201
|
Bao Y, Ge Y, Wu M, Mao Z, Ye J, Tong W. Record-High Ultrasound-Sensitive NO Nanogenerators for Cascade Tumor Pyroptosis and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302278. [PMID: 37400368 PMCID: PMC10502831 DOI: 10.1002/advs.202302278] [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: 04/10/2023] [Revised: 05/30/2023] [Indexed: 07/05/2023]
Abstract
Pyroptosis is a pro-inflammatory cell death that is associated with innate immunity promotion against tumors. Excess nitric oxide (NO)-triggered nitric stress has potential to induce pyroptosis, but the precise delivery of NO is challenging. Ultrasound (US)-responsive NO production has dominant priority due to its deep penetration, low side effects, noninvasion, and local activation manner. In this work, US-sensitive NO donor N-methyl-N-nitrosoaniline (NMA) with thermodynamically favorable structure is selected and loaded into hyaluronic acid (HA)-modified hollow manganese dioxide nanoparticles (hMnO2 NPs) to fabricate hMnO2 @HA@NMA (MHN) nanogenerators (NGs). The obtained NGs have a record-high NO generation efficiency under US irradiation and can release Mn2+ after targeting the tumor sites. Later on, cascade tumor pyroptosis and cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING)-based immunotherapy is achieved and tumor growth is effectively inhibited.
Collapse
Affiliation(s)
- Yuheng Bao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationMinistry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Yanni Ge
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityZhejiang Provincial Key Laboratory of OphthalmologyZhejiang Provincial Clinical Research Center for Eye DiseasesZhejiang Provincial Engineering Institute on Eye DiseasesHangzhouZhejiang310009China
| | - Mengjie Wu
- Stomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiang310058China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationMinistry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Juan Ye
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityZhejiang Provincial Key Laboratory of OphthalmologyZhejiang Provincial Clinical Research Center for Eye DiseasesZhejiang Provincial Engineering Institute on Eye DiseasesHangzhouZhejiang310009China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationMinistry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| |
Collapse
|
202
|
Li F, Gong J, Shi T, Ren X, Cui X, Xiao L, Liu J, Qiu F. The design and straightforward synthesis of multifunctional DNA microgels for the improved targeted delivery of antitumor drugs. Int J Pharm 2023; 643:123242. [PMID: 37467815 DOI: 10.1016/j.ijpharm.2023.123242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/28/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
Multifunctional drug delivery platforms represent ideal approaches to reliably targeting pharmacological agents of interest to the complex tumor microenvironment (TME), yet the complicated synthesis processes, high costs, and toxicities associated with these agents have hindered their clinical application to date. In this study, the properties of the TME are leveraged to develop a multifunctional pNAB/AS DNA microgel that is able to actively target tumors. This microgel is generated by a straightforward one-step free radical precipitation polymerization procedure, exhibiting extremely high drug encapsulation efficiency (∼90%), and is responsive to three environmental stimuli including temperature, reduction, and an acidic pH while showing minimal drug leakage under physiological conditions. Through a synergistic combination of appropriate size and aptamer recognition, this microgel is able to reliably facilitate intratumoral drug accumulation and nuclear drug delivery. Critically, pNAB/AS-Dox treatment is associated with specific antitumor activity in vitro and in vivo while retaining a good biosafety profile and causing lower levels of off-target toxicity as compared to free drug treatment. Together, these findings emphasize the potential value of this multifunctional pNAB/AS DNA microgel as a platform amenable to targeted drug delivery to the TME, providing a foundation for further efforts to readily develop multifunctional drug delivery systems.
Collapse
Affiliation(s)
- Fengyun Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jianing Gong
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Taoran Shi
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyi Cui
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Li Xiao
- Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
203
|
Cui S, Wang B, Zhai C, Wei S, Zhang H, Sun G. A double rare earth doped CD nanoplatform for nanocatalytic/starving-like synergistic therapy with GSH-depletion and enhanced reactive oxygen species generation. J Mater Chem B 2023; 11:7986-7997. [PMID: 37523206 DOI: 10.1039/d3tb00959a] [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: 08/01/2023]
Abstract
Cancer has been one of the principal diseases threatening human health in the world. Traditional chemotherapy, radiotherapy and surgery in clinical applications have some disadvantages, such as inefficiency, low specificity, and serious side effects. Therefore, some emerging synergistic therapies have been developed for more accurate diagnosis and more efficient treatment of cancer. Herein, novel Ce-Gd@CDs-GOx nanozymes were obtained by combining magnetic resonance/fluorescence (MR/FL) imaging and nanocatalytic/starving-like synergistic therapy for tumor tissue imaging and efficient cancer treatment. The as-prepared Ce-Gd@CDs-GOx nanozymes with a diameter of 25.0 ± 0.8 nm exhibited favorable physiological stability, negligible toxicity, bright fluorescence and strong T1-weighted MR imaging (MRI) performance (10.97 mM-1 s-1). Moreover, the nanozymes could not only cut off the nutrient supply of tumor cells, but also generate ROS to synergistically enhance antitumor efficacy. The coexistence of Ce3+/Ce4+ in Ce-Gd@CDs-GOx endowed them with attractive capacity for alleviating hypoxia and enhancing GSH consumption to induce the apoptosis of tumor cells. Furthermore, most of the 4T1 cells treated with Ce-Gd@CDs-GOx nanozymes were damaged in the CCK-8 and Calcein-AM/PI staining assays, indicating the excellent efficiency of intracellular synergistic therapy. In summary, this study offered a promising strategy to design a nanoplatform for MR/FL imaging-guided nanocatalytic and starvation-like synergistic therapy of cancer.
Collapse
Affiliation(s)
- Shufeng Cui
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
- Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| | - Bin Wang
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
- Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| | - Changyu Zhai
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
| | - Shanshan Wei
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
| | - Hongyuan Zhang
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
| | - Guoying Sun
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
- Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| |
Collapse
|
204
|
Luo G, Li X, Lin J, Ge G, Fang J, Song W, Xiao GG, Zhang B, Peng X, Duo Y, Tang BZ. Multifunctional Calcium-Manganese Nanomodulator Provides Antitumor Treatment and Improved Immunotherapy via Reprogramming of the Tumor Microenvironment. ACS NANO 2023; 17:15449-15465. [PMID: 37530575 PMCID: PMC10448754 DOI: 10.1021/acsnano.3c01215] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/12/2023] [Indexed: 08/03/2023]
Abstract
Ions play a vital role in regulating various biological processes, including metabolic and immune homeostasis, which involves tumorigenesis and therapy. Thus, the perturbation of ion homeostasis can induce tumor cell death and evoke immune responses, providing specific antitumor effects. However, antitumor strategies that exploit the effects of multiion perturbation are rare. We herein prepared a pH-responsive nanomodulator by coloading curcumin (CU, a Ca2+ enhancer) with CaCO3 and MnO2 into nanoparticles coated with a cancer cell membrane. This nanoplatform was aimed at reprogramming the tumor microenvironment (TME) and providing an antitumor treatment through ion fluctuation. The obtained nanoplatform, called CM NPs, could neutralize protons by decomposing CaCO3 and attenuating cellular acidity, they could generate Ca2+ and release CU, elevating Ca2+ levels and promoting ROS generation in the mitochondria and endoplasmic reticulum, thus, inducing immunogenic cell death. Mn2+ could decompose the endogenous H2O2 into O2 to relieve hypoxia and enhance the sensitivity of cGAS, activating the cGAS-STING signaling pathway. In addition, this strategy allowed the reprogramming of the immune TME, inducing macrophage polarization and dendritic cell maturation via antigen cross-presentation, thereby increasing the immune system's ability to combat the tumor effectively. Moreover, the as-prepared nanoparticles enhanced the antitumor responses of the αPD1 treatment. This study proposes an effective strategy to combat tumors via the reprogramming of the tumor TME and the alteration of essential ions concentrations. Thus, it shows great potential for future clinical applications as a complementary approach along with other multimodal treatment strategies.
Collapse
Affiliation(s)
- Guanghong Luo
- School of
Medicine, The 2nd Affiliated Hospital, The
Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
- Department
of Radiation Oncology, Shenzhen People’s
Hospital (The Second Clinical Medical College, Jinan University; The
First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong China
| | - Xing Li
- School
of
Medicine, Southern University of Science
and Technology, Shenzhen 518055, China
| | - Jihui Lin
- School of
Medicine, The 2nd Affiliated Hospital, The
Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
- School
of
Nursing, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Gao Ge
- Department
of Laboratory Medicine, The Third Xiangya
Hospital, Central South University, Changsha, 410013, China
| | - Jiangli Fang
- Department
of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Wangze Song
- State Key
Laboratory of Fine Chemicals, Department of Pharmacology, School of
Chemical Engineering, Dalian University
of Technology, Dalian, 116024, China
| | - Gary Guishan Xiao
- Research
Center for Cancer Metabolism, College of Pharmacology, Shenzhen University of Technology, Chinese Academy
of Sciences, Shenzhen, 518118, China
- State Key
Laboratory of Fine Chemicals, Department of Pharmacology, School of
Chemical Engineering, Dalian University
of Technology, Dalian, 116024, China
| | - Bo Zhang
- School of
Medicine, The 2nd Affiliated Hospital, The
Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
- Department
of Neurosurgery, The Shenzhen Luohu Hospital Group, The Third Affiliated Hospital of Shenzhen University, Shenzhen 518001, China
| | - Xiaojun Peng
- State
Key
Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Yanhong Duo
- Department
of Radiation Oncology, Shenzhen People’s
Hospital (The Second Clinical Medical College, Jinan University; The
First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong China
- Department
of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, 171 77, Sweden
- Key Lab for
New Drug Research of TCM, Research Institute
of Tsinghua University in Shenzhen, Shenzhen 518057, Guangdong China
| | - Ben Zhong Tang
- Shenzhen
Institute of Aggregate Science and Technology, School of Science and
Engineering, The Chinese University of Hong
Kong, Shenzhen. Shenzhen 518172, Guangdong China
| |
Collapse
|
205
|
Deng C, Zheng M, Han S, Wang Y, Xin J, Aras O, Cheng L, An F. GSH-activated Porphyrin Sonosensitizer Prodrug for Fluorescence Imaging-guided Cancer Sonodynamic Therapy. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2300348. [PMID: 38045635 PMCID: PMC10691834 DOI: 10.1002/adfm.202300348] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 12/05/2023]
Abstract
Sonodynamic therapy (SDT), which uses ultrasound to trigger a sonosensitizer to generate reactive oxygen species (ROS), is a promising form of cancer therapy with outstanding tissue penetration depth. However, the sonosensitizer may inevitably spread to surrounding healthy tissue beyond the tumor, resulting in undesired side effects under an ultrasound stimulus. Herein, as glutathione (GSH) is overexpressed in the tumor microenvironment, a GSH-activatable sonosensitizer prodrug was designed by attaching a quencher to tetraphydroxy porphyrin for tumor therapy. The prodrug exhibited poor fluorescence and low ROS generation capacity under ultrasound irradiation but it can be activated by GSH to simultaneously switch on fluorescence emission and ROS generation in tumor site. Compared with the non-quenched sonosensitizer, the designed prodrug exhibited significantly higher tumor/healthy organ fluorescence ratios, due to the specific fluorescence and ROS activation by overexpressed GSH in the tumor. Finally, the prodrug exhibited efficient tumor growth inhibition under ultrasound irradiation, further demonstrating its promise as a GSH-activated sonosensitizer prodrug for highly effective cancer treatment.
Collapse
Affiliation(s)
- Caiting Deng
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No. 76 Yanta West Road, Xi’an 710061, Shaanxi, China
- School of Public Health, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi’an 710061 Shaanxi, China
| | - Meichen Zheng
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No. 76 Yanta West Road, Xi’an 710061, Shaanxi, China
- School of Public Health, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi’an 710061 Shaanxi, China
| | - Shupeng Han
- School of Public Health, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi’an 710061 Shaanxi, China
| | - Yuanjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jingqi Xin
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No. 76 Yanta West Road, Xi’an 710061, Shaanxi, China
- School of Public Health, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi’an 710061 Shaanxi, China
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No. 76 Yanta West Road, Xi’an 710061, Shaanxi, China
- School of Public Health, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi’an 710061 Shaanxi, China
| |
Collapse
|
206
|
Zhang R, Xu S, Yuan M, Guo L, Xie L, Liao Y, Xu Y, Fu X. An ultrasmall PVP-Fe-Cu-Ni-S nano-agent for synergistic cancer therapy through triggering ferroptosis and autophagy. NANOSCALE 2023; 15:12598-12611. [PMID: 37462439 DOI: 10.1039/d3nr02708b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Photothermal therapy (PTT) is an emerging field where photothermal agents could convert visible or near-infrared (NIR) radiation into heat to kill tumor cells. However, the low photothermal conversion efficiency of photothermal agents and their limited antitumor activities hinder the development of these agents into monotherapies for cancer. Herein, we have fabricated an ultrasmall polyvinylpyrrolidone (PVP)-Fe-Cu-Ni-S (PVP-NP) nano-agent via a simple hot injection method with excellent photothermal conversion efficiency (∼96%). Photothermal therapy with this nano-agent effectively inhibits tumor growth without apparent toxic side-effects. Mechanistically, our results demonstrated that, after NIR irradiation, PVP-NPs can induce ROS/singlet oxygen generation, decrease the mitochondrial membrane potential, release extracellular Fe2+, and consume glutathione, triggering autophagy and ferroptosis of cancer cells. Moreover, PVP-NPs exhibit excellent contrast enhancement according to magnetic resonance imaging (MRI) analysis. In summary, PVP-NPs have a high photothermal conversion efficiency and can be applied for MRI-guided synergistic photothermal/photodynamic/chemodynamic cancer therapy, resolving the bottleneck of existing phototherapeutic agents.
Collapse
Affiliation(s)
- Rongjun Zhang
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Shuxiang Xu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.
- Binjiang Research Institute of Zhejiang University, Hangzhou, Zhejiang 310052, China
| | - Miaomiao Yuan
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Lihao Guo
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Luoyijun Xie
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Yingying Liao
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Yang Xu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.
- Binjiang Research Institute of Zhejiang University, Hangzhou, Zhejiang 310052, China
| | - Xuemei Fu
- International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
207
|
Zhang J, Wang S, Zhang D, He X, Wang X, Han H, Qin Y. Nanoparticle-based drug delivery systems to enhance cancer immunotherapy in solid tumors. Front Immunol 2023; 14:1230893. [PMID: 37600822 PMCID: PMC10435760 DOI: 10.3389/fimmu.2023.1230893] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Immunotherapy has developed rapidly in solid tumors, especially in the areas of blocking inhibitory immune checkpoints and adoptive T-cell transfer for immune regulation. Many patients benefit from immunotherapy. However, the response rate of immunotherapy in the overall population are relatively low, which depends on the characteristics of the tumor and individualized patient differences. Moreover, the occurrence of drug resistance and adverse reactions largely limit the development of immunotherapy. Recently, the emergence of nanodrug delivery systems (NDDS) seems to improve the efficacy of immunotherapy by encapsulating drug carriers in nanoparticles to precisely reach the tumor site with high stability and biocompatibility, prolonging the drug cycle of action and greatly reducing the occurrence of toxic side effects. In this paper, we mainly review the advantages of NDDS and the mechanisms that enhance conventional immunotherapy in solid tumors, and summarize the recent advances in NDDS-based therapeutic strategies, which will provide valuable ideas for the development of novel tumor immunotherapy regimen.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Daidi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin He
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Wang
- Academy of Medical Science, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
208
|
Zhou J, Cao C, Zhang X, Zhang X, Li J, Deng H, Wang S. Gas-assisted phototherapy for cancer treatment. J Control Release 2023; 360:564-577. [PMID: 37442200 DOI: 10.1016/j.jconrel.2023.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
Phototherapies, mainly including photodynamic and photothermal therapy, have made considerable strides in the field of cancer treatment. With the aid of phototherapeutic agents, reactive oxygen species (ROS) or heat are generated under light irradiation to selectively damage cancer cells. However, sole-modality phototherapy faces certain drawbacks, such as limited penetration of phototherapeutic agents into tumor tissues, inefficient ROS generation due to hypoxia, treatment-induced inflammation and resistance of tumor to treatment (e.g., high levels of antioxidants, expression of heat shock protein). Gas therapy, an emerging therapy approach that damages cancer cells by improving the level of certain gas at the tumor site, shows potential to overcome the challenges associated with phototherapies. In addition, with the rapid development of nanotechnology, gas-assisted phototherapy based on nanomedicines has emerged as a promising strategy to enhance the treatment efficacy. This review summarizes recent advances in gas-assisted phototherapy and discusses the prospects and challenges of this strategy in cancer phototherapy.
Collapse
Affiliation(s)
- Jun Zhou
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Chen Cao
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Xinlu Zhang
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Xu Zhang
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Jiansen Li
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Hongzhang Deng
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China.
| | - Sheng Wang
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
209
|
Wu H, Wei M, Hu S, Cheng P, Shi S, Xia F, Xu L, Yin L, Liang G, Li F, Ling D. A Photomodulable Bacteriophage-Spike Nanozyme Enables Dually Enhanced Biofilm Penetration and Bacterial Capture for Photothermal-Boosted Catalytic Therapy of MRSA Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301694. [PMID: 37310410 PMCID: PMC10460864 DOI: 10.1002/advs.202301694] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/22/2023] [Indexed: 06/14/2023]
Abstract
Nanozymes, featuring intrinsic biocatalytic effects and broad-spectrum antimicrobial properties, are emerging as a novel antibiotic class. However, prevailing bactericidal nanozymes face a challenging dilemma between biofilm penetration and bacterial capture capacity, significantly impeding their antibacterial efficacy. Here, this work introduces a photomodulable bactericidal nanozyme (ICG@hMnOx ), composed of a hollow virus-spiky MnOx nanozyme integrated with indocyanine green, for dually enhanced biofilm penetration and bacterial capture for photothermal-boosted catalytic therapy of bacterial infections. ICG@hMnOx demonstrates an exceptional capability to deeply penetrate biofilms, owing to its pronounced photothermal effect that disrupts the compact structure of biofilms. Simultaneously, the virus-spiky surface significantly enhances the bacterial capture capacity of ICG@hMnOx . This surface acts as a membrane-anchored generator of reactive oxygen species and a glutathione scavenger, facilitating localized photothermal-boosted catalytic bacterial disinfection. Effective treatment of methicillin-resistant Staphylococcus aureus-associated biofilm infections is achieved using ICG@hMnOx , offering an appealing strategy to overcome the longstanding trade-off between biofilm penetration and bacterial capture capacity in antibacterial nanozymes. This work presents a significant advancement in the development of nanozyme-based therapies for combating biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Haibin Wu
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhou311399P. R. China
| | - Min Wei
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Shen Hu
- Department of Obstetrics and GynaecologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Pu Cheng
- Department of Obstetrics and GynaecologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Shuhan Shi
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhou311399P. R. China
| | - Fan Xia
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Lenan Xu
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhou311399P. R. China
| | - Lina Yin
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhou311399P. R. China
| | - Guang Liang
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhou311399P. R. China
| | - Fangyuan Li
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
- Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310012P. R. China
- World Laureates Association (WLA) LaboratoriesShanghai201203P. R. China
| | - Daishun Ling
- Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310012P. R. China
- World Laureates Association (WLA) LaboratoriesShanghai201203P. R. China
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240P. R. China
| |
Collapse
|
210
|
Zhang J, Wei L, Ma X, Wang J, Liang S, Chen K, Wu M, Niu L, Zhang Y. pH-sensitive tumor-tropism hybrid membrane-coated nanoparticles for reprogramming the tumor microenvironment and boosting the antitumor immunity. Acta Biomater 2023; 166:470-484. [PMID: 37253416 DOI: 10.1016/j.actbio.2023.05.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/08/2023] [Accepted: 05/23/2023] [Indexed: 06/01/2023]
Abstract
Metabolic dysregulation contributes not only to cancer development but also to a tumor immune microenvironment (TIME), which poses great challenges to chemo- and immunotherapy. Targeting metabolic reprogramming has recently emerged as a promising strategy for cancer treatment, but the lethality against solid tumors appears to be fairly restricted, partially due to the poor solubility of small molecule drugs. Herein, we construct a versatile biomimetic nanoplatform (referred to as HM-BPT) employing pH-sensitive tumor-tropism hybrid membrane-coated Manganese oxide (MnO2) nanoparticles for the delivery of BPTES, a glutamine metabolism inhibitor. Basically, hybrid membranes consisting of mesenchymal stem cell membranes (MSCm) and pH-sensitive liposomes (pSL) enable the biomimetic nanoplatform to target TME and escape from endo/lysosomes after endocytosis. The results reveal that HM-BPT treatment leads to remarkable tumor inhibition, cytotoxic T lymphocyte (CTL) infiltration, as well as M1 phenotype repolarization and stimulator of IFN genes (STING) pathway activation in macrophages in a 4T1 xenograft model. Furthermore, glutathione (GSH) depletion and oxygen (O2) supply synergistically ameliorate the immunosuppressive status of the TME, boosting potent antitumor immune responses. Overall, our study explores an integrated therapeutic platform for TME reprogramming and immune activation, offering tremendous promise for cancer combination therapy. STATEMENT OF SIGNIFICANCE: Metabolic abnormalities and the tumor immune microenvironment (TIME) lead to hyporesponsiveness to conventional therapies, ultimately resulting in refractory malignancies. In the current work, a biomimetic nanoplatform (HM-BPT) was developed for TME metabolic reprogramming in favor of immunotherapy. Particularly, hybrid membrane camouflage endowed the nanoplatform with TME targeting, endo/lysosomal escape, and sensitive release properties. The impact of hybrid membrane fusion ratio on cellular uptake and cell viability was explored, yielding beneficial references for the future development of bioactive nanomaterials. Intravenous administration of HM-BPT substantially relieved tumor burden and restored innate and acquired immune activation in 4T1 xenograft models. In conclusion, the created HM-BPT system has the potential to be a promising nanoplatform for combining cancer therapies.
Collapse
Affiliation(s)
- Jie Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P R China
| | - Liwen Wei
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P R China
| | - Xiaocao Ma
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P R China
| | - Jingguo Wang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P R China
| | - Siping Liang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P R China
| | - Kang Chen
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, Zhongshan 528403, P R China.
| | - Minhao Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P R China.
| | - Li Niu
- Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P R China.
| | - Yuanqing Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P R China.
| |
Collapse
|
211
|
Yoon J, Le XT, Kim J, Lee H, Nguyen NT, Lee WT, Lee ES, Oh KT, Choi HG, Youn YS. Macrophage-reprogramming upconverting nanoparticles for enhanced TAM-mediated antitumor therapy of hypoxic breast cancer. J Control Release 2023; 360:482-495. [PMID: 37423526 DOI: 10.1016/j.jconrel.2023.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/10/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
In an attempt to achieve antitumor effects by switching the phenotype of macrophages from the tumor-promoting M2 type to the tumor-suppressing M1 type, we fabricated mannose-decorated/macrophage membrane-coated, silica-layered NaErF4@NaLuF4 upconverting nanoparticles (UCNPs) co-doped with perfluorocarbon (PFC)/chlorin e6 (Ce6) and loaded with paclitaxel (PTX) (UCNP@mSiO2-PFC/Ce6@RAW-Man/PTX: ∼61 nm; -11.6 mV). These nanoparticles were designed to have two major functionalities, (i) efficient singlet oxygen generation aided by an oxygen supply and (ii) good targeting to tumor-associated macrophage (TAMs) (M2-type), to induce polarization to M1 type macrophages that release proinflammatory cytokines and suppress breast cancers. The primary UCNPs consisted of lanthanide elements (erbium and lutetium) in a core@shell structure, and they facilely emitted 660 nm light in response to a deep-penetrating 808 nm near-infrared laser. Moreover, the UCNPs@mSiO2-PFC/Ce6@RAW-Man/PTX were able to release O2 and generate 1O2 because of the co-doped PFC/Ce6 and upconversion. Our nanocarriers' excellent uptake to RAW 264.7 macrophage cells (M2 type) and efficient M1-type polarization activity were clearly demonstrated using qRT-PCR and immunofluorescence-based confocal laser scanning microscopy. Our nanocarriers displayed significant cytotoxicity to 4T1 cells in 2D culture and 3D co-culture systems of 4T1/RAW 264.7 cells. More importantly, UCNPs@mSiO2-PFC/Ce6@RAW-Man/PTX (+808 nm laser) noticeably suppressed tumor growth in 4T1-xenografted mice, compared with the other treatment groups (332.4 vs. 709.5-1185.5 mm3). We attribute this antitumor efficacy to the prominent M1-type macrophage polarization caused by our nanocarriers through efficient ROS/O2 generation and targeting of M2-type TAMs via mannose ligands on coated macrophage-membrane.
Collapse
Affiliation(s)
- Johyun Yoon
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Juho Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hyunjun Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Eun Seong Lee
- Department of Biotechnology and Department of Biomedical-Chemical Engineering, Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
212
|
Zhang Y, Lei H, Wang P, Zhou Q, Yu J, Leng X, Ma R, Wang D, Dong K, Xing J, Dong Y. Restoration of dysregulated intestinal barrier and inflammatory regulation through synergistically ameliorating hypoxia and scavenging reactive oxygen species using ceria nanozymes in ulcerative colitis. Biomater Res 2023; 27:75. [PMID: 37507801 PMCID: PMC10375752 DOI: 10.1186/s40824-023-00412-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) overproduction and excessive hypoxia play pivotal roles in the initiation and progression of ulcerative colitis (UC). Synergistic ROS scavenging and generating O2 could be a promising strategy for UC treatment. METHODS Ceria nanozymes (PEG-CNPs) are fabricated using a modified reverse micelle method. We investigate hypoxia attenuating and ROS scavenging of PEG-CNPs in intestinal epithelial cells and RAW 264.7 macrophages and their effects on pro-inflammatory macrophages activation. Subsequently, we investigate the biodistribution, pharmacokinetic properties and long-term toxicity of PEG-CNPs in mice. PEG-CNPs are administered intravenously to mice with 2,4,6-trinitrobenzenesulfonic acid-induced colitis to test their colonic tissue targeting and assess their anti-inflammatory activity and mucosal healing properties in UC. RESULTS PEG-CNPs exhibit multi-enzymatic activity that can scavenge ROS and generate O2, promote intestinal epithelial cell healing and inhibit pro-inflammatory macrophage activation, and have good biocompatibility. After intravenous administration of PEG-CNPs to colitis mice, they can enrich at the site of colonic inflammation, and reduce hypoxia-induced factor-1α expression in intestinal epithelial cells by scavenging ROS to generate O2, thus further promoting disrupted intestinal mucosal barrier restoration. Meanwhile, PEG-CNPs can effectively scavenge ROS in impaired colon tissues and relieve colonic macrophage hypoxia to suppress the pro-inflammatory macrophages activation, thereby preventing UC occurrence and development. CONCLUSION This study has provided a paradigm to utilize metallic nanozymes, and suggests that further materials engineering investigations could yield a facile method based on the pathological characteristics of UC for clinically managing UC.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hengyu Lei
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pengchong Wang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Qinyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jie Yu
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xue Leng
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ruirui Ma
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Danyang Wang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Kai Dong
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Jianfeng Xing
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
213
|
Zou J, Zhang F, Fan W, Li L, Yang Z. Editorial: Synthesis of novel photosensitizers for cancer theranostics. Front Chem 2023; 11:1188243. [PMID: 37492526 PMCID: PMC10364599 DOI: 10.3389/fchem.2023.1188243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023] Open
Affiliation(s)
- Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, Miami, FL, United States
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Ling Li
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Zhen Yang
- Fujian Cross Strait Institute of Flexible Electronics (Future Technologies), Fujian Normal University, Fuzhou, China
| |
Collapse
|
214
|
Li Z, Guo L, Lin L, Wang T, Jiang Y, Song J, Feng J, Huang J, Li H, Bai Z, Liu W, Zhang J. Porous SiO 2-Based Reactor with Self-Supply of O 2 and H 2O 2 for Synergistic Photo-Thermal/Photodynamic Therapy. Int J Nanomedicine 2023; 18:3623-3639. [PMID: 37427365 PMCID: PMC10327690 DOI: 10.2147/ijn.s387505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose Although the combined photo-thermal (PTT) and photodynamic therapy (PDT) of tumors have demonstrated promise as effective cancer therapy, the hypoxic and insufficient H2O2 supply of tumors seriously limits the efficacy of PDT, and the acidic environment reduces the catalytic activity of nanomaterial in the tumor microenvironment. To develop a platform for efficiently addressing these challenges, we constructed a nanomaterial of Aptamer@dox/GOD-MnO2-SiO2@HGNs-Fc@Ce6 (AMS) for combination tumor therapy. The treatment effects of AMS were evaluated both in vitro and in vivo. Methods In this work, Ce6 and hemin were loaded on graphene (GO) through π-π conjugation, and Fc was connected to GO via amide bond. The HGNs-Fc@Ce6 was loaded into SiO2, and coated with dopamine. Then, MnO2 was modified on the SiO2. Finally, AS1411-aptamer@dox and GOD were fixed to gain AMS. We characterized the morphology, size, and zeta potential of AMS. The oxygen and reactive oxygen species (ROS) production properties of AMS were analyzed. The cytotoxicity of AMS was detected by MTT and calcein-AM/PI assays. The apoptosis of AMS to a tumor cell was estimated with a JC-1 probe, and the ROS level was detected with a 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA) probe. The anticancer efficacy in vivo was analyzed by the changes in the tumor size in different treatment groups. Results AMS was targeted to the tumor cell and released doxorubicin. It decomposed glucose to produce H2O2 in the GOD-mediated reaction. The generated sufficient H2O2 was catalyzed by MnO2 and HGNs-Fc@Ce6 to produce O2 and free radicals (•OH), respectively. The increased oxygen content improved the hypoxic environment of the tumor and effectively reduced the resistance to PDT. The generated •OH enhanced the ROS treatment. Moreover, AMS depicted a good photo-thermal effect. Conclusion The results revealed that AMS had an excellent enhanced therapy effect by combining synergistic PTT and PDT.
Collapse
Affiliation(s)
- Zhengzhao Li
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Lianshan Guo
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Liqiao Lin
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Tongting Wang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Yanqiu Jiang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jin Song
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jihua Feng
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jianfeng Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Haoyu Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Zhihao Bai
- College of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, China
| | - Wenqi Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Jianfeng Zhang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| |
Collapse
|
215
|
Fan H, McGhee CE, Lake RJ, Yang Z, Guo Z, Zhang XB, Lu Y. A Highly Selective Mn(II)-Specific DNAzyme and Its Application in Intracellular Sensing. JACS AU 2023; 3:1615-1622. [PMID: 37388692 PMCID: PMC10302744 DOI: 10.1021/jacsau.3c00062] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 07/01/2023]
Abstract
Manganese is an essential trace element in the human body that acts as a cofactor in many enzymes and metabolisms. It is important to develop methods to detect Mn2+ in living cells. While fluorescent sensors have been very effective in detecting other metal ions, Mn2+-specific fluorescent sensors are rarely reported due to nonspecific fluorescence quenching by the paramagnetism of Mn2+ and poor selectivity against other metal ions such as Ca2+ and Mg2+. To address these issues, we herein report in vitro selection of an RNA-cleaving DNAzyme with exceptionally high selectivity for Mn2+. Through converting it into a fluorescent sensor using a catalytic beacon approach, Mn2+ sensing in immune cells and tumor cells has been achieved. The sensor is also used to monitor degradation of manganese-based nanomaterials such as MnOx in tumor cells. Therefore, this work provides an excellent tool to detect Mn2+ in biological systems and monitor the Mn2+-involved immune response and antitumor therapy.
Collapse
Affiliation(s)
- Huanhuan Fan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), Nanjing University, Nanjing 210023, China
| | - Claire E. McGhee
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Ryan J. Lake
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Zhenglin Yang
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), Nanjing University, Nanjing 210023, China
| | - Xiao-Bing Zhang
- Molecular
Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing
and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative
Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Yi Lu
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
216
|
Hu T, Huang Y, Liu J, Shen C, Wu F, He Z. Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy. Pharmaceutics 2023; 15:1821. [PMID: 37514008 PMCID: PMC10383408 DOI: 10.3390/pharmaceutics15071821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy can significantly prevent tumor growth and metastasis by activating the autoimmune system without destroying normal cells. Although cancer immunotherapy has made some achievements in clinical cancer treatment, it is still restricted by systemic immunotoxicity, immune cell dysfunction, cancer heterogeneity, and the immunosuppressive tumor microenvironment (ITME). Biomimetic cell-derived nanoparticles are attracting considerable interest due to their better biocompatibility and lower immunogenicity. Moreover, biomimetic cell-derived nanoparticles can achieve different preferred biological effects due to their inherent abundant source cell-relevant functions. This review summarizes the latest developments in biomimetic cell-derived nanoparticles for cancer immunotherapy, discusses the applications of each biomimetic system in cancer immunotherapy, and analyzes the challenges for clinical transformation.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuezhou Huang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chao Shen
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fengbo Wu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
217
|
Kesharwani P, Ma R, Sang L, Fatima M, Sheikh A, Abourehab MAS, Gupta N, Chen ZS, Zhou Y. Gold nanoparticles and gold nanorods in the landscape of cancer therapy. Mol Cancer 2023; 22:98. [PMID: 37344887 DOI: 10.1186/s12943-023-01798-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
Cancer is a grievous disease whose treatment requires a more efficient, non-invasive therapy, associated with minimal side effects. Gold nanoparticles possessing greatly impressive optical properties have been a forerunner in bioengineered cancer therapy. This theranostic system has gained immense popularity and finds its application in the field of molecular detection, biological imaging, cancer cell targeting, etc. The photothermal property of nanoparticles, especially of gold nanorods, causes absorption of the light incident by the light source, and transforms it into heat, resulting in tumor cell destruction. This review describes the different optical features of gold nanoparticles and summarizes the advance research done for the application of gold nanoparticles and precisely gold nanorods for combating various cancers including breast, lung, colon, oral, prostate, and pancreatic cancer.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, China
| | - Mahak Fatima
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Neelima Gupta
- Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York City, NY, 11439, USA
| | - Yun Zhou
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
218
|
Wang H, Shang J, Yang F, Zhang S, Cui J, Hou X, Li Y, Liu W, Shu X, Liu Y, Sun B. Modulation of tumour hypoxia by ultrasound-responsive microbubbles to enhance the sono-photodynamic therapy effect on triple-negative breast cancer. Photodiagnosis Photodyn Ther 2023:103642. [PMID: 37271488 DOI: 10.1016/j.pdpdt.2023.103642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Sono-photodynamic therapy (SPDT) is an oxidative stress-dependent antitumour treatment modality. Due to the hypoxic tumour microenvironment, the antitumour effect of SPDT is limited. In this study, we developed lipid vesicles to transport a photosensitizer (chlorin e6, Ce6) and oxygen into tumours to promote SPDT efficiency on triple-negative breast cancer in vitro and in vivo. The results showed that compared with the same concentration of free Ce6, Lipo-Ce6 produced a higher singlet oxygen level under light irradiation. Cellular Lipo-Ce6 accumulation was 4-fold higher than that of free Ce6. The cytotoxicity on 4T1 cells caused by Lipo-Ce6-SPDT was significantly stronger than that caused by free Ce6-SPDT, and oxygen microbubbles (O2MB) further enhanced the cytotoxicity of Lipo-Ce6-SPDT under hypoxic conditions. Cellular ROS production in the Lipo-Ce6-SPDT+O2MB group was approximately 2.5-fold higher than that in the Lipo-Ce6-SPDT+C3F8MB group. Furthermore, O2MB rapidly relieved 4T1 subcutaneous xenograft hypoxia conditions under ultrasound exposure and significantly improved the antitumour activity of SPDT in vivo. These results indicate that the combination of O2MB and a high-activity liposome photosensitizer can significantly enhance the antitumour efficiency of SPDT for hypoxic tumours.
Collapse
Affiliation(s)
- Haiping Wang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China
| | - Jinting Shang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China; Huazhong University of Science and Technology; Advanced Technology Institute of Suzhou Chinese Academy of Science,Co.,Ltd
| | - Fang Yang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China
| | - Song Zhang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Jingsong Cui
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xiaoying Hou
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, Guangxi, China
| | - Wei Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xiji Shu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Yuchen Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China.
| | - Binlian Sun
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China.
| |
Collapse
|
219
|
Dong H, Li Q, Zhang Y, Ding M, Teng Z, Mou Y. Biomaterials Facilitating Dendritic Cell-Mediated Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301339. [PMID: 37088780 PMCID: PMC10288267 DOI: 10.1002/advs.202301339] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.
Collapse
Affiliation(s)
- Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
220
|
Li Q, Qin S, Tian H, Liu R, Qiao L, Liu S, Li B, Yang M, Shi J, Nice EC, Li J, Lang T, Huang C. Nano-Econazole Enhanced PD-L1 Checkpoint Blockade for Synergistic Antitumor Immunotherapy against Pancreatic Ductal Adenocarcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207201. [PMID: 36899444 DOI: 10.1002/smll.202207201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/04/2023] [Indexed: 06/08/2023]
Abstract
Insufficienct T lymphocyte infiltration and unresponsiveness to immune checkpoint blockade therapy are still major difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). Although econazole has shown promise in inhibiting PDAC growth, its poor bioavailability and water solubility limit its potential as a clinical therapy for PDAC. Furthermore, the synergistic role of econazole and biliverdin in immune checkpoint blockade therapy in PDAC remains elusive and challenging. Herein, a chemo-phototherapy nanoplatform is designed by which econazole and biliverdin can be co-assembled (defined as FBE NPs), which significantly improve the poor water solubility of econazole and enhance the efficacy of PD-L1 checkpoint blockade therapy against PDAC. Mechanistically, econazole and biliverdin are directly released into the acidic cancer microenvironment, to activate immunogenic cell death via biliverdin-induced PTT/PDT and boost the immunotherapeutic response of PD-L1 blockade. In addition, econazole simultaneously enhances PD-L1 expression to sensitize anti-PD-L1 therapy, leading to suppression of distant tumors, long-term immune memory effects, improved dendritic cell maturation, and tumor infiltration of CD8+ T lymphocytes. The combined FBE NPs and α-PDL1 show synergistic antitumor efficacy. Collectively, FBE NPs show excellent biosafety and antitumor efficacy by combining chemo-phototherapy with PD-L1 blockade, which has promising potential in a precision medicine approach as a PDAC treatment strategy.
Collapse
Affiliation(s)
- Qiong Li
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Siyuan Qin
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Hailong Tian
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Ruolan Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Ling Qiao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Shanshan Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, 563006, P. R. China
| | - Bowen Li
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Mei Yang
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Jiayan Shi
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Jingquan Li
- Department of Gastrointestinal Oncology Surgery, the First Affiliated Hospital of Hainan Medical University, Hainan Province, Haikou, 570216, P. R. China
| | - Tingyuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, Chongqing, 400030, P. R. China
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, 400042, P. R. China
| | - Canhua Huang
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, and West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| |
Collapse
|
221
|
Liu J, Li RS, Zhang L, Wang J, Dong Q, Xu Z, Kang Y, Xue P. Enzyme-Activatable Polypeptide for Plasma Membrane Disruption and Antitumor Immunity Elicitation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206912. [PMID: 36932931 DOI: 10.1002/smll.202206912] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/01/2023] [Indexed: 06/15/2023]
Abstract
Enzyme-instructed self-assembly of bioactive molecules into nanobundles inside cells is conceived to potentially disrupt plasma membrane and subcellular structure. Herein, an alkaline phosphatase (ALP)-activatable hybrid of ICG-CF4 KYp is facilely synthesized by conjugating photosensitizer indocyanine green (ICG) with CF4 KYp peptide via classical Michael addition reaction. ALP-induced dephosphorylation of ICG-CF4 KYp enables its transformation from small-molecule precursor into rigid nanofibrils, and such fibrillation in situ causes severe mechanical disruption of cytomembrane. Besides, ICG-mediated photosensitization causes additional oxidative damage of plasma membrane by lipid peroxidation. Hollow MnO2 nanospheres devote to deliver ICG-CF4 KYp into tumorous tissue through tumor-specific acidity/glutathione-triggered degradation of MnO2 , which is monitored by fluorescent probing and magnetic resonance imaging. The burst release of damage-associated molecular patterns and other tumor antigens during therapy effectively triggers immunogenetic cell death and improves immune stimulatory, as demonstrated by the promotion of dendritic cell maturation and CD8+ lymphocyte infiltration, as well as constraint of regulatory T cell population. Taken together, such cytomembrane injury strategy based on peptide fibrillation in situ holds high clinical promise for lesion-specific elimination of primary, abscopal, and metastatic tumors, which may enlighten more bioinspired nanoplatforms for anticancer theranostics.
Collapse
Affiliation(s)
- Jiahui Liu
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Rong Sheng Li
- National Demonstration Center for Experimental Chemistry and Chemical Engineering Education, School of Chemical Science and Engineering, Yunnan University, Kunming, 650091, China
| | - Lei Zhang
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Jie Wang
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Qi Dong
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Zhigang Xu
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Yuejun Kang
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Peng Xue
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| |
Collapse
|
222
|
Zhang Y, Dong P, Yang L. The role of nanotherapy in head and neck squamous cell carcinoma by targeting tumor microenvironment. Front Immunol 2023; 14:1189323. [PMID: 37292204 PMCID: PMC10244756 DOI: 10.3389/fimmu.2023.1189323] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) refers to a group of highly malignant and pathogenically complex tumors. Traditional treatment methods include surgery, radiotherapy, and chemotherapy. However, with advancements in genetics, molecular medicine, and nanotherapy, more effective and safer treatments have been developed. Nanotherapy, in particular, has the potential to be an alternative therapeutic option for HNSCC patients, given its advantageous targeting capabilities, low toxicity and modifiability. Recent research has highlighted the important role of the tumor microenvironment (TME) in the development of HNSCC. The TME is composed of various cellular components, such as fibroblasts, vascular endothelial cells, and immune cells, as well as non-cellular agents such as cytokines, chemokines, growth factors, extracellular matrix (ECM), and extracellular vesicles (EVs). These components greatly influence the prognosis and therapeutic efficacy of HNSCC, making the TME a potential target for treatment using nanotherapy. By regulating angiogenesis, immune response, tumor metastasis and other factors, nanotherapy can potentially alleviate HNSCC symptoms. This review aims to summarize and discuss the application of nanotherapy that targets HNSCC's TME. We highlight the therapeutic value of nanotherapy for HNSCC patients.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Radiation Oncology, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Pengbo Dong
- School of Energy and Power Engineering, Dalian University of Technology, Dalian, China
| | - Lu Yang
- Department of Internal Medicine, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
223
|
Wang D, Liu J, Wang C, Zhang W, Yang G, Chen Y, Zhang X, Wu Y, Gu L, Chen H, Yuan W, Chen X, Liu G, Gao B, Chen Q, Zhao Y. Microbial synthesis of Prussian blue for potentiating checkpoint blockade immunotherapy. Nat Commun 2023; 14:2943. [PMID: 37221237 DOI: 10.1038/s41467-023-38796-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 05/16/2023] [Indexed: 05/25/2023] Open
Abstract
Cancer immunotherapy is revolutionizing oncology. The marriage of nanotechnology and immunotherapy offers a great opportunity to amplify antitumor immune response in a safe and effective manner. Here, electrochemically active Shewanella oneidensis MR-1 can be applied to produce FDA-approved Prussian blue nanoparticles on a large-scale. We present a mitochondria-targeting nanoplatform, MiBaMc, which consists of Prussian blue decorated bacteria membrane fragments having further modifications with chlorin e6 and triphenylphosphine. We find that MiBaMc specifically targets mitochondria and induces amplified photo-damages and immunogenic cell death of tumor cells under light irradiation. The released tumor antigens subsequently promote the maturation of dendritic cells in tumor-draining lymph nodes, eliciting T cell-mediated immune response. In two tumor-bearing mouse models using female mice, MiBaMc triggered phototherapy synergizes with anti-PDL1 blocking antibody for enhanced tumor inhibition. Collectively, the present study demonstrates biological precipitation synthetic strategy of targeted nanoparticles holds great potential for the preparation of microbial membrane-based nanoplatforms to boost antitumor immunity.
Collapse
Affiliation(s)
- Dongdong Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, 230026, Hefei, P.R. China
| | - Jiawei Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
- The Institute of Geology and Geophysics, Chinese Academy of Sciences, 100029, Beijing, P.R. China
| | - Changlai Wang
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, 230026, Hefei, P.R. China
| | - Weiyun Zhang
- School of Biomedical Engineering, Shenzhen University, 518060, Shenzhen, P.R. China
| | - Guangbao Yang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Yun Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Xiaodong Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Yinglong Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Long Gu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Hongzhong Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Wei Yuan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Xiaokai Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Guofeng Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Bin Gao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Qianwang Chen
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, 230026, Hefei, P.R. China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore.
| |
Collapse
|
224
|
Xiao M, Tang Q, Zeng S, Yang Q, Yang X, Tong X, Zhu G, Lei L, Li S. Emerging biomaterials for tumor immunotherapy. Biomater Res 2023; 27:47. [PMID: 37194085 PMCID: PMC10189985 DOI: 10.1186/s40824-023-00369-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The immune system interacts with cancer cells in various intricate ways that can protect the individual from overproliferation of cancer cells; however, these interactions can also lead to malignancy. There has been a dramatic increase in the application of cancer immunotherapy in the last decade. However, low immunogenicity, poor specificity, weak presentation efficiency, and off-target side effects still limit its widespread application. Fortunately, advanced biomaterials effectively contribute immunotherapy and play an important role in cancer treatment, making it a research hotspot in the biomedical field. MAIN BODY This review discusses immunotherapies and the development of related biomaterials for application in the field. The review first summarizes the various types of tumor immunotherapy applicable in clinical practice as well as their underlying mechanisms. Further, it focuses on the types of biomaterials applied in immunotherapy and related research on metal nanomaterials, silicon nanoparticles, carbon nanotubes, polymer nanoparticles, and cell membrane nanocarriers. Moreover, we introduce the preparation and processing technologies of these biomaterials (liposomes, microspheres, microneedles, and hydrogels) and summarize their mechanisms when applied to tumor immunotherapy. Finally, we discuss future advancements and shortcomings related to the application of biomaterials in tumor immunotherapy. CONCLUSION Research on biomaterial-based tumor immunotherapy is booming; however, several challenges remain to be overcome to transition from experimental research to clinical application. Biomaterials have been optimized continuously and nanotechnology has achieved continuous progression, ensuring the development of more efficient biomaterials, thereby providing a platform and opportunity for breakthroughs in tumor immunotherapy.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
225
|
Sisakhtnezhad S, Rahimi M, Mohammadi S. Biomedical applications of MnO 2 nanomaterials as nanozyme-based theranostics. Biomed Pharmacother 2023; 163:114833. [PMID: 37150035 DOI: 10.1016/j.biopha.2023.114833] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/30/2023] [Accepted: 05/01/2023] [Indexed: 05/09/2023] Open
Abstract
Manganese dioxide (MnO2) nanoenzymes/nanozymes (MnO2-NEs) are 1-100 nm nanomaterials that mimic catalytic, oxidative, peroxidase, and superoxide dismutase activities. The oxidative-like activity of MnO2-NEs makes them suitable for developing effective and low-cost colorimetric detection assays of biomolecules. Interestingly, MnO2-NEs also demonstrate scavenging properties against reactive oxygen species (ROS) in various pathological conditions. In addition, due to the decomposition of MnO2-NEs in the tumor microenvironment (TME) and the production of Mn2+, they can act as a contrast agent for improving clinical imaging diagnostics. MnO2-NEs also can use as an in situ oxygen production system in TME, thereby overcoming hypoxic conditions and their consequences in the progression of cancer. Furthermore, MnO2-NEs as a shell and coating make the nanosystems smart and, therefore, in combination with other nanomaterials, the MnO2-NEs can be used as an intelligent nanocarrier for delivering drugs, photosensitizers, and sonosensitizers in vivo. Moreover, these capabilities make MnO2-NEs a promising candidate for the detection and treatment of different human diseases such as cancer, metabolic, infectious, and inflammatory pathological conditions. MnO2-NEs also have ROS-scavenging and anti-bacterial properties against Gram-positive and Gram-negative bacterial strains, which make them suitable for wound healing applications. Given the importance of nanomaterials and their potential applications in biomedicine, this review aimed to discuss the biochemical properties and the theranostic roles of MnO2-NEs and recent advances in their use in colorimetric detection assays of biomolecules, diagnostic imaging, drug delivery, and combinatorial therapy applications. Finally, the challenges of MnO2-NEs applications in biomedicine will be discussed.
Collapse
Affiliation(s)
| | - Matin Rahimi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Soheila Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
226
|
Zhang K, Qi C, Cai K. Manganese-Based Tumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2205409. [PMID: 36121368 DOI: 10.1002/adma.202205409] [Citation(s) in RCA: 116] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/26/2022] [Indexed: 05/12/2023]
Abstract
As an essential micronutrient, manganese (Mn) participates in various physiological processes and plays important roles in host immune system, hematopoiesis, endocrine function, and oxidative stress regulation. Mn-based nanoparticles are considered to be biocompatible and show versatile applications in nanomedicine, in particular utilized in tumor immunotherapy in the following ways: 1) acting as a biocompatible nanocarrier to deliver immunotherapeutic agents for tumor immunotherapy; 2) serving as an adjuvant to regulate tumor immune microenvironment and enhance immunotherapy; 3) activating host's immune system through the cGAS-STING pathway to trigger tumor immunotherapy; 4) real-time monitoring tumor immunotherapy effect by magnetic resonance imaging (MRI) since Mn2+ ions are ideal MRI contrast agent which can significantly enhance the T1 -weighted MRI signal after binding to proteins. This comprehensive review focuses on the most recent progress of Mn-based nanoplatforms in tumor immunotherapy. The characteristics of Mn are first discussed to guide the design of Mn-based multifunctional nanoplatforms. Then the biomedical applications of Mn-based nanoplatforms, including immunotherapy alone, immunotherapy-involved multimodal synergistic therapy, and imaging-guided immunotherapy are discussed in detail. Finally, the challenges and future developments of Mn-based tumor immunotherapy are highlighted.
Collapse
Affiliation(s)
- Ke Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chao Qi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
227
|
Yue L, Gao C, Li J, Chen H, Lee SMY, Luo R, Wang R. Chemotaxis-guided Self-propelled Macrophage Motor for Targeted Treatment of Acute Pneumonia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211626. [PMID: 36905923 DOI: 10.1002/adma.202211626] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/07/2023] [Indexed: 05/19/2023]
Abstract
Immune cells exhibit great potential as carriers of nanomedicine, attributed to their high tolerance to internalized nanomaterials and targeted accumulation in inflammatory tissues. However, the premature efflux of internalized nanomedicine during systemic delivery and slow infiltration into inflammatory tissues have limited their translational applications. Herein, a motorized cell platform as a nanomedicine carrier for highly efficient accumulation and infiltration in the inflammatory lungs and effective treatment of acute pneumonia are reported. β-Cyclodextrin and adamantane respectively modified manganese dioxide nanoparticles are intracellularly self-assembled into large aggregates mediated via host-guest interactions, to effectively inhibit the efflux of nanoparticles, catalytically consume/deplete H2 O2 to alleviate inflammation, and generate O2 to propel macrophage movement for rapid tissue infiltration. With curcumin loaded into MnO2 nanoparticles, macrophages carry the intracellular nano-assemblies rapidly into the inflammatory lungs via chemotaxis-guided, self-propelled movement, for effective treatment of acute pneumonia via immunoregulation induced by curcumin and the aggregates.
Collapse
Affiliation(s)
- Ludan Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Junyan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Hanbin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Simon M Y Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Ruifeng Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| |
Collapse
|
228
|
Li J, Wang S, Fontana F, Tapeinos C, Shahbazi MA, Han H, Santos HA. Nanoparticles-based phototherapy systems for cancer treatment: Current status and clinical potential. Bioact Mater 2023; 23:471-507. [PMID: 36514388 PMCID: PMC9727595 DOI: 10.1016/j.bioactmat.2022.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 12/11/2022] Open
Abstract
Remarkable progress in phototherapy has been made in recent decades, due to its non-invasiveness and instant therapeutic efficacy. In addition, with the rapid development of nanoscience and nanotechnology, phototherapy systems based on nanoparticles or nanocomposites also evolved as an emerging hotspot in nanomedicine research, especially in cancer. In this review, first we briefly introduce the history of phototherapy, and the mechanisms of phototherapy in cancer treatment. Then, we summarize the representative development over the past three to five years in nanoparticle-based phototherapy and highlight the design of the innovative nanoparticles thereof. Finally, we discuss the feasibility and the potential of the nanoparticle-based phototherapy systems in clinical anticancer therapeutic applications, aiming to predict future research directions in this field. Our review is a tutorial work, aiming at providing useful insights to researchers in the field of nanotechnology, nanoscience and cancer.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Shiqi Wang
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Flavia Fontana
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Christos Tapeinos
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Huijie Han
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Hélder A Santos
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
229
|
Chen W, Li C, Jiang X. Advanced Biomaterials with Intrinsic Immunomodulation Effects for Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201404. [PMID: 36811240 DOI: 10.1002/smtd.202201404] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/17/2023] [Indexed: 05/17/2023]
Abstract
In recent years, tumor immunotherapy has achieved significant success in tumor treatment based on immune checkpoint blockers and chimeric antigen receptor T-cell therapy. However, about 70-80% of patients with solid tumors do not respond to immunotherapy due to immune evasion. Recent studies found that some biomaterials have intrinsic immunoregulatory effects, except serve as carriers for immunoregulatory drugs. Moreover, these biomaterials have additional advantages such as easy functionalization, modification, and customization. In this review, the recent advances of these immunoregulatory biomaterials in cancer immunotherapy and their interaction with cancer cells, immune cells, and the immunosuppressive tumor microenvironment are summarized. Finally, the opportunities and challenges of immunoregulatory biomaterials used in the clinic and the prospect of their future in cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210023, P. R. China
| | - Cheng Li
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210023, P. R. China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
230
|
Zhao X, Zhang J, Chen B, Ding X, Zhao N, Xu FJ. Rough Nanovaccines Boost Antitumor Immunity Through the Enhancement of Vaccination Cascade and Immunogenic Cell Death Induction. SMALL METHODS 2023; 7:e2201595. [PMID: 36869418 DOI: 10.1002/smtd.202201595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/13/2023] [Indexed: 05/17/2023]
Abstract
Nanovaccines have attracted intense interests for efficient antigen delivery and tumor-specific immunity. It is challenging to develop a more efficient and personalized nanovaccine to maximize all steps of the vaccination cascade by exploiting the intrinsic properties of nanoparticles. Here, biodegradable nanohybrids (MP) composed of manganese oxide nanoparticles and cationic polymers are synthesized to load a model antigen ovalbumin to form MPO nanovaccines. More interestingly, MPO could serve as autologous nanovaccines for personalized tumor treatment taking advantage of in situ released tumor-associated antigens induced by immunogenic cell death (ICD). The intrinsic properties of MP nanohybrids including morphology, size, surface charge, chemical, and immunoregulatory functions are fully exploited to enhance of all steps of the cascade and induce ICD. MP nanohybrids are designed to efficiently encapsulate antigens by cationic polymers, drain to lymph nodes by appropriate size, be internalized by dendritic cells (DCs) by rough morphology, induce DC maturation through cGAS-STING pathway, and enhance lysosomal escape and antigen cross-presentation through the "proton sponge effect". The MPO nanovaccines are found to efficiently accumulate in lymph nodes and elicit robust specific T-cell immune responses to inhibit the occurrence of ovalbumin-expressing B16-OVA melanoma. Furthermore, MPO demonstrate great potential to serve as personalized cancer vaccines through the generation of autologous antigen depot through ICD induction, activation of potent antitumor immunity, and reversal of immunosuppression. This work provides a facile strategy for the construction of personalized nanovaccines by exploiting the intrinsic properties of nanohybrids.
Collapse
Affiliation(s)
- Xiaoyi Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jiatong Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Beibei Chen
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaokang Ding
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
231
|
Shao M, Wang Y, Dong H, Wang L, Zhang X, Han X, Sang X, Bao Y, Peng M, Cao G. From liver fibrosis to hepatocarcinogenesis: Role of excessive liver H2O2 and targeting nanotherapeutics. Bioact Mater 2023; 23:187-205. [PMID: 36406254 PMCID: PMC9663332 DOI: 10.1016/j.bioactmat.2022.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/23/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022] Open
Abstract
Liver fibrosis and hepatocellular carcinoma (HCC) have been worldwide threats nowadays. Liver fibrosis is reversible in early stages but will develop precancerosis of HCC in cirrhotic stage. In pathological liver, excessive H2O2 is generated and accumulated, which impacts the functionality of hepatocytes, Kupffer cells (KCs) and hepatic stellate cells (HSCs), leading to genesis of fibrosis and HCC. H2O2 accumulation is associated with overproduction of superoxide anion (O2•−) and abolished antioxidant enzyme systems. Plenty of therapeutics focused on H2O2 have shown satisfactory effects against liver fibrosis or HCC in different ways. This review summarized the reasons of liver H2O2 accumulation, and the role of H2O2 in genesis of liver fibrosis and HCC. Additionally, nanotherapeutics targeting H2O2 were summarized for further consideration of antifibrotic or antitumor therapy. Liver fibrosis and HCC are closely related because ROS induced liver damage and inflammation, especially over-cumulated H2O2. Excess H2O2 diffusion in pathological liver was due to increased metabolic rate and diminished cellular antioxidant systems. Freely diffused H2O2 damaged liver-specific cells, thereby leading to fibrogenesis and hepatocarcinogenesis. Nanotherapeutics targeting H2O2 are summarized for treatment of liver fibrosis and HCC, and also challenges are proposed.
Collapse
|
232
|
Meng Y, Huang J, Ding J, Yan B, Li Y, Gao X, Zhou W. Poly-thymine DNA templated MnO 2 biomineralization as a high-affinity anchoring enabling tumor targeting delivery. J Colloid Interface Sci 2023; 637:441-452. [PMID: 36716668 DOI: 10.1016/j.jcis.2023.01.089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Manganese oxide nanomaterials (MONs) are emerging as a type of highly promising nanomaterials for diseases diagnosis, and surface modification is the basis for colloidal stability and targeting delivery of the nanomaterials. Here, we report the in-situ functionalization of MnO2 with DNA through a biomineralization process. Using adsorption-oxidation method, DNA templated Mn2+ precursor to biomineralize into nano-cubic seed, followed by the growth of MnO2 to form cube/nanosheet hybrid nanostructure. Among four types of DNA homopolymers, poly-thymine (poly-T) was found to stably attach on MnO2 surface to resist various biological displacements (phosphate, serum, and complementary DNA). Capitalized on this finding, a di-block DNA was rationally designed, in which the poly-T block stably anchored on MnO2 surface, while the AS1411 aptamer block was not only an active ligand for tumor targeting delivery, but also a carrier for photosensitizer (Ce6) loading. Upon targeting delivery into tumor cells, the MnO2 acted as catalase-mimic nanozyme for oxygenation to sensitize photodynamic therapy, and the released Mn2+ triggered chemodynamic therapy via Fenton-like reaction, achieving synergistic anti-tumor effect with full biocompatibility. This work provides a simple yet robust strategy to functionalize metal oxides nanomaterials for biological applications via DNA-templated biomineralization.
Collapse
Affiliation(s)
- Yingcai Meng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jiaxin Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Bohua Yan
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha 410004, Hunan, China.
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China.
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
233
|
Perez-Potti A, Rodríguez-Pérez M, Polo E, Pelaz B, Del Pino P. Nanoparticle-based immunotherapeutics: from the properties of nanocores to the differential effects of administration routes. Adv Drug Deliv Rev 2023; 197:114829. [PMID: 37121275 DOI: 10.1016/j.addr.2023.114829] [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: 02/22/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
The engagement with the immune system is one of the main cornerstones in the development of nanotechnologies for therapy and diagnostics. Recent advances have made possible the tuning of features like size, shape and biomolecular modifications that influence such interactions, however, the capabilities for immune modulation of nanoparticles are still not well defined and exploited. This review focuses on recent advances made in preclinical research for the application of nanoparticles to modulate immune responses, and the main features making them relevant for such applications. We review and discuss newest evidence in the field, which include in vivo experiments with an extensive physicochemical characterization as well as detailed study of the induced immune response. We emphasize the need of incorporating knowledge about immune response development and regulation in the design and application of nanoparticles, including the effect by parameters such as the administration route and the differential interactions with immune subsets.
Collapse
Affiliation(s)
- André Perez-Potti
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Manuel Rodríguez-Pérez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
234
|
Naletova I, Tomasello B, Attanasio F, Pleshkan VV. Prospects for the Use of Metal-Based Nanoparticles as Adjuvants for Local Cancer Immunotherapy. Pharmaceutics 2023; 15:1346. [PMID: 37242588 PMCID: PMC10222518 DOI: 10.3390/pharmaceutics15051346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Immunotherapy is among the most effective approaches for treating cancer. One of the key aspects for successful immunotherapy is to achieve a strong and stable antitumor immune response. Modern immune checkpoint therapy demonstrates that cancer can be defeated. However, it also points out the weaknesses of immunotherapy, as not all tumors respond to therapy and the co-administration of different immunomodulators may be severely limited due to their systemic toxicity. Nevertheless, there is an established way through which to increase the immunogenicity of immunotherapy-by the use of adjuvants. These enhance the immune response without inducing such severe adverse effects. One of the most well-known and studied adjuvant strategies to improve immunotherapy efficacy is the use of metal-based compounds, in more modern implementation-metal-based nanoparticles (MNPs), which are exogenous agents that act as danger signals. Adding innate immune activation to the main action of an immunomodulator makes it capable of eliciting a robust anti-cancer immune response. The use of an adjuvant has the peculiarity of a local administration of the drug, which positively affects its safety. In this review, we will consider the use of MNPs as low-toxicity adjuvants for cancer immunotherapy, which could provide an abscopal effect when administered locally.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, 95125 Catania, Italy
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| |
Collapse
|
235
|
Wang M, Liang Y, Jiang X, Shen J. α-Fe 2O 3@Au-PEG-Ce6-Gd Nanoparticles as Acidic H 2O 2-Driven Oxygenators for Multimodal Imaging and Synergistic Tumor Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:5333-5341. [PMID: 37018043 DOI: 10.1021/acs.langmuir.2c03388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Nanoparticles with visual imaging capabilities and synergistic therapeutics have a bright future in antitumor applications. However, most of the current nanomaterials lack multiple imaging-guided therapeutic capabilities. In this study, a novel enhanced photothermal photodynamic antitumor nanoplatform with photothermal imaging, fluorescence (FL) imaging, and MRI-guided therapeutic capabilities was constructed by grafting gold, dihydroporphyrin Ce6, and Gd onto α-iron trioxide. This antitumor nanoplatform can convert NIR light into local hyperthermia at a temperature of up to 53 °C under NIR light irradiation, while Ce6 can generate singlet oxygen, which further synergizes the tumor-killing effect. At the same time, α-Fe2O3@Au-PEG-Ce6-Gd can also have significant photothermal imaging effect under light irradiation, which can guide to see the temperature change near the tumor tissue. It is worth noting that α-Fe2O3@Au-PEG-Ce6-Gd can have obvious MRI and FL imaging effects after tail vein injection in mice with blood circulation, realizing imaging-guided synergistic antitumor therapy. α-Fe2O3@Au-PEG-Ce6-Gd NPs provide a new solution for tumor imaging and treatment.
Collapse
Affiliation(s)
- Mingqian Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
| | - Ying Liang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
| | - Xuefeng Jiang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
- Jiangsu Engineering Research Center of Interfacial Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China
| |
Collapse
|
236
|
Li J, Gong C, Chen X, Guo H, Tai Z, Ding N, Gao S, Gao Y. Biomimetic liposomal nanozymes improve breast cancer chemotherapy with enhanced penetration and alleviated hypoxia. J Nanobiotechnology 2023; 21:123. [PMID: 37038165 PMCID: PMC10084658 DOI: 10.1186/s12951-023-01874-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/26/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Doxorubicin (Dox) has been recommended in clinical guidelines for the standard-of-care treatment of breast cancer. However, Dox therapy faces challenges such as hypoxia, acidosis, H2O2-rich conditions and condensed extracellular matrix in TME as well as low targeted ability. METHODS We developed a nanosystem H-MnO2-Dox-Col NPs based on mesoporous manganese dioxide (H-MnO2) in which Dox was loaded in the core and collagenase (Col) was wrapped in the surface. Further the H-MnO2-Dox-Col NPs were covered by a fusion membrane (MP) of inflammation-targeted RAW264.7 cell membrane and pH-sensitive liposomes to form biomimetic MP@H-MnO2-Dox-Col for in vitro and in vivo study. RESULTS Our results shows that MP@H-MnO2-Dox-Col can increase the Dox effect with low cardiotoxicity based on multi-functions of effective penetration in tumor tissue, alleviating hypoxia in TME, pH sensitive drug release as well as targeted delivery of Dox. CONCLUSIONS This multifunctional biomimetic nanodelivery system exhibited antitumor efficacy in vivo and in vitro, thus having potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Juanjuan Li
- School of Pharmacy & Zhong Shan Hospital, Fudan University, Shanghai, 201206, China
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, P. R. China
| | - Xinlu Chen
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Huanhuan Guo
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| | - Nan Ding
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shen Gao
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Yuan Gao
- School of Pharmacy & Zhong Shan Hospital, Fudan University, Shanghai, 201206, China.
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
237
|
Jia Y, Sun J, Yang J, Chen C, Zhang Z, Yang K, Shen P, Qu S, He B, Song Y, Han X. Tumor Microenvironment-Responsive Nanoherb Delivery System for Synergistically Inhibition of Cancer Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16329-16342. [PMID: 36946515 DOI: 10.1021/acsami.2c19029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Multidrug resistance in cancer stem cells (CSCs) is a major barrier to chemotherapy; hence, developing CSC-specific targeted nanocarriers for efficient drug delivery is critical. In this study, monodisperse hollow-structured MnO2 (H-MnO2) with a mesoporous shell was created for efficient targeted drug delivery. An effective therapeutic compound isoliquiritigenin (ISL) was confirmed to inhibit the lung cancer stem-cell phenotype by natural compound screening based on integrated microfluidic devices. The resultant H-MnO2 showed a high drug-loading content of the potent CSC-targeting compound ISL and near-infrared fluorescent dye indocyanine green (ICG). In addition, H-MnO2 was successively modified with hyaluronic acid (HA) to enhance targeting CSCs with high CD44 expression levels. The H-MnO2@(ICG + ISL)@HA nanocomposites displayed promising chemotherapeutic and photothermal treatment capabilities, as well as NIR-triggered drug release, which showed excellent CSC-killing effects and tumor inhibition efficacy. Meanwhile, the development of the tumor was effectively restrained by NIR-triggered phototherapy and prominent chemotherapy without obvious side effects after tail vein injection of the nanocomposites in vivo. In summary, the prepared nanocomposites accomplished synergistic cancer therapy that targets CSCs, offering a versatile platform for lung cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yuanyuan Jia
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia Sun
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jingjing Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chao Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhenyu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Kaiyong Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Peiliang Shen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Suchen Qu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yanni Song
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China
| | - Xin Han
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
238
|
Ge YX, Zhuang HJ, Zhang TW, Liang HF, Ding W, Zhou L, Dong ZR, Hu ZC, Chen Q, Dong J, Jiang LB, Yin XF. Precise manipulation of circadian clock using MnO 2 nanocapsules to amplify photodynamic therapy for osteosarcoma. Mater Today Bio 2023; 19:100547. [PMID: 36896415 PMCID: PMC9988696 DOI: 10.1016/j.mtbio.2023.100547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
Circadian rhythm (CR) disruption contributes to tumor initiation and progression, however the pharmacological targeting of circadian regulators reversely inhibits tumor growth. Precisely controlling CR in tumor cells is urgently required to investigate the exact role of CR interruption in tumor therapy. Herein, based on KL001, a small molecule that specifically interacts with the clock gene cryptochrome (CRY) functioning at disruption of CR, we fabricated a hollow MnO2 nanocapsule carrying KL001 and photosensitizer BODIPY with the modification of alendronate (ALD) on the surface (H-MnSiO/K&B-ALD) for osteosarcoma (OS) targeting. The H-MnSiO/K&B-ALD nanoparticles reduced the CR amplitude in OS cells without affecting cell proliferation. Furthermore, nanoparticles-controlled oxygen consumption by inhibiting mitochondrial respiration via CR disruption, thus partially overcoming the hypoxia limitation for photodynamic therapy (PDT) and significantly promoting PDT efficacy. An orthotopic OS model demonstrated that KL001 significantly enhanced the inhibitory effect of H-MnSiO/K&B-ALD nanoparticles on tumor growth after laser irradiation. CR disruption and oxygen level enhancement induced by H-MnSiO/K&B-ALD nanoparticles under laser irradiation were also confirmed in vivo. This discovery first demonstrated the potential of CR controlling for tumor PDT ablation and provided a promising strategy for overcoming tumor hypoxia.
Collapse
Affiliation(s)
- Yu-Xiang Ge
- Department of Orthopedics Surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hong-Jun Zhuang
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Tai-Wei Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wang Ding
- Department of Orthopedics Surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Lei Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhi-rui Dong
- Department of Orthopedics Surgery, Jinshan Hospital, Fudan University, Shanghai 201512, China
| | - Zhi-Chao Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qing Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li-Bo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiao-Fan Yin
- Department of Orthopedics Surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| |
Collapse
|
239
|
Zhang Y, Wang R, Liu R, Xie S, Jiao F, Li Y, Xin J, Zhang H, Wang Z, Yan Y. Delivery of miR-3529-3p using MnO 2 -SiO 2 -APTES nanoparticles combined with phototherapy suppresses lung adenocarcinoma progression by targeting HIGD1A. Thorac Cancer 2023; 14:913-928. [PMID: 36808485 PMCID: PMC10067359 DOI: 10.1111/1759-7714.14823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND The present study aimed to investigate the function of miR-3529-3p in lung adenocarcinoma and MnO2 -SiO2 -APTES (MSA) as a promising multifunctional delivery agent for lung adenocarcinoma therapy. METHODS Expression levels of miR-3529-3p were evaluated in lung carcinoma cells and tissues by qRT-PCR. The effects of miR-3529-3p on apoptosis, proliferation, metastasis and neovascularization were assessed by CCK-8, FACS, transwell and wound healing assays, tube formation and xenografts experiments. Luciferase reporter assays, western blot, qRT-PCR and mitochondrial complex assay were used to determine the targeting relationship between miR-3529-3p and hypoxia-inducible gene domain family member 1A (HIGD1A). MSA was fabricated using MnO2 nanoflowers, and its heating curves, temperature curves, IC50, and delivery efficiency were examined. The hypoxia and reactive oxygen species (ROS) production was investigated by nitro reductase probing, DCFH-DA staining and FACS. RESULTS MiR-3529-3p expression was reduced in lung carcinoma tissues and cells. Transfection of miR-3529-3p could promote apoptosis and suppress cell proliferation, migration and angiogenesis. As a target of miR-3529-3p, HIGD1A expression was downregulated, through which miR-3529-3p could disrupt the activities of complexes III and IV of the respiratory chain. The multifunctional nanoparticle MSA could not only efficiently deliver miR-3529-3p into cells, but also enhance the antitumor function of miR-3529-3p. The underlying mechanism may be that MSA alleviates hypoxia and has synergistic effects in cellular ROS promotion with miR-3529-3p. CONCLUSIONS Our results establish the antioncogenic role of miR-3529-3p, and demonstrate that miR-3529-3p delivered by MSA has enhanced tumor suppressive effects, probably through elevating ROS production and thermogenesis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
- Oncology DepartmentBinzhou Medical University HospitalBinzhouP. R. China
| | - Ran‐Ran Wang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Rui Liu
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Shu‐Yang Xie
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Fei Jiao
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - You‐Jie Li
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Jiaxuan Xin
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Han Zhang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Zhenbo Wang
- Oncology DepartmentBinzhou Medical University HospitalBinzhouP. R. China
| | - Yun‐Fei Yan
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| |
Collapse
|
240
|
Hosseini S, Mohammadnejad J, Salamat S, Beiram Zadeh Z, Tanhaei M, Ramakrishna S. Theranostic polymeric nanoparticles as a new approach in cancer therapy and diagnosis: a review. MATERIALS TODAY CHEMISTRY 2023; 29:101400. [DOI: 10.1016/j.mtchem.2023.101400] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
241
|
Fan H, Guo Z. Tumor microenvironment-responsive manganese-based nanomaterials for cancer treatment. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
242
|
Zhang HL, Wang Y, Tang Q, Ren B, Yang SP, Liu JG. A mesoporous MnO 2-based nanoplatform with near infrared light-controlled nitric oxide delivery and tumor microenvironment modulation for enhanced antitumor therapy. J Inorg Biochem 2023; 241:112133. [PMID: 36708626 DOI: 10.1016/j.jinorgbio.2023.112133] [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: 12/11/2022] [Revised: 01/06/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
A hollow mesoporous manganese dioxide-based (H-MnO2) multifunctional nanoplatform, H-MnO2 @AFIPB@PDA@Ru-NO@FA (MAPRF NPs), was prepared for synergistic cancer treatment, in which a histone deacetylase inhibitor AFIPB was loaded in its hollow cavity and a ruthenium nitrosyl donor (Ru-NO) and a folic acid (FA) targeting group were covalently decorated on its covered polydopamine (PDA) layer. The MAPRF NPs showed tumor microenvironment (TME)-responsive properties of depletion of glutathione (GSH) to disrupt the antioxidant defense system and on-demand drug delivery. And the released Mn2+ further catalyzed the decomposition of endogenous H2O2 to produce highly toxic hydroxyl radicals (·OH) for enhanced chemodynamic therapy (CDT). Furthermore, upon 808 nm light irradiation MAPRF NPs exhibited controlled nitric oxide (NO) delivery and simultaneously produced significant photothermal effect. Consequently, MAPRF NPs showed high mortality toward cancer cells in the presence of 808 nm light irradiation. This work provides a paradigm of multimodal synergistic therapy that combines NO-based gas therapy with TME modulation for efficient antitumor therapy.
Collapse
Affiliation(s)
- Hai-Lin Zhang
- Key Laboratory for Advanced Materials, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yi Wang
- Key Laboratory for Advanced Materials, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Qi Tang
- Key Laboratory for Advanced Materials, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Bing Ren
- Key Laboratory for Advanced Materials, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Shi-Ping Yang
- Key Lab of Resource Chemistry of MOE & Shanghai Key Lab of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, P. R. China
| | - Jin-Gang Liu
- Key Laboratory for Advanced Materials, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| |
Collapse
|
243
|
Kim H, Yoon J, Kim HK, Lee WT, Nguyen NT, Le XT, Lee EH, Lee ES, Oh KT, Choi HG, Youn YS. Upconverting nanoparticle-containing erythrocyte-sized hemoglobin microgels that generate heat, oxygen and reactive oxygen species for suppressing hypoxic tumors. Bioact Mater 2023; 22:112-126. [PMID: 36203958 PMCID: PMC9526021 DOI: 10.1016/j.bioactmat.2022.09.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
Inspired by erythrocytes that contain oxygen-carrying hemoglobin (Hb) and that exhibit photo-driven activity, we introduce homogenous-sized erythrocyte-like Hb microgel (μGel) systems (5–6 μm) that can (i) emit heat, (ii) supply oxygen, and (iii) generate reactive oxygen species (ROS; 1O2) in response to near-infrared (NIR) laser irradiation. Hb μGels consist of Hb, bovine serum albumin (BSA), chlorin e6 (Ce6) and erbium@lutetium upconverting nanoparticles (UCNPs; ∼35 nm) that effectively convert 808 nm NIR light to 660 nm visible light. These Hb μGels are capable of releasing oxygen to help generate sufficient reactive oxygen species (1O2) from UCNPs/Ce6 under severely hypoxic condition upon NIR stimulation for efficient photodynamic activity. Moreover, the Hb μGels emit heat and increase surface temperature due to NIR light absorption by heme (iron protoporphyrin IX) and display photothermal activity. By changing the Hb/UCNP/Ce6 ratio and controlling the amount of NIR laser irradiation, it is possible to formulate bespoke Hb μGels with either photothermal or photodynamic activity or both in the context of combined therapeutic effect. These Hb μGels effectively suppress highly hypoxic 4T1 cell spheroid growth and xenograft mice tumors in vivo. Erythrocyte-like hemoglobin μGels are prepared with upconverting nanoparticles. The μGels respond to the 808 nm near-infrared laser irradiation. The μGels emit heat, supply oxygen, and generate reactive oxygen species. The μGels have combined photothermal and photodynamic activity. The μGels suppress the growth of severe hypoxic 4T1 xenograft tumors.
Collapse
|
244
|
He M, Wang M, Xu T, Zhang M, Dai H, Wang C, Ding D, Zhong Z. Reactive oxygen species-powered cancer immunotherapy: Current status and challenges. J Control Release 2023; 356:623-648. [PMID: 36868519 DOI: 10.1016/j.jconrel.2023.02.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Reactive oxygen species (ROS) are crucial signaling molecules that can arouse immune system. In recent decades, ROS has emerged as a unique therapeutic strategy for malignant tumors as (i) it can not only directly reduce tumor burden but also trigger immune responses by inducing immunogenic cell death (ICD); and (ii) it can be facilely generated and modulated by radiotherapy, photodynamic therapy, sonodynamic therapy and chemodynamic therapy. The anti-tumor immune responses are, however, mostly downplayed by the immunosuppressive signals and dysfunction of effector immune cells within the tumor microenvironment (TME). The past years have seen fierce developments of various strategies to power ROS-based cancer immunotherapy by e.g. combining with immune checkpoints inhibitors, tumor vaccines, and/or immunoadjuvants, which have shown to potently inhibit primary tumors, metastatic tumors, and tumor relapse with limited immune-related adverse events (irAEs). In this review, we introduce the concept of ROS-powered cancer immunotherapy, highlight the innovative strategies to boost ROS-based cancer immunotherapy, and discuss the challenges in terms of clinical translation and future perspectives.
Collapse
Affiliation(s)
- Mengying He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengyuan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), Dublin D02 NY74, Ireland
| | - Mengyao Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Dawei Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| | - Zhiyuan Zhong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
245
|
Duan J, Liao T, Xu X, Liu Y, Kuang Y, Li C. Metal-polyphenol nanodots loaded hollow MnO 2 nanoparticles with a "dynamic protection" property for enhanced cancer chemodynamic therapy. J Colloid Interface Sci 2023; 634:836-851. [PMID: 36565625 DOI: 10.1016/j.jcis.2022.12.088] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/15/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Chemodynamic therapy (CDT) is a novel cancer therapeutic strategy. However, barriers such as high glutathione (GSH) concentration and low concentration of metal ions intracellular reduce its treatment effect. In this work, a nanosystem named GA-Fe@HMDN-PEI-PEG with a "dynamic protection" property was reported for enhanced cancer CDT. Mesoporous hollow manganese dioxide (MnO2) nanoparticle (HMDN) was prepared to load gallic acid-ferrous (GA-Fe) nanodots fabricated from gallic acid (GA) and ferrous ion (Fe2+). Then the pores of HMDN were blocked by polyethyleneimine (PEI), which was then grafted with methoxy poly(ethylene glycol) (mPEG) through a pH-sensitive benzoic imine bond. mPEG could protect the nanoparticles (NPs) against the nonspecific uptake by normal cells and enhance their accumulation in the tumor. However, in the slightly acidic tumor microenvironment, hydrolysis of benzoic imine led to DePEGylation to reveal PEI for enhanced uptake by cancer cells. The reaction between HMDN and GSH could consume GSH and obtain manganese ion (Mn2+) for the Fenton-like reaction for CDT. GA-Fe nanodots could also offer Fe for the Fenton reaction, and reductive GA could reduce the high-valence ions to low-valence for reusing in Fenton and Fenton-like reactions. These properties allowed GA-Fe@HMDN-PEI-PEG for precise medicine with a high utilization rate and common side effects.
Collapse
Affiliation(s)
- Junlin Duan
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China
| | - Tao Liao
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China
| | - Xiangyu Xu
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China
| | - Yun Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, 524023 Zhanjiang, China
| | - Ying Kuang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China.
| | - Cao Li
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; School of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| |
Collapse
|
246
|
Bag N, Bardhan S, Roy S, Roy J, Mondal D, Guo B, Das S. Nanoparticle-mediated stimulus-responsive antibacterial therapy. Biomater Sci 2023; 11:1994-2019. [PMID: 36748318 DOI: 10.1039/d2bm01941h] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The limitations associated with conventional antibacterial therapies and the subsequent amplification of multidrug-resistant (MDR) microorganisms have increased, necessitating the urgent development of innovative antibacterial techniques. Accordingly, nanoparticle-mediated therapeutics have emerged as potential candidates for antibacterial treatment due to their suitable dimensions, penetration capacity, and high efficiency in targeted drug delivery. However, although nanoparticle-based drug delivery systems have been demonstrated to be effective, they are limited by their overuse and unwanted side effects. Thus, to overcome these drawbacks, stimulus-responsive antibiotic delivery has been extended as a promising strategy for site-specific restricted drug exemption. Nano-formulations that are triggered by various stimuli, such as intrinsic, extrinsic, and bacterial stimuli, have been developed. Thus, by harnessing the physicochemical properties of various nanoparticles, the selective release of therapeutic cargoes can be achieved through the application of a variety of local stimuli such as light, sound, irradiation, pH, and magnetic field. In this review, we also highlight the progress and perspectives of stimulus-responsive combination therapy, with special emphasis on the eradication of MDR strains and biofilms. Hence, this review addresses the advancement and challenges in the applications of stimulus-responsive nanoparticles together with the various future prospects of this technique.
Collapse
Affiliation(s)
- Neelanjana Bag
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| | - Souravi Bardhan
- Department of Physics, Jadavpur University, Kolkata-700032, India. .,Department of Environmental Science, Netaji Nagar College for Women, Kolkata-700092, India
| | - Shubham Roy
- Department of Physics, Jadavpur University, Kolkata-700032, India. .,Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Jhilik Roy
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| | - Dhananjoy Mondal
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Sukhen Das
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| |
Collapse
|
247
|
Zhu Y, Gong P, Wang J, Cheng J, Wang W, Cai H, Ao R, Huang H, Yu M, Lin L, Chen X. Amplification of Lipid Peroxidation by Regulating Cell Membrane Unsaturation To Enhance Chemodynamic Therapy. Angew Chem Int Ed Engl 2023; 62:e202218407. [PMID: 36708200 DOI: 10.1002/anie.202218407] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/29/2023]
Abstract
Lipid peroxidation (LPO) is one of the most damaging processes in chemodynamic therapy (CDT). Although it is well known that polyunsaturated fatty acids (PUFAs) are much more susceptible than saturated or monounsaturated ones to LPO, there is no study exploring the effect of cell membrane unsaturation degree on CDT. Here, we report a self-reinforcing CDT agent (denoted as OA@Fe-SAC@EM NPs), consisting of oleanolic acid (OA)-loaded iron single-atom catalyst (Fe-SAC)-embedded hollow carbon nanospheres encapsulated by an erythrocyte membrane (EM), which promotes LPO to improve chemodynamic efficacy via modulating the degree of membrane unsaturation. Upon uptake of OA@Fe-SAC@EM NPs by cancer cells, Fe-SAC-catalyzed conversion of endogenous hydrogen peroxide into hydroxyl radicals, in addition to initiating the chemodynamic therapeutic process, causes the dissociation of the EM shell and the ensuing release of OA that can enrich cellular membranes with PUFAs, enabling LPO amplification-enhanced CDT.
Collapse
Affiliation(s)
- Yang Zhu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.,Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.,Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Peng Gong
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jun Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Junjie Cheng
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Wenyu Wang
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Huilan Cai
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Rujiang Ao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Hongwei Huang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Meili Yu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Lisen Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.,Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
248
|
Zhang J, Tang K, Fang R, Liu J, Liu M, Ma J, Wang H, Ding M, Wang X, Song Y, Yang D. Nanotechnological strategies to increase the oxygen content of the tumor. Front Pharmacol 2023; 14:1140362. [PMID: 36969866 PMCID: PMC10034070 DOI: 10.3389/fphar.2023.1140362] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Hypoxia is a negative prognostic indicator of solid tumors, which not only changes the survival state of tumors and increases their invasiveness but also remarkably reduces the sensitivity of tumors to treatments such as radiotherapy, chemotherapy and photodynamic therapy. Thus, developing therapeutic strategies to alleviate tumor hypoxia has recently been considered an extremely valuable target in oncology. In this review, nanotechnological strategies to elevate oxygen levels in tumor therapy in recent years are summarized, including (I) improving the hypoxic tumor microenvironment, (II) oxygen delivery to hypoxic tumors, and (III) oxygen generation in hypoxic tumors. Finally, the challenges and prospects of these nanotechnological strategies for alleviating tumor hypoxia are presented.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Kaiyuan Tang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Runqi Fang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Jiaming Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Ming Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma’anshan, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| | - Yanni Song
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| |
Collapse
|
249
|
Shi Y, Zhou M, Zhang Y, Wang Y, Cheng J. MRI-guided dual-responsive anti-tumor nanostructures for synergistic chemo-photothermal therapy and chemodynamic therapy. Acta Biomater 2023; 158:571-582. [PMID: 36586501 DOI: 10.1016/j.actbio.2022.12.053] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Image-guided stimulus-responsive theranostics are beneficial for identifying malignant lesions and integrating multiple cell-killing mechanisms to enhance tumor cell clearance. Herein, an intelligent dual-responsive nanostructure (HSPMH-DOX) was developed for magnetic resonance imaging (MRI)-guided synergistic chemo-photothermal therapy (PTT) and chemodynamic therapy (CDT). The core-shell nanostructure was synthesized by layering polydopamine (PDA), manganese oxide (MnO2), and hyaluronic acid (HA) onto drug-loaded hollow mesoporous silica nanoparticles (HS). The constructed nanoagent has both endogenous and external dual responses. The tumor microenvironment (pH/GSH) can trigger the degradation of gatekeeper (MnO2 and PDA), resulting in the release of anti-tumor drugs, whereas external near-infrared light irradiation can accelerate the degradation process and generate local overheating, resulting in PTT. Notably, MnO2 can not only consume intracellular GSH to enhance CDT but also release Mn2+ for precise localization of tumor tissues using MRI. Both in vitro and in vivo experiments showed that the prepared dual-response nanoagent satisfied biocompatibility, targeting, and the great efficiency of MRI-guided combined therapy. In animal models, combining chemo-PTT and CDT can eradicate tumors in less than two weeks. This work could pave the way for a wide range of stimulus-responsive synergistic theranostic applications, including MRI, chemo-photothermal therapy, and chemodynmic therapy. STATEMENT OF SIGNIFICANCE: Low bioavailability and severe side effects remain the major limitations of conventional cancer chemotherapy. Image-guided combination therapy can alleviate these problems and improve tumor-specific therapy. In the present study, the anticancer drug doxorubicin was encapsulated in a core-shell hollow mesoporous silica nanostructure (HSPMH-DOX), enabling MRI-guided targeted release under both endogenous and external dual stimuli. Moreover, the photothermal and nanoenzymatic effects of nanomedicine can cause local overheating in the tumor and amplify the intracellular CDT effect, accelerating tumor eradication. Systematic evaluations in vitro and in vivo confirmed that nanomedicine enables highly effective MRI-guided synergistic chemo-photothermal and chemodynamic therapy. This work offers a promising therapeutic strategy for precise anti-tumor applications.
Collapse
Affiliation(s)
- Yupeng Shi
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Mengyang Zhou
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yong Zhang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yifei Wang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
250
|
Ding B, Chen H, Tan J, Meng Q, Zheng P, Ma P, Lin J. ZIF-8 Nanoparticles Evoke Pyroptosis for High-Efficiency Cancer Immunotherapy. Angew Chem Int Ed Engl 2023; 62:e202215307. [PMID: 36629270 DOI: 10.1002/anie.202215307] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Although zeolitic imidazolate framework-8 (ZIF-8) has been applied in various tumor therapies, the intrinsic immunogenicity remains unclear. Here, we initiatively discover that ZIF-8 nanoparticles (NPs) can intrinsically induce pyroptosis by a caspase-1/gasdermin D (GSDMD)-dependent pathway. The pyroptotic cell death is accompanied by necrosis and immunogenic cell death (ICD) simultaneously for efficient in situ immunity initiation. Meanwhile, carbonyl cyanide m-chlorophenyl hydrazone (CCCP), a mitochondrial depolarizing agent, is successfully loaded into ZIF-8 NPs and found to further enhance the pyroptosis process. Collectively, the obtained Pluronic F127-modified CCCP-incorporated ZIF-8 NPs (F127 ZIF-8CCCP NPs) activate antitumor immunity and reprogram immunosuppressive tumor microenvironment (TME), realizing high-efficiency tumor growth inhibition. This work will facilitate biomedicine applications of ZIF-8 and provide good inspiration for pyroptosis-induced cancer therapy.
Collapse
Affiliation(s)
- Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Institute of Frontier and Interdisciplinarity Science and Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|