1
|
Liu Y, Li B, Yang R, Shang C, Bai Y, Zheng B, Zhao L. Ultrasound-triggered lysosomal alkalinization to block autophagy in tumor therapy. Biomaterials 2025; 320:123250. [PMID: 40081223 DOI: 10.1016/j.biomaterials.2025.123250] [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: 02/14/2025] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Lysosomes play a crucial role in regulating cancer progression and drug resistance. However, there is a pressing need for the development of drugs that can safely and effectively modulate the pH of cancerous lysosomes in a controlled manner. In this study, we propose a novel strategy for lysosomal alkalinization triggered by piezoelectricity. Our findings indicate that the electrons generated by (BaTiO3/Zr/Ca) BCZT under sonication effectively alkalinize the lysosomes. Molecular dynamics simulations further demonstrate that alterations in lysosomal pH lead to modifications in the conformation of V-ATPase (proton pump), enhancing its interaction with sodium ions while partially excluding hydrogen ions from entering the lysosomes. This mechanism helps maintain lysosomal alkalization, resulting in reduced hydrolase activity and preventing the degradation of proteins and damaged organelles. The accumulation of nanoparticles within the lysosomes causes swelling and gradual destruction of the lysosomal membrane. Consequently, this lysosomal dysfunction hampers the fusion with autophagosomes, inhibiting autophagy in tumor cells and promoting apoptosis in various tumor types. Our strategy significantly inhibited tumor volume growth in mice during animal studies. In conclusion, our piezoelectric-triggered lysosomal alkalinization strategy holds promise for innovative breakthroughs in the treatment of multiple cancers.
Collapse
Affiliation(s)
- Yong Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P R China; Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Bowen Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Run Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Chenxu Shang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Yang Bai
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China; Department of Stomatology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China; School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, China; FANGZHOU Biotechnology (Tianjin) Co. Ltd (FZBio), Building 5, Tianjin Science and Technology Square, East Research Road, Nankai District, Tianjin, 300192, China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P R China; Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121000, China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
2
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
3
|
Ismail M, Liu J, Wang N, Zhang D, Qin C, Shi B, Zheng M. Advanced nanoparticle engineering for precision therapeutics of brain diseases. Biomaterials 2025; 318:123138. [PMID: 39914193 DOI: 10.1016/j.biomaterials.2025.123138] [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: 09/18/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
Despite the increasing global prevalence of neurological disorders, the development of nanoparticle (NP) technologies for brain-targeted therapies confronts considerable challenges. One of the key obstacles in treating brain diseases is the blood-brain barrier (BBB), which restricts the penetration of NP-based therapies into the brain. To address this issue, NPs can be installed with specific ligands or bioengineered to boost their precision and efficacy in targeting brain-diseased cells by navigating across the BBB, ultimately improving patient treatment outcomes. At the outset of this review, we highlighted the critical role of ligand-functionalized or bioengineered NPs in treating brain diseases from a clinical perspective. We then identified the key obstacles and challenges NPs encounter during brain delivery, including immune clearance, capture by the reticuloendothelial system (RES), the BBB, and the complex post-BBB microenvironment. Following this, we overviewed the recent progress in NPs engineering, focusing on ligand-functionalization or bionic designs to enable active BBB transcytosis and targeted delivery to brain-diseased cells. Lastly, we summarized the critical challenges hindering clinical translation, including scalability issues and off-target effects, while outlining future opportunities for designing cutting-edge brain delivery technologies.
Collapse
Affiliation(s)
- Muhammad Ismail
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ningyang Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Changjiang Qin
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China.
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meng Zheng
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
4
|
Pu J, Yuan K, Tao J, Qin Y, Li Y, Fu J, Li Z, Zhou H, Tang Z, Li L, Gai X, Qin D. Glioblastoma multiforme: an updated overview of temozolomide resistance mechanisms and strategies to overcome resistance. Discov Oncol 2025; 16:731. [PMID: 40353925 PMCID: PMC12069213 DOI: 10.1007/s12672-025-02567-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with high lethality. The typical treatment regimen includes post-surgical radiotherapy and temozolomide (TMZ) chemotherapy, which helps extend survival. Nevertheless, TMZ resistance occurs in approximately 50% of patients. This resistance is primarily associated with the expression of O6-methylguanine-DNA methyltransferase (MGMT), which repairs O6-methylguanine lesions generated by TMZ and is thought to be the major mechanism of drug resistance. Additionally, the mismatch repair and base excision repair pathways play crucial roles in TMZ resistance. Emerging studies also point to drug transport mechanisms, glioma stem cells, and the heterogeneous tumor microenvironment as additional influences on TMZ resistance in gliomas. A better understanding of these mechanisms is vital for developing new treatments to improve TMZ effectiveness, such as DNA repair inhibitors, inhibitors of multidrug transporting proteins, TMZ analogs, and combination therapies targeting multiple pathways. This article discusses the main resistance mechanisms and potential strategies to counteract resistance in GBM patients, aiming to broaden the understanding of these mechanisms for future research and to explore the therapeutic effects of traditional Chinese medicines and their active components in overcoming TMZ resistance.
Collapse
Affiliation(s)
- Jianlin Pu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Tao
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Yongxin Li
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Jing Fu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhong Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Haimei Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhengxiu Tang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Li
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xuesong Gai
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
5
|
Xu Q, Hu X, Ullah I, Lin T, Liu Y, Dong H, Zong Q, Yuan Y, Wang J. Biomimetic Hybrid PROTAC Nanovesicles Block Multiple DNA Repair Pathways to Overcome Temozolomide Resistance Against Orthotopic Glioblastoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504253. [PMID: 40347032 DOI: 10.1002/adma.202504253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/28/2025] [Indexed: 05/12/2025]
Abstract
Glioblastoma (GBM) remains one of the deadliest forms of cancer due to its high rates of postoperative recurrence and resistance to treatment. Temozolomide (TMZ) is the standard chemotherapy for GBM. However, the therapeutic efficacy of TMZ is significantly compromised by the activation of various intracellular DNA repair mechanisms that facilitate TMZ resistance. Herein, the upregulation of bromodomain-containing protein 4 (BRD4) expression is demonstrated to be a key contributor to TMZ resistance in GBM. To address this challenge, a biomimetic hybrid PROteolysis TArgeting Chimeras (PROTAC) liposome delivery system (M@TP) is developed. This system efficiently penetrates the blood-brain barrier (BBB) and specifically targets GBM cells through homotypic recognition. Once within TMZ-resistant GBM cells, the released PROTAC from M@TP can specifically degrade BRD4, effectively inhibiting multiple DNA repair pathways and restoring sensitivity to TMZ treatment. In vivo, studies showed that M@TP demonstrated significant efficacy in suppressing tumor growth in both TMZ-resistant and postoperative GBM, with prolonged mouse survival times. These findings highlight the potential of M@TP as a promising strategy to overcome TMZ resistance and improve therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Qing Xu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Xiangyu Hu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Taian Lin
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Ye Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - He Dong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Qingyu Zong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
6
|
Mandal M, Banerjee I, Mandal M. Effective approaches in conquering chemoresistance of glioblastoma: potential for nanoformulations. Drug Deliv Transl Res 2025:10.1007/s13346-025-01859-z. [PMID: 40259195 DOI: 10.1007/s13346-025-01859-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Glioblastoma Multiforme is an aggressive and complex cancer affecting mostly elderly patients above the age of 60 years. Originally classified as the fourth stage of glioma, it has an abysmal prognosis along with limited therapeutic options. Surgical removal of tumors, radiotherapy, and chemotherapy are prevalent treatment strategies with numerous therapeutic obstacles, including undefined boundary of tumor mass leaving traces even after excision, chances of secondary cancer formation, and presence of blood-brain barrier. These blood-brain and blood-brain tumor barriers actively restrict the permeability of many molecules from blood circulation to enter the central nervous system. Therefore, many conventional antineoplastic drugs fail to reach the tumor periphery except temozolomide. Meanwhile, active stem cells in the tumor microenvironment, genetic mutation inducing tumor growth, and epigenetic pattern alteration make this cancer chemoresistant. Our review delineates the recent approaches to resensitize the existing clinical drugs through specifically designed nanoformulations. Nanoparticles with modified physiological characteristics and modified through technological parameters can reduce the tumor's stemness, which increases tumor cells' apoptosis rate. Moreover, these nanoparticles can efficiently traverse the blood-brain barrier and escape from endosomal degradation with minimum toxicological impact. Overall, this review discusses the cancer chemoresistance phenomena and related pathways and highlights the potential of nanoformulation in reversing chemoresistance. Also, the existing limitations of this unique approach and suggestions are discussed at the end of the article, which may facilitate the identification of new directions for advancement of the nanoparticle-mediated reversal of chemoresistance.
Collapse
Affiliation(s)
- Madhurima Mandal
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Indranil Banerjee
- Department of Pharmaceutical Technology, JIS University, 81, Nilgunj Road, Agarpara, Kolkata, West Bengal, 700109, India.
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
7
|
Shi Y, Yu Q, Tan L, Wang Q, Zhu WH. Tumor Microenvironment-Responsive Polymer Delivery Platforms for Cancer Therapy. Angew Chem Int Ed Engl 2025:e202503776. [PMID: 40214115 DOI: 10.1002/anie.202503776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
Most chemotherapeutic and bioimaging agents struggle with inadequate bioavailability, primarily due to their limited biocompatibility and lack of specificity in targeting, leading to low or decreased anticancer efficacy and inaccurate imaging. To surmount these obstacles, the development of stimuli-responsive polymer delivery platforms, predominantly leveraging the tumor microenvironment (TME), has emerged as a promising strategy. Therapeutic and diagnostic agents can be released controllably at the tumor site by virtue of the bond cleavage or hydrophobic to hydrophilic transformation of TME-sensitive linkages in TME-responsive systems, thus augmenting cancer treatment and imaging precision, while simultaneously attenuating the damage to healthy tissues and false imaging signals caused by non-specific drug leakage. In this comprehensive review, we scrutinize recent studies of TME-responsive polymer delivery platforms, encompassing pH-, ROS-, GSH-, enzyme-, and hypoxia-responsive vectors, significantly from the perspective of their molecular design and responsive mechanism, and further summarizing their bio-application in drug delivery and diagnostic imaging. Moreover, this review encapsulates the critical challenges and offers an insightful perspective on the future prospects of TME-responsive polymer delivery platforms in terms of molecular and vector design.
Collapse
Affiliation(s)
- Yiqi Shi
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Qianqian Yu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Lijie Tan
- Department of Thoracic Surgery, Cancer Center, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Qi Wang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Wei-Hong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
- Center of Photosensitive Chemicals Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| |
Collapse
|
8
|
Zheng G, Ding L, Gan J, Li T, Liu X, Zhang X, Wang P, Wei D. Mesoporous bowl-shaped polydopamine co-loaded temozolomide and indocyanine green for synergistically inhibiting glioblastoma. SCIENCE CHINA MATERIALS 2025. [DOI: 10.1007/s40843-025-3311-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/04/2025] [Indexed: 05/03/2025]
|
9
|
Mao M, Lei Y, Ma X, Xie HY. Challenges and Emerging Strategies of Immunotherapy for Glioblastoma. Chembiochem 2025; 26:e202400848. [PMID: 39945240 DOI: 10.1002/cbic.202400848] [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: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
Glioblastoma (GBM) is recognized as the most lethal primary malignant tumor of the central nervous system. Although traditional treatments can somewhat prolong patient survival, the overall prognosis remains grim. Immunotherapy has become an effective method for GBM treatment. Oncolytic virus, checkpoint inhibitors, CAR T cells and tumor vaccines have all been applied in this field. Moreover, the combining of immunotherapy with traditional radiotherapy, chemotherapy, or gene therapy can further improve the treatment outcome. This review systematically summarizes the features of GBM, the recent progress of immunotherapy in overcoming GBM.
Collapse
Affiliation(s)
- Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yao Lei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Hai-Yan Xie
- Chemical Biology Center, Peking University, Beijing, 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
10
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [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: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|
11
|
Zhou L, Li Z, Zhou S, Wang B, Liang Z, Hu S, Zhang H, Duan L, Zhao D, Cheng L, Ren H, Wakimoto H, Li M. Targeting NAD + biosynthesis suppresses TGF-β1/Smads/RAB26 axis and potentiates cisplatin cytotoxicity in non-small cell lung cancer brain metastasis. Acta Neuropathol Commun 2025; 13:56. [PMID: 40069888 PMCID: PMC11895195 DOI: 10.1186/s40478-025-01967-4] [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: 07/14/2024] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) plays an important role in tumor progression, but its role in non-small cell lung cancer with brain metastasis (NSCLC BM) remains unclear. Herein, we investigated NAD+ biosynthesis targeting as a new therapeutic strategy for NSCLC BM. Therapeutic activity of nicotinamide phosphoribosyl transferase (NAMPT) inhibitors was evaluated in mouse models of NSCLC BM and using various assays such as NAD+ quantitation, cell viability, and apoptosis assays. To explore impact on downstream signaling, RNA sequencing was used in NAMPT inhibitor-treated and control cells, followed by validation with genetic knockdown, western blot and qRT-PCR. Expression of NAMPT and downstream proteins in human NSCLC BM and its association with patient prognosis were examined. Finally, combination of NAMPT inhibitor and cisplatin was tested in vivo. Systemic treatment with NAMPT inhibitors demonstrated intracranial activity in an NSCLC BM model. NAMPT inhibitors decreased cellular NAD levels and suppressed proliferation and invasion, and induced apoptosis in NSCLC cells. Supplementation with NAD+ precursor NMN rescued these NAMPT inhibitor effects. Mechanistically, disruption of NAMPT-mediated NAD+ biosynthesis suppressed TGF-β1/Smads/RAB26 signaling, leading to inhibition of NSCLC cells. Expression of NAMPT/TGF-β1/Smads/RAB26 axis proteins was upregulated in NSCLC BM tissues and correlated with poor prognosis. Combining NAMPT inhibitors with cisplatin further extended the survival of NSCLC BM-bearing mice. Targeting NAD+ biosynthesis provides a new therapeutic strategy for NSCLC BM and can be effectively combined with cisplatin. Our studies identified the TGF-β1/Smads/RAB26 signaling downstream of NAMPT, which was targeted by NAMPT inhibition to mediate anti-cancer effects.
Collapse
Affiliation(s)
- Liyun Zhou
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Zhiying Li
- Department of Neurosurgery, The 7th People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Shengli Zhou
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Bin Wang
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Zhen Liang
- Department of Neurosurgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Sen Hu
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Hang Zhang
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Lin Duan
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Dongxu Zhao
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Luyao Cheng
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Hang Ren
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| | - Ming Li
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, People's Republic of China.
| |
Collapse
|
12
|
Zheng H, Zhang L, Bai X, Zhu J, Liu S, Ke Y, Lin Q, Yuan Y, Ji T. GCN5-targeted dual-modal probe across the blood-brain barrier for borders display in invasive glioblastoma. Nat Commun 2025; 16:2345. [PMID: 40057495 PMCID: PMC11890771 DOI: 10.1038/s41467-025-57598-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
Glioblastoma (GBM) is a highly invasive malignancy with a poor prognosis, primarily attributable to its diffuse infiltration into adjacent brain tissue, thereby complicating effective surgical resection. Current imaging modalities often struggle to accurately identify tumor boundaries. Here, we identify general control non-repressed protein 5 (GCN5) as a promising molecular target for GBM imaging, as it is expressed in GBM lesions within brain tissue, and its expression levels are significantly correlated with GBM grading. We develop a dual-modal probe with a particle size of 20 nm, capable of efficiently traversing the blood-brain barrier (BBB) to target GCN5 through adsorptive-mediated transcytosis (AMT). The probe employs dendrimers (Den) as carriers, which are loaded with a small molecule inhibitor specifically designed to target GCN5. This probe enhances the preoperative delineation of GBM boundaries using magnetic resonance imaging (MRI) and facilitates intraoperative fluorescence image-guided surgical procedures. Our work introduces a promising tool for boundary delineation, offering new opportunities for the precise resection of GBM.
Collapse
Affiliation(s)
- Haiyan Zheng
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Zhang
- Nuclear Medical Department, Changhai Hospital affiliated to Naval Medical University, Shanghai, China
| | - Xinning Bai
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinchao Zhu
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanshan Liu
- Chenggong Hospital, Xiamen University, Xiamen, China
| | - Yao Ke
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingyuan Lin
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Yuan
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianhai Ji
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Su X, Liu Y, Zhong Y, Shangguan P, Liu J, Luo Z, Qi C, Guo J, Li X, Lin D, Wang G, Wang D, Han T, Wang J, Shi B, Tang BZ. A Brain-Targeting NIR-II Polymeric Phototheranostic Nanoplatform toward Orthotopic Drug-Resistant Glioblastoma. NANO LETTERS 2025; 25:3445-3454. [PMID: 39992704 DOI: 10.1021/acs.nanolett.4c05470] [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: 02/26/2025]
Abstract
Glioblastoma is the most common and devastating brain tumor owing to its high invasiveness and high-frequency drug resistance. Near infrared-II (NIR-II) imaging-guided phototherapy based on polymer luminogens provides a promising remedy against drug-resistant glioma, but it is difficult to maximize photoenergy utilization. Herein, we designed a series of semiconducting polymers to boost the visualization and ablation of glioblastoma. By subtly engineering the side chains or substituents on the phenothiazine and thiophene moieties, an NIR-II polymer luminogen with high-quality fluorescence performance, good solubility, superior photothermal conversion, and balanced reactive oxygen species generation is achieved. The optimal polymer possesses a branched alkyl chain and tetraphenylethylene pendant to manipulate the equilibrium between the radiative and nonradiative energy-dissipating channels. High-sensitivity NIR-II imaging was used to monitor the blood-brain barrier penetration and glioma cell targeting of apolipoprotein E-modified polymer nanoparticles. The NIR irradiation triggers and maximizes the photon utilization in prominent photodynamic/photothermal synergistic therapy in orthotopic drug-resistant glioblastoma.
Collapse
Affiliation(s)
- Xiang Su
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yisheng Liu
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Yong Zhong
- Key Laboratory for Special Functional Materials of Ministry of Education, Henan University, Kaifeng, 475004, China
| | - Ping Shangguan
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Junkai Liu
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Zhengqun Luo
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Cai Qi
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Jincheng Guo
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Xi Li
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Danmin Lin
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Gaoyang Wang
- Key Laboratory for Special Functional Materials of Ministry of Education, Henan University, Kaifeng, 475004, China
| | - Dong Wang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Ting Han
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jiefei Wang
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Bingyang Shi
- The Zhongzhou Laboratory for Integrative Biology, Henan Provincial Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Department of Clinical Laboratory of Huaihe Hospital, Henan University, Kaifeng, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen, 518172, China
| |
Collapse
|
14
|
Wang L, Gu M, Zhang X, Kong T, Liao J, Zhang D, Li J. Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413367. [PMID: 39854126 PMCID: PMC11905078 DOI: 10.1002/advs.202413367] [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: 10/21/2024] [Revised: 12/14/2024] [Indexed: 01/26/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
Collapse
Affiliation(s)
- Liyin Wang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Min Gu
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Xiaoli Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | | | - Jun Liao
- Institute of Systems BiomedicineBeijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Dan Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Jingwu Li
- The First Hospital of China Medical UniversityLiaoning110001China
| |
Collapse
|
15
|
Liu Y, Wu H, Liang G. Combined Strategies for Nanodrugs Noninvasively Overcoming the Blood-Brain Barrier and Actively Targeting Glioma Lesions. Biomater Res 2025; 29:0133. [PMID: 39911305 PMCID: PMC11794768 DOI: 10.34133/bmr.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 02/07/2025] Open
Abstract
Drugs for tumor treatment face various challenges, including poor solubility, poor stability, short blood half-life, nontargeting ability, and strong toxic side effects. Fortunately, nanodrug delivery systems provide excellent solution to these problems. However, nanodrugs for glioma treatment also face some key challenges including overcoming the blood-brain barrier (BBB) and, specifically, accumulation in glioma lesions. In this review, we systematically summarize the advantages and disadvantages of combined strategies for nanodrugs noninvasively overcoming BBB and actively targeting glioma lesions to achieve effective glioma therapy. Common noninvasive strategies for nanodrugs overcoming the BBB include bypassing the BBB via the nose-to-brain route, opening the tight junction of the BBB by focused ultrasound with microbubbles, and transendothelial cell transport by intact cell loading, ligand decoration, or cell membrane camouflage of nanodrugs. Actively targeting glioma lesions after overcoming the BBB is another key factor helping nanodrugs accurately treat in situ gliomas. This aim can also be achieved by loading nanodrugs into intact cells and modifying ligand or cell membrane fragments on the surface of nanodrugs. Targeting decorated nanodrugs can guarantee precise glioma killing and avoid side effects on normal brain tissues that contribute to the specific recognition of glioma lesions. Furthermore, the challenges and prospects of nanodrugs in clinical glioma treatment are discussed.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| | - Haigang Wu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan Province 475004, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| |
Collapse
|
16
|
Sun B, Li R, Ji N, Liu H, Wang H, Chen C, Bai L, Su J, Chen J. Brain-targeting drug delivery systems: The state of the art in treatment of glioblastoma. Mater Today Bio 2025; 30:101443. [PMID: 39866779 PMCID: PMC11759563 DOI: 10.1016/j.mtbio.2025.101443] [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: 08/25/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant brain tumor, characterized by a high mortality rate and a poor prognosis. The blood-brain barrier (BBB) and the blood-tumor barrier (BTB) present significant obstacles to the efficacy of tumor-targeted pharmacotherapy, thereby impeding the therapeutic potential of numerous candidate drugs. Targeting delivery of adequate doses of drug across the BBB to treat GBM has become a prominent research area in recent years. This emphasis has driven the exploration and evaluation of diverse technologies for GBM pharmacotherapy, with some already undergoing clinical trials. This review provides a thorough overview of recent advancements and challenges in targeted drug delivery for GBM treatment. It specifically emphasizes systemic drug administration strategies to assess their potential and limitations in GBM treatment. Furthermore, this review highlights promising future research directions in the development of intelligent drug delivery systems aimed at overcoming current challenges and enhancing therapeutic efficacy against GBM. These advancements not only support foundational research on targeted drug delivery systems for GBM but also offer methodological approaches for future clinical applications.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Rong Li
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chao Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
17
|
Tokhtueva MD, Melekhin VV, Abramov VM, Ponomarev AI, Prokofyeva AV, Grzhegorzhevskii KV, Paramonova AV, Makeev OG, Eltsov OS. The arylbipyridine platinum (II) complex increases the level of ROS and induces lipid peroxidation in glioblastoma cells. Biometals 2025; 38:185-202. [PMID: 39397212 DOI: 10.1007/s10534-024-00646-6] [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: 05/14/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Here we present the biological properties of the arylbipyridine platinum (II) complex (arylbipy-Pt) and describe the potential mechanism of its antitumor action which differs from that of the well-known cisplatin. Leading to the oxidative stress and lipid peroxidation, the arylbipyridine platinum (II) complex showcases the significant cytotoxicity against the glioblastoma cells as shown by the MTT test. Using the 5-ethyl-2-deoxyuridine we study the proliferative activity of glioblastoma cells to affirm that arylbipyridine platinum (II) complex does not impede cell division or DNA replication. Staining by the MitoCLox dye and 2',7'-dichlorodihydrofluorescein diacetate demonstrates that the glioblastoma cells treated with arylbipy-Pt exhibit a strong increase of the lipid peroxidation and the stimulation of the reactive oxygen species formation. The hypothesis that arylbipy-Pt promotes oxidative death of tumor cells is confirmed by control experiments using N-acetyl-L-cysteine as an antioxidant. Further evidence for the oxidative mechanism of action is provided by real-time PCR, which shows high expression levels for genes associated with the heat shock proteins HSP27 and HSP70, which can be used as markers of tumor cell ferroptosis. To elucidate the chemical nature of the arylbipy-Pt complex activity, we perform 195Pt NMR spectroscopy and cyclic voltammetry measurements under biologically relevant conditions. The results obtained clearly indicate the structural transformation of the arylbipy-Pt complex in the DMSO-saline mixture, which is crucial for its further antitumor activity via the oxidative pathway. The found correlation between the molecular structure of arylbipy-Pt and its effect on the tumor cell cycle paves the way for the rational design of Pt complexes possessing the alternative mechanism of antitumor activity as compared to DNA intercalation, providing possible solutions to the major problems such as toxicity and drug resistance.
Collapse
Affiliation(s)
- Maria D Tokhtueva
- Scientific, Educational and Innovative Center of Chemical and Pharmaceutical Technologies, Ural Federal University, 620002, Yekaterinburg, Russian Federation.
| | - Vsevolod V Melekhin
- Scientific, Educational and Innovative Center of Chemical and Pharmaceutical Technologies, Ural Federal University, 620002, Yekaterinburg, Russian Federation
- Department of Medical Biology and Genetics, Ural State Medical University, Yekaterinburg, Russian Federation
| | - Vladislav M Abramov
- Scientific, Educational and Innovative Center of Chemical and Pharmaceutical Technologies, Ural Federal University, 620002, Yekaterinburg, Russian Federation
| | - Alexander I Ponomarev
- Department of Medical Biology and Genetics, Ural State Medical University, Yekaterinburg, Russian Federation
- Molecular Biology, Immunophenotyping and Pathomorphology Department, Regional Children's Hospital, Yekaterinburg, Russian Federation
| | - Anna V Prokofyeva
- Institute of Natural Sciences and Mathematics, Ural Federal University, Yekaterinburg, Russian Federation
| | - Kirill V Grzhegorzhevskii
- Institute of Natural Sciences and Mathematics, Ural Federal University, Yekaterinburg, Russian Federation
| | - Anastasia V Paramonova
- Scientific, Educational and Innovative Center of Chemical and Pharmaceutical Technologies, Ural Federal University, 620002, Yekaterinburg, Russian Federation
| | - Oleg G Makeev
- Department of Medical Biology and Genetics, Ural State Medical University, Yekaterinburg, Russian Federation
- Laboratory of Cell and Gene Therapy Technologies, Institute of Medical Cell Technologies, Institute of Medical Cell Technologies, Yekaterinburg, Russian Federation
| | - Oleg S Eltsov
- Scientific, Educational and Innovative Center of Chemical and Pharmaceutical Technologies, Ural Federal University, 620002, Yekaterinburg, Russian Federation
| |
Collapse
|
18
|
Cheng W, Duan Z, Chen H, Wang Y, Wang C, Pan Y, Wu J, Wang N, Qu H, Xue X. Macrophage membrane-camouflaged pure-drug nanomedicine for synergistic chemo- and interstitial photodynamic therapy against glioblastoma. Acta Biomater 2025; 193:392-405. [PMID: 39800099 DOI: 10.1016/j.actbio.2025.01.016] [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: 09/23/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Glioblastoma (GBM) persists as a highly fatal malignancy, with current clinical treatments showing minimal progress over years. Interstitial photodynamic therapy (iPDT) holds promise due to its minimally invasive nature and low toxicity but is impeded by poor photosensitizer penetration and inadequate GBM targeting. Here, we developed a biomimetic pure-drug nanomedicine (MM@CT), which co-assembles the photosensitizer chlorin e6 (Ce6) and the first-line chemotherapeutic drug (temozolomide, TMZ) for GBM, then camouflaged with macrophage membranes. This design eliminates the need for traditional excipients, ensuring formulation safety and achieving exceptionally high drug loading with 73.2 %. By leveraging the biomimetic properties of macrophage membranes, MM@CT evades clearance by the mononuclear phagocyte system and can overcome blood circulatory barriers to target intracranial GBM tumors due to its inherent tumor-homing ability. Consequently, this targeted strategy enables precise delivery of TMZ to the tumor site while significantly enhancing Ce6 accumulation within the tumor tissue. Upon intra-tumoral irradiation using an optical fiber, activated Ce6 synergizes with TMZ to exert both cytotoxic effects from chemotherapy and unique advantages from iPDT simultaneously attacking GBM tumors in a dual manner. In subcutaneous and intracranial GBM mouse models, MM@CT exhibits remarkable anti-tumor efficacy with minimal systemic toxicity, emerging as a promising GBM treatment strategy. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) remains a formidable brain cancer, posing significant therapeutic challenges due to the presence of the blood-brain barrier (BBB) and tumor heterogeneity. To overcome these obstacles, we have developed MM@CT, a biomimetic nanomedicine with exceptional drug loading efficiency of 73.2 %. MM@CT incorporates the photosensitizer Ce6 and chemotherapy agent TMZ, encapsulated within nanoparticles and camouflaged with macrophage membranes. This innovative design enables efficient BBB penetration, precise tumor targeting, and synergistic application of chemotherapy and photodynamic therapy. Encouragingly, preclinical evaluations have demonstrated substantial antitumor activity with minimal systemic toxicity, positioning MM@CT as a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanjun Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ning Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
19
|
Liu H, Jin X, Liu S, Liu X, Pei X, Sun K, Li M, Wang P, Chang Y, Wang T, Wang B, Yu XA. Recent advances in self-targeting natural product-based nanomedicines. J Nanobiotechnology 2025; 23:31. [PMID: 39833846 PMCID: PMC11749302 DOI: 10.1186/s12951-025-03092-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Natural products, recognized for their potential in disease prevention and treatment, have been integrated with advanced nano-delivery systems to create natural product-based nanomedicines, offering innovative approaches for various diseases. Natural products derived from traditional Chinese medicine have their own targeting effect and remarkable therapeutic effect on many diseases, but there are some shortcomings such as poor physical and chemical properties. The construction of nanomedicines using the active ingredients of natural products has become a key step in the modernization research process, which could be used to make up for the defects of natural products such as low solubility, large dosage, poor bioavailability and poor targeting. Nanotechnology enhances the safety, selectivity, and efficacy of natural products, positioning natural product-based nanomedicines as promising candidates in medicine. This review outlines the current status of development, the application in different diseases, and safety evaluation of natural product-based nanomedicines, providing essential insights for further exploration of the synergy between natural products and nano-delivery systems in disease treatment.
Collapse
Affiliation(s)
- Haifan Liu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xingyue Jin
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Suyi Liu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinyue Liu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiao Pei
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Kunhui Sun
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Meifang Li
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Ping Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Yanxu Chang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tiejie Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Bing Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xie-An Yu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
| |
Collapse
|
20
|
Qin W, Li H, Chen J, Qiu Y, Ma L, Nie L. Amphiphilic hemicyanine molecular probes crossing the blood-brain barrier for intracranial optical imaging of glioblastoma. SCIENCE ADVANCES 2025; 11:eadq5816. [PMID: 39813352 PMCID: PMC11734739 DOI: 10.1126/sciadv.adq5816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Intracranial optical imaging of glioblastoma (GBM) is challenging due to the scarcity of effective probes with blood-brain barrier (BBB) permeability and sufficient imaging depth. Herein, we describe a rational strategy for designing optical probes crossing the BBB based on an electron donor-π-acceptor system to adjust the lipid/water partition coefficient and molecular weight of probes. The amphiphilic hemicyanine dye (namely, IVTPO), which exhibits remarkable optical properties and effective BBB permeability, is chosen as an efficient fluorescence/photoacoustic probe for in vivo real-time imaging of orthotopic GBM with high resolution through the intact skull. Abnormal leakage of IVTPO adjacent to the developing tumor is unambiguously observed at an early stage of tumor development prior to impairment of BBB integrity, as assessed by commercial Evans blue (EB). Compared with EB, IVTPO demonstrates enhanced optical imaging capability and improved tumor-targeting efficacy. These results offer encouraging insights into medical diagnosis of intracranial GBM.
Collapse
Affiliation(s)
- Wei Qin
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Honghui Li
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Jiali Chen
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yang Qiu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Limin Ma
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Liming Nie
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Cardiovascular Institute, Guangzhou 510080, China
| |
Collapse
|
21
|
Cheng W, Qu H, Yang J, Chen H, Pan Y, Duan Z, Xue X. Hierarchically Engineered Self-Adaptive Nanoplatform Guided Intuitive and Precision Interventions for Deep-Seated Glioblastoma. ACS NANO 2025; 19:557-579. [PMID: 39754309 DOI: 10.1021/acsnano.4c11006] [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: 01/06/2025]
Abstract
Glioblastoma multiforme (GBM), particularly the deep-seated tumor where surgical removal is not feasible, poses great challenges for clinical treatments due to complicated biological barriers and the risk of damaging healthy brain tissue. Here, we hierarchically engineer a self-adaptive nanoplatform (SAN) that overcomes delivery barriers by dynamically adjusting its structure, surface charge, particle size, and targeting moieties to precisely distinguish between tumor and parenchyma cells. We further devise a SAN-guided intuitive and precision intervention (SGIPi) strategy which obviates the need for sophisticated facilities, skilled operations, and real-time magnetic resonance imaging (MRI) guidance required by current MRI-guided laser or ultrasound interventions. In a preclinical intracranial GBM mouse model, SGIPi-based photodynamic therapy effectively impedes GBM progression with high tumor specificity and significantly extends overall survival. Moreover, the SGIPi potentiates chemotherapy while minimizing adverse effects; it eradicates intracranial GBM lesions in 100% cases solely through Temozolomide chemotherapy. This SGIPi strategy holds potential to improve the clinical management of GBM, with the possibility of extending survival rates and even achieving complete remission, and may inspire research focus from expensive and complex hardware development to simpler, delivery-based GBM interventions.
Collapse
Affiliation(s)
- Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiaojiao Yang
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
22
|
Ma J, Qiu J, Wright GA, Wang S. Oxygen/Nitric Oxide Dual-Releasing Nanozyme for Augmenting TMZ-Mediated Apoptosis and Necrosis. Mol Pharm 2025; 22:168-180. [PMID: 39571173 PMCID: PMC11707740 DOI: 10.1021/acs.molpharmaceut.4c00817] [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: 07/23/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 01/07/2025]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant brain tumor, with a poor prognosis. Temozolomide (TMZ) represents the standard chemotherapy for GBM but has limited efficacy due to poor targeting and a hypoxic tumor microenvironment (TME). To address these challenges, we developed a dual-gas-releasing, cancer-cell-membrane-camouflaged nanoparticle to deliver TMZ. This nanoceria, camouflaged with a cancer cell membrane (CCM-CeO2), targets explicitly GBM cells and accumulates in lysosomes, triggering the rapid release of TMZ. Additionally, CCM-CeO2 could release oxygen (O2) and nitric oxide (NO) in response to the TME. Synthesized using d-arginine, catalytic nanoceria could decompose excessive hydrogen peroxide (H2O2) in the TME to produce O2, while d-arginine could nonenzymatically react with H2O2 to generate NO. CCM-CeO2 could penetrate GBM spheroids to a depth of 148.3 ± 31 μm, with the O2 and NO produced, reducing HIF-1α protein expression. When loaded with TMZ, CCM-CeO2 could increase the intracellular ROS produced by TMZ, leading to lysosome membrane permeabilization and notably augmented apoptosis and necrosis in GBM cells. An in vitro antitumor assay using spheroids showed that CCM-CeO2 reduced the IC50 value of TMZ from 174.5 to 42.6 μg/mL, likely due to the catalase-like activity of nanoceria. These results suggest that alleviating hypoxia and increasing ROS produced by chemotherapeutics could be an effective therapeutic strategy for treating GBM.
Collapse
Affiliation(s)
- Jun Ma
- Department
of Biomedical Engineering, Texas A&M
University, College Station, Texas 77843, United States
| | - Jingjing Qiu
- Department
of Mechanical Engineering & Department of Materials Science and
Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Gus A. Wright
- Flow
Cytometry Facility, College of Veterinary Medicine & Biomedical
Sciences, Texas A&M University, College Station, Texas 77843, United States
| | - Shiren Wang
- Department
of Industrial Systems and Engineering & Department of Materials
Science and Engineering & Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
23
|
Wang S, Yang L, He W, Zheng M, Zou Y. Cell Membrane Camouflaged Biomimetic Nanoparticles as a Versatile Platform for Brain Diseases Treatment. SMALL METHODS 2025; 9:e2400096. [PMID: 38461538 DOI: 10.1002/smtd.202400096] [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: 01/19/2024] [Revised: 02/27/2024] [Indexed: 03/12/2024]
Abstract
Although there are various advancements in biomedical in the past few decades, there are still challenges in the treatment of brain diseases. The main difficulties are the inability to deliver a therapeutic dose of the drug to the brain through the blood-brain barrier (BBB) and the serious side effects of the drug. Thus, it is essential to select biocompatible drug carriers and novel therapeutic tools to better enhance the effect of brain disease treatment. In recent years, biomimetic nanoparticles (BNPs) based on natural cell membranes, which have excellent biocompatibility and low immunogenicity, are widely used in the treatment of brain diseases to enable the drug to successfully cross the BBB and target brain lesions. BNPs can prolong the circulation time in vivo, are more conducive to drug aggregation in brain lesions. Cell membranes (CMs) from cancer cells (CCs), red blood cells (RBCs), white blood cells (WBCs), and so on are used as biomimetic coatings for nanoparticles (NPs) to achieve the ability to target, evade clearance, or stimulate the immune system. This review summarizes the application of different cell sources as BNPs coatings in the treatment of brain diseases and discusses the possibilities and challenges of clinical translation.
Collapse
Affiliation(s)
- Shiyu Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Longfei Yang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Wenya He
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meng Zheng
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan Zou
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
24
|
Wei F, Lin Y, Pan R, Peng Y, Chen E, Kang J, Zhu J, Wang J, Wu B, Shen W, Lin J, Gao H, Tian X. Virus-Inspired Biodegradable Tetrasulfide-Bridged Mesoporous Organosilica with GSH Depletion for Fluorescence Imaging-Guided Sonodynamic Chemotherapy of Glioblastoma Multiforme. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70407-70418. [PMID: 39670856 DOI: 10.1021/acsami.4c19480] [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: 12/14/2024]
Abstract
Glioblastoma multiforme (GBM), a highly prevalent and lethal form of malignant tumor, is typically treated with Temozolomide (TMZ), a chemotherapeutic agent. Nevertheless, the effectiveness of TMZ is hampered by inadequate cell entry, systemic adverse effects, and monotherapy constraints. Previous clinical studies have demonstrated that combination therapy can significantly enhance the therapeutic efficacy. Herein, we developed ultrasmall virus-inspired biodegradable tetrasulfide-bridged mesoporous organosilica coloaded with TMZ and indocyanine green (ICG) (designated as vMSTI) for fluorescence imaging-guided sonodynamic chemotherapy and glutathione (GSH) depletion, aiming to enhance the therapeutic efficiency of GBM. Once accumulated within the tumors, the vMSTI nanosystem efficiently entered tumor cells via "spike surface"-assisted endocytosis. Subsequently, intracellular overproduction of GSH within tumor cells triggered the degradation of vMSTI, resulting in the release of both TMZ and ICG, while simultaneously depleting intracellular GSH levels. Upon ultrasound (US) irradiation, the released ICG generated abundant reactive oxygen species (ROS) for sonodynamic therapy, which could be further potentiated by GSH depletion. Furthermore, released TMZ effectively elicited DNA damage to enable chemotherapy. Consequently, the vMSTI effectively triggered apoptosis, suppressing GBM growth under the guidance of fluorescence imaging. Our nanosystems offered a promising strategy for imaging-guided combination therapy for GBM.
Collapse
Affiliation(s)
- Feng Wei
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen University, School of Medicine, Xiamen, Fujian 361004, China
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Yanling Lin
- Department of Occupational and Environmental Health, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Rujun Pan
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fujian 350001, China
| | - Yilong Peng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
- Department of Neurosurgery, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, China
| | - E Chen
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen University, School of Medicine, Xiamen, Fujian 361004, China
| | - Junlong Kang
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen University, School of Medicine, Xiamen, Fujian 361004, China
| | - Jiang Zhu
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen University, School of Medicine, Xiamen, Fujian 361004, China
| | - Jiayin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Baofang Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Wenwen Shen
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen University, School of Medicine, Xiamen, Fujian 361004, China
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Jinyan Lin
- Department of Occupational and Environmental Health, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hongzhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Xinhua Tian
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen University, School of Medicine, Xiamen, Fujian 361004, China
| |
Collapse
|
25
|
Wang X, Gao Y, Wang T, Wang Z, Hang H, Li S, Feng F. Photoactivated hydride therapy under hypoxia beyond ROS. Chem Sci 2024; 15:20292-20302. [PMID: 39568933 PMCID: PMC11575613 DOI: 10.1039/d4sc06576j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
As compared to oxidative phototherapy, studies on reactive reductive species-participating photodynamic therapy (PDT) are rare. Porphyrins are typical photosensitizers restricted by the oxygen level, but efficacy and selectivity are always incompatible in PDT. Herein, we report that phlorins are ideal hydride (H-) donors and explore a water-soluble triphenylphosphonium-modified zinc-coordinated porphyrin (mitoZnPor) for in situ photogeneration of zinc-cored phlorin (mitoZnPhl). Driven by 1,4-dihydronicotinamide adenine dinucleotide (NADH), the mitoZnPor/mitoZnPhl couple can reduce electron acceptors like iron heme and ubiquinone that play key roles in the mitochondrial electron transport chain (Mito-ETC). Under hypoxia, mitoZnPor showed excellent cancer-selectivity and a highly efficient in vitro PDT effect with IC50 at nanomolar levels and potent tumor growth inhibition in a 4T1 tumor-xenografted mouse model with good biosafety, which underlines the great potential of Mito-ETC targeted non-classical PDT via a H--transfer mechanism beyond reactive oxygen species (ROS) in precision cancer phototherapy using NADH as a biomarker and original electron donor.
Collapse
Affiliation(s)
- Xia Wang
- MOE Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Yijian Gao
- College of Pharmaceutical Sciences, Soochow University Suzhou 215123 China
| | - Ting Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Zhaobin Wang
- MOE Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University Nanjing 210023 China
| | - He Hang
- MOE Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University Suzhou 215123 China
| | - Fude Feng
- MOE Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University Nanjing 210023 China
| |
Collapse
|
26
|
Chen Z, Liu C, Zheng W, Fang Y, Zhang H, Luo J, Li J, Qiu Y, Peng J, Xia Y, Miao C, Luo Q. Deciphering the Role of SLFN12: A Novel Biomarker for Predicting Immunotherapy Outcomes in Glioma Patients Through Artificial Intelligence. J Cell Mol Med 2024; 28:e70317. [PMID: 39740094 DOI: 10.1111/jcmm.70317] [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: 11/13/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/02/2025] Open
Abstract
Gliomas are the most prevalent form of primary brain tumours. Recently, targeting the PD-1 pathway with immunotherapies has shown promise as a novel glioma treatment. However, not all patients experience long-lasting benefits, underscoring the necessity to discover reliable biomarkers for predicting treatment outcomes. This study applied a range of advanced artificial intelligence methods to identify a new biomarker linked to the effectiveness of anti-PD-1 immunotherapy in glioma patients. Through an extensive analysis of single-cell RNA sequencing and bulk transcriptomic data from over 3000 patients, the gene SLFN12 emerged as a significant and independent predictor of immunotherapy response. Our results indicate that elevated SLFN12 expression is associated with worse overall survival across various glioma cohorts. Notably, we found that patients with high SLFN12 levels are less likely to respond favourably to anti-PD-1 treatment, positioning SLFN12 as a clinically valuable biomarker for personalised treatment decisions. Functional studies revealed that SLFN12 is involved in key immune-related pathways, shedding light on its potential role in altering the tumour microenvironment and impacting immunotherapy outcomes. Additional laboratory experiments confirmed the role of SLFN12 in promoting glioma cell proliferation, migration and macrophage recruitment. In summary, this study identifies SLFN12 as a novel biomarker for predicting immunotherapy response in glioma patients, offering new insights for precision immunotherapy approaches.
Collapse
Affiliation(s)
- Zigui Chen
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China
| | - Wei Zheng
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China
| | - Yuhua Fang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, Guangxi, China
| | - He Zhang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, Guangxi, China
| | - Jiawei Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, Guangxi, China
| | - Jiale Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Yingqi Qiu
- Department of Clinical Research Center, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Changfeng Miao
- Department of Neurosurgery Second Branche, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Qisheng Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Engineering Research Center for Biomaterials in Bone and Joint Degenerative Diseases, Baise, Guangxi, China
| |
Collapse
|
27
|
Yi L, Zhang Z, Zhou W, Zhang Y, Hu Y, Guo A, Cheng Y, Qian Z, Zhou P, Gao X. BRD4 Degradation Enhanced Glioma Sensitivity to Temozolomide by Regulating Notch1 via Glu-Modified GSH-Responsive Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409753. [PMID: 39544152 DOI: 10.1002/advs.202409753] [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: 08/19/2024] [Revised: 10/20/2024] [Indexed: 11/17/2024]
Abstract
Temozolomide (TMZ) serves as the principal chemotherapeutic agent for glioma; nonetheless, its therapeutic efficacy is compromised by the rapid emergence of drug resistance, the inadequate targeting of glioma cells, and significant systemic toxicity. ARV-825 may play a role in modulating drug resistance by degrading the BRD4 protein, thereby exerting anti-glioma effects. Therefore, to surmount TMZ resistance and achieve efficient and specific drug delivery, a dual-targeted glutathione (GSH)-responsive nanoparticle system (T+A@Glu-NP) is designed and synthesized for the co-delivery of ARV-825 and TMZ. As anticipated, T+A@Glu-NPs significantly enhanced penetration of the blood-brain barrier (BBB), facilitated drug uptake by glioma cells, and exhibited efficient accumulation in brain tissue. Additionally, T+A@Glu-NPs exhibited augmented efficacy against glioma both in vitro and in vivo through the induction of apoptosis, inhibition of proliferation, and cell cycle arrest. Furthermore, mechanistic exploration revealed that T+A@Glu-NPs degraded the BRD4 protein, leading to the downregulation of Notch1 gene transcription and the inhibition of the Notch1 signaling pathway, thereby augmenting the therapeutic efficacy of glioma chemotherapy. Taken together, the findings suggest that T+A@Glu-NPs represents a novel and promising therapeutic strategy for glioma chemotherapy.
Collapse
Affiliation(s)
- Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhenyu Zhang
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjie Zhou
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China
| | - Yunchu Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yuzhu Hu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Anjie Guo
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Peizhi Zhou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| |
Collapse
|
28
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
29
|
Liang Y, Feng L, Zheng Y, Gao Y, Shi R, Zhang Z, Ying X, Zeng Y. Targeted Liposomal Co-Delivery Dopamine with 3-n-Butylphthalide for Effective Against Parkinson's Disease in Mice Model. Int J Nanomedicine 2024; 19:12851-12870. [PMID: 39640048 PMCID: PMC11618862 DOI: 10.2147/ijn.s483595] [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: 06/24/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Parkinson's disease (PD) is a multifactor-induced neurodegenerative disease with high incidence in the elderly population. We found for the first time that the combination of dopamine (DA) and 3-n-butylphthalide (NBP) has great potential for the synergistic treatment of PD. To further improve the therapeutic performance of the drugs, a brain-targeting liposomal co-delivery system encapsulating NBP and DA ((NBP+DA)-Lips-RVG29) was designed using a rabies virus polypeptide with 29 amino acids (RVG29) as the targeting ligand. Methods The synergistic neuroprotective effects of NBP and DA were assessed in 6-OHDA-induced PC12 cells. Then, (NBP+DA)-Lips-RVG29 loading with NBP and DA at an optimal ratio was prepared using the thin-film hydration and sonication method. The physicochemical and biological characterization of (NBP+DA)-Lips-RVG29 were systemically investigated, and the therapeutic efficiency and underlying mechanisms of (NBP+DA)-Lips-RVG29 were also explored in vitro and in vivo. Finally, the safety of (NBP+DA)-Lips-RVG29 was evaluated. Results The synergistic effects of NBP and DA peaked at 1:1 (NBP/DA, mol/mol). The functionalized liposomes showed significantly higher uptake efficiency and blood-brain barrier (BBB) penetration efficiency in vitro. After systemic administration, (NBP+DA)-Lips-RVG29 prolonged the blood circulation of drugs, enhanced BBB penetration and increased drug accumulation in the striatum, substantia nigra and hippocampus. Moreover, (NBP+DA)-Lips-RVG29 showed excellent neuroprotective effects in a cellular PD model of PC12 cells and improved therapeutic efficacy in a PD mouse model. Furthermore, the safety evaluation of (NBP+DA)-Lips-RVG29 revealed no systemic toxicity. Conclusion NBP and DA exhibited the synergistic anti-PD effects. The RVG29-modified liposomes encapsulating NBP and DA contributed to the accumulation of drugs in the brain lesions area of PD and further improved treatment efficacy. Therefore, (NBP+DA)-Lips-RVG29 represents a promising strategy for the treatment of PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Liang
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Liping Feng
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Yue Zheng
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Yunzhen Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Rongying Shi
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xue Ying
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Yingchun Zeng
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| |
Collapse
|
30
|
Hegde MM, Palkar P, Mutalik SP, Mutalik S, Goda JS, Rao BSS. Enhancing glioblastoma cytotoxicity through encapsulating O6-benzylguanine and temozolomide in PEGylated liposomal nanocarrier: an in vitro study. 3 Biotech 2024; 14:275. [PMID: 39450422 PMCID: PMC11499494 DOI: 10.1007/s13205-024-04123-2] [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: 07/12/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Glioblastoma (GBM) (grade IV glioma) is the most fatal brain tumor, with a median survival of just 14 months despite current treatments. Temozolomide (TMZ), an alkylating agent used with radiation, faces challenges such as systemic toxicity, poor absorption, and drug resistance. To enhance TMZ effectiveness, we developed poly(ethylene glycol) (PEG) liposomes co-loaded with TMZ and O6-benzylguanine (O6-BG) for targeted glioma therapy. These liposomes, prepared using the thin-layer hydration method, had an average size of 146.33 ± 6.75 nm and a negative zeta potential (-49.6 ± 3.1 mV). Drug release was slower at physiological pH, with 66.84 ± 4.62% of TMZ and 69.70 ± 2.88% of O6-BG released, indicating stability at physiological conditions. The liposomes showed significantly higher cellular uptake (p < 0.05) than the free dye. The dual drug-loaded liposomes exhibited superior cytotoxicity against U87 glioma cells, with a lower IC50 value (3.99µg/mL) than the free drug combination, demonstrating enhanced anticancer efficacy. The liposome formulation induced higher apoptosis (19.42 ± 3.5%) by causing sub-G0/G1 cell cycle arrest. The novelty of our study lies in co-encapsulating TMZ and O6-BG within PEGylated liposomes, effectively overcoming drug resistance and improving targeted delivery for glioma treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04123-2.
Collapse
Affiliation(s)
- Manasa Manjunath Hegde
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Pranoti Palkar
- Advance Centre for Treatment Research and Education in Cancer, Tata Memorial Centre & Homi Bhaba National Institute, Navi Mumbai, India
| | - Sadhana P. Mutalik
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Srinivas Mutalik
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Jayant Sastri Goda
- Advance Centre for Treatment Research and Education in Cancer, Tata Memorial Centre & Homi Bhaba National Institute, Navi Mumbai, India
- Department of Radiation Oncology, Advanced Centre for Treatment Research Education in Cancer, Tata Memorial Centre & Homi Bhaba National Institute, Navi Mumbai, India
| | - B. S. Satish Rao
- Manipal School of Life Sciences & Director-Research, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
31
|
Chen C, Xie B, Sun S, Guo S, Yang Z, Yang L, Zhang Y, Li SA, Sun W, Wang Z, Qin S, Ji Y. Bovine serum albumin-bound homologous targeted nanoparticles for breast cancer combinatorial therapy. Int J Biol Macromol 2024; 281:136090. [PMID: 39343270 DOI: 10.1016/j.ijbiomac.2024.136090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/01/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Breast cancer, the most common lethal cancer among women, is characterized by the uncontrolled growth of abnormal cells in breast tissue. Therefore, synergistic anticancer strategies are essential, particularly for maximizing drug delivery to tumor sites. Herein, bovine serum albumin (BSA)-bound nanoparticles encapsulating the photosensitizer chlorin e6 (Ce6) (BC) with a CuO2 core (BC/CuO2 NPs) were developed for cuproptosis-promoted cancer photodynamic therapy (PDT). The cancer cell membrane (CC) was then coated onto the surfaces to produce BC/CuO2@CC NPs for breast cancer combinatorial therapy. BSA serves dual functions as both a stabilizing scaffold for metal peroxide nanomaterials and a molecular connector for Ce6. The BC/CuO2@CC NPs group showed the stronger internalization capability than the other groups. BC/CuO2@CC NPs could effectively induce the greatest degree of apoptosis and death ratio (81.77 %), and lead to cuproptosis by downregulating the expression of DLAT, LIAS, and FDX1 protein in vitro. The intra-tumoral accumulation of BC/CuO2@CC NPs was 8.3- and 7.7-fold higher than that of Ce6 and BC/CuO2@CC NPs at 24 h postinjection, respectively. Moreover, synergistic efficacy of cuproptosis and PDT not only inhibited tumor growth but also prevented liver metastases. Thus, our work may be a novel approach for efficient and targeted cancer treatment.
Collapse
Affiliation(s)
- Caili Chen
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, China; Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453000, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Bohong Xie
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453000, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Shuming Sun
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453000, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Sheng Guo
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453000, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Zishan Yang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453000, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Liuzhong Yang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Yana Zhang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Shu-Ang Li
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wei Sun
- Department of Burn and Repair Reconstruction, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zihao Wang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453000, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Shuang Qin
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, China.
| | - Yinghua Ji
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, China.
| |
Collapse
|
32
|
Yang T, Zhang N, Liu Y, Yang R, Wei Z, Liu F, Song D, Wang L, Wei J, Li Y, Shen D, Liang G. Nanoplatelets modified with RVG for targeted delivery of miR-375 and temozolomide to enhance gliomas therapy. J Nanobiotechnology 2024; 22:623. [PMID: 39402578 PMCID: PMC11476726 DOI: 10.1186/s12951-024-02895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Gliomas are one of the most frequent primary brain tumors and pose a serious threat to people's lives and health. Platelets, a crucial component of blood, have been applied as drug delivery carriers for disease diagnosis and treatment. In this study, we designed engineered nanoplatelets for targeted delivery of therapeutic miR-375 and temozolomide (TMZ, a first-line glioma treatment agent) to enhance glioma therapy. Nanoplatelets were prepared through mild ultrasound, TMZ and miR-375 were co-loaded through ultrasound and electrostatic interactions, respectively, to combine chemotherapy with gene therapy against glioma. To improve the blood brain barrier (BBB) crossing efficiency and glioma targeting ability, the nanoplatelets were modified with central nervous system-specific rabies viral glycoprotein peptide (RVG) through thiol-maleimide click reaction. The RVG modified nanoplatelets co-loaded TMZ and miR-375 (NR/TMZ/miR-375) not only inherited the good stability and remarkable biocompatibility of platelets, but also promoted the cellular uptake and penetration of glioma tissues, and effectively induced cell apoptosis to enhance the therapeutic effect of drugs. In vivo studies showed that NR/TMZ/miR-375 significantly increased the circulation time of TMZ, and exhibited superior combined antitumor effects. In summary, this multifunctional 'natural' nanodrug delivery system provides a potent, scalable, and safety approach for platelet-based combined cancer chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Tingting Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
- Zhumadian Cental Hospital, Zhumadian, 463000, China
| | - Nan Zhang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Yuanyuan Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Ruyue Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Zhaoyi Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Futai Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Dan Song
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Longwei Wang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Jiangyan Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Yuanpei Li
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Deliang Shen
- Key Laboratory of Cardiac Injury and Repair of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China.
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China.
| |
Collapse
|
33
|
Ismail M, Wang Y, Li Y, Liu J, Zheng M, Zou Y. Stimuli-Responsive Polymeric Nanocarriers Accelerate On-Demand Drug Release to Combat Glioblastoma. Biomacromolecules 2024; 25:6250-6282. [PMID: 39259212 DOI: 10.1021/acs.biomac.4c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with a poor prognosis and limited treatment options. Drug delivery by stimuli-responsive nanocarriers holds great promise for improving the treatment modalities of GBM. At the beginning of the review, we highlighted the stimuli-active polymeric nanocarriers carrying therapies that potentially boost anti-GBM responses by employing endogenous (pH, redox, hypoxia, enzyme) or exogenous stimuli (light, ultrasonic, magnetic, temperature, radiation) as triggers for controlled drug release mainly via hydrophobic/hydrophilic transition, degradability, ionizability, etc. Modifying these nanocarriers with target ligands further enhanced their capacity to traverse the blood-brain barrier (BBB) and preferentially accumulate in glioma cells. These unique features potentially lead to more effective brain cancer treatment with minimal adverse reactions and superior therapeutic outcomes. Finally, the review summarizes the existing difficulties and future prospects in stimuli-responsive nanocarriers for treating GBM. Overall, this review offers theoretical guidelines for developing intelligent and versatile stimuli-responsive nanocarriers to facilitate precise drug delivery and treatment of GBM in clinical settings.
Collapse
Affiliation(s)
- Muhammad Ismail
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan Zou
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
34
|
Li S, Li X, Wang N, Zhang C, Sang Y, Sun Y, Xia X, Zheng M. Brain targeted biomimetic siRNA nanoparticles for drug resistance glioblastoma treatment. J Control Release 2024; 376:67-78. [PMID: 39368706 DOI: 10.1016/j.jconrel.2024.10.004] [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: 06/04/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma multiforme (GBM), the most aggressive intracranial neoplasm, remains incurable at present, primarily due to drug resistance, which significantly contributes to elevated recurrence rates and dismal prognosis. Signal transducer and activator of transcription 3 (STAT3) is a critical gene closely associated with GBM drug resistance and the progression of GBM stem cells (GSCs), making it a promising therapeutic target. In this study, we developed cancer cell membrane-cloaked biomimetic nanoparticles to deliver STAT3 siRNA to reverse drug resistance in homologous GBM. These biomimetic nanoparticles leverage homotypic targeting, rapid endosome escape, and fast siRNA release, leading to efficient in vitro STAT3 knockdown in both temozolomide-resistant U251-TR cells and X01 GSCs. Moreover, benefited from the membrane functionalization, significant prolonged blood circulation, improved blood brain barrier (BBB) penetration and GBM tumor accumulation are achieved by these siRNA biomimetic nanoparticles. Importantly, these nanoparticles effectively inhibit tumor proliferation, significantly extending median survival time in orthotopic U251-TR (43.5 d versus 20 d for PBS control) and X01 GSC-bearing mouse xenografts (52 d versus 19.5 d for PBS control). Altogether, this biomimetic siRNA platform offers a promising strategy for gene therapy targeting drug-resistant GBM.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Xiaozhe Li
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ningyang Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Chen Zhang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yujing Sang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yajing Sun
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
35
|
Dai Y, Min Y, Zhou L, Cheng L, Ni H, Yang Y, Zhou W. Brain-targeting redox-sensitive micelles for codelivery of TMZ and β-lapachone for glioblastoma therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 61:102772. [PMID: 38960367 DOI: 10.1016/j.nano.2024.102772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Glioblastoma (GBM) is a central nervous system cancer with high incidence and poor survival rates. Enhancing drug penetration of the blood-brain barrier (BBB) and targeting efficacy is crucial for improving treatment outcomes. In this study, we developed a redox-sensitive targeted nano-delivery system (HCA-A2) for temozolomide (TMZ) and β-lapachone (β-Lapa). This system used hyaluronic acid (HA) as the hydrophilic group, arachidonic acid (CA) as the hydrophobic group, and angiopep-2 (A2) as the targeting group. Control systems included non-redox sensitive (HDA-A2) and non-targeting (HCA) versions. In vitro, HCA-TMZ-Lapa micelles released 100 % of their payload in a simulated tumor microenvironment within 24 h, compared to 43.97 % under normal conditions. HCA-A2 micelles, internalized via clathrin-mediated endocytosis, showed stronger cytotoxicity and better BBB penetration and cellular uptake than controls. In vivo studies demonstrated superior tumor growth inhibition with HCA-A2 micelles, indicating their potential for GBM treatment.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Yuanping Min
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Lu Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Longyang Cheng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hongbin Ni
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Wendi Zhou
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| |
Collapse
|
36
|
Wu X, Tang N, Zhao Q, Xiong J. Spatiotemporal evolutionary process of osteosarcoma immune microenvironment remodeling and C1QBP-driven drug resistance deciphered through single-cell multi-dimensional analysis. Bioeng Transl Med 2024; 9:e10654. [PMID: 39553438 PMCID: PMC11561849 DOI: 10.1002/btm2.10654] [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: 10/15/2023] [Revised: 01/24/2024] [Accepted: 02/12/2024] [Indexed: 11/19/2024] Open
Abstract
The tumor immune microenvironment has manifested a crucial correlation with tumor occurrence, development, recurrence, and metastasis. To explore the mechanisms intrinsic to osteosarcoma (OS) initiation and progression, this study synthesizes multiple single-cell RNA sequencing data sets, constructing a comprehensive landscape of the OS microenvironment. Integrating single-cell RNA sequencing with bulk RNA sequencing data has enabled the identification of a significant correlation between heightened expression of the fatty acid metabolism-associated gene (C1QBP) and patient survival in OS. C1QBP not only amplifies the proliferation, migration, invasion, and anti-apoptotic properties of OS but also instigates cisplatin resistance. Subsequent investigations suggest that C1QBP potentially promotes macrophage polarization from monocytes/macrophages toward M2 and M3 phenotypes. Consequently, C1QBP may emerge as a novel target for modulating OS progression and resistance therapy.
Collapse
Affiliation(s)
- Xin Wu
- Department of Spine Surgery, Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ning Tang
- Department of Orthopaedics, Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Qiangqiang Zhao
- Department of HematologyLiuzhou People's Hospital affiliated to Guangxi Medical UniversityLiuzhouGuangxiChina
- Department of HematologyThe Qinghai Provincial People's HospitalXiningQinghaiChina
| | - Jianbin Xiong
- Department of OrthopaedicsLiuzhou Municipal Liutie Central HospitalLiuzhouGuangxiChina
| |
Collapse
|
37
|
Hang Z, Zhou L, Bian X, Liu G, Cui F, Du H, Wen Y. Potential application of aptamers combined with DNA nanoflowers in neurodegenerative diseases. Ageing Res Rev 2024; 100:102444. [PMID: 39084322 DOI: 10.1016/j.arr.2024.102444] [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: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
The efficacy of neurotherapeutic drugs hinges on their ability to traverse the blood-brain barrier and access the brain, which is crucial for treating or alleviating neurodegenerative diseases (NDs). Given the absence of definitive cures for NDs, early diagnosis and intervention become paramount in impeding disease progression. However, conventional therapeutic drugs and existing diagnostic approaches must meet clinical demands. Consequently, there is a pressing need to advance drug delivery systems and early diagnostic methods tailored for NDs. Certain aptamers endowed with specific functionalities find widespread utility in the targeted therapy and diagnosis of NDs. DNA nanoflowers (DNFs), distinctive flower-shaped DNA nanomaterials, are intricately self-assembled through rolling ring amplification (RCA) of circular DNA templates. Notably, imbuing DNFs with diverse functionalities becomes seamlessly achievable by integrating aptamer sequences with specific functions into RCA templates, resulting in a novel nanomaterial, aptamer-bound DNFs (ADNFs) that amalgamates the advantageous features of both components. This article delves into the characteristics and applications of aptamers and DNFs, exploring the potential or application of ADNFs in drug-targeted delivery, direct treatment, early diagnosis, etc. The objective is to offer prospective ideas for the clinical treatment or diagnosis of NDs, thereby contributing to the ongoing efforts in this critical field.
Collapse
Affiliation(s)
- Zhongci Hang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| | - Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guotao Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Fenghe Cui
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangdingdong Road, Zhifu District, Yantai, Shandong 264000, China.
| | - Hongwu Du
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
38
|
Ruan W, Xu S, An Y, Cui Y, Liu Y, Wang Y, Ismail M, Liu Y, Zheng M. Brain-Targeted Cas12a Ribonucleoprotein Nanocapsules Enable Synergetic Gene Co-Editing Leading to Potent Inhibition of Orthotopic Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402178. [PMID: 38943253 PMCID: PMC11434219 DOI: 10.1002/advs.202402178] [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: 02/29/2024] [Revised: 04/26/2024] [Indexed: 07/01/2024]
Abstract
Gene-editing technology shows great potential in glioblastoma (GBM) therapy. Due to the complexity of GBM pathogenesis, a single gene-editing-based therapy is unlikely to be successful; therefore, a multi-gene knockout strategy is preferred for effective GBM inhibition. Here, a non-invasive, biodegradable brain-targeted CRISPR/Cas12a nanocapsule is used that simultaneously targeted dual oncogenes, EGFR and PLK1, for effective GBM therapy. This cargo nanoencapsulation technology enables the CRISPR/Cas12a system to achieve extended blood half-life, efficient blood-brain barrier (BBB) penetration, active tumor targeting, and selective release. In U87MG cells, the combinatorial gene editing system resulted in 61% and 33% knockout of EGFR and PLK1, respectively. Following systemic administration, the CRISPR/Cas12a system demonstrated promising brain tumor accumulation that led to extensive EGFR and PLK1 gene editing in both U87MG and patient-derived GSC xenograft mouse models with negligible off-target gene editing detected through NGS. Additionally, CRISPR/Cas12a nanocapsules that concurrently targeted the EGFR and PLK1 oncogenes showed superior tumor growth suppression and significantly improved the median survival time relative to nanocapsules containing single oncogene knockouts, signifying the potency of the multi-oncogene targeting strategy. The findings indicate that utilization of the CRISPR/Cas12a combinatorial gene editing technique presents a practical option for gene therapy in GBM.
Collapse
Affiliation(s)
- Weimin Ruan
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Sen Xu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yang An
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Science, Henan University, Kaifeng, Henan, 475004, China
| | - Yingxue Cui
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yang Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Muhammad Ismail
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yong Liu
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, 270 Xuanyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Meng Zheng
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| |
Collapse
|
39
|
Li C, Niu C, Chen L, Yu B, Luo F, Qie J, Yang H, Qian J, Chu Y. Classical Monocytes Shuttling for Precise Delivery of Nanotherapeutics to Glioblastoma. Adv Healthc Mater 2024:e2400925. [PMID: 39212635 DOI: 10.1002/adhm.202400925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor for which current therapies have limited efficacy. Immunosuppression and difficulties in accessing tumors with therapeutic agents are major obstacles for GBM treatments. Classical monocytes (CMs) possess the strongest infiltration among myeloid cells recruited into tumors during tumorigenesis. In this study, CMs are utilized to deliver the small-molecule CUDC-907 encapsulated in nanoparticles (907-NPs@CMs) for GBM therapy. Hitchhiking on CMs enables more 907-NPs to successfully penetrate the blood-brain barrier (BBB) and reach the interior of tumors. Results demonstrate that 907-NPs@CMs significantly improve the survival rates by suppressing tumor growth and reversing the immunosuppression of tumor microenvironment (TME). Furthermore, the high delivery efficiency of CMs reduces the amount of CUDC-907 required for treatments, reducing the physiological toxicity and off-target effects caused by high doses. 907-NPs@CMs is a safe and versatile therapeutic system that provides a platform for targeted drug delivery to tumors and the ability to treat GBM through a combination of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Congwen Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Congyi Niu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lin Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingbo Qie
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hui Yang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiawen Qian
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Science, Fudan University, Shanghai, 200030, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, 200032, China
| |
Collapse
|
40
|
Zhao C, Zhu X, Yang H, Tan J, Gong R, Mei C, Cai X, Su Z, Kong F. Lactoferrin/CD133 antibody conjugated nanostructured lipid carriers for dual targeting of blood-brain-barrier and glioblastoma stem cells. Biomed Mater 2024; 19:055041. [PMID: 39134023 DOI: 10.1088/1748-605x/ad6e47] [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: 04/09/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
The main reasons for the difficulty in curing and high recurrence rate of glioblastoma multiforme (GBM) include: 1. The difficulty of chemotherapy drugs in penetrating the blood-brain barrier (BBB) to target tumor cells; 2. The presence of glioma stem cells (GSCs) leading to chemotherapy resistance. Therefore, breaking through the limitations of the BBB and overcoming the drug resistance caused by GSCs are the main strategies to address this problem. This study presents our results on the development of lactoferrin (Lf)/CD133 antibody conjugated nanostructured lipid carriers (Lf/CD133-NLCS) for simultaneously targeting BBB and GSCs. Temozolomide (TMZ) loaded Lf/CD133-NLCS (Lf/CD133-NLCS-TMZ) exhibited high-efficiencyin vitroanti-tumor effects toward malignant glioma cells (U87-MG) and GSCs, while demonstrating no significant toxicity to normal cells at concentrations lower than 200 μg ml-1. The results of thein vitrotargeting GBM study revealed a notably higher cellular uptake of Lf/CD133-NLCS-TMZ in U87-MG cells and GSCs in comparison to Lf/CD133 unconjugated counterpart (NLCS-TMZ). In addition, increased BBB permeability were confirmed for Lf/CD133-NLCS-TMZ compared to NLCS-TMZ bothin vitroandin vivo. Taking together, Lf/CD133-NLCS-TMZ show great potential for dual targeting of BBB and GSCs, as well as GBM therapy based on this strategy.
Collapse
Affiliation(s)
- Changhong Zhao
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, Hubei 435003, People's Republic of China
- Lantian Pharmaceuticals Co., Ltd, Huangshi, Hubei 435000, People's Republic of China
| | - Xinshu Zhu
- School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an 223005, People's Republic of China
| | - Huili Yang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, Hubei 435003, People's Republic of China
| | - Jianmei Tan
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, Hubei 435003, People's Republic of China
| | - Ruohan Gong
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, Hubei 435003, People's Republic of China
| | - Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, People's Republic of China
| | - Xiang Cai
- Lantian Pharmaceuticals Co., Ltd, Huangshi, Hubei 435000, People's Republic of China
| | - Zhenhong Su
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, Hubei 435003, People's Republic of China
| | - Fei Kong
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|
41
|
Miao K, Xia X, Zou Y, Shi B. Small Scale, Big Impact: Nanotechnology-Enhanced Drug Delivery for Brain Diseases. Mol Pharm 2024; 21:3777-3799. [PMID: 39038108 DOI: 10.1021/acs.molpharmaceut.4c00387] [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] [Indexed: 07/24/2024]
Abstract
Central nervous system (CNS) diseases, ranging from brain cancers to neurodegenerative disorders like dementia and acute conditions such as strokes, have been heavily burdening healthcare and have a direct impact on patient quality of life. A significant hurdle in developing effective treatments is the presence of the blood-brain barrier (BBB), a highly selective barrier that prevents most drugs from reaching the brain. The tight junctions and adherens junctions between the endothelial cells and various receptors expressed on the cells make the BBB form a nonfenestrated and highly selective structure that is crucial for brain homeostasis but complicates drug delivery. Nanotechnology offers a novel pathway to circumvent this barrier, with nanoparticles engineered to ferry drugs across the BBB, protect drugs from degradation, and deliver medications to the designated area. After years of development, nanoparticle optimization, including sizes, shapes, surface modifications, and targeting ligands, can enable nanomaterials tailored to specific brain drug delivery settings. Moreover, smart nano drug delivery systems can respond to endogenous and exogenous stimuli that control subsequent drug release. Here, we address the importance of the BBB in brain disease treatment, summarize different delivery routes for brain drug delivery, discuss the cutting-edge nanotechnology-based strategies for brain drug delivery, and further offer valuable insights into how these innovations in nanoparticle technology could revolutionize the treatment of CNS diseases, presenting a promising avenue for noninvasive, targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kaiting Miao
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xue Xia
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yan Zou
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bingyang Shi
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
42
|
Cai X, Cai D, Wang X, Zhang D, Qiu L, Diao Z, Liu Y, Sun J, Cui D, Liu Y, Yin T. Manganese self-boosting hollow nanoenzymes with glutathione depletion for synergistic cancer chemo-chemodynamic therapy. Biomater Sci 2024; 12:3622-3632. [PMID: 38855985 DOI: 10.1039/d4bm00386a] [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: 06/11/2024]
Abstract
Chemodynamic therapy (CDT) has outstanding potential as a combination therapy to treat cancer. However, the effectiveness of CDT in the treatment of solid tumors is limited by the overexpression of glutathione (GSH) in the tumor microenvironment (TME). GSH overexpression diminishes oxidative stress and attenuates chemotherapeutic drug-induced apoptosis in cancer cells. To counter these effects, a synergistic CDT/chemotherapy cancer treatment, involving the use of a multifunctional bioreactor of hollow manganese dioxide (HMnO2) loaded with cisplatin (CDDP), was developed. Metal nanoenzymes that can auto-degrade to produce Mn2+ exhibit Fenton-like, GSH-peroxidase-like activity, which effectively depletes GSH in the TME to attenuate the tumor antioxidant capacity. In an acidic environment, Mn2+ catalyzed the decomposition of intra-tumor H2O2 into highly toxic ·OH as a CDT. HMnO2 with large pores, pore volume, and surface area exhibited a high CDDP loading capacity (>0.6 g-1). Treatment with CDDP-loaded HMnO2 increased the intratumor Pt-DNA content, leading to the up-regulation of γ-H2Aχ and an increase in tumor tissue damage. The decreased GSH triggered by HMnO2 auto-degradation protected Mn2+-generated ·OH from scavenging to amplify oxidative stress and enhance the efficacy of CDT. The nanoenzymes with encapsulated chemotherapeutic agents deplete GSH and remodel the TME. Thus, tumor CDT/chemotherapy combination therapy is an effective therapeutic strategy.
Collapse
Affiliation(s)
- Xinyi Cai
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| | - Deng Cai
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, P. R. China
| | - Xiaozhen Wang
- Respiratory department, Tsinghua University Yuquan Hospital, Beijing, 100040, P. R. China
| | - Dou Zhang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| | - Long Qiu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| | - Zhenying Diao
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| | - Yong Liu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| | - Jianbo Sun
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| | - Daxiang Cui
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, P. R. China.
| | - Yanlei Liu
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China.
| | - Ting Yin
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, P. R. China.
| |
Collapse
|
43
|
Sarkar S, Greer J, Marlowe NJ, Medvid A, Ivan ME, Kolishetti N, Dhar S. Stemness, invasion, and immunosuppression modulation in recurrent glioblastoma using nanotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1976. [PMID: 39091260 DOI: 10.1002/wnan.1976] [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: 08/07/2023] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 08/04/2024]
Abstract
The recurrent nature of glioblastoma negatively impacts conventional treatment strategies leading to a growing need for nanomedicine. Nanotherapeutics, an approach designed to deliver drugs to specific sites, is experiencing rapid growth and gaining immense popularity. Having potential in reaching the hard-to-reach disease sites, this field has the potential to show high efficacy in combatting glioblastoma progression. The presence of glioblastoma stem cells (GSCs) is a major factor behind the poor prognosis of glioblastoma multiforme (GBM). Stemness potential, heterogeneity, and self-renewal capacity, are some of the properties that make GSCs invade across the distant regions of the brain. Despite advances in medical technology and MRI-guided maximal surgical resection, not all GSCs residing in the brain can be removed, leading to recurrent disease. The aggressiveness of GBM is often correlated with immune suppression, where the T-cells are unable to infiltrate the cancer initiating GSCs. Standard of care therapies, including surgery and chemotherapy in combination with radiation therapy, have failed to tackle all the challenges of the GSCs, making it increasingly important for researchers to develop strategies to tackle their growth and proliferation and reduce the recurrence of GBM. Here, we will focus on the advancements in the field of nanomedicine that has the potential to show positive impact in managing glioblastoma tumor microenvironment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Shrita Sarkar
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Jessica Greer
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Nathaniel J Marlowe
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Angeline Medvid
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Michael E Ivan
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Institute of Neuroimmune Pharmacology, Miami, Florida International University, Florida, USA
| | - Shanta Dhar
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Chemistry, University of Miami, Coral Gables, Florida, USA
| |
Collapse
|
44
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
45
|
Susa F, Arpicco S, Pirri CF, Limongi T. An Overview on the Physiopathology of the Blood-Brain Barrier and the Lipid-Based Nanocarriers for Central Nervous System Delivery. Pharmaceutics 2024; 16:849. [PMID: 39065547 PMCID: PMC11279990 DOI: 10.3390/pharmaceutics16070849] [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: 05/11/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood-brain barrier (BBB), blood-retinal barrier, blood-nerve barrier, blood-lymph barrier, and blood-cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
Collapse
Affiliation(s)
- Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| | - Candido Fabrizio Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Tania Limongi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| |
Collapse
|
46
|
Zhang L, Teng F, Xin H, Xu W, Wu W, Yao C, Wang Z. A Big Prospect for Hydrogel Nano-System in Glioma. Int J Nanomedicine 2024; 19:5605-5618. [PMID: 38882547 PMCID: PMC11179662 DOI: 10.2147/ijn.s470315] [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: 04/17/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024] Open
Abstract
Patients diagnosed with glioma typically face a limited life expectancy (around 15 months on average), a bleak prognosis, and a high likelihood of recurrence. As such, glioma is recognized as a significant form of malignancy. Presently, the treatment options for glioma include traditional approaches such as surgery, chemotherapy, and radiotherapy. Regrettably, the efficacy of these treatments has been less than optimal. Nevertheless, a promising development in glioma treatment lies in the use of hydrogel nano-systems as sophisticated delivery systems. These nano-systems have demonstrated exceptional therapeutic effects in the treatment of glioma by various responsive ways, including temperature-response, pH-response, liposome-response, ROS-response, light-response, and enzyme-response. This study seeks to provide a comprehensive summary of both the therapeutic application of hydrogel nano-systems in managing glioma and the underlying immune action mechanisms.
Collapse
Affiliation(s)
- Lu Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- The State Key Laboratory of Power Transmission Equipment and System Security and New Technology, College of Electrical Engineering, Chongqing University, Chongqing, 400044, People’s Republic of China
| | - Fei Teng
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Huajie Xin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Wei Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Wei Wu
- College of Biological Engineering, Chongqing University, Chongqing, 400030, People’s Republic of China
| | - Chenguo Yao
- The State Key Laboratory of Power Transmission Equipment and System Security and New Technology, College of Electrical Engineering, Chongqing University, Chongqing, 400044, People’s Republic of China
| | - Zhiqiang Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| |
Collapse
|
47
|
Wang Q, Li H, Wu T, Yu B, Cong H, Shen Y. Nanodrugs based on co-delivery strategies to combat cisplatin resistance. J Control Release 2024; 370:14-42. [PMID: 38615892 DOI: 10.1016/j.jconrel.2024.04.020] [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: 12/30/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Qiubo Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Taixia Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bio-nanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
48
|
Ye J, Fan Y, She Y, Shi J, Yang Y, Yuan X, Li R, Han J, Liu L, Kang Y, Ji X. Biomimetic Self-Propelled Asymmetric Nanomotors for Cascade-Targeted Treatment of Neurological Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310211. [PMID: 38460166 PMCID: PMC11165487 DOI: 10.1002/advs.202310211] [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: 01/16/2024] [Revised: 02/19/2024] [Indexed: 03/11/2024]
Abstract
The precise targeted delivery of therapeutic agents to deep regions of the brain is crucial for the effective treatment of various neurological diseases. However, achieving this goal is challenging due to the presence of the blood‒brain barrier (BBB) and the complex anatomy of the brain. Here, a biomimetic self-propelled nanomotor with cascade targeting capacity is developed for the treatment of neurological inflammatory diseases. The self-propelled nanomotors are designed with biomimetic asymmetric structures with a mesoporous SiO2 head and multiple MnO2 tentacles. Macrophage membrane biomimetic modification endows nanomotors with inflammatory targeting and BBB penetration abilities The MnO2 agents catalyze the degradation of H2O2 into O2, not only by reducing brain inflammation but also by providing the driving force for deep brain penetration. Additionally, the mesoporous SiO2 head is loaded with curcumin, which actively regulates macrophage polarization from the M1 to the M2 phenotype. All in vitro cell, organoid model, and in vivo animal experiments confirmed the effectiveness of the biomimetic self-propelled nanomotors in precise targeting, deep brain penetration, anti-inflammatory, and nervous system function maintenance. Therefore, this study introduces a platform of biomimetic self-propelled nanomotors with inflammation targeting ability and active deep penetration for the treatment of neurological inflammation diseases.
Collapse
Affiliation(s)
- Jiamin Ye
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yaoguang She
- Department of General Surgerythe First Medical CenterChinese People's Liberation Army General HospitalBeijing100853China
| | - Jiacheng Shi
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yiwen Yang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Xue Yuan
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Ruiyan Li
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Jingwen Han
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Luntao Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin100730China
| | - Yong Kang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
- Medical CollegeLinyi UniversityLinyi276000China
| |
Collapse
|
49
|
Thomas RG, Kim S, Tran TAT, Kim YH, Nagareddy R, Jung TY, Kim SK, Jeong YY. Magnet-Guided Temozolomide and Ferucarbotran Loaded Nanoparticles to Enhance Therapeutic Efficacy in Glioma Model. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:939. [PMID: 38869565 PMCID: PMC11173836 DOI: 10.3390/nano14110939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024]
Abstract
Background. The aim of the study was to synthesize liposomal nanoparticles loaded with temozolomide and ferucarbotran (LTF) and to evaluate the theranostic effect of LTF in the glioma model. Methods. We synthesized an LTF that could pass through the Blood Brain Barrier (BBB) and localize in brain tumor tissue with the help of magnet guidance. We examined the chemical characteristics. Cellular uptake and cytotoxicity studies were conducted in vitro. A biodistribution and tumor inhibition study was conduted using an in vivo glioma model. Results. The particle size and surface charge of LTF show 108 nm and -38 mV, respectively. Additionally, the presence of ferucarbotran significantly increased the contrast agent effect of glioma compared to the control group in MR imaging. Magnet-guided LTF significantly reduced the tumor size compared to control and other groups. Furthermore, compared to the control group, our results demonstrate a significant inhibition in brain tumor size and an increase in lifespan. Conclusions. These findings suggest that the LTF with magnetic guidance represents a novel approach to address current obstacles, such as BBB penetration of nanoparticles and drug resistance. Magnet-guided LTF is able to enhance therapeutic efficacy in mouse brain glioma.
Collapse
Affiliation(s)
- Reju George Thomas
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
| | - Subin Kim
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea;
| | - Thi-Anh-Thuy Tran
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea
- Brain Tumor Research Laboratory, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea (T.-Y.J.)
| | - Young Hee Kim
- Brain Tumor Research Laboratory, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea (T.-Y.J.)
| | - Raveena Nagareddy
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
| | - Tae-Young Jung
- Brain Tumor Research Laboratory, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea (T.-Y.J.)
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea
| | - Seul Kee Kim
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
- Department of Radiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
- Department of Radiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
50
|
Chen ZA, Wu CH, Wu SH, Huang CY, Mou CY, Wei KC, Yen Y, Chien IT, Runa S, Chen YP, Chen P. Receptor Ligand-Free Mesoporous Silica Nanoparticles: A Streamlined Strategy for Targeted Drug Delivery across the Blood-Brain Barrier. ACS NANO 2024; 18:12716-12736. [PMID: 38718220 PMCID: PMC11112986 DOI: 10.1021/acsnano.3c08993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024]
Abstract
Mesoporous silica nanoparticles (MSNs) represent a promising avenue for targeted brain tumor therapy. However, the blood-brain barrier (BBB) often presents a formidable obstacle to efficient drug delivery. This study introduces a ligand-free PEGylated MSN variant (RMSN25-PEG-TA) with a 25 nm size and a slight positive charge, which exhibits superior BBB penetration. Utilizing two-photon imaging, RMSN25-PEG-TA particles remained in circulation for over 24 h, indicating significant traversal beyond the cerebrovascular realm. Importantly, DOX@RMSN25-PEG-TA, our MSN loaded with doxorubicin (DOX), harnessed the enhanced permeability and retention (EPR) effect to achieve a 6-fold increase in brain accumulation compared to free DOX. In vivo evaluations confirmed the potent inhibition of orthotopic glioma growth by DOX@RMSN25-PEG-TA, extending survival rates in spontaneous brain tumor models by over 28% and offering an improved biosafety profile. Advanced LC-MS/MS investigations unveiled a distinctive protein corona surrounding RMSN25-PEG-TA, suggesting proteins such as apolipoprotein E and albumin could play pivotal roles in enabling its BBB penetration. Our results underscore the potential of ligand-free MSNs in treating brain tumors, which supports the development of future drug-nanoparticle design paradigms.
Collapse
Affiliation(s)
- Zih-An Chen
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Research
Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Cheng-Hsun Wu
- Nano
Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Si-Han Wu
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiung-Yin Huang
- Neuroscience
Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chung-Yuan Mou
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Nano
Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Kuo-Chen Wei
- Neuroscience
Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- School
of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, New Taipei Municipal TuCheng
Hospital, New Taipei City 23652, Taiwan
| | - Yun Yen
- Center
for Cancer Translational Research, Tzu Chi
University, Hualien 970374, Taiwan
- Cancer
Center, Taipei Municipal WanFang Hospital, Taipei 116081, Taiwan
| | - I-Ting Chien
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Sabiha Runa
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- SRS Medical Communications,
LLC, Cleveland, Ohio 44124, United States
| | - Yi-Ping Chen
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Peilin Chen
- Research
Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|