonin-induced necroptosis. As a result, tumor cells that produced apoptosis resistance were destroyed by necroptosis. Results The results indicated a successful preparation of ICG-NBs with a uniform particle size of 273.0 ± 118.9 nm spherical structures. ICG-NB-mediated SDT, in combination with shikonin treatment, inhibited the viability, invasion, and migration of tumor cells. SDT + shikonin treatment group caused a substantial increase in necroptotic cells. The increased degree of tumor necrosis and the upregulated expression of receptor-interacting protein 3 kinase were observed in vivo studies, which indicated that the antitumor effect was accompanied by enhanced necroptosis in the SDT + shikonin treatment group. Conclusion ICG-NB-mediated SDT combined with shikonin inhibits the growth of HCC by increasing the necroptosis of tumor cells. Therefore, this combination therapy is a promising treatment strategy against the specific cancer.
Collapse
Affiliation(s)
- Huimin Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Haitao Shang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yichi Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Bolin Wu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Xiaodong Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
19
|
Wang H, He Z, Gao Y, Feng D, Wei X, Huang Y, Hou J, Li S, Zhang W. Dual-Pronged Attack: pH-Driven Membrane-Anchored NIR Dual-Type Nano-Photosensitizer Excites Immunogenic Pyroptosis and Sequester Immune Checkpoint for Enhanced Prostate Cancer Photo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302422. [PMID: 37544896 PMCID: PMC10558672 DOI: 10.1002/advs.202302422] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/14/2023] [Indexed: 08/08/2023]
Abstract
Prostate cancer (PCa) is a frustrating immunogenic "cold" tumor and generally receives unsatisfied immunotherapy outcomes in the clinic. Pyroptosis is an excellent immunogenic cell death form that can effectively activate the antitumor immune response, promote cytotoxic T-lymphocyte infiltration, and convert tumors from "cold" to "hot." However, the in vivo application of pyroptosis drugs is seriously limited, and the upregulation of tumor PD-L1 caused by photo-immunotherapy further promotes immune escape. Herein, a new nano-photosensitizer (YBS-BMS NPs-RKC) with pH-response integrating immunogenic pyroptosis induction and immune checkpoint blockade is developed. The pH-responsive polymer equipped with the cell membrane anchoring peptide RKC is used as the carrier and further encapsulated with the near-infrared-activated semiconductor polymer photosensitizer YBS and a PD-1/PD-L1 complex small molecule inhibitor BMS-202. The pH-driven membrane-anchoring and pyroptosis activation of YBS-BMS NPs-RKC is clearly demonstrated. In vitro and in vivo studies have shown that this dual-pronged therapy stimulates a powerful antitumor immune response to suppress primary tumor progression and evokes long-term immune memory to inhibit tumor relapse and metastasis. This work provides an effective self-synergistic platform for PCa immunotherapy and a new idea for developing more biocompatible photo-controlled pyroptosis inducers.
Collapse
Affiliation(s)
- He Wang
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhou215006China
| | - Zhangxin He
- Department of UrologyDushu Lake Hospital Affiliated to Soochow UniversityMedical Center of Soochow UniversitySuzhou Dushu Lake HospitalSuzhou215000China
| | - Yijian Gao
- College of Pharmaceutical SciencesSoochow UniversitySuzhou215000China
| | - Dexiang Feng
- Department of UrologyDushu Lake Hospital Affiliated to Soochow UniversityMedical Center of Soochow UniversitySuzhou Dushu Lake HospitalSuzhou215000China
| | - Xuedong Wei
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhou215006China
| | - Yuhua Huang
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhou215006China
| | - Jianquan Hou
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhou215006China
- Department of UrologyDushu Lake Hospital Affiliated to Soochow UniversityMedical Center of Soochow UniversitySuzhou Dushu Lake HospitalSuzhou215000China
| | - Shengliang Li
- College of Pharmaceutical SciencesSoochow UniversitySuzhou215000China
| | - Weijie Zhang
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhou215006China
- Department of UrologyDushu Lake Hospital Affiliated to Soochow UniversityMedical Center of Soochow UniversitySuzhou Dushu Lake HospitalSuzhou215000China
| |
Collapse
|
20
|
Iacobucci I, La Manna S, Cipollone I, Monaco V, Canè L, Cozzolino F. From the Discovery of Targets to Delivery Systems: How to Decipher and Improve the Metallodrugs' Actions at a Molecular Level. Pharmaceutics 2023; 15:1997. [PMID: 37514183 PMCID: PMC10385150 DOI: 10.3390/pharmaceutics15071997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Metals are indispensable for the life of all organisms, and their dysregulation leads to various disorders due to the disruption of their homeostasis. Nowadays, various transition metals are used in pharmaceutical products as diagnostic and therapeutic agents because their electronic structure allows them to adjust the properties of molecules differently from organic molecules. Therefore, interest in the study of metal-drug complexes from different aspects has been aroused, and numerous approaches have been developed to characterize, activate, deliver, and clarify molecular mechanisms. The integration of these different approaches, ranging from chemoproteomics to nanoparticle systems and various activation strategies, enables the understanding of the cellular responses to metal drugs, which may form the basis for the development of new drugs and/or the modification of currently used drugs. The purpose of this review is to briefly summarize the recent advances in this field by describing the technological platforms and their potential applications for identifying protein targets for discovering the mechanisms of action of metallodrugs and improving their efficiency during delivery.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- UMR7042 CNRS-Unistra-UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), European School of Chemistry, Polymers and Materials (ECPM), 67087 Strasbourg, France
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Sara La Manna
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Irene Cipollone
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Vittoria Monaco
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Luisa Canè
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Flora Cozzolino
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| |
Collapse
|
21
|
Wahnou H, Youlyouz-Marfak I, Liagre B, Sol V, Oudghiri M, Duval RE, Limami Y. Shining a Light on Prostate Cancer: Photodynamic Therapy and Combination Approaches. Pharmaceutics 2023; 15:1767. [PMID: 37376215 DOI: 10.3390/pharmaceutics15061767] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer is a major health concern worldwide, and current treatments, such as surgery, radiation therapy, and chemotherapy, are associated with significant side effects and limitations. Photodynamic therapy (PDT) is a promising alternative that has the potential to provide a minimally invasive and highly targeted approach to treating prostate cancer. PDT involves the use of photosensitizers (PSs) that are activated by light to produce reactive oxygen species (ROS), which can induce tumor cell death. There are two main types of PSs: synthetic and natural. Synthetic PSs are classified into four generations based on their structural and photophysical properties, while natural PSs are derived from plant and bacterial sources. Combining PDT with other therapies, such as photothermal therapy (PTT), photoimmunotherapy (PIT), and chemotherapy (CT), is also being explored as a way to improve its efficacy. This review provides an overview of conventional treatments for prostate cancer, the underlying principles of PDT, and the different types of PSs used in PDT as well as ongoing clinical studies. It also discusses the various forms of combination therapy being explored in the context of PDT for prostate cancer, as well as the challenges and opportunities associated with this approach. Overall, PDT has the potential to provide a more effective and less invasive treatment option for prostate cancer, and ongoing research is aimed at improving its selectivity and efficacy in clinical settings.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P. 2693, Maarif, Casablanca 20100, Morocco
| | - Ibtissam Youlyouz-Marfak
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco
| | | | - Vincent Sol
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France
| | - Mounia Oudghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P. 2693, Maarif, Casablanca 20100, Morocco
| | | | - Youness Limami
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P. 2693, Maarif, Casablanca 20100, Morocco
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco
| |
Collapse
|
22
|
Li Y, Lin L, Xie J, Wei L, Xiong S, Yu K, Zhang B, Wang S, Li Z, Tang Y, Chen G, Li Z, Yu Z, Wang X. ROS-Triggered Self-Assembled Nanoparticles Based on a Chemo-Sonodynamic Combinational Therapy Strategy for the Noninvasive Elimination of Hypoxic Tumors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15893-15906. [PMID: 36940438 DOI: 10.1021/acsami.3c00990] [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: 06/18/2023]
Abstract
The hypopermeability and hypoxia in the tumor milieu are important factors that limit multiple treatments. Herein, the reactive oxygen species (ROS)-triggered self-assembled nanoparticles (RP-NPs) was constructed. The natural small molecule Rhein (Rh) was encapsulated into RP-NPs as a sonosensitizer highly accumulated at the tumor site. Then highly tissue-permeable ultrasound (US) irradiation induced apoptosis of tumor cells through the excitation of Rh and acoustic cavitation, which prompted the rapid production of large amounts of ROS in the hypoxic tumor microenvironment. In addition, the thioketal bond structures in the innovatively designed prodrug LA-GEM were triggered and broken by ROS to achieve rapid targeted release of the gemcitabine (GEM). Sonodynamic therapy (SDT) increased the tissue permeability of solid tumors and actively disrupted redox homeostasis via mitochondrial pathways to kill hypoxic tumor cells, and the triggered response mechanism to GEM synergistically amplified the effect of chemotherapy. The chemo-sonodynamic combinational treatment approach is highly effective and noninvasive, with promising applications for hypoxic tumor elimination, such as in cervical cancer (CCa) patients who want to maintain their reproductive function.
Collapse
Affiliation(s)
- Yibing Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, China
| | - Ling Lin
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Jiashan Xie
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Lixue Wei
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Shuping Xiong
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Kunyi Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Bingchen Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Shengtao Wang
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University (Foshan Maternity & Child Healthcare Hospital), Foshan, 528000, China
| | - Zibo Li
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Yan Tang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Guimei Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, Guangdong 523058, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Xuefeng Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| |
Collapse
|