1
|
Li Q, Chen Q, Xiao S, Wang S, Ge X, Wang Q, Zheng L, Wei Q, Du W, Shen W, Wu Y, Song J. A Salidroside-Based Radiosensitizer Regulates the Nrf2/ROS Pathway for X-Ray Activated Synergistic Cancer Precise Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2413226. [PMID: 40195850 DOI: 10.1002/adma.202413226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/26/2025] [Indexed: 04/09/2025]
Abstract
The hypoxic microenvironment and radioresistance of tumor cells, as well as the delay in efficacy evaluation, significantly limit the effect of clinical radiotherapy. Therefore, developing effective radiosensitizers with monitoring of tumor response is of great significance for precise radiotherapy. Herein, a novel radiosensitizer (term as: SCuFs) is developed, consisting of traditional Chinese medicine (TCM) compounds salidroside, Cu2+, and hydroxyl radical (•OH) activated second near-infrared window fluorescence (NIR-II FL) molecules, which make the radiosensitization effect and boosted chemodynamic therapy (CDT) efficacy. The overexpressed glutathione in the tumor induces the SCuFs dissociation, allowing deep penetration of the drug to the whole tumor region. After X-ray irradiation, salidroside inhibits the Nuclear factor erythroid 2-like 2 (Nrf2)protein expression and blocks cells in the G2/M phase with the highest radiosensitivity, which amplifies the reactive oxygen species (ROS) generation to exacerbate DNA damage, thus achieving radiosensitization. Meanwhile, the upregulated ROS provides sufficient chemical fuel for Cu+-mediated CDT to produce more •OH. NIR-II FL imaging can monitor the •OH changes during the therapy process, confirming the radiosensitization effect and CDT process related to •OH. This study not only achieves effective radiosensitization and cascaded ROS-mediated CDT efficacy, but also provides a useful tool for monitoring therapeutic efficacy, showing great prospects for clinical application.
Collapse
Affiliation(s)
- Qingqing Li
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qing Chen
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Shenggan Xiao
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Shuhan Wang
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoguang Ge
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qian Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Liting Zheng
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qiaoqiao Wei
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wei Du
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Wenbin Shen
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
2
|
Cai Y, Chai T, Nguyen W, Liu J, Xiao E, Ran X, Ran Y, Du D, Chen W, Chen X. Phototherapy in cancer treatment: strategies and challenges. Signal Transduct Target Ther 2025; 10:115. [PMID: 40169560 PMCID: PMC11961771 DOI: 10.1038/s41392-025-02140-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/11/2024] [Accepted: 01/13/2025] [Indexed: 04/03/2025] Open
Abstract
Phototherapy has emerged as a promising modality in cancer treatment, garnering considerable attention for its minimal side effects, exceptional spatial selectivity, and optimal preservation of normal tissue function. This innovative approach primarily encompasses three distinct paradigms: Photodynamic Therapy (PDT), Photothermal Therapy (PTT), and Photoimmunotherapy (PIT). Each of these modalities exerts its antitumor effects through unique mechanisms-specifically, the generation of reactive oxygen species (ROS), heat, and immune responses, respectively. However, significant challenges impede the advancement and clinical application of phototherapy. These include inadequate ROS production rates, subpar photothermal conversion efficiency, difficulties in tumor targeting, and unfavorable physicochemical properties inherent to traditional phototherapeutic agents (PTs). Additionally, the hypoxic microenvironment typical of tumors complicates therapeutic efficacy due to limited agent penetration in deep-seated lesions. To address these limitations, ongoing research is fervently exploring innovative solutions. The unique advantages offered by nano-PTs and nanocarrier systems aim to enhance traditional approaches' effectiveness. Strategies such as generating oxygen in situ within tumors or inhibiting mitochondrial respiration while targeting the HIF-1α pathway may alleviate tumor hypoxia. Moreover, utilizing self-luminescent materials, near-infrared excitation sources, non-photoactivated sensitizers, and wireless light delivery systems can improve light penetration. Furthermore, integrating immunoadjuvants and modulating immunosuppressive cell populations while deploying immune checkpoint inhibitors holds promise for enhancing immunogenic cell death through PIT. This review seeks to elucidate the fundamental principles and biological implications of phototherapy while discussing dominant mechanisms and advanced strategies designed to overcome existing challenges-ultimately illuminating pathways for future research aimed at amplifying this intervention's therapeutic efficacy.
Collapse
Affiliation(s)
- Yeyu Cai
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Tian Chai
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi Province, China
| | - William Nguyen
- School of Chips, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, Taicang, Suzhou, China
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Enhua Xiao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Xin Ran
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuping Ran
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dan Du
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Chen
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi Province, China.
| | - Xiangyu Chen
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
3
|
Zhang K, Wang B, Sun F, Yang Z. Semiconducting Perylene Diimide J-aggregates Cross-linked Hydrogel Enables High-Efficiency Photothermal Controlled Release of Nitric Oxide for Antibiofilm Therapy. Adv Healthc Mater 2025; 14:e2404754. [PMID: 39924765 DOI: 10.1002/adhm.202404754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/18/2025] [Indexed: 02/11/2025]
Abstract
Antibiofilm treatment, particularly drug-containing wound healing dressings, does not typically penetrate the robust protective extracellular polymeric substance of biofilm and eradicate the bacteria. Here, a rational design of nitric oxide (NO) donor N,N'-di-sec-butyl-N,N'-dinitroso-1,4-phenylenediamine (BNN6)-based injectable hydrogel, is reported in which the NO release can be triggered by a photothermal effect owing to semiconducting perylene diimide (PDI) J-aggregation fibers. The synthetic PDI derivatives self-assembling into 0D nanoparticles and then aggregating to 1D J fiber is accompanied by absorbance red-shifting from 700 to 790 nm and then to 852 nm. After encapsulating BNN6, a "sandwich roll" (SR) like structure is evenly crosslinked into an injectable hydrogel (SRH) exhibiting a high photothermal convenience efficiency of 72%, which enables the SRH to achieve highly efficient photocontrol NO release. The SRH shows excellent injectability, shape adaptability, and effective antibacterial efficacy over 99% to the E.coli and S. aureus. and remarkable in vivo antibiofilm efficiency of 99.58% by laser irradiation. Furthermore, the synergistic treatment displays the ability to eliminate inflammation, facilitate angiogenesis, and promote collagen deposition, thereby significantly stimulating the healing process of wounds. The semiconducting J-aggregation injectable hydrogel can be a versatile strategy for the treatment of biofilm.
Collapse
Affiliation(s)
- Kangxin Zhang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Bo Wang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Fengwei Sun
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| |
Collapse
|
4
|
Sangavi R, Jothi R, Malligarjunan N, Raja V, Pandian SK, Gowrishankar S. Cetyltrimethylammonium Chloride (CTAC) and Its Formulated Mouthwash Reduce the Infectivity of Streptococcus mutans and Candida albicans in Mono and Dual State. Appl Biochem Biotechnol 2025; 197:2274-2300. [PMID: 39731640 DOI: 10.1007/s12010-024-05119-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/30/2024]
Abstract
Early childhood caries (ECC), a severe form of dental caries, is exacerbated by the synergistic interaction between Streptococcus mutans and Candida albicans, leading to greater disease severity than their individual effects. This underscores the need for more targeted and potent therapeutic alternatives. Given the promising anti-infective properties of quaternary ammonium surfactants (QAS), this study explores the microbicidal properties of one such QAS, cetyltrimethylammonium chloride (CTAC), against both individual- and dual-species cultures of S. mutans and C. albicans for effective ECC treatment. Initially, the minimal inhibitory concentrations (MICs) of CTAC were determined to range from 4 to 8 µg/mL against S. mutans, C. albicans, and dual-species cultures. Time-kill kinetics, assessed via spot assays and spectrometry, demonstrated that CTAC completely eradicated both individual- and dual-species cultures within 30 min of exposure. Furthermore, at sub-MIC concentrations, CTAC effectively reduced biofilm formation and virulence traits in S. mutans (including acidogenicity and aciduricity) and C. albicans (including yeast-to-hyphal transition and filamentation). To explore therapeutic application, a mouthwash containing CTAC was formulated. The results showed that the formulated CTAC mouthwash was as effective at eradicating pathogens as a commercially available mouthwash containing 0.075% cetylpyridinium chloride (CPC). Moreover, the CTAC mouthwash maintained stable physicochemical characteristics and antimicrobial activity over 4 weeks. It exhibited rapid killing activity against pathogens, achieving efficacy within just 2 min of exposure. Fluorescence microscopy and SEM micrographs confirmed the strong biofilm eradication potential of the CTAC mouthwash. The non-toxic nature of the formulated mouthwash was validated using human buccal epithelial cells, and in vivo studies further demonstrated that CTAC mouthwash significantly reduced bacterial and fungal loads in Galleria mellonella. Overall, the findings of this study highlight the potential application of QAS-CTAC in the treatment of ECC.
Collapse
Affiliation(s)
- Ravichellam Sangavi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, India
| | - Ravi Jothi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, India
| | | | - Veerapandian Raja
- Department of Molecular and Translational Medicine, Center of Emphasis in Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, 79905, USA
| | | | | |
Collapse
|
5
|
Wang S, Li J, Zhang Z, Cao S, Zhang Z, Bian Y, Xu Y, Ma C. Advances in nanomedicine and delivery systems for gastric cancer research. Front Bioeng Biotechnol 2025; 13:1565999. [PMID: 40190709 PMCID: PMC11968739 DOI: 10.3389/fbioe.2025.1565999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
The early diagnosis rate of gastric cancer is low, and most patients are already at an advanced stage by the time they are diagnosed, posing significant challenges for treatment and exhibiting high recurrence rates, which notably diminish patients' survival time and quality of life. Therefore, there is an urgent need to identify methods that can enhance treatment efficacy. Nanomedicine, distinguished by its small size, high targeting specificity, and strong biological compatibility, is particularly well-suited to address the toxic side effects associated with current diagnostic and therapeutic approaches for gastric cancer. Consequently, the application of nanomedicine and delivery systems in the diagnosis and treatment of gastric cancer has garnered increasing interest from researchers. This review provides an overview of recent advancements in the use of nanomaterials as drugs or drug delivery systems in gastric cancer research, encompassing their applications in diagnosis, chemotherapy, radiotherapy, surgery, and phototherapy, and explores the promising prospects of nanomedicine in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Sizhe Wang
- Henan University of Chinese Medicine(The Second Clinical Medical College of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Jilei Li
- Henan Province Hospital of TCM, Zhengzhou(The Second Affiliated Hospital of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Zhenyu Zhang
- Henan University of Chinese Medicine(The Second Clinical Medical College of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Shasha Cao
- Henan University of Chinese Medicine(The Second Clinical Medical College of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Zihan Zhang
- Henan University of Chinese Medicine(The Second Clinical Medical College of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Yifan Bian
- Henan University of Chinese Medicine(The Second Clinical Medical College of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Yanchao Xu
- Henan Province Hospital of TCM, Zhengzhou(The Second Affiliated Hospital of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| | - Chunzheng Ma
- Henan Province Hospital of TCM, Zhengzhou(The Second Affiliated Hospital of Henan University of Chinese Medicine), Zhengzhou, Henan, China
| |
Collapse
|
6
|
Zheng Y, Zhang T, Shao J, Du Y, Li Z, Zhang L, Gao J. Antibiotic-free responsive biomaterials for specific and targeted Helicobacter pylori eradication. J Control Release 2025; 379:708-729. [PMID: 39863021 DOI: 10.1016/j.jconrel.2025.01.054] [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: 08/03/2024] [Revised: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Gastric cancer is highly correlated with Helicobacter pylori (H. pylori) infection. Approximately 50 % of the population worldwide is infected with H. pylori. However, current treatment regimens face severe challenges including drug resistance and gut microbiota disruption. An integrative treatment with slight negative influences on intestinal flora, conforming with concepts of integrative prevention of gastric cancer, is urgently needed. Non-antibiotic responsive biomaterials can respond to different stimuli, including pH, enzymes, light, ultrasound and magnetism, under which biomaterials are specifically activated to perform antibacterial capabilities, while neutral intestinal microenvironments differ from gastric microenvironments or inflammatory sites and have no or minimal irradiation via precisely controlled exogenous stimuli, which may not only overcome antibiotic resistance but also avoid gut microbiota disorders. First, the latest progress in responsive biomaterials against H. pylori without gut microbiome disturbance and their anti-H. pylori performances are profoundly summarized. Second, the mechanisms against planktonic bacteria, biofilms and intracellular bacteria are discussed respectively. Finally, the strategies of specific and targeted H. pylori elimination by responsive biomaterials are introduced. Additionally, the challenges and the focus of future research on translation into clinical application are fully proposed. Antibiotic-free responsive biomaterials for specific and targeted H. pylori eradication represent an innovative approach.
Collapse
Affiliation(s)
- Yating Zheng
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Yangzhou Branch of Jiangsu Provincial Corps of Chinese People's Armed Police Force, Yangzhou 225007, Jiangsu, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Juan Shao
- Yangzhou Branch of Jiangsu Provincial Corps of Chinese People's Armed Police Force, Yangzhou 225007, Jiangsu, China
| | - Yiqi Du
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Li Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China.
| |
Collapse
|
7
|
Zhang S, Wang X, Chen X, Shu D, Lin Q, Zou H, Dong J, Wang B, Tang Q, Li H, Chen X, Pu J, Gu B, Liu P. An on-Demand Oxygen Nano-vehicle Sensitizing Protein and Nucleic Acid Drug Augment Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2409378. [PMID: 39840472 DOI: 10.1002/adma.202409378] [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: 07/01/2024] [Revised: 11/26/2024] [Indexed: 01/23/2025]
Abstract
Hypoxia severely limits the antitumor immunotherapy for breast cancer. Although efforts to alleviate tumor hypoxia and drug delivery using diverse nanostructures achieve promising results, the creation of a versatile controllable oxygen-releasing nano-platform for co-delivery with immunostimulatory molecules remains a persistent challenge. To address this problem, a versatile oxygen controllable releasing vehicle PFOB@F127@PDA (PFPNPs) is developed, which effectively co-delivered either protein drug lactate oxidase (LOX) or nucleic acids drug unmethylated cytosine-phosphate-guanine oligonucleotide (CpG ODNs). Upon photothermal heating, this platform triggered oxygen release, thereby augmenting LOX-mediated lactate detection rates, and improving T cells infiltrating and cytokine expression. Moreover, under an oxygenated tumor microenvironment (TME), PFPNPs co-delivered with CpG ODNs effectively reprogrammed the immunosuppressive TME by repolarizing macrophages to an M1-like phenotype, promoting dendritic cells maturation, and increasing tumor-infiltrating T cells while decreasing the ratio of regulatory T cells (Tregs). Our study demonstrated that this controlled oxygen-releasing platform possessed adaptive drug-loading capabilities to meet varied immunotherapeutic demands in clinical settings.
Collapse
Affiliation(s)
- Sidi Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
- State Key Laboratory for Cancer Systems Regulation and Clinical Translation, Jiading District Central Medicine Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, Shanghai, 201800, P. R. China
| | - Xinghui Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Xiaojing Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
- Central Laboratory Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Duohuo Shu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Quankun Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Hanbing Zou
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
- Central Laboratory Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Jialin Dong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Bing Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Qianyun Tang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
- State Key Laboratory for Cancer Systems Regulation and Clinical Translation, Jiading District Central Medicine Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, Shanghai, 201800, P. R. China
- Central Laboratory Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Huishan Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Xiaoxiang Chen
- Allergy Department Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Jun Pu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Bin Gu
- Department of Urology, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, P. R. China
| | - Peifeng Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute. Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
- State Key Laboratory for Cancer Systems Regulation and Clinical Translation, Jiading District Central Medicine Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, Shanghai, 201800, P. R. China
- Central Laboratory Ren Ji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| |
Collapse
|
8
|
Huang X, Ge W, Li S, Huang R, Wang F. Transferrin-Based Bismuth Nanoparticles for Radiotherapy with Immunomodulation Against Orthotopic Glioma. Adv Healthc Mater 2025; 14:e2404144. [PMID: 39797464 DOI: 10.1002/adhm.202404144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/31/2024] [Indexed: 01/13/2025]
Abstract
Modern radiotherapy frequently employs radiosensitizers for radiation dose deposition and triggers an immunomodulatory effect to enhance tumor destruction. However, developing glioma-targeted sensitizers remains challenging due to the blood-brain barrier (BBB) and multicomponent instability. This study aims to green-synthesize transferrin-bismuth nanoparticles (TBNPs) as biosafe radiosensitizers to enhance X-ray absorption by tumors and stimulate the immune response for glioma therapy. The proposed protein-based strategy provides TBNPs with BBB-crossing ability and prevents off-target toxicity. Cellular experiments following 4 Gy of X-ray irradiation reveal that TBNPs increase DNA damage in glioma cells and trigger immunomodulation, thereby inducing immunogenic cell death. Furthermore, TBNPs effectively inhibit tumor growth through synergistic radiotherapy and immunotherapy in an orthotopic glioma mouse model. The findings highlight TBNPs as promising radiosensitizers for effective and biosafe radiotherapy with immunomodulation.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Wei Ge
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shuxian Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Ruofan Huang
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, 200240, P. R. China
| | - Fu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
9
|
Wu J, Zhao X, Ren B, Duan X, Sun J. Radioresistance in Hepatocellular Carcinoma: Biological Bases and Therapeutic Implications. Int J Mol Sci 2025; 26:1839. [PMID: 40076465 PMCID: PMC11899467 DOI: 10.3390/ijms26051839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Radiotherapy technology is a common treatment modality that can be used in all stages of HCC. However, in some cases, radiotherapy fails in clinical practice mainly because of the patient's resistance to radiotherapy, creating a bottleneck for future breakthroughs. HCC radiosensitivity is primarily related to DNA double-strand break repair, cellular autophagy, cell cycle, cellular metabolism, and hypoxic environmental regulators. Therefore, a comprehensive understanding of its molecular mechanisms will be of immense importance in reversing HCC radioresistance. In this review, we provide a comprehensive overview of the mechanism of action of radiotherapy on HCC, the cellular and molecular basis of radiation resistance in HCC, related treatment modalities, and future prospects.
Collapse
Affiliation(s)
- Jianhui Wu
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| | - Xiaofang Zhao
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| | - Bowen Ren
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| | - Xuezhang Duan
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
| | - Jing Sun
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| |
Collapse
|
10
|
Fu H, Xie Y, Ren S, Zhang Q, Cheng J, Liang Q, Xiao X. Multifunctional Cu 3BiS 3-BP@PEI Radiosensitizer with Enhanced Reactive Oxygen Species Activity for Multimodal Synergistic Therapy. ACS Biomater Sci Eng 2025; 11:930-941. [PMID: 39792365 DOI: 10.1021/acsbiomaterials.4c01907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Development of radiosensitizers with high-energy deposition efficiency, electron transfer, and oxidative stress amplification will help to improve the efficiency of radiotherapy. To overcome the drawbacks of radiotherapy alone, it is also crucial to design a multifunctional radiosensitizer that simultaneously realizes multimodal treatment and tumor microenvironment modulation. Herein, a multifunctional radiosensitizer based on the Cu3BiS3-BP@PEI nanoheterostructure (NHS) for multimodal cancer treatment is designed. Cu3BiS3-BP@PEI NHS is able to deposit a high radiation dose into cancer cells, enhancing the radiotherapy effect. Due to the heterostructure and the synergistic effect of Cu3BiS3 and black phosphorus (BP), significantly boosted 1O2 and •OH generation is obtained under X-ray irradiation, which is promising for extremely efficient radiodynamic therapy. More importantly, the acidic tumor microenvironment (TME) can induce the cycle conversion of Cu2+ to Cu+, oxidizing glutathione (GSH) and catalyzing intracellular overproduction of H2O2 into highly toxic •OH, which thus further enhances reactive oxygen species (ROS) production and reduces GSH-associated radioresistance. Furthermore, Cu3BiS3-BP@PEI NHS has an excellent photothermal effect and can effectively transform light into heat. The outcomes of the in vitro and in vivo research confirm that the as-prepared Cu3BiS3-BP@PEI NHS has a high synergistic therapeutic efficacy at a low radiation dose. This work provides a viable approach to constructing a multifunctional radiosensitizer for deep tumor treatment with TME-triggered multiple synergistic therapies.
Collapse
Affiliation(s)
- Hanping Fu
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Yan Xie
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Shufen Ren
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Qing Zhang
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Jiayun Cheng
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Qingshuang Liang
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Xiufeng Xiao
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| |
Collapse
|
11
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [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: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
12
|
Huang Y, Zhang C, Zhang L, Chen X, Fan W. Chemical Synthesis and Multihybridization of Small-Sized Hollow Mesoporous Organosilica Nanoparticles Toward Advanced Theranostics. Acc Chem Res 2024; 57:3465-3477. [PMID: 39576957 DOI: 10.1021/acs.accounts.4c00502] [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: 11/24/2024]
Abstract
As one of the most widely used nanomaterials, mesoporous silica nanoparticles (MSNs) have received extensive attraction due to their desirable physicochemical performances of high stability, large surface area, and tunable pore sizes. Besides, the U.S. Food and Drug Administration (FDA) has recognized that silica-based nanoparticles are generally safe for biomedical applications. However, the poor biodegradation and inert Si-O-Si framework of inorganic MSNs severely impair their diverse biomedical applications. A promising strategy to improve the physicochemical properties of MSNs is the incorporation of functional organic moieties into their framework to construct mesoporous organosilica nanoparticles (MONs), which exhibit distinct advantages over traditional inorganic MSNs, such as adjustable organosilica framework, excellent biocompatibility, stimuli-responsive biodegradability, and even improved therapeutic effects. Moreover, the emerging hollow-structured MONs (HMONs) with an internal cavity can offer a large drug loading capacity and thus become increasingly attractive and promising theranostic nanoplatforms in biomedicine. In recent years, numerous studies have delved into establishing multifunctional HMONs with sizes ranging in diameters from 50 to 200 nm for desirable biological responses. With the gradual deepening of research, small-sized HMONs with diameters below 50 nm (sub-50 nm HMONs) demonstrate unparalleled advantages in extending blood circulation time, reducing the risk of vascular occlusion, and achieving high tumor accumulation, thus leading to a growing interest in the design, development, and translation of sub-50 nm HMONs. However, the mechanism of the chemical synthesis and structural regulation of sub-50 nm HMONs is still unclear, which is detrimental to further structural hybridization and surface functionalization. In this account, we will focus on the structural design, chemical synthesis, adjustable framework hybridization, multifunctional surface modification, and versatile biomedical applications of small-sized HMONs. First, we will illustrate the chemical approaches for controllable synthesis of HMONs and the underlying mechanism of particle size regulation below 50 nm. Subsequently, the basic principles and design strategies of multihybridization of sub-50 nm HMONs based on framework hybridization, surface modulation, and in situ polymerization will be systematically discussed. Through diverse functionalization strategies, a series of sub-50 nm multihybridized HMONs-based nanotheranostics are established, and their applications in multimodal biomedical imaging and highly efficient synergistic treatment of various diseases (e.g., cancer, glaucoma, bacterial infection, etc.) will be accounted. Finally, we will summarize the current status and potential challenges of HMONs in clinic trials, as well as provide a comprehensive outlook on the future development of sub-50 nm HMONs. These innovative sub-50 nm HMONs hold the potential to introduce novel theranostic modalities for a variety of systemic disorders and to advance smart promising nanomedicine in the near future.
Collapse
Affiliation(s)
- Yuhang Huang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Cheng Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Liyuan Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, Singapore 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
13
|
Meng W, Huang L, Guo J, Xin Q, Liu J, Hu Y. Innovative Nanomedicine Delivery: Targeting Tumor Microenvironment to Defeat Drug Resistance. Pharmaceutics 2024; 16:1549. [PMID: 39771528 PMCID: PMC11728492 DOI: 10.3390/pharmaceutics16121549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/24/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
Nanodrug delivery systems have revolutionized tumor therapy like never before. By overcoming the complexity of the tumor microenvironment (TME) and bypassing drug resistance mechanisms, nanotechnology has shown great potential to improve drug efficacy and reduce toxic side effects. This review examines the impact of the TME on drug resistance and recent advances in nanomedicine delivery systems to overcome this challenge. Characteristics of the TME such as hypoxia, acidity, and high interstitial pressure significantly reduce the effectiveness of chemotherapy and radiotherapy, leading to increased drug resistance in tumor cells. Then, this review summarizes innovative nanocarrier designs for these microenvironmental features, including hypoxia-sensitive nanoparticles, pH-responsive carriers, and multifunctional nanosystems that enable targeted drug release and improved drug penetration and accumulation in tumors. By combining nanotechnology with therapeutic strategies, this review offers a novel perspective by focusing on the innovative design of nanocarriers that interact with the TME, a dimension often overlooked in similar reviews. We highlight the dual role of these nanocarriers in therapeutic delivery and TME modulation, emphasize their potential to overcome drug resistance, and look at future research directions.
Collapse
Affiliation(s)
- Wenjun Meng
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China (J.L.)
| | - Li Huang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China (J.L.)
| | - Jiamin Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Xin
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China (J.L.)
| | - Yuzhu Hu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Zhang J, Liu Z, Zhang Z, Yang H, Wang H, Yang Z, Xu Y, Li S, Yang D. Recent Advances in Silica-Based Nanomaterials for Enhanced Tumor Imaging and Therapy. ACS APPLIED BIO MATERIALS 2024; 7:7133-7169. [PMID: 39495482 DOI: 10.1021/acsabm.4c01318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Cancer remains a formidable challenge, inflicting profound physical, psychological, and financial burdens on patients. In this context, silica-based nanomaterials have garnered significant attention for their potential in tumor imaging and therapy owing to their exceptional properties, such as biocompatibility, customizable porosity, and versatile functionalization capabilities. This review meticulously examines the latest advancements in the application of silica-based nanomaterials for tumor imaging and therapy. It underscores their potential in enhancing various cancer imaging modalities, including fluorescence imaging, magnetic resonance imaging, computed tomography, positron emission tomography, ultrasound imaging, and multimodal imaging approaches. Moreover, the review delves into their therapeutic efficacy in chemotherapy, radiotherapy, phototherapy, immunotherapy, gas therapy, sonodynamic therapy, chemodynamic therapy, starvation therapy, and gene therapy. Critical evaluations of the biosafety profiles and degradation pathways of these nanomaterials within biological environments are also presented. By discussing the current challenges and prospects, this review aims to provide a nuanced perspective on the clinical translation of silica-based nanomaterials, thereby highlighting their promise in revolutionizing cancer diagnostics, enabling real-time monitoring of therapeutic responses, and advancing personalized medicine.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zilu Liu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhijing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhenlu Yang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Yunjian Xu
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an 271000, China
- Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Shengke Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| |
Collapse
|
15
|
Qiao K, Pan Y, Zhang S, Shi G, Yang J, Zhang Z, Wang K, Chen X, Ning S. Cold Exposure Therapy Sensitizes Nanodrug-Mediated Radioimmunotherapy of Breast Cancer. ACS NANO 2024; 18:29689-29703. [PMID: 39401104 DOI: 10.1021/acsnano.4c09021] [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: 10/30/2024]
Abstract
Cold exposure (CE) therapy can quickly induce tumor starvation by brown adipose tissue (BAT) thermogenesis. Exploring the combined antitumor mechanism of CE and traditional therapies (such as radiotherapy (RT)) is exciting and promising. In this study, we investigated the effect of CE in combination with nitric oxide (NO) gas therapy on sensitizing tumors to RT and promoting tumor radio-immunotherapy. We first constructed a liposome (SL) loaded with the NO prodrug S-nitroso-N-acetylpenicillamine (SNAP). When SL is injected, the glutathione (GSH) within the tumor region promotes the release of NO from SNAP. Subsequently, the superoxide anion produced by RT reacts with NO to generate peroxynitrite (ONOO-), which has strong oxidative properties and induces cell death. Meanwhile, the mice were exposed to a CE environment of 4 °C. CE-mediated BAT thermogenesis induced tumor starvation, which led to a decrease in ATP and GSH content within the tumor as well as an improvement in the hypoxic microenvironment and a decrease in myeloid-derived suppressor cells. All of the above have promoted the effectiveness of RT and activated the systemic antitumor immunity. In the bilateral tumor experiment, treatment of the primary tumor inhibited the growth of the distant tumor and promoted the infiltration of CD8+ T cells into the tumor. These findings reveal that the synergy of CE, NO gas therapy, and RT could confer high effective anticancer effects, providing possibilities in personalized cancer treatment.
Collapse
Affiliation(s)
- Kun Qiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - You Pan
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Guangfu Shi
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Jinglin Yang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Zhenlin Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| |
Collapse
|
16
|
Song J, Feng Y, Yan J, Wang Y, Yan W, Yang N, Wu T, Liu S, Wang Y, Zheng N, He L, Zhang Y. Computed Tomography Imaging Guided Microenvironment-Responsive Ir@WO 3-x Dual-Catalytic Nanoreactor for Selective Radiosensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405192. [PMID: 39102342 PMCID: PMC11481196 DOI: 10.1002/advs.202405192] [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: 05/13/2024] [Revised: 07/28/2024] [Indexed: 08/07/2024]
Abstract
Radiotherapy (RT) is often administered, either alone or in combination with other therapies, for most malignancies. However, the degree of tumor oxygenation, damage to adjacent healthy tissues, and inaccurate guidance remain issues that result in discontinuation or failure of RT. Here, a multifunctional therapeutic platform based on Ir@WO3-x is developed which simultaneously addresses these critical issues above for precision radiosensitization. Ir@WO3-x nanoreactors exhibit strong absorption of X-ray, acting as radiosensitizers. Moreover, ultrasmall Ir enzyme-mimic nanocrystals (NCs) are decorated onto the surface of the nanoreactor, where NCs have catalyst-like activity and are sensitive to H2O2 in the tumor microenvironment (TME) under near infrared-II (NIR-II) light stimulation. They efficiently catalyze the conversion of H2O2 to O2, thereby ameliorating hypoxia, inhibiting the expression of HIF-1α, and enhancing RT-induced DNA damage in cancerous tissue, further improving the efficiency of RT. Additionally, in response to high H2O2 levels in TME, the Ir@WO3-x nanoreactor also exerts peroxidase-like activity, boosting exogenous ROS, which increases oxidative damage and enhances ROS-dependent death signaling. Furthermore, Ir@WO3-x can serve as a high-quality computed tomography contrast agent due to its high X-ray attenuation coefficient and generation of pronounced tumor-tissue contrast. This report highlights the potential of advanced health materials to enhance precision therapeutic modalities.
Collapse
Affiliation(s)
- Jiayu Song
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
| | - Yue Feng
- Department of Gynecological OncologyZhejiang Cancer HospitalZhengzhouZhejiang310022China
| | - Jiazhuo Yan
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Ying Wang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Weixiao Yan
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
| | - Nan Yang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Tusheng Wu
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Sijia Liu
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Yuan Wang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Nannan Zheng
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhouHenan450000China
| | - Liangcan He
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhouHenan450000China
| | - Yunyan Zhang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| |
Collapse
|
17
|
Dong Y, Tao J, Wang B, Zhang A, Xiang G, Li S, Jiang T, Zhao X. Partitioned Microneedle Patch Based on NO Release and HSP Inhibition for mPTT/GT Combination Treatment of Melanoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:49104-49113. [PMID: 39234752 DOI: 10.1021/acsami.4c10141] [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: 09/06/2024]
Abstract
Photothermal therapy (PTT) shows promise in cancer treatments due to its good spatiotemporal selectivity and minimal invasiveness. However, PTT has some problems such as excessive heat damage to normal tissues, tumor thermo-resistance caused by heat shock proteins (HSPs), and limited efficacy of monotherapy. Here, we construct a patch named "partitioned microneedles" (PMN-SNAP/CuS), which separates the "catalyst" bovine serum albumin-based copper sulfide nanoparticles (CuS@BSA NPs) and the "reactant" S-nitroso-N-acetylpenicillamine (SNAP) into different regions of microneedles, for enhancing mild PTT (mPTT) of melanoma. PMN-SNAP/CuS showed an excellent photothermal effect, Fenton-like catalytic activity, and nitric oxide (NO) generation ability. The combination of NO and reactive oxygen species (ROS) produced by PMN-SNAP/CuS effectively blocked the synthesis of HSPs at the source and enhanced the efficacy of mPTT. Both in vitro and in vivo results proved that PMN-SNAP/CuS significantly enhanced the inhibition of melanoma under 808 nm laser irradiation. In conclusion, our partitioned microneedle strategy based on the combination of enhanced mPTT and gas therapy (GT) provides a promising approach to enhance the therapeutic effect on melanoma.
Collapse
Affiliation(s)
- Yu Dong
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jiaojiao Tao
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Bingjie Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Aijia Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Guangli Xiang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Shuang Li
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Tianze Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Xia Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
18
|
Lu F, Li L, Zhang M, Yu C, Pan Y, Cheng F, Hu W, Lu X, Wang Q, Fan Q. Confined semiconducting polymers with boosted NIR light-triggered H 2O 2 production for hypoxia-tolerant persistent photodynamic therapy. Chem Sci 2024; 15:12086-12097. [PMID: 39092116 PMCID: PMC11290442 DOI: 10.1039/d4sc01609b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/11/2024] [Indexed: 08/04/2024] Open
Abstract
Hypoxia featured in malignant tumors and the short lifespan of photo-induced reactive oxygen species (ROS) are two major issues that limit the efficiency of photodynamic therapy (PDT) in oncotherapy. Developing efficient type-I photosensitizers with long-term ˙OH generation ability provides a possible solution. Herein, a semiconducting polymer-based photosensitizer PCPDTBT was found to generate 1O2, ˙OH, and H2O2 through type-I/II PDT paths. After encapsulation within a mesoporous silica matrix, the NIR-II fluorescence and ROS generation are enhanced by 3-4 times compared with the traditional phase transfer method, which can be attributed to the excited-state lifetime being prolonged by one order of magnitude, resulting from restricted nonradiative decay channels, as confirmed by femtosecond spectroscopy. Notably, H2O2 production reaches 15.8 μM min-1 under a 730 nm laser (80 mW cm-2). Further adsorption of Fe2+ ions on mesoporous silica not only improves the loading capacity of the chemotherapy drug doxorubicin but also triggers a Fenton reaction with photo-generated H2O2 in situ to produce ˙OH continuously after the termination of laser irradiation. Thus, semiconducting polymer-based nanocomposites enables NIR-II fluorescence imaging guided persistent PDT under hypoxic conditions. This work provides a promising paradigm to fabricate persistent photodynamic therapy platforms for hypoxia-tolerant phototheranostics.
Collapse
Affiliation(s)
- Feng Lu
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| | - Lili Li
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| | - Meng Zhang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| | - Chengwu Yu
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| | - Yonghui Pan
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| | - Fangfang Cheng
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Wenbo Hu
- Institute of Flexible Electronics (IFE), Northwestern Polytechnical University Xi'an 710072 China
| | - Xiaomei Lu
- Institute of Advanced Materials (IAM) & School of Flexible Electronics (Future Technologies), Nanjing Tech University Nanjing 211816 China
- Zhengzhou Institute of Biomedical Engineering and Technology Zhengzhou 450001 China
| | - Qi Wang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| | - Quli Fan
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications Nanjing 210023 China
| |
Collapse
|
19
|
Zhou Z, Jiang X, Yi L, Li C, Wang H, Xiong W, Li Z, Shen J. Mitochondria Energy Metabolism Depression as Novel Adjuvant to Sensitize Radiotherapy and Inhibit Radiation Induced-Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401394. [PMID: 38715382 PMCID: PMC11234447 DOI: 10.1002/advs.202401394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Indexed: 07/11/2024]
Abstract
Currently, the typical combination therapy of programmed death ligand-1 (PD-L1) antibodies with radiotherapy (RT) still exhibits impaired immunogenic antitumor response in clinical due to lessened DNA damage and acquired immune tolerance via the upregulation of some other immune checkpoint inhibitors. Apart from this, such combination therapy may raise the occurrence rate of radiation-induced lung fibrosis (RIPF) due to enhanced systemic inflammation, leading to the ultimate death of cancer patients (average survival time of about 3 years). Therefore, it is newly revealed that mitochondria energy metabolism regulation can be used as a novel effective PD-L1 and transforming growth factor-β (TGF-β) dual-downregulation method. Following this, IR-TAM is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with oxidative phosphorylation (OXPHOS) inhibitor Tamoxifen (TAM), which then self-assembled with albumin (Alb) to form IR-TAM@Alb nanoparticles. By doing this, tumor-targeting IR-TAM@Alb nanoparticle effectively reversed tumor hypoxia and depressed PD-L1 and TGF-β expression to sensitize RT. Meanwhile, due to the capacity of heptamethine cyanine dye in targeting RIPF and the function of TAM in depressing TGF-β, IR-TAM@Alb also ameliorated fibrosis development induced by RT.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lei Yi
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Cheng Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wei Xiong
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Zhipeng Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
20
|
Yao Y, Zheng Y, Wu M, Gao Y, Yu Q, Liu M, Luo X, Wang R, Jiang L. CD133-targeted multifunctional nanomicelles for dual-modality imaging and synergistic high-intensity focus ultrasound (HIFU) ablation on pancreatic cancer in nude mice. J Mater Chem B 2024; 12:5884-5897. [PMID: 38775254 DOI: 10.1039/d4tb00091a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Pancreatic cancer is an aggressive and highly fatal malignant tumor. Recent studies have shown that cancer stem cells (CSCs) play an important role in resisting current therapeutic modalities. Furthermore, CD133 is highly expressed in CSCs. High-intensity focused ultrasound (HIFU) is a promising non-invasive therapeutic strategy for unresectable pancreatic cancers. In our study, we synthesized targeted CD133 organosilane nanomicelles by encapsulating perfluorohexane (PFH). The CD133 antibody on the surface could specifically bind to CD133-positive pancreatic cancer cells and selectively concentrate in pancreatic cancer tumor tissues. PFH was introduced to improve the ablation effect of HIFU due to its liquid-gas phase transition properties. By combining with the dorsal skinfold window chamber model (DSWC) of pancreatic cancer in nude mice, multiphoton fluorescence microscopy was used to evaluate the targeting effect of nanomicelles on pancreatic cancer tumor tissue. These multifunctional nanomicelles synergistically affected HIFU treatment of pancreatic cancer, providing an integrated research platform for diagnosing and treating pancreatic cancer with HIFU.
Collapse
Affiliation(s)
- Yijing Yao
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Yiwen Zheng
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Mingtai Wu
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Yihui Gao
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Qian Yu
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Mengyao Liu
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Xiaoxiao Luo
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Rui Wang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Lixin Jiang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
21
|
Sun H, Bai Y, Zhao D, Wang J, Qiu L. Transition-Metal-Oxide-Based Nanozymes for Antitumor Applications. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2896. [PMID: 38930266 PMCID: PMC11205014 DOI: 10.3390/ma17122896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Transition metal oxide (TMO)-based nanozymes have appeared as hopeful tools for antitumor applications due to their unique catalytic properties and ability to modulate the tumor microenvironment (TME). The purpose of this review is to provide an overview of the latest progress made in the field of TMO-based nanozymes, focusing on their enzymatic activities and participating metal ions. These nanozymes exhibit catalase (CAT)-, peroxidase (POD)-, superoxide dismutase (SOD)-, oxidase (OXD)-, and glutathione oxidase (GSH-OXD)-like activities, enabling them to regulate reactive oxygen species (ROS) levels and glutathione (GSH) concentrations within the TME. Widely studied transition metals in TMO-based nanozymes include Fe, Mn, Cu, Ce, and the hybrid multimetallic oxides, which are also summarized. The review highlights several innovative nanozyme designs and their multifunctional capabilities. Despite the significant progress in TMO-based nanozymes, challenges such as long-term biosafety, targeting precision, catalytic mechanisms, and theoretical supports remain to be addressed, and these are also discussed. This review contributes to the summary and understanding of the rapid development of TMO-based nanozymes, which holds great promise for advancing nanomedicine and improving cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| |
Collapse
|
22
|
Zou J, Li Z, Zhu Y, Tao Y, You Q, Cao F, Wu Q, Wu M, Cheng J, Zhu J, Chen X. pH/GSH dual responsive nanosystem for nitric oxide generation enhanced type I photodynamic therapy. Bioact Mater 2024; 34:414-421. [PMID: 38292411 PMCID: PMC10825229 DOI: 10.1016/j.bioactmat.2023.12.023] [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: 10/07/2023] [Revised: 12/08/2023] [Accepted: 12/25/2023] [Indexed: 02/01/2024] Open
Abstract
Tumor hypoxia diminishes the effectiveness of traditional type II photodynamic therapy (PDT) due to oxygen consumption. Type I PDT, which can operate independently of oxygen, is a viable option for treating hypoxic tumors. In this study, we have designed and synthesized JSK@PEG-IR820 NPs that are responsive to the tumor microenvironment (TME) to enhance type I PDT through glutathione (GSH) depletion. Our approach aims to expand the sources of therapeutic benefits by promoting the generation of superoxide radicals (O2-.) while minimizing their consumption. The diisopropyl group within PEG-IR820 serves a dual purpose: it functions as a pH sensor for the disassembly of the NPs to release JSK and enhances intermolecular electron transfer to IR820, facilitating efficient O2-. generation. Simultaneously, the release of JSK leads to GSH depletion, resulting in the generation of nitric oxide (NO). This, in turn, contributes to the formation of highly cytotoxic peroxynitrite (ONOO-.), thereby enhancing the therapeutic efficacy of these NPs. NIR-II fluorescence imaging guided therapy has achieved successful tumor eradication with the assistance of laser therapy.
Collapse
Affiliation(s)
- Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zheng Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yang Zhu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yucen Tao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Qinghe Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Min Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, PR China
| | - Junjie Cheng
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Department of Chemistry Center for Bioanalytical Chemistry, University of Science and Technology of China, Hefei, 230026, PR China
| | - Jianwei Zhu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- College of Life Science, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, PR China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
23
|
Song Y, Zou J, Castellanos EA, Matsuura N, Ronald JA, Shuhendler A, Weber WA, Gilad AA, Müller C, Witney TH, Chen X. Theranostics - a sure cure for cancer after 100 years? Theranostics 2024; 14:2464-2488. [PMID: 38646648 PMCID: PMC11024861 DOI: 10.7150/thno.96675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
Cancer has remained a formidable challenge in medicine and has claimed an enormous number of lives worldwide. Theranostics, combining diagnostic methods with personalized therapeutic approaches, shows huge potential to advance the battle against cancer. This review aims to provide an overview of theranostics in oncology: exploring its history, current advances, challenges, and prospects. We present the fundamental evolution of theranostics from radiotherapeutics, cellular therapeutics, and nanotherapeutics, showcasing critical milestones in the last decade. From the early concept of targeted drug delivery to the emergence of personalized medicine, theranostics has benefited from advances in imaging technologies, molecular biology, and nanomedicine. Furthermore, we emphasize pertinent illustrations showcasing that revolutionary strategies in cancer management enhance diagnostic accuracy and provide targeted therapies customized for individual patients, thereby facilitating the implementation of personalized medicine. Finally, we describe future perspectives on current challenges, emerging topics, and advances in the field.
Collapse
Affiliation(s)
- Yangmeihui Song
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, 81675, Germany
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | | | - Naomi Matsuura
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Materials Science & Engineering, University of Toronto, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - John A. Ronald
- Imaging Laboratories, Department of Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Adam Shuhendler
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Assaf A. Gilad
- Department of Chemical Engineering and Materials Sciences, Michigan State University, East Lansing, MI, USA
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Timothy H. Witney
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
24
|
Zhang JA, Haddleton D, Wilson P, Zhu LH, Dai CY, Zhao LL. pH-Responsive Amphiphilic Triblock Fluoropolymers as Assemble Oxygen Nanoshuttles for Enhancing PDT against Hypoxic Tumor. Bioconjug Chem 2024; 35:400-411. [PMID: 38366969 DOI: 10.1021/acs.bioconjchem.4c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
Photodynamic therapy (PDT) is a cancer treatment strategy that utilizes photosensitizers to convert oxygen within tumors into reactive singlet oxygen (1O2) to lyse tumor cells. Nevertheless, pre-existing tumor hypoxia and oxygen consumption during PDT can lead to an insufficient oxygen supply, potentially reducing the photodynamic efficacy. In response to this issue, we have devised a pH-responsive amphiphilic triblock fluorinated polymer (PDP) using copper-mediated RDRP. This polymer, composed of poly(ethylene glycol) methyl ether acrylate, 2-(diethylamino)ethyl methacrylate, and (perfluorooctyl)ethyl acrylate, self-assembles in an aqueous environment. Oxygen, chlorine e6 (Ce6), and doxorubicin (DOX) can be codelivered efficiently by PDP. The incorporation of perfluorocarbon into the formulation enhances the oxygen-carrying capacity of PDP, consequently extending the lifetime of 1O2. This increased lifetime, in turn, amplifies the PDT effect and escalates the cellular cytotoxicity. Compared with PDT alone, PDP@Ce6-DOX-O2 NPs demonstrated significant inhibition of tumor growth. This study proposes a novel strategy for enhancing the efficacy of PDT.
Collapse
Affiliation(s)
- Jun-An Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - David Haddleton
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Paul Wilson
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Lin-Hua Zhu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Key Laboratory of Functional Organic Polymers of Haikou, Tropical Functional Polymer Materials Engineering Research Center of Hainan, Haikou 571158, China
| | - Chun-Yan Dai
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Key Laboratory of Functional Organic Polymers of Haikou, Tropical Functional Polymer Materials Engineering Research Center of Hainan, Haikou 571158, China
| | - Lin-Lu Zhao
- College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
25
|
Wang L, Zhou H, Chen Q, Lin Z, Jiang C, Chen X, Chen M, Liu L, Shao L, Liu X, Pan J, Wu J, Song J, Wu J, Zhang D. STING Agonist-Loaded Nanoparticles Promotes Positive Regulation of Type I Interferon-Dependent Radioimmunotherapy in Rectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307858. [PMID: 38063844 PMCID: PMC10870073 DOI: 10.1002/advs.202307858] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/14/2023] [Indexed: 02/17/2024]
Abstract
Hypoxia-associated radioresistance in rectal cancer (RC) has severely hampered the response to radioimmunotherapy (iRT), necessitating innovative strategies to enhance RC radiosensitivity and improve iRT efficacy. Here, a catalytic radiosensitizer, DMPtNPS, and a STING agonist, cGAMP, are integrated to overcome RC radioresistance and enhance iRT. DMPtNPS promotes efficient X-ray energy transfer to generate reactive oxygen species, while alleviating hypoxia within tumors, thereby increasing radiosensitivity. Mechanistically, the transcriptomic and immunoassay analysis reveal that the combination of DMPtNPS and RT provokes bidirectional regulatory effects on the immune response, which may potentially reduce the antitumor efficacy. To mitigate this, cGAMP is loaded into DMPtNPS to reverse the negative impact of DMPtNPS and RT on the tumor immune microenvironment (TiME) through the type I interferon-dependent pathway, which promotes cancer immunotherapy. In a bilateral tumor model, the combination treatment of RT, DMPtNPS@cGAMP, and αPD-1 demonstrates a durable complete response at the primary site and enhanced abscopal effect at the distant site. This study highlights the critical role of incorporating catalytic radiosensitizers and STING agonists into the iRT approach for RC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
- Department of Oncologythe Second Affiliated Hospital of Nanchang UniversityNanchang360000P. R. China
| | - Han Zhou
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenGuangdong518053P. R. China
| | - Qingjing Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Department of Hepatopancreatobiliary SurgeryFirst Affiliated Hospital of Fujian Medical UniversityFuzhou350004P.R. China
| | - Zhiwen Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Department of Hepatopancreatobiliary SurgeryFirst Affiliated Hospital of Fujian Medical UniversityFuzhou350004P.R. China
| | - Chenwei Jiang
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Xingte Chen
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Mingdong Chen
- Department of Radiation OncologyMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Libin Liu
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Lingdong Shao
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- CAS Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Jianji Pan
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Jingcheng Wu
- Department of Health ScienceTechnology and EducationNational Health Commission of the People's Republic of ChinaBeijing100088China
| | - Jibin Song
- State Key Laboratory of Chemical Resource EngineeringCollege of ChemistryBeijing University of Chemical TechnologyBeijing10010P. R. China
| | - Junxin Wu
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| |
Collapse
|
26
|
Yan Z, Liu Z, Zhang H, Guan X, Xu H, Zhang J, Zhao Q, Wang S. Current trends in gas-synergized phototherapy for improved antitumor theranostics. Acta Biomater 2024; 174:1-25. [PMID: 38092250 DOI: 10.1016/j.actbio.2023.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
Phototherapy, such as photothermal therapy (PTT) and photodynamic therapy (PDT), has been considered an elegant solution to eradicate tumors due to its minimal invasiveness and low systemic toxicity. Nevertheless, it is still challenging for phototherapy to achieve ideal outcomes and clinical translation due to its inherent drawbacks. Owing to the unique biological functions, diverse gases have attracted growing attention in combining with phototherapy to achieve super-additive therapeutic effects. Specifically, gases such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) have been proven to kill tumor cells by inducing mitochondrial damage in synergy with phototherapy. Additionally, several gases not only enhance the thermal damage in PTT and the reactive oxygen species (ROS) production in PDT but also improve the tumor accumulation of photoactive agents. The inflammatory responses triggered by hyperthermia in PTT are also suppressed by the combination of gases. Herein, we comprehensively review the latest studies on gas-synergized phototherapy for cancer therapy, including (1) synergistic mechanisms of combining gases with phototherapy; (2) design of nanoplatforms for gas-synergized phototherapy; (3) multimodal therapy based on gas-synergized phototherapy; (4) imaging-guided gas-synergized phototherapy. Finally, the current challenges and future opportunities of gas-synergized phototherapy for tumor treatment are discussed. STATEMENT OF SIGNIFICANCE: 1. The novelty and significance of the work with respect to the existing literature. (1) Strategies to design nanoplatforms for gas-synergized anti-tumor phototherapy have been summarized for the first time. Meanwhile, the integration of various imaging technologies and therapy modalities which endow these nanoplatforms with advanced theranostic capabilities has been summarized. (2) The mechanisms by which gases synergize with phototherapy to eradicate tumors are innovatively and comprehensively summarized. 2. The scientific impact and interest. This review elaborates current trends in gas-synergized anti-tumor phototherapy, with special emphases on synergistic anti-tumor mechanisms and rational design of therapeutic nanoplatforms to achieve this synergistic therapy. It aims to provide valuable guidance for researchers in this field.
Collapse
Affiliation(s)
- Ziwei Yan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Zhu Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Haotian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Xinyao Guan
- Experimental Teaching Center, Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Hongwei Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jinghai Zhang
- Department of Biomedical Engineering, School of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
27
|
He S, Gou X, Zhang S, Zhang X, Huang H, Wang W, Yi L, Zhang R, Duan Z, Zhou P, Qian Z, Gao X. Nanodelivery Systems as a Novel Strategy to Overcome Treatment Failure of Cancer. SMALL METHODS 2024; 8:e2301127. [PMID: 37849248 DOI: 10.1002/smtd.202301127] [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: 08/24/2023] [Revised: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Despite the tremendous progress in cancer treatment in recent decades, cancers often become resistant due to multiple mechanisms, such as intrinsic or acquired multidrug resistance, which leads to unsatisfactory treatment effects or accompanying metastasis and recurrence, ultimately to treatment failure. With a deeper understanding of the molecular mechanisms of tumors, researchers have realized that treatment designs targeting tumor resistance mechanisms would be a promising strategy to break the therapeutic deadlock. Nanodelivery systems have excellent physicochemical properties, including highly efficient tissue-specific delivery, substantial specific surface area, and controllable surface chemistry, which endow nanodelivery systems with capabilities such as precise targeting, deep penetration, responsive drug release, multidrug codelivery, and multimodal synergy, which are currently widely used in biomedical researches and bring a new dawn for overcoming cancer resistance. Based on the mechanisms of tumor therapeutic resistance, this review summarizes the research progress of nanodelivery systems for overcoming tumor resistance to improve therapeutic efficacy in recent years and offers prospects and challenges of the application of nanodelivery systems for overcoming cancer resistance.
Collapse
Affiliation(s)
- Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xinyu Gou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Shuheng Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Hongyi Huang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Rui Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhongxin Duan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Peizhi Zhou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| |
Collapse
|
28
|
Cai J, Hu G, Hu L, Chen J, Chen D, Liu D, Wang X, Hu B, Li C. A CaCO 3-based nanoplatform with sonodynamic and tumor microenvironment activated for combined in vitro cancer therapy. Transl Oncol 2023; 38:101771. [PMID: 37729741 PMCID: PMC10518365 DOI: 10.1016/j.tranon.2023.101771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 09/22/2023] Open
Abstract
INTRODUCTION Sonodynamic therapy (SDT) has potential clinical applications for cancer therapy, and is yet restricted by complex tumor microenvironmental (TME) factors. Thus, the research problem of TME modulation as well as efficient tumor treatment still needs to be clarified. METHOD In this study, a calcium carbonate (CaCO3) nanoplatform was designed for ultrasound (US) and TME response-triggered, which encapsulated Ag2S and loaded with l-Arg, and further wrapped with RBC/Platelet membrane, named as QD@Ca/ML-Arg. RESULTS Non-invasive US-triggered SDT by Ag2S and acidic environment-responsive drug release were achieved. In vitro experiments validated the efficacy of SDT, Ca-ion interference and nitric oxide (NO) gas therapy as combined therapy for cancer treatment. By means of RNA sequencing, the cancer therapeutic mechanism of SDT in redox-related pathways was elucidated. The immunosuppressive TME was simulated with a M2-macrophage/cancer cell co-culture system to confirm the immune activating effect of immunogenic cell death (ICD). CONCLUSION Accordingly, the potential of QD@Ca/ML-Arg-was demonstrated for in vitro TME modulation, cancer therapeutic efficacy and clinical translation.
Collapse
Affiliation(s)
- Jiale Cai
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Guiping Hu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Lihua Hu
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Dan Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Dan Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Xiaolei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Boxian Hu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Cheng Li
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China.
| |
Collapse
|
29
|
Lu J, Chen F, Xie X, Wu Z, Chen Y, Zhang Y, Fang H, Ruan F, Shao D, Wang Z, Pei R. X-ray-controllable release of carbon monoxide potentiates radiotherapy by ultrastable hybrid nanoreservoirs. Biomaterials 2023; 302:122313. [PMID: 37672998 DOI: 10.1016/j.biomaterials.2023.122313] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
Carbon monoxide (CO) exhibits unique abilities in sensitizing cancer radiotherapy (RT). However, the development of a highly stable CO-delivery nanosystem with sustained CO release in tumor tissues and the prevention of CO leakage into normal tissues remains a challenge. Herein, an organic-inorganic hybrid strategy is proposed to create ultrastable CO nanoreservoirs by locking an unstable iron carbonyl (FeCO) prodrug in a stable mesoporous silica matrix. Different from traditional FeCO-loading nanoplatforms, FeCO-bridged nanoreservoirs not only tethered labile FeCO in the framework to prevent unwanted FeCO leakage, but also achieved sustained CO release in response to X-ray and endogenous H2O2. Importantly, FeCO-bridged nanoreservoirs exhibited the sequential release of CO and Fe2+, thereby performing highly efficient chemodynamic therapy. Such a powerful combination of RT, gas therapy, and chemodynamic therapy boosts robust immunogenic cell death, thus enabling the elimination of deeply metastatic colon tumors with minimal side effects. The proposed organic-inorganic hybrid strategy opens a new window for the development of stable nanoreservoirs for the on-demand delivery of unstable gases and provides a feasible approach for the sequential release of CO and metal ions from metal carbonyl complexes.
Collapse
Affiliation(s)
- Junna Lu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Fangman Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China.
| | - Xiaochun Xie
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Ziping Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yinglu Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yidan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Hui Fang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Feixia Ruan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangdong, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and NanoBionics Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and NanoBionics Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
30
|
Pan MM, Li P, Yu YP, Jiang M, Yang X, Zhang P, Nie J, Hu J, Yu X, Xu L. Bimetallic Ions Functionalized Metal-Organic-Framework Nanozyme for Tumor Microenvironment Regulating and Enhanced Photodynamic Therapy for Hypoxic Tumor. Adv Healthc Mater 2023; 12:e2300821. [PMID: 37199497 DOI: 10.1002/adhm.202300821] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Photodynamic therapy (PDT), as a light irradiation inducing reactive oxygen species (ROS) generation for cancer treatment, offers facile and promising solutions with respect to spatiotemporal control of ROS generation, and minimizes the systemic toxicity and side effects for highly precise tumor therapy. However, the PDT efficiency is often severely compromised by the complex tumor microenvironment (TME), such as the hypoxic condition and overexpressed antioxidants. Here, for the first time, a bimetallic ion-modified metal-organic framework nanozyme (Zr4+ -MOF-Ru3+ /Pt4+ -Ce6@HA, ZMRPC@HA) is designed. ZMRPC@HA with catalase (CAT) and glutathione oxidase (GSHOx) mimetic activities, can efficiently regulate TME by generation of O2 and deplete the GSH synergistically for enhancing the long-term PDT efficacy toward the hypoxic tumor. The in vitro cell inhibition and in vivo on tumor xenograft evaluations demonstrate the PDT strategy by using ZMRPC@HA can successfully inhibit the differentiation and proliferation of tumor cells under a 660 nm laser irradiation in deep tissues. These findings open a new avenue for the design of multimetallic ions functionalized MOF-based nanozymes with multienzyme mimetic activities toward the antitumor and various other biological applications.
Collapse
Affiliation(s)
- Meng-Meng Pan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Puze Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yan-Ping Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Pei Zhang
- Wuhan Institute of Virology, CAS, Wuhan, 430071, China
| | - Jing Nie
- Hubei Medical Devices Quality Supervision and Test Institute, High-Tech Avenue 507#, Wuhan, 430075, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Jiangxia Laboratory, Wuhan, 430200, China
| | - Xu Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
31
|
Zhang J, Jiang X, Luo W, Mo Y, Dai C, Zhu L. PEGA-BA@Ce6@PFCE Micelles as Oxygen Nanoshuttles for Tumor Hypoxia Relief and Enhanced Photodynamic Therapy. Molecules 2023; 28:6697. [PMID: 37764473 PMCID: PMC10535279 DOI: 10.3390/molecules28186697] [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: 05/26/2023] [Revised: 08/27/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Tumor hypoxia, which is mainly caused by the inefficient microvascular systems induced by rapid tumor growth, is a common characteristic of most solid tumors and has been found to hinder treatment outcomes for many types of cancer therapeutics. In this study, an amphiphilic block copolymer, poly (ethylene glycol) methyl ether acrylate-block-n-butyl acrylate (PEGA-BA), was prepared via the ATRP method and self-assembled into core-shell micelles as nano radiosensitizers. These micelles encapsulated a photosensitizer, Chlorin e6 (Ce6), and demonstrated well-defined morphology, a uniform size distribution, and high oxygen loading capacity. Cell experiments showed that PEGA-BA@Ce6@PFCE micelles could effectively enter cells. Further in vitro anticancer studies demonstrated that the PEGA-BA@Ce6@PFCE micelles significantly suppressed the tumor cell survival rate when exposed to a laser.
Collapse
Affiliation(s)
- Junan Zhang
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (J.Z.); (W.L.); (Y.M.)
| | - Xiaoyun Jiang
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (J.Z.); (W.L.); (Y.M.)
| | - Wenyue Luo
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (J.Z.); (W.L.); (Y.M.)
| | - Yongjie Mo
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (J.Z.); (W.L.); (Y.M.)
| | - Chunyan Dai
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (J.Z.); (W.L.); (Y.M.)
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, Haikou 571158, China
| | - Linhua Zhu
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (J.Z.); (W.L.); (Y.M.)
- Key Laboratory of Functional Organic Polymers of Haikou, Haikou 571158, China
| |
Collapse
|
32
|
Zhang C, Hu X, Jin L, Lin L, Lin H, Yang Z, Huang W. Strategic Design of Conquering Hypoxia in Tumor for Advanced Photodynamic Therapy. Adv Healthc Mater 2023; 12:e2300530. [PMID: 37186515 DOI: 10.1002/adhm.202300530] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/08/2023] [Indexed: 05/17/2023]
Abstract
Photodynamic therapy (PDT), with its advantages of high targeting, minimally invasive, and low toxicity side effects, has been widely used in the clinical therapy of various tumors, especially superficial tumors. However, the tumor microenvironment (TME) presents hypoxia due to the low oxygen (O2 ) supply caused by abnormal vascularization in neoplastic tissues and high O2 consumption induced by the rapid proliferation of tumor cells. The efficacy of oxygen-consumping PDT can be hampered by a hypoxic TME. To address this problem, researchers have been developing advanced nanoplatforms and strategies to enhance the therapeutic effect of PDT in tumor treatment. This review summarizes recent advanced PDT therapeutic strategies to against the hypoxic TME, thus enhancing PDT efficacy, including increasing O2 content in TME through delivering O2 to the tumors and in situ generations of O2 ; decreasing the O2 consumption during PDT by design of type I photosensitizers. Moreover, recent synergistically combined therapy of PDT and other therapeutic methods such as chemotherapy, photothermal therapy, immunotherapy, and gas therapy is accounted for by addressing the challenging problems of mono PDT in hypoxic environments, including tumor resistance, proliferation, and metastasis. Finally, perspectives of the opportunities and challenges of PDT in future clinical research and translations are provided.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Xiaoming Hu
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
- Jiangxi Key Laboratory of Nanobiomaterials, School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, P. R. China
| | - Long Jin
- Department of Pathology, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Lisheng Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Hongxin Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Zhen Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
| | - Wei Huang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
- Frontiers Science Center for Flexible Electronics (FSCFE), MIIT Key Laboratory of Flexible Electronics (KLoFE) Northwestern Polytechnical University Xi'an, Xi'an, 710072, P. R. China
| |
Collapse
|
33
|
Lu J, Shi T, Shi C, Chen F, Yang C, Xie X, Wang Z, Shen H, Xu J, Leong KW, Shao D. Thiol-Disulfide Exchange Coordinates the Release of Nitric Oxide and Dexamethasone for Synergistic Regulation of Intestinal Microenvironment in Colitis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0204. [PMID: 37533463 PMCID: PMC10393581 DOI: 10.34133/research.0204] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023]
Abstract
The cell-specific functions of nitric oxide (NO) in the intestinal microenvironment orchestrate its therapeutic effects in ulcerative colitis. While most biomaterials show promise by eliciting the characteristics of NO, the insufficient storage, burst release, and pro-inflammatory side effects of NO remain as challenges. Herein, we report the development of thiol-disulfide hybrid mesoporous organosilica nanoparticles (MONs) that improve the storage and sustained release of NO, broadening the therapeutic window of NO-based therapy against colitis. The tailored NO-storing nanomaterials coordinated the release of NO and the immunoregulator dexamethasone (Dex) in the intestinal microenvironment, specifically integrating the alleviation of oxidative stress in enterocytes and the reversal of NO-exacerbated macrophage activation. Mechanistically, such a synchronous operation was achieved by a self-motivated process wherein the thiyl radicals produced by NO release cleaved the disulfide bonds to degrade the matrix and release Dex via thiol-disulfide exchange. Specifically, the MON-mediated combination of NO and Dex greatly ameliorated intractable colitis compared with 5-aminosalicylic acid, even after delayed treatment. Together, our results reveal a key contribution of synergistic modulation of the intestinal microenvironment in NO-based colitis therapy and introduce thiol-disulfide hybrid nanotherapeutics for the management of inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Junna Lu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Tongfei Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Chengxin Shi
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou 310000, China
| | - Fangman Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
| | - Chao Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Xiaochun Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and NanoBionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - He Shen
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and NanoBionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
34
|
Shen J, Chen G, Zhao L, Huang G, Liu H, Liu B, Miao Y, Li Y. Recent Advances in Nanoplatform Construction Strategy for Alleviating Tumor Hypoxia. Adv Healthc Mater 2023; 12:e2300089. [PMID: 37055912 DOI: 10.1002/adhm.202300089] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Hypoxia is a typical feature of most solid tumors and has important effects on tumor cells' proliferation, invasion, and metastasis. This is the key factor that leads to poor efficacy of different kinds of therapy including chemotherapy, radiotherapy, photodynamic therapy, etc. In recent years, the construction of hypoxia-relieving functional nanoplatforms through nanotechnology has become a new strategy to reverse the current situation of tumor microenvironment hypoxia and improve the effectiveness of tumor treatment. Here, the main strategies and recent progress in constructing nanoplatforms are focused on to directly carry oxygen, generate oxygen in situ, inhibit mitochondrial respiration, and enhance blood perfusion to alleviate tumor hypoxia. The advantages and disadvantages of these nanoplatforms are compared. Meanwhile, nanoplatforms based on organic and inorganic substances are also summarized and classified. Through the comprehensive overview, it is hoped that the summary of these nanoplatforms for alleviating hypoxia could provide new enlightenment and prospects for the construction of nanomaterials in this field.
Collapse
Affiliation(s)
- Jing Shen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Guobo Chen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Linghao Zhao
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Guoyang Huang
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Hui Liu
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuhao Li
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
35
|
Cui X, Ruan Q, Zhuo X, Xia X, Hu J, Fu R, Li Y, Wang J, Xu H. Photothermal Nanomaterials: A Powerful Light-to-Heat Converter. Chem Rev 2023. [PMID: 37133878 DOI: 10.1021/acs.chemrev.3c00159] [Citation(s) in RCA: 324] [Impact Index Per Article: 162.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
All forms of energy follow the law of conservation of energy, by which they can be neither created nor destroyed. Light-to-heat conversion as a traditional yet constantly evolving means of converting light into thermal energy has been of enduring appeal to researchers and the public. With the continuous development of advanced nanotechnologies, a variety of photothermal nanomaterials have been endowed with excellent light harvesting and photothermal conversion capabilities for exploring fascinating and prospective applications. Herein we review the latest progresses on photothermal nanomaterials, with a focus on their underlying mechanisms as powerful light-to-heat converters. We present an extensive catalogue of nanostructured photothermal materials, including metallic/semiconductor structures, carbon materials, organic polymers, and two-dimensional materials. The proper material selection and rational structural design for improving the photothermal performance are then discussed. We also provide a representative overview of the latest techniques for probing photothermally generated heat at the nanoscale. We finally review the recent significant developments of photothermal applications and give a brief outlook on the current challenges and future directions of photothermal nanomaterials.
Collapse
Affiliation(s)
- Ximin Cui
- State Key Laboratory of Radio Frequency Heterogeneous Integration, College of Electronics and Information Engineering, Shenzhen University, Shenzhen 518060, China
| | - Qifeng Ruan
- Ministry of Industry and Information Technology Key Lab of Micro-Nano Optoelectronic Information System & Guangdong Provincial Key Laboratory of Semiconductor Optoelectronic Materials and Intelligent Photonic Systems, Harbin Institute of Technology, Shenzhen 518055, China
| | - Xiaolu Zhuo
- Guangdong Provincial Key Lab of Optoelectronic Materials and Chips, School of Science and Engineering, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
| | - Xinyue Xia
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Jingtian Hu
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Runfang Fu
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Yang Li
- State Key Laboratory of Radio Frequency Heterogeneous Integration, College of Electronics and Information Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Hongxing Xu
- School of Physics and Technology and School of Microelectronics, Wuhan University, Wuhan 430072, Hubei, China
- Henan Academy of Sciences, Zhengzhou 450046, Henan, China
- Wuhan Institute of Quantum Technology, Wuhan 430205, Hubei, China
| |
Collapse
|
36
|
Roy S, Bag N, Bardhan S, Hasan I, Guo B. Recent Progress in NIR-II Fluorescence Imaging-guided Drug Delivery for Cancer Theranostics. Adv Drug Deliv Rev 2023; 197:114821. [PMID: 37037263 DOI: 10.1016/j.addr.2023.114821] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 04/12/2023]
Abstract
Fluorescence imaging in the second near-infrared window (NIR-II) has become a prevalent choice owing to its appealing advantages like deep penetration depth, low autofluorescence, decent spatiotemporal resolution, and a high signal-to-background ratio. This would expedite the innovation of NIR-II imaging-guided drug delivery (IGDD) paradigms for the improvement of the prognosis of patients with tumors. This work systematically reviews the recent progress of such NIR-II IGDD-mediated cancer therapeutics and collectively brings its essence to the readers. Special care has been taken to assess their performances based on their design approach, such as enhancing their drug loading and triggering release, designing intrinsic and extrinsic fluorophores, and/ or overcoming biological barriers. Besides, the state-of-the-art NIR-II IGDD platforms for different therapies like chemo-, photodynamic, photothermal, chemodynamic, immuno-, ion channel, gas-therapies, and multiple functions such as stimulus-responsive imaging and therapy, and monitoring of drug release and therapeutic response, have been updated. In addition, for boosting theranostic outcomes and clinical translation, the innovation directions of NIR-II IGDD platforms are summarized, including renal-clearable, biodegradable, sub-cellular targeting, and/or afterglow, chemiluminescence, X-ray excitable NIR-IGDD, and even cell therapy. This review will propel new directions for safe and efficient NIR-II fluorescence-mediated anticancer drug delivery.
Collapse
Affiliation(s)
- Shubham Roy
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China
| | - Neelanjana Bag
- Department of Physics, Jadavpur University, Kolkata-700032, India
| | - Souravi Bardhan
- Department of Physics, Jadavpur University, Kolkata-700032, India
| | - Ikram Hasan
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| |
Collapse
|
37
|
Ren SN, Zhang ZY, Guo RJ, Wang DR, Chen FF, Chen XB, Fang XD. Application of nanotechnology in reversing therapeutic resistance and controlling metastasis of colorectal cancer. World J Gastroenterol 2023; 29:1911-1941. [PMID: 37155531 PMCID: PMC10122790 DOI: 10.3748/wjg.v29.i13.1911] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
Colorectal cancer (CRC) is the most common digestive malignancy across the world. Its first-line treatments applied in the routine clinical setting include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, resistance to therapy has been identified as the major clinical challenge that fails the treatment method, leading to recurrence and distant metastasis. An increasing number of studies have been attempting to explore the underlying mechanisms of the resistance of CRC cells to different therapies, which can be summarized into two aspects: (1) The intrinsic characters and adapted alterations of CRC cells before and during treatment that regulate the drug metabolism, drug transport, drug target, and the activation of signaling pathways; and (2) the suppressive features of the tumor microenvironment (TME). To combat the issue of therapeutic resistance, effective strategies are warranted with a focus on the restoration of CRC cells’ sensitivity to specific treatments as well as reprogramming impressive TME into stimulatory conditions. To date, nanotechnology seems promising with scope for improvement of drug mobility, treatment efficacy, and reduction of systemic toxicity. The instinctive advantages offered by nanomaterials enable the diversity of loading cargoes to increase drug concentration and targeting specificity, as well as offer a platform for trying the combination of different treatments to eventually prevent tumor recurrence, metastasis, and reversion of therapy resistance. The present review intends to summarize the known mechanisms of CRC resistance to chemotherapy, radiotherapy, immunotherapy, and targeted therapy, as well as the process of metastasis. We have also emphasized the recent application of nanomaterials in combating therapeutic resistance and preventing metastasis either by combining with other treatment approaches or alone. In summary, nanomedicine is an emerging technology with potential for CRC treatment; hence, efforts should be devoted to targeting cancer cells for the restoration of therapeutic sensitivity as well as reprogramming the TME. It is believed that the combined strategy will be beneficial to achieve synergistic outcomes contributing to control and management of CRC in the future.
Collapse
Affiliation(s)
- Sheng-Nan Ren
- Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Zhan-Yi Zhang
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Rui-Jie Guo
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Da-Ren Wang
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Fang-Fang Chen
- Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xue-Bo Chen
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xue-Dong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
38
|
Zhu Y, Li Q, Wang C, Hao Y, Yang N, Chen M, Ji J, Feng L, Liu Z. Rational Design of Biomaterials to Potentiate Cancer Thermal Therapy. Chem Rev 2023. [PMID: 36912061 DOI: 10.1021/acs.chemrev.2c00822] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Cancer thermal therapy, also known as hyperthermia therapy, has long been exploited to eradicate mass lesions that are now defined as cancer. With the development of corresponding technologies and equipment, local hyperthermia therapies such as radiofrequency ablation, microwave ablation, and high-intensity focused ultrasound, have has been validated to effectively ablate tumors in modern clinical practice. However, they still face many shortcomings, including nonspecific damages to adjacent normal tissues and incomplete ablation particularly for large tumors, restricting their wide clinical usage. Attributed to their versatile physiochemical properties, biomaterials have been specially designed to potentiate local hyperthermia treatments according to their unique working principles. Meanwhile, biomaterial-based delivery systems are able to bridge hyperthermia therapies with other types of treatment strategies such as chemotherapy, radiotherapy and immunotherapy. Therefore, in this review, we discuss recent progress in the development of functional biomaterials to reinforce local hyperthermia by functioning as thermal sensitizers to endow more efficient tumor-localized thermal ablation and/or as delivery vehicles to synergize with other therapeutic modalities for combined cancer treatments. Thereafter, we provide a critical perspective on the further development of biomaterial-assisted local hyperthermia toward clinical applications.
Collapse
Affiliation(s)
- Yujie Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Quguang Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Chunjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| |
Collapse
|
39
|
Sun T, Jiang C. Stimuli-responsive drug delivery systems triggered by intracellular or subcellular microenvironments. Adv Drug Deliv Rev 2023; 196:114773. [PMID: 36906230 DOI: 10.1016/j.addr.2023.114773] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Drug delivery systems (DDS) triggered by local microenvironment represents the state-of-art of nanomedicine design, where the triggering hallmarks at intracellular and subcellular levels could be employed to exquisitely recognize the diseased sites, reduce side effects, and expand the therapeutic window by precisely tailoring the drug-release kinetics. Though with impressive progress, the DDS design functioning at microcosmic levels is fully challenging and underexploited. Here, we provide an overview describing the recent advances on stimuli-responsive DDSs triggered by intracellular or subcellular microenvironments. Instead of focusing on the targeting strategies as listed in previous reviews, we herein mainly highlight the concept, design, preparation and applications of stimuli-responsive systems in intracellular models. Hopefully, this review could give useful hints in developing nanoplatforms proceeding at a cellular level.
Collapse
Affiliation(s)
- Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China.
| |
Collapse
|
40
|
Tang Z, Jiang S, Tang W, He Q, Wei H, Jin C, Wang S, Zhang H. H 2O 2 Self-Supplying and GSH-Depleting Nanocatalyst for Copper Metabolism-Based Synergistic Chemodynamic Therapy and Chemotherapy. Mol Pharm 2023; 20:1717-1728. [PMID: 36809003 DOI: 10.1021/acs.molpharmaceut.2c00937] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Chemodynamic therapy (CDT) that involves the use of Fenton catalysts to convert endogenous hydrogen peroxide (H2O2) to hydroxyl radicals (·OH) constitutes a promising strategy for cancer therapy; however, insufficient endogenous H2O2 and glutathione (GSH) overexpression render its efficiency unsatisfactory. Herein, we present an intelligent nanocatalyst that comprises copper peroxide nanodots and DOX-loaded mesoporous silica nanoparticles (MSNs) (DOX@MSN@CuO2) and can self-supply exogenous H2O2 and respond to specific tumor microenvironments (TME). Following endocytosis into tumor cells, DOX@MSN@CuO2 initially decomposes into Cu2+ and exogenous H2O2 in the weakly acidic TME. Subsequently, Cu2+ reacts with high GSH concentrations, thereby inducing GSH depletion and reducing Cu2+ to Cu+ Next, the generated Cu+ undergoes Fenton-like reactions with exogenous H2O2 to accelerate toxic ·OH production, which exhibits a rapid reaction rate and is responsible for tumor cell apoptosis, thereby enhancing CDT. Furthermore, the successful delivery of DOX from the MSNs achieves chemotherapy and CDT integration. Thus, this excellent strategy can resolve the problem of insufficient CDT efficacy due to limited H2O2 and GSH overexpression. Integrating H2O2 self-supply and GSH deletion enhances CDT, and DOX-induced chemotherapy endows DOX@MSN@CuO2 with effective tumor growth-inhibiting properties alongside minimal side effects in vivo.
Collapse
Affiliation(s)
- Zhaomin Tang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Shuting Jiang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Wanlan Tang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Qian He
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Huangzhao Wei
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Chengyu Jin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Shuai Wang
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China School of Medicine, Chengdu 610500, China
| | - Hui Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
41
|
Wu W, Xu M, Qiao B, Huang T, Guo H, Zhang N, Zhou L, Li M, Tan Y, Zhang M, Xie X, Shuai X, Zhang C. Nanodroplet-enhanced sonodynamic therapy potentiates immune checkpoint blockade for systemic suppression of triple-negative breast cancer. Acta Biomater 2023; 158:547-559. [PMID: 36539109 DOI: 10.1016/j.actbio.2022.12.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/21/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Immune checkpoint blockade (ICB) has shown great promise in treating various advanced malignancies including triple-negative breast cancer (TNBC). However, only limited number of patients could benefit from it due to the low immune response rate caused by insufficient matured dendritic cells (DCs) and inadequate tumor infiltration of cytotoxic T lymphocytes (CTLs). Here, we report a combination therapeutic strategy which integrates STING pathway activation, hypoxia relief and sonodynamic therapy (SDT) with anti-PD-L1 therapy to improve the therapeutic outcome. The synthesized nanodroplet consisted of a O2-filled Perfluorohexane (PFH) core and a lipid membrane carrying sonosensitizer IR-780 and STING agonist Vadimezan (DMXAAs). It released O2 inside the hypoxic tumor tissue, thereby enhancing SDT which relied on O2 to generate cytotoxic reactive oxygen species (ROS). The co-delivered STING agonist DMXAAs promoted the maturation and tumor antigen cross-presenting of DCs for priming of CTLs. Moreover, SDT induced immunogenic cell death (ICD) of tumor to release abundant tumor-associated antigens, which increased tumor immunogenicity to promote tumor infiltration of CTLs. Consequently, not only a robust adaptive immune response was elicited but also the immunologically "cold" TNBC was turned "hot" to enable a potent anti-PD-L1 therapy. The nanodroplet demonstrated strong efficacy to systemically suppress TNBC growth and mimic distant tumor in vivo. STATEMENT OF SIGNIFICANCE: Only a limited number of triple-negative breast cancer (TNBC) patients can benefit from immune checkpoint blockade therapy due to its low immune response rate caused by insufficient matured DCs and inadequate tumor infiltration of cytotoxic T lymphocytes (CTLs). Interestingly, compelling evidence has shown that sonodynamic therapy (SDT) not only directly kills cancer cells but also elicits immunogenic cell death (ICD), which promotes tumor infiltration of cytotoxic T lymphocytes to transform an immunosuppressive "cold" tumor into a "hot" one. However, the hypoxic tumor microenvironment severely restricts the therapeutic efficiency of SDT, wherein, oxygen is indispensable in the process of ROS generation. Here, we report an O2-filled nanodroplet-enhanced sonodynamic therapy that significantly potentiated immune checkpoint blockade for systemic suppression of TNBC.
Collapse
Affiliation(s)
- Wenxin Wu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ming Xu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Qiao
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tongyi Huang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huanling Guo
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Luyao Zhou
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Manying Li
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Tan
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Minru Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Chunyang Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
42
|
Chang M, Wang M, Liu Y, Liu M, Kheraif AAA, Ma P, Zhao Y, Lin J. Dendritic Plasmonic CuPt Alloys for Closed-Loop Multimode Cancer Therapy with Remarkably Enhanced Efficacy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206423. [PMID: 36567272 DOI: 10.1002/smll.202206423] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/22/2022] [Indexed: 06/17/2023]
Abstract
The outcome of laser-triggered plasmons-induced phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is significantly limited by the hypoxic tumor microenvironment and the upregulation of heat shock proteins (HSPs) in response to heat stress. Mitochondria, the biological battery of cells, can serve as an important breakthrough to overcome these obstacles. Herein, dendritic triangular pyramidal plasmonic CuPt alloys loaded with heat-sensitive NO donor N, N'-di-sec-butyl-N, N'-dinitroso-1,4-phenylenediamine (BNN) is developed. Under 808 nm laser irradiation, plasmonic CuPt can generate superoxide anion free radicals (·O2 - ) and heat simultaneously. The heat generated can then trigger the release of NO gas, which not only enables gas therapy but also damages the mitochondrial respiratory chain. Impaired mitochondrial respiration leads to reduced oxygen consumption and insufficient intracellular ATP supply, which effectively alleviates tumor hypoxia and undermines the synthesis of HSPs, in turn boosting plasmonic CuPt-based PDT and mild PTT. Additionally, the generated NO and ·O2 - can react to form more cytotoxic peroxynitrite (ONOO- ). This work describes a plasmonic CuPt@BNN (CPB) triggered closed-loop NO gas, free radicals, and mild photothermal therapy strategy that is highly effective at reciprocally promoting antitumor outcomes.
Collapse
Affiliation(s)
- Mengyu Chang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Man Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Yuhui Liu
- State Key Laboratory of Nuclear Resources and Environment, East China University of Technology, Nanchang, 330013, P. R. China
| | - Min Liu
- Department of Periodontology, Stomatological Hospital, Jilin University, Changchun, 130021, P. R. China
| | - Abdulaziz A Al Kheraif
- Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, 12372, Saudi Arabia
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
43
|
Li X, Wang H, Li Z, Tao F, Wu J, Guan W, Liu S. Oxygen switches: Refueling for cancer radiotherapy. Front Oncol 2023; 12:1085432. [PMID: 36873299 PMCID: PMC9978393 DOI: 10.3389/fonc.2022.1085432] [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: 10/31/2022] [Accepted: 12/28/2022] [Indexed: 02/18/2023] Open
Abstract
Radiotherapy remains the major therapeutic intervention for tumor patients. However, the hypoxic tumor microenvironment leads to treatment resistance. Recently, a burgeoning number of nano-radiosensitizers designed to increase the oxygen concentration in tumors were reported. These nano radiosensitizers served as oxygen carriers, oxygen generators, and even sustained oxygen pumps, attracting increased research interest. In this review, we focus on the novel oxygen-enrich nano radiosensitizers, which we call oxygen switches, and highlight their influence in radiotherapy through different strategies. Physical strategies-based oxygen switches carried O2 into the tumor via their high oxygen capacity. The chemical reactions to generate O2 in situ were triggered by chemical strategies-based oxygen switches. Biological strategies-based oxygen switches regulated tumor metabolism, remodeled tumor vasculature, and even introduced microorganisms-mediated photosynthesis for long-lasting hypoxia alleviating. Moreover, the challenges and perspectives of oxygen switches-mediated oxygen-enrich radiotherapy were discussed.
Collapse
Affiliation(s)
- Xianghui Li
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
- Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
| | - Haoran Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyan Li
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
| | - Feng Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
| | - Wenxian Guan
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
| | - Song Liu
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
| |
Collapse
|
44
|
Wen D, Li K, Deng R, Feng J, Zhang H. Defect-Rich Glassy IrTe 2 with Dual Enzyme-Mimic Activities for Sono-Photosynergistic-Enhanced Oncotherapy. J Am Chem Soc 2023; 145:3952-3960. [PMID: 36757875 DOI: 10.1021/jacs.2c09967] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The complexity, diversity, and heterogeneity of malignant tumors pose a formidable challenge for antitumor therapy. To achieve the goal of significantly enhancing the antitumor effect, nanomedicine-based synergistic therapy is one of the important strategies. Herein, we innovatively report a defect-rich glassy IrTe2 (G-IrTe2) with weak Ir-Te bond strength for synergistic sonodynamic therapy (SDT), chemodynamic therapy (CDT), and mild photothermal therapy (PTT). G-IrTe2 sonosensitizer under ultrasound (US) stimuli exhibits excellent reactive oxygen species (ROS) production performance. Besides, catalase (CAT)-like activity of G-IrTe2 can provide abundant oxygen to enhance the SDT effect. Then, the theoretical calculation verifies that US stimuli can easily make the irregular Ir-Te bond to be broken in amorphous IrTe2 and free electrons will be released to combine with the oxygen and further form singlet oxygen (1O2). Meanwhile, G-IrTe2 with peroxidase (POD)-like activity can also catalyze endogenous H2O2 to produce more ROS for chemodynamic therapy (CDT), which is conducive to better tumor ablation. Furthermore, the ROS produced by sono-/chemodynamic processes can cause mitochondrial dysfunction and further give rise to heat shock protein (HSP) downregulated expression, maximizing the efficiency of mild PTT. Therefore, such glassy IrTe2 with rich defect could be significantly involved in synergistic oncotherapy and then effectively achieve outstanding antitumor efficacy. This study provides a new research idea for expanding the application of inorganic glassy nanomaterials in promoting the therapeutic effect of tumors.
Collapse
Affiliation(s)
- Ding Wen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Kai Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Ruiping Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Jing Feng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
45
|
Chen Z, Liu Z, Zhang Q, Huang S, Zhang Z, Feng X, Zeng L, Lin D, Wang L, Song H. Hypoxia-ameliorated photothermal manganese dioxide nanoplatform for reversing doxorubicin resistance. Front Pharmacol 2023; 14:1133011. [PMID: 36909187 PMCID: PMC9998484 DOI: 10.3389/fphar.2023.1133011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
Drug resistance is a huge hurdle in tumor therapy. Tumor hypoxia contributes to chemotherapy resistance by inducing the hypoxia-inducible factor-1α (HIF-1α) pathway. To reduce tumor hypoxia, novel approaches have been devised, providing significant importance to reverse therapeutic resistance and improve the effectiveness of antitumor therapies. Herein, the nanosystem of bovine serum albumin (BSA)-templated manganese dioxide (MnO2) nanoparticles (BSA/MnO2 NPs) loaded with doxorubicin (DOX) (DOX-BSA/MnO2 NPs) developed in our previous report was further explored for their physicochemical properties and capacity to reverse DOX resistance because of their excellent photothermal and tumor microenvironment (TME) response effects. The DOX-BSA/MnO2 NPs showed good biocompatibility and hemocompatibility. Meanwhile, DOX-BSA/MnO2 NPs could greatly affect DOX pharmacokinetic properties, with prolonged circulation time and reduced cardiotoxicity, besides enhancing accumulation at tumor sites. DOX-BSA/MnO2 NPs can interact with H2O2 and H+ in TME to form oxygen and exhibit excellent photothermal effect to further alleviate hypoxia due to MnO2, reversing DOX resistance by down-regulating HIF-1α expression and significantly improving the antitumor efficiency in DOX-resistant human breast carcinoma cell line (MCF-7/ADR) tumor model. The hypoxia-ameliorated photothermal MnO2 platform is a promising strategy for revering DOX resistance.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Zhihong Liu
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Qian Zhang
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Sheng Huang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Zaizhong Zhang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Xianquan Feng
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Lingjun Zeng
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Ding Lin
- Department of Pharmacy, Jiaxing Maternal and Child Healthcare Hospital, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lie Wang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Hongtao Song
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| |
Collapse
|
46
|
Liu S, Fang L, Ding H, Zhang Y, Li W, Liu B, Dong S, Tian B, Feng L, Yang P. Alternative Strategy to Optimize Cerium Oxide for Enhanced X-ray-Induced Photodynamic Therapy. ACS NANO 2022; 16:20805-20819. [PMID: 36378717 DOI: 10.1021/acsnano.2c08047] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The emergence of X-ray-induced photodynamic therapy (X-PDT) holds tremendous promise for clinical deep-penetrating cancer therapy. However, the clinical application of X-PDT in cancer treatment is still limited due to the hypoxic property of cancerous tissue, the inherent antioxidant system of tumor cells, and the difficulty in matching the absorption spectra of photosensitizers. Herein, a versatile core-shell radiosensitizer (SCNPs@DMSN@CeOx-PEG, denoted as SSCP) was elaborately designed and constructed to enhance X-PDT by coating tunable mesoporous silica on nanoscintillators, followed by embedding the cerium oxide nanoparticles in situ. The obtained SSCP radiosensitizer demonstrated a distinct blue-shift in the ultraviolet light region, so that it could perfectly absorb the ultraviolet light converted by the SCNPs core, resulting in the formation of photoinduced electron-hole (e--h+) pairs separation to generate reactive oxygen species (ROS). In addition, the cerium oxide exhibits high glutathione consumption to heighten ROS accumulation, and catalase-like activity to alleviate the hypoxia, which further enhances the efficiency of radiotherapy. Benefiting from the abundant Lu and Ce elements, the computed tomography imaging performance of SSCP is about 3.79-fold that of the clinical contrast agent (iohexol), which has great potential in both preclinical imaging and clinical translation.
Collapse
Affiliation(s)
- Shikai Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Linyang Fang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - He Ding
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Yangyang Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Wenting Li
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Bin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Shuming Dong
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Boshi Tian
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| |
Collapse
|
47
|
Wang D, Liao Y, Yan H, Zhu S, Liu Y, Li J, Wang X, Guo X, Gu Z, Sun B. In Situ Formed Z-Scheme Graphdiyne Heterojunction Realizes NIR-Photocatalytic Oxygen Evolution and Selective Radiosensitization for Hypoxic Tumors. ACS NANO 2022; 16:21186-21198. [PMID: 36445074 DOI: 10.1021/acsnano.2c09169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Photon radiotherapy is a common tool in the armory against tumors, but it is limited by hypoxia-related radioresistance of tumors and radiotoxicity to normal tissues. Here, we constructed a spatiotemporally controlled synergistic therapy platform based on the heterostructured CuO@Graphdiyne (CuO@GDY) nanocatalyst for simultaneously addressing the two key problems above in radiotherapy. First, the in situ formed Z-scheme CuO@GDY heterojunction performs highly efficient and controlled photocatalytic O2 evolution upon near-infrared (NIR) laser stimulation for tumor hypoxia alleviation. Subsequently, the CuO@GDY nanocatalyst with X-ray-stimulated Cu+ active sites can accelerate Fenton-like catalysis of ·OH production by responding to endogenous H2O2 for the selective killing of tumor cells rather than normal cells. In this way, the sequential combination of NIR-triggered photocatalytic O2 production and X-ray-accelerated Fenton-like reaction can lead to a comprehensive radiosensitization. Overall, this synergism underscores a controllable and precise therapy modality for simultaneously unlocking the hypoxia and non-selectivity in radiotherapy.
Collapse
Affiliation(s)
- Dongmei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - You Liao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Haili Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Yunpeng Liu
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
| | - Jian Li
- Laboratory of Renewable Energy Science and Engineering, Institute of Mechanical Engineering, EPFL, Station 9, 1015Lausanne, Switzerland
| | - Xue Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Xihong Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Baoyun Sun
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing100049, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
48
|
Xu Y, Yao Y, Deng W, Fang JC, Dupont RL, Zhang M, Čopar S, Tkalec U, Wang X. Magnetocontrollable droplet mobility on liquid crystal-infused porous surfaces. NANO RESEARCH 2022:1-10. [PMID: 36570861 DOI: 10.1007/s12274-022-5239-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/29/2022] [Accepted: 11/13/2022] [Indexed: 05/22/2023]
Abstract
UNLABELLED Magnetocontrollable droplet mobility on surfaces of both solids and simple fluids have been widely used in a wide range of applications. However, little is understood about the effect of the magnetic field on the wettability and mobility of droplets on structured fluids. Here, we report the manipulation of the dynamic behaviors of water droplets on a film of thermotropic liquid crystals (LCs). We find that the static wetting behavior and static friction of water droplets on a 4'-octyl-4-biphenylcarbonitrile (8CB) film strongly depend on the LC mesophases, and that a magnetic field caused no measurable change to these properties. However, we find that the droplet dynamics can be affected by a magnetic field as it slides on a nematic 8CB film, but not on isotropic 8CB, and is dependent on both the direction and strength of the magnetic field. By measuring the dynamic friction of a droplet sliding on a nematic 8CB film, we find that a magnetic field alters the internal orientational ordering of the 8CB which in turn affects its viscosity. We support this interpretation with a scaling argument using the LC magnetic coherence length that includes (i) the elastic energy from the long-range orientational ordering of 8CB and (ii) the free energy from the interaction between 8CB and a magnetic field. Overall, these results advance our understanding of droplet mobility on LC films and enable new designs for responsive surfaces that can manipulate the mobility of water droplets. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (further details of the stability of LCIPS against water-induced dewetting, the interfacial tension and contact angle measurement using a goniometer, the estimation of the thickness of LC wrapping layer at air-water interface on droplets, SEM measurements, the average sliding velocity of a water droplet on 5CB, E7, silicone oil, and mineral oil films with and without a magnetic field, representative force diagram (F d versus time) of a 3-µL water droplet moving at a speed of 0.1 mm/s on a nematic 8CB film, F dynamic acting on 3 µL water droplets moving at speeds of 0.1-1 mm/s on an isotropic 8CB film, the calculated magnetic coherence length as a function of the magnitude of the magnetic field applied to the nematic LCIPS, and the apparent advancing and receding contact angles of a moving water droplet on nematic LCIPS as a function of time, and polarized light micrographs (top view) of a nematic 8CB film between two DMOAP-functionalized glass slides before and after applying a horizontal magnetic field) is available in the online version of this article at 10.1007/s12274-022-5318-y.
Collapse
Affiliation(s)
- Yang Xu
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Yuxing Yao
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125 USA
| | - Weichen Deng
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Jen-Chun Fang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Robert L Dupont
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Meng Zhang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Simon Čopar
- Department of Physics, Faculty of Mathematics and Physics, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Uroš Tkalec
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Department of Condensed Matter Physics, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
| | - Xiaoguang Wang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210 USA
- Sustainability Institute, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
49
|
Cui Y, Yan H, Wang H, Zhang Y, Li M, Cui K, Xiao Z, Liu L, Xie W. CuS- 131I-PEG Nanotheranostics-Induced "Multiple Mild-Hyperthermia" Strategy to Overcome Radio-Resistance in Lung Cancer Brachytherapy. Pharmaceutics 2022; 14:pharmaceutics14122669. [PMID: 36559162 PMCID: PMC9785376 DOI: 10.3390/pharmaceutics14122669] [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: 10/20/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
Brachytherapy is one mainstay treatment for lung cancer. However, a great challenge in brachytherapy is radio-resistance, which is caused by severe hypoxia in solid tumors. In this research, we have developed a PEGylated 131I-labeled CuS nanotheranostics (CuS-131I-PEG)-induced "multiple mild-hyperthermia" strategy to reverse hypoxia-associated radio-resistance. Specifically, after being injected with CuS-131I-PEG nanotheranostics, tumors were irradiated by NIR laser to mildly increase tumor temperature (39~40 °C). This mild hyperthermia can improve oxygen levels and reduce expression of hypoxia-induced factor-1α (HIF-1α) inside tumors, which brings about alleviation of tumor hypoxia and reversion of hypoxia-induced radio-resistance. During the entire treatment, tumors are treated by photothermal brachytherapy three times, and meanwhile mild hyperthermia stimulation is conducted before each treatment of photothermal brachytherapy, which is defined as a "multiple mild-hyperthermia" strategy. Based on this strategy, tumors have been completely inhibited. Overall, our research presents a simple and effective "multiple mild-hyperthermia" strategy for reversing radio-resistance of lung cancer, achieving the combined photothermal brachytherapy.
Collapse
Affiliation(s)
- Yanna Cui
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui Yan
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haoze Wang
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200233, China
| | - Yongming Zhang
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Meng Li
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200233, China
| | - Kai Cui
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zeyu Xiao
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (Z.X.); (L.L.); (W.X.)
| | - Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (Z.X.); (L.L.); (W.X.)
| | - Wenhui Xie
- Department of Nuclear Medicine, Shanghai Chest Hospital & Department of Pharmacology and Chemical Biology, Translational Medicine Collaborative Innovation Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (Z.X.); (L.L.); (W.X.)
| |
Collapse
|
50
|
Fu J, Mao Y, Han J, Zhang P, Tan Y, Hu J, Seeberger PH, Yin J. A nitric oxide and hydrogen sulfide dual-donating nanosystem for highly synergistic gas-radiotherapy against hepatocellular carcinoma. BIOMATERIALS ADVANCES 2022; 144:213209. [PMID: 36473350 DOI: 10.1016/j.bioadv.2022.213209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
A drug delivery system (DDS) based on gold-capped mesoporous silica nanoparticles (MSN) is fabricated for loading NOSH-aspirin, a nitric oxide (NO) and hydrogen sulfide (H2S) dual-donating cytotoxic molecule. The liver targeting and tumor microenvironment responsive properties of the nanosystem enable, for the first time, the concurrent delivery of NO and H2S from a DDS into hepatocellular carcinoma (HCC) cells. Combined gas-radiotherapy (GT-RT) from drug-loaded DDS (NOSH@MSN-Au-Gal) and X-ray irradiation shows highly synergistic anti-cancer activity against both normoxic and hypoxic HCC cells. Further studies revealed that the combined GT-RT not only retains the well-known anticancer mechanism of NO, H2S, and X-ray individually, but also alleviates HCC hypoxia via NO- and H2S- involved unique pathways. In mice, the GT-RT greatly slows the growth of both subcutaneous and orthotopic HCC tumors and shows high biocompatibility. The current work is expected to promote the clinical application of combined GT-RT as an effective cancer treatment.
Collapse
Affiliation(s)
- Junjie Fu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214062, PR China
| | - Jing Han
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Pengfei Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China; Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214062, PR China
| | - Yunying Tan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China
| | - Jing Hu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, PR China
| | - Peter H Seeberger
- Biomolecular Systems Department, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|