1
|
Chen W, Feng H, Mo Y, Pan Z, Ji S, Liang H, Shen XC, Jiang BP. Hyaluronic acid-functionalized ruthenium photothermal nanoenzyme for enhancing osteosarcoma chemotherapy: Cascade targeting and bidirectional modulation of drug resistance. Carbohydr Polym 2025; 349:122945. [PMID: 39643406 DOI: 10.1016/j.carbpol.2024.122945] [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: 08/17/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 12/09/2024]
Abstract
Insufficient drug delivery efficiency in vivo and robust drug resistance are two major factors to induce suboptimal efficacy in chemotherapy of osteosarcoma (OS). To address these challenges, we developed polysaccharide hyaluronic acid (HA)-functionalized ruthenium nanoaggregates (Ru NAs) to enhance the chemotherapy of doxorubicin (DOX) for OS. These NAs, comprising Ru nanoparticles (NPs) and alendronate-modified HA (HA-ALN), effectively load DOX, resulting in DOX@Ru-HA-ALN NAs. The combination of HA and ALN in NAs ensures outstanding cascade targeting towards tumor-invaded bone tissues and CD44-overexpressing tumor cells, maximizing therapeutic efficacy while minimizing off-target effects. Concurrently, the Ru NPs in NAs function as "smart" photoenzymatic agent to not only in situ relieve hypoxia of OS via the catalysis of overexpressed H2O2 to produce O2, but also generate mild photothermal effect under 808-nm laser irradiation. They can bidirectionally overcome drug resistance of DOX via downregulation of resistance-related factors including multi-drug resistant associate protein, P-glycoprotein, heat shock factor 1, etc. The integration of cascade targeting with bidirectional modulation of drug resistance positions Ru-HA-ALN NAs to substantially enhance DOX chemotherapy for OS. Therefore, the present work highlights the potential of polysaccharide-functionalized nanomaterials in advancing tumor chemotherapy by addressing challenges of both delivery efficiency and drug resistance.
Collapse
Affiliation(s)
- Weifeng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Hao Feng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Yinyin Mo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Zhihui Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Shichen Ji
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China.
| | - Bang-Ping Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
2
|
Du Q, Yuen HY, Pan J, Sun C, Wu D, Liu J, Wu G, Zhao X, Wang S. Metronidazole-modified Au@BSA nanocomposites for dual sensitization of radiotherapy in solid tumors. J Mater Chem B 2024; 12:9686-9694. [PMID: 39193619 DOI: 10.1039/d4tb00910j] [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: 08/29/2024]
Abstract
The hypoxic microenvironment of solid tumors can lead to reduced therapeutic DNA damage to the tumor cells, thus diminishing tumor sensitivity to radiotherapy. Although hypoxic radiosensitizers can improve radiotherapy efficacy by enhancing the role of oxygen, their effects are limited by the uneven distribution of oxygen within solid tumor tissues. In this study, a novel radiosensitizer via leveraging gold complexes and metronidazole (MN) was synthesized to improve radiotherapeutic efficacy. The gold atoms incorporated in the radiosensitizer enabled efficient deposition of high-energy radiation; the hydrophobic metronidazole was reduced to hydrophilic aminoimidazole under hypoxia conditions and further promoted radiotherapy sensitization. The results of CCK-8 assays, Live/Dead assays, γ-H2AX immunofluorescence indicated that metronidazole-modified Au@BSA nanocomposites (NCs) exhibited excellent antitumor effects. The in vivo antitumor tests further showed an inhibition rate of 100%. These results demonstrated that the NCs successfully enhanced radiotherapy efficacy by the dual sensitization strategy. Overall, we believe this multimodal radiosensitizing nanocomplex can significantly inhibit tumor growth and metastasis, with their hypoxia-oriented characteristics ensuring a higher efficacy and safety.
Collapse
Affiliation(s)
- Qijun Du
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Ho-Yin Yuen
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| | - Jingke Pan
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Chenwei Sun
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Di Wu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Jie Liu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Guohua Wu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| | - Shuqi Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu 641400, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
| |
Collapse
|
3
|
Gong Z, Fu Y, Gao Y, Jiao F, Su Q, Sang X, Chen B, Deng X, Liu X. "Abraxane-Like" Radiosensitizer for In Situ Oral Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309569. [PMID: 38973195 PMCID: PMC11425904 DOI: 10.1002/advs.202309569] [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: 12/08/2023] [Revised: 06/06/2024] [Indexed: 07/09/2024]
Abstract
Radiotherapy plays a vital role in cancer therapy. However, the hypoxic microenvironment of tumors greatly limits the effectiveness, thus it is crucial to develop a simple, efficient, and safe radiosensitizer to reverse hypoxia and ameliorate the efficacy of radiotherapy. Inspired by the structure of canonical nanodrug Abraxane, herein, a native HSA-modified CaO2 nanoparticle system (CaO2-HSA) prepared by biomineralization-induced self-assembly is developed. CaO2-HSA will accumulate in tumor tissue and decompose to produce oxygen, altering the hypoxic condition inside the tumor. Simultaneously, ROS and calcium ions will lead to calcium overload and further trigger immunogenic cell death. Notably, its sensitizing enhancement ratio (SER = 3.47) is much higher than that of sodium glycididazole used in the clinic. Furthermore, in animal models of in situ oral cancer, CaO2-HSA can effectively inhibit tumor growth. With its high efficacy, facile preparation, and heavy-metal free biosafety, the CaO2-HSA-based radiosensitizer holds enormous potential for oral cancer therapy.
Collapse
Affiliation(s)
- Zijian Gong
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yixuan Fu
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yuan Gao
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Fei Jiao
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Qinzhi Su
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiao Sang
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Binglin Chen
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Biomedical Engineering DepartmentPeking UniversityBeijing100191P. R. China
| | - Xinyu Liu
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
4
|
Zhou J, Yang R, Chen Y, Chen D. Efficacy tumor therapeutic applications of stimuli-responsive block copolymer-based nano-assemblies. Heliyon 2024; 10:e28166. [PMID: 38571609 PMCID: PMC10987934 DOI: 10.1016/j.heliyon.2024.e28166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024] Open
Abstract
Block copolymers are composed of two or more blocks or segments with different chemical properties via various chemical bonds, which can assemble into nanoparticles with a "core-shell" structure. Due to the benefits of simple functionalization, superior drug-loading capacity, and good biocompatibility, various nano-assemblies based on block copolymers have become widely applied in the treatment of cancers in recent years. These nano-assemblies serve as carriers for anti-tumor bioactive, enhancing drug stability and prolonging their circulation time in vivo, which can reduce the toxic side effects of drugs and improve the therapeutic effect. However, the complex and heterogeneous tumor microenvironment poses challenges to the therapeutic efficacy of these nano-assemblies, having the result in the occurrence of drug resistance and the recurrence of tumors. Consequently, a diverse array of stimuli-responsive nano-assemblies has been devised in order to surmount these obstacles. This article provides a comprehensive overview of the utilization of stimuli-responsive nano-assemblies derived from block copolymers in the context of tumor treatment. The review summarizes block polymers responsive to internal stimuli (like ROS, redox, pH, and enzymes) and external stimuli (like light, and temperature), and discusses current challenges and prospects in this field, aiming to provide novel insights for clinical applications.
Collapse
Affiliation(s)
- Jie Zhou
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Jiangsu, 214002, China
| | - Rui Yang
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Jiangsu, 214002, China
| | - Yu Chen
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Jiangsu, 214002, China
| | - Daozhen Chen
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Jiangsu, 214002, China
- Department of Laboratory, Haidong Second People's Hospital, Haidong, 810699, China
| |
Collapse
|
5
|
Ding Y, Yu W, Shen R, Zheng X, Zheng H, Yao Y, Zhang Y, Du C, Yi H. Hypoxia-Responsive Tetrameric Supramolecular Polypeptide Nanoprodrugs for Combination Therapy. Adv Healthc Mater 2024; 13:e2303308. [PMID: 37924332 DOI: 10.1002/adhm.202303308] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Despite the intense progress of photodynamic and chemotherapy, however, they cannot prevent solid tumor invasion, metastasis, and relapse, along with inferior efficacy and severe side effects. The hypoxia-responsive nanoprodrugs integrating photodynamic functions are highly sought to address the above-mentioned problems and overcome the tumor hypoxia-reduced efficacy. Herein, a hypoxia-responsive tetrameric supramolecular polypeptide nanoprodrug (SPN-TAPP-PCB4) is constructed from the self-assembly of tetrameric porphyrin-central poly(l-lysine-azobenzene-chlorambucil) (TAPP-(PLL-Azo-CB)4) and an anionic water-soluble [2]biphenyl-extended-pillar[6]arene (AWBpP6) via the synergy of hydrophobic, π-π stacking, and host-guest interactions. Upon laser irradiation, the central TAPP can convert oxygen to generate single oxygen (1 O2 ) to kill tumor cells. Furthermore, under the acidic and PDT-aggravated hypoxia tumor cell microenvironment, SPN-TAPP-PCB4 is rapidly disassembled, and then efficiently releases activated CB through the hypoxic-responsive cleavage of azobenzene linkages. Both in vitro and in vivo biological studies showcase synergistic cancer-killing actions between photodynamic therapy (PDT) and chemotherapy (CT) with negligible toxicity. Consequently, this supramolecular polypeptide nanoprodrug offers an effective strategy to design a hypoxia-responsive nanoprodrug for a potential combo PDT-CT transition.
Collapse
Affiliation(s)
- Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Wei Yu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Rongkai Shen
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, 20, Chazhong Rd., Fuzhou, Fujian, 350005, China
| | - Xiangqin Zheng
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| | - Hui Zheng
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Yuehua Zhang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Chang Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Huan Yi
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| |
Collapse
|
6
|
Li H, Huang H, Tan H, Jia Q, Song W, Zhang Q, Zhou B, Bai J. Key processes in tumor metastasis and therapeutic strategies with nanocarriers: a review. Mol Biol Rep 2024; 51:197. [PMID: 38270746 DOI: 10.1007/s11033-023-08910-7] [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: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 01/26/2024]
Abstract
Cancer metastasis is the leading cause of cancer-related death. Metastasis occurs at all stages of tumor development, with unexplored changes occurring at the primary site and distant colonization sites. The growing understanding of the metastatic process of tumor cells has contributed to the emergence of better treatment options and strategies. This review summarizes a range of features related to tumor cell metastasis and nanobased drug delivery systems for inhibiting tumor metastasis. The mechanisms of tumor metastasis in the ideal order of metastatic progression were summarized. We focus on the prominent role of nanocarriers in the treatment of tumor metastasis, summarizing the latest applications of nanocarriers in combination with drugs to target important components and processes of tumor metastasis and providing ideas for more effective nanodrug delivery systems.
Collapse
Affiliation(s)
- Hongjie Li
- School of Clinical Medicine, Weifang Medical University, 261053, Weifang, China
| | - Haiqin Huang
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, 250012, Jinan, China
| | - Qitao Jia
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China
| | - Weina Song
- Department of Pediatric Respiratory and Critical Care, Qilu Hospital of Shandong University Dezhou Hospital, 253000, Dezhou, China
| | - Qingdong Zhang
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China.
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China.
| |
Collapse
|
7
|
Liu J, Liu C, Tang J, Chen Q, Yu Y, Dong Y, Hao J, Wu W. Synergistic cerium oxide nanozymes: targeting DNA damage and alleviating tumor hypoxia for improved NSCLC radiotherapy efficiency. J Nanobiotechnology 2024; 22:25. [PMID: 38195456 PMCID: PMC10777519 DOI: 10.1186/s12951-023-02196-4] [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: 06/05/2023] [Accepted: 11/05/2023] [Indexed: 01/11/2024] Open
Abstract
Radiotherapy (RT) is one of the important treatment modalities for non-small cell lung cancer (NSCLC). However, the maximum radiation dose that NSCLC patient can receive varies little. Therefore, the exploitation of novel RT sensitization approaches is a critical need for the clinical treatment. RT resistance in NSCLC is linked to tumor microenvironment (TME) hypoxia, cell cycle arrest and associated genetic alterations. Here, we designed a novel method for targeted delivery of quercetin (QT) and CeO2 to enhance RT sensitivity. We loaded QT into CeO2@ZIF-8-HA nanoparticles to prevent its degradation in the circulatory system and successfully delivered QT and CeO2 targeted to NSCLC tumors. Under the protection and targeted delivery of Zeolitic Imidazolate Framework-8 (ZIF-8), the nanocomplexes exhibited excellent catalytic mimetic activity in decomposing H2O2 into O2, thus significantly reversing the hypoxia of TME, while the radiosensitizer QT caused DNA damage directly after RT. In a subcutaneous tumor model, CeO2@ZIF-8-HA overcame radiation resistance and enhanced therapeutic efficacy. This multiple sensitization strategy combining delivery of QT and CeO2@ZIF-8-HA nanozymes opens a promising approach for RT of NSCLC.
Collapse
Affiliation(s)
- Jie Liu
- Department of Cardiothoracic Surgery, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China
| | - Chengxiang Liu
- Department of Oncology, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China
| | - Jinghua Tang
- Department of Cardiothoracic Surgery, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China
| | - Qiao Chen
- Department of Cardiothoracic Surgery, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China
| | - Yan Yu
- Department of Cardiothoracic Surgery, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China
| | - Yan Dong
- Department of Oncology, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China.
| | - Jie Hao
- Department of Oncology, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China.
| | - Wei Wu
- Department of Cardiothoracic Surgery, Southwest Hospital, Army Medical University, 30 Gaotanyan Main St, Chongqing, 400038, China.
| |
Collapse
|
8
|
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
|
9
|
Bai W, Hu Y, Zhao J, Shi L, Ge C, Zhu Z, Rao J. Precision Dosing of Antibiotics and Potentiators by Hypoxia-Responsive Nanoparticles for Overcoming Antibiotic Resistance in Gram-Negative Bacteria. ACS Macro Lett 2023; 12:1193-1200. [PMID: 37590266 DOI: 10.1021/acsmacrolett.3c00365] [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: 08/19/2023]
Abstract
The stalling development of antibiotics, especially against intrinsically resistant Gram-negative pathogens associated with outer membranes, leads to an emerging antibiotic crisis across the globe. To breathe life into existing drugs, we herein report a hypoxia-responsive nanoparticle (NP) that encapsulates a hydrophobic antibiotic, rifampicin, and a cationic potentiator, polysulfonium. The simultaneous release of antibiotics and potentiators can be promoted and inhibited in response to the severity of bacterial-induced hypoxia, leading to antimicrobial dosing in a precision manner. Under the synergism of polysulfoniums with membrane-disruption capability, the NPs can intensively decrease the antibiotic dose by up to 66-95% in eliminating planktonic Gram-negative P. aeruginosa bacteria and achieve an 8-log reduction of bacteria in mature biofilms at rifampicin MIC. The NP formulation demonstrates that precision dosing of antibiotics and potentiators regulated by hypoxia provides a promising strategy to maximize efficacy and minimize toxicity in treating Gram-negative bacterial infection.
Collapse
Affiliation(s)
- Weiguang Bai
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
- College of Chemistry, Liaoning University, Shenyang 110036, PR China
| | - Yongjin Hu
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Jinghua Zhao
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Liuqi Shi
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Chunhua Ge
- College of Chemistry, Liaoning University, Shenyang 110036, PR China
| | - Zhiyuan Zhu
- Taizhou Research Institute, Southern University of Science and Technology, Taizhou, Zhejiang 318001, PR China
| | - Jingyi Rao
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| |
Collapse
|
10
|
Li D, Ren T, Ge Y, Wang X, Sun G, Zhang N, Zhao L, Zhong R. A multi-functional hypoxia/esterase dual stimulus responsive and hyaluronic acid-based nanomicelle for targeting delivery of chloroethylnitrosouea. J Nanobiotechnology 2023; 21:291. [PMID: 37612719 PMCID: PMC10464291 DOI: 10.1186/s12951-023-02062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023] Open
Abstract
Carmustine (BCNU), a vital type of chloroethylnitrosourea (CENU), inhibits tumor cells growth by inducing DNA damage at O6 position of guanine and eventually forming dG-dC interstrand cross-links (ICLs). However, the clinical application of BCNU is hindered to some extent by the absence of tumor selectivity, poor stability and O6-alkylguanine-DNA alkyltransferase (AGT) mediated drug resistance. In recent years, tumor microenvironment has been widely utilized for advanced drug delivery. In the light of the features of tumor microenvironment, we constructed a multifunctional hypoxia/esterase-degradable nanomicelle with AGT inhibitory activity named HACB NPs for tumor-targeting BCNU delivery and tumor sensitization. HACB NPs was self-assembled from hyaluronic acid azobenzene AGT inhibitor conjugates, in which O6-BG analog acted as an AGT inhibitor, azobenzene acted as a hypoxia-responsive linker and carboxylate ester bond acted as both an esterase-sensitive switch and a connector with hyaluronic acid (HA). The obtained HACB NPs possessed good stability, favorable biosafety and hypoxia/esterase-responsive drug-releasing ability. BCNU-loaded HACB/BCNU NPs exhibited superior cytotoxicity and apoptosis-inducing ability toward the human uterine cervix carcinoma HeLa cells compared with traditional combined medication of BCNU plus O6-BG. In vivo studies further demonstrated that after a selective accumulation in the tumor site, the micelles could respond to hypoxic tumor tissue for rapid drug release to an effective therapeutic dosage. Thus, this multifunctional stimulus-responsive nanocarrier could be a new promising strategy to enhance the anticancer efficacy and reduce the side effects of BCNU and other CENUs.
Collapse
Affiliation(s)
- Duo Li
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| | - Yunxuan Ge
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| | - Xiaoli Wang
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| | - Na Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China.
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, China
| |
Collapse
|
11
|
Geng H, Chen K, Cao L, Liu L, Huang Y, Liu J. Hypoxia-Responsive Aggregation of Gold Nanoparticles for Near-Infrared-II Photoacoustic Imaging-Guided Enhanced Radiotherapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:4037-4048. [PMID: 36907993 DOI: 10.1021/acs.langmuir.2c03399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
By directly harming cancer cells, radiotherapy (RT) is a crucial therapeutic approach for the treatment of cancers. However, the efficacy of RT is reduced by the limited accumulation and short retention time of the radiosensitizer in the tumor. Herein, we developed hypoxia-triggered in situ aggregation of nanogapped gold nanospheres (AuNNP@PAA/NIC NPs) within the tumor, resulting in second near-infrared window (NIR-II) photoacoustic (PA) imaging and enhanced radiosensitization. AuNNP@PAA/NIC NPs demonstrated increased accumulation and retention in hypoxic tumors, mainly due to the hypoxia-triggered aggregation. After aggregation of AuNNP@PAA/NIC NPs, the absorption of the system extended from visible light to NIR-II light owing to the plasmon coupling effects between adjacent nanoparticles. Compared to the normoxic tumor, the PA intensity at 1200 nm in the hypoxic tumor increased from 0.42 to 1.88 at 24 h postintravenous injection of AuNNP@PAA/NIC NPs, leading to an increase of 4.5 times. This indicated that the hypoxic microenvironment in the tumor successfully triggered the in situ aggregation of AuNNP@PAA/NIC NPs. The in vivo radiotherapeutic effect demonstrated that this hypoxia-triggered in situ aggregation of radiosensitizers significantly enhanced radiosensitization and thus resulted in superior cancer radiotherapeutic outcomes.
Collapse
Affiliation(s)
- Huafeng Geng
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun 130033, China
| | - Ke Chen
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun 130033, China
| | - Lu Cao
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun 130033, China
| | - Luntao Liu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Yue Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Junbao Liu
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun 130033, China
| |
Collapse
|
12
|
Zhou J, Xu D, Tian G, He Q, Zhang X, Liao J, Mei L, Chen L, Gao L, Zhao L, Yang G, Yin W, Nie G, Zhao Y. Coordination-Driven Self-Assembly Strategy-Activated Cu Single-Atom Nanozymes for Catalytic Tumor-Specific Therapy. J Am Chem Soc 2023; 145:4279-4293. [PMID: 36744911 DOI: 10.1021/jacs.2c13597] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
How to optimize the enzyme-like catalytic activity of nanozymes to improve their applicability has become a great challenge. Herein, we present an l-cysteine (l-Cys) coordination-driven self-assembly strategy to activate polyvinylpyrrolidone (PVP)-modified Cu single-atom nanozymes MoOx-Cu-Cys (denoted as MCCP SAzymes) aiming at catalytic tumor-specific therapy. The Cu single atom content of MCCP can be rationally modulated to 10.10 wt %, which activates the catalase (CAT)-like activity of MoOx nanoparticles to catalyze the decomposition of H2O2 in acidic microenvironments to increase O2 production. Excitingly, the maximized CAT-like catalytic efficiency of MCCP is 138-fold higher than that of typical MnO2 nanozymes and exhibits 14.3-fold higher affinity than natural catalase, as demonstrated by steady-state kinetics. We verify that the well-defined l-Cys-Cu···O active sites optimize CAT-like activity to match the active sites of natural catalase through an l-Cys bridge-accelerated electron transfer from Cys-Cu to MoOx disclosed by density functional theory calculations. Simultaneously, the high loading Cu single atoms in MCCP also enable generation of •OH via a Fenton-like reaction. Moreover, under X-ray irradiation, MCCP converts O2 to 1O2 for cascading radiodynamic therapy, thereby facilitating the multiple reactive oxygen species (ROS) for radiosensitization to achieve substantial antitumor.
Collapse
Affiliation(s)
- Jie Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, Jiangxi Province Key Laboratory of Synthetic Chemistry, East China University of Technology, Nanchang, Jiangxi 330013, China
| | - Deting Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Gan Tian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing 400038, China
| | - Qian He
- College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi 530004, China
| | - Xiao Zhang
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing 400038, China
| | - Jing Liao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Linqiang Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Lizeng Gao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Lina Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Guoping Yang
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, Jiangxi Province Key Laboratory of Synthetic Chemistry, East China University of Technology, Nanchang, Jiangxi 330013, China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Wang Y, Gao D, Jin L, Ren X, Ouyang Y, Zhou Y, He X, Jia L, Tian Z, Wu D, Yang Z. NADPH Selective Depletion Nanomedicine-Mediated Radio-Immunometabolism Regulation for Strengthening Anti-PDL1 Therapy against TNBC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203788. [PMID: 36403210 PMCID: PMC9875612 DOI: 10.1002/advs.202203788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/08/2022] [Indexed: 05/25/2023]
Abstract
Anti-PD(L)1 immunotherapy recently arises as an effective treatment against triple-negative breast cancer (TNBC) but is only applicable to a small portion of TNBC patients due to the low PD-L1 expression and the immunosuppressive tumor microenvironment (TME). To address these challenges, a multifunctional "drug-like" copolymer that possesses the auto-changeable upper critical solution temperature and the capacity of scavenging reduced nicotinamide adenine dinucleotide phosphate (NADPH) inside tumor cells is synthesized and employed to develop a hypoxia-targeted and BMS202 (small molecule antagonist of PD-1/PD-L1 interactions)-loaded nanomedicine (BMS202@HZP NPs), combining the anti-PD-L1 therapy and the low-dose radiotherapy (LDRT) against TNBC. In addition to the controlled release of BMS202 in the hypoxic TNBC, BMS202@HZP NPs benefit the LDRT by upregulating the pentose phosphate pathway (PPP, the primary cellular source for NADPH) of TME whereas scavenging the NADPH inside tumor cells. As a result, the BMS202@HZP NPs-mediated LDRT upregulate the PD-L1 expression of tumor to promote anti-PD-L1 therapy response while reprogramming the immunometabolism of TME to alleviate its immunosuppression. This innovative nanomedicine-mediated radio-immunometabolism regulation provides a promising strategy to reinforce the anti-PD-L1 therapy against TNBC.
Collapse
Affiliation(s)
- Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Lin Jin
- International Joint Research Laboratory for Biomedical Nanomaterials of HenanZhoukou Normal UniversityZhoukou466001P. R. China
| | - Xuechun Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Yanan Ouyang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Ying Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Xinyu He
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Dingcai Wu
- PCFM LabSchool of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
- Center of Accurate DiagnosisTreatment and Transformation of Bone and Joint DiseasesThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518000P. R. China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| |
Collapse
|
14
|
Qian L, Li Q, Ding Z, Luo K, Su J, Chen J, Zhu G, Gan Z, Yu Q. Prodrug Nanosensitizer Overcomes the Radiation Resistance of Hypoxic Tumor. ACS APPLIED MATERIALS & INTERFACES 2022; 14:56454-56470. [PMID: 36525559 DOI: 10.1021/acsami.2c14628] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Clinical radiation therapy (RT) is often hindered by the low radiation energy absorption coefficient and the hypoxic features of tumor tissues. Among the tremendous efforts devoted to overcoming the barriers to efficient RT, the application of hypoxic radiosensitizers and cell-cycle-specific chemotherapeutics has shown great potential. However, their effectiveness is often compromised by their limited bioavailability, especially in the hypoxic region, which plays a major role in radioresistance. Herein, to simultaneously improve the delivery efficacy of both hypoxic radiosensitizer and cell-cycle-specific drug, a gambogic acid (GA) metronidazole (MN) prodrug (GM) was designed and synthesized based on GA, a naturally occurring chemotherapeutic and multiple pathway inhibitor, and MN, a typical hypoxic radiosensitizer. In combination with MN-containing block copolymers, the prodrug nanosensitizer (NS) of GM was obtained. Owing to the bioreduction of MN, the as-designed prodrug could be efficiently delivered to hypoxic cells and act on mitochondria to cause the accumulation of reactive oxygen species. The strong G2/M phase arrest caused by the prodrug NS could further sensitize treated cells to external radiation under hypoxic conditions by increasing DNA damage and delaying DNA repair. After coadministration of the NS with a well-established tissue-penetrating peptide, efficient tumor accumulation, deep tumor penetration, and highly potent chemoradiotherapy could be achieved.
Collapse
Affiliation(s)
- Lili Qian
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| | - Qian Li
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| | - Zhenshan Ding
- Department of Urology, China-Japan Friendship Hospital, Beijing100029, China
| | - Kejun Luo
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| | - Jiamin Su
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| | - Jiawei Chen
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| | - Guangying Zhu
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing100029, China
| | - Zhihua Gan
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| | - Qingsong Yu
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), Beijing University of Chemical Technology, Beijing100029, China
| |
Collapse
|
15
|
Bin Y, Meng Z, Huang LL, Hu XY, Song JM, Xie YT, Kang M, Wang RS. Prognostic value of the cervical lymph node necrosis ratio in nasopharyngeal carcinoma. Radiother Oncol 2022; 177:185-190. [PMID: 36375560 DOI: 10.1016/j.radonc.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/01/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
PURPOSE Whether cervical lymph node necrosis (CNN) is an independent adverse prognostic factor in nasopharyngeal carcinoma (NPC) has not been determined. In this study, the CNN ratio was graded quantitatively to explore the prognostic value in NPC. PARTICIPANTS AND METHODS We retrospectively reviewed a total of 648 pathologically confirmed as NPC. We outlined metastatic lymph nodes and necrotic area of lymph nodes slice by slice on the magneticresonanceimages (MRI) cross section, and calculated the corresponding CNN ratio. RESULTS The median CNN ratio (17.37 %) was taken as the cut-off point, 256 (39.51 %) patients were divided into CNN1 group (<17.37 %, n = 128) and CNN2 group (≥17.37 %, n = 128), 392 (60.49 %) patients without lymph nodes necrosis were CNN0. Among the CNN0, CNN1 and CNN2 groups, five-year overall survival (OS) was 82.4 %, 76.6 % and 71.1 %, locoregional recurrence-free survival (LRRFS) was 91.3 %, 91.1 % and 90.5 %, distant metastasis-free survival (DMFS) was 83.7 %, 78.5 % and 68.7 %, progression-free survival (PFS) was 78.3 %, 71.7 % and 61.6 % respectively. By multivariate analysis, CNN was an independent prognostic factor for OS (P = 0.003), DMFS (P = 0.019) and PFS (P = 0.007). More than 3 cycles of chemotherapy significantly increased OS (P = 0.024) and DMFS (P = 0.015) in the CNN1 group. CONCLUSIONS This study indicated that CNN is one of the factors with the negative prognosis of NPC. The CNN ratio might be used as one of the reference factors in the formulation of individualized treatment plan.
Collapse
Affiliation(s)
- Ying Bin
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Zhen Meng
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China; Department of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530021, Guangxi, China
| | - Lu-Lu Huang
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Xue-Ying Hu
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
| | - Jun-Mei Song
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Yi-Ting Xie
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China; Department of Radiation Oncology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Min Kang
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
| | - Ren-Sheng Wang
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China.
| |
Collapse
|
16
|
Yu Q, Tu L, Zhu T, Zhu H, Liu S, Sun Y, Zhao Q. Hypoxia-Activatable Nanovesicles as In Situ Bombers for Combined Hydrogen-Sulfide-Mediated Respiration Inhibition and Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50637-50648. [PMID: 36326806 DOI: 10.1021/acsami.2c15844] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Photothermal therapy (PTT) has emerged as a promising alternative or supplement to cancer treatments. While PTT induces the ablation of solid tumors, its efficiency is hampered by self-recovery within impaired cancer cells through glycolysis and respiration metabolism. Based on this, the introduction of hydrogen sulfide (H2S)-mediated respiration inhibition is a good choice to make up for the PTT limitation. Herein, nanovesicles (NP1) are integrated by a hypoxia-responsive conjugated polymer (P1), polymetric H2S donor (P2), and near-infrared (NIR) light-harvesting aza-BODIPY dye (B1) for the delivery of H2S and synergistic H2S gas therapy/PTT. The scaffold of NP1 undergoes disassembly in the hypoxic environments, thus triggering the hydrolysis of P2 to continuously long-term release H2S. Dependent on the superior photothermal ability of B1, NP1 elicits high photothermal conversion efficiency (η = 19.9%) under NIR light irradiation for PTT. Moreover, NP1 serves as in situ H2S bombers in the hypoxic tumor environment and suppresses the mitochondrial respiration through inhibiting expression of cytochrome c oxidase (COX IV) and cutting off the adenosine triphosphate (ATP) generation. Both in vitro and in vivo results demonstrate good antitumor efficacy of H2S gas therapy/PTT, which will be recommended as an advanced strategy for cancer therapeutics.
Collapse
Affiliation(s)
- Qi Yu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Food and Biological Engineering, Hubei University of Technology, Wuhan 430068, P.R. China
| | - Le Tu
- Key Laboratory of Pesticide and Chemical Biology (CCNU), Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Ting Zhu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM) & Institute of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P.R. China
| | - Hongda Zhu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Food and Biological Engineering, Hubei University of Technology, Wuhan 430068, P.R. China
| | - Shujuan Liu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM) & Institute of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P.R. China
| | - Yao Sun
- Key Laboratory of Pesticide and Chemical Biology (CCNU), Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Qiang Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM) & Institute of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P.R. China
| |
Collapse
|
17
|
Rasel MSI, Mohona FA, Akter W, Kabir S, Chowdhury AA, Chowdhury JA, Hassan MA, Al Mamun A, Ghose DK, Ahmad Z, Khan FS, Bari MF, Rahman MS, Amran MS. Exploration of Site-Specific Drug Targeting-A Review on EPR-, Stimuli-, Chemical-, and Receptor-Based Approaches as Potential Drug Targeting Methods in Cancer Treatment. JOURNAL OF ONCOLOGY 2022; 2022:9396760. [PMID: 36284633 PMCID: PMC9588330 DOI: 10.1155/2022/9396760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
Cancer has been one of the most dominant causes of mortality globally over the last few decades. In cancer treatment, the selective targeting of tumor cells is indispensable, making it a better replacement for conventional chemotherapies by diminishing their adverse side effects. While designing a drug to be delivered selectively in the target organ, the drug development scientists should focus on various factors such as the type of cancer they are dealing with according to which drug, targeting moieties, and pharmaceutical carriers should be targeted. All published articles have been collected regarding cancer and drug-targeting approaches from well reputed databases including MEDLINE, Embase, Cochrane Library, CENTRAL and ClinicalTrials.gov, Science Direct, PubMed, Scopus, Wiley, and Springer. The articles published between January 2010 and December 2020 were considered. Due to the existence of various mechanisms, it is challenging to choose which one is appropriate for a specific case. Moreover, a combination of more than one approach is often utilized to achieve optimal drug effects. In this review, we have summarized and highlighted central mechanisms of how the targeted drug delivery system works in the specific diseased microenvironment, along with the strategies to make an approach more effective. We have also included some pictorial illustrations to have a precise idea about different types of drug targeting. The core contribution of this work includes providing a cancer drug development scientist with a broad preliminary idea to choose the appropriate approach among the various targeted drug delivery mechanisms. Also, the study will contribute to improving anticancer treatment approaches by providing a pathway for lesser side effects observed in conventional chemotherapeutic techniques.
Collapse
Affiliation(s)
- Md. Shamiul Islam Rasel
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Farhana Afrin Mohona
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Wahida Akter
- College of Pharmacy, University of Houston, Houston, USA
| | - Shaila Kabir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Abu Asad Chowdhury
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Md. Abul Hassan
- Department of Science & Technology, Tokushima University Graduate School, Tokushima, Japan
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Dipayon Krisna Ghose
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, King Khalid University, Abha 61413, Saudi Arabia
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Md. Fazlul Bari
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Shah Amran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| |
Collapse
|
18
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
19
|
Wang Z, Mu X, Yang Q, Luo J, Zhao Y. Hypoxia-responsive nanocarriers for chemotherapy sensitization via dual-mode inhibition of hypoxia-inducible factor-1 alpha. J Colloid Interface Sci 2022; 628:106-115. [PMID: 35987150 DOI: 10.1016/j.jcis.2022.08.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/23/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
The overexpression of hypoxia-inducible factor-1 alpha (HIF-1α) in solid tumor compromises the potency of chemotherapy under hypoxia. The high level of HIF-1α arises from the stabilization effect of reduced nicotinamideadeninedinucleotide(phosphate) NAD(P)H: quinone oxidoreductase 1 (NQO1). It was postulated that the inhibition of NQO1 could degrade HIF-1α and sensitize hypoxic cancer cells to antineoplastic agents. In the current work, we report hypoxia-responsive polymer micelles, i.e. methoxyl poly(ethylene glycol)-co-poly(aspartate-nitroimidazole) orchestrate with a NQO1 inhibitor (dicoumarol) to sensitize the ovarian cancer cell line (SKOV3) to a model anticancer agent (sorafenib) at low oxygen conditions. Both cargos were physically encapsulated in the nanoscale micelles. The placebo micelles transiently induced the depletion of reduced nicotinamideadeninedinucleotidephosphate (NADPH) as well as glutathione and thioredoxin under hypoxia, which further inactivated NQO1 because NADPH was the cofactor of NQO1. As a consequence, the expression of HIF-1α was repressed due to the dual action of dicoumarol and polymer. The degradation of HIF-1α significantly increased the vulnerability of SKOV3 cells to sorafenib-induced apoptosis, as indicated by the enhancement of cytotoxicity, and increase of caspase 3 and cytochrome C. The current work opens new avenues of addressing hypoxia-induced drug resistance in chemotherapy.
Collapse
Affiliation(s)
- Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Xuewen Mu
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Qian Yang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Jiajia Luo
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
20
|
Hypoxia responsive and tumor-targeted mixed micelles for enhanced cancer therapy and real-time imaging. Colloids Surf B Biointerfaces 2022; 215:112526. [PMID: 35512561 DOI: 10.1016/j.colsurfb.2022.112526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022]
Abstract
Most chemotherapy agents have serious side effects due to lack of tumor targeting, which affects their clinical application. In addition, as an essential characteristic of malignant tumor, hypoxia is attracting exclusive research focus regarding its non-invasive real-time tracing in novel targeting delivery system. Herein, we designed a mixed micelle with tumor targeting and hypoxia responsiveness for tumor therapy and imaging. In particular, the dual-modified mix micelles were self-assembled by folic acid (FA) and 2-(2-nitroimidazole) ethylamine (NI) conjugated polymers, in which paclitaxel (PTX) and quantum dots (QDs) were co-loaded into the hydrophobic core. The drug loaded micelles showed satisfactory drug encapsulation, good storage stability, and sustained release properties. In vitro cell experiments showed that the mixed micelles exhibited enhanced cytotoxic effect and improved the cellular uptake, especially under hypoxic conditions, which was due to the FA mediated active targeting effect and NI induced hypoxic responsive release. In vivo experiments further proved that the mixed micelles possessed outstanding tumor targeting and hypoxia responsive properties. Furthermore, the drug loaded micelles showed excellent anti-tumor effect and can realize real-time in vivo imaging. This work demonstrates that the dual-modified mixed micelles co-loading with PTX and QDs might provide a novel approach for tumor therapy and imaging.
Collapse
|
21
|
Lin J, Yin M, Liu X, Meng F, Luo L. Nanomaterials Based on Functional Polymers for Sensitizing Cancer Radiotherapy. Macromol Rapid Commun 2022; 43:e2200194. [PMID: 35578790 DOI: 10.1002/marc.202200194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/21/2022] [Indexed: 11/12/2022]
Abstract
Despite being the mainstay treatment for many types of cancer in clinic, radiotherapy is undertaking great challenges in overcoming a series of limitations. Radiosensitizers are promising agents capable of depositing irradiation energy and generating free radicals to enhance the radiosensitivity of tumor cells. Combining radiosensitizers with functional polymer-based nanomaterials holds great potential to improve biodistribution, circulation time, and stability in vivo. The derived polymeric nano-radiosensitizers can significantly improve the efficiency of tumor targeting and radiotherapy, and reduce the side effect to healthy tissues. In this review, we provide an overview of functional polymer-based nanomaterials for radiosensitization in recent years. Particular emphases are given to the action mechanisms, drug loading methods, targeting efficiencies, the impact on therapeutic effects and biocompatibility of various radiosensitizing polymers, which are classified as polymeric micelles, dendrimers, polymeric nanospheres, nanoscale coordination polymers, polymersomes, and nanogels. The challenges and outlooks of polymeric nano-radiosensitizers are also discussed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jinfeng Lin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mingming Yin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
22
|
Zhu X, Wang X, Li B, Zhang Y, Chen Y, Zhang W, Wang Y, Zhai W, Liu Z, Liu S, Sun J, Chen Z, Gao Y. A Three-In-One Assembled Nanoparticle Containing Peptide-Radio-Sensitizer Conjugate and TLR7/8 Agonist Can Initiate the Cancer-Immunity Cycle to Trigger Antitumor Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107001. [PMID: 35434938 DOI: 10.1002/smll.202107001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/24/2022] [Indexed: 06/14/2023]
Abstract
Radiotherapy (RT) has been shown to cause immunogenic cell death (ICD) of cancer cells, which promote the release of tumor-associated antigens, and trigger the cancer-immunity cycle (CIC). However, ICD induced by RT usually does not occur in hypoxic tumor cells due to their resistance to radiation. Moreover, RT also induces programmed death ligand 1 (PD-L1) upregulation on tumor cells, which has an inhibitory effect on T lymphocytes. Therefore, therapy based on CIC must selectively target the restricted steps of antitumor immunity. Herein, the authors design a versatile three-in-one assembling nanoparticle that can simultaneously execute these obstacles. The amphiphilic peptide drug conjugate NIA-D1, containing the hydrophobic radio-sensitizer 2-(2-nitroimidazol-1-yl) acetic acid (NIA), a peptide substrate of matrix metalloproteinase-2, and a hydrophilic PD-L1 antagonist D PPA-1, is constructed and co-assembled with hydrophobic Toll-like receptor (TLR) 7/8 agonist R848 to form nanoparticle NIA-D1@R848. The NIA-D1@R848 nanoparticles combined with RT can trigger the apoptosis of tumor cells and initiate the CIC. In the presence of R848, it promotes the maturation of dendritic cells, which together with protein programmed cell death protein 1 (PD-1) and its ligand PD-L1 blockade to relieve T cell suppression, and amplify the antitumor immune cycle. In conclusion, a functionalized three-in-one nanoparticle NIA-D1@R848 is successfully constructed, which can induce strong systemic antitumor immune response.
Collapse
Affiliation(s)
- Xueqin Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoxi Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bingyu Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yun Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yalan Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenyan Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yan Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou, 450001, China
- International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou, 450001, China
| | - Zimai Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Sijia Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiaxin Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenzhen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou, 450001, China
- International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou, 450001, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
23
|
Javani S, Barsbay M, Ghaffarlou M, Mousazadeh N, Mohammadi A, Mozafari F, Rezaeejam H, Nasehi L, Nosrati H, Kavetskyy T, Danafar H. Metronidazole conjugated bismuth sulfide nanoparticles for enhanced X-ray radiation therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Roy I, Krishnan S, Kabashin AV, Zavestovskaya IN, Prasad PN. Transforming Nuclear Medicine with Nanoradiopharmaceuticals. ACS NANO 2022; 16:5036-5061. [PMID: 35294165 DOI: 10.1021/acsnano.1c10550] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nuclear medicine is expected to make major advances in cancer diagnosis and therapy; tumor-targeted radiopharmaceuticals preferentially eradicate tumors while causing minimal damage to healthy tissues. The current scope of nuclear medicine can be significantly expanded by integration with nanomedicine, which utilizes nanoparticles for cancer diagnosis and therapy by capitalizing on the increased surface area-to-volume ratio, the passive/active targeting ability and high loading capacity, the greater interaction cross section with biological tissues, the rich surface properties of nanomaterials, the facile decoration of nanomaterials with a plethora of functionalities, and the potential for multiplexing several functionalities within one construct. This review provides a comprehensive discussion of nuclear nanomedicine using tumor-targeted nanoparticles for cancer radiation therapy with either pre-embedded radionuclides or nonradioactive materials which can be extrinsically triggered using various external nuclear particle sources to produce in situ radioactivity. In addition, it describes the prospect of combining nuclear nanomedicine with other modalities to enable synergistically enhanced combination therapies. The review also discusses advances in the fabrication of radionuclides as well as describes laser ablation technologies for producing nanoradiopharmaceuticals, which combine the ease of production with exceptional purity and rapid biodegradability, along with additional imaging or therapeutic functionalities. From a practical standpoint, these attributes of nanoradiopharmaceuticals may provide distinct advantages in diagnostic/therapeutic sensitivity and specificity, imaging resolution, and scalability of turnkey platforms. Coupling image-guided targeted radiation therapy with the possibility of in situ activation of nanomaterials as well as combining with other therapeutic modalities using a multifunctional nanoplatform could herald an era of exciting technological and therapeutic advances to radically transform the landscape of nuclear medicine. The review concludes with a discussion of current challenges and presents the authors' views on future opportunities to stimulate further research in this rewarding field of high societal impact.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy - Case 917, 13288 Marseille, France
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
| | - Irina N Zavestovskaya
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Nuclear Physics and Astrophysics Department, LPI of RAS, 119991 Moscow, Russia
| | - Paras N Prasad
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Department of Chemistry and Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
25
|
Wang Y, Huo J, Li S, Huang R, Fan D, Cheng H, Wan B, Du Y, He H, Zhang G. Self-Rectifiable and Hypoxia-Assisted Chemo-Photodynamic Nanoinhibitor for Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10092-10101. [PMID: 35170301 DOI: 10.1021/acsami.1c23121] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) can eradicate cancer cells under light irradiation, mainly because of reactive singlet oxygen (1O2) being transformed from intratumoral oxygen. Nonetheless, the consumption of oxygen during PDT results in serious hypoxic conditions and an elevated hypoxia-inducing factor-1α (HIF-1α) level that hamper further photodynamic efficacy and induce tumor metastasis. To address this problem, we developed hypoxia-assisted NP-co-encapsulating Ce6 (photosensitizer) and YC-1 (HIF-1α inhibitor) as a self-rectifiable nanoinhibitor for synergistic antitumor treatment. PDT-aggravated intracellular hypoxic stress facilitated NP dissociation to release the drug (YC-1), which achieved tumor killing and HIF-1α inhibition to further enhance the therapeutic effect of PDT and prevent tumor metastasis. Besides, in vivo studies revealed that the HC/PI@YC-1 NPs afforded synergistic anticancer efficacy with minimal toxicity. Therefore, this study provides a prospective approach against PDT drawbacks and combination cancer therapy.
Collapse
Affiliation(s)
- Yanan Wang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Jian Huo
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Shuang Li
- Department of Pathology, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou 450003, China
| | - Ran Huang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Daopeng Fan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hanghang Cheng
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Bo Wan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yongkun Du
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hua He
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Gaiping Zhang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
26
|
Pharmaceutical nanoformulation strategies to spatiotemporally manipulate oxidative stress for improving cancer therapies — exemplified by polyunsaturated fatty acids and other ROS-modulating agents. Drug Deliv Transl Res 2022; 12:2303-2334. [DOI: 10.1007/s13346-021-01104-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 12/18/2022]
|
27
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
28
|
Zhou Q, Mohammed F, Wang Y, Wang J, Lu N, Li J, Ge Z. Hypoxia-responsive block copolymer polyprodrugs for complementary photodynamic-chemotherapy. J Control Release 2021; 339:130-142. [PMID: 34560158 DOI: 10.1016/j.jconrel.2021.09.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/18/2021] [Indexed: 01/12/2023]
Abstract
The inherent hypoxic microenvironment of solid tumors has an important influence on tumor growth, distant metastasis, and invasiveness. The heterogeneous distribution of hypoxic regions inside tumors limits the therapeutic efficacy of O2-assisted therapeutic strategy (e.g. photodynamic therapy (PDT)). On the other hand, the hypoxia-activable prodrugs cannot work effectively in the regions with enough O2 concentration. To address the issues, we prepare a block copolymer polyprodrug consisting of polyethylene glycol (PEG) and copolymerized segments of nitroimidazole-linked camptothecin (CPT) methacrylate and 5,10,15,20-tetraphenylporphyrin (TPP)-containing methacrylate monomers for complementary photodynamic-chemotherapy. The polyprodrug can self-assemble into polymeric micelles in aqueous solution with suitable size and high stability. After intravenous injection, the polyprodrug micelles show tumor accumulation. Followed by light irradiation (650 nm) at tumor sites, TPP moieties induce singlet oxygen (1O2) production in the oxygen-rich area to exert PDT and cause transformation of the oxygen-rich areas into hypoxia. Simultaneously, in the hypoxic areas, the hypoxia-responsive polyprodrugs can be activated to release free CPT due to the cleavage of nitroimidazole linkages. The polyprodrug micelles with the segments for PDT and hypoxia-activable CPT efficiently suppress the growth of HeLa tumors. The well-defined polyprodrug amphiphiles offer an effective strategy to overcome the disadvantages of single treatment of PDT or hypoxia-responsive prodrugs for complementary photodynamic-chemotherapy of cancers.
Collapse
Affiliation(s)
- Qinghao Zhou
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jingbo Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Nannan Lu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Junjie Li
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
29
|
Tinajero-Díaz E, Kimmins SD, García-Carvajal ZY, Martínez de Ilarduya A. Polypeptide-based materials prepared by ring-opening polymerisation of anionic-based α-amino acid N-carboxyanhydrides: A platform for delivery of bioactive-compounds. REACT FUNCT POLYM 2021. [DOI: 10.1016/j.reactfunctpolym.2021.105040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
30
|
Wang J, Liu J, Yang Z. Recent advances in peptide-based nanomaterials for targeting hypoxia. NANOSCALE ADVANCES 2021; 3:6027-6039. [PMID: 36133944 PMCID: PMC9418673 DOI: 10.1039/d1na00637a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 06/16/2023]
Abstract
Hypoxia is a prominent feature of many severe diseases such as malignant tumors, ischemic strokes, and rheumatoid arthritis. The lack of oxygen has a paramount impact on angiogenesis, invasion, metastasis, and chemotherapy resistance. The potential of hypoxia as a therapeutic target has been increasingly recognized over the last decade. In order to treat these disease states, peptides have been extensively investigated due to their advantages in safety, target specificity, and tumor penetrability. Peptides can overcome difficulties such as low drug/energy delivery efficiency, hypoxia-induced drug resistance, and tumor nonspecificity. There are three main strategies for targeting hypoxia through peptide-based nanomaterials: (i) using peptide ligands to target cellular environments unique to hypoxic conditions, such as cell surface receptors that are upregulated in cells under hypoxic conditions, (ii) utilizing peptide linkers sensitive to the hypoxic microenvironment that can be cleaved to release therapeutic or diagnostic payloads, and (iii) a combination of the above where targeting peptides will localize the system to a hypoxic environment for it to be selectively cleaved to release its payload, forming a dual-targeting system. This review focuses on recent developments in the design and construction of novel peptide-based hypoxia-targeting nanomaterials, followed by their mechanisms and potential applications in diagnosis and treatment of hypoxic diseases. In addition, we address challenges and prospects of how peptide-based hypoxia-targeting nanomaterials can achieve a wider range of clinical applications.
Collapse
Affiliation(s)
- Jun Wang
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| | - Jing Liu
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| | - Zhongxing Yang
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| |
Collapse
|
31
|
Bhardwaj P, Goda JS, Pai V, Chaudhari P, Mohanty B, Pai T, Vishwakarma K, Thorat R, Wadasadawala T, Banerjee R. Ultrasound augments on-demand breast tumor radiosensitization and apoptosis through a tri-responsive combinatorial delivery theranostic platform. NANOSCALE 2021; 13:17077-17092. [PMID: 34622906 DOI: 10.1039/d1nr04211d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Advanced inoperable triple-negative breast cancer (TNBC) comprises aggressive tumors with a modest pathological response to neoadjuvant chemotherapy. The concomitant use of chemoradiotherapy improves the pathological response rates. However, the dose-dependent systemic toxicity of clinical radiosensitizers with poor circulation half-life and limited passive bioavailability limits their clinical utility. We address these challenges by rationally designing a stealth and tumor microenvironment responsive nano-conjugate platform for the ultrasound-mediated on-demand spatio-temporal delivery of plant flavonoid curcumin as a combinatorial regimen with clinically approved paclitaxel for the neoadjuvant chemoradiotherapy of locally advanced triple-negative breast cancer (TNBC). Interestingly, the focused application of ultrasound at the orthotopic TNBC xenograft of NOD-SCID mice facilitated the immediate infiltration of nano-conjugates at the tumor interstitium, and conferred in vivo safety over marketed paclitaxel formulation. In addition, curcumin significantly potentiated the in vivo chemoradiotherapeutic efficacy of paclitaxel upon loading into nano-conjugates. This gets further enhanced by the concurrent pulse of ultrasound, as confirmed by PET-CT imaging, along with a significant improvement in the mice survival. The quadrapeutic apoptotic effect by the combination of paclitaxel, curcumin, radiation, and ultrasound, along with a reduction in the tumor microvessel density and cell proliferation marker, confers the broad chemo-radiotherapeutic potential of this regimen for radio-responsive solid tumors, as well as metastatic niches.
Collapse
Affiliation(s)
- Prateek Bhardwaj
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India.
| | - Jayant Sastri Goda
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Venkatesh Pai
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Pradip Chaudhari
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Bhabani Mohanty
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Trupti Pai
- Department of Pathology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Komal Vishwakarma
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Rahul Thorat
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Tabassum Wadasadawala
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India.
| |
Collapse
|
32
|
Liu W, Chen B, Zheng H, Xing Y, Chen G, Zhou P, Qian L, Min Y. Advances of Nanomedicine in Radiotherapy. Pharmaceutics 2021; 13:pharmaceutics13111757. [PMID: 34834172 PMCID: PMC8622383 DOI: 10.3390/pharmaceutics13111757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) remains one of the current main treatment strategies for many types of cancer. However, how to improve RT efficiency while reducing its side effects is still a large challenge to be overcome. Advancements in nanomedicine have provided many effective approaches for radiosensitization. Metal nanoparticles (NPs) such as platinum-based or hafnium-based NPs are proved to be ideal radiosensitizers because of their unique physicochemical properties and high X-ray absorption efficiency. With nanoparticles, such as liposomes, bovine serum albumin, and polymers, the radiosensitizing drugs can be promoted to reach the tumor sites, thereby enhancing anti-tumor responses. Nowadays, the combination of some NPs and RT have been applied to clinical treatment for many types of cancer, including breast cancer. Here, as well as reviewing recent studies on radiotherapy combined with inorganic, organic, and biomimetic nanomaterials for oncology, we analyzed the underlying mechanisms of NPs radiosensitization, which may contribute to exploring new directions for the clinical translation of nanoparticle-based radiosensitizers.
Collapse
Affiliation(s)
- Wei Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
| | - Bo Chen
- Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China; (B.C.); (Y.M.)
| | - Haocheng Zheng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Yun Xing
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Guiyuan Chen
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Peijie Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
| | - Liting Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
- Correspondence:
| | - Yuanzeng Min
- Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China; (B.C.); (Y.M.)
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
33
|
Zhao J, Peng YY, Diaz-Dussan D, White J, Duan W, Kong L, Narain R, Hao X. Zwitterionic Block Copolymer Prodrug Micelles for pH Responsive Drug Delivery and Hypoxia-Specific Chemotherapy. Mol Pharm 2021; 19:1766-1777. [PMID: 34473523 DOI: 10.1021/acs.molpharmaceut.1c00518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tirapazamine (TPZ) and its derivatives (TPZD) have shown their great potential for efficiently killing hypoxic cancer cells. However, unsatisfactory clinical outcomes resulting from the low bioavailability of the low-molecular TPZ and TPZD limited their further applications. Precise delivery and release of these prodrugs via functional nanocarriers can significantly improve the therapeutic effects due to the targeted drug delivery and enhanced permeability and retention (EPR) effect. Herein, zwitterionic block copolymer (BCP) micelles with aldehyde functional groups are prepared from the self-assembly of poly(2-methacryloyloxyethyl phosphorylcholine-b-poly(di(ethylene glycol) methyl ether methacrylate-co-4-formylphenyl methacrylate) [PMPC-b-P(DEGMA-co-FPMA)]. TPZD is then grafted onto PMPC-b-P(DEGMA-co-FPMA) to obtain a polymer-drug conjugate, PMPC-b-P(DEGMA-co-FPMA-g-TPZD) (BCP-TPZ), through the formation of a pH-responsive imine bond, exhibiting a pH-dependent drug release profile owing to the cleavage of the imine bond under acidic conditions. Outstandingly, BCP-TPZ shows around 13.7-fold higher cytotoxicity to hypoxic cancer cells in comparison to normoxic cancer cells evaluated through an in vitro cytotoxicity assay. The pH-responsiveness and hypoxia-specific cytotoxicity confer BCP-TPZ micelles a great potential to achieve precise delivery of TPZD and thus enhance the therapeutic effect toward tumor-hypoxia.
Collapse
Affiliation(s)
- Jianyang Zhao
- Institute for Frontier Materials, Deakin University, Geelong, Victoria 3216, Australia.,Manufacturing, CSIRO, Research Way, Clayton, Victoria 3168, Australia
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering, University of Alberta, 116 Street and 85th Avenue, Edmonton T6G 2G6, Alberta, Canada
| | - Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, University of Alberta, 116 Street and 85th Avenue, Edmonton T6G 2G6, Alberta, Canada
| | - Jacinta White
- Manufacturing, CSIRO, Research Way, Clayton, Victoria 3168, Australia
| | - Wei Duan
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia
| | - Lingxue Kong
- Institute for Frontier Materials, Deakin University, Geelong, Victoria 3216, Australia
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, 116 Street and 85th Avenue, Edmonton T6G 2G6, Alberta, Canada
| | - Xiaojuan Hao
- Manufacturing, CSIRO, Research Way, Clayton, Victoria 3168, Australia
| |
Collapse
|
34
|
Photothermal photodynamic therapy and enhanced radiotherapy of targeting copolymer-coated liquid metal nanoparticles on liver cancer. Colloids Surf B Biointerfaces 2021; 207:112023. [PMID: 34403983 DOI: 10.1016/j.colsurfb.2021.112023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/13/2021] [Accepted: 08/04/2021] [Indexed: 02/08/2023]
Abstract
The maximized therapeutic efficacy in tumor treatment can be achieved with combination therapy. Herein, a metronidazole (MN) and RGD peptides were linked with the copolymer chains of polyacrylic acid (PAA) and polyethylene glycol (PEG) by condensation and Michael addition reactions, respectively, named as RGD-PEG-PAA-MN. Subsequently, liquid-metal (LM) nanoparticles broken by ultrasonication were coated with modified copolymer, forming RGD-PEG-PAA-MN@LM nanoparticles. These nanoparticles with the degradation under an acidic condition could target to tumor cells, and LM of these composited nanoparticles could not only efficiently convert the photoenergy of near infrared (NIR) into thermal energy, but also produce more reactive oxygen species under NIR or X ray irradiation. Furthermore, MN in the composited nanoparticles could enhance their radiation sensitivity of tumor tissues with hypoxia condition. The synergic effect of these nanoparticles on cancer limitation after the sequential radiations of NIR and X ray was significantly higher than the single radiation. In the experiments of tumor bearing mice, the volume of the tumor in RGD-PEG-PAA-MN@LM group at 14th day after two radiations of NIR and X-ray were significantly smaller than LM group, and the tumor of RGD-PEG-PAA-MN@LM group at 14th day after two radiations almost disappeared, suggesting better synergistic effect of RGD-PEG-PAA-MN@LM nanoparticles on photothermal conversion, photodynamics under two irradiations and their enhanced sensitization of X-ray radiation. Our results indicated that the prepared nanoparticles would be applied in the combinational therapy of liver tumor by the photothermal, photodynamic and sensitized radiation.
Collapse
|
35
|
Karayianni M, Pispas S. Block copolymer solution self‐assembly: Recent advances, emerging trends, and applications. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210430] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maria Karayianni
- Theoretical and Physical Chemistry Institute National Hellenic Research Foundation Athens Greece
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute National Hellenic Research Foundation Athens Greece
| |
Collapse
|
36
|
Shrestha B, Wang L, Brey EM, Uribe GR, Tang L. Smart Nanoparticles for Chemo-Based Combinational Therapy. Pharmaceutics 2021; 13:853. [PMID: 34201333 PMCID: PMC8227511 DOI: 10.3390/pharmaceutics13060853] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/27/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Traditional cancer therapy is associated with low therapeutic index, acquired resistance, and various adverse effects. With the increasing understanding of cancer biology and technology advancements, more strategies have been exploited to optimize the therapeutic outcomes. The rapid development and application of nanomedicine have motivated this progress. Combinational regimen, for instance, has become an indispensable approach for effective cancer treatment, including the combination of chemotherapeutic agents, chemo-energy, chemo-gene, chemo-small molecules, and chemo-immunology. Additionally, smart nanoplatforms that respond to external stimuli (such as light, temperature, ultrasound, and magnetic field), and/or to internal stimuli (such as changes in pH, enzymes, hypoxia, and redox) have been extensively investigated to improve precision therapy. Smart nanoplatforms for combinational therapy have demonstrated the potential to be the next generation cancer treatment regimen. This review aims to highlight the recent advances in smart combinational therapy.
Collapse
Affiliation(s)
| | | | | | - Gabriela Romero Uribe
- Department of Biomedical and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (B.S.); (L.W.); (E.M.B.)
| | - Liang Tang
- Department of Biomedical and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (B.S.); (L.W.); (E.M.B.)
| |
Collapse
|
37
|
Exploiting a New Approach to Destroy the Barrier of Tumor Microenvironment: Nano-Architecture Delivery Systems. Molecules 2021; 26:molecules26092703. [PMID: 34062992 PMCID: PMC8125456 DOI: 10.3390/molecules26092703] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 01/04/2023] Open
Abstract
Recent findings suggest that tumor microenvironment (TME) plays an important regulatory role in the occurrence, proliferation, and metastasis of tumors. Different from normal tissue, the condition around tumor significantly altered, including immune infiltration, compact extracellular matrix, new vasculatures, abundant enzyme, acidic pH value, and hypoxia. Increasingly, researchers focused on targeting TME to prevent tumor development and metastasis. With the development of nanotechnology and the deep research on the tumor environment, stimulation-responsive intelligent nanostructures designed based on TME have attracted much attention in the anti-tumor drug delivery system. TME-targeted nano therapeutics can regulate the distribution of drugs in the body, specifically increase the concentration of drugs in the tumor site, so as to enhance the efficacy and reduce adverse reactions, can utilize particular conditions of TME to improve the effect of tumor therapy. This paper summarizes the major components and characteristics of TME, discusses the principles and strategies of relevant nano-architectures targeting TME for the treatment and diagnosis systematically.
Collapse
|
38
|
Zhou H, Qin F, Chen C. Designing Hypoxia-Responsive Nanotheranostic Agents for Tumor Imaging and Therapy. Adv Healthc Mater 2021; 10:e2001277. [PMID: 32985141 DOI: 10.1002/adhm.202001277] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/06/2020] [Indexed: 12/15/2022]
Abstract
Hypoxia, a common feature of most solid tumors, plays an important role in tumor proliferation, metastasis, and invasion, leading to drug, radiation, and photodynamic therapy resistance, and resulting in a sharp reduction in the disease-free survival rate of tumor patients. The lack of sufficient blood supply to the interior regions of tumors hinders the delivery of traditional drugs and contrast agents, interfering with their accumulation in the hypoxic region, and preventing efficient theranostics. Thus, there is a need for the fabrication of novel tumor theranostic agents that overcome these obstacles. Reports, in recent years, of hypoxia-responsive nanomaterials may provide with such means. In this review, a comprehensive description of the physicochemical and biological characteristics of hypoxic tumor tissues is provided, the principles of designing the hypoxia-responsive tumor theranostic agents are discussed, and the recent research into hypoxia-triggered nanomaterials is examined. Additionally, other hypoxia-associated responsive strategies, the current limitations, and future prospects for hypoxia-responsive nanotheranostic agents in tumor treatment are discussed.
Collapse
Affiliation(s)
- Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| | - Fenglan Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| |
Collapse
|
39
|
Yang Y, Zeng W, Huang P, Zeng X, Mei L. Smart materials for drug delivery and cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200042] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yao Yang
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Weiwei Zeng
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Ping Huang
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Xiaowei Zeng
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Lin Mei
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
- 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 China
| |
Collapse
|
40
|
Gao Y, Gao J, Mu G, Zhang Y, Huang F, Zhang W, Ren C, Yang C, Liu J. Selectively enhancing radiosensitivity of cancer cells via in situ enzyme-instructed peptide self-assembly. Acta Pharm Sin B 2020; 10:2374-2383. [PMID: 33354508 PMCID: PMC7745053 DOI: 10.1016/j.apsb.2020.07.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/20/2020] [Accepted: 06/23/2020] [Indexed: 01/20/2023] Open
Abstract
The radiotherapy modulators used in clinic have disadvantages of high toxicity and low selectivity. For the first time, we used the in situ enzyme-instructed self-assembly (EISA) of a peptide derivative (Nap-GDFDFpYSV) to selectively enhance the sensitivity of cancer cells with high alkaline phosphatase (ALP) expression to ionizing radiation (IR). Compared with the in vitro pre-assembled control formed by the same molecule, assemblies formed by in situ EISA in cells greatly sensitized the ALP-high-expressing cancer cells to γ-rays, with a remarkable sensitizer enhancement ratio. Our results indicated that the enhancement was a result of fixing DNA damage, arresting cell cycles and inducing cell apoptosis. Interestingly, in vitro pre-formed assemblies mainly localized in the lysosomes after incubating with cells, while the assemblies formed via in situ EISA scattered in the cell cytosol. The accumulation of these molecules in cells could not be inhibited by endocytosis inhibitors. We believed that this molecule entered cancer cells by diffusion and then in situ self-assembled to form nanofibers under the catalysis of endogenous ALP. This study provides a successful example to utilize intracellular in situ EISA of small molecules to develop selective tumor radiosensitizers. The intracellular in situ enzyme-instructed self-assembly (in situ EISA) was firstly used for selective cancer radiosensitization. Compared with the in vitro pre-assembled control formed by the same molecule, assemblies formed by in-situ EISA in cells greatly sensitized the ALP-high-expressing cancer cells to γ-rays. This work provides a successful example to utilize intracellular in situ EISA of small molecules to develop selective tumor radiosensitizers.
Collapse
Affiliation(s)
- Yang Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jie Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Ganen Mu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yumin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Fan Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Wenxue Zhang
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunhua Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors.
| | - Cuihong Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors.
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors.
| |
Collapse
|
41
|
Hypoxia-activated ROS burst liposomes boosted by local mild hyperthermia for photo/chemodynamic therapy. J Control Release 2020; 328:100-111. [DOI: 10.1016/j.jconrel.2020.08.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/25/2020] [Accepted: 08/18/2020] [Indexed: 12/21/2022]
|
42
|
Azo-inserted responsive hybrid liposomes for hypoxia-specific drug delivery. Acta Biomater 2020; 115:343-357. [PMID: 32771598 DOI: 10.1016/j.actbio.2020.07.061] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 11/21/2022]
Abstract
Stimuli-responsive drug delivery systems using endogenous stimuli from tumor microenvironments such as acidic pH, over-expressed enzyme, and high redox potential as triggers have shown tremendous promise in cancer therapy. However, their clinical application is severely limited because of tumor heterogeneity. Hypoxia, a physiological feature observed in almost all solid tumors and even in nodules with very small size, has currently emerged as a more general but efficient stimulus to trigger release. Herein, we developed hypoxia-responsive hybrid liposomes (HR-HLPs), composed of azo-inserted organokoxysilane-based lipid analogue as a responsive component and commercial phospholipid for reducing the rigidity of liposomal membrane caused by azo, for drug delivery targeting tumor hypoxia. HR-HLPs had the advantages of high structural stability to avoid premature drug leakage when circulating in the blood and high sensitivity in responding to hypoxia once reaching tumor sites. HR-HLPs exhibit deep tumor penetration capability, enabling effective delivery to hypoxic regions distant from tumor vessels. Moreover, HR-HLPs could selectively release their payload, co-localizing with over-expressed hypoxia inducible factor 1α (HIF-1α) in vitro and in vivo. As a result, HR-HLPs showed improved therapeutic outcome accompanied by reduced adverse effects. The results highlighted the potential application of azo-inserted responsive hybrid liposomes for hypoxia-targeted drug delivery. STATEMENT OF SIGNIFICANCE.
Collapse
|
43
|
Li Y, Jeon J, Park JH. Hypoxia-responsive nanoparticles for tumor-targeted drug delivery. Cancer Lett 2020; 490:31-43. [DOI: 10.1016/j.canlet.2020.05.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/02/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
|
44
|
Han L, Wang Y, Huang X, Liu F, Ma C, Feng F, Zhang J, Liu W, Qu W, Pang H, Xue J. Specific-oxygen-supply functionalized core-shell nanoparticles for smart mutual-promotion between photodynamic therapy and gambogic acid-induced chemotherapy. Biomaterials 2020; 257:120228. [DOI: 10.1016/j.biomaterials.2020.120228] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
|
45
|
Intracellular delivery of cytochrome C using hypoxia-responsive polypeptide micelles for efficient cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111069. [DOI: 10.1016/j.msec.2020.111069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 01/13/2023]
|
46
|
Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J Control Release 2020; 322:566-592. [DOI: 10.1016/j.jconrel.2020.03.051] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022]
|
47
|
Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020; 10:4557-4588. [PMID: 32292515 PMCID: PMC7150471 DOI: 10.7150/thno.38069] [Citation(s) in RCA: 314] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
In recent years, much progress has been motivated in stimuli-responsive nanocarriers, which could response to the intrinsic physicochemical and pathological factors in diseased regions to increase the specificity of drug delivery. Currently, numerous nanocarriers have been engineered with physicochemical changes in responding to external stimuli, such as ultrasound, thermal, light and magnetic field, as well as internal stimuli, including pH, redox potential, hypoxia and enzyme, etc. Nanocarriers could respond to stimuli in tumor microenvironments or inside cancer cells for on-demanded drug delivery and accumulation, controlled drug release, activation of bioactive compounds, probes and targeting ligands, as well as size, charge and conformation conversion, etc., leading to sensing and signaling, overcoming multidrug resistance, accurate diagnosis and precision therapy. This review has summarized the general strategies of developing stimuli-responsive nanocarriers and recent advances, presented their applications in drug delivery, tumor imaging, therapy and theranostics, illustrated the progress of clinical translation and made prospects.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, 610041, China
| |
Collapse
|
48
|
Weng J, Huang Z, Pu X, Chen X, Yin G, Tian Y, Song Y. Preparation of polyethylene glycol-polyacrylic acid block copolymer micelles with pH/hypoxic dual-responsive for tumor chemoradiotherapy. Colloids Surf B Biointerfaces 2020; 191:110943. [PMID: 32203861 DOI: 10.1016/j.colsurfb.2020.110943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023]
Abstract
Block copolymers of poly(ethylene glycol)-poly(acrylic acid) linked with metronidazole (MN-PAA-PEG) were prepared via carbodiimide and esterification methods, and self-assembled into core-shell micelles as nano radiosensitizers and carriers of doxorubicin (DOX) delivery. These DOX/MN-PAA-PEG micelles exhibited good pH value and hypoxia dual-responsive properties via analyzing the change of micelle size and drug‒release behavior under hypoxia humor condition. The results of the cell test indicated that DOX was efficiently delivered by DOX/MN-PAA-PEG micelles into the cell nuclei. Compared to 22.4 % of their DOX release under pH 7.4, the rate of DOX release from DOX/MN-PAA-PEG micelles under reducing condition (pH 5.0) was up to 55.9 %. DOX-loaded micelles under 600 MU electron radiation and hypoxia induced the rapidest apoptosis of the tumor-cells, indicating the synergistic effect of their radiotherapy and chemotherapy from the prepared micelles. In vivo investigation and fluorescence imaging revealed that MN-PAA-PEG possessed no toxicity on main organs, and DOX/MN-PAA-PEG micelles were mainly accumulated in the tumor site at 10 h of post-injection, suggesting their good passive tumor-targeted effect. These results suggested that DOX/MN-PAA-PEG micelles were promising candidates for chemoradiotherapy on tumor.
Collapse
Affiliation(s)
- Jiajia Weng
- College of Materials Science and Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Zhongbing Huang
- College of Materials Science and Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China.
| | - Ximing Pu
- College of Materials Science and Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Xianchun Chen
- College of Materials Science and Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Guangfu Yin
- College of Materials Science and Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Yaping Tian
- College of Materials Science and Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Ying Song
- Department of Radiology, West China Hospital of Sichuan University, No. 37, Guangxue Lane, Chengdu 610093, China.
| |
Collapse
|
49
|
Denkova AG, Liu H, Men Y, Eelkema R. Enhanced Cancer Therapy by Combining Radiation and Chemical Effects Mediated by Nanocarriers. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900177] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Antonia G. Denkova
- Department of Radiation Science and TechnologyDelft University of Technology Mekelweg 15 2629 JB Delft The Netherlands
| | - Huanhuan Liu
- Department of Radiation Science and TechnologyDelft University of Technology Mekelweg 15 2629 JB Delft The Netherlands
| | - Yongjun Men
- Department of Chemical EngineeringDelft University of Technology van der Maasweg 9 2629 HZ Delft The Netherlands
| | - Rienk Eelkema
- Department of Chemical EngineeringDelft University of Technology van der Maasweg 9 2629 HZ Delft The Netherlands
| |
Collapse
|
50
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|