1
|
Chen Z, Tai Y, Deng C, Sun Y, Chen H, Luo T, Lin J, Chen W, Xu H, Song G, Tang Q, Lu J, Zhu X, Wen S, Wang J. Innovative sarcoma therapy using multifaceted nano-PROTAC-induced EZH2 degradation and immunity enhancement. Biomaterials 2025; 321:123344. [PMID: 40262462 DOI: 10.1016/j.biomaterials.2025.123344] [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: 11/03/2024] [Revised: 04/05/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Sarcomas are highly malignant tumors characterized by their heterogeneity and resistance to conventional therapies, which significantly limit treatment options. EZH2 is highly expressed in sarcomas, but targeting it is difficult. In this study, we uncovered the non-canonical transcriptional mechanisms of EZH2 in sarcoma and highlighted the essential role of EZH2 in regulating YAP1 through non-canonical transcriptional pathways in the progression of sarcoma. Building on this, we developed YM@VBM, a novel and versatile nano-PROTAC (proteolysis-targeting chimera), by integrating a polyphenol-vanadium oxide system with the EZH2 degrader YM281 PROTAC, encapsulated in methoxy polyethylene glycol-NH2 to enhance biocompatibility. To further facilitate targeted drug delivery to tumors, YM@VBM nano-PROTACs were incorporated into microneedle patches. Our engineered YM@VBM exhibited multiple functionalities, including the peroxidase-like activity to generate reactive oxygen species, depletion of glutathione, and photothermal effects, specifically targeting sarcoma characteristics. YM@VBM significantly enhanced targeting efficacy via inducing potent EZH2 degradation. Most importantly, it can also activate anti-tumor immunity via excluding myeloid-derived suppressor cells, maturing dendritic cells, and forming tertiary lymphoid structures. Hence, we reveal that YM@VBM presents a promising treatment strategy for sarcoma, offering a multifaceted approach to combat this challenging malignancy.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Yi Tai
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China; Surgical Department of Colorectal Cancer, Zhejiang Cancer Hospital, 1st BanShan East Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, PR China
| | - Chuangzhong Deng
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Yameng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Hongmin Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Tianqi Luo
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Jiaming Lin
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Weiqing Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Huaiyuan Xu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Guohui Song
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Xiaojun Zhu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China.
| | - Jin Wang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China.
| |
Collapse
|
2
|
Li J, Zhang S, Yu C, Chen X, Zhong W, Shen Y. Arjunolic acid inhibits Wnt3a-mediated macrophage M2 polarization to suppress osteosarcoma progression. GENES & NUTRITION 2025; 20:11. [PMID: 40399780 PMCID: PMC12096606 DOI: 10.1186/s12263-025-00771-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Osteosarcoma (OS) is a bone tumor characterized by a high recurrence rate and poor prognosis. Arjunolic acid (AA), the most abundant triterpene component in Cyclocarya paliurus, is reported to have anti-tumor effects. Its specific role in OS is still unknown, which we aim to investigate in our study. METHODS An OS mouse model was established to investigate the effects of AA. Subsequently, M2 macrophages and M0 macrophages pretreated with AA were co-cultured with OS cells. The impact of AA on OS cell behavior (proliferation, apoptosis, migration, and invasion) was evaluated via EdU staining, flow cytometry, and Transwell assays. Concurrently, the expression of M1- and M2-associated genes (CD86, CD163, IL-6, Arg1) was quantified to assess AA's regulatory role in macrophages within the tumor microenvironment (TME). Knockdown or overexpression of Wnt3a in AA-treated M0 macrophages to determine whether AA modulates Wnt3a-mediated M2 polarization, which was further validated in vivo. RESULTS In vivo, AA inhibited tumor progression in OS mice. Concurrently, AA-treated macrophages inhibited OS cell malignant behavior, and AA inhibited OS cell-mediated macrophage M2-type polarization. Mechanistically, AA inhibits the malignant behavior of OS cells and inhibits tumor progression in OS mice by suppressing Wnt3a-mediated macrophage M2 polarization. Additionally, AA-induced macrophage conversion to a pro-inflammatory phenotype in the TME of OS mice. CONCLUSION Our experiment demonstrated that AA from Cyclocarya paliurus inhibits Wnt3a-mediated M2 macrophage polarization to suppress the progression of osteosarcoma, providing a pharmacological foundation for developing therapies against OS.
Collapse
Affiliation(s)
- Jun Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong District, Changsha, Hunan, China
| | - Shuang Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong District, Changsha, Hunan, China
| | - Chao Yu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong District, Changsha, Hunan, China
| | - Xia Chen
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong District, Changsha, Hunan, China
| | - Weiye Zhong
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong District, Changsha, Hunan, China
| | - Yi Shen
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong District, Changsha, Hunan, China.
| |
Collapse
|
3
|
Fang H, Zhu D, Chen Y, Zhang C, Li G, Fang Q, Chang M, Chen Y, Gao Y. Ultrasound-Responsive 4D Bioscaffold for Synergistic Sonopiezoelectric-Gaseous Osteosarcoma Therapy and Enhanced Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417208. [PMID: 40178027 DOI: 10.1002/advs.202417208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/02/2025] [Indexed: 04/05/2025]
Abstract
Various antitumor strategies have emerged to address the escalating need for effective tumor eradication. However, achieving precise and spatiotemporally controlled dynamic therapies remains promising yet challenging. Sonopiezoelectric nanotherapy eliminates tumor cells by generating reactive oxygen species (ROS) through ultrasound stimulation, enabling spatiotemporal control and ensuring safety during deep tissue penetration. In this study, a hybrid bioscaffold incorporating few-layer black phosphorus (BP) and nitric oxide (NO) donors are rationally designed and engineered for sonopiezoelectric-gaseous synergistic therapy. This ultrasound-responsive system provides a stepwise countermeasure against tumor invasion in bone tissues. Ultrasonic vibration induces mechanical strain in BP nanosheets, leading to piezoelectric polarization and subsequent ROS generation. Moreover, ultrasound-triggered NO burst release from the donors enables spatiotemporally controlled gas therapy. The synergistic effects of sonopiezoelectric therapy and ultrasound-excited gas therapy enhance tumor eradication, effectively inhibiting tumor proliferation and metastasis while minimizing off-target cytotoxicity. Additionally, the biomineralization capability of degradable BP and proangiogenic effects of low-concentration NO establish the hybrid bioscaffold as a bioactive platform that facilitates subsequent bone regeneration. The development of this 4D multifunctional therapeutic platform, characterized by superior sonopiezoelectric efficacy, controlled NO release, and stimulatory effects on tissue regeneration, offers new insights into the comprehensive treatment of invasive bone tumors.
Collapse
Affiliation(s)
- Haoyu Fang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Gan Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qihang Fang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
4
|
Wang MF, Guo J, Yuan SJ, Li K, Zhang Q, Lei HM, Wu JL, Zhao L, Xu YH, Chen X. Targeted sonodynamic therapy induces tumor cell quasi-immunogenic ferroptosis and macrophage immunostimulatory autophagy in glioblastoma. Biomaterials 2025; 315:122913. [PMID: 39471712 DOI: 10.1016/j.biomaterials.2024.122913] [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: 06/19/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
In this study, we demonstrated the mechanism of a glioblastoma (GBM)-targeted sonodynamic therapy (SDT) strategy employing platelets loaded with a sonosensitizer based on functionalized boron nitride nanoparticles carrying chlorin e6 (BNPD-Ce6). In the in vitro study, we first found that the BNPD-Ce6-mediated sonodynamic action (SDA) induced remarkable viability loss, DNA damage, and cell death in the GBM cells (GBCs) but not macrophages. Surprisingly, the SDA-exposed GBCs displayed a ferroptotic phenotype while the SDA-exposed macrophages underwent immuno-stimulatory autophagy and potently potentiated the SDA's toxicity to the GBCs. The ferroptotic GBCs induced by the SDA were found to be quasi-immunogenic, characterized by the emission of some alarmins such as ATP, HSP90, and CRT, but absent HMGB1, a potent endogenous adjuvant. As such, the SDA-stressed GBCs were unable to stimulate the BMDMs. This defect, interestingly, could be rescued by platelets as a donor of HMGB1 which markedly enhanced the BNPD-Ce6's sonotoxicity to the GBCs. In the in vivo study, we first employed BNPD-Ce6-loaded platelets to achieve ultrasound-triggered, targeted delivery of BNPD-Ce6 in grafted intra-cranial GBMs and subsequent sonodynamic tumor damage. An SDT regimen designed based on these results slowed the growth of grafted intra-cranial GBMs and significantly increased the survival of the host animals. Pathological examination of the SDT-treated GBMs revealed tissue necrosis and destruction and validated the in vitro observations. Finally, the depletion of macrophages was found to abrogate the efficacy of the SDT in subcutaneous GBC grafts. In conclusion, the BNPD-Ce6@Plt-mediated SDT is a practicable and efficacious anti-GBM therapy. Its therapeutic mechanism critically involves a synergy of tumor cell ferroptosis, macrophage stimulation, and platelet activation induced by the SDA.
Collapse
Affiliation(s)
- Meng-Fei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Jie Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Shen-Jun Yuan
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Ke Li
- Center for Lab Teaching, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Quan Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Hui-Mei Lei
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Jia-Lin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Li Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RADX), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yong-Hong Xu
- Institute of Ophthalmological Research, Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430072, China.
| |
Collapse
|
5
|
Sunbul FS, Almuqbil RM, Zhang H, Alhudaithi SS, Fernandez ME, Aldaqqa RR, Garcia VA, Robila V, Halquist MS, Gordon SW, Bos PD, da Rocha SRP. An improved experimental model of osteosarcoma lung metastases to investigate innovative therapeutic interventions and sex as a biological variable. Int J Pharm 2025; 673:125372. [PMID: 39971171 DOI: 10.1016/j.ijpharm.2025.125372] [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: 11/15/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy, with OS lung metastasis (OSLM) being the leading cause of death in OS patients. No curative pharmacotherapies for OSLM are available, highlighting the clinical need for new therapies. Improved and rigorous preclinical models of OSLM are key in supporting advancements in this field. We aimed to develop an immunocompetent mouse model of OSLM that allows monitoring pharmacotherapies' effect on the lung metastatic burden over time and assessing the impact of sex as a biological variable in tumor growth and response to therapy. We transformed K7M2 cells to express bioluminescence and fluorescence, enabling real-time tracking of OSLM in BALB/c mice following tail vein injection. Metastasis was confined to the lungs and exhibited exponential growth with typical downregulated Fas receptor expression. In vivo bioluminescence correlated strongly with ex vivo, suggesting its reliability for evaluating metastatic progression and therapy response. Fluorescence from tdT was stable upon tissue processing, providing unique opportunities to probe the tumor characteristics ex vivo. We also assessed the effect of local lung-delivered gemcitabine, which was well-tolerated and significantly reduced OSLM burden without causing pulmonary toxicity. However, treatment did not resolve metastatic disease. We also explored the effect of sex on tumor growth and response to therapy; while no difference was observed in tumor growth between male and female mice, females showed a better response to local gemcitabine administration. In sum, we established a robust and rigorous immunocompetent mouse model of OSLM that will facilitate exploring new pharmacotherapies for OSLM.
Collapse
Affiliation(s)
- Fatemah S Sunbul
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Rashed M Almuqbil
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Hanming Zhang
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sulaiman S Alhudaithi
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew E Fernandez
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Raneem R Aldaqqa
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Victoria A Garcia
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Valentina Robila
- Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew S Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sarah W Gordon
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Thomas JeffersonUniversity, Philadelphia, PA, the United States of America
| | - Paula D Bos
- Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sandro R P da Rocha
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America.
| |
Collapse
|
6
|
Lai G, Zhao X, Chen Y, Xie T, Su Z, Lin J, Chen Y, Chen K. The origin and polarization of Macrophages and their role in the formation of the Pre-Metastatic niche in osteosarcoma. Int Immunopharmacol 2025; 150:114260. [PMID: 39938167 DOI: 10.1016/j.intimp.2025.114260] [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: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Osteosarcoma, a primary malignant bone tumor commonly found in adolescents, is highly aggressive, with a high rate of disability and mortality. It has a profound negative impact on both the physical and psychological well-being of patients. The standard treatment approach, comprising surgery and chemotherapy, has seen little improvement in patient outcomes over the past several decades. Once relapse or metastasis occurs, prognosis worsens significantly. Therefore, there is an urgent need to explore new therapeutic approaches. In recent years, the successful application of immunotherapy in certain cancers has demonstrated its potential in the field of cancer treatment. Macrophages are the predominant components of the immune microenvironment in osteosarcoma and represent critical targets for immunotherapy. Macrophages exhibit dual characteristics; while they play a key role in maintaining tumor-promoting properties within the microenvironment, such as inflammation, angiogenesis, and immune suppression, they also possess antitumor potential as part of the innate immune system. A deeper understanding of macrophages and their relationship with osteosarcoma is essential for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Guisen Lai
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Xinyi Zhao
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanquan Chen
- Department of Orthopaedic Sun Yat-sen Memorial Hospital Sun Yat-sen University PR China
| | - Tianwei Xie
- The People's Hospital of Hezhou, No.150 Xiyue Street, Hezhou 542800 PR China
| | - Zepeng Su
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Jiajie Lin
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanhai Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Keng Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China.
| |
Collapse
|
7
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
8
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
9
|
Lian H, Zhang J, Hou S, Ma S, Yu J, Zhao W, Zhao D, Zhang Z. Immunotherapy of osteosarcoma based on immune microenvironment modulation. Front Immunol 2025; 15:1498060. [PMID: 39916962 PMCID: PMC11799554 DOI: 10.3389/fimmu.2024.1498060] [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: 09/19/2024] [Accepted: 12/30/2024] [Indexed: 02/09/2025] Open
Abstract
Osteosarcoma is a highly malignant tumor with unsatisfactory therapeutic outcomes achieved by chemotherapy, radiotherapy, and surgery. As an emerging oncological treatment, immunotherapy has shown potential in the clinical management of many tumors but has a poor response rate in osteosarcoma. The immunosuppressive microenvironment in osteosarcoma is the main reason for the ineffectiveness of immunotherapy, in which the low immune response rate of immune effector cells and the high activation of immunosuppressive cells contribute to this outcome. Therefore, modulating the function of the immune microenvironment in osteosarcoma is expected to remodel the immunosuppressive microenvironment of osteosarcoma and enhance the efficacy of immunotherapy. This article reviews the role of immune cells in the progression of osteosarcoma, describes the corresponding regulatory tools for the characteristics of different cells to enhance the efficacy of osteosarcoma immunotherapy, and concludes the prospects and future challenges of osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Heping Lian
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, China
| | - Jiakui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuna Hou
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuang Ma
- Nursing Department, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiachen Yu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Tian QS, Zhang C, Bao ZJ, Pei Z. The role of CD47 in immune escape of colon cancer and its correlation with heterogeneity of tumor immune microenvironment. PeerJ 2024; 12:e18579. [PMID: 39670101 PMCID: PMC11636535 DOI: 10.7717/peerj.18579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024] Open
Abstract
Background Cluster of differentiation 47 (CD47), a transmembrane protein, plays a critical role in regulating cellular functions and maintaining immune homeostasis. Its expression has been shown to influence cancer prognosis. In this study, we investigated the role of CD47 in tumor progression in colon adenocarcinoma (COAD) and evaluated its potential as a target for immunotherapy. Materials and Methods We analyzed surgical samples from 96 COAD patients. Immunohistochemical analysis was performed on 90 samples, while the remaining 6 were subjected to multiplex immunofluorescence. To explore the association between CD47 expression and clinicopathological characteristics, we integrated transcriptome data from The Cancer Genome Atlas and the Gene Expression Omnibus using R software. The Tumor Immune Estimation Resource and Kaplan-Meier plotter were utilized to assess the relationship between CD47 expression, patient prognosis, and immune infiltration. Furthermore, the single-cell Tumor Immune System Interaction Database was used to examine the correlation between CD47 expression and the tumor microenvironment (TME). All included patients gave oral and written informed consent. The study was approved by the Ethics Committee of 3201 Hospital (full name: Medical Ethics Committee of 3201 Hospital). Results CD47 was found to be overexpressed in various tumors, including COAD. Higher CD47 expression was significantly associated with more advanced tumor stages, including TNM staging, T staging, and N staging (P < 0.05). A robust correlation was observed between CD47 expression and immune cell infiltration in COAD. Patients with elevated CD47 expression tended to have longer disease-free intervals, although this benefit was diminished in cases with high infiltration of M1 macrophages. The immunosuppressive function of CD47 primarily acted through the CD47/SIRPα pathway. Additionally, distinct cellular compositions and distributions were identified between primary and metastatic COAD, underscoring the heterogeneity of the TME. CD47 also influenced the TME by modulating cytokine and cytokine receptor interactions. Conclusion CD47 represents a promising prognostic biomarker and a potential target for immunotherapy in COAD. These findings provide new insights into therapeutic strategies aimed at modulating the TME and improving patient outcomes.
Collapse
Affiliation(s)
- Qiu-Si Tian
- Department of Neurosurgery, 3201 Hospital of Xi’an Jiaotong University Health Science Center, HanZhong, ShaanXi, China
| | - ChunMei Zhang
- Department of Pathology, Chongqing University Jiangjin Hospital, ChongQing, China
| | - Zhi-Jun Bao
- Department of Neurosurgery, 3201 Hospital of Xi’an Jiaotong University Health Science Center, HanZhong, ShaanXi, China
| | - ZhiGang Pei
- Department of Pathology, Chongqing University Jiangjin Hospital, ChongQing, China
| |
Collapse
|
11
|
Zhang M, Sun D, Huang H, Yang D, Song X, Feng W, Jing X, Chen Y. Nanosonosensitizer Optimization for Enhanced Sonodynamic Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409663. [PMID: 39308222 DOI: 10.1002/adma.202409663] [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: 07/05/2024] [Revised: 08/16/2024] [Indexed: 11/16/2024]
Abstract
Low-intensity ultrasound-mediated sonodynamic therapy (SDT), which, by design, integrates sonosensitizers and molecular oxygen to generate therapeutic substances (e.g., toxic hydroxyl radicals, superoxide anions, or singlet oxygen) at disease sites, has shown enormous potential for the effective treatment of a variety of diseases. Nanoscale sonosensitizers play a crucial role in the SDT process because their structural, compositional, physicochemical, and biological characteristics are key determinants of therapeutic efficacy. In particular, advances in materials science and nanotechnology have invigorated a series of optimization strategies for augmenting the therapeutic efficacy of nanosonosensitizers. This comprehensive review systematically summarizes, discusses, and highlights state-of-the-art studies on the current achievements of nanosonosensitizer optimization in enhanced sonodynamic disease treatment, with an emphasis on the general design principles of nanosonosensitizers and their optimization strategies, mainly including organic and inorganic nanosonosensitizers. Additionally, recent advancements in optimized nanosonosensitizers for therapeutic applications aimed at treating various diseases, such as cancer, bacterial infections, atherosclerosis, and autoimmune diseases, are clarified in detail. Furthermore, the biological effects of the improved nanosonosensitizers for versatile SDT applications are thoroughly discussed. The review concludes by highlighting the current challenges and future opportunities in this rapidly evolving research field to expedite its practical clinical translation and application.
Collapse
Affiliation(s)
- Min Zhang
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Dandan Sun
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Hui Huang
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Dayan Yang
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Xinran Song
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Wei Feng
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiangxiang Jing
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Yu Chen
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| |
Collapse
|
12
|
Wang N, Luo L, Xu X, Zhou H, Li F. Focused ultrasound-induced cell apoptosis for the treatment of tumours. PeerJ 2024; 12:e17886. [PMID: 39184389 PMCID: PMC11344538 DOI: 10.7717/peerj.17886] [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: 02/01/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Cancer is a serious public health problem worldwide. Traditional treatments, such as surgery, radiotherapy, chemotherapy, and immunotherapy, do not always yield satisfactory results; therefore, an efficient treatment for tumours is urgently needed. As a convenient and minimally invasive modality, focused ultrasound (FUS) has been used not only as a diagnostic tool but also as a therapeutic tool in an increasing number of studies. FUS can help treat malignant tumours by inducing apoptosis. This review describes the three apoptotic pathways, apoptotic cell clearance, and how FUS affects these three apoptotic pathways. This review also discusses the role of thermal and cavitation effects on apoptosis, including caspase activity, mitochondrial dysfunction, and Ca2+ elease. Finally, this article reviews various aspects of FUS combination therapy, including sensitization by radiotherapy and chemotherapy, gene expression upregulation, and the introduction of therapeutic gases, to provide new ideas for clinical tumour therapy.
Collapse
Affiliation(s)
- Na Wang
- Chongqing University, School of Medicine, Chongqing, China
- Chongqing University Cancer Hospital, Ultrasound Department, Chongqing, China
| | - Li Luo
- Chongqing University Cancer Hospital, Ultrasound Department, Chongqing, China
| | - Xinzhi Xu
- Chongqing University Cancer Hospital, Ultrasound Department, Chongqing, China
| | - Hang Zhou
- Chongqing University Cancer Hospital, Ultrasound Department, Chongqing, China
| | - Fang Li
- Chongqing University Cancer Hospital, Ultrasound Department, Chongqing, China
| |
Collapse
|
13
|
Wang D, Deng X, Wang J, Che S, Ma X, Zhang S, Dong Q, Huang C, Chen J, Shi C, Zhang MR, Hu K, Luo L, Xiao Z. Environmentally responsive hydrogel promotes vascular normalization to enhance STING anti-tumor immunity. J Control Release 2024; 372:403-416. [PMID: 38914207 DOI: 10.1016/j.jconrel.2024.06.052] [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: 05/28/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
The immunosuppressive microenvironment of malignant tumors severely hampers the effectiveness of anti-tumor therapy. Moreover, abnormal tumor vasculature interacts with immune cells, forming a vicious cycle that further interferes with anti-tumor immunity and promotes tumor progression. Our pre-basic found excellent anti-tumor effects of c-di-AMP and RRx-001, respectively, and we further explored whether they could be combined synergistically for anti-tumor immunotherapy. We chose to load these two drugs on PVA-TSPBA hydrogel scaffolds that expressly release drugs within the tumor microenvironment by in situ injection. Studies have shown that c-di-AMP activates the STING pathway, enhances immune cell infiltration, and reverses tumor immunosuppression. Meanwhile, RRx-001 releases nitric oxide, which increases oxidative stress injury in tumor cells and promotes apoptosis. Moreover, the combination of the two presented more powerful pro-vascular normalization and reversed tumor immunosuppression than the drug alone. This study demonstrates a new design option for anti-tumor combination therapy and the potential of tumor environmentally responsive hydrogel scaffolds in combination with anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Xiujiao Deng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Pharmacy, The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jinghao Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Pharmacy, The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuang Che
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Xiaocong Ma
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Radiology, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan 517000, China
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qiu Dong
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Cuiqing Huang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Changzheng Shi
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical, Science, National Institutes for Quantum Science and Technology, Chiba 2638555, Japan
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Radiology, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan 517000, China.
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| |
Collapse
|
14
|
Li X, Sun X, Chen H, Wang Y, Chen H, Gao Y. Boron Dipyrromethene-Based Nanotheranostic System for Sonophotoassisted Therapy and Simultaneous Monitoring of Tumor Immune Microenvironment Reprogramming. ACS NANO 2024; 18:18230-18245. [PMID: 38950337 DOI: 10.1021/acsnano.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Therapy-induced modulation of the tumor microenvironment (TME) to overcome the immunosuppressive TME is considered to be an opportunity for cancer treatment. However, monitoring of TME modulation during the therapeutic process to accurately determine immune responses and adjust treatment plans in a timely manner remains to be challenging. Herein, we report a carrier-free nanotheranostic system (CANPs) assembled by two boron dipyrromethene (BODIPY) dyes, a sonophotosensitizer C-BDP, and a nitric oxide (NO) probe amino-BODIPY (A-BDP). CANPs can exert combined sonophototherapeutic effects of C-BDP under ultrasound and light irradiation and simultaneously induce inflammatory TME, as well as emit bright fluorescence via A-BDP by monitoring tumor-associated macrophages (TAMs) repolarization through the released NO in vitro and in vivo. Of note, transforming growth factor-β (TGF-β) could be the key cytokine involved in the sonophototherapy-induced TME reprogramming. By virtue of high physiological stability, good biocompatibility, and effective tumor targetability, CANPs could be a potential nanotheranostic system for the simultaneous induction and detection of TME reprogramming triggered by sonophototherapy.
Collapse
Affiliation(s)
- Xudong Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and 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, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Hui Chen
- College of Chemistry, Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), Fuzhou University, Fuzhou, Fujian 350116, China
| | - Ya Wang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Haijun Chen
- College of Chemistry, Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
15
|
Guo S, Li Z, Zhou R, Feng J, Huang L, Ren B, Zhu J, Huang Y, Wu G, Cai H, Zhang Q, Ke Y, Guan T, Chen P, Xu Y, Yan C, Ou C, Shen Z. MRI-Guided Tumor Therapy Based on Synergy of Ferroptosis, Immunosuppression Reversal and Disulfidptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309842. [PMID: 38431935 DOI: 10.1002/smll.202309842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/19/2024] [Indexed: 03/05/2024]
Abstract
Triple negative breast cancer (TNBC) cells have a high demand for oxygen and glucose to fuel their growth and spread, shaping the tumor microenvironment (TME) that can lead to a weakened immune system by hypoxia and increased risk of metastasis. To disrupt this vicious circle and improve cancer therapeutic efficacy, a strategy is proposed with the synergy of ferroptosis, immunosuppression reversal and disulfidptosis. An intelligent nanomedicine GOx-IA@HMON@IO is successfully developed to realize this strategy. The Fe release behaviors indicate the glutathione (GSH)-responsive degradation of HMON. The results of titanium sulfate assay, electron spin resonance (ESR) spectra, 5,5'-Dithiobis-(2-nitrobenzoic acid (DTNB) assay and T1-weighted magnetic resonance imaging (MRI) demonstrate the mechanism of the intelligent iron atom (IA)-based cascade reactions for GOx-IA@HMON@IO, generating robust reactive oxygen species (ROS). The results on cells and mice reinforce the synergistic mechanisms of ferroptosis, immunosuppression reversal and disulfidptosis triggered by the GOx-IA@HMON@IO with the following steps: 1) GSH peroxidase 4 (GPX4) depletion by disulfidptosis; 2) IA-based cascade reactions; 3) tumor hypoxia reversal; 4) immunosuppression reversal; 5) GPX4 depletion by immunotherapy. Based on the synergistic mechanisms of ferroptosis, immunosuppression reversal and disulfidptosis, the intelligent nanomedicine GOx-IA@HMON@IO can be used for MRI-guided tumor therapy with excellent biocompatibility and safety.
Collapse
Affiliation(s)
- Shuai Guo
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Zongheng Li
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Ruilong Zhou
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jie Feng
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Lin Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Bin Ren
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jiaoyang Zhu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Ya Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Guochao Wu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Haobin Cai
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Qianqian Zhang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Yushen Ke
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Tianwang Guan
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Peier Chen
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Yikai Xu
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Chenggong Yan
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Caiwen Ou
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
16
|
Wang J, Liang S, Chen S, Ma T, Chen M, Niu C, Leng Y, Wang L. Bacterial outer membrane vesicle-cancer cell hybrid membrane-coated nanoparticles for sonodynamic therapy in the treatment of breast cancer bone metastasis. J Nanobiotechnology 2024; 22:328. [PMID: 38858780 PMCID: PMC11165797 DOI: 10.1186/s12951-024-02619-w] [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: 03/11/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
Breast cancer bone metastasis is a terminal-stage disease and is typically treated with radiotherapy and chemotherapy, which causes severe side effects and limited effectiveness. To improve this, Sonodynamic therapy may be a more safe and effective approach in the future. Bacterial outer membrane vesicles (OMV) have excellent immune-regulating properties, including modulating macrophage polarization, promoting DC cell maturation, and enhancing anti-tumor effects. Combining OMV with Sonodynamic therapy can result in synergetic anti-tumor effects. Therefore, we constructed multifunctional nanoparticles for treating breast cancer bone metastasis. We fused breast cancer cell membranes and bacterial outer membrane vesicles to form a hybrid membrane (HM) and then encapsulated IR780-loaded PLGA with HM to produce the nanoparticles, IR780@PLGA@HM, which had tumor targeting, immune regulating, and Sonodynamic abilities. Experiments showed that the IR780@PLGA@HM nanoparticles had good biocompatibility, effectively targeted to 4T1 tumors, promoted macrophage type I polarization and DC cells activation, strengthened anti-tumor inflammatory factors expression, and presented the ability to effectively kill tumors both in vitro and in vivo, which showed a promising therapeutic effect on breast cancer bone metastasis. Therefore, the nanoparticles we constructed provided a new strategy for effectively treating breast cancer bone metastasis.
Collapse
Affiliation(s)
- Jiahao Wang
- The School of Medicine, Nankai University, Tianjin, 300071, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shuailong Liang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, Second Xiangya Hospital, Central South University, Changsha, China
| | - Tianliang Ma
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mingyu Chen
- Department of Ultrasound Diagnosis, Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Leng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China.
| |
Collapse
|
17
|
Liang H, Cui M, Tu J, Chen X. Advancements in osteosarcoma management: integrating immune microenvironment insights with immunotherapeutic strategies. Front Cell Dev Biol 2024; 12:1394339. [PMID: 38915446 PMCID: PMC11194413 DOI: 10.3389/fcell.2024.1394339] [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: 03/01/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Osteosarcoma, a malignant bone tumor predominantly affecting children and adolescents, presents significant therapeutic challenges, particularly in metastatic or recurrent cases. Conventional surgical and chemotherapeutic approaches have achieved partial therapeutic efficacy; however, the prognosis for long-term survival remains bleak. Recent studies have highlighted the imperative for a comprehensive exploration of the osteosarcoma immune microenvironment, focusing on the integration of diverse immunotherapeutic strategies-including immune checkpoint inhibitors, tumor microenvironment modulators, cytokine therapies, tumor antigen-specific interventions, cancer vaccines, cellular therapies, and antibody-based treatments-that are directly pertinent to modulating this intricate microenvironment. By targeting tumor cells, modulating the tumor microenvironment, and activating host immune responses, these innovative approaches have demonstrated substantial potential in enhancing the effectiveness of osteosarcoma treatments. Although most of these novel strategies are still in research or clinical trial phases, they have already demonstrated significant potential for individuals with osteosarcoma, suggesting the possibility of developing new, more personalized and effective treatment options. This review aims to provide a comprehensive overview of the current advancements in osteosarcoma immunotherapy, emphasizing the significance of integrating various immunotherapeutic methods to optimize therapeutic outcomes. Additionally, it underscores the imperative for subsequent research to further investigate the intricate interactions between the tumor microenvironment and the immune system, aiming to devise more effective treatment strategies. The present review comprehensively addresses the landscape of osteosarcoma immunotherapy, delineating crucial scientific concerns and clinical challenges, thereby outlining potential research directions.
Collapse
Affiliation(s)
- Hang Liang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Cui
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Bai Z, Huang J, Lu H, Wang N, Li H, Zhu Y. Based on polydopamine-coated metal organic framework multifunctional nanoplatform for enhanced photothermal/sonodynamicand treatment combined with checkpoint blockade therapy. Int J Biol Macromol 2024; 269:132207. [PMID: 38723823 DOI: 10.1016/j.ijbiomac.2024.132207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
To overcome the low efficacy of sonodynamic therapy (SDT) caused by hypoxia in the tumor microenvironment, we developed a multiple anti-tumor nanoplatform with synergistic SDT, photothermal therapy (PTT), and ferroptosis effects. PCN-224@FcCaO2/Mn/dihydroartemisinin/imiquimod/PDA (PFC) was prepared by modified with dihydroartemisinin (DHA), imiquimod (R837), CaO2, ferrocene (Fc) and Mn2+ on the PCN-224 (Cu) to achieve self-replenishment of H2O2/O2 and GSH consumption. FcCaO2 decomposed into H2O2 in the tumor microenvironment, triggering the Fenton effect to produce OH, and Cu2+ reduced the potential loss of OH by the depletion of GSH. Under ultrasonic (US) and laser irradiation, PFC exhibits exciting PTT and SDT effects from polydopamine (PDA) and PCN-224. Mn2+ not only promoted the reaction of H2O2 to produce O2 to effectively enhance SDT but also induced tumor cell apoptosis by Mn2+ combined with DHA. PFC induced ferroptosis via Fe interaction with DHA to produce ROS and reduce the expression of GPX4. The released R837 and tumor-associated antigens from SDT/PTT can produce damage associated molecular patterns (DAMPs), which can initiate adaptive immune responses to kill cancer cells, and released again to promote the tumor immune cycle. What's more, SDT/PTT and ferroptosis combined with aPD-L1 can effectively suppress both primary and distant tumor growth.
Collapse
Affiliation(s)
- Zhihao Bai
- College of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, China; State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - JianFeng Huang
- Department of nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - HaiZhen Lu
- Department of nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China.
| | - HaoYu Li
- Department of nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China.
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter EX4 4QF, UK.
| |
Collapse
|
19
|
Qi H, Li Y, Geng Y, Wan X, Cai X. Nanoparticle-mediated immunogenic cell death for cancer immunotherapy. Int J Pharm 2024; 656:124045. [PMID: 38561134 DOI: 10.1016/j.ijpharm.2024.124045] [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: 11/05/2023] [Revised: 03/01/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
The field of cancer therapy is witnessing the emergence of immunotherapy, an innovative approach that activates the body own immune system to combat cancer. Immunogenic cell death (ICD) has emerged as a prominent research focus in the field of cancer immunotherapy, attracting significant attention in recent years. The activation of ICD can induce the release of damage-associated molecular patterns (DAMPs), such as calreticulin (CRT), adenosine triphosphate (ATP), high mobility group box protein 1 (HMGB1), and heat shock proteins (HSP). Subsequently, this process promotes the maturation of innate immune cells, including dendritic cells (DCs), thereby triggering a T cell-mediated anti-tumor immune response. The activation of the ICD ultimately leads to the development of long-lasting immune responses against tumors. Studies have demonstrated that partial therapeutic approaches, such as chemotherapy with doxorubicin, specific forms of radiotherapy, and phototherapy, can induce the generation of ICD. The main focus of this article is to discuss and review the therapeutic methods triggered by nanoparticles for ICD, while briefly outlining their anti-tumor mechanism. The objective is to provide a comprehensive reference for the widespread application of ICD.
Collapse
Affiliation(s)
- Haolong Qi
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Yuan Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Yingjie Geng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Xinhuan Wan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Xiaoqing Cai
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China.
| |
Collapse
|
20
|
Liu B, Du F, Feng Z, Xiang X, Guo R, Ma L, Zhu B, Qiu L. Ultrasound-augmented cancer immunotherapy. J Mater Chem B 2024; 12:3636-3658. [PMID: 38529593 DOI: 10.1039/d3tb02705h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Cancer is a growing worldwide health problem with the most broadly studied treatments, in which immunotherapy has made notable advancements in recent years. However, innumerable patients have presented a poor response to immunotherapy and simultaneously experienced immune-related adverse events, with failed therapeutic results and increased mortality rates. Consequently, it is crucial to develop alternate tactics to boost therapeutic effects without producing negative side effects. Ultrasound is considered to possess significant therapeutic potential in the antitumor field because of its inherent characteristics, including cavitation, pyrolysis, and sonoporation. Herein, this timely review presents the comprehensive and systematic research progress of ultrasound-enhanced cancer immunotherapy, focusing on the various ultrasound-related mechanisms and strategies. Moreover, this review summarizes the design and application of current sonosensitizers based on sonodynamic therapy, with an attempt to provide guidance on new directions for future cancer therapy.
Collapse
Affiliation(s)
- Bingjie Liu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Fangxue Du
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ziyan Feng
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xi Xiang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruiqian Guo
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bihui Zhu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
21
|
Zhang S, Zhang X, Gao H, Zhang X, Sun L, Huang Y, Zhang J, Ding B. Cell Membrane-Coated Biomimetic Nanoparticles in Cancer Treatment. Pharmaceutics 2024; 16:531. [PMID: 38675192 PMCID: PMC11055162 DOI: 10.3390/pharmaceutics16040531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Nanoparticle-based drug delivery systems hold promise for cancer treatment by enhancing the solubility and stability of anti-tumor drugs. Nonetheless, the challenges of inadequate targeting and limited biocompatibility persist. In recent years, cell membrane nano-biomimetic drug delivery systems have emerged as a focal point of research and development, due to their exceptional traits, including precise targeting, low toxicity, and good biocompatibility. This review outlines the categorization and advantages of cell membrane bionic nano-delivery systems, provides an introduction to preparation methods, and assesses their applications in cancer treatment, including chemotherapy, gene therapy, immunotherapy, photodynamic therapy, photothermal therapy, and combination therapy. Notably, the review delves into the challenges in the application of various cell membrane bionic nano-delivery systems and identifies opportunities for future advancement. Embracing cell membrane-coated biomimetic nanoparticles presents a novel and unparalleled avenue for personalized tumor therapy.
Collapse
Affiliation(s)
- Shu Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 214122, China;
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Xiaojuan Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Huan Gao
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Xiaoqin Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Lidan Sun
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Yueyan Huang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Jie Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Baoyue Ding
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| |
Collapse
|
22
|
Zanata SM, El-Shafai NM, Beltagi AM, Alshehri S, El-Sherbiny M, El-Mehasseb IM. Bio-study: Modeling of natural nanomolecules as a nanocarrier surface for antioxidant and glucose biosensor. Int J Biol Macromol 2024; 264:130634. [PMID: 38460624 DOI: 10.1016/j.ijbiomac.2024.130634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 03/11/2024]
Abstract
The nature of nano molecules as a self-assembled nanocomposite surface depends on the nanoparticles of sodium butyrate, cellulose, and pycnogenol; the synthesis is achieved via precipitation and grinding methods. The excellent functionalized surface of nanocomposite (NCP) enables the loading of the selected drugs, where the efficiency of the NCP surface arrived at 92.2 %. The electrochemical behavior emphasized the success of a functionalized NCP surface for incorporation with drugs for the drug delivery system, the results of cytotoxicity detect the effect of NCP on the mouse normal liver (BNL) cells, where the high and low concentrations on the BNL cells have a safe dose. Cell viability with BNL cells was reported at 101.8 % with10 μL and 100.12 % with 100 μL, the interaction between the NCP and the human serum albumin (HSA) at room temperature. The low interaction rate with the glutamate and increased binding with the oxidized glutathione disulfide (GSSG) and reduced glutathione (SGH) reflect the antioxidant activity of NCP. The strong binding of NCP with biomolecules such as glucose is referred to as the biosensor property. The results recommend that NCP is an excellent nanocarrier for drug delivery and glucose biosensors for diabetes.
Collapse
Affiliation(s)
- Samar M Zanata
- Nanotechnology Center, Chemistry Department, Faculty of Science, Kafrelsheikh University 33516, Egypt
| | - Nagi M El-Shafai
- Nanotechnology Center, Chemistry Department, Faculty of Science, Kafrelsheikh University 33516, Egypt.
| | - Amr M Beltagi
- Nanotechnology Center, Chemistry Department, Faculty of Science, Kafrelsheikh University 33516, Egypt
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia
| | - Ibrahim M El-Mehasseb
- Nanotechnology Center, Chemistry Department, Faculty of Science, Kafrelsheikh University 33516, Egypt
| |
Collapse
|
23
|
Yang W, Cui Y, Pan Q, Peng Y, Li K, Huang W, Zhang Y, Hu H, Shao Z, Zhang Z. Biomimetic engineered nanoparticles target drug-resistant tumor cells and heterogeneous blood vessels for combination therapy of osteosarcoma. CHEMICAL ENGINEERING JOURNAL 2024; 485:149761. [DOI: 10.1016/j.cej.2024.149761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2025]
|
24
|
Li SL, Hou HY, Chu X, Zhu YY, Zhang YJ, Duan MD, Liu J, Liu Y. Nanomaterials-Involved Tumor-Associated Macrophages' Reprogramming for Antitumor Therapy. ACS NANO 2024; 18:7769-7795. [PMID: 38420949 DOI: 10.1021/acsnano.3c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Tumor-associated macrophages (TAMs) play pivotal roles in tumor development. As primary contents of tumor environment (TME), TAMs secrete inflammation-related substances to regulate tumoral occurrence and development. There are two kinds of TAMs: the tumoricidal M1-like TAMs and protumoral M2-like TAMs. Reprogramming TAMs from immunosuppressive M2 to immunocompetent M1 phenotype is considered a feasible way to improve immunotherapeutic efficiency. Notably, nanomaterials show great potential for biomedical fields due to their controllable structures and properties. There are many types of nanomaterials that exhibit great regulatory activities for TAMs' reprogramming. In this review, the recent progress of nanomaterials-involved TAMs' reprogramming is comprehensively discussed. The various nanomaterials for TAMs' reprogramming and the reprogramming strategies are summarized and introduced. Additionally, the challenges and perspectives of TAMs' reprogramming for efficient therapy are discussed, aiming to provide inspiration for TAMs' regulator design and promote the development of TAMs-mediated immunotherapy.
Collapse
Affiliation(s)
- Shu-Lan Li
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Hua-Ying Hou
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Xu Chu
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Juan Zhang
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Meng-Die Duan
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Junyi Liu
- Albany Medical College, New York 12208, United States
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| |
Collapse
|
25
|
Fu Y, Zhang H, Ye J, Chen C, Yang Y, Wu B, Yin X, Shi J, Zhu Y, Zhao C, Zhang W. An "all-in-one" treatment and imaging nanoplatform for breast cancer with photothermal nanoparticles. NANOSCALE ADVANCES 2024; 6:1423-1435. [PMID: 38419880 PMCID: PMC10898424 DOI: 10.1039/d3na00814b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024]
Abstract
Drug delivery systems based on nanoparticles still face challenges of low efficacy and an inability to track treatment effects in tumor therapy due to biological barriers. This limitation hinders clinicians' ability to determine treatment effects and proper drug dosages, thus, ultimately impeding the further application and transformation of nanoplatforms. To address this challenge, an all-in-one nanoplatform for therapy and imaging is proposed. The nanoplatform is constructed by using nanoparticles through the co-encapsulation of the photothermal therapeutic agent IR780, the passively targeted drug OA@Fe3O4, and the chemotherapeutic drug paclitaxel. Under the guidance of magnetic navigation, the nanoparticles can enhance local enrichment of the drug, while the luminescence properties of IR780 enable drug tracking at the same time. Remarkably, the nanoparticles exhibit improved photothermal-chemotherapy synergy under magnetic targeting guidance, demonstrating antitumor effects in both in vitro and in vivo experiments. It is demonstrated that the use of these polymeric nanoparticles has significant potential for future biomedical applications and clinical decisions.
Collapse
Affiliation(s)
- Yuping Fu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing 210008 China
| | - Hongmei Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing 210008 China
| | - Jiahui Ye
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Changrong Chen
- Division of Emergency Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Yaxuan Yang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Baojuan Wu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Xi Yin
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Jiajun Shi
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Yun Zhu
- Division of Pharmacy Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Cheng Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing 210008 China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University Nanjing 210008 China
| |
Collapse
|
26
|
Lu Y, Fan L, Wang J, Hu M, Wei B, Shi P, Li J, Feng J, Zheng Y. Cancer Cell Membrane-Based Materials for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306540. [PMID: 37814370 DOI: 10.1002/smll.202306540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/18/2023] [Indexed: 10/11/2023]
Abstract
The nanodelivery system provides a novel direction for disease diagnosis and treatment; however, its delivery effectiveness is restricted by the short biological half-life and inadequate tumor targeting. The immune evasion properties and homologous targeting capabilities of natural cell membranes, particularly those of cancer cell membranes (CCM), have gained significant interest. The integration of CCM and nanoparticles has resulted in the emergence of CCM-based nanoplatforms (CCM-NPs), which have gained significant attention due to their unique properties. CCM-NPs not only prolong the blood circulation time of core nanoparticles, but also direct them for homologous tumor targeting. Herein, the history and development of CCM-NPs as well as how these platforms have been used for biomedical applications are discussed. The application of CCM-NPs for cancer therapy will be described in detail. Translational efforts are currently under way and further research to address key areas of need will ultimately be required to facilitate the successful clinical adoption of CCM-NPs.
Collapse
Affiliation(s)
- Yongping Lu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
- Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Linming Fan
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jun Wang
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Mingxiang Hu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Baogang Wei
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Ping Shi
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Jinyan Feng
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Yu Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
27
|
Xie X, Zhang J, Wang Y, Shi W, Tang R, Tang Q, Sun S, Wu R, Xu S, Wang M, Liang X, Cui L. Nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. Mater Today Bio 2024; 24:100926. [PMID: 38179429 PMCID: PMC10765306 DOI: 10.1016/j.mtbio.2023.100926] [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: 10/05/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024] Open
Abstract
Immunotherapy as a milestone in cancer treatment has made great strides in the past decade, but it is still limited by low immune response rates and immune-related adverse events. Utilizing bioeffects of ultrasound to enhance tumor immunotherapy has attracted more and more attention, including sonothermal, sonomechanical, sonodynamic and sonopiezoelectric immunotherapy. Moreover, the emergence of nanomaterials has further improved the efficacy of ultrasound mediated immunotherapy. However, most of the summaries in this field are about a single aspect of the biological effects of ultrasound, which is not comprehensive and complete currently. This review proposes the recent progress of nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. The concept of immunotherapy and the application of bioeffects of ultrasound in cancer immunotherapy are initially introduced. Then, according to different bioeffects of ultrasound, the representative paradigms of nanomaterial augmented sono-immunotherapy are described, and their mechanisms are discussed. Finally, the challenges and application prospects of nanomaterial augmented ultrasound mediated cancer immunotherapy are discussed in depth, hoping to pave the way for cancer immunotherapy and promote the clinical translation of ultrasound mediated cancer immunotherapy through the reasonable combination of nanomaterials augmented ultrasonic bioeffects.
Collapse
Affiliation(s)
- Xinxin Xie
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Jinxia Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yuan Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Wanrui Shi
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Rui Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Qingshuang Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ruiqi Wu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Shuyu Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Mengxin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ligang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| |
Collapse
|
28
|
Ling H, Zhang Q, Luo Q, Ouyang D, He Z, Sun J, Sun M. Dynamic immuno-nanomedicines in oncology. J Control Release 2024; 365:668-687. [PMID: 38042376 DOI: 10.1016/j.jconrel.2023.11.052] [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: 09/25/2023] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Anti-cancer therapeutics have achieved significant advances due to the emergence of immunotherapies that rely on the identification of tumors by the patients' immune system and subsequent tumor eradication. However, tumor cells often escape immunity, leading to poor responsiveness and easy tolerance to immunotherapy. Thus, the potentiated anti-tumor immunity in patients resistant to immunotherapies remains a challenge. Reactive oxygen species-based dynamic nanotherapeutics are not new in the anti-tumor field, but their potential as immunomodulators has only been demonstrated in recent years. Dynamic nanotherapeutics can distinctly enhance anti-tumor immune response, which derives the concept of the dynamic immuno-nanomedicines (DINMs). This review describes the pivotal role of DINMs in cancer immunotherapy and provides an overview of the clinical realities of DINMs. The preclinical development of emerging DINMs is also outlined. Moreover, strategies to synergize the antitumor immunity by DINMs in combination with other immunologic agents are summarized. Last but not least, the challenges and opportunities related to DINMs-mediated immune responses are also discussed.
Collapse
Affiliation(s)
- Hao Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qinyi Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001, China
| | - Defang Ouyang
- Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
29
|
Yang M, Xie M, Guo J, Zhang Y, Qiu Y, Wang Z, Du Y. Mucus-Permeable Sonodynamic Therapy Mediated Amphotericin B-Loaded PEGylated PLGA Nanoparticles Enable Eradication of Candida albicans Biofilm. Int J Nanomedicine 2023; 18:7941-7963. [PMID: 38169688 PMCID: PMC10758343 DOI: 10.2147/ijn.s437726] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Background Candida albicans (C. albicans) forms pathogenic biofilms, and the dense mucus layer secreted by the epithelium is a major barrier to the traditional antibiotic treatment of mucosa-associated C. albicans infections. Herein, we report a novel anti-biofilm strategy of mucus-permeable sonodynamic therapy (mp-SDT) based on ultrasound (US)-mediated amphotericin B-loaded PEGylated PLGA nanoparticles (AmB-NPs) to overcome mucus barrier and enable the eradication of C. albicans biofilm. Methods AmB-NPs were fabricated using ultrasonic double emulsion method, and their physicochemical and sonodynamic properties were determined. The mucus and biofilm permeability of US-mediated AmB-NPs were further investigated. Moreover, the anti-biofilm effect of US-mediated AmB-NPs treatment was thoroughly evaluated on mucus barrier abiotic biofilm, epithelium-associated biotic biofilm, and C. albicans-induced rabbit vaginal biofilms model. In addition, the ultrastructure and secreted cytokines of epithelial cells and the polarization of macrophages were analyzed to investigate the regulation of local cellular immune function by US-mediated AmB-NPs treatment. Results Polymeric AmB-NPs display excellent sonodynamic performance with massive singlet oxygen (1O2) generation. US-mediated AmB-NPs could rapidly transport through mucus and promote permeability in biofilms, which exhibited excellent eradicating ability to C. albicans biofilms. Furthermore, in the vaginal epithelial cells (VECs)-associated C. albicans biofilm model, the mp-SDT scheme showed the strongest biofilm eradication effect, with up to 98% biofilm re-formation inhibition rate, improved the ultrastructural damage, promoted local immune defense enhancement of VECs, and regulated the polarization of macrophages to the M1 phenotype to enhance macrophage-associated antifungal immune responses. In addition, mp-SDT treatment exhibited excellent therapeutic efficacy against C. albicans-induced rabbit vaginitis, promoted the recovery of mucosal epithelial ultrastructure, and contributed to the reshaping of a healthier vaginal microbiome. Conclusion The synergistic anti-biofilm strategies of mp-SDT effectively eradicated C. albicans biofilm and simultaneously regulated local antifungal immunity enhancement, which may provide a new approach to treat refractory drug-resistant biofilm-associated mucosal candidiasis.
Collapse
Affiliation(s)
- Min Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Mengyao Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jiajun Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yuqing Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yan Qiu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
30
|
Wang X, Xu X, Yang Z, Xu X, Han S, Zhang H. Improvement of the effectiveness of sonodynamic therapy: by optimizing components and combination with other treatments. Biomater Sci 2023; 11:7489-7511. [PMID: 37873617 DOI: 10.1039/d3bm00738c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Sonodynamic therapy (SDT) is an emerging treatment method. In comparison with photodynamic therapy (PDT), SDT exhibits deep penetration, high cell membrane permeability, and free exposure to light capacity. Unfortunately, owing to inappropriate ultrasound parameter selection, poor targeting of sonosensitizers, and the complex tumor environment, SDT is frequently ineffective. In this review, we describe the approaches for selecting ultrasound parameters and how to develop sonosensitizers to increase targeting and improve adverse tumor microenvironments. Furthermore, the potential of combining SDT with other treatment methods, such as chemotherapy, chemodynamic therapy, photodynamic therapy, photothermal therapy, and immunotherapy, is discussed to further increase the treatment efficiency of SDT.
Collapse
Affiliation(s)
- Xiangting Wang
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Xiaohong Xu
- Department of Ultrasound, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhe Yang
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Xuanshou Xu
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Shisong Han
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Heng Zhang
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| |
Collapse
|
31
|
Li S, Mok GSP, Dai Y. Lipid bilayer-based biological nanoplatforms for sonodynamic cancer therapy. Adv Drug Deliv Rev 2023; 202:115110. [PMID: 37820981 DOI: 10.1016/j.addr.2023.115110] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/01/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Sonodynamic therapy (SDT) has been developed as a promising alternative therapeutic modality for cancer treatment, involving the synergetic application of sonosensitizers and low-intensity ultrasound. However, the antitumor efficacy of SDT is significantly limited due to the poor performance of conventional sonosensitizers in vivo and the constrained tumor microenvironment (TME). Recent breakthroughs in lipid bilayer-based nanovesicles (LBBNs), including multifunctional liposomes, exosomes, and isolated cellular membranes, have brought new insights into the advancement of SDT. Despite their distinct sources and preparation methods, the lipid bilayer structure in common allows them to be functionalized in many comparable ways to serve as ideal nanocarriers against challenges arising from the tumor-specific sonosensitizer delivery and the complicated TME. In this review, we provide a comprehensive summary of the recent advances in LBBN-based SDT, with particular attention on how LBBNs can be engineered to improve the delivery efficiency of sonosensitizers and overcome physical, biological, and immune barriers within the TME for enhanced sonodynamic cancer therapy. We anticipate that this review will offer valuable guidance in the construction of LBBN-based nanosonosensitizers and contribute to the development of advanced strategies for next-generation sonodynamic cancer therapy.
Collapse
Affiliation(s)
- Songhao Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Greta S P Mok
- Biomedical Imaging Laboratory (BIG), Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Macau SAR 999078, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
32
|
Li J, Hu Z, Zhu J, Lin X, Gao X, Lv G. Antitumor Effects of Pegylated Zinc Protoporphyrin-Mediated Sonodynamic Therapy in Ovarian Cancer. Pharmaceutics 2023; 15:2275. [PMID: 37765244 PMCID: PMC10534787 DOI: 10.3390/pharmaceutics15092275] [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: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Sonodynamic therapy (SDT) induces reactive oxygen species (ROS) to kill tumor cells. Heme oxygenase-1 (HO-1), as an important antioxidant enzyme, resists killing by scavenging ROS. Zinc protoporphyrin (ZnPP) not only effectively inhibits HO-1 activity, but also becomes a potential sonosensitizer. However, its poor water solubility limits its applications. Herein, we developed an improved water-soluble method. It was proved that pegylated zinc protoporphyrin-mediated SDT (PEG-ZnPP-SDT) could significantly enhance ROS production by destroying the HO-1 antioxidant system in ovarian cancer. Increased ROS could cause mitochondrial membrane potential collapse, release cytochrome c from mitochondria to the cytoplasm, and trigger the mitochondrial-caspase apoptotic pathway. In conclusion, our results demonstrated that PEG-ZnPP-SDT, as a novel sonosensitizer, could improve the antitumor effects by destroying the HO-1 antioxidant system. It provided a new therapeutic strategy for SDT to treat cancers, especially those with higher HO-1 expression.
Collapse
Affiliation(s)
- Jia Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| | - Zheng Hu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China;
| | - Jiwei Zhu
- Department of Forensic Medicine, Harbin Medical University, Harbin 150086, China;
| | - Xin Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| | - Guixiang Lv
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| |
Collapse
|