1
|
Wang C, Li R, Dong C, Shi S. A locally-adapted nanoreactor for autophagy inhibition-enhanced cascade starvation-chemodynamic therapy. J Colloid Interface Sci 2025; 695:137820. [PMID: 40354733 DOI: 10.1016/j.jcis.2025.137820] [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: 03/06/2025] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Cutting off the energy supply of tumors and disrupting their redox homeostasis have emerged as two promising therapeutic strategies in oncology. However, a persistent challenge lies in overcoming tumor self-protection mechanisms while minimizing toxic side effects. To overcome this limitation, a locally-adapted nanoreactor (MCGH) is developed for autophagy inhibition-enhanced cascade starvation-chemodynamic therapy, which not only selectively enhances tumor treatment by inhibiting autophagy, but also minimizes damage to normal tissues through tissue-specific cascade catalytic reactions. Specifically, glucose oxidase (GOx)-loaded MCGH catalyzes intratumoral glucose conversion into hydrogen peroxide (H2O2), thereby simultaneously inducing starvation therapy while generating substrates for chemodynamic therapy (CDT). When exposed to the tumor microenvironment, the low pH and high levels of glutathione (GSH) trigger the degradation of MCGH to release Mn2+ ions. These ions subsequently catalyze Fenton-like reaction that transform H2O2 into highly toxic hydroxyl radicals (OH) for CDT. Additionally, concurrent autophagy inhibition by MCGH blocks OH clearance and prevents nutrient replenishment, thereby amplifying the therapeutic efficacy of this cascade strategy. Importantly, MCGH exhibits catalase (CAT)-like activity in normal tissues, effectively detoxifying H2O2 through its decomposition. By engineering a single material platform to execute different catalytic cascades, this work provides a new perspective for precision nanomedicine design.
Collapse
Affiliation(s)
- Chunhui Wang
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai 200092, PR China
| | - Ruihao Li
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai 200092, PR China
| | - Chunyan Dong
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai 200092, PR China.
| | - Shuo Shi
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai 200092, PR China.
| |
Collapse
|
2
|
Wang C, Ye P, Chen M, Li R, Wen Y, Wang Y, Tong X, Dong C, Shi S. Reducing the availability of endogenous copper and glucose for cascade starvation therapy and chemodynamic therapy. Mater Today Bio 2025; 32:101702. [PMID: 40230642 PMCID: PMC11995123 DOI: 10.1016/j.mtbio.2025.101702] [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: 01/14/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
The rapid growth of tumors relies heavily on a continuous supply of essential nutrients, including glucose and copper. Disrupting the nutrient supply to tumors has become an increasingly focal point in tumor therapy. However, solely blocking the energy supply typically only hinders further tumor growth and may not effectively eliminate existing tumor cells. Herein, a multifunctional cascade nanoreactor (HPP/TPEN@GC) endowed with N, N, N', N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine (TPEN, a copper chelator) and glucose oxidase (GOx) is designed to disrupt both glycolysis and mitochondrial metabolism, which further induce cascade chemodynamic therapy (CDT). HPP/TPEN@GC can react with endogenous copper and glucose, thereby reducing their availability. The absence of copper prevents proper assembly and function of mitochondrial complex IV (CIV), hindering mitochondrial metabolism; the lack of glucose cuts off glycolysis and leads to a tumor specific starvation. Meanwhile, the reactions catalyzed by HPP/TPEN@GC contribute to the generation of Fenton-like catalysts and hydrogen peroxide (H2O2), which can further react to produce highly toxic hydroxyl radical (·OH) for CDT. Taken together, the multifunctional cascade nanoreactor reduces the availability of endogenous copper and glucose, and further takes advantage of them to generate ·OH for cascade starvation-chemodynamic therapy. Collectively, this work represents a distinctive therapeutic paradigm to harness endogenous copper and glucose, which should inspire further studies to take full advantage of endogenous nutrients to combat various diseases, including tumors.
Collapse
Affiliation(s)
| | | | - Mengyao Chen
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| | - Ruihao Li
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| | - Yixuan Wen
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| | - Yu Wang
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| | - Xiaohan Tong
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| | - Chunyan Dong
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| | - Shuo Shi
- School of Chemical Science and Engineering, Breast Cancer Center, Shanghai East Hospital, Tongji University, Shanghai, 200092, PR China
| |
Collapse
|
3
|
Gao X, Tang X, Tu Z, Yu J, Bao Y, Long G, Sheu WC, Wu H, Liu J, Zhou J. Tertiary amine modification enables triterpene nanoparticles to target the mitochondria and treat glioblastoma via pyroptosis induction. Biomaterials 2025; 317:123035. [PMID: 39731842 PMCID: PMC11827167 DOI: 10.1016/j.biomaterials.2024.123035] [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: 05/01/2024] [Revised: 12/03/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Glioblastoma (GBM), the most common primary brain tumor, lacks effective treatments. Emerging evidence suggests mitochondria as a promising therapeutic target, albeit successfully targeting represents a major challenge. Recently, we discovered a group of triterpenes that can self-assemble into nanoparticles (NPs) for cancer treatment. However, unmodified triterpene NPs lack affinity for mitochondria. In this study, using oleanolic acid (OA) as an example, we demonstrated that tertiary amine modification enabled triterpene NPs to selectively target the mitochondria through interaction with translocase of outer mitochondrial membrane 70 (TOM70) leading to effective killing of GBM cells via pyroptosis. We showed that the NPs could be engineered for preferentially penetrating brain tumors through surface conjugation of iRGD, and treatment with the resulting NPs significantly prolonged the survival of tumor-bearing mice. We found that the efficacy could be further improved by encapsulating lonidamine, a mitochondrial hexokinase inhibitor. Furthermore, the observed mitochondria targeting effect through tertiary amine modification could be extended to other triterpenes, including lupeol and glycyrrhetinic acid. Collectively, this study reveals a novel strategy for targeting the mitochondria through tertiary amine modification of triterpenes, offering a promising avenue for the effective treatment of GBM.
Collapse
Affiliation(s)
- Xingchun Gao
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Xiangjun Tang
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Zewei Tu
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Jiang Yu
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Youmei Bao
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Gretchen Long
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Wendy C Sheu
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Haoan Wu
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Jia Liu
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT, 06511, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA.
| |
Collapse
|
4
|
Yang M, Hu Y, Hao X, Chen Q, Cao Y, Ran H, Zhang W. Ultrasound-actuated ion homeostasis perturbator for oxidative damage-augmented Ca 2+ interference therapy and combined immunotherapy. Mater Today Bio 2025; 32:101666. [PMID: 40166376 PMCID: PMC11957807 DOI: 10.1016/j.mtbio.2025.101666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Calcium ion therapy has shown promise for cancer treatment, but its efficacy is limited by the cellular calcium buffering mechanism. Herein, an ion homeostasis disruptor (PCCa) was synthesized using an in situ mineralization method. The surface of the porphyrin-metal-organic framework PCN was coated with calcium carbonate (CaCO3), aimed at causing Ca2+ overload and disrupting the self-defense mechanism during ion imbalance. Upon internalization into tumor cells, PCCa undergoes lysosomal acidification-induced CaCO3 decomposition, leading to instantaneous Ca2+ overload. Simultaneously, under ultrasonic irradiation, the meso-tetra-(4-carboxyphenyl)porphine (TCPP) within the ion homeostasis disruptor generates reactive oxygen species (ROS), which impairs cellular calcium buffering capacity and amplifies cell damage caused by calcium overload. In addition, PCCa could also induce immunogenic cell death, release tumor-associated antigens (TAA), and act as an adjuvant, thereby promoting dendritic cell maturation and enhancing the antitumor activity of CD8+ T cells. In mouse models, PCCa not only led to significant regression of subcutaneous mammary tumors but also demonstrated substantial anti-metastatic effects. In summary, the proposed ultrasound-actuated Ca2+ interference strategy is promising to deactivate the ion homeostasis maintenance system, contributing to the attainment of splendid tumor treatment outcome with reliable biosafety, which may provide useful insights in cancer therapy.
Collapse
Affiliation(s)
- Mi Yang
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| | - Yaqin Hu
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| | - Xiuxiu Hao
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| | - Qiaoqi Chen
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| | - Yang Cao
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| | - Haitao Ran
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| | - Wei Zhang
- Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University. Chongqing, 400010, China
| |
Collapse
|
5
|
Liu G, Liu J, Li S, Zhang Y, He R. Exosome-Mediated Chemoresistance in Cancers: Mechanisms, Therapeutic Implications, and Future Directions. Biomolecules 2025; 15:685. [PMID: 40427578 PMCID: PMC12108986 DOI: 10.3390/biom15050685] [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: 03/15/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Chemotherapy resistance represents a formidable obstacle in oncological therapeutics, substantially compromising the efficacy of adjuvant chemotherapy regimens and contributing to unfavorable clinical prognoses. Emerging evidence has elucidated the pivotal involvement of exosomes in the dissemination of chemoresistance phenotypes among tumor cells and within the tumor microenvironment. This review delineates two distinct intra-tumoral resistance mechanisms orchestrated by exosomes: (1) the exosome-mediated sequestration of chemotherapeutic agents coupled with enhanced drug efflux in neoplastic cells, and (2) the horizontal transfer of chemoresistance to drug-sensitive cells through the delivery of bioactive molecular cargo, thereby facilitating the propagation of resistance phenotypes across the tumor population. Furthermore, the review covers current in vivo experimental data focusing on targeted interventions against specific genetic elements and exosomal secretion pathways, demonstrating their potential in mitigating chemotherapy resistance. Additionally, the therapeutic potential of inhibiting exosome-mediated transporter transfer strategy is particularly examined as a promising strategy to overcome tumor resistance mechanisms.
Collapse
Affiliation(s)
| | | | | | - Yumiao Zhang
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| | - Ren He
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| |
Collapse
|
6
|
Zhou H, Zhu C, Li Y, Zhao F, Feng Q, Liu S, Jia S, Ji J, Ye L, Zhai G, Yang X. Exosome/liposome hybrid nanovesicles for enhanced phototherapy and boosted anti-tumor immunity against melanoma. Eur J Med Chem 2025; 289:117485. [PMID: 40081104 DOI: 10.1016/j.ejmech.2025.117485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Although phototherapy shows great potential as a safe ablative modality for treatment of cutaneous melanoma, there remain serious flaws restricting its therapeutic outcomes, such as cellular resistance against apoptosis, tumor hypoxia, rewritten cellular metabolism and abnormal angiogenesis. To cope with these challenges, this work combines hemin and IR780 (phototherapy agent) and designs an orchestrated liposome/macrophage-derived exosome hybrid delivery system (named IHEL) for tumor-specific delivery of these two drugs and synchronous tumor microenvironment (TME) reprogramming. As the experimental data suggest, by triggering iron overload and up-regulating HMOX-1, hemin drives a shift from an apoptosis-dominant anti-cancer mode to a combined ferroptosis/apoptosis mode of IR780 treatment, which helps to avoid apoptosis resistance. Also, the catalase-like activity of hemin strengthens PDT effect by alleviating hypoxia. In addition to the above-mentioned enhanced direct cell-killing effect, IHEL also provokes anti-cancer immunity by triggering immunogenic cell death (ICD), intervening glycometabolism and polarizing tumor-associated macrophages (TAMs) in TME to M1-type. This work strongly demonstrated the rationality of IR780/hemin combination and delicately designed immunostimulatory nanocarriers for their tumor-specific delivery, providing both theoretical foundation and practical strategies for advanced anti-cancer phototherapy.
Collapse
Affiliation(s)
- He Zhou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Chuanxiu Zhu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yingchao Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Feiyan Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qixiang Feng
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shangui Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shuangxu Jia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
7
|
Wang T, Du M, Chen Z. Sonosensitizers for Sonodynamic Therapy: Current Progress and Future Perspectives. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:727-734. [PMID: 39909788 DOI: 10.1016/j.ultrasmedbio.2025.01.011] [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: 05/22/2024] [Revised: 01/08/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025]
Abstract
Sonodynamic therapy (SDT) is a novel non-invasive treatment method that combines low-intensity ultrasound and sonosensitizers. Compared with photodynamic therapy, SDT has the advantages of deeper tissue penetration, higher accuracy and fewer adverse reactions. Sonosensitizers are essential for the efficacy of SDT. Sonosensitizers have the advantages of clear structure, easy monitoring, evaluation of drug metabolism and clinical transformation, etc. Notably, biochemical techniques can be used in the field of sonosensitizers and SDT to overcome inherent barriers and achieve sustainable innovation. This article first summarizes the molecular mechanism of SDT, focusing on organic sonosensitizers, inorganic nano-sonosensitizers and multi-functional drug delivery systems with targeting, penetration and imaging functions after a series of modifications. This review provides ideas and references for the design of sonosensitizers and SDT and promotes their future transformation into clinical applications.
Collapse
Affiliation(s)
- Ting Wang
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China; Department of Medical Imaging, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China.
| |
Collapse
|
8
|
Lee J, Lee S, Jo G, Hwang E, Lee J, Han J, Jung HS. A Novel BODIPY-Zn Complex as Innovative Sonosensitizer for Enhanced Sonodynamic Therapy. Molecules 2025; 30:1587. [PMID: 40286210 PMCID: PMC11990734 DOI: 10.3390/molecules30071587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Ultrasound (US)-based sonodynamic therapy (SDT) presents a promising and secure approach to treating cancer with the advantage of enhanced tissue penetration, making it a favorable option compared with traditional photodynamic therapy. However, the search for innovative sonosensitizers that exhibit both high sonosensitizing efficacy and good biocompatibility poses a formidable challenge. In this research, we prepared a novel BODIPY-Zn complex (BSS-Zn) incorporating a hydrophilic short polyethylene glycol unit and explored its feasibility as a sonosensitizer. BSS-Zn exhibited enhanced reactive oxygen species (ROS) generation behavior upon US irradiation, outperforming a control sensitizer, BSS (an analog lacking the Zn complex), and a commercial sonosensitizer, ZnPc (currently undergoing clinical testing), with regard to sonosensitizing properties. The enhanced effect of BSS-Zn was attributed to increased levels of ROS, such as hydroxyl radicals, singlet oxygen, and superoxide, mediated by US exposure in aqueous media. The SDT effect of BSS-Zn on MDA-MB-231 cells was verified by confirming the intracellular types of generated ROS and evaluating the cytotoxicity to MDA-MB-231 cancer cells. This pioneering study highlights the potential of BSS-Zn as an innovative sonosensitizer for SDT. Our findings provide valuable guidance for the design of efficient sonosensitizers.
Collapse
Affiliation(s)
- Jungmin Lee
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
| | - Soeun Lee
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
| | - Gihoon Jo
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
| | - Eunbin Hwang
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Junhyoung Lee
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Jiyou Han
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
| | - Hyo Sung Jung
- Department of Biomedical & Chemical Sciences, Hyupsung University, Hwasung-si 18330, Republic of Korea; (J.L.); (S.L.); (G.J.); (E.H.); (J.L.)
| |
Collapse
|
9
|
Nguyen VN, Nguyen Cao TG, Jeong H, Truong Hoang Q, Pham BTT, Bang J, Koh CW, Kang JH, Lee JH, Wu X, Rhee WJ, Ko YT, Swamy KMK, Park S, Park J, Shim MS, Yoon J. Tumor-Targeted Exosome-Based Heavy Atom-Free Nanosensitizers With Long-Lived Excited States for Safe and Effective Sono-Photodynamic Therapy of Solid Tumors. Adv Healthc Mater 2025:e2500927. [PMID: 40165690 DOI: 10.1002/adhm.202500927] [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: 02/19/2025] [Revised: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Theranostic nanosensitizers with combined near-infrared (NIR) fluorescence imaging and sono-photodynamic effects have great potential for use in the personalized treatment of deep-seated tumors. However, developing effective nanosensitizers for NIR fluorescence image-guided sono-photodynamic therapy remains a considerable challenge, including the low generation efficacy of reactive oxygen species (ROS), poor photostability, and the absence of cancer specificity. Herein, a novel heavy atom-free nanosensitizer is developed, which exhibits intense NIR fluorescence, high ROS generation efficiency, and improved aqueous stability. By conjugating a bulky and electron-rich group, 4-(1,2,2-triphenylvinyl)-1,1'-biphenyl (TPE), to the IR820 backbone, the resulting IR820 bearing TPE (IR820-TPE) effectively generates ROS via type I and II photochemical mechanisms under 808 nm laser irradiation. Moreover, TPE conjugation considerably increases the sono-photodynamic performance of IR820. To improve the intracellular delivery and tumor-targeting ability of IR820-TPE, biotin-conjugated exosome (B-Exo) is used as a natural nanocarrier. In vitro studies demonstrate the outstanding therapeutic performance of IR820-TPE-loaded B-Exo (IR820-TPE@B-Exo) in synergistic sono-photodynamic cancer therapy. In vivo studies reveal that IR820-TPE@B-Exo shows enhanced tumor accumulation, strong fluorescence signals, and effective sono-photodynamic therapeutic activity with high biosafety. This work demonstrates that IR820-TPE@B-Exo is a promising sono-phototheranostic agent for safe and targeted cancer therapy and NIR fluorescence imaging.
Collapse
Affiliation(s)
- Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Department of Chemistry, School of Chemistry and Life Sciences, Hanoi University of Science and Technology, Ha Noi, 100000, Vietnam
| | - Thuy Giang Nguyen Cao
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Hyunsun Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Quan Truong Hoang
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Binh T T Pham
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Jieun Bang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Chang Woo Koh
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hee Kang
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Jeong Hyun Lee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Xiaofeng Wu
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon, 22012, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - K M K Swamy
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sungnam Park
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - JaeHong Park
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| |
Collapse
|
10
|
Li M, Liu Q, Xie S, Weng D, He J, Yang X, Liu Y, You J, Liao J, Wang P, Lu X, Zhao J. Transformable Tumor Microenvironment-Responsive Oxygen Vacancy-Rich MnO 2@Hydroxyapatite Nanospheres for Highly Efficient Cancer Sonodynamic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414162. [PMID: 39960349 PMCID: PMC11984894 DOI: 10.1002/advs.202414162] [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: 11/02/2024] [Revised: 12/31/2024] [Indexed: 04/12/2025]
Abstract
Despite the promise of sonodynamic therapy (SDT)-mediated immunotherapy, the anticancer efficacy of current sonosensitizers is greatly limited by the immunosuppressive tumor microenvironment (TME) and their inability to selectively respond to it. Herein, oxygen vacancy-rich MnO2@hydroxyapatite (Ca10(PO4)6(OH)2) core-shell nanospheres (denoted as Ov-MO@CPO) as an advanced TME-responsive sonosensitizer for sonodynamic immunotherapy is demonstrated. The Ov-MO@CPO maintains its structural integrity under neutral conditions but dissolves the pH-sensitive hydroxyapatite shell under acidic TME to release active oxygen vacancy-rich MnO2 core, which reinvigorates H2O2 consumption and hypoxia alleviation due to its catalase-like activity. Furthermore, the introduced oxygen vacancies optimize the electronic structure of Ov-MO@CPO, with active electronic states near the Fermi level and higher d-band center. It results in accelerated electron-hole pair separation and lower catalytic energy barriers to boost ultrasound (US)-initiated ROS production. These multimodal synergistic effects effectively reverse the immunosuppressive tumor microenvironment, inhibiting tumor growth and metastasis in 4T1 tumor-bearing mice. No evident toxic effects are observed in normal mouse tissues. Additionally, when combined with an immune checkpoint inhibitor, Ov-MO@CPO-mediated SDT further improves the effectiveness of immunotherapy. This work affords a new avenue for developing TME-dependent sonosensitizers for SDT-mediated immunotherapy.
Collapse
Affiliation(s)
- Minxing Li
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Qiyu Liu
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Songzuo Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Desheng Weng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinjun He
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Xinyi Yang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Yuanyuan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinqi You
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinghao Liao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Peng Wang
- Department of Emergency MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Xihong Lu
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Jingjing Zhao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
11
|
Li M, Liu Y, Liu F, Chen Q, Xu L, Cheng Z, Tan Y, Liu Z. Extracellular Vesicle-Based Antitumor Nanomedicines. Adv Healthc Mater 2025; 14:e2403903. [PMID: 39935134 DOI: 10.1002/adhm.202403903] [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: 10/08/2024] [Revised: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Extracellular vesicles (EVs) have emerged as promising bioactive carriers for delivering therapeutic agents, including nucleic acids, proteins, and small-molecule drugs, owing to their excellent physicochemical stability and biocompatibility. However, comprehensive reviews on the various types of EV-based nanomedicines for cancer therapy remain scarce. This review explores the potential of EVs as antitumor nanomedicines. Methods for EV extraction, drug loading, and engineering modifications are systematically examined, and the strengths and limitations of these technical approaches are critically assessed. Additionally, key strategies for developing EV-based antitumor therapies are highlighted. Finally, the opportunities and challenges associated with advancing EVs toward clinical translation are discussed. With the integration of multiple disciplines, robust EV-based therapeutic platforms are expected to be manufactured to provide more personalized and effective solutions for oncology patients.
Collapse
Affiliation(s)
- Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Fei Liu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhongyu Cheng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
12
|
Yuan W, Lu G, Zhao Y, He X, Liao S, Wang Z, Lei X, Xie Z, Yang X, Tang S, Tang G, Deng X. Intranuclear TCA and mitochondrial overload: The nascent sprout of tumors metabolism. Cancer Lett 2025; 613:217527. [PMID: 39909232 DOI: 10.1016/j.canlet.2025.217527] [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: 11/27/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Abnormal glucose metabolism in tumors is a well-known form of metabolic reprogramming in tumor cells, the most representative of which, the Warburg effect, has been widely studied and discussed since its discovery. However, contradictions in a large number of studies and suboptimal efficacy of drugs targeting glycolysis have prompted us to further deepen our understanding of glucose metabolism in tumors. Here, we review recent studies on mitochondrial overload, nuclear localization of metabolizing enzymes, and intranuclear TCA (nTCA) in the context of the anomalies produced by inhibition of the Warburg effect. We provide plausible explanations for many of the contradictory points in the existing studies, including the causes of the Warburg effect. Furthermore, we provide a detailed prospective discussion of these studies in the context of these new findings, providing new ideas for the use of nTCA and mitochondrial overload in tumor therapy.
Collapse
Affiliation(s)
- Weixi Yuan
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guozhong Lu
- 922nd Hospital of Hengyang, 421001, Hunan, China
| | - Yin Zhao
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiang He
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Senyi Liao
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyong Lei
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Xiangnan University, Chenzhou, 423000, China
| | - Zhizhong Xie
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoyan Yang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Xiangnan University, Chenzhou, 423000, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery Systems (2018TP1044), Hunan, 410007, China.
| | - Guotao Tang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
13
|
Zhang Q, Ma RF, Chen SW, Cao K, Wang Y, Xu ZR. Biomineralized and metallized small extracellular vesicles encapsulated in hydrogels for mitochondrial-targeted synergistic tumor therapy. Acta Biomater 2025; 194:428-441. [PMID: 39870149 DOI: 10.1016/j.actbio.2025.01.041] [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: 09/19/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Targeted organelle therapy is a promising therapeutic method for significantly regulating the tumor microenvironment, yet it often lacks effective strategies for leveraging synergistic enhancement effect. Engineered small extracellular vesicles (sEVs) are expected to address this challenge due to their notable advantages in drug delivery, extended circulation time, and intercellular information transmission. Herein, we prepare sEVs with pH and photothermal dual-responsiveness, which are encapsulated with hydrogels for a quadruple-efficient synergistic therapy. M1-phenotype macrophages-derived sEVs, which carry cytokines that inhibit tumor progression, were separately encapsulated with calcium phosphates (CaPs) and Au@Pt nanoparticles (Au@Pt NPs), endowing them with pH and photothermal dual-responsiveness. Subsequently, they were assembled into sEV-Au@Pt NPs/CaPs nanohybrids, and functionalized with mitochondria-targeting peptides. Within tumor cells, mitochondrial targeting enhances Ca2+ accumulation, resulting in mitochondrial homeostasis imbalance. The release of Pt2+ causes nuclear damage and exacerbates mitochondrial dysfunction. Furthermore, under laser irradiation, the sEV-Au@Pt NPs absorb light, generating hyperthermia that promotes the release of Ca2+ and Pt2+ from the hydrogel and cytokines from the sEVs, thereby achieving a quadruple-efficient synergistic therapy. The hydrogel effectively prolongs the retention time of nanohybrids, aiding in the prevention of tumor recurrence. These nanohybrids exhibit favorable mitochondrial targeting ability, with a Pearson's co-localization coefficient of 0.877. In experimental trials, tumor growth was significantly inhibited after only five treatments, with the tumor volume reduced to 0.16-fold that of the control group. This strategy presents a potential tailored platform for engineered sEVs in mitochondrial-targeted therapy and holds great promise for advancing organelle-targeted therapeutic strategies. STATEMENT OF SIGNIFICANCE: Engineering small extracellular vesicles (sEVs) can significantly enhance the synergistic effects of organelle-targeted therapy, thereby improving therapeutic efficacy and reducing side effects. However, their full development is still pending. In this study, we present a promising strategy that involves engineering sEVs with pH and photothermal dual-responsiveness through biomineralization and metallization, enabling quadruple synergistic tumor therapy. Our study demonstrates the remarkable synergistic effects of mitochondrial homeostasis imbalance caused by Ca2+ bursts and nuclear damage due to Pt2+ release. After five treatments, the tumor volume in the experimental group was reduced to 0.16-fold that of the control group. This strategy holds great promise for the design of engineered sEVs as organelle-targeted therapeutic systems.
Collapse
Affiliation(s)
- Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Ruo-Fei Ma
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Si-Wen Chen
- Center for Molecular Science and Engineering, College of Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Ke Cao
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Yue Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Zhang-Run Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
14
|
Ali-Khiavi P, Mohammadi M, Masoumi S, Saffarfar H, Kheradmand R, Mobed A, Hatefnia F. The Therapeutic Potential of Exosome Therapy in Sepsis Management: Addressing Complications and Improving Outcomes". Cell Biochem Biophys 2025; 83:307-326. [PMID: 39363035 DOI: 10.1007/s12013-024-01564-7] [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] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Infection occurs when pathogens penetrate tissues, reproduce, and trigger a host response to both the infectious agents and their toxins. A diverse array of pathogens, including viruses and bacteria, can cause infections. The host's immune system employs several mechanisms to combat these infections, typically involving an innate inflammatory response. Inflammation is a complex biological reaction that can affect various parts of the body and is a key component of the response to harmful stimuli. Sepsis arises when the body's response to infection leads to widespread damage to tissues and organs, potentially resulting in severe outcomes or death. The initial phase of sepsis involves immune system suppression. Early identification and targeted management are crucial for improving sepsis outcomes. Common treatment approaches include antibiotics, intravenous fluids, blood cultures, and monitoring urine output. This study explores the potential of exosome therapy in enhancing the management and alleviation of sepsis symptoms.
Collapse
Affiliation(s)
- Payam Ali-Khiavi
- Medical faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Masoumi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kheradmand
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Faezeh Hatefnia
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Luo X, McAndrews KM, Kalluri R. Natural and Bioengineered Extracellular Vesicles in Diagnosis, Monitoring and Treatment of Cancer. ACS NANO 2025; 19:5871-5896. [PMID: 39869032 PMCID: PMC12002402 DOI: 10.1021/acsnano.4c11630] [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] [Indexed: 01/28/2025]
Abstract
Extracellular vesicles (EVs) are cell derived nanovesicles which are implicated in both physiological and pathological intercellular communication, including the initiation, progression, and metastasis of cancer. The exchange of biomolecules between stromal cells and cancer cells via EVs can provide a window to monitor cancer development in real time for better diagnostic and interventional strategies. In addition, the process of secretion and internalization of EVs by stromal and cancer cells in the tumor microenvironment (TME) can be exploited for delivering therapeutics. EVs have the potential to provide a targeted, biocompatible, and efficient delivery platform for the treatment of cancer and other diseases. Natural as well as engineered EVs as nanomedicine have immense potential for disease intervention. Here, we provide an overview of current knowledge of EVs' function in cancer progression, diagnostic and therapeutic applications for EVs in the cancer setting, as well as current EV engineering strategies.
Collapse
Affiliation(s)
- Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Kathleen M. McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
16
|
Wang J, Wang M, Zeng X, Li Y, Lei L, Chen C, Lin X, Fang P, Guo Y, Jiang X, Wang Y, Chen L, Long J. Targeting membrane contact sites to mediate lipid dynamics: innovative cancer therapies. Cell Commun Signal 2025; 23:89. [PMID: 39955542 PMCID: PMC11830217 DOI: 10.1186/s12964-025-02089-z] [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: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
Membrane contact sites (MCS) are specialized regions where organelles are closely interconnected through membrane structures, facilitating the transfer and exchange of ions, lipids, and other molecules. This proximity enables a synergistic regulation of cellular homeostasis and functions. The formation and maintenance of these contact sites are governed by specific proteins that bring organelle membranes into close apposition, thereby enabling functional crosstalk between cellular compartments. In eukaryotic cells, lipids are primarily synthesized and metabolized within distinct organelles and must be transported through MCS to ensure proper cellular function. Consequently, MCS act as pivotal platforms for lipid synthesis and trafficking, particularly in cancer cells and immune cells within the tumor microenvironment, where dynamic alterations are critical for maintaining lipid homeostasis. This article provides a comprehensive analysis of how these cells exploit membrane contact sites to modulate lipid synthesis, metabolism, and transport, with a specific focus on how MCS-mediated lipid dynamics influence tumor progression. We also examine the differences in MCS and associated molecules across various cancer types, exploring novel therapeutic strategies targeting MCS-related lipid metabolism for the development of anticancer drugs, while also addressing the challenges involved.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China.
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Xueni Zeng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Yanhan Li
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Lingzhi Lei
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Changan Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Xi Lin
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Peiyuan Fang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Yuxuan Guo
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Yian Wang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
| | - Lihong Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China.
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China.
| | - Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
17
|
Guo X, Song J, Liu M, Ou X, Guo Y. The interplay between the tumor microenvironment and tumor-derived small extracellular vesicles in cancer development and therapeutic response. Cancer Biol Ther 2024; 25:2356831. [PMID: 38767879 PMCID: PMC11110713 DOI: 10.1080/15384047.2024.2356831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/14/2024] [Indexed: 05/22/2024] Open
Abstract
The tumor microenvironment (TME) plays an essential role in tumor cell survival by profoundly influencing their proliferation, metastasis, immune evasion, and resistance to treatment. Extracellular vesicles (EVs) are small particles released by all cell types and often reflect the state of their parental cells and modulate other cells' functions through the various cargo they transport. Tumor-derived small EVs (TDSEVs) can transport specific proteins, nucleic acids and lipids tailored to propagate tumor signals and establish a favorable TME. Thus, the TME's biological characteristics can affect TDSEV heterogeneity, and this interplay can amplify tumor growth, dissemination, and resistance to therapy. This review discusses the interplay between TME and TDSEVs based on their biological characteristics and summarizes strategies for targeting cancer cells. Additionally, it reviews the current issues and challenges in this field to offer fresh insights into comprehending tumor development mechanisms and exploring innovative clinical applications.
Collapse
Affiliation(s)
- Xuanyu Guo
- The Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Jiajun Song
- Department of Clinical Laboratory Medicine, the Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Miao Liu
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| | - Xinyi Ou
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| | - Yongcan Guo
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| |
Collapse
|
18
|
Xu J, Liu Y, Wang H, Hao J, Cao Y, Liu Z. Titanium boride nanosheets with photo-enhanced sonodynamic efficiency for glioblastoma treatment. Acta Biomater 2024; 188:344-357. [PMID: 39307260 DOI: 10.1016/j.actbio.2024.09.025] [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: 05/27/2024] [Revised: 08/20/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Sonodynamic therapy (SDT) has garnered significant attention in cancer treatment, however, the low-yield reactive oxygen species (ROS) generation from sonosensitizers remains a major challenge. In this study, titanium boride nanosheets (TiB2 NSs) with photo-enhanced sonodynamic efficiency was fabricated for SDT of glioblastoma (GBM). Compared with commonly-used TiO2 nanoparticles, the obtained TiB2 NSs exhibited much higher ROS generation efficiency under ultrasound (US) irradiation due to their narrower band gap (2.50 eV). Importantly, TiB2 NSs displayed strong localized surface plasmon resonance (LSPR) effect in the second near-infrared (NIR II) window, which facilitated charge transfer rate and improved the separation efficiency of US-triggered electron-hole pairs, leading to photo-enhanced ROS generation efficiency. Furthermore, TiB2 NSs were encapsulated with macrophage cell membranes (CM) and then modified with RGD peptide to construct biomimetic nanoagents (TiB2@CM-RGD) for efficient blood-brain barrier (BBB) penetrating and GBM targeting. After intravenous injection into the tumor-bearing mouse, TiB2@CM-RGD can efficiently cross BBB and accumulate in the tumor sites. The tumor growth was significantly inhibited under simultaneous NIR II laser and US irradiation without causing appreciable long-term toxicity. Our work highlighted a new type of multifunctional titanium-based sonosensitizer with photo-enhanced sonodynamic efficiency for GBM treatment. STATEMENT OF SIGNIFICANCE: Titanium boride nanosheets (TiB2 NSs) with photo-enhanced sonodynamic efficiency was fabricated for SDT of glioblastoma (GBM). The obtained TiB2 NSs displayed strong localized surface plasmon resonance (LSPR) effect in the second near-infrared (NIR II) window, which facilitated charge transfer rate and improved the separation efficiency of US-triggered electron-hole pairs, leading to photo-enhanced ROS generation efficiency. Furthermore, TiB2 NSs were encapsulated with macrophage cell membranes (CM) and then modified with RGD peptide to construct biomimetic nanoagents (TiB2@CM-RGD) for efficient blood-brain barrier (BBB) penetrating and GBM targeting. After intravenous injection into the tumor-bearing mouse, TiB2@CM-RGD can efficiently cross BBB and accumulate in the tumor sites. The tumor growth was significantly inhibited under simultaneous NIR II laser and US irradiation without causing appreciable long-term toxicity.
Collapse
Affiliation(s)
- Jiaqing Xu
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Ying Liu
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Han Wang
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Junxing Hao
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Yu Cao
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China.
| | - Zhihong Liu
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China.
| |
Collapse
|
19
|
Ding H, Zhou C, Li T. Nanomedicines with Versatile GSH-Responsive Linkers for Cancer Theranostics. ACS Biomater Sci Eng 2024; 10:5977-5994. [PMID: 39298132 DOI: 10.1021/acsbiomaterials.4c00897] [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: 10/15/2024]
Abstract
Glutathione (GSH)-responsive nanomedicines have generated significant interest in biochemistry, oncology, and material sciences due to their diverse applications, including chemical and biological sensors, diagnostics, and drug delivery systems. The effectiveness of these smart GSH-responsive nanomedicines depends critically on the choice of GSH-responsive linkers. Despite their crucial role, comprehensive reviews of GSH-responsive linkers are scarce, revealing a gap in the current literature. This review addresses this gap by systematically summarizing various GSH-responsive linkers and exploring their potential applications in cancer treatment. We provide an overview of the mechanisms of action of these linkers and their bioapplications, evaluating their advantages and limitations. The insights presented aim to guide the development of advanced GSH-responsive agents for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Huamin Ding
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai 200125, China
| | - Can Zhou
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai 200125, China
| |
Collapse
|
20
|
Chen Z, Chen L, Lyu TD, Weng S, Xie Y, Jin Y, Wu O, Jones M, Kwan K, Makvnadi P, Li B, Sharopov F, Ma C, Li H, Wu A. Targeted mitochondrial nanomaterials in biomedicine: Advances in therapeutic strategies and imaging modalities. Acta Biomater 2024; 186:1-29. [PMID: 39151665 DOI: 10.1016/j.actbio.2024.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/15/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
Mitochondria, pivotal organelles crucial for energy generation, apoptosis regulation, and cellular metabolism, have spurred remarkable advancements in targeted material development. This review surveys recent breakthroughs in targeted mitochondrial nanomaterials, illuminating their potential in drug delivery, disease management, and biomedical imaging. This review approaches from various application perspectives, introducing the specific applications of mitochondria-targeted materials in cancer treatment, probes and imaging, and diseases treated with mitochondria as a therapeutic target. Addressing extant challenges and elucidating potential therapeutic mechanisms, it also outlines future development trajectories and obstacles. By comprehensively exploring the diverse applications of targeted mitochondrial nanomaterials, this review aims to catalyze innovative treatment modalities and diagnostic approaches in medical research. STATEMENT OF SIGNIFICANCE: This review presents the latest advancements in mitochondria-targeted nanomaterials for biomedical applications, covering diverse fields such as cancer therapy, bioprobes, imaging, and the treatment of various systemic diseases. The novelty and significance of this work lie in its systematic analysis of the intricate relationship between mitochondria and different diseases, as well as the ingenious design strategies employed to harness the therapeutic potential of nanomaterials. By providing crucial insights into the development of mitochondria-targeted nanomaterials and their applications, this review offers a valuable resource for researchers working on innovative treatment modalities and diagnostic approaches. The scientific impact and interest to the readership lie in the identification of promising avenues for future research and the potential for clinical translation of these cutting-edge technologies.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China
| | - Linjie Chen
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Tai Dong Lyu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Shoutao Weng
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Yihao Xie
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Yuxin Jin
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Ouqiang Wu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham B31 2AP, UK
| | - Kenny Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pooyan Makvnadi
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India; Centre of Research Impact and Outreach, Chitkara University, Rajpura, Punjab 140417, India
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College Soochow University, PR China
| | - Farukh Sharopov
- V.I. Nikitin Chemistry Institute of Tajikistan National Academy of Sciences, Dushanbe 734063, Tajikistan
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | - Huaqiong Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China.
| | - Aimin Wu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China.
| |
Collapse
|
21
|
Al-Ani SA, Lee QY, Maheswaran D, Sin YM, Loh JS, Foo JB, Hamzah S, Ng JF, Tan LKS. Potential of Exosomes as Multifunctional Nanocarriers for Targeted Drug Delivery. Mol Biotechnol 2024:10.1007/s12033-024-01268-6. [PMID: 39269575 DOI: 10.1007/s12033-024-01268-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024]
Abstract
Exosomes are small vesicles that form when multivesicular bodies fuse with the plasma membrane and are released into body fluids. They play a vital role in facilitating communication between cells by transferring different biomolecules, including DNA, RNA, proteins, and lipids, over both short and long distances. They also function as vital mediators in both states of health and disease, exerting an impact on several physiological processes. Exosomes have been modified to overcome the limitations of natural exosomes to enhance their potential as carriers for drug delivery systems, and these modifications aim to improve the drug delivery efficiency, enhance tissue and organ targeting, and prolong the circulating half-life of exosomes. This review discussed recent advancements in exosome nanotechnology, as well as the progression and use of exosomes for drug delivery. The potential commercialisation and challenges associated with the use of exosome-based drug delivery systems were also discussed, aiming to motivate the development of exosome-based theranostic nanoplatforms and nanotechnology for improved healthcare treatments.
Collapse
Affiliation(s)
- Safa Ali Al-Ani
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia
| | - Qiao Ying Lee
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia
| | - Danesha Maheswaran
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia
| | - Yuh Miin Sin
- Faculty of Medicine, AIMST University, Jalan Bedong, 08100, Semeling, Kedah Darulaman, Malaysia
| | - Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia
- Digital Health and Medical Advancements Impact Lab, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor campus, 42300 Puncak Alam, Selangor, Malaysia
| | - Sharina Hamzah
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia
- Digital Health and Medical Advancements Impact Lab, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Jeck Fei Ng
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia
- Digital Health and Medical Advancements Impact Lab, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, 47500, Subang Jaya, Selangor, Malaysia.
- Digital Health and Medical Advancements Impact Lab, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
22
|
Liu J, Bai Y, Li Y, Li X, Luo K. Reprogramming the immunosuppressive tumor microenvironment through nanomedicine: an immunometabolism perspective. EBioMedicine 2024; 107:105301. [PMID: 39178747 PMCID: PMC11388279 DOI: 10.1016/j.ebiom.2024.105301] [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: 06/22/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
Increasing evidence indicates that immunotherapy is hindered by a hostile tumor microenvironment (TME) featured with deprivation of critical nutrients and pooling of immunosuppressive metabolites. Tumor cells and immunosuppressive cells outcompete immune effector cells for essential nutrients. Meanwhile, a wide range of tumor cell-derived toxic metabolites exerts negative impacts on anti-tumor immune response, diminishing the efficacy of immunotherapy. Nanomedicine with excellent targetability offers a novel approach to improving cancer immunotherapy via metabolically reprogramming the immunosuppressive TME. Herein, we review recent strategies of enhancing immunotherapeutic effects through rewiring tumor metabolism via nanomedicine. Attention is drawn on immunometabolic tactics for immune cells and stromal cells in the TME via nanomedicine. Additionally, we discuss future directions of developing metabolism-regulating nanomedicine for precise and efficacious cancer immunotherapy.
Collapse
Affiliation(s)
- Jieyu Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinan Bai
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinggang Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoling Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| |
Collapse
|
23
|
Li J, Wang T, Hou X, Li Y, Zhang J, Bai W, Qian H, Sun Z. Extracellular vesicles: opening up a new perspective for the diagnosis and treatment of mitochondrial dysfunction. J Nanobiotechnology 2024; 22:487. [PMID: 39143493 PMCID: PMC11323404 DOI: 10.1186/s12951-024-02750-8] [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: 11/14/2023] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
Mitochondria are crucial organelles responsible for energy generation in eukaryotic cells. Oxidative stress, calcium disorders, and mitochondrial DNA abnormalities can all cause mitochondrial dysfunction. It is now well documented that mitochondrial dysfunction significantly contributes to the pathogenesis of numerous illnesses. Hence, it is vital to investigate innovative treatment methods targeting mitochondrial dysfunction. Extracellular vesicles (EVs) are cell-derived nanovesicles that serve as intercellular messengers and are classified into small EVs (sEVs, < 200 nm) and large EVs (lEVs, > 200 nm) based on their sizes. It is worth noting that certain subtypes of EVs are rich in mitochondrial components (even structurally intact mitochondria) and possess the ability to transfer them or other contents including proteins and nucleic acids to recipient cells to modulate their mitochondrial function. Specifically, EVs can modulate target cell mitochondrial homeostasis as well as mitochondria-controlled apoptosis and ROS generation by delivering relevant substances. In addition, the artificial modification of EVs as delivery carriers for therapeutic goods targeting mitochondria is also a current research hotspot. In this article, we will focus on the ability of EVs to modulate the mitochondrial function of target cells, aiming to offer novel perspectives on therapeutic approaches for diverse conditions linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jiali Li
- Department of Gerontology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Tangrong Wang
- Department of Gerontology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaomei Hou
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450000, China
| | - Yu Li
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiaxin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Wenhuan Bai
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Zixuan Sun
- Department of Gerontology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
24
|
Lin P, Lu Y, Zheng J, Lin Y, Zhao X, Cui L. Strategic disruption of cancer's powerhouse: precise nanomedicine targeting of mitochondrial metabolism. J Nanobiotechnology 2024; 22:318. [PMID: 38849914 PMCID: PMC11162068 DOI: 10.1186/s12951-024-02585-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/26/2024] [Indexed: 06/09/2024] Open
Abstract
Mitochondria occupy a central role in the biology of most eukaryotic cells, functioning as the hub of oxidative metabolism where sugars, fats, and amino acids are ultimately oxidized to release energy. This crucial function fuels a variety of cellular activities. Disruption in mitochondrial metabolism is a common feature in many diseases, including cancer, neurodegenerative conditions and cardiovascular diseases. Targeting tumor cell mitochondrial metabolism with multifunctional nanosystems emerges as a promising strategy for enhancing therapeutic efficacy against cancer. This review comprehensively outlines the pathways of mitochondrial metabolism, emphasizing their critical roles in cellular energy production and metabolic regulation. The associations between aberrant mitochondrial metabolism and the initiation and progression of cancer are highlighted, illustrating how these metabolic disruptions contribute to oncogenesis and tumor sustainability. More importantly, innovative strategies employing nanomedicines to precisely target mitochondrial metabolic pathways in cancer therapy are fully explored. Furthermore, key challenges and future directions in this field are identified and discussed. Collectively, this review provides a comprehensive understanding of the current state and future potential of nanomedicine in targeting mitochondrial metabolism, offering insights for developing more effective cancer therapies.
Collapse
Affiliation(s)
- Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
25
|
Kim HI, Park J, Zhu Y, Wang X, Han Y, Zhang D. Recent advances in extracellular vesicles for therapeutic cargo delivery. Exp Mol Med 2024; 56:836-849. [PMID: 38556545 PMCID: PMC11059217 DOI: 10.1038/s12276-024-01201-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 01/07/2024] [Accepted: 01/15/2024] [Indexed: 04/02/2024] Open
Abstract
Exosomes, which are nanosized vesicles secreted by cells, are attracting increasing interest in the field of biomedical research due to their unique properties, including biocompatibility, cargo loading capacity, and deep tissue penetration. They serve as natural signaling agents in intercellular communication, and their inherent ability to carry proteins, lipids, and nucleic acids endows them with remarkable therapeutic potential. Thus, exosomes can be exploited for diverse therapeutic applications, including chemotherapy, gene therapy, and photothermal therapy. Moreover, their capacity for homotypic targeting and self-recognition provides opportunities for personalized medicine. Despite their advantages as novel therapeutic agents, there are several challenges in optimizing cargo loading efficiency and structural stability and in defining exosome origins. Future research should include the development of large-scale, quality-controllable production methods, the refinement of drug loading strategies, and extensive in vivo studies and clinical trials. Despite the unresolved difficulties, the use of exosomes as efficient, stable, and safe therapeutic delivery systems is an interesting area in biomedical research. Therefore, this review describes exosomes and summarizes cutting-edge studies published in high-impact journals that have introduced novel or enhanced therapeutic effects using exosomes as a drug delivery system in the past 2 years. We provide an informative overview of the current state of exosome research, highlighting the unique properties and therapeutic applications of exosomes. We also emphasize challenges and future directions, underscoring the importance of addressing key issues in the field. With this review, we encourage researchers to further develop exosome-based drugs for clinical application, as such drugs may be among the most promising next-generation therapeutics.
Collapse
Affiliation(s)
- Hyo In Kim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Yohan Han
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Department of Microbiology, Wonkwang University School of Medicine, Iksan, 54538, Republic of Korea.
- Sarcopenia Total Solution Center, Wonkwang University, Iksan, 54538, Republic of Korea.
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
26
|
Huang Z, Liu X, Guo Q, Zhou Y, Shi L, Cai Q, Tang S, Ouyang Q, Zheng J. Extracellular vesicle-mediated communication between CD8 + cytotoxic T cells and tumor cells. Front Immunol 2024; 15:1376962. [PMID: 38562940 PMCID: PMC10982391 DOI: 10.3389/fimmu.2024.1376962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Tumors pose a significant global public health challenge, resulting in numerous fatalities annually. CD8+ T cells play a crucial role in combating tumors; however, their effectiveness is compromised by the tumor itself and the tumor microenvironment (TME), resulting in reduced efficacy of immunotherapy. In this dynamic interplay, extracellular vesicles (EVs) have emerged as pivotal mediators, facilitating direct and indirect communication between tumors and CD8+ T cells. In this article, we provide an overview of how tumor-derived EVs directly regulate CD8+ T cell function by carrying bioactive molecules they carry internally and on their surface. Simultaneously, these EVs modulate the TME, indirectly influencing the efficiency of CD8+ T cell responses. Furthermore, EVs derived from CD8+ T cells exhibit a dual role: they promote tumor immune evasion while also enhancing antitumor activity. Finally, we briefly discuss current prevailing approaches that utilize functionalized EVs based on tumor-targeted therapy and tumor immunotherapy. These approaches aim to present novel perspectives for EV-based tumor treatment strategies, demonstrating potential for advancements in the field.
Collapse
Affiliation(s)
- Zeyu Huang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuehui Liu
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qinghao Guo
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yihang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Linlin Shi
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shupei Tang
- Department of Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
27
|
Wan X, Wang W, Zhou Y, Ma X, Guan M, Liu F, Chen S, Fan JX, Yan GP. Self-Delivery Nanoplatform Based on Amphiphilic Apoptosis Peptide for Precise Mitochondria-Targeting Photothermal Therapy. Mol Pharm 2024; 21:1537-1547. [PMID: 38356224 DOI: 10.1021/acs.molpharmaceut.3c01243] [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: 02/16/2024]
Abstract
Mitochondria-targeting photothermal therapy could significantly enhance the tumor cell killing effect. However, since therapeutic reagents need to overcome a series of physiological obstacles to arrive at mitochondria accurately, precise mitochondria-targeting photothermal therapy still faces great challenges. In this study, we developed a self-delivery nanoplatform that specifically targeted the mitochondria of tumor cells for precise photothermal therapy. Photothermal agent IR780 was encapsulated by amphiphilic apoptotic peptide KLA with mitochondria-targeting ability to form nanomicelle KI by self-assembly through hydrophilic and hydrophobic interactions. Subsequently, negatively charged tumor-targeting polymer HA was coated on the surface of KI through electrostatic interactions, to obtain tumor mitochondria-targeting self-delivery nanoplatform HKI. Through CD44 receptor-mediated recognition, HKI was internalizated by tumor cells and then disassembled in an acidic environment with hyaluronidase in endosomes, resulting in the release of apoptotic peptide KLA and photothermal agent IR780 with mitochondria anchoring capacity, which achieved precise mitochondria guidance and destruction. This tumor mitochondria-targeting self-delivery nanoplatform was able to effectively deliver photothermal agents and apoptotic peptides to tumor cell mitochondria, resulting in precise destruction to mitochondria and enhancing tumor cell inhibition at the subcellular organelle level.
Collapse
Affiliation(s)
- Xin Wan
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Wensong Wang
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yutian Zhou
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Xiaoyu Ma
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Meng Guan
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fan Liu
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Si Chen
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Jin-Xuan Fan
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Guo-Ping Yan
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Material Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
- College of Chemical and Material Engineering, Quzhou University, Quzhou 324000, China
| |
Collapse
|
28
|
Chang M, Zhang L, Wang Z, Chen L, Dong Y, Yang J, Chen Y. Nanomedicine/materdicine-enabled sonocatalytic therapy. Adv Drug Deliv Rev 2024; 205:115160. [PMID: 38110153 DOI: 10.1016/j.addr.2023.115160] [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: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/20/2023]
Abstract
The advent of numerous treatment modalities with desirable therapeutic efficacy has been made possible by the fast development of nanomedicine and materdicine, among which the ultrasound (US)-triggered sonocatalytic process as minimal or non-invasive method has been frequently employed for diagnostic and therapeutic purposes. In comparison to phototherapeutic approaches with inherent penetration depth limitations, sonocatalytic therapy shatters the depth limit of photoactivation and offers numerous remarkable prospects and advantages, including mitigated side effects and appropriate tissue-penetration depth. Nevertheless, the optimization of sonosensitizers and therapies remains a significant issue in terms of precision, intelligence and efficiency. In light of the fact that nanomedicine and materdicine can effectively enhance the theranostic efficiency, we herein aim to furnish a cutting-edge review on the latest progress and development of nanomedicine/materdicine-enabled sonocatalytic therapy. The design methodologies and biological features of nanomedicine/materdicine-based sonosensitizers are initially introduced to reveal the underlying relationship between composition/structure, sonocatalytic function and biological effect, in accompany with a thorough discussion of nanomedicine/materdicine-enabled synergistic therapy. Ultimately, the facing challenges and future perspectives of this intriguing sonocatalytic therapy are highlighted and outlined to promote technological advancements and clinical translation in efficient disease treatment.
Collapse
Affiliation(s)
- Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, PR China
| | - Lu Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Hebei University, Baoding 071000, PR China
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yang Dong
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| | - Jishun Yang
- Naval Medical Center of PLA, Medical Security Center, Shanghai 200052, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
29
|
Liu X, Wang J, Wu Y, Wu M, Song J. Ultrasound activated probe for disease imaging and therapy In-Vivo. Adv Drug Deliv Rev 2024; 205:115158. [PMID: 38104895 DOI: 10.1016/j.addr.2023.115158] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/19/2023]
Abstract
Sonodynamic therapy (SDT) is the use of ultrasound (US) to excite sonosensitizers to produce reactive oxygen species (ROS) to induce tumor cell death, thereby achieving therapeutic purposes. Based on the strong tissue penetration ability of ultrasound, SDT can realize the treatment of deeper tumors, and it is targeted, can be specifically concentrated at the tumor site, and has little impact on surrounding normal tissues. It has broad clinical transformation prospects. Therefore, sonosensitizers are the key to SDT, and the exploration of sonosensitizers with excellent therapeutic performance has received great attention. We reviewed the development of ultrasound-inspired sound sensitizers for imaging and treatment. First, different types of sonosensitizers are introduced, the construction and performance of inorganic, organic and hybrid types of sonosensitizers are evaluated, followed by a review of different image-guided SDT, and finally the key problems and solutions in this field are discussed in detail.
Collapse
Affiliation(s)
- Xing Liu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jimei Wang
- College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, P. R. China
| | - Ying Wu
- College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, P. R. China.
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jibin Song
- College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, P. R. China.
| |
Collapse
|
30
|
Li X, Sun X, Chen H, Chen X, Li Y, Li D, Zhang Z, Chen H, Gao Y. Exploring BODIPY derivatives as sonosensitizers for anticancer sonodynamic therapy. Eur J Med Chem 2024; 264:116035. [PMID: 38101040 DOI: 10.1016/j.ejmech.2023.116035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
Sonodynamic therapy (SDT) is an emerging non-invasive and effective therapeutic modality for cancer treatment bearing benefit of deep tissue-penetration in comparison to photo-inspired therapy. However, exploring novel sonosensitizers with high sonosensitivity and desirable biosafety remains a significant challenge. Although boron dipyrromethene (BODIPY) dyes have been widely used in biomedical filed, no BODIPY-based sonosensitizers have been reported yet. Herein, we synthesized four BODIPY dyes (BDP1-BDP4) and investigated their potential applications in SDT. BDP4 exhibited superb sonosensitivity and high SDT efficiency against cancer cells and tumors in tumor-bearing mice. The types of the generated reactive oxygen species, cavitation effect, and cell apoptosis were investigated to figure out the sonodynamic therapeutic mechanisms of BDP4. This work for the first time demonstrates the potential of BODIPY dyes as novel sonosensitizers for SDT, which may pave an avenue for developing more efficient and safer sonosensitizers in future.
Collapse
Affiliation(s)
- Xudong Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Xianbin Sun
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Hui Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Xinyu Chen
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Yuanming Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Dongmiao Li
- State Key Lab of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Zizhong Zhang
- State Key Lab of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Haijun Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China.
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
31
|
Chen Z, Wang X, Zhao N, Chen H, Guo G. Advancements in pH-responsive nanocarriers: enhancing drug delivery for tumor therapy. Expert Opin Drug Deliv 2023; 20:1623-1642. [PMID: 38059646 DOI: 10.1080/17425247.2023.2292678] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Tumors pose a significant global economic and health burden, with conventional cancer treatments lacking tumor specificity, leading to limited efficiency and undesirable side effects. Targeted tumor therapy is imminent. Tumor cells produce lactate and hydrogen ions (H+) by Warburg effect, forming an acidic tumor microenvironment (TME), which can be employed to design targeted tumor therapy. Recently, progress in nanotechnology has led to the development of pH-responsive nanocarriers, which have gathered significant attention. Under acidic tumor conditions, they exhibit targeted accumulation within tumor sites and controlled release profiles of therapeutic reagents, enabling precise tumor therapy. AREAS COVERED This review comprehensively summarize the principles underlying pH-responsive features, discussing various types of pH-responsive nanocarriers, their advantages, and limitations. Innovative therapeutic drugs are also examined, followed by an exploration of recent advancements in applying various pH-responsive nanocarriers as delivery systems for enhanced tumor therapy. EXPERT OPINIONS pH-responsive nanocarriers have garnered significant attention for their capability to achieve targeted accumulation of therapeutic agents at tumor sites and controlled drug delivery profiles, ultimately increasing the efficiency of tumor eradication. It is anticipated that the employment of pH-responsive nanocarriers will elevate the effectiveness and safety of tumor therapy, contributing to improved overall outcomes.
Collapse
Affiliation(s)
- Zhouyun Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxiao Wang
- West China School of Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Zhao
- School of Pharmacy, Shihezi University, Shihezi, China
| | - Haifeng Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Guo
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|