1
|
Hiep Tran T, Thu Phuong Tran T. Current status of nanoparticle-mediated immunogenic cell death in cancer immunotherapy. Int Immunopharmacol 2024; 142:113085. [PMID: 39276455 DOI: 10.1016/j.intimp.2024.113085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
Immunogenic cell death (ICD) encompasses various forms of cell death modalities, including apoptosis, necroptosis, ferroptosis, and pyroptosis. It arises from a harmonious interplay of adjuvant (damage-associated molecular patterns-DAMPs and chemokines/cytokines) and antigenicity (tumor-associated antigens-TAA) to induce immune-reaction toward cancer cells. Inducing ICD stands out as a promising approach in cancer immunotherapy, capable of directly eliminating cancer cells and of eliciting enduring antitumor immune responses. Conventional tumor therapies like radiation therapy, photodynamic therapy, and chemotherapy can also induce ICD which could amplify their activities. The development of effective ICD inducers like nano-systems is crucial for ensuring safe and efficacious immunotherapy. Nanoparticles hold considerable promise in cancer therapy, offering enhanced therapeutic outcomes and mitigated side effects. They could be the capacity to adjust systemic biodistribution, augment the accumulation of therapeutic agents at the intended site and protect active agents from the complexity of human biofluid. This review aims to outline the role of nanoparticles in triggering ICD for cancer immunotherapy that potentially pave the way for cancer treatment.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
| | - Thi Thu Phuong Tran
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
2
|
Lin J, He Y, Li Y, Chen J, Liu X. Oxygen-Evolving Radiotherapy-Radiodynamic Therapy Synergized with NO Gas Therapy by Cerium-Based Rare-Earth Metal-Porphyrin Framework. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310957. [PMID: 38698608 DOI: 10.1002/smll.202310957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/12/2024] [Indexed: 05/05/2024]
Abstract
The efficacy of traditional radiotherapy (RT) has been severely limited by its significant side effects, as well as tumor hypoxia. Here, the nanoscale cerium (Ce)-based metaloxo clusters (Ce(IV)6)-porphyrin (meso-tetra (4-carboxyphenyl) porphyrin, TCPP) framework loaded with L-arginine (LA) (denoted as LA@Ce(IV)6-TCPP) is developed to serve as a multifarious radio enhancer to heighten X-ray absorption and energy transfer accompanied by O2/NO generation for hypoxia-improved RT-radiodynamic therapy (RDT) and gas therapy. Within tumor cells, LA@Ce(IV)6-TCPP will first react with endogenous H2O2 and inducible NO synthase (iNOS) to produce O2 and NO to respectively increase the oxygen supply and reduce oxygen consumption, thus alleviating tumor hypoxia. Then upon X-ray irradiation, LA@Ce(IV)6-TCPP can significantly enhance hydroxyl radical (•OH) generation from Ce(IV)6 metaloxo clusters for RT and synchronously facilitate singlet oxygen (1O2) generation from adjacently-coordinated TCPP for RDT. Moreover, both the •OH and 1O2 can further react with NO to generate more toxic peroxynitrite anions (ONOO-) to inhibit tumor growth for gas therapy. Benefitting from the alleviation of tumor hypoxia and intensified RT-RDT synergized with gas therapy, LA@Ce(IV)6-TCPP elicited superior anticancer outcomes. This work provides an effective RT strategy by using low doses of X-rays to intensify tumor suppression yet reduce systemic toxicity.
Collapse
Affiliation(s)
- Jinyan Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- State Key Laboratory of Structural Chemistry & CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, P. R. China
- Department of Translational Medicine, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, 361021, P. R. China
| | - Yueyang He
- State Key Laboratory of Structural Chemistry & CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, P. R. China
- Department of Translational Medicine, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, 361021, P. R. China
- Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361100, P. R. China
| | - Yang Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- State Key Laboratory of Structural Chemistry & CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, P. R. China
- Department of Translational Medicine, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, 361021, P. R. China
| | - Jianwu Chen
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, 350004, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- State Key Laboratory of Structural Chemistry & CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, P. R. China
- Department of Translational Medicine, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, 361021, P. R. China
| |
Collapse
|
3
|
Manoharan D, Wang LC, Chen YC, Li WP, Yeh CS. Catalytic Nanoparticles in Biomedical Applications: Exploiting Advanced Nanozymes for Therapeutics and Diagnostics. Adv Healthc Mater 2024; 13:e2400746. [PMID: 38683107 DOI: 10.1002/adhm.202400746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Catalytic nanoparticles (CNPs) as heterogeneous catalyst reveals superior activity due to their physio-chemical features, such as high surface-to-volume ratio and unique optical, electric, and magnetic properties. The CNPs, based on their physio-chemical nature, can either increase the reactive oxygen species (ROS) level for tumor and antibacterial therapy or eliminate the ROS for cytoprotection, anti-inflammation, and anti-aging. In addition, the catalytic activity of nanozymes can specifically trigger a specific reaction accompanied by the optical feature change, presenting the feasibility of biosensor and bioimaging applications. Undoubtedly, CNPs play a pivotal role in pushing the evolution of technologies in medical and clinical fields, and advanced strategies and nanomaterials rely on the input of chemical experts to develop. Herein, a systematic and comprehensive review of the challenges and recent development of CNPs for biomedical applications is presented from the viewpoint of advanced nanomaterial with unique catalytic activity and additional functions. Furthermore, the biosafety issue of applying biodegradable and non-biodegradable nanozymes and future perspectives are critically discussed to guide a promising direction in developing span-new nanozymes and more intelligent strategies for overcoming the current clinical limitations.
Collapse
Affiliation(s)
- Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Liu-Chun Wang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Peng Li
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
4
|
Wu J, Zhu X, Li Q, Fu Q, Wang B, Li B, Wang S, Chang Q, Xiang H, Ye C, Li Q, Huang L, Liang Y, Wang D, Zhao Y, Li Y. Enhancing radiation-resistance and peroxidase-like activity of single-atom copper nanozyme via local coordination manipulation. Nat Commun 2024; 15:6174. [PMID: 39039047 PMCID: PMC11263674 DOI: 10.1038/s41467-024-50416-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
The inactivation of natural enzymes by radiation poses a great challenge to their applications for radiotherapy. Single-atom nanozymes (SAzymes) with high structural stability under such extreme conditions become a promising candidate for replacing natural enzymes to shrink tumors. Here, we report a CuN3-centered SAzyme (CuN3-SAzyme) that exhibits higher peroxidase-like catalytic activity than a CuN4-centered counterpart, by locally regulating the coordination environment of single copper sites. Density functional theory calculations reveal that the CuN3 active moiety confers optimal H2O2 adsorption and dissociation properties, thus contributing to high enzymatic activity of CuN3-SAzyme. The introduction of X-ray can improve the kinetics of the decomposition of H2O2 by CuN3-SAzyme. Moreover, CuN3-SAzyme is very stable after a total radiation dose of 500 Gy, without significant changes in its geometrical structure or coordination environment, and simultaneously still retains comparable peroxidase-like activity relative to natural enzymes. Finally, this developed CuN3-SAzyme with remarkable radioresistance can be used as an external field-improved therapeutics for enhancing radio-enzymatic therapy in vitro and in vivo. Overall, this study provides a paradigm for developing SAzymes with improved enzymatic activity through local coordination manipulation and high radioresistance over natural enzymes, for example, as sensitizers for cancer therapy.
Collapse
Affiliation(s)
- Jiabin Wu
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xianyu Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Qun Li
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiang Fu
- School of Future Technology, University of Science and Technology of China, Hefei, 230026, China.
- Hefei National Laboratory, University of Science and Technology of China, Hefei, 230088, China.
| | - Bingxue Wang
- School of Future Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Beibei Li
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shanshan Wang
- Institute of Quality Standards & Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qingchao Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, China
| | - Huandong Xiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, China
| | - Chengliang Ye
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Qiqiang Li
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Liang Huang
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yan Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, China.
| | - Dingsheng Wang
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, China
| | - Yadong Li
- Department of Chemistry, Tsinghua University, Beijing, 100084, China.
- College of Chemistry, Beijing Normal University, Beijing, 100875, China.
- The Key Laboratory of Functional Molecular Solids, Ministry of Education, College of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241002, China.
| |
Collapse
|
5
|
Cai Q, He Y, Zhou Y, Zheng J, Deng J. Nanomaterial-Based Strategies for Preventing Tumor Metastasis by Interrupting the Metastatic Biological Processes. Adv Healthc Mater 2024; 13:e2303543. [PMID: 38411537 DOI: 10.1002/adhm.202303543] [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: 10/17/2023] [Revised: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Tumor metastasis is the primary cause of cancer-related deaths. The prevention of tumor metastasis has garnered notable interest and interrupting metastatic biological processes is considered a potential strategy for preventing tumor metastasis. The tumor microenvironment (TME), circulating tumor cells (CTCs), and premetastatic niche (PMN) play crucial roles in metastatic biological processes. These processes can be interrupted using nanomaterials due to their excellent physicochemical properties. However, most studies have focused on only one aspect of tumor metastasis. Here, the hypothesis that nanomaterials can be used to target metastatic biological processes and explore strategies to prevent tumor metastasis is highlighted. First, the metastatic biological processes and strategies involving nanomaterials acting on the TME, CTCs, and PMN to prevent tumor metastasis are briefly summarized. Further, the current challenges and prospects of nanomaterials in preventing tumor metastasis by interrupting metastatic biological processes are discussed. Nanomaterial-and multifunctional nanomaterial-based strategies for preventing tumor metastasis are advantageous for the long-term fight against tumor metastasis and their continued exploration will facilitate rapid progress in the prevention, diagnosis, and treatment of tumor metastasis. Novel perspectives are outlined for developing more effective strategies to prevent tumor metastasis, thereby improving the outcomes of patients with cancer.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijia He
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
6
|
Wang M, Chen G, Hu B, Zhang F, Xu Q, Li L, Xi Q, Liu J, Yu Z, Cao P, Wang Y, Yu M. Electrically activated polymetallic nanocrystals for long-term tumor suppression via oxygen-independent ROS generation and electro-immunotherapy. J Control Release 2024; 370:677-690. [PMID: 38740093 DOI: 10.1016/j.jconrel.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
The low oxidation level and immunosuppressive microenvironment within hypoxic tumor tissue are critical factors contributing to the inefficacy of various anti-tumor strategies. Herein, we have designed a novel intravenous injection nanoplatform to conduct electro-immunotherapy, based on phospholipid-modified PtPd nanocrystals loaded with the immunoregulator IPI549 (LP@Pt-Pd@IPI549 nanoparticles, LPPI). LPPI responds to reactive oxygen species (ROS), triggering a cascade of therapeutic effects that overcome hypoxia-related resistance and effectively eradicate hypoxic tumors. Firstly, under electric field exposure, LPPI relied on water rather than oxygen to generate abundant ROS under hypoxic conditions for tumor electrodynamic therapy (EDT). Moreover, the generated ROS further induced the disintegration of the outer phospholipid membrane of LPPI, leading to the release of the immunoregulator and inhibition of myeloid-derived suppressor cells (MDSCs), triggering cascade immune responses. Additionally, the immunomodulatory effects of IPI549, in synergy with the immunogenic cell death (ICD) induced by EDT, reversed the immunosuppressive microenvironment contributing to tumor resistance. In summary, EDT transiently killed tumor cells while simultaneously generating antigen release, instigating an adaptive immune response for electro-immunotherapy, resulting in a potent and long-lasting tumor inhibition effect.
Collapse
Affiliation(s)
- Manchun Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Gui Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), Dongguan 523018, China
| | - Ben Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fengling Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qinqin Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lei Li
- Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Qiye Xi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jun Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang 110000, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), Dongguan 523018, China
| | - Peng Cao
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang 110000, China.
| | - Yongxia Wang
- Breast Department, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China.
| | - Meng Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
7
|
Han J, Dong H, Zhu T, Wei Q, Wang Y, Wang Y, Lv Y, Mu H, Huang S, Zeng K, Xu J, Ding J. Biochemical hallmarks-targeting antineoplastic nanotherapeutics. Bioact Mater 2024; 36:427-454. [PMID: 39044728 PMCID: PMC11263727 DOI: 10.1016/j.bioactmat.2024.05.042] [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: 02/20/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 07/25/2024] Open
Abstract
Tumor microenvironments (TMEs) have received increasing attention in recent years as they play pivotal roles in tumorigenesis, progression, metastases, and resistance to the traditional modalities of cancer therapy like chemotherapy. With the rapid development of nanotechnology, effective antineoplastic nanotherapeutics targeting the aberrant hallmarks of TMEs have been proposed. The appropriate design and fabrication endow nanomedicines with the abilities for active targeting, TMEs-responsiveness, and optimization of physicochemical properties of tumors, thereby overcoming transport barriers and significantly improving antineoplastic therapeutic benefits. This review begins with the origins and characteristics of TMEs and discusses the latest strategies for modulating the TMEs by focusing on the regulation of biochemical microenvironments, such as tumor acidosis, hypoxia, and dysregulated metabolism. Finally, this review summarizes the challenges in the development of smart anti-cancer nanotherapeutics for TME modulation and examines the promising strategies for combination therapies with traditional treatments for further clinical translation.
Collapse
Affiliation(s)
- Jing Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - He Dong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Tianyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Yongheng Wang
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Yun Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Yu Lv
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Shandeng Huang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Ke Zeng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
8
|
Cui Z, Huang B, Zheng J, Tian J, Zhang W. A TME-enlightened protein-binding photodynamic nanoinhibitor for highly effective oncology treatment. Proc Natl Acad Sci U S A 2024; 121:e2321545121. [PMID: 38713621 PMCID: PMC11098098 DOI: 10.1073/pnas.2321545121] [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: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 05/09/2024] Open
Abstract
The efficiency of photodynamic therapy (PDT) is greatly dependent on intrinsic features of photosensitizers (PSs), but most PSs suffer from narrow diffusion distances and short life span of singlet oxygen (1O2). Here, to conquer this issue, we propose a strategy for in situ formation of complexes between PSs and proteins to deactivate proteins, leading to highly effective PDT. The tetrafluorophenyl bacteriochlorin (FBC), a strong near-infrared absorbing photosensitizer, can tightly bind to intracellular proteins to form stable complexes, which breaks through the space-time constraints of PSs and proteins. The generated singlet oxygen directly causes the protein dysfunction, leading to high efficiency of PSs. To enable efficient delivery of PSs, a charge-conversional and redox-responsive block copolymer POEGMA-b-(PAEMA/DMMA-co-BMA) (PB) was designed to construct a protein-binding photodynamic nanoinhibitor (FBC@PB), which not only prolongs blood circulation and enhances cellular uptake but also releases FBC on demand in tumor microenvironment (TME). Meanwhile, PDT-induced destruction of cancer cells could produce tumor-associated antigens which were capable to trigger robust antitumor immune responses, facilitating the eradication of residual cancer cells. A series of experiments in vitro and in vivo demonstrated that this multifunctional nanoinhibitor provides a promising strategy to extend photodynamic immunotherapy.
Collapse
Affiliation(s)
- Zepeng Cui
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Baoxuan Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Jiahao Zheng
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| |
Collapse
|
9
|
Liu Z, Huang L, Qi L, Wang J, Xu H, Yang H, Liu L, Feng G, Zhang L. Activating Angiogenesis and Immunoregulation to Propel Bone Regeneration via Deferoxamine-Laden Mg-Mediated Tantalum Oxide Nanoplatform. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38709640 DOI: 10.1021/acsami.4c04316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Vascularization and inflammation management are essential for successful bone regeneration during the healing process of large bone defects assisted by artificial implants/fillers. Therefore, this study is devoted to the optimization of the osteogenic microenvironment for accelerated bone healing through rapid neovascularization and appropriate inflammation inhibition that were achieved by applying a tantalum oxide (TaO)-based nanoplatform carrying functional substances at the bone defect. Specifically, TaO mesoporous nanospheres were first constructed and then modified by functionalized metal ions (Mg2+) with the following deferoxamine (DFO) loading to obtain the final product simplified as DFO-Mg-TaO. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) revealed that the product was homogeneously dispersed hollow nanospheres with large specific surface areas and mesoporous shells suitable for loading Mg2+ and DFO. The biological assessments indicated that DFO-Mg-TaO could enhance the adhesion, proliferation, and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). The DFO released from DFO-Mg-TaO promoted angiogenetic activity by upregulating the expressions of hypoxia-inducible factor-1 (HIF-1α) and vascular endothelial growth factor (VEGF). Notably, DFO-Mg-TaO also displayed anti-inflammatory activity by reducing the expressions of pro-inflammatory factors, benefiting from the release of bioactive Mg2+. In vivo experiments demonstrated that DFO-Mg-TaO integrated with vascular regenerative, anti-inflammatory, and osteogenic activities significantly accelerated the reconstruction of bone defects. Our findings suggest that the optimized DFO-Mg-TaO nanospheres are promising as multifunctional fillers to speed up the bone healing process.
Collapse
Affiliation(s)
- Zheng Liu
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Leizhen Huang
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Lin Qi
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jing Wang
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Huilun Xu
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Hao Yang
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Limin Liu
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Ganjun Feng
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Li Zhang
- Analytical & Testing Center, Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| |
Collapse
|
10
|
Hajfathalian M, Mossburg KJ, Radaic A, Woo KE, Jonnalagadda P, Kapila Y, Bollyky PL, Cormode DP. A review of recent advances in the use of complex metal nanostructures for biomedical applications from diagnosis to treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1959. [PMID: 38711134 PMCID: PMC11114100 DOI: 10.1002/wnan.1959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/31/2024] [Accepted: 04/01/2024] [Indexed: 05/08/2024]
Abstract
Complex metal nanostructures represent an exceptional category of materials characterized by distinct morphologies and physicochemical properties. Nanostructures with shape anisotropies, such as nanorods, nanostars, nanocages, and nanoprisms, are particularly appealing due to their tunable surface plasmon resonances, controllable surface chemistries, and effective targeting capabilities. These complex nanostructures can absorb light in the near-infrared, enabling noteworthy applications in nanomedicine, molecular imaging, and biology. The engineering of targeting abilities through surface modifications involving ligands, antibodies, peptides, and other agents potentiates their effects. Recent years have witnessed the development of innovative structures with diverse compositions, expanding their applications in biomedicine. These applications encompass targeted imaging, surface-enhanced Raman spectroscopy, near-infrared II imaging, catalytic therapy, photothermal therapy, and cancer treatment. This review seeks to provide the nanomedicine community with a thorough and informative overview of the evolving landscape of complex metal nanoparticle research, with a specific emphasis on their roles in imaging, cancer therapy, infectious diseases, and biofilm treatment. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Diagnostic Tools > Diagnostic Nanodevices.
Collapse
Affiliation(s)
- Maryam Hajfathalian
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ 07102
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305
| | - Katherine J. Mossburg
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Allan Radaic
- School of Dentistry, University of California Los Angeles
| | - Katherine E. Woo
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305
| | - Pallavi Jonnalagadda
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yvonne Kapila
- School of Dentistry, University of California Los Angeles
| | - Paul L. Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University
| | - David P. Cormode
- Department of Radiology, Department of Bioengineering, University of Pennsylvania
| |
Collapse
|
11
|
Du Y, Zhao L, Geng Z, Huo Z, Li H, Shen X, Peng X, Yan R, Cui J, Jia S. Construction of catalase@hollow silica nanosphere: Catalase with immobilized but not rigid state for improving catalytic performances. Int J Biol Macromol 2024; 263:130381. [PMID: 38395291 DOI: 10.1016/j.ijbiomac.2024.130381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Enzyme immobilization usually make use of nanomaterials to hold up biocatalysis stability in various unamiable reaction conditions, but also lead large discount on enzyme activity. Thus, there are abundant researches focus on how to deal with the relation of enzyme molecules and supports. In this work, a new state of highly active enzymes has been established through facile and novel in situ immobilization and soft template removal method to construct enzyme contained hollow silica nanosphere (catalase@HSN) biocatalysts where enzymes in the cavity exhibit "immobilized but not rigid state". The obtained catalase@HSN was characterized by transmission electron microscopy, scanning electron microscopy and confocal laser scanning microscopy et al. Catalase@HSN exhibits excellent activity (about 80 % activity recovery rate) and stability suffers from extreme pH, temperature, and organic solvents. Moreover, the reusability and storage stability of catalase@HSN also are satisfactory. This proposed strategy provides a facile method for preparing biocatalysts under mild conditions, facilitating the applications of immobilized enzyme in the fields of real biocatalytic industry with high apparent activity and passable stability.
Collapse
Affiliation(s)
- Yingjie Du
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, China; Tianjin UBasio Biotechnology Group, China
| | - Lixue Zhao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China
| | - Zixin Geng
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China
| | - Zibei Huo
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China
| | - Huihui Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China
| | - Xuejian Shen
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China
| | - Xiaogang Peng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, China
| | - Renyi Yan
- Tianjin UBasio Biotechnology Group, China
| | - Jiandong Cui
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China.
| | - Shiru Jia
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, State Key Laboratory of Biobased Fiber Manufacturing Technology, Tianjin University of Science and Technology, China
| |
Collapse
|
12
|
Li J, Yue Z, Tang M, Wang W, Sun Y, Sun T, Chen C. Strategies to Reverse Hypoxic Tumor Microenvironment for Enhanced Sonodynamic Therapy. Adv Healthc Mater 2024; 13:e2302028. [PMID: 37672732 DOI: 10.1002/adhm.202302028] [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: 06/27/2023] [Revised: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Sonodynamic therapy (SDT) has emerged as a highly effective modality for the treatment of malignant tumors owing to its powerful penetration ability, noninvasiveness, site-confined irradiation, and excellent therapeutic efficacy. However, the traditional SDT, which relies on oxygen availability, often fails to generate a satisfactory level of reactive oxygen species because of the widespread issue of hypoxia in the tumor microenvironment of solid tumors. To address this challenge, various approaches are developed to alleviate hypoxia and improve the efficiency of SDT. These strategies aim to either increase oxygen supply or prevent hypoxia exacerbation, thereby enhancing the effectiveness of SDT. In view of this, the current review provides an overview of these strategies and their underlying principles, focusing on the circulation of oxygen from consumption to external supply. The detailed research examples conducted using these strategies in combination with SDT are also discussed. Additionally, this review highlights the future prospects and challenges of the hypoxia-alleviated SDT, along with the key considerations for future clinical applications. These considerations include the development of efficient oxygen delivery systems, the accurate methods for hypoxia detection, and the exploration of combination therapies to optimize SDT outcomes.
Collapse
Affiliation(s)
- Jialun Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Zhengya Yue
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Minglu Tang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Wenxin Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, 150076, P. R. China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chunxia Chen
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
13
|
Moloudi K, Abrahamse H, George BP. Nanotechnology-mediated photodynamic therapy: Focus on overcoming tumor hypoxia. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1937. [PMID: 38072393 DOI: 10.1002/wnan.1937] [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: 06/15/2023] [Revised: 10/05/2023] [Accepted: 11/20/2023] [Indexed: 03/09/2024]
Abstract
The oxygen level in the tumor is a critical marker that determines response to different treatments. Cancerous cells can adapt to hypoxia and low pH conditions within the tumor microenvironment (TME) to regulate tumor metabolism, proliferation, and promote tumor metastasis as well as angiogenesis, consequently leading to treatment failure and recurrence. In recent years, widespread attempts have been made to overcome tumor hypoxia through different methods, such as hyperbaric oxygen therapy (HBOT), hyperthermia, O2 carriers, artificial hemoglobin, oxygen generator hydrogels, and peroxide materials. While oxygen is found to be an essential agent to improve the treatment response of photodynamic therapy (PDT) and other cancer treatment modalities, the development of hypoxia within the tumor is highly associated with PDT failure. Recently, the use of nanoparticles has been a hot topic for researchers and exploited to overcome hypoxia through Oxygen-generating hydrogels, O2 nanocarriers, and O2 -generating nanoparticles. This review aimed to discuss the role of nanotechnology in tumor oxygenation and highlight the challenges, prospective, and recent advances in this area to improve PDT outcomes. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Kave Moloudi
- Laser Research Centre (LRC), Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre (LRC), Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Blassan P George
- Laser Research Centre (LRC), Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
14
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
15
|
Zhang D, Liu D, Wang C, Su Y, Zhang X. Nanoreactor-based catalytic systems for therapeutic applications: Principles, strategies, and challenges. Adv Colloid Interface Sci 2023; 322:103037. [PMID: 37931381 DOI: 10.1016/j.cis.2023.103037] [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: 08/02/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023]
Abstract
Inspired by natural catalytic compartments, various synthetic compartments that seclude catalytic reactions have been developed to understand complex multistep biosynthetic pathways, bestow therapeutic effects, or extend biosynthetic pathways in living cells. These emerging nanoreactors possessed many advantages over conventional biomedicine, such as good catalytic activity, specificity, and sustainability. In the past decade, a great number of efficient catalytic systems based on diverse nanoreactors (polymer vesicles, liposome, polymer micelles, inorganic-organic hybrid materials, MOFs, etc.) have been designed and employed to initiate in situ catalyzed chemical reactions for therapy. This review aims to present the recent progress in the development of catalytic systems based on nanoreactors for therapeutic applications, with a special emphasis on the principles and design strategies. Besides, the key components of nanoreactor-based catalytic systems, including nanocarriers, triggers or energy inputs, and products, are respectively introduced and discussed in detail. Challenges and prospects in the fabrication of therapeutic catalytic nanoreactors are also discussed as a conclusion to this review. We believe that catalytic nanoreactors will play an increasingly important role in modern biomedicine, with improved therapeutic performance and minimal side effects.
Collapse
Affiliation(s)
- Dan Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Dongcheng Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Chunfei Wang
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yanhong Su
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Xuanjun Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; MOE Frontiers Science Centre for Precision Oncology, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
16
|
Hu C, Qiu Y, Guo J, Cao Y, Li D, Du Y. An Oxygen Supply Strategy for Sonodynamic Therapy in Tuberculous Granuloma Lesions Using a Catalase-Loaded Nanoplatform. Int J Nanomedicine 2023; 18:6257-6274. [PMID: 37936950 PMCID: PMC10627092 DOI: 10.2147/ijn.s430019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Purpose Tuberculosis (TB) is a chronic disease caused by Mycobacterium tuberculosis (MTB) that remains a major global health challenge. One of the main obstacles to effective treatment is the heterogeneous microenvironment of TB granulomas. This study aimed to investigate the potential of a hypoxic remission-based strategy to enhance the outcome of tuberculosis treatment when implemented in combination with ultrasound. Methods A PLGA nanoparticle (LEV@CAT-NPs) loaded with levofloxacin (LEV) and catalase (CAT) was fabricated by a double emulsification method, and its physical characteristics, oxygen production capacity, drug release capacity, and biosafety were thoroughly investigated. The synergistic therapeutic effects of ultrasound (US)-mediated LEV@CAT-NPs were evaluated using an experimental mouse model of subcutaneous tuberculosis granuloma induced by Bacille Calmette-Guérin (BCG) as a substitute for MTB. Results LEV@CAT-NPs exhibited excellent oxygen production capacity, biosafety, and biocompatibility. Histological analysis revealed that ultrasound-mediated LEV@CAT-NPs could effectively remove bacteria from tuberculous granulomas, significantly alleviate the hypoxia state, reduce the necrotic area and inflammatory cells within the granuloma, and increase the penetration of dyes in granuloma tissues. The combined treatment also reduced the serum levels of inflammatory cytokines (eg, TNF-α, IL-6, and IL-8), and significantly downregulated the expression of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF). These results suggested that the synergistic treatment of ultrasound-mediated LEV@CAT-NPs effectively eradicated the bacterial infection and reversed the hypoxic microenvironment of tuberculous granulomas, further promoting tissue repair. Conclusion This study provides a non-invasive and new avenue for treating refractory tuberculosis infections. The potential role of regulating hypoxia within infected lesions as a therapeutic target for infection deserves further exploration in future studies.
Collapse
Affiliation(s)
- Can Hu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yan Qiu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jiajun Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yuchao Cao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Dairong Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
17
|
Ma T, Li W, Ye J, Huang C, Li Y, Qiu H, Yin S. GSH/pH dual response drug delivery system for photothermal enhanced gene-immunotherapy. NANOSCALE 2023; 15:16947-16958. [PMID: 37779508 DOI: 10.1039/d3nr03881e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Breast cancer has emerged as a leading cause of mortality among women. Photothermal therapy represents a recent therapeutic modality for eradicating localized tumors, albeit hindered by its limited penetration into tumor tissues. Recognizing the potential of photothermal therapy to induce immunogenic cell death in tumor cells, we explored a gene delivery approach utilizing small interfering RNA targeting programmed death ligand 1 (PD-L1), abbreviated as siPD-L1, to bolster the anti-tumor immune response elicited by this therapy. Nonetheless, the suboptimal release efficiency and inherent instability of RNA molecules have posed challenges to their therapeutic efficacy. In this study, we designed a glutathione (GSH)/pH-responsive micelle system, employing biocompatible and low-toxicity polyethyleneimine in conjunction with structurally robust pluronic P123, to encapsulate both indocyanine green (ICG) and siPD-L1 for precise targeting in breast cancer treatment. The resulting PSP/ICG/siPD-L1 nanocarrier demonstrated admirable biocompatibility and stability. Upon internalization into tumor cells, this nanocarrier exhibited rapid release of both ICG and siPD-L1, responding to the acidic tumor microenvironment and GSH conditions. The inclusion of siPD-L1 effectively downregulated the expression of PD-L1 on the tumor cell surface, thereby impeding tumor growth. Additionally, ICG demonstrated a photothermal effect when exposed to near-infrared light. Both in vitro and in vivo investigations substantiated the nanocarrier's efficacy against tumor cells, culminating in the complete ablation of 4T1 tumors in situ. Consequently, PSP/ICG/siPD-L1 emerges as a promising nanocarrier candidate for augmenting anti-tumor immunity through the synergistic combination of photothermal therapy and gene-based intervention.
Collapse
Affiliation(s)
- Tiantian Ma
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| | - Wen Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| | - Jingtao Ye
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| | - Chenchen Huang
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| | - Yang Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| | - Huayu Qiu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| | - Shouchun Yin
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou 311121, Zhejiang Province, P. R. China.
| |
Collapse
|
18
|
Moghassemi S, Dadashzadeh A, Jafari H, Ghaffari-Bohlouli P, Shavandi A, Amorim CA. Liposomal oxygen-generating hydrogel for enhancing cell survival under hypoxia condition. Colloids Surf B Biointerfaces 2023; 231:113562. [PMID: 37774524 DOI: 10.1016/j.colsurfb.2023.113562] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/06/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023]
Abstract
The inadequate oxygen supply to engineered tissues has been a persistent challenge in tissue engineering and regenerative medicine. To overcome this limitation, we developed a scaffold combined with an oxygen-releasing liposomal system comprising catalase-loaded liposomes (CAT@Lip) and H2O2-loaded liposomes (H2O2@Lip). This oxygenation system has shown high cytocompatibility when they were applied to human stromal cells. Under hypoxic conditions, the cell viability enclosed in the oxygen-releasing liposomal alginate hydrogel (94.62 ± 3.46 %) was significantly higher than that of cells enclosed in hydrogel without liposomes (47.18 ± 9.68 %). There was no significant difference in cell viability and apoptosis rate compared to normoxia conditions after three days, indicating the effectiveness of the oxygen-releasing approach in hypoxic conditions. In conclusion, our study demonstrates that the use of liposomal oxygen-releasing scaffolds can overcome the oxygen diffusion challenge in tissue implant fabrication, providing a simple solution for cellular oxygenation that could be a crucial element in tissue engineering.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hafez Jafari
- BioMatter unit - École polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Pejman Ghaffari-Bohlouli
- BioMatter unit - École polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Amin Shavandi
- BioMatter unit - École polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
19
|
Najafi A, Keykhaee M, Kazemi MH, Karimi MY, Khorramdelazad H, Aghamohamadi N, Bolouri MR, Ghaffari-Nazari H, Mirsharif ES, Karimi M, Dehghan Manshadi HR, Mahdavi SR, Safari E, Jalali SA, Falak R, Khoobi M. Catalase-gold nanoaggregates manipulate the tumor microenvironment and enhance the effect of low-dose radiation therapy by reducing hypoxia. Biomed Pharmacother 2023; 167:115557. [PMID: 37757491 DOI: 10.1016/j.biopha.2023.115557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Radiotherapy as a standard method for cancer treatment faces tumor recurrence and antitumoral unresponsiveness. Suppressive tumor microenvironment (TME) and hypoxia are significant challenges affecting efficacy of radiotherapy. Herein, a versatile method is introduced for the preparation of pH-sensitive catalase-gold cross-linked nanoaggregate (Au@CAT) having acceptable stability and selective activity in tumor microenvironment. Combining Au@CAT with low-dose radiotherapy enhanced radiotherapy effects via polarizing protumoral immune cells to the antitumoral landscape. This therapeutic approach also attenuated hypoxia, confirmed by downregulating hypoxia hallmarks, such as hypoxia-inducible factor α-subunits (HIF-α), vascular endothelial growth factor (VEGF), and EGF. Catalase stability against protease digestion was improved significantly in Au@CAT compared to the free catalase. Moreover, minimal toxicity of Au@CAT on normal cells and increased reactive oxygen species (ROS) were confirmed in vitro compared with radiotherapy. Using the nanoaggregates combined with radiotherapy led to a significant reduction of immunosuppressive infiltrating cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (T-regs) compared to the other groups. While, this combined therapy could significantly increase the frequency of CD8+ cells as well as M1 to M2 macrophages (MQs) ratio. The combination therapy also reduced the tumor size and increased survival rate in mice models of colorectal cancer (CRC). Our results indicate that this innovative nanocomposite could be an excellent system for catalase delivery, manipulating the TME and providing a potential therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Alireza Najafi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Keykhaee
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khorramdelazad
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nazanin Aghamohamadi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Bolouri
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Ghaffari-Nazari
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Milad Karimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Seied Rabi Mahdavi
- Radiation Biology Research Center& Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Jalali
- Immunology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Wang Z, Wang D, Ren X, Liu Z, Liu A, Li X, Guan L, Shen Y, Jin S, Zvyagin AV, Yang B, Wang T, Lin Q. One Stone, Three Birds: Multifunctional Nanodots as "Pilot Light" for Guiding Surgery, Enhanced Radiotherapy, and Brachytherapy of Tumors. ACS CENTRAL SCIENCE 2023; 9:1976-1988. [PMID: 37901175 PMCID: PMC10604975 DOI: 10.1021/acscentsci.3c00994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 10/31/2023]
Abstract
Surgery, radiotherapy (RT), and brachytherapy are crucial treatments for localized deep tumors. However, imprecise tumor location often leads to issues such as positive surgical margins, extended radiotherapy target volumes, and radiation damage to healthy tissues. Reducing side effects in healthy tissue and enhancing RT efficacy are critical challenges. To address these issues, we developed a multifunctional theranostic platform using Au/Ag nanodots (Au/AgNDs) that act as a "pilot light" for real-time guided surgery, high-efficiency RT, and brachytherapy, achieving a strategy of killing three birds with one stone. First, dual-mode imaging of Au/AgNDs enabled precision RT, minimizing damage to adjacent normal tissue during X-ray irradiation. Au/AgNDs enhanced ionizing radiation energy deposition, increased intracellular reactive oxygen species (ROS) generation, regulated the cell cycle, promoted DNA damage formation, and inhibited DNA repair in tumor cells, significantly improving RT efficacy. Second, in brachytherapy, precise guidance provided by dual-mode imaging addressed challenges related to non-visualization of existing interstitial brachytherapy and multiple adjustments of insertion needle positions. Meanwhile, the effect of brachytherapy was improved. Third, the excellent fluorescence imaging of Au/AgNDs accurately distinguished tumors from normal tissue, facilitating their use as a powerful tool for assisting surgeons during tumor resection. Taken together, our multifunctional theranostic platform offers real-time guidance for surgery and high-efficiency RT, and improves brachytherapy precision, providing a novel strategy and vision for the clinical diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Ze Wang
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Dongzhou Wang
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Xiaojun Ren
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Zhongshan Liu
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Annan Liu
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xingchen Li
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lin Guan
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yannan Shen
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Shunzi Jin
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Andrei V. Zvyagin
- Australian
Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute
of Biology and Biomedicine, Lobachevsky
Nizhny Novgorod State University, 603105 Nizhny Novgorod, Russia
| | - Bai Yang
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Tiejun Wang
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Quan Lin
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
21
|
Lei H, Pei Z, Jiang C, Cheng L. Recent progress of metal-based nanomaterials with anti-tumor biological effects for enhanced cancer therapy. EXPLORATION (BEIJING, CHINA) 2023; 3:20220001. [PMID: 37933288 PMCID: PMC10582613 DOI: 10.1002/exp.20220001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/06/2022] [Indexed: 11/08/2023]
Abstract
Metal-based nanomaterials have attracted broad attention recently due to their unique biological physical and chemical properties after entering tumor cells, namely biological effects. In particular, the abilities of Ca2+ to modulate T cell receptors activation, K+ to regulate stem cell differentiation, Mn2+ to activate the STING pathway, and Fe2+/3+ to induce tumor ferroptosis and enhance catalytic therapy, make the metal ions and metal-based nanomaterials play crucial roles in the cancer treatments. Therefore, due to the superior advantages of metal-based nanomaterials and the characteristics of the tumor microenvironment, we will summarize the recent progress of the anti-tumor biological effects of metal-based nanomaterials. Based on the different effects of metal-based nanomaterials on tumor cells, this review mainly focuses on the following five aspects: (1) metal-enhanced radiotherapy sensitization, (2) metal-enhanced catalytic therapy, (3) metal-enhanced ferroptosis, (4) metal-enhanced pyroptosis, and (5) metal-enhanced immunotherapy. At the same time, the shortcomings of the biological effects of metal-based nanomaterials on tumor therapy are also discussed, and the future research directions have been prospected. The highlights of promising biosafety, potent efficacy on biological effects for tumor therapy, and the in-depth various biological effects mechanism studies of metal-based nanomaterials provide novel ideas for the future biological application of the nanomaterials.
Collapse
Affiliation(s)
- Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| | - Chenyu Jiang
- School of Optical and Electronic InformationSuzhou City UniversitySuzhouChina
- Department of ChemistryNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| |
Collapse
|
22
|
Liao H, Yang S, Liang Z, Xiao L, Xie S, Lin P, Xia F, Fang C, Chen Q, Ling D, Li F. A Cancer Cell Selective Replication Stress Nano Amplifier Promotes Replication Fork Catastrophe to Overcome Radioresistance. ACS NANO 2023; 17:18548-18561. [PMID: 37706454 DOI: 10.1021/acsnano.3c06774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Replication stress (RS) induced by DNA damage plays a significant role in conferring the anticancer effects of radiotherapy and is tightly associated with radioresistance of cancer cells. Amplification of RS represents an effective approach to improving the efficacy of radiotherapy, although the development of selective RS amplifiers remains an unexplored frontier. We herein present an RS nano amplifier (RSNA) consisting of a catalytic FePt nanoparticle loaded with the chemotherapeutic doxorubicin (DOX), which selectively exacerbates RS in cancer cells by promoting replication fork (RF) catastrophe. RSNA converts the excessive reactive oxygen species (ROS) in cancer cells into oxygen, enhancing the DNA-damaging effects of radiotherapy to create more template lesions that impede RF progression in coalition with DOX. After radiation, ROS scavenging by RSNA accelerates RF progression through damaged template strands, increasing the frequency of RF collapse into double-strand breaks. Moreover, pretreatment with RSNA accumulates cancer cells in the S phase, exposing more RFs to radiation-induced RS. These effects of RSNA convergently maximize RS in cancer cells, effectively overcoming the radioresistance of cancer cells without affecting normal cells. Our study demonstrates the feasibility of selectively amplifying RS to boost radiotherapy.
Collapse
Affiliation(s)
- Hongwei Liao
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shengfei Yang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zeyu Liang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lin Xiao
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shangzhi Xie
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Lin
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daishun Ling
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- WLA Laboratories, Shanghai 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- WLA Laboratories, Shanghai 201203, China
| |
Collapse
|
23
|
Wang YP, Duan XH, Huang YH, Hou YJ, Wu K, Zhang F, Pan M, Shen J, Su CY. Radio- and Photosensitizing Os(II)-Based Nanocage for Combined Radio-/Chemo-/X-ray-Induced Photodynamic Therapies, NIR Imaging, and Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43479-43491. [PMID: 37694454 DOI: 10.1021/acsami.3c08503] [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: 09/12/2023]
Abstract
Integration of clinical imaging and collaborative multimodal therapies into a single nanomaterial for multipurpose diagnosis and treatment is of great interest to theranostic nanomedicine. Here, we report a rational design of a discrete Os-based metal-organic nanocage Pd6(OsL3)828+ (MOC-43) as a versatile theranostic nanoplatform to meet the following demands simultaneously: (1) synergistic treatments of radio-, chemo-, and X-ray-induced photodynamic therapies (X-PDT) for breast cancer, (2) NIR imaging for cancer cell tracking and tumor-targeting, and (3) anticancer drug transport through a host-guest strategy. The nanoscale MOC-43 incorporates high-Z Os-element to interact with X-ray irradiation for dual radiosensitization and photosensitization, showing efficient energy transfer to endogenous oxygen in cancer cells to enhance X-PDT efficacy. It also features intrinsic NIR emission originating from metal-to-ligand charge transfer (MLCT) as an excellent imaging probe. Meanwhile, its 12 pockets can capture and concentrate low-water-soluble molecules for anticancer drug delivery. These multifunctions are implemented and demonstrated by micellization of coumarin-loaded cages with DSPE-PEG2000 into coumarin ⊂ MOC-43 nanoparticles (CMNPs) for efficient subcellular endocytosis and uptake. The cancer treatments in vitro/in vivo show promising antitumor performance, providing a conceptual protocol to combine cage-cargo drug transport with diagnosis and treatment for collaborative cancer theranostics by virtue of multifunction synergism on a single-nanomaterial platform.
Collapse
Affiliation(s)
- Ya-Ping Wang
- MOE Laboratory of Bioinorganic and Synthetic Chemistry, Lehn Institute of Functional Materials, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiao-Hui Duan
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510030, China
| | - Yin-Hui Huang
- MOE Laboratory of Bioinorganic and Synthetic Chemistry, Lehn Institute of Functional Materials, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Ya-Jun Hou
- MOE Laboratory of Bioinorganic and Synthetic Chemistry, Lehn Institute of Functional Materials, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Kai Wu
- MOE Laboratory of Bioinorganic and Synthetic Chemistry, Lehn Institute of Functional Materials, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Fang Zhang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510030, China
| | - Mei Pan
- MOE Laboratory of Bioinorganic and Synthetic Chemistry, Lehn Institute of Functional Materials, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jun Shen
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510030, China
| | - Cheng-Yong Su
- MOE Laboratory of Bioinorganic and Synthetic Chemistry, Lehn Institute of Functional Materials, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
24
|
Kim S, Sundaram A, Mathew AP, Hareshkumar VS, Mohapatra A, Thomas RG, Bui TTM, Moon K, Kweon S, Park IK, Jeong YY. In situ hypoxia modulating nano-catalase for amplifying DNA damage in radiation resistive colon tumors. Biomater Sci 2023; 11:6177-6192. [PMID: 37504889 DOI: 10.1039/d3bm00618b] [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: 07/29/2023]
Abstract
Radiation therapy (RT) is a mainstream clinical approach in cancer treatment. However, the therapeutic efficacy of RT is greatly hindered by the presence of excessive hydrogen peroxide (H2O2) in the hypoxic region of the solid tumor, thus leading to tumor recurrence and metastasis. Herein, a thioketal-linked amphiphilic nano-assembly (MTS) loaded with hydrophobic manganese oxide (HMO) nanoparticles (MTS@HMO) is examined as a promising multi-purpose reactive oxygen species (ROS)-catalytic nanozyme for transforming an RT-resistant hypoxic tumor microenvironment (TME) into an RT-susceptible one by scavenging ROS in the hypoxic core of the solid tumor. After intravenous injection, the MTS@HMO nano-assembly was able to sense and be degraded by the abundant ROS in the hypoxic TME, thereby releasing HMO particles for subsequent scavenging of H2O2. The oxygen generated during peroxide scavenging then relieved the hypoxic TME, thereby resulting in an increased sensitivity of the hypoxic tumor tissue towards RT. Moreover, the in situ hypoxic status was monitored via the T1-enhanced magnetic resonance (MR) imaging of the Mn2+ ions generated by the ROS-mediated degradation of HMO. The in vitro results demonstrated a significant H2O2 elimination and enhanced oxygen generation after the treatment of the MTS@HMO nano-assembly with tumor cells under hypoxic conditions, compared to the control MTS group. In addition, the combination of RT and pre-treatment with MTS@HMO nano-assembly significantly amplified the permanent DNA strand breaks in tumor cells compared to the control RT group. More importantly, the in vivo results proved that the systemic injection of the MTS@HMO nano-assembly prior to RT irradiation enhanced the RT-mediated tumor suppression and down-regulated the hypoxic marker of HIF-1α in the solid tumor compared to the control RT group. Overall, the present work demonstrates the great potential of the versatile ROS-catalytic hypoxia modulating strategy using the MTS@HMO nano-assembly to enhance the RT-induced antitumor efficacy in hypoxic solid tumors.
Collapse
Affiliation(s)
- Subin Kim
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
- Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- DR Cure Inc., Hwasun 58128, Republic of Korea
| | - Aravindkumar Sundaram
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Ansuja Pulickal Mathew
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Vasvani Shyam Hareshkumar
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
- Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- DR Cure Inc., Hwasun 58128, Republic of Korea
| | - Adityanarayan Mohapatra
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Reju George Thomas
- Department of Radiology, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea.
| | - Thinh T M Bui
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, South Korea
| | - Kyuho Moon
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, South Korea
| | - Seho Kweon
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
- Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- DR Cure Inc., Hwasun 58128, Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea.
- DR Cure Inc., Hwasun 58128, Republic of Korea
| |
Collapse
|
25
|
Huang L, Su Y, Zhang D, Zeng Z, Hu X, Hong S, Lin X. Recent theranostic applications of hydrogen peroxide-responsive nanomaterials for multiple diseases. RSC Adv 2023; 13:27333-27358. [PMID: 37705984 PMCID: PMC10496458 DOI: 10.1039/d3ra05020c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023] Open
Abstract
It is well established that hydrogen peroxide (H2O2) is associated with the initiation and progression of many diseases. With the rapid development of nanotechnology, the diagnosis and treatment of those diseases could be realized through a variety of H2O2-responsive nanomaterials. In order to broaden the application prospects of H2O2-responsive nanomaterials and promote their development, understanding and summarizing the design and application fields of such materials has attracted much attention. This review provides a comprehensive summary of the types of H2O2-responsive nanomaterials including organic, inorganic and organic-inorganic hybrids in recent years, and focused on their specific design and applications. Based on the type of disease, such as tumors, bacteria, dental diseases, inflammation, cardiovascular diseases, bone injury and so on, key examples for above disease imaging diagnosis and therapy strategies are introduced. In addition, current challenges and the outlook of H2O2-responsive nanomaterials are also discussed. This review aims to stimulate the potential of H2O2-responsive nanomaterials and provide new application ideas for various functional nanomaterials related to H2O2.
Collapse
Affiliation(s)
- Linjie Huang
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Yina Su
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Dongdong Zhang
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Zheng Zeng
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Xueqi Hu
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Shanni Hong
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Xiahui Lin
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| |
Collapse
|
26
|
Zhuo X, Liu Z, Aishajiang R, Wang T, Yu D. Recent Progress of Copper-Based Nanomaterials in Tumor-Targeted Photothermal Therapy/Photodynamic Therapy. Pharmaceutics 2023; 15:2293. [PMID: 37765262 PMCID: PMC10534922 DOI: 10.3390/pharmaceutics15092293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Nanotechnology, an emerging and promising therapeutic tool, may improve the effectiveness of phototherapy (PT) in antitumor therapy because of the development of nanomaterials (NMs) with light-absorbing properties. The tumor-targeted PTs, such as photothermal therapy (PTT) and photodynamic therapy (PDT), transform light energy into heat and produce reactive oxygen species (ROS) that accumulate at the tumor site. The increase in ROS levels induces oxidative stress (OS) during carcinogenesis and disease development. Because of the localized surface plasmon resonance (LSPR) feature of copper (Cu), a vital trace element in the human body, Cu-based NMs can exhibit good near-infrared (NIR) absorption and excellent photothermal properties. In the tumor microenvironment (TME), Cu2+ combines with H2O2 to produce O2 that is reduced to Cu1+ by glutathione (GSH), causing a Fenton-like reaction that reduces tumor hypoxia and simultaneously generates ROS to eliminate tumor cells in conjunction with PTT/PDT. Compared with other therapeutic modalities, PTT/PDT can precisely target tumor location to kill tumor cells. Moreover, multiple treatment modalities can be combined with PTT/PDT to treat a tumor using Cu-based NMs. Herein, we reviewed and briefly summarized the mechanisms of actions of tumor-targeted PTT/PDT and the role of Cu, generated from Cu-based NMs, in PTs. Furthermore, we described the Cu-based NMs used in PTT/PDT applications.
Collapse
Affiliation(s)
| | | | | | - Tiejun Wang
- Department of Radiotherapy, The Second Affiliated Hospital of Jilin University, Changchun 130062, China; (X.Z.); (Z.L.); (R.A.)
| | - Duo Yu
- Department of Radiotherapy, The Second Affiliated Hospital of Jilin University, Changchun 130062, China; (X.Z.); (Z.L.); (R.A.)
| |
Collapse
|
27
|
Ding S, Chen L, Liao J, Huo Q, Wang Q, Tian G, Yin W. Harnessing Hafnium-Based Nanomaterials for Cancer Diagnosis and Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300341. [PMID: 37029564 DOI: 10.1002/smll.202300341] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/01/2023] [Indexed: 06/19/2023]
Abstract
With the rapid development of nanotechnology and nanomedicine, there are great interests in employing nanomaterials to improve the efficiency of disease diagnosis and treatment. The clinical translation of hafnium oxide (HfO2 ), commercially namedas NBTXR3, as a new kind of nanoradiosensitizer for radiotherapy (RT) of cancers has aroused extensive interest in researches on Hf-based nanomaterials for biomedical application. In the past 20 years, Hf-based nanomaterials have emerged as potential and important nanomedicine for computed tomography (CT)-involved bioimaging and RT-associated cancer treatment due to their excellent electronic structures and intrinsic physiochemical properties. In this review, a bibliometric analysis method is employed to summarize the progress on the synthesis technology of various Hf-based nanomaterials, including HfO2 , HfO2 -based compounds, and Hf-organic ligand coordination hybrids, such as metal-organic frameworks or nanoscaled coordination polymers. Moreover, current states in the application of Hf-based CT-involved contrasts for tissue imaging or cancer diagnosis are reviewed in detail. Importantly, the recent advances in Hf-based nanomaterials-mediated radiosensitization and synergistic RT with other current mainstream treatments are also generalized. Finally, current challenges and future perspectives of Hf-based nanomaterials with a view to maximize their great potential in the research of translational medicine are also discussed.
Collapse
Affiliation(s)
- Shuaishuai Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education of China, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Lei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jing Liao
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education of China, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- Laboratory for Micro-sized Functional Materials, Department of Chemistry and College of Elementary Education, Capital Normal University, Beijing, 100048, P. R. China
| | - Qing Huo
- College of Biochemical and Engineering, Beijing Union University, Beijing, 100023, China
| | - Qiang Wang
- Laboratory for Micro-sized Functional Materials, Department of Chemistry and College of Elementary Education, Capital Normal University, Beijing, 100048, P. R. China
| | - Gan Tian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education of China, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing, 401329, P. R. China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
28
|
Cao Y, Si J, Zheng M, Zhou Q, Ge Z. X-ray-responsive prodrugs and polymeric nanocarriers for multimodal cancer therapy. Chem Commun (Camb) 2023. [PMID: 37318285 DOI: 10.1039/d3cc01398g] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Radiotherapy as one of the most important cancer treatment modalities has been widely used in the therapy of various cancers. The clinically used radiation (e.g. X-ray) for radiotherapy has the advantages of precise spatiotemporal controllability and deep tissue penetration. However, traditional radiotherapy is frequently limited by the high side effects and tumor hypoxia. The combination of radiotherapy and other cancer treatment modalities may overcome the disadvantages of radiotherapy and improve the final therapeutic efficacy. In recent years, X-ray-activable prodrugs and polymeric nanocarriers have been extensively explored to introduce other treatment modalities in the precise position during radiotherapy, which can reduce the side toxicity of the drugs and improve the combination therapeutic efficacy. In this review, we focus on recent advances in X-ray-activable prodrugs and polymeric nanocarriers to boost X-ray-based multimodal synergistic therapy with reduced toxicity. The design strategies of prodrugs and polymeric nanocarriers are highlighted. Finally, challenges and outlooks of X-ray-activable prodrugs and polymeric nanocarriers are discussed.
Collapse
Affiliation(s)
- Yufei Cao
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Jiale Si
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Moujiang Zheng
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Qinghao Zhou
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Zhishen Ge
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
29
|
Zhen W, Weichselbaum RR, Lin W. Nanoparticle-Mediated Radiotherapy Remodels the Tumor Microenvironment to Enhance Antitumor Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206370. [PMID: 36524978 PMCID: PMC10213153 DOI: 10.1002/adma.202206370] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/12/2022] [Indexed: 05/26/2023]
Abstract
Radiotherapy (RT) uses ionizing radiation to eradicate localized tumors and, in rare cases, control tumors outside of the irradiated fields via stimulating an antitumor immune response (abscopal effect). However, the therapeutic effect of RT is often limited by inherent physiological barriers of the tumor microenvironment (TME), such as hypoxia, abnormal vasculature, dense extracellular matrix (ECM), and an immunosuppressive TME. Thus, it is critical to develop new RT strategies that can remodel the TME to overcome radio-resistance and immune suppression. In the past decade, high-Z-element nanoparticles have been developed to increase radiotherapeutic indices of localized tumors by reducing X-ray doses and side effects to normal tissues and enhance abscopal effects by activating the TME to elicit systemic antitumor immunity. In this review, the principles of RT and radiosensitization, the mechanisms of radio-resistance and immune suppression, and the use of various nanoparticles to sensitize RT and remodel TMEs for enhanced antitumor efficacy are discussed. The challenges in clinical translation of multifunctional TME-remodeling nanoradiosensitizers are also highlighted.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
30
|
Wen X, Bi S, Zeng S. NIR-II Light-Activated Gold Nanorods for Synergistic Thermodynamic and Photothermal Therapy of Tumor. ACS APPLIED BIO MATERIALS 2023; 6:1934-1942. [PMID: 37032485 DOI: 10.1021/acsabm.3c00134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
There are tricky challenges in tumor therapy due to the hypoxic tumor microenvironment, inevitably inhibiting the treatment efficacy of the traditional photodynamic therapy (PDT), radiation therapy (RT), and sonodynamic therapy (SDT). Herein, to overcome tumor hypoxia limitation, we constructed a near-infrared II (NIR-II) light-triggered thermodynamic therapy (TDT) nanoplatform of Au@mSiO2-AIPH@PCM/PEG (ASAPP) by integrating the Au nanorods (Au NRs) and thermally activated alkyl free radical-releasing molecules (AIPH). Au NRs@mSiO2 was used as a photothermally responsive material and AIPH carrier, and the hot-melt phase-change material (PCM) was used as a capping agent to prevent leakage of AIPH during blood circulation. Upon NIR-II light irradiation, heat-triggered free radical release from AIPH was successfully achieved for killing cancer cells in vitro and in vivo without oxygen dependence, leading to synergistically enhanced antitumor therapy.
Collapse
Affiliation(s)
- Xingwang Wen
- School of Physics and Electronics, Key Laboratory of Low-dimensional Quantum Structures and Quantum Control of Ministry of Education, Key Laboratory for Matter Microstructure and Function of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, P. R. China
| | - Shenghui Bi
- School of Physics and Electronics, Key Laboratory of Low-dimensional Quantum Structures and Quantum Control of Ministry of Education, Key Laboratory for Matter Microstructure and Function of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, P. R. China
| | - Songjun Zeng
- School of Physics and Electronics, Key Laboratory of Low-dimensional Quantum Structures and Quantum Control of Ministry of Education, Key Laboratory for Matter Microstructure and Function of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, P. R. China
| |
Collapse
|
31
|
Zhang M, Hu S, Liu L, Dang P, Liu Y, Sun Z, Qiao B, Wang C. Engineered exosomes from different sources for cancer-targeted therapy. Signal Transduct Target Ther 2023; 8:124. [PMID: 36922504 PMCID: PMC10017761 DOI: 10.1038/s41392-023-01382-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/31/2023] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Exosome is a subgroup of extracellular vesicles, which has been serving as an efficient therapeutic tool for various diseases. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. After appropriate modification, engineered exosomes are able to deliver antitumor drugs to tumor sites efficiently and precisely with fewer treatment-related adverse effects. However, there still exist many challenges for the clinical translation of engineered exosomes. For instance, what sources and modification strategies could endow exosomes with the most efficient antitumor activity is still poorly understood. Additionally, how to choose appropriately engineered exosomes in different antitumor therapies is another unresolved problem. In this review, we summarized the characteristics of engineered exosomes, especially the spatial and temporal properties. Additionally, we concluded the recent advances in engineered exosomes in the cancer fields, including the sources, isolation technologies, modification strategies, and labeling and imaging methods of engineered exosomes. Furthermore, the applications of engineered exosomes in different antitumor therapies were summarized, such as photodynamic therapy, gene therapy, and immunotherapy. Consequently, the above provides the cancer researchers in this community with the latest ideas on engineered exosome modification and new direction of new drug development, which is prospective to accelerate the clinical translation of engineered exosomes for cancer-targeted therapy.
Collapse
Affiliation(s)
- Menghui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Pengyuan Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, 450001, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Bingbing Qiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
32
|
Zu Y, Wang Z, Yao H, Yan L. Oxygen-generating biocatalytic nanomaterials for tumor hypoxia relief in cancer radiotherapy. J Mater Chem B 2023; 11:3071-3088. [PMID: 36920849 DOI: 10.1039/d2tb02751h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Radiotherapy (RT), the most commonly used treatment method in clinics, shows unique advantages such as strong penetration, high energy intensity, and low systemic side effects. However, in vivo tumor hypoxia seriously hinders the therapeutic effect of RT. Hypoxia is a common characteristic of locally advanced solid tumor microenvironments, which leads to the proliferation, invasion and metastasis of tumor cells. In addition, oxygen consumption during RT will further aggravate tumor hypoxia, causing a variety of adverse side effects. In recent years, various biocatalytic nanomaterials (BCNs) have been explored to regulate and reverse tumor hypoxia microenvironments during RT. In this review, the most recent efforts toward developing oxygen-generating BCNs in relieving tumor hypoxia in RT are focused upon. The classification, engineering nanocatalytical activity of oxygen-generating BCNs and combined therapy based on these BCNs are systematically introduced and discussed. The challenges and prospects of these oxygen-generating BCNs in RT applications are also summarized.
Collapse
Affiliation(s)
- Yan Zu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.
| | - Ziyu Wang
- College of Medical and Biological lnformation Engineering, Northeastern University, Shenyang 110170, China
| | - Huiqin Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| | - Liang Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
33
|
Deng Z, Xi M, Zhang C, Wu X, Li Q, Wang C, Fang H, Sun G, Zhang Y, Yang G, Liu Z. Biomineralized MnO 2 Nanoplatforms Mediated Delivery of Immune Checkpoint Inhibitors with STING Pathway Activation to Potentiate Cancer Radio-Immunotherapy. ACS NANO 2023; 17:4495-4506. [PMID: 36848115 DOI: 10.1021/acsnano.2c10352] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Radiotherapy (RT), as one of the main methods in the clinical treatment of various malignant tumors, would induce systemic immunotherapeutic effects by triggering immunogenic cell death (ICD) of cancer cells. However, the antitumor immune responses produced by RT-induced ICD alone usually are not robust enough to eliminate distant tumors and thus ineffective against cancer metastases. Herein, a biomimetic mineralization method for facile synthesis of MnO2 nanoparticles with high anti-programmed death ligand 1 (αPDL1) encapsulation efficiency (αPDL1@MnO2) is proposed to reinforce RT-induced systemic antitumor immune responses. This therapeutic nanoplatforms-mediated RT can significantly improve the killing of tumor cells and effectively evoke ICD by overcoming hypoxia-induced radio-resistance and reprogramming the immunosuppressive tumor microenvironment (TME). Furthermore, the released Mn2+ ions from αPDL1@MnO2 under acidic tumor pH can activate the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and facilitate the dendritic cells (DCs) maturation. Meanwhile, αPDL1 released from αPDL1@MnO2 nanoparticles would further promote the intratumoral infiltration of cytotoxic T lymphocytes (CTLs) and trigger systemic antitumor responses, resulting in a strong abscopal effect to effectively inhibit tumor metastases. Overall, the biomineralized MnO2-based nanoplatforms offer a simple strategy for TME modulation and immune activation, which are promising for enhanced RT immunotherapy.
Collapse
Affiliation(s)
- Zheng Deng
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Min Xi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xirui Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Quguang Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chunjie Wang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huapan Fang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guanting Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yifan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
34
|
Zhao J, Chen Y, Xiong T, Han S, Li C, He Y, He Y, Zhao G, Wang T, Wang L, Cheng T, Wang C, Wang J. Clustered Cobalt Nanodots Initiate Ferroptosis by Upregulating Heme Oxygenase 1 for Radiotherapy Sensitization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206415. [PMID: 36627264 DOI: 10.1002/smll.202206415] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/18/2022] [Indexed: 06/17/2023]
Abstract
High cobalt (Co) levels in tumors are associated with good clinical prognosis. An anticancer regimen that increases intratumoral Co through targeted nanomaterial delivery is proposed in this study. Bovine serum albumin and cobalt dichloride are applied to prepare cobaltous oxide nanodots using a facile biomineralization strategy. After iRGD peptide conjugation, the nanodots are loaded into dendritic mesoporous silica nanoparticles, generating a biocompatible product iCoDMSN. This nanocomposite accumulates in tumors after intravenous injection by deep tissue penetration and can be used for photoacoustic imaging. Proteomics research and molecular biology experiments reveal that iCoDMSN is a potent ferroptosis inducer in cancer cells. Mechanistically, iCoDMSNs upregulate heme oxygenase 1 (HMOX1), which increases transferrin receptors and reduces solute carrier family 40 member 1 (SLC40A1), resulting in Fe2+ accumulation and ferroptosis initiation. Furthermore, upregulated nuclear factor erythroid 2-related factor 2 (NRF2), arising from the reduction in Kelch-like ECH-associated protein 1 (KEAP1) expression, is responsible for HMOX1 enhancement after iCoDMSN treatment. Owing to intensified ferroptosis, iCoDMSN acts as an efficient radiotherapy enhancer to eliminate cancer cells in vitro and in vivo. This study demonstrates a versatile Co-based nanomaterial that primes ferroptosis by expanding the labile iron pool in cancer cells, providing a promising tumor radiotherapy sensitizer.
Collapse
Affiliation(s)
- Jianqi Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Yin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Tainong Xiong
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Songling Han
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Chenwenya Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Yingjuan He
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Yongwu He
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Gaomei Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Tao Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Tianmin Cheng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Cheng Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| |
Collapse
|
35
|
Wang Z, Ren X, Wang D, Guan L, Li X, Zhao Y, Liu A, He L, Wang T, Zvyagin AV, Yang B, Lin Q. Novel strategies for tumor radiosensitization mediated by multifunctional gold-based nanomaterials. Biomater Sci 2023; 11:1116-1136. [PMID: 36601661 DOI: 10.1039/d2bm01496c] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Radiotherapy (RT) is one of the most effective and commonly used cancer treatments for malignant tumors. However, the existing radiosensitizers have a lot of side effects and poor efficacy, which limits the curative effect and further application of radiotherapy. In recent years, emerging nanomaterials have shown unique advantages in enhancing radiosensitization. In particular, gold-based nanomaterials, with high X-ray attenuation capacity, good biocompatibility, and promising chemical, electronic and optical properties, have become a new type of radiotherapy sensitizer. In addition, gold-based nanomaterials can be used as a carrier to load a variety of drugs and immunosuppressants; in particular, its photothermal therapy, photodynamic therapy and multi-mode imaging functions aid in providing excellent therapeutic effect in coordination with RT. Recently, many novel strategies of radiosensitization mediated by multifunctional gold-based nanomaterials have been reported, which provides a new idea for improving the efficacy and reducing the side effects of RT. In this review, we systematically summarize the recent progress of various new gold-based nanomaterials that mediate radiosensitization and describe the mechanism. We further discuss the challenges and prospects in the field. It is hoped that this review will help researchers understand the latest progress of gold-based nanomaterials for radiosensitization, and encourage people to optimize the existing methods or explore novel approaches for radiotherapy.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Xiaojun Ren
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Dongzhou Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Lin Guan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Xingchen Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Yue Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Liang He
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia.,Institute of Biology and Biomedicine, Lobachevsky Nizhny Novgorod State University, 603105, Nizhny Novgorod, Russia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| |
Collapse
|
36
|
Zhuang Y, Liu K, He Q, Gu X, Jiang C, Wu J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e203. [PMID: 36703877 PMCID: PMC9870816 DOI: 10.1002/mco2.203] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoxia is a persistent physiological feature of many different solid tumors and a key driver of malignancy, and in recent years, it has been recognized as an important target for cancer therapy. Hypoxia occurs in the majority of solid tumors due to a poor vascular oxygen supply that is not sufficient to meet the needs of rapidly proliferating cancer cells. A hypoxic tumor microenvironment (TME) can reduce the effectiveness of other tumor therapies, such as radiotherapy, chemotherapy, and immunotherapy. In this review, we discuss the critical role of hypoxia in tumor development, including tumor metabolism, tumor immunity, and tumor angiogenesis. The treatment methods for hypoxic TME are summarized, including hypoxia-targeted therapy and improving oxygenation by alleviating tumor hypoxia itself. Hyperoxia therapy can be used to improve tissue oxygen partial pressure and relieve tumor hypoxia. We focus on the underlying mechanisms of hyperoxia and their impact on current cancer therapies and discuss the prospects of hyperoxia therapy in cancer treatment.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Kua Liu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Qinyu He
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Xiaosong Gu
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| |
Collapse
|
37
|
Wang C, Wu L, Yuan H, Yu H, Xu J, Chen S, Yan S, Wang X. A powerful antitumor "trident": the combination of radio-, immuno- and anti-angiogenesis therapy based on mesoporous silica single coated gold nanoparticles. J Mater Chem B 2023; 11:879-889. [PMID: 36594928 DOI: 10.1039/d2tb02046g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although immunotherapy in combination with anti-angiogenesis therapy has made a breakthrough in the first-line treatment of cancer, considering the low responder rate and the adverse events, it is vital to propose a new combination modality. In this study, we report single encapsulated mesoporous silica coated gold nanoparticles that synergize sensitizing radiotherapy with the current combination therapy. Distinguished from simply combining two treatments, the nanoparticle-mediated "trident" therapy resolved the problem of matching the dose between radiation and drug, which determines the outcome since drug demand rises with immunosuppression from increased sensitivity to radiotherapy. The nanomedicine produced energy depositions when radiation was introduced, and released the loaded toripalimab and bevacizumab, exhibiting significant anti-tumor properties. In vitro tumor cell viability results indicated the highest inhibition by the "trident" therapy and in vivo animal models also revealed the earliest decrease in tumor tissue volume. As a result, the "trident" therapy is expected to further improve the anti-tumor benefits of the combination of immunotherapy and anti-angiogenesis therapy and provides a versatile perspective on cancer treatment.
Collapse
Affiliation(s)
- Cheng Wang
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| | - Lingyun Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Huili Yuan
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| | - Hao Yu
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Jiaqi Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Si Chen
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| | - Senxiang Yan
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Xu Wang
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| |
Collapse
|
38
|
Chen Y, Liu S, Liao Y, Yang H, Chen Z, Hu Y, Fu S, Wu J. Albumin-Modified Gold Nanoparticles as Novel Radiosensitizers for Enhancing Lung Cancer Radiotherapy. Int J Nanomedicine 2023; 18:1949-1964. [PMID: 37070100 PMCID: PMC10105590 DOI: 10.2147/ijn.s398254] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/01/2023] [Indexed: 04/19/2023] Open
Abstract
Background Considering the strong attenuation of photons and the potential to increase the deposition of radiation, high-atomic number nanomaterials are often used as radiosensitizers in cancer radiotherapy, of which gold nanoparticles (GNPs) are widely used. Materials and Methods We prepared albumin-modified GNPs (Alb-GNPs) and observed their radiosensitizing effects and biotoxicity in human non-small-cell lung carcinoma tumor-bearing mice models. Results The prepared nanoparticles (Alb-GNPs) demonstrated excellent colloidal stability and biocompatibility at the mean size of 205.06 ± 1.03 nm. Furthermore, clone formation experiments revealed that Alb-GNPs exerted excellent radiosensitization, with a sensitization enhancement ratio (SER) of 1.432, which is higher than X-ray alone. Our in vitro and in vivo data suggested that Alb-GNPs enabled favorable accumulation in tumors, and the combination of Alb-GNPs and radiotherapy exhibited a relatively greater radiosensitizing effect and anti-tumor activity. In addition, no toxicity or abnormal irritating response resulted from the application of Alb-GNPs. Conclusion Alb-GNPs can be used as an effective radiosensitizer to improve the efficacy of radiotherapy with minimal damage to healthy tissues.
Collapse
Affiliation(s)
- Yao Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Yin Liao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Hanshan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Zhuo Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Yuru Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Correspondence: Shaozhi Fu; Jingbo Wu, Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China, Tel/Fax +86 8303165696, Email ;
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, People’s Republic of China
| |
Collapse
|
39
|
Li Z, Wang Q, Huang X, Yang M, Zhou S, Li Z, Fang Z, Tang Y, Chen Q, Hou H, Li L, Fei F, Wang Q, Wu Y, Gong A. Lactate in the tumor microenvironment: A rising star for targeted tumor therapy. Front Nutr 2023; 10:1113739. [PMID: 36875841 PMCID: PMC9978120 DOI: 10.3389/fnut.2023.1113739] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Metabolic reprogramming is one of fourteen hallmarks of tumor cells, among which aerobic glycolysis, often known as the "Warburg effect," is essential to the fast proliferation and aggressive metastasis of tumor cells. Lactate, on the other hand, as a ubiquitous molecule in the tumor microenvironment (TME), is generated primarily by tumor cells undergoing glycolysis. To prevent intracellular acidification, malignant cells often remove lactate along with H+, yet the acidification of TME is inevitable. Not only does the highly concentrated lactate within the TME serve as a substrate to supply energy to the malignant cells, but it also works as a signal to activate multiple pathways that enhance tumor metastasis and invasion, intratumoral angiogenesis, as well as immune escape. In this review, we aim to discuss the latest findings on lactate metabolism in tumor cells, particularly the capacity of extracellular lactate to influence cells in the tumor microenvironment. In addition, we examine current treatment techniques employing existing medications that target and interfere with lactate generation and transport in cancer therapy. New research shows that targeting lactate metabolism, lactate-regulated cells, and lactate action pathways are viable cancer therapy strategies.
Collapse
Affiliation(s)
- Zhangzuo Li
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Mengting Yang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zhengrui Li
- School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhengzou Fang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yidan Tang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Qian Chen
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hanjin Hou
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Li
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fei Fei
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiaowei Wang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuqing Wu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
40
|
A macrophage membrane-coated mesoporous silica nanoplatform inhibiting adenosine A2AR via in situ oxygen supply for immunotherapy. J Control Release 2023; 353:535-548. [PMID: 36481693 DOI: 10.1016/j.jconrel.2022.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
Immunotherapy has achieved remarkable research outcomes and shows the potential to cure cancer. However, its therapeutic response is limited in terms of the immunosuppressive tumor microenvironment induced by hypoxia, in which the adenosinergic A2A receptor (A2AR) pathway is mainly participated. Here, we developed a novel core/shell structured nanoplatform composed of macrophage membrane-coated mesoporous silica nanoparticles which loaded catalase, doxorubicin (Dox), and resiquimod (R848), to promote the efficacy of immunotherapy. The nanoplatform is able to actively target the tumor site via ligand binding, and the A2AR of T regulatory (Treg) cells can further be blocked due to in situ oxygen production by hydrogen peroxide catalysis. Meanwhile, Dox and R848 released from the nanoplatform can induce immunogenic cell death and enhance the activation of dendritic cells (DCs), respectively. Thus, the improved microenvironment by A2AR blockade and the stimulation of the DCs to enhance the CD8+ T cells mediated immune response were achieved. Consequently, the expression of Treg cells decreased to 9.79% in tumor tissue and the inhibition rate of tumor growth reached 73.58%. Therefore, this nanoplatform provides a potential strategy for clinical application in cancer immunotherapy.
Collapse
|
41
|
Duan Y, Shen C, Zhang Y, Luo Y. Advanced diagnostic and therapeutic strategies in nanotechnology for lung cancer. Front Oncol 2022; 12:1031000. [PMID: 36568152 PMCID: PMC9767962 DOI: 10.3389/fonc.2022.1031000] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
As a highly invasive thoracic malignancy with increasing prevalence, lung cancer is also the most lethal cancer worldwide due to the failure of effective early detection and the limitations of conventional therapeutic strategies for advanced-stage patients. Over the past few decades, nanotechnology has emerged as an important technique to obtain desired features by modifying and manipulating different objects on a molecular level and gained a lot of attention in many fields of medical applications. Studies have shown that in lung cancer, nanotechnology may be more effective and specific than traditional methods for detecting extracellular cancer biomarkers and cancer cells in vitro, as well as imaging cancer in vivo; Nanoscale drug delivery systems have developed rapidly to overcome various forms of multi-drug resistance and reduce detrimental side effects to normal tissues by targeting cancerous tissue precisely. There is no doubt that nanotechnology has the potential to enhance healthcare systems by simplifying and improving cancer diagnostics and treatment. Throughout this review, we summarize and highlight recent developments in nanotechnology applications for lung cancer in diagnosis and therapy. Moreover, the prospects and challenges in the translation of nanotechnology-based diagnostic and therapeutic methods into clinical applications are also discussed.
Collapse
Affiliation(s)
- Yujuan Duan
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China,School of Chemical Science and Engineering, Tongji University, Shanghai, China,Department of Laboratory Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Shen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai, China,*Correspondence: Yao Luo, ; Yinan Zhang,
| | - Yao Luo
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Yao Luo, ; Yinan Zhang,
| |
Collapse
|
42
|
Hu X, Ha E, Ai F, Huang X, Yan L, He S, Ruan S, Hu J. Stimulus-responsive inorganic semiconductor nanomaterials for tumor-specific theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Liu H, Wang H, Ni D, Xu Y. Lactic acid modified rare earth-based nanomaterials for enhanced radiation therapy by disturbing the glycolysis. J Nanobiotechnology 2022; 20:490. [PMCID: PMC9675198 DOI: 10.1186/s12951-022-01694-1] [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: 07/30/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Deficient deposition of X-rays and strong capacity of repairing damage DNA of cancer cells limit the effect of radiation therapy (RT). Herein, we synthesize CsLu2F7 nanoparticles with lactic acid (LA) ligands (CsLu2F7-LA) to overcome these limitations. The high-Z atoms of Lu and Cs can deposit more X-rays for generating enhanced hydroxyl radicals (·OH). Meanwhile, the LA ligand will guide CsLu2F7-LA to target monocarboxylic acid transporter (MCT) and impede the transportation of free LA, leading to decreased glycolysis and DNA damage repair. Consequently, the curative effect of RT will be enhanced and the strategy of LA accumulation induced radiosensitization is proved by in vivo and in vitro experiments, which will bring prospects for enhanced RT with nanomedicine.
Collapse
Affiliation(s)
- Hu Liu
- grid.452666.50000 0004 1762 8363Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004 Jiangsu China ,grid.459351.fDepartment of Orthopedics, Yancheng Third People’s Hospital, Yancheng, 224001 Jiangsu China
| | - Han Wang
- grid.16821.3c0000 0004 0368 8293Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Dalong Ni
- grid.16821.3c0000 0004 0368 8293Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Youjia Xu
- grid.452666.50000 0004 1762 8363Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004 Jiangsu China ,grid.459351.fDepartment of Orthopedics, Yancheng Third People’s Hospital, Yancheng, 224001 Jiangsu China
| |
Collapse
|
44
|
Wang Y, Zeng W, Liang H, Wu X, Li H, Chen T, Yang M, Wang X, Li W, Zhang F, Li Q, Ye F, Guan J, Mei L. Targeted Wolfram-Doped Polypyrrole for Photonic Hyperthermia-Synergized Radiotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50557-50568. [PMID: 36322879 DOI: 10.1021/acsami.2c15015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Single ionizing radiation at a tolerable dose is ineffectual in eliminating malignancies but readily generates harmful effects on surrounding normal tissues. Herein, we intelligently fabricated novel wolfram-doped polypyrrole (WPPy) through a simple oxidative polymerization method with WCl6 as an oxidizing catalyst, which possessed good biocompatibility, high photothermal conversion, and intensive radiosensitivity capacities to concurrently serve as a photothermal reagent and a radiosensitizer for hyperthermia-synergized radiotherapy (RT) against a malignant tumor. In comparison with traditional polypyrrole without noble metal doping, the innovative introduction of WCl6 not only successfully launched the polymerization of a pyrrole monomer but also endowed WPPy with additional radiosensitization. More importantly, after further decoration with an active targeted component (SP94 polypeptide), the obtained WPPy@SP94 significantly increased tumor internalization and accumulation in vitro and in vivo and induced obvious DNA damage as well as robust ROS generation under X-ray irradiation, which meanwhile synergized with strong photonic hyperthermia to effectively inhibit tumor growth by single drug injection. Moreover, such biocompatible WPPy@SP94 showed negligible adverse effects on normal cells and tissues. WPPy@SP94 developed in this study not only expands the category of polypyrrole chemical syntheses but also sheds light on WPPy@SP94-based radiosensitizers for cancer RT.
Collapse
Affiliation(s)
- Yin Wang
- Department of Radiation Oncology, Nanfang Hospital Southern Medical University, Guangzhou 510515, China
| | - Weiwei Zeng
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Huazhen Liang
- The First Tumor Department, Maoming People's Hospital, Maoming 525000, China
| | - Xixi Wu
- Department of Radiation Oncology, Nanfang Hospital Southern Medical University, Guangzhou 510515, China
| | - Hanyue Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Ting Chen
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Mi Yang
- Department of Radiation Oncology, Nanfang Hospital Southern Medical University, Guangzhou 510515, China
| | - Xiaoqing Wang
- Department of Radiation Oncology, Nanfang Hospital Southern Medical University, Guangzhou 510515, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Fan Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Qianqian Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Feng Ye
- Department of Radiation Oncology, Nanfang Hospital Southern Medical University, Guangzhou 510515, China
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital Southern Medical University, Guangzhou 510515, China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
45
|
He P, Yang G, Zhu D, Kong H, Corrales-Ureña YR, Colombi Ciacchi L, Wei G. Biomolecule-mimetic nanomaterials for photothermal and photodynamic therapy of cancers: Bridging nanobiotechnology and biomedicine. J Nanobiotechnology 2022; 20:483. [PMID: 36384717 PMCID: PMC9670580 DOI: 10.1186/s12951-022-01691-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022] Open
Abstract
Nanomaterial-based phototherapy has become an important research direction for cancer therapy, but it still to face some obstacles, such as the toxic side effects and low target specificity. The biomimetic synthesis of nanomaterials using biomolecules is a potential strategy to improve photothermal therapy (PTT) and photodynamic therapy (PDT) techniques due to their endowed biocompatibility, degradability, low toxicity, and specific targeting. This review presents recent advances in the biomolecule-mimetic synthesis of functional nanomaterials for PTT and PDT of cancers. First, we introduce four biomimetic synthesis methods via some case studies and discuss the advantages of each method. Then, we introduce the synthesis of nanomaterials using some biomolecules such as DNA, RNA, protein, peptide, polydopamine, and others, and discuss in detail how to regulate the structure and functions of the obtained biomimetic nanomaterials. Finally, potential applications of biomimetic nanomaterials for both PTT and PDT of cancers are demonstrated and discussed. We believe that this work is valuable for readers to understand the mechanisms of biomimetic synthesis and nanomaterial-based phototherapy techniques, and will contribute to bridging nanotechnology and biomedicine to realize novel highly effective cancer therapies.
Collapse
Affiliation(s)
- Peng He
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Guozheng Yang
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Danzhu Zhu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Hao Kong
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Yendry Regina Corrales-Ureña
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, University of Bremen, 28359, Bremen, Germany.
| | - Lucio Colombi Ciacchi
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, University of Bremen, 28359, Bremen, Germany
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
46
|
Chai R, Yu L, Dong C, Yin Y, Wang S, Chen Y, Zhang Q. Oxygen-evolving photosynthetic cyanobacteria for 2D bismuthene radiosensitizer-enhanced cancer radiotherapy. Bioact Mater 2022; 17:276-288. [PMID: 35386463 PMCID: PMC8965086 DOI: 10.1016/j.bioactmat.2022.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
The local hypoxic tumor environment substantially hampers the therapeutic efficiency of radiotherapy, which typically requires the large X-ray doses for tumor treatment but induces the serious side effects. Herein, a biomimetic radiosensitized platform based on a natural in-situ oxygen-evolving photosynthetic cyanobacteria combined with two-dimensional (2D) bismuthene with high atomic-number (Z) components, is designed and engineered to effectively modulate the radiotherapy-resistant hypoxic tumor environment and achieve sufficient radiation energy deposition into tumor. Upon the exogenous sequential irradiation of 660 nm laser and X-ray beam, continuous photosynthetic oxygen evolution by the cyanobacteria and considerable generation of reactive oxygen species by the 2D bismuthene radiosensitizer substantially augmented the therapeutic efficacy of radiotherapy and suppressed the in vivo tumor growth, as demonstrated on both LLC-lung tumor xenograft-bearing C57/B6 mice model and 4T1-breast tumor xenograft-bearing Balb/c mice model, further demonstrating the photosynthetic hypoxia-alleviation capability and radiosensitization performance of the engineered biomimetic radiosensitized platform. This work exemplifies a distinct paradigm on the construction of microorganism-enabled tumor-microenvironment modulation and nanoradiosensitizer-augmented radiotherapy for efficient tumor treatment. The unique microorganism-based and 2D bismuthene-mediated biomimetic radiosensitization platform has been engineered for enhanced tumor nanotherapy. The photosynthetic production of oxygen has been utilized via natural microorganism cyanobacteria to effectively modulate the radiotherapy-resistant hypoxic tumor environment with high biocompatibility. The biomimetic dual-radiosensitized platform has achieved sufficient radiation energy deposition for inducing overproduction of reactive oxygen species to augment the RT efficacy.
Collapse
Affiliation(s)
- Rong Chai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Luodan Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Caihong Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, 200032, PR China
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| |
Collapse
|
47
|
Platinum-based nanocomposites loaded with MTH1 inhibitor amplify oxidative damage for cancer therapy. Colloids Surf B Biointerfaces 2022; 218:112715. [PMID: 35932557 DOI: 10.1016/j.colsurfb.2022.112715] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/10/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022]
Abstract
Photodynamic therapy (PDT) is a promising therapeutic strategy for tumor ablation by generating highly toxic reactive oxygen species (ROS) to damage DNA and other biomacromolecules. However, the local hypoxic microenvironment of the tumor and the presence of ROS-defensing system, such as the mobilization of mutt homolog 1 (MTH1) to sanitize ROS-oxidized nucleotide pool, severely limit the efficiency of PDT. Therefore, a novel tumor ablation strategy was developed that not only focused on the enhancement of ROS generation but also weakened the ROS-defensing system by inhibiting MTH1 enzyme activity. In our work, a simple one-step reduction approach was applied to enable platinum nanoparticles (Pt NPs) with catalase activity to grow in situ in the nanochannels of mesoporous silica nanoparticles (MSNs). After physical encapsulation of photosensitizer chlorin e6 (Ce6) and MTH1 inhibitor TH588, the drug loading nanoplatform was modified with an arginine-glycine-aspartic acid (RGD) functionalized liposome shell, resulting in the fabrication of amplified oxidative damage nanoplatform MSN-Pt@Ce6/TH588 @Liposome-RGD (MPCT@Li-R). The prepared MPCT@Li-R NPs could continuously catalyze the decomposition of hydrogen peroxide (H2O2) into oxygen (O2) in tumor, thus promoting the generation of singlet oxygen during PDT process for improved oxidative damage of bases. Simultaneously, acid responsive released TH588 hindered MTH1-mediated scavenging of oxidative bases, further aggravating DNA oxidative damage. Consequently, this cascade therapy strategy exhibited excellent tumor suppression efficiency both in vitro and in vivo.
Collapse
|
48
|
Yuan CS, Teng Z, Yang S, He Z, Meng LY, Chen XG, Liu Y. Reshaping hypoxia and silencing CD73 via biomimetic gelatin nanotherapeutics to boost immunotherapy. J Control Release 2022; 351:255-271. [PMID: 36165836 DOI: 10.1016/j.jconrel.2022.09.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 10/31/2022]
Abstract
The ubiquitous hypoxic microenvironment at the tumor site helps to regulate hypoxic inducible factor (HIF-1α), up-regulate downstream CD73-adenosine (CD73-ADO) pathways, and further result in effector T cell function exhaustion, which is regarded as a crucial adverse factor in the poor clinical efficacy of immune checkpoint blockade therapy (ICB). How to reshape hypoxic microenvironment and silence CD73 remains a huge challenge to improve ICB therapeutic outcomes. In this study, cancer cell membrane-camouflaged gelatin nanoparticles (CSG@B16F10) were designed to co-deliver oxygen-generating agent catalase (CAT) and CD73siRNA, thus enhancing tumor oxygenation and alleviating CD73-ADO pathway-mediated T cell immunosuppression. The fabricated biomimetic nanoparticles could efficiently achieve immune evading and homologous targeting by virtue of the retention of cancer cell membrane protein. Matrix metalloproteinases (MMP)-responsive gelatin nanoparticles were gradually disintegrated to accelerate the release of payloads. Rapidly released CAT was found to relieve tumor hypoxia by generating endogenous oxygen, while CD73siRNA effectively silenced target gene, synergically inhibiting CD73 protein expression and facilitating T-cell-specific immunity. Upon introduction of CSG@B16F10 in melanoma-bearing mice, PD-L1 checkpoint blockade achieved optimal tumor suppression (∼83%). The enhanced immune efficacy was mainly manifested by enhanced cytotoxic T cell (CTL), reduced regulatory T cells (Tregs), and increased anti-tumor cytokine secretion. This work presents a new paradigm for the ideal design of biomimetic nanoplatforms and the synergistic treatment of hypoxia alleviation and CD73 silence, greatly promising for enhancing clinical immune potency of PD-1/PD-L1 immune checkpoint blockade.
Collapse
Affiliation(s)
- Cong-Shan Yuan
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Zhuang Teng
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Shuang Yang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Zheng He
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Ling-Yang Meng
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Xi-Guang Chen
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, PR China
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China.
| |
Collapse
|
49
|
Yao X, Lu S, Feng C, Suo R, Li H, Zhang Y, Chen Q, Lu J, Wu B, Guo J. Tumor oxygenation nanoliposome synergistic hypoxia-inducible-factor-1 inhibitor enhanced Iodine-125 seed brachytherapy for esophageal cancer. Biomaterials 2022; 289:121801. [PMID: 36137416 DOI: 10.1016/j.biomaterials.2022.121801] [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: 05/12/2022] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/02/2022]
Abstract
Iodine-125 (125I) brachytherapy has become one of the most effective palliative treatment options for advanced esophageal cancer. However, resistance toward 125I brachytherapy caused by pre-existing tumor hypoxia and hypoxia-inducible factor 1 (HIF-1) signaling pathway activation represents a significant limitation in esophageal cancer treatment. To circumvent these problems, herein, we proposed an innovative strategy to alleviate radioresistance of brachytherapy by co-encapsulating catalase (CAT) and HIF-1 inhibitor-acriflavine (ACF) into the hydrophilic cavities of liposome, termed as "ACF-CAT@Lipo". Under overexpressed H2O2 stimulation in the tumor region, the fabricated ACF-CAT@Lipo can generate an amount of O2 and alleviate tumor hypoxia in vitro and in vivo. Furthermore, cooperating with ACF, the expression of hypoxia-related protein (e.g. HIF-1α, VEGF, MMP-2) are obviously decreased. Importantly, the copious oxygenation and the significant inhibition expression of HIF-1α can further improve the radiosensitivity of 125I brachytherapy and finally realize the eradication of esophageal cancer in vivo. The oxygen enrichment and HIF-1 inhibition function of ACF-CAT@Lipo provides a new strategy to overcome the brachytherapy resistance of esophageal cancer therapy.
Collapse
Affiliation(s)
- Xijuan Yao
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Shuting Lu
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Cheng Feng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Ruiyang Suo
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Hang Li
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Yi Zhang
- School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Qi Chen
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Jian Lu
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Jinhe Guo
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
50
|
Hu B, Xiao X, Chen P, Qian J, Yuan G, Ye Y, Zeng L, Zhong S, Wang X, Qin X, Yang Y, Pan Y, Zhang Y. Enhancing anti-tumor effect of ultrasensitive bimetallic RuCu nanoparticles as radiosensitizers with dual enzyme-like activities. Biomaterials 2022; 290:121811. [DOI: 10.1016/j.biomaterials.2022.121811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/02/2022]
|