1
|
Wang Y, Ma K, Kang M, Yan D, Niu N, Yan S, Sun P, Zhang L, Sun L, Wang D, Tan H, Tang BZ. A new era of cancer phototherapy: mechanisms and applications. Chem Soc Rev 2024. [PMID: 39494674 DOI: 10.1039/d4cs00708e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
The past decades have witnessed great strides in phototherapy as an experimental option or regulation-approved treatment in numerous cancer indications. Of particular interest is nanoscale photosensitizer-based phototherapy, which has been established as a prominent candidate for advanced tumor treatment by virtue of its high efficacy and safety. Despite considerable research progress on materials, methods and devices in nanoscale photosensitizing agent-based phototherapy, their mechanisms of action are not always clear, which impedes their practical application in cancer treatment. Hence, from a new perspective, this review elaborates the working mechanisms, involving impairment and moderation effects, of diverse phototherapies on cells, organelles, organs, and tissues. Furthermore, the most current available phototherapy modalities are categorized as photodynamic, photothermal, photo-immune, photo-gas, and radio therapies in this review. A comprehensive understanding of the inferiority and superiority of various phototherapies will facilitate the advent of a new era of cancer phototherapy.
Collapse
Affiliation(s)
- Yuanwei Wang
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Ke Ma
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Miaomiao Kang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Dingyuan Yan
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Niu Niu
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Saisai Yan
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Panpan Sun
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Luzhi Zhang
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Lijie Sun
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Dong Wang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Hui Tan
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong Shenzhen, (CUHK-Shenzhen), Guangdong 518172, China.
| |
Collapse
|
2
|
Song J, Feng Y, Yan J, Wang Y, Yan W, Yang N, Wu T, Liu S, Wang Y, Zheng N, He L, Zhang Y. Computed Tomography Imaging Guided Microenvironment-Responsive Ir@WO 3-x Dual-Catalytic Nanoreactor for Selective Radiosensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405192. [PMID: 39102342 PMCID: PMC11481196 DOI: 10.1002/advs.202405192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/28/2024] [Indexed: 08/07/2024]
Abstract
Radiotherapy (RT) is often administered, either alone or in combination with other therapies, for most malignancies. However, the degree of tumor oxygenation, damage to adjacent healthy tissues, and inaccurate guidance remain issues that result in discontinuation or failure of RT. Here, a multifunctional therapeutic platform based on Ir@WO3-x is developed which simultaneously addresses these critical issues above for precision radiosensitization. Ir@WO3-x nanoreactors exhibit strong absorption of X-ray, acting as radiosensitizers. Moreover, ultrasmall Ir enzyme-mimic nanocrystals (NCs) are decorated onto the surface of the nanoreactor, where NCs have catalyst-like activity and are sensitive to H2O2 in the tumor microenvironment (TME) under near infrared-II (NIR-II) light stimulation. They efficiently catalyze the conversion of H2O2 to O2, thereby ameliorating hypoxia, inhibiting the expression of HIF-1α, and enhancing RT-induced DNA damage in cancerous tissue, further improving the efficiency of RT. Additionally, in response to high H2O2 levels in TME, the Ir@WO3-x nanoreactor also exerts peroxidase-like activity, boosting exogenous ROS, which increases oxidative damage and enhances ROS-dependent death signaling. Furthermore, Ir@WO3-x can serve as a high-quality computed tomography contrast agent due to its high X-ray attenuation coefficient and generation of pronounced tumor-tissue contrast. This report highlights the potential of advanced health materials to enhance precision therapeutic modalities.
Collapse
Affiliation(s)
- Jiayu Song
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
| | - Yue Feng
- Department of Gynecological OncologyZhejiang Cancer HospitalZhengzhouZhejiang310022China
| | - Jiazhuo Yan
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Ying Wang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Weixiao Yan
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
| | - Nan Yang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Tusheng Wu
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Sijia Liu
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Yuan Wang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Nannan Zheng
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhouHenan450000China
| | - Liangcan He
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhouHenan450000China
| | - Yunyan Zhang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| |
Collapse
|
3
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self-Catalytic NO/O 2 Gas-Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403921. [PMID: 39101290 DOI: 10.1002/adma.202403921] [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: 03/17/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Radiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor-targeted "prosthetic-Arginine" coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO-donating Fmoc-protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu-HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2-transporting ability of heme, HRRu-HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu-HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu-HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment, Henan, 450001, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
4
|
Sun W, Wang H, Qi Y, Li M, Zhang R, Gao Z, Cui J, Yu D. Metal-Phenolic Vehicles Potentiate Cycle-Cascade Activation of Pyroptosis and cGAS-STING Pathway for Tumor Immunotherapy. ACS NANO 2024; 18:23727-23740. [PMID: 39155444 DOI: 10.1021/acsnano.4c08613] [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: 08/20/2024]
Abstract
The treatment of triple-negative breast cancer (TNBC) faces challenges due to its limited immune response and weak tumor immunogenicity. A collaborative strategy involves combining the activation of pyroptosis and the stimulator of interferon genes (STING) pathway to enhance tumor immunogenicity and fortify the antitumor immune response, which may improve therapeutic outcomes in TNBC. In this study, we report the fabrication of a zinc-phenolic nanocapsule (RMP@Cap), which is loaded with mitoxantrone (MTO) and anti-PD-L1 antibodies (aPD-L1) and coated with erythrocyte membrane, for TNBC immunotherapy. The delivery of RMP@Cap can induce tumor cell pyroptosis and, therefore, trigger the release of mitochondrial DNA, which further combines with zinc agonists to intensify STING activation, resulting in a cascade amplification of the therapeutic effect on tumors. Additionally, the incorporation of aPD-L1 into the zinc-phenolic nanocapsule relieves the inhibitory effect of tumor cells on recruited cytotoxic T cells, thereby improving the tumor-killing capacity. Furthermore, the incorporation of a camouflaged erythrocyte membrane coating enables nanocapsules to achieve prolonged in vivo circulation, resulting in improved tumor accumulation for effective antitumor therapy. This study demonstrates a synergistic therapeutic modality involving pyroptosis, coupled with the simultaneous activation and cyclic amplification of the STING pathway in immunotherapy.
Collapse
Affiliation(s)
- Weikai Sun
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Hong Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yafei Qi
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Mengqi Li
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Ruyue Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Zhiliang Gao
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging, Jinan, Shandong 250012, China
- Research Institute of Shandong University: Magnetic Field-Free Medicine & Functional Imaging, Jinan, Shandong 250012, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Jinan, Shandong 250012, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging, Jinan, Shandong 250012, China
- Research Institute of Shandong University: Magnetic Field-Free Medicine & Functional Imaging, Jinan, Shandong 250012, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Jinan, Shandong 250012, China
| |
Collapse
|
5
|
Qiao R, Yuan Z, Yang M, Tang Z, He L, Chen T. Selenium-Doped Nanoheterojunctions for Highly Efficient Cancer Radiosensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402039. [PMID: 38828705 PMCID: PMC11304322 DOI: 10.1002/advs.202402039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/30/2024] [Indexed: 06/05/2024]
Abstract
Exploring efficient and low-toxicity radiosensitizers to break through the bottleneck of radiation tolerance, immunosuppression and poor prognosis remains one of the critical developmental challenges in radiotherapy. Nanoheterojunctions, due to their unique physicochemical properties, have demonstrated excellent radiosensitization effects in radiation energy deposition and in lifting tumor radiotherapy inhibition. Herein, they doped selenium (Se) into prussian blue (PB) to construct a nano-heterojunction (Se@PB), which could promote the increase of Fe2+/Fe3+ ratio and conversion of Se to a high valence state with Se introduction. The Fe2+-Se-Fe3+ electron transfer chain accelerates the rate of electron transfer on the surface of the nanoparticles, which in turn endows it with efficient X-ray energy transfer and electron transport capability, and enhances radiotherapy physical sensitivity. Furthermore, Se@PB induces glutathione (GSH) depletion and Fe2+ accumulation through pro-Fenton reaction, thereby disturbs the redox balance in tumor cells and enhances biochemical sensitivity of radiotherapy. As an excellent radiosensitizer, Se@PB effectively enhances X-ray induced mitochondrial dysfunction and DNA damage, thereby promotes cell apoptosis and synergistic cervical cancer radiotherapy. This study elucidates the radiosensitization mechanism of Se-doped nanoheterojunction from the perspective of the electron transfer chain and biochemistry reaction, which provides an efficient and low-toxic strategy in radiotherapy.
Collapse
Affiliation(s)
- Rui Qiao
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Zhongwen Yuan
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Meijin Yang
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Zhiying Tang
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Lizhen He
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Tianfeng Chen
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| |
Collapse
|
6
|
Yue R, Li Z, Liu H, Wang Y, Li Y, Yin R, Yin B, Qian H, Kang H, Zhang X, Song G. Imaging-guided companion diagnostics in radiotherapy by monitoring APE1 activity with afterglow and MRI imaging. Nat Commun 2024; 15:6349. [PMID: 39068156 PMCID: PMC11283504 DOI: 10.1038/s41467-024-50688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Companion diagnostics using biomarkers have gained prominence in guiding radiotherapy. However, biopsy-based techniques fail to account for real-time variations in target response and tumor heterogeneity. Herein, we design an activated afterglow/MRI probe as a companion diagnostics tool for dynamically assessing biomarker apurinic/apyrimidinic endonuclease 1(APE1) during radiotherapy in vivo. We employ ultrabright afterglow nanoparticles and ultrasmall FeMnOx nanoparticles as dual contrast agents, significantly broadening signal change range and enhancing the sensitivity of APE1 imaging (limit of detection: 0.0092 U/mL in afterglow imaging and 0.16 U/mL in MRI). We devise longitudinally and transversely subtraction-enhanced imaging (L&T-SEI) strategy to markedly enhance MRI contrast and signal-to-noise ratio between tumor and normal tissue of living female mice. The combined afterglow and MRI facilitate both anatomical and functional imaging of APE1 activity. This probe enables correlation of afterglow and MRI signals with APE1 expression, radiation dosage, intratumor ROS, and DNA damage, enabling early prediction of radiotherapy outcomes (as early as 3 h), significantly preceding tumor size reduction (6 days). By monitoring APE1 levels, this probe allows for early and sensitive detection of liver organ injury, outperforming histopathological analysis. Furthermore, MRI evaluates APE1 expression in radiation-induced abscopal effects provides insights into underlying mechanisms, and supports the development of treatment protocols.
Collapse
Affiliation(s)
- Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, PR China
| | - Zhe Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Huiyi Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Youjuan Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Yuhang Li
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, PR China
| | - Rui Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Baoli Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, Seoul, South Korea
| | - Xiaobing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China.
- Shenzhen Research Institute, Hunan University, Shenzhen, China.
| |
Collapse
|
7
|
Wang D, Jia H, Cao H, Hou X, Wang Q, Lin J, Liu J, Yang L, Liu J. A Dual-Channel Ca 2+ Nanomodulator Induces Intracellular Ca 2+ Disorders via Endogenous Ca 2+ Redistribution for Tumor Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401222. [PMID: 38690593 DOI: 10.1002/adma.202401222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/21/2024] [Indexed: 05/02/2024]
Abstract
Tumor cells harness Ca2+ to maintain cellular homeostasis and withstand external stresses from various treatments. Here, a dual-channel Ca2+ nanomodulator (CAP-P-NO) is constructed that can induce irreversible intracellular Ca2+ disorders via the redistribution of tumor-inherent Ca2+ for disrupting cellular homeostasis and thus improving tumor radiosensitivity. Stimulated by tumor-overexpressed acid and glutathione, capsaicin and nitric oxide are successively escaped from CAP-P-NO to activate the transient receptor potential cation channel subfamily V member 1 and the ryanodine receptor for the influx of extracellular Ca2+ and the release of Ca2+ in the endoplasmic reticulum, respectively. The overwhelming level of Ca2+ in tumor cells not only impairs the function of organelles but also induces widespread changes in the gene transcriptome, including the downregulation of a set of radioresistance-associated genes. Combining CAP-P-NO treatment with radiotherapy achieves a significant suppression against both pancreatic and patient-derived hepatic tumors with negligible side effects. Together, the study provides a feasible approach for inducing tumor-specific intracellular Ca2+ overload via endogenous Ca2+ redistribution and demonstrates the great potential of Ca2+ disorder therapy in enhancing the sensitivity for tumor radiotherapy.
Collapse
Affiliation(s)
- Dianyu Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Haixue Jia
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Hongmei Cao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Xiaoxue Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Qian Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jia Lin
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jinjian Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Lijun Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
8
|
Yang DH, Nah H, Lee D, Min SJ, Park S, An SH, Wang J, He H, Choi KS, Ko WK, Lee JS, Kwon IK, Lee SJ, Heo DN. A review on gold nanoparticles as an innovative therapeutic cue in bone tissue engineering: Prospects and future clinical applications. Mater Today Bio 2024; 26:101016. [PMID: 38516171 PMCID: PMC10952045 DOI: 10.1016/j.mtbio.2024.101016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/19/2024] [Accepted: 03/02/2024] [Indexed: 03/23/2024] Open
Abstract
Bone damage is a complex orthopedic problem primarily caused by trauma, cancer, or bacterial infection of bone tissue. Clinical care management for bone damage remains a significant clinical challenge and there is a growing need for more advanced bone therapy options. Nanotechnology has been widely explored in the field of orthopedic therapy for the treatment of a severe bone disease. Among nanomaterials, gold nanoparticles (GNPs) along with other biomaterials are emerging as a new paradigm for treatment with excellent potential for bone tissue engineering and regenerative medicine applications. In recent years, a great deal of research has focused on demonstrating the potential for GNPs to provide for enhancement of osteogenesis, reduction of osteoclastogenesis/osteomyelitis, and treatment of bone cancer. This review details the latest understandings in regards to GNPs based therapeutic systems, mechanisms, and the applications of GNPs against various bone disorders. The present review aims to summarize i) the mechanisms of GNPs in bone tissue remodeling, ii) preparation methods of GNPs, and iii) functionalization of GNPs and its decoration on biomaterials as a delivery vehicle in a specific bone tissue engineering for future clinical application.
Collapse
Affiliation(s)
- Dae Hyeok Yang
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Haram Nah
- Department of Dentistry, Graduate School, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Donghyun Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Sung Jun Min
- Department of Dentistry, Graduate School, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Seulki Park
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Sang-Hyun An
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Kyu-Sun Choi
- Department of Neurosurgery, College of Medicine, Hanyang University, 222, Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Wan-Kyu Ko
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jae Seo Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul 02447, Republic of Korea
- Kyung Hee University Medical Science Research Institute, Kyung Hee University, 23 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Sang Jin Lee
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, China
| | - Dong Nyoung Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul 02447, Republic of Korea
- Biofriends Inc, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| |
Collapse
|
9
|
Zhang R, Jia M, Lv H, Li M, Ding G, Cheng G, Li J. Assembling Au 8 clusters on surfaces of bifunctional nanoimmunomodulators for synergistically enhanced low dose radiotherapy of metastatic tumor. J Nanobiotechnology 2024; 22:20. [PMID: 38183048 PMCID: PMC10768385 DOI: 10.1186/s12951-023-02279-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Radiotherapy is one of the mainstays of cancer therapy and has been used for treating 65-75% of patients with solid tumors. However, radiotherapy of tumors has two limitations: high-dose X-rays damage adjacent normal tissue and tumor metastases cannot be prevented. RESULTS Therefore, to overcome the two limitations of radiotherapy, a multifunctional core-shell R837/BMS@Au8 nanoparticles as a novel radiosensitizer were fabricated by assembling Au8NCs on the surface of a bifunctional nanoimmunomodulator R837/BMS nanocore using nanoprecipitation followed by electrostatic assembly. Formed R837/BMS@Au8 NP composed of R837, BMS-1, and Au8 clusters. Au8NC can enhance X-ray absorption at the tumor site to reduce X-ray dose and releases a large number of tumor-associated antigens under X-ray irradiation. With the help of immune adjuvant R837, dendritic cells can effectively process and present tumor-associated antigens to activate effector T cells, meanwhile, a small-molecule PD-L1 inhibitor BMS-1 can block PD-1/PD-L1 pathway to reactivate cytotoxic T lymphocyte, resulting in a strong systemic antitumor immune response that is beneficial for limiting tumor metastasis. According to in vivo and in vitro experiments, radioimmunotherapy based on R837/BMS@Au8 nanoparticles can increase calreticulin expression on of cancer cells, reactive oxygen species generation, and DNA breakage and decrease colony formation. The results revealed that distant tumors were 78.2% inhibited depending on radioimmunotherapy of primary tumors. Therefore, the use of a novel radiosensitizer R837/BMS@Au8 NPs realizes low-dose radiotherapy combined with immunotherapy against advanced cancer. CONCLUSION In conclusion, the multifunctional core-shell R837/BMS@Au8 nanoparticles as a novel radiosensitizer effectively limiting tumor metastasis and decrease X-ray dose to 1 Gy, providing an efective strategy for the construction of nanosystems with radiosensitizing function.
Collapse
Affiliation(s)
- Rui Zhang
- School of Public Health, Jilin University, Chang Chun, 130021, China.
| | - Mengchao Jia
- School of Public Health, Jilin University, Chang Chun, 130021, China
| | - Hongying Lv
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Radiation Medicine Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Mengxuan Li
- School of Public Health, Jilin University, Chang Chun, 130021, China
| | - Guanwen Ding
- School of Public Health, Jilin University, Chang Chun, 130021, China
| | - Ge Cheng
- School of Public Health, Jilin University, Chang Chun, 130021, China
| | - Juan Li
- School of Public Health, Jilin University, Chang Chun, 130021, China.
| |
Collapse
|
10
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
11
|
A R, Wang H, Nie C, Han Z, Zhou M, Atinuke OO, Wang K, Wang X, Liu S, Zhao J, Qiao W, Sun X, Wu L, Sun X. Glycerol-weighted chemical exchange saturation transfer nanoprobes allow 19F /1H dual-modality magnetic resonance imaging-guided cancer radiotherapy. Nat Commun 2023; 14:6644. [PMID: 37863898 PMCID: PMC10589257 DOI: 10.1038/s41467-023-42286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023] Open
Abstract
Recently, radiotherapy (RT) has entered a new realm of precision cancer therapy with the introduction of magnetic resonance (MR) imaging guided radiotherapy systems into the clinic. Nonetheless, identifying an optimized radiotherapy time window (ORTW) is still critical for the best therapeutic efficacy of RT. Here we describe pH and O2 dual-sensitive, perfluorooctylbromide (PFOB)-based and glycerol-weighted chemical exchange saturation transfer (CEST) nano-molecular imaging probes (Gly-PFOBs) with dual fluorine and hydrogen proton based CEST MR imaging properties (19F/1H-CEST). Oxygenated Gly-PFOBs ameliorate tumor hypoxia and improve O2-dependent radiotherapy. Moreover, the pH and O2 dual-sensitive properties of Gly-PFOBs could be quantitatively, spatially, and temporally monitored by 19F/1H-CEST imaging to optimize ORTW. In this study, we describe the CEST signal characteristics exhibited by the glycerol components of Gly-PFOBs. The pH and O2 dual-sensitive Gly-PFOBs with19F/1H-CEST MR dual-modality imaging properties, with superior therapeutic efficacy and biosafety, are employed for sensitive imaging-guided lung cancer RT, illustrating the potential of multi-functional imaging to noninvasively monitor and enhance RT-integrated effectiveness.
Collapse
Affiliation(s)
- Rong A
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Haoyu Wang
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Chaoqun Nie
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Zhaoguo Han
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Meifang Zhou
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Olagbaju Oluwatosin Atinuke
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Kaiqi Wang
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Xiance Wang
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Shuang Liu
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Jingshi Zhao
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Wenju Qiao
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Xiaohong Sun
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Lina Wu
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China
| | - Xilin Sun
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Heilongjiang Province, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Heilongjiang Province, China.
| |
Collapse
|
12
|
Gerken LRH, Gerdes ME, Pruschy M, Herrmann IK. Prospects of nanoparticle-based radioenhancement for radiotherapy. MATERIALS HORIZONS 2023; 10:4059-4082. [PMID: 37555747 PMCID: PMC10544071 DOI: 10.1039/d3mh00265a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
Radiotherapy is a key pillar of solid cancer treatment. Despite a high level of conformal dose deposition, radiotherapy is limited due to co-irradiation of organs at risk and subsequent normal tissue toxicities. Nanotechnology offers an attractive opportunity for increasing the efficacy and safety of cancer radiotherapy. Leveraging the freedom of design and the growing synthetic capabilities of the nanomaterial-community, a variety of engineered nanomaterials have been designed and investigated as radiosensitizers or radioenhancers. While research so far has been primarily focused on gold nanoparticles and other high atomic number materials to increase the absorption cross section of tumor tissue, recent studies are challenging the traditional concept of high-Z nanoparticle radioenhancers and highlight the importance of catalytic activity. This review provides a concise overview on the knowledge of nanoparticle radioenhancement mechanisms and their quantification. It critically discusses potential radioenhancer candidate materials and general design criteria for different radiation therapy modalities, and concludes with research priorities in order to advance the development of nanomaterials, to enhance the efficacy of radiotherapy and to increase at the same time the therapeutic window.
Collapse
Affiliation(s)
- Lukas R H Gerken
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Maren E Gerdes
- Karolinska Institutet, Solnavägen 1, 171 77 Stockholm, Sweden
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Inge K Herrmann
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
13
|
Xiao L, Chen B, Wang W, Tian T, Qian H, Li X, Yu Y. Multifunctional Au@AgBiS 2 Nanoparticles as High-Efficiency Radiosensitizers to Induce Pyroptosis for Cancer Radioimmunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302141. [PMID: 37688340 PMCID: PMC10602534 DOI: 10.1002/advs.202302141] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/09/2023] [Indexed: 09/10/2023]
Abstract
Radiotherapy (RT), a widely used clinical treatment modality for cancer, uses high-energy irradiation for reactive oxygen species (ROS) production and DNA damage. However, its therapeutic effect is primarily limited owing to insufficient DNA damage to tumors and harmful effects on normal tissues. Herein, a core-shell structure of metal-semiconductors (Au@AgBiS2 nanoparticles) that can function as pyroptosis inducers to both kill cancer cells directly and trigger a robust anti-tumor immune against 4T1 triple-negative murine breast cancer and metastasis is rationally designed. Metal-semiconductor composites can enhance the generation of considerable ROS and simultaneously DNA damage for RT sensitization. Moreover, Au@AgBiS2 , a pyroptosis inducer, induces caspase-3 protein activation, gasdermin E cleavage, and the release of damage-associated molecular patterns. In vivo studies in BALB/c mice reveal that Au@AgBiS2 nanoparticles combined with RT exhibit remarkable antitumor immune activity, preventing tumor growth, and lung metastasis. Therefore, this core-shell structure is an alternative for designing highly effective radiosensitizers for radioimmunotherapy.
Collapse
Affiliation(s)
- Liang Xiao
- Department of RadiologyResearch Center of Clinical Medical ImagingAnhui Province Clinical Image Quality Control CenterThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022P. R. China
| | - Benjin Chen
- Department of PharmacologySchool of Basic Medical SciencesAnhui Medical UniversityHefei230032P. R. China
| | - Wanni Wang
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Engineering Research Center for Medical Micro‐Nano DevicesAnhui Medical UniversityHefei230011P. R. China
| | - Tian Tian
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230036P. R. China
| | - Haisheng Qian
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Engineering Research Center for Medical Micro‐Nano DevicesAnhui Medical UniversityHefei230011P. R. China
| | - Xiaohu Li
- Department of RadiologyResearch Center of Clinical Medical ImagingAnhui Province Clinical Image Quality Control CenterThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022P. R. China
| | - Yongqiang Yu
- Department of RadiologyResearch Center of Clinical Medical ImagingAnhui Province Clinical Image Quality Control CenterThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022P. R. China
| |
Collapse
|
14
|
Fan X, Li K, Liu S, Wang T, Ma Y, Li Z, He C. Protein Nanotubes Assembled from Imidazole-Grafted Horseradish Peroxidase Nanogels. ACS Macro Lett 2023; 12:1031-1036. [PMID: 37433040 DOI: 10.1021/acsmacrolett.3c00198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Protein assembly, a common phenomenon in nature, plays an important role in the evolution of life. Inspired by nature, assembling protein monomers into delicate nanostructures has emerged as an attractive research area. However, sophisticated protein assemblies usually need complicated designs or templates. In this work, we successfully fabricated protein nanotubes in a facile way by coordination interactions between imidazole-grafted horseradish peroxidase (HRP) nanogels (iHNs) and Cu2+. The iHNs were synthesized by polymerization on the surface of HRP by employing vinyl imidazole as a comonomer. By direct addition of Cu2+ into iHN solution, protein tubes were therefore formed. The size of the protein tubes could be adjusted by changing the added Cu2+ amount, and the mechanism behind the formation of protein nanotubes was elucidated. Furthermore, a highly sensitive H2O2 detection system was established based on the protein tubes. This work provides a facile method to construct diverse sophisticated functional protein nanomaterials.
Collapse
Affiliation(s)
- Xiaotong Fan
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Singapore
| | - Ke Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore
| | - Siqi Liu
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Tingting Wang
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Yedong Ma
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Zibiao Li
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Singapore
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore
| | - Chaobin He
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore
| |
Collapse
|
15
|
Varzandeh M, Sabouri L, Mansouri V, Gharibshahian M, Beheshtizadeh N, Hamblin MR, Rezaei N. Application of nano-radiosensitizers in combination cancer therapy. Bioeng Transl Med 2023; 8:e10498. [PMID: 37206240 PMCID: PMC10189501 DOI: 10.1002/btm2.10498] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 11/08/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Radiosensitizers are compounds or nanostructures, which can improve the efficiency of ionizing radiation to kill cells. Radiosensitization increases the susceptibility of cancer cells to radiation-induced killing, while simultaneously reducing the potentially damaging effect on the cellular structure and function of the surrounding healthy tissues. Therefore, radiosensitizers are therapeutic agents used to boost the effectiveness of radiation treatment. The complexity and heterogeneity of cancer, and the multifactorial nature of its pathophysiology has led to many approaches to treatment. The effectiveness of each approach has been proven to some extent, but no definitive treatment to eradicate cancer has been discovered. The current review discusses a broad range of nano-radiosensitizers, summarizing possible combinations of radiosensitizing NPs with several other types of cancer therapy options, focusing on the benefits and drawbacks, challenges, and future prospects.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials EngineeringIsfahan University of TechnologyIsfahanIran
| | - Leila Sabouri
- AmitisGen TECH Dev GroupTehranIran
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Vahid Mansouri
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical SciencesTehranIran
| | - Maliheh Gharibshahian
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeSchool of Medicine, Shahroud University of Medical SciencesShahroudIran
| | - Nima Beheshtizadeh
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Tissue EngineeringSchool of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
| | - Michael R. Hamblin
- Laser Research Center, Faculty of Health ScienceUniversity of JohannesburgDoornfonteinSouth Africa
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
- Research Center for ImmunodeficienciesChildren's Medical Center, Tehran University of Medical SciencesTehranIran
- Department of ImmunologySchool of Medicine, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
16
|
Shukla AK, Randhawa S, Saini TC, Acharya A. Carbon nanosphere based bifunctional oxidoreductase nano-catalytic agent to mitigate hypoxia in cancer cells. Int J Biol Macromol 2023; 233:123466. [PMID: 36739044 DOI: 10.1016/j.ijbiomac.2023.123466] [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: 11/03/2022] [Revised: 01/06/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Developing metal-free carbon nanozyme for tumor hypoxia is difficult. In biomedical applications, especially in the case of biomolecular detection, extensive research has been done on nanozymes with enzyme-mimicking catalytic activity. However, there are considerably fewer investigations on targeted nano-catalytic tumor therapy. Nano catalytic medicine-enabled chemotherapy is a safe and promising treatment strategy that involves the conversion of excess H2O2 into O2 in a tumor environment. Here we have synthesized carbon nanosphere (CNS) using the Camellia sinensis plant (CS-CNS). Further surface functionalization was achieved via nitrilotriacetic acid conjugation (NTA@CS-CNS). A stability study of synthesized nanozyme in the presence of various cations, anions, and 5 different pH range suggested the robustness of carbon based nanoassembly. The catalytic in vitro study shows that NTA@CS-CNS mimics peroxidase and catalase using TMB and H2O2 as substrates. NTA@CS-CNS showed Km and Vmax values of ~ 193.2 μM and 0.43 μM/s, ~ 413 μM and 1.42 μM/s, and ~ 378 μM and 1.63 μM/s, respectively when H2O2 and TMB was used for CAT and POD activity. Results showed that NTA@CS-CNS in combination with SFN and laser irradiation reduces hypoxia. Hence, our study could pave the path for the development of different non-toxic nano catalytic therapy for tumors in cancerous cells.
Collapse
Affiliation(s)
- Ashish K Shukla
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
17
|
Zhang G, Guo M, Ma H, Wang J, Zhang XD. Catalytic nanotechnology of X-ray photodynamics for cancer treatments. Biomater Sci 2023; 11:1153-1181. [PMID: 36602259 DOI: 10.1039/d2bm01698b] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Photodynamic therapy (PDT) has been applied in cancer treatment because of its high selectivity, low toxicity, and non-invasiveness. However, the limited penetration depth of the light still hampers from reaching deep-seated tumors. Considering the penetrating ability of high-energy radiotherapy, X-ray-induced photodynamic therapy (X-PDT) has evolved as an alternative to overcome tissue blocks. As the basic principle of X-PDT, X-rays stimulate the nanoparticles to emit scintillating or persistent luminescence and further activate the photosensitizers to generate reactive oxygen species (ROS), which would cause a series of molecular and cellular damages, immune response, and eventually break down the tumor tissue. In recent years, catalytic nanosystems with unique structures and functions have emerged that can enhance X-PDT therapeutic effects via an immune response. The anti-cancer effect of X-PDT is closely related to the following factors: energy conversion efficiency of the material, the radiation dose of X-rays, quantum yield of the material, tumor resistance, and biocompatibility. Based on the latest research in this field and the classical theories of nanoscience, this paper systematically elucidates the current development of the X-PDT and related immunotherapy, and highlights its broad prospects in medical applications, discussing the connection between fundamental science and clinical translation.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China.
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China.
| | - Huizhen Ma
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China.
| | - Junying Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiao-Dong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China. .,Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
18
|
Wang Z, Ren X, Wang D, Guan L, Li X, Zhao Y, Liu A, He L, Wang T, Zvyagin AV, Yang B, Lin Q. Novel strategies for tumor radiosensitization mediated by multifunctional gold-based nanomaterials. Biomater Sci 2023; 11:1116-1136. [PMID: 36601661 DOI: 10.1039/d2bm01496c] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Radiotherapy (RT) is one of the most effective and commonly used cancer treatments for malignant tumors. However, the existing radiosensitizers have a lot of side effects and poor efficacy, which limits the curative effect and further application of radiotherapy. In recent years, emerging nanomaterials have shown unique advantages in enhancing radiosensitization. In particular, gold-based nanomaterials, with high X-ray attenuation capacity, good biocompatibility, and promising chemical, electronic and optical properties, have become a new type of radiotherapy sensitizer. In addition, gold-based nanomaterials can be used as a carrier to load a variety of drugs and immunosuppressants; in particular, its photothermal therapy, photodynamic therapy and multi-mode imaging functions aid in providing excellent therapeutic effect in coordination with RT. Recently, many novel strategies of radiosensitization mediated by multifunctional gold-based nanomaterials have been reported, which provides a new idea for improving the efficacy and reducing the side effects of RT. In this review, we systematically summarize the recent progress of various new gold-based nanomaterials that mediate radiosensitization and describe the mechanism. We further discuss the challenges and prospects in the field. It is hoped that this review will help researchers understand the latest progress of gold-based nanomaterials for radiosensitization, and encourage people to optimize the existing methods or explore novel approaches for radiotherapy.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Xiaojun Ren
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Dongzhou Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Lin Guan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Xingchen Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Yue Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Liang He
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia.,Institute of Biology and Biomedicine, Lobachevsky Nizhny Novgorod State University, 603105, Nizhny Novgorod, Russia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| |
Collapse
|
19
|
Ghosh R, Satarifard V, Lipowsky R. Different pathways for engulfment and endocytosis of liquid droplets by nanovesicles. Nat Commun 2023; 14:615. [PMID: 36739277 PMCID: PMC9899248 DOI: 10.1038/s41467-023-35847-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/04/2023] [Indexed: 02/06/2023] Open
Abstract
During endocytosis of nanoparticles by cells, the cellular membranes engulf the particles, thereby forming a closed membrane neck that subsequently undergoes fission. For solid nanoparticles, these endocytic processes have been studied in some detail. Recently, such processes have also been found for liquid and condensate droplets, both in vitro and in vivo. These processes start with the spreading of the droplet onto the membrane followed by partial or complete engulfment of the droplet. Here, we use molecular dynamics simulations to study these processes at the nanoscale, for nano-sized droplets and vesicles. For both partial and complete engulfment, we observe two different endocytic pathways. Complete engulfment leads to a closed membrane neck which may be formed in a circular or strongly non-circular manner. A closed circular neck undergoes fission, thereby generating two nested daughter vesicles whereas a non-circular neck hinders the fission process. Likewise, partial engulfment of larger droplets leads to open membrane necks which can again have a circular or non-circular shape. Two key parameters identified here for these endocytic pathways are the transbilayer stress asymmetry of the vesicle membrane and the positive or negative line tension of the membrane-droplet contact line.
Collapse
Affiliation(s)
- Rikhia Ghosh
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14424, Potsdam, Germany.,Icahn School of Medicine Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Vahid Satarifard
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14424, Potsdam, Germany.,Yale Institute for Network Science, Yale University, New Haven, CT, 06520, USA
| | - Reinhard Lipowsky
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14424, Potsdam, Germany.
| |
Collapse
|
20
|
Zhuang Y, Liu K, He Q, Gu X, Jiang C, Wu J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e203. [PMID: 36703877 PMCID: PMC9870816 DOI: 10.1002/mco2.203] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoxia is a persistent physiological feature of many different solid tumors and a key driver of malignancy, and in recent years, it has been recognized as an important target for cancer therapy. Hypoxia occurs in the majority of solid tumors due to a poor vascular oxygen supply that is not sufficient to meet the needs of rapidly proliferating cancer cells. A hypoxic tumor microenvironment (TME) can reduce the effectiveness of other tumor therapies, such as radiotherapy, chemotherapy, and immunotherapy. In this review, we discuss the critical role of hypoxia in tumor development, including tumor metabolism, tumor immunity, and tumor angiogenesis. The treatment methods for hypoxic TME are summarized, including hypoxia-targeted therapy and improving oxygenation by alleviating tumor hypoxia itself. Hyperoxia therapy can be used to improve tissue oxygen partial pressure and relieve tumor hypoxia. We focus on the underlying mechanisms of hyperoxia and their impact on current cancer therapies and discuss the prospects of hyperoxia therapy in cancer treatment.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Kua Liu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Qinyu He
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Xiaosong Gu
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| |
Collapse
|
21
|
Liang Z, Li X, Chen X, Zhou J, Li Y, Peng J, Lin Z, Liu G, Zeng X, Li C, Hang L, Li H. Fe/MOF based platform for NIR laser induced efficient PDT/PTT of cancer. Front Bioeng Biotechnol 2023; 11:1156079. [PMID: 37064235 PMCID: PMC10098195 DOI: 10.3389/fbioe.2023.1156079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/15/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction: Photodynamic therapy (PDT) and photothermal therapy (PTT) are widely used in the treatment of tumors. However, their application in the treatment of clinical tumors is limited by the complexity and irreversible hypoxia environment generated by tumor tissues. To overcome this limitation, a nanoparticle composed of indocyanine green (ICG) and Fe-MOF-5 was developed. Methods: We prepared F-I@FM5 and measured its morphology, particle size, and stability. Its enzyme like ability and optical effect was verified. Then we used MTT, staining and flow cytometry to evaluated the anti-tumor effect on EMT-6 cells in vitro. Finally, the anti-tumor effect in vivo has been studied on EMT-6 tumor bearing mice. Results: For the composite nanoparticle, we confirmed that Fe-MOF-5 has the best nanozyme activity. In addition, it has excellent photothermal conversion efficiency and generates reactive oxygen species (ROS) under near-infrared light irradiation (808 nm). The composite nanoparticle showed good tumor inhibition effect in vitro and in vivo, which was superior to the free ICG or Fe-MOF-5 alone. Besides, there was no obvious cytotoxicity in major organs within the effective therapeutic concentration. Discussion: Fe-MOF-5 has the function of simulating catalase, which can promote the decomposition of excessive H2O2 in the tumor microenvironment and produce oxygen to improve the hypoxic environment. The improvement of tumor hypoxia can enhance the efficacy of PDT and PTT. This research not only provides an efficient and stable anti-tumor nano platform, but also has broad application prospects in the field of tumor therapy, and provides a new idea for the application of MOF as an important carrier material in the field of photodynamic therapy.
Collapse
Affiliation(s)
- Zixing Liang
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofeng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofang Chen
- Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiawei Zhou
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yanan Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jianhui Peng
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhousheng Lin
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Gai Liu
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiancheng Zeng
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Cheng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
- Jinan University, Guangzhou, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| | - Lifeng Hang
- Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| | - Hailiang Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| |
Collapse
|
22
|
Wang L, Fu H, Song L, Wu Z, Yu J, Guo Q, Chen C, Yang X, Zhang J, Wang Q, Duan Y, Yang Y. Overcoming AZD9291 Resistance and Metastasis of NSCLC via Ferroptosis and Multitarget Interference by Nanocatalytic Sensitizer Plus AHP-DRI-12. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204133. [PMID: 36420659 DOI: 10.1002/smll.202204133] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/03/2022] [Indexed: 06/16/2023]
Abstract
The acquired resistance to Osimertinib (AZD9291) greatly limits the clinical benefit of patients with non-small cell lung cancer (NSCLC), whereas AZD9291-resistant NSCLCs are prone to metastasis. It's challenging to overcome AZD9291 resistance and suppress metastasis of NSCLC simultaneously. Here, a nanocatalytic sensitizer (VF/S/A@CaP) is proposed to deliver Vitamin c (Vc)-Fe(II), si-OTUB2, ASO-MALAT1, resulting in efficient inhibition of tumor growth and metastasis of NSCLC by synergizing with AHP-DRI-12, an anti-hematogenous metastasis inhibitor by blocking the amyloid precursor protein (APP)/death receptor 6 (DR6) interaction designed by our lab. Fe2+ released from Vc-Fe(II) generates cytotoxic hydroxyl radicals (•OH) through Fenton reaction. Subsequently, glutathione peroxidase 4 (GPX4) is consumed to sensitize AZD9291-resistant NSCLCs with high mesenchymal state to ferroptosis due to the glutathione (GSH) depletion caused by Vc/dehydroascorbic acid (DHA) conversion. By screening NSCLC patients' samples, metastasis-related targets (OTUB2, LncRNA MALAT1) are confirmed. Accordingly, the dual-target knockdown plus AHP-DRI-12 significantly suppresses the metastasis of AZD9291-resistant NSCLC. Such modality leads to 91.39% tumor inhibition rate in patient-derived xenograft (PDX) models. Collectively, this study highlights the vulnerability to ferroptosis of AZD9291-resistant tumors and confirms the potential of this nanocatalytic-medicine-based modality to overcome critical AZD9291 resistance and inhibit metastasis of NSCLC simultaneously.
Collapse
Affiliation(s)
- Liting Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Liwei Song
- Shanghai Pulmonary Tumor Medical Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Zhihua Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Chuanrong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xupeng Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Jiali Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Quan Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200032, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yunhai Yang
- Shanghai Pulmonary Tumor Medical Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| |
Collapse
|
23
|
Chen J, Zhao B, Zou J, Yang J, Yang L, Zhang J, Chen W, Huang D, Zhong Y. Macromolecular NO-Donor Micelles for Targeted and Augmented Chemotherapy against Prostate Cancer. Adv Healthc Mater 2023; 12:e2202266. [PMID: 36415059 DOI: 10.1002/adhm.202202266] [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: 09/05/2022] [Revised: 11/01/2022] [Indexed: 11/24/2022]
Abstract
Mitoxantrone (MTO) is clinically utilized for treating hormone-refractory prostate cancer (PCa), however, the therapeutic outcome is far from optimal due to the lack of proper drug carrier as well as the inherent MTO detoxification mechanisms of DNA lesion repair and anti-oxidation. Herein, a bombesin-installed nanoplatform combining the chemotherapeutic MTO and the chemotherapeutic sensitizer of nitric oxide (NO) is developed based on MTO-loaded macromolecular NO-donor-containing polymeric micelles (BN-NMMTO ) for targeted NO-sensitized chemotherapy against PCa. BN-NMMTO actively target and accumulates in PCa sites and are internalized into the tumor cells. The macromolecular NO-donor of BN-NMMTO undergoes a reductive reaction to unleash NO upon intracellular glutathione (GSH), accompanying by micelle swelling and MTO release. The targeted intracellular MTO release induces DNA lesion and reactive oxygen species (ROS) generation in tumor cells without damage to the normal cells, and MTO's cytotoxicity is further augmented by NO release via the inhibition of both DNA repair and anti-oxidation pathways as compared with traditional MTO therapies.
Collapse
Affiliation(s)
- Jiaxin Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Bingbing Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Junhui Zou
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiachen Yang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Lifen Yang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Junmei Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.,Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.,Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
24
|
Liu S, Fang L, Ding H, Zhang Y, Li W, Liu B, Dong S, Tian B, Feng L, Yang P. Alternative Strategy to Optimize Cerium Oxide for Enhanced X-ray-Induced Photodynamic Therapy. ACS NANO 2022; 16:20805-20819. [PMID: 36378717 DOI: 10.1021/acsnano.2c08047] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The emergence of X-ray-induced photodynamic therapy (X-PDT) holds tremendous promise for clinical deep-penetrating cancer therapy. However, the clinical application of X-PDT in cancer treatment is still limited due to the hypoxic property of cancerous tissue, the inherent antioxidant system of tumor cells, and the difficulty in matching the absorption spectra of photosensitizers. Herein, a versatile core-shell radiosensitizer (SCNPs@DMSN@CeOx-PEG, denoted as SSCP) was elaborately designed and constructed to enhance X-PDT by coating tunable mesoporous silica on nanoscintillators, followed by embedding the cerium oxide nanoparticles in situ. The obtained SSCP radiosensitizer demonstrated a distinct blue-shift in the ultraviolet light region, so that it could perfectly absorb the ultraviolet light converted by the SCNPs core, resulting in the formation of photoinduced electron-hole (e--h+) pairs separation to generate reactive oxygen species (ROS). In addition, the cerium oxide exhibits high glutathione consumption to heighten ROS accumulation, and catalase-like activity to alleviate the hypoxia, which further enhances the efficiency of radiotherapy. Benefiting from the abundant Lu and Ce elements, the computed tomography imaging performance of SSCP is about 3.79-fold that of the clinical contrast agent (iohexol), which has great potential in both preclinical imaging and clinical translation.
Collapse
Affiliation(s)
- Shikai Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Linyang Fang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - He Ding
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Yangyang Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Wenting Li
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Bin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Shuming Dong
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Boshi Tian
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| |
Collapse
|
25
|
Han D, Zhang X, Ma Y, Yang X, Li Z. The development of live microorganism-based oxygen shuttles for enhanced hypoxic tumor therapy. Mater Today Bio 2022; 18:100517. [PMID: 36578285 PMCID: PMC9791452 DOI: 10.1016/j.mtbio.2022.100517] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Hypoxia is a prominent feature of malignant tumors and contributes to tumor proliferation, metastasis, and drug resistance in various solid tumors. Therefore, improving tumor oxygenation is crucial for curing tumors. To date, multiple strategies, including oxygen delivering and producing materials, have been designed to increase the oxygen concentration in hypoxic tumors. However, the unsustainable supply of oxygen is still the main obstacle, resulting in a suboptimal outcome in treating oxygen-deprived tumors. Thus, a sufficient oxygen supply is highly desirable in the treatment of hypoxic tumors. Photosynthesis, as the main source of oxygen in nature through the conversion of light energy into chemical energy and oxygen, has been widely studied in scientific research. Moreover, photosynthetic microorganisms have been increasingly applied in cancer therapy by increasing oxygenation, which improves the therapeutic effect of oxygen-consuming tumor therapeutic tools such as radiotherapy and photodynamic therapy. In this review, we summarize recent advances in the design and manufacture of live bacteria as oxygen shuttles for a new generation of hypoxic tumor treatment strategies. Finally, current challenges and future directions are also discussed for successfully addressing hypoxic tumor issues.
Collapse
Affiliation(s)
- Dandan Han
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China,College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xing Zhang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Yichuan Ma
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xinjian Yang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China,Corresponding author.
| | - Zhenhua Li
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangdong, 510515, PR China,Corresponding author. Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China.
| |
Collapse
|
26
|
Zhu L, Zhao Y, Liu T, Chen M, Qian WP, Jiang B, Barwick BG, Zhang L, Styblo TM, Li X, Yang L. Inhibition of NADPH Oxidase-ROS Signal using Hyaluronic Acid Nanoparticles for Overcoming Radioresistance in Cancer Therapy. ACS NANO 2022; 16:18708-18728. [PMID: 36256454 PMCID: PMC9764083 DOI: 10.1021/acsnano.2c07440] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Upregulation of NADPH oxidases (NOXs) in cancer cells leads to chronic increase in intracellular reactive oxygen species (ROS) and adaptation to a high ROS level for cell survival and, thereby, low sensitivity to radiotherapy. To overcome resistance to radiotherapy, we have developed a bioactive and CD44 targeted hyaluronic acid nanoparticle encapsulated with an NOX inhibitor, GKT831 (HANP/GKT831). We found that HANP/GKT831 had stronger inhibitory effects on ROS generation and cell proliferation than that of GKT831 alone in cancer cells. Systemic delivery of HANP/GKT831 led to the targeted accumulation in breast cancer patient derived xenograft (PDX) tumors in nude mice. Importantly, the combination of systemic delivery of HANP/GKT831 with a low dose of local radiotherapy significantly enhanced tumor growth inhibition in breast cancer PDX models. Our results showed that HANP/GKT831 primed tumor cells to radiation-induced DNA damage and cell death by downregulation of DNA repair function and oncogenic signal pathways.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Yi Zhao
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Tongrui Liu
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Minglong Chen
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Wei Ping Qian
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Binghua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia 19107, Pennsylvania, United States
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Lumeng Zhang
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Toncred M Styblo
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Lily Yang
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| |
Collapse
|
27
|
Garcia-Peiro JI, Bonet-Aleta J, Santamaria J, Hueso JL. Platinum nanoplatforms: classic catalysts claiming a prominent role in cancer therapy. Chem Soc Rev 2022; 51:7662-7681. [PMID: 35983786 DOI: 10.1039/d2cs00518b] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Platinum nanoparticles (Pt NPs) have a well-established role as a classic heterogeneous catalyst. Also, Pt has traditionally been employed as a component of organometallic drug formulations for chemotherapy. However, a new role in cancer therapy is emerging thanks to its outstanding catalytic properties, enabling novel approaches that are surveyed in this review. Herein, we critically discuss results already obtained and attempt to ascertain future perspectives for Pt NPs as catalysts able to modify key processes taking place in the tumour microenvironment (TME). In addition, we explore relevant parameters affecting the cytotoxicity, biodistribution and clearance of Pt nanosystems. We also analyze pros and cons in terms of biocompatibility and potential synergies that emerge from combining the catalytic capabilities of Pt with other agents such as co-catalysts, external energy sources (near-infrared light, X-ray, electric currents) and conventional therapies.
Collapse
Affiliation(s)
- Jose I Garcia-Peiro
- Instituto de Nanociencia y Materiales de Aragon (INMA) CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I + D, C/Poeta Mariano Esquillor, s/n, 50018, Zaragoza, Spain. .,Department of Chemical and Environmental Engineering, University of Zaragoza, Spain, Campus Rio Ebro, C/ María de Luna, 3, 50018 Zaragoza, Spain.,Networking Res. Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Javier Bonet-Aleta
- Instituto de Nanociencia y Materiales de Aragon (INMA) CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I + D, C/Poeta Mariano Esquillor, s/n, 50018, Zaragoza, Spain. .,Department of Chemical and Environmental Engineering, University of Zaragoza, Spain, Campus Rio Ebro, C/ María de Luna, 3, 50018 Zaragoza, Spain.,Networking Res. Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Jesus Santamaria
- Instituto de Nanociencia y Materiales de Aragon (INMA) CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I + D, C/Poeta Mariano Esquillor, s/n, 50018, Zaragoza, Spain. .,Department of Chemical and Environmental Engineering, University of Zaragoza, Spain, Campus Rio Ebro, C/ María de Luna, 3, 50018 Zaragoza, Spain.,Networking Res. Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Jose L Hueso
- Instituto de Nanociencia y Materiales de Aragon (INMA) CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I + D, C/Poeta Mariano Esquillor, s/n, 50018, Zaragoza, Spain. .,Department of Chemical and Environmental Engineering, University of Zaragoza, Spain, Campus Rio Ebro, C/ María de Luna, 3, 50018 Zaragoza, Spain.,Networking Res. Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
28
|
Zhao Y, Feng Y, Li J, Cui C, Wang A, Fang J, Zhang Y, Ye S, Mao Q, Wang X, Shi H. Endogenous ROS-Mediated Covalent Immobilization of Gold Nanoparticles in Mitochondria: A “Sharp Sword” in Tumor Radiotherapy. ACS Chem Biol 2022; 17:2355-2365. [DOI: 10.1021/acschembio.2c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yan Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yali Feng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Jiachen Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Chaoxiang Cui
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Jing Fang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Shuyue Ye
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Qiulian Mao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Xiaoyan Wang
- Department of Ultrasound, Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046000, P. R. China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
29
|
Hao Y, Peng B, Si C, Wang B, Luo C, Chen M, Luo C, Gong B, Li Z. PVP-Modified Multifunctional Bi 2WO 6 Nanosheets for Enhanced CT Imaging and Cancer Radiotherapy. ACS OMEGA 2022; 7:18795-18803. [PMID: 35694478 PMCID: PMC9178605 DOI: 10.1021/acsomega.2c01591] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
Malignant tumors are one of the main causes of human death. The clinical treatment of malignant tumors is usually surgery, chemotherapy, radiotherapy, and so forth. Radiotherapy, as a traditional and effective treatment method for cancer, is widely used in clinical practice, but the radiation resistance of tumor cells and the toxic side effects to normal cells are still the Achilles heel of radiotherapy. Multifunctional inorganic high-atom nanomaterials are expected to enhance the effect of tumor radiotherapy. Tungsten and bismuth, which contain elements with high atomic coefficients, have strong X-ray energy attenuation capability. We synthesized Bi2WO6 nanosheets (NSs) using a hydrothermal synthesis method and modified polyvinylpyrrolidone (PVP) on their surface to make them more stable. PVP-Bi2WO6 NSs have a variety of effects after absorbing X-rays (such as the photoelectric effect and Compton effect) and release a variety of particles such as photoelectrons, Compton electrons, auger electrons, and so forth, which can react with organic molecules or water in cells, generate a large number of free radicals, and promote cell apoptosis, thereby improving the effect of radiotherapy. We show through γ-H2AX and DCFH-DA probe analysis experiments that PVP-Bi2WO6 NSs can effectively increase cell DNA damage and reactive oxygen species formation under X-ray irradiation. Clone formation analysis showed that PVP-Bi2WO6 NSs can effectively suppress cell colony formation under X-ray irradiation. These versatile functions endow PVP-Bi2WO6 NSs with enhanced radiotherapy efficacy in animal models. In addition, PVP-Bi2WO6 NSs can also be used as contrast agents for X-ray computed tomography (CT) imaging with obvious effects. Therefore, PVP-Bi2WO6 NSs can be used as CT imaging contrast agents and tumor radiotherapy sensitizers and have potential medical applications.
Collapse
Affiliation(s)
- Yifan Hao
- Department
of Oral Radiology, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
- Jilin
Provincial Key Laboratory of Tooth Development and Bone Remodeling,
School and Hospital of Stomatology, Jilin
University, Changchun 130021, P. R. China
| | - Bo Peng
- Department
of Oral Radiology, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| | - Chao Si
- Department
of Oral Radiology, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
- Jilin
Provincial Key Laboratory of Tooth Development and Bone Remodeling,
School and Hospital of Stomatology, Jilin
University, Changchun 130021, P. R. China
| | - Bo Wang
- Department
of Oral Radiology, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| | - Chengfeng Luo
- Department
of Oral Radiology, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| | - Menghao Chen
- Department
of Oral Radiology, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| | - Cheng Luo
- Department
of Orthodontics, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| | - Baijuan Gong
- Department
of Orthodontics, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| | - Zhimin Li
- Department
of Oral Radiology, School of Stomatology, China Medical University, Shenyang 110002, P. R. China
| |
Collapse
|
30
|
Qi Y, Yu Z, Hu K, Wang D, Zhou T, Rao W. Rigid metal/liquid metal nanoparticles: Synthesis and application for locally ablative therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 42:102535. [PMID: 35181527 DOI: 10.1016/j.nano.2022.102535] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 12/15/2022]
Abstract
Locally ablative therapy, as the main therapy for advanced tumors, has fallen into a bottleneck in recent years. The breakthrough of metal nanoparticles provides a novel approach for ablative therapy. Previous studies have mostly focused on the combined field of rigid metal nanoparticles and ablation. However, with the maturity of the preparation process of liquid metal nanoparticles, liquid metal nanoparticles not only have metallic properties but also have fluid properties, showing the potential to be combined with ablation. At present, there is no review on the combination of liquid metal nanoparticles and ablation. In this article, we first review the preparation, characterization and application characteristics of rigid metal and liquid metal nanoparticles in ablation applications, and then summarize the advantages, disadvantages and possible future development trends of rigid and liquid metal nanoparticles.
Collapse
Affiliation(s)
- Yuxia Qi
- Beijing University of Chinese Medicine, Beijing, China.
| | - Zhongyang Yu
- Beijing University of Chinese Medicine, Beijing, China.
| | - Kaiwen Hu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing,, China.
| | - Dawei Wang
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, China; Beijing Key Laboratory of Cryo-Biomedical Engineering, Beijing, China.
| | - Tian Zhou
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing,, China.
| | - Wei Rao
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, China; Beijing Key Laboratory of Cryo-Biomedical Engineering, Beijing, China.
| |
Collapse
|
31
|
Niu N, Yu Y, Zhang Z, Kang M, Wang L, Zhao Z, Wang D, Tang BZ. A cell membrane-targeting AIE photosensitizer as a necroptosis inducer for boosting cancer theranostics. Chem Sci 2022; 13:5929-5937. [PMID: 35685806 PMCID: PMC9132078 DOI: 10.1039/d2sc01260j] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/12/2022] [Indexed: 11/21/2022] Open
Abstract
The exploration of cellular organelle-specific anchoring photosensitizers with both prominent fluorescence imaging behavior and extraordinary reactive oxygen species (ROS) production capability is highly in demand but remains a severe challenge for effective cancer theranostics involving photodynamic therapy (PDT). In this contribution, we developed a cell membrane-targeting and NIR-emission photosensitizer having an aggregation-induced emission (AIE) tendency. The AIE photosensitizer, namely TBMPEI, is capable of lighting up and ablating cancer cells by means of a necroptosis procedure enabling cell membrane rupture and DNA degradation upon light irradiation, endowing TBMPEI with impressive performance for both in vitro and in vivo fluorescence imaging-guided PDT.
Collapse
Affiliation(s)
- Niu Niu
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University Shenzhen 518060 China
- College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 China
| | - Ying Yu
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong Shenzhen Guangdong 518172 China
| | - Zhijun Zhang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University Shenzhen 518060 China
- College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 China
| | - Miaomiao Kang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University Shenzhen 518060 China
- College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 China
| | - Lei Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University Shenzhen 518060 China
- College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 China
| | - Zheng Zhao
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong Shenzhen Guangdong 518172 China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University Shenzhen 518060 China
- College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong Shenzhen Guangdong 518172 China
| |
Collapse
|
32
|
Sivasubramanian M, Lo LW. Assessment of Nanoparticle-Mediated Tumor Oxygen Modulation by Photoacoustic Imaging. BIOSENSORS 2022; 12:336. [PMID: 35624636 PMCID: PMC9138624 DOI: 10.3390/bios12050336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 06/01/2023]
Abstract
Photoacoustic imaging (PAI) is an invaluable tool in biomedical imaging, as it provides anatomical and functional information in real time. Its ability to image at clinically relevant depths with high spatial resolution using endogenous tissues as contrast agents constitutes its major advantage. One of the most important applications of PAI is to quantify tissue oxygen saturation by measuring the differential absorption characteristics of oxy and deoxy Hb. Consequently, PAI can be utilized to monitor tumor-related hypoxia, which is a crucial factor in tumor microenvironments that has a strong influence on tumor invasiveness. Reactive oxygen species (ROS)-based therapies, such as photodynamic therapy, radiotherapy, and sonodynamic therapy, are oxygen-consuming, and tumor hypoxia is detrimental to their efficacy. Therefore, a persistent demand exists for agents that can supply oxygen to tumors for better ROS-based therapeutic outcomes. Among the various strategies, NP-mediated supplemental tumor oxygenation is especially encouraging due to its physio-chemical, tumor targeting, and theranostic properties. Here, we focus on NP-based tumor oxygenation, which includes NP as oxygen carriers and oxygen-generating strategies to alleviate hypoxia monitored by PAI. The information obtained from quantitative tumor oxygenation by PAI not only supports optimal therapeutic design but also serves as a highly effective tool to predict therapeutic outcomes.
Collapse
Affiliation(s)
| | - Leu-Wei Lo
- Department of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 350, Taiwan;
| |
Collapse
|
33
|
Yang R, Wang P, Lou K, Dang Y, Tian H, Li Y, Gao Y, Huang W, Zhang Y, Liu X, Zhang G. Biodegradable Nanoprobe for NIR-II Fluorescence Image-Guided Surgery and Enhanced Breast Cancer Radiotherapy Efficacy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104728. [PMID: 35170876 PMCID: PMC9036023 DOI: 10.1002/advs.202104728] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Indexed: 05/19/2023]
Abstract
Positive resection margin frequently exists in breast-conserving treatment (BCT) of early-stage breast cancer, and insufficient therapeutic efficacy is common during radiotherapy (RT) in advanced breast cancer patients. Moreover, a multimodal nanotherapy platform is urgently required for precision cancer medicine. Therefore, a biodegradable cyclic RGD pentapeptide/hollow virus-like gadolinium (Gd)-based indocyanine green (R&HV-Gd@ICG) nanoprobe is developed to improve fluorescence image-guided surgery and breast cancer RT efficacy. R&HV-Gd exhibits remarkably improved aqueous stability, tumor retention, and target specificity of ICG, and achieves outstanding magnetic resonance/second near-infrared (NIR-II) window multimodal imaging in vivo. The nanoprobe-based NIR-II fluorescence image guidance facilitates complete tumor resection, improves the overall mouse survival rate, and effectively discriminates between benign and malignant breast tissues in spontaneous breast cancer transgenic mice (area under the curve = 0.978; 95% confidence interval: 0.952, 1.0). Moreover, introducing the nanoprobe to tumors generated more reactive oxygen species under X-ray irradiation, improved RT sensitivity, and reduced mouse tumor progression. Notably, the nanoprobe is biodegradable in vivo and exhibits accelerated bodily clearance, which is expected to reduce the potential long-term inorganic nanoparticle toxicity. Overall, the nanoprobe provides a basis for developing precision breast cancer treatment strategies.
Collapse
Affiliation(s)
- Rui‐Qin Yang
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| | - Pei‐Yuan Wang
- Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhouFujian350000China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouFujian350025China
| | - Kang‐Liang Lou
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| | - Yong‐Ying Dang
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| | - Hai‐Na Tian
- Department of BiomaterialsCollege of MaterialsResearch Center of Biomedical Engineering of Xiamen and Key Laboratory of Biomedical Engineering of Fujian Province and Fujian Provincial Key Laboratory for Soft Functional Materials ResearchXiamen UniversityXiamenFujian361005China
| | - Yang Li
- Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhouFujian350000China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouFujian350025China
| | - Yi‐Yang Gao
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| | - Wen‐He Huang
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| | - Yong‐Qu Zhang
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| | - Xiao‐Long Liu
- Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhouFujian350000China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouFujian350025China
| | - Guo‐Jun Zhang
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361100China
- Key Laboratory for Endocrine‐Related Cancer Precision Medicine of XiamenXiang'an Hospital of Xiamen UniversityXiamenFujian361100China
- Cancer Research CenterSchool of MedicineXiamen UniversityXiamenFujian361100China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid CancersXiamenFujian361100China
| |
Collapse
|
34
|
Ye M, Gao Y, Liang M, Qiu W, Ma X, Xu J, Hu J, Xue P, Kang Y, Xu Z. Microenvironment-responsive chemotherapeutic nanogels for enhancing tumor therapy via DNA damage and glutathione consumption. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.01.086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
35
|
Yun M, Yingzi L, Jie G, Guanxin L, Zimei Z, Zhen C, Zhi L, Yingjie N, Lunquan S, Tao C, Yuezhen D, Chengzhi Z. PPDPF Promotes the Progression and acts as an Antiapoptotic Protein in Non-Small Cell Lung Cancer. Int J Biol Sci 2022; 18:214-228. [PMID: 34975328 PMCID: PMC8692159 DOI: 10.7150/ijbs.65654] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023] Open
Abstract
Resistance to radiotherapy is frequently observed in the clinic and leads to poor prognosis of non-small cell lung cancer (NSCLC). How to overcome resistance to radiotherapy is a challenge in the treatment of NSCLC. In this study, PPDPF was found to be upregulated in NSCLC tissues and cell lines, and its expression negatively correlated with the overall survival of patients with NSCLC. PPDPF promoted the growth, colony formation and invasion of lung cancer cells. Moreover, knockout of PPDPF inhibited tumorigenesis in the KL (KrasG12D; LKB1f/f) mouse model of lung cancer. Additionally, overexpression of PPDPF led to radioresistance in lung cancer cells, and knockdown of PPDPF sensitized lung cancer cells to radiotherapy. Mechanistically, PPDPF interacted with BABAM2 (an antiapoptotic protein) and blocked its ubiquitination by MDM2, thus stabilizing BABAM2 and promoting the radioresistance of lung cancer cells. Our present study suggested PPDPF as a therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Mu Yun
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Li Yingzi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Gao Jie
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Liu Guanxin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Zeng Zimei
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Cao Zhen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Li Zhi
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Nie Yingjie
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital; Guiyang 550000, China
| | - Sun Lunquan
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Chen Tao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Deng Yuezhen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Zhou Chengzhi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
36
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, 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
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, 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, NUS Center for Nanomedicine, 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
|
37
|
Zhou M, Yuan M, Zhang M, Lei C, Aras O, Zhang X, An F. Combining histone deacetylase inhibitors (HDACis) with other therapies for cancer therapy. Eur J Med Chem 2021; 226:113825. [PMID: 34562854 DOI: 10.1016/j.ejmech.2021.113825] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022]
Abstract
Histone deacetylases (HDACs) play an important role in regulating the expression of genes involved in tumorigenesis and tumor maintenance, and hence they have been considered as key targets in cancer therapy. As a novel category of antitumor agents, histone deacetylase inhibitors (HDACis) can induce cell cycle arrest, apoptosis, and differentiation in cancer cells, ultimately combating cancer. Although in the United States, the use of HDACis for the treatment of certain cancers has been approved, the therapeutic efficacy of HDACis as a single therapeutic agent in solid tumorshas been unsatisfactory and drug resistance may yet occur. To enhance therapeutic efficacy and limit drug resistance, numerous combination therapies involving HDACis in synergy with other antitumor therapies have been studied. In this review, we describe the classification of HDACs. Moreover, we summarize the antitumor mechanism of the HDACis for targeting key cellular processes of cancers (cell cycle, apoptosis, angiogenesis, DNA repair, and immune response). In addition, we outline the major developments of other antitumor therapies in combination with HDACis, including chemotherapy, radiotherapy, phototherapy, targeted therapy, and immunotherapy. Finally, we discuss the current state and challenges of HDACis-drugs combinations in future clinical studies, with the aim of optimizing the antitumor effect of such combinations.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Minjian Yuan
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Meng Zhang
- 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, People's Republic of China
| | - Chenyi Lei
- 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, People's Republic of China
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, United States
| | - Xiaohong Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, PR 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, People's Republic of China; Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, PR China.
| |
Collapse
|
38
|
Xu Q, Zhang H, Liu H, Han Y, Qiu W, Li Z. Inhibiting autophagy flux and DNA repair of tumor cells to boost radiotherapy of orthotopic glioblastoma. Biomaterials 2021; 280:121287. [PMID: 34864449 DOI: 10.1016/j.biomaterials.2021.121287] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/25/2021] [Indexed: 12/27/2022]
Abstract
Radio-resistance of glioblastoma (GBM) remains a leading cause of radiotherapy failure because of the protective autophagy induced by X-Ray irradiation and tumor cells' strong capability of repairing damaged DNA. It is of great importance to overcome the radio-resistance for improving the efficacy of radiotherapy. Herein, we report the novel mechanism of core-shell copper selenide coated gold nanoparticles (Au@Cu2-xSe NPs) inhibiting the protective autophagy and DNA repair of tumor cells to drastically boost the radiotherapy efficacy of glioblastoma. We reveal that the core-shell Au@Cu2-xSe NPs can inhibit the autophagy flux by effectively alkalizing lysosomes. They can increase the SQSTM1/p62 protein levels of tumor cells without influencing their mRNA. We also reveal that Au@Cu2-xSe NPs can increase the ubiquitination of DNA repair protein Rad51, and promote the degradation of Rad51 by proteasomes to prevent the DNA repair. The simultaneous inhibition of protective autophagy and DNA repair significantly suppress the growth of orthotopic GBM by using radiotherapy and our novel Au@Cu2-xSe NPs. Our work provides a new insight and paradigm to significantly improve the efficacy of radiotherapy by rationally designing theranostic nano-agents to simultaneously inhibit protective autophagy and DNA repair of tumor cells.
Collapse
Affiliation(s)
- Qi Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China; College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P.R. China.
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China
| | - Weibao Qiu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China.
| |
Collapse
|
39
|
Zhu Z, Wu M, Sun J, Huangfu Z, Yin L, Yong W, Sun J, Wang G, Meng F, Zhong Z. Redox-sensitive iodinated polymersomes carrying histone deacetylase inhibitor as a dual-functional nano-radiosensitizer for enhanced radiotherapy of breast cancer. Drug Deliv 2021; 28:2301-2309. [PMID: 34730060 PMCID: PMC8567935 DOI: 10.1080/10717544.2021.1995080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 01/14/2023] Open
Abstract
Radiotherapy (RT) is a frequently used means in clinical tumor treatment. The outcome of RT varies, however, to a great extent, due to RT resistance or intolerable dose, which might be resolved by the development of radio-sensitizing strategies. Here, we report redox-sensitive iodinated polymersomes (RIP) carrying histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA, vorinostat), as a new dual-functional nano-radiosensitizer for breast cancer radiotherapy. SAHA-loaded RIP (RIP-SAHA) with a size of about 101 nm exhibited good colloidal stability while the reduction-activated release of SAHA, giving rise to better antitumor effect to 4T1 breast carcinoma cells than free SAHA. Accordingly, RIP-SAHA combined with a 4 Gy dose of X-ray radiation led to significantly enhanced suppression of 4T1 cells compared with SAHA combined 4 Gy of X-ray radiation, as a result of enhanced DNA damage and impeded DNA damage repair. The pharmacokinetics and biodistribution studies by single-photon emission computed tomography (SPECT) with 125I-labeled SAHA (125I-SAHA) showed a 17.3-fold longer circulation and 237.7-fold better tumor accumulation of RIP-SAHA over SAHA. The systemic administration of RIP-SAHA greatly sensitized radiotherapy of subcutaneous 4T1 breast tumors and brought about significant inhibition of tumor growth, without causing damages to major organs, compared with radiotherapy alone. RIP not only enhanced SAHA delivery but also acted as a radiosensitizer. RIP-SAHA emerges as a smart dual-functional nano-radiosensitizer to effectively enhance tumor radiotherapy.
Collapse
Affiliation(s)
- Zhehong Zhu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Manran Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Juan Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Zhengyuan Huangfu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Lingling Yin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Weipeng Yong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jing Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
40
|
Dou Y, Zhao F, Li X, Guo Y. Monitoring Nitric Oxide-Induced Hypoxic Tumor Radiosensitization by Radiation-Activated Nanoagents under BOLD/DWI Imaging. ACS Biomater Sci Eng 2021; 7:5242-5254. [PMID: 34612040 DOI: 10.1021/acsbiomaterials.1c00543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tumor heterogeneity leads to unpredictable radiotherapeutic outcomes although multiple sensitization strategies have been developed. Real-time monitoring of treatment response through noninvasive imaging methods is critical and a great challenge in optimizing radiotherapy. Herein, we propose a combined functional magnetic resonance imaging approach (blood-oxygen-level-dependent/diffusion-weighted (BOLD/DWI) imaging) for monitoring tumor response to nitric oxide (NO)-induced hypoxic radiosensitization achieved by radiation-activated nanoagents (NSC@SiO2-SNO NPs). This nanoagent carrying NO donors can efficiently concentrate in tumors and specifically produce high concentrations of NO under radiation. In vitro and in vivo studies show that this nanoagent can effectively reduce tumor hypoxia, promote radiation-induced apoptosis and DNA damage under hypoxia, and ultimately inhibit tumor growth. In vivo BOLD/DWI imaging enables noninvasive monitoring of improvements in tumor oxygen levels and radiosensitivity during treatment with this nanostrategy by quantifying functional parameters. This work demonstrates that BOLD/DWI imaging is a useful tool for evaluating tumor response and monitoring the effectiveness of radiotherapeutic strategies aimed at improving hypoxia, with great clinical potential.
Collapse
Affiliation(s)
- Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Fangshi Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Xue Li
- Department of Radiology and Department of Radiation Oncology, Tianjin Medical University Second Hospital, Tianjin 300211, P. R. China
| | - Yanyan Guo
- Department of Radiology and Department of Radiation Oncology, Tianjin Medical University Second Hospital, Tianjin 300211, P. R. China
| |
Collapse
|
41
|
Bhardwaj P, Goda JS, Pai V, Chaudhari P, Mohanty B, Pai T, Vishwakarma K, Thorat R, Wadasadawala T, Banerjee R. Ultrasound augments on-demand breast tumor radiosensitization and apoptosis through a tri-responsive combinatorial delivery theranostic platform. NANOSCALE 2021; 13:17077-17092. [PMID: 34622906 DOI: 10.1039/d1nr04211d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Advanced inoperable triple-negative breast cancer (TNBC) comprises aggressive tumors with a modest pathological response to neoadjuvant chemotherapy. The concomitant use of chemoradiotherapy improves the pathological response rates. However, the dose-dependent systemic toxicity of clinical radiosensitizers with poor circulation half-life and limited passive bioavailability limits their clinical utility. We address these challenges by rationally designing a stealth and tumor microenvironment responsive nano-conjugate platform for the ultrasound-mediated on-demand spatio-temporal delivery of plant flavonoid curcumin as a combinatorial regimen with clinically approved paclitaxel for the neoadjuvant chemoradiotherapy of locally advanced triple-negative breast cancer (TNBC). Interestingly, the focused application of ultrasound at the orthotopic TNBC xenograft of NOD-SCID mice facilitated the immediate infiltration of nano-conjugates at the tumor interstitium, and conferred in vivo safety over marketed paclitaxel formulation. In addition, curcumin significantly potentiated the in vivo chemoradiotherapeutic efficacy of paclitaxel upon loading into nano-conjugates. This gets further enhanced by the concurrent pulse of ultrasound, as confirmed by PET-CT imaging, along with a significant improvement in the mice survival. The quadrapeutic apoptotic effect by the combination of paclitaxel, curcumin, radiation, and ultrasound, along with a reduction in the tumor microvessel density and cell proliferation marker, confers the broad chemo-radiotherapeutic potential of this regimen for radio-responsive solid tumors, as well as metastatic niches.
Collapse
Affiliation(s)
- Prateek Bhardwaj
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India.
| | - Jayant Sastri Goda
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Venkatesh Pai
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Pradip Chaudhari
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Bhabani Mohanty
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Trupti Pai
- Department of Pathology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Komal Vishwakarma
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Rahul Thorat
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Tabassum Wadasadawala
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India.
| |
Collapse
|
42
|
Veroniaina H, Wu Z, Qi X. Innate tumor-targeted nanozyme overcoming tumor hypoxia for cancer theranostic use. J Adv Res 2021; 33:201-213. [PMID: 34603790 PMCID: PMC8463960 DOI: 10.1016/j.jare.2021.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 01/11/2023] Open
Abstract
Introduction Hypoxic tumor microenvironment (TME) is the major contributor to cancer metastasis, resistance to chemotherapy, and recurrence of tumors. So far, no approved treatment has been available to overcome tumor hypoxia. Objectives The present study aimed to relieve tumor hypoxia via a nanozyme theranostic nanomaterial as well as providing magnetic resonance imaging (MRI)-guided therapy. Methods Manganese dioxide (MnO2) was used for its intrinsic enzymatic activity co-loaded with the anti-cancer drug Doxorubicin (Dox) within the recombinant heavy-chain apoferritin cavity to form MnO2-Dox@HFn. Following the synthesis of the nanomaterial, different characterizations were performed as well as its nanozyme-like ability. This nanoplatform recognizes tumor cells through the transferrin receptors 1 (TfR1) which are highly expressed on the surface of most cancer cells. The cellular uptake was confirmed by flow cytometry and fluorescence spectroscopy. In vitro and in vivo studies have been investigated to evaluate the hypoxia regulation, MRI ability and anti-tumor activity of MnO2-Dox@HFn. Results Being a TME-responsive nanomaterial, MnO2-Dox@HFn exerted both peroxidase and catalase activity that mainly produce massive oxygen and Mn2+ ions. Respectively, these products relieve the unfavorable tumor hypoxia and also exhibit T1-weighted MRI with a high longitudinal relaxivity of 33.40 mM. s−1. The utility of MnO2-Dox@HFn was broadened with their efficient anti-cancer activity proved both in vitro and in vivo. Conclusions MnO2-Dox@HFn successfully overcome tumor hypoxia with double potentials enzymatic ability and diagnostic capacity. This investigation could ignite the future application for cancer theranostic nanozyme therapy.
Collapse
Affiliation(s)
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
43
|
Kayani Z, Islami N, Behzadpour N, Zahraie N, Imanlou S, Tamaddon P, Salehi F, Daneshvar F, Perota G, Sorati E, Mohammadi S, Sattarahmady N. Combating cancer by utilizing noble metallic nanostructures in combination with laser photothermal and X-ray radiotherapy. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
44
|
Li L, Cao L, Xiang X, Wu X, Ma L, Chen F, Cao S, Cheng C, Deng D, Qiu L. ROS‐Catalytic Transition‐Metal‐Based Enzymatic Nanoagents for Tumor and Bacterial Eradication. ADVANCED FUNCTIONAL MATERIALS 2021. [DOI: 10.1002/adfm.202107530] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Ling Li
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
- Department of Ultrasound Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Lijian Cao
- Department of Pharmaceutical Engineering School of Engineering China Pharmaceutical University Nanjing 211198 P. R. China
| | - Xi Xiang
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
| | - Xizheng Wu
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
| | - Lang Ma
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
- Department of Chemistry and Biochemistry Freie Universität Berlin Takustrasse 3 14195 Berlin Germany
| | - Fan Chen
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
| | - Sujiao Cao
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
| | - Chong Cheng
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
- Department of Chemistry and Biochemistry Freie Universität Berlin Takustrasse 3 14195 Berlin Germany
- State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 China
| | - Dawei Deng
- Department of Pharmaceutical Engineering School of Engineering China Pharmaceutical University Nanjing 211198 P. R. China
| | - Li Qiu
- Department of Ultrasound National Clinical Research Center for Geriatrics West China Hospital College of Polymer Science and Engineering Sichuan University Chengdu 610041 China
| |
Collapse
|
45
|
Wang X, Niu X, Sha W, Feng X, Yu L, Zhang Z, Wang W, Yuan Z. An oxidation responsive nano-radiosensitizer increases radiotherapy efficacy by remolding tumor vasculature. Biomater Sci 2021; 9:6308-6324. [PMID: 34519724 DOI: 10.1039/d1bm00834j] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
As an excellent candidate material for nano-sensitizers, gold nanostructures have shown great potential in radiotherapy. Nevertheless, severe hypoxia and low accumulation of nanomedicine caused by poor perfusion at the tumor site have significantly reduced radiotherapy efficacy. Vascular normalization has gained attention owing to its ability to relieve hypoxia and increase perfusion. The synergistic therapy of tumor vascular normalization and radiotherapy has become a new option to increase anti-cancer efficacy. However, the commonly used strategy of suppressing a single growth factor to induce vascular normalization is limited by tumor compensatory effects. In this work, we developed a strategy to inhibit oxidative stress in tumors by generating chelating agents in response to hydrogen peroxide, thereby inhibiting multi-angiogenic factors simultaneously to normalize blood vessels. Concretely, sodium alginate (SA) reacted with 8-quinoline boric acid (QBA) to form SA-QBA. Then gold nanoparticles (Au NPs) were modified with SA-QBA to obtain Au@SA-QBA. The system was simple in structure and could generate 8HQ in response to H2O2in vitro to inhibit oxidative stress and reduce the expression of VEGF, bFGF, and Ang-2. In vivo, the perfusion unit (PU) increased by 78% after Au@SA-QBA treatment, and the coverage of pericytes increased by 32%, which in turn induced vascular normalization. In addition, blood routine and blood biochemical tests confirmed its good biocompatibility and 8HQ was not detected in the supernatant after homogenization of major organs. More importantly, after the synergistic treatment of vascular normalization and radiotherapy (4 Gy), the tumor growth inhibition rate was increased by 38.6% compared to the Au@SA-treated group with negligible side effects to normal tissues.
Collapse
Affiliation(s)
- Xiaohui Wang
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Xiaoyan Niu
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Weizhou Sha
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Xiaoyue Feng
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Licheng Yu
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Zhenjie Zhang
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Wei Wang
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Zhi Yuan
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| |
Collapse
|
46
|
Liu Y, Li X, Shi Y, Wang Y, Zhao X, Gong X, Cai R, Song G, Chen M, Zhang X. Two-dimensional intermetallic PtBi/Pt core/shell nanoplates overcome tumor hypoxia for enhanced cancer therapy. NANOSCALE 2021; 13:14245-14253. [PMID: 34477707 DOI: 10.1039/d1nr02561a] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The design of multifunctional nanoplatforms is of great importance for improving hypoxia-induced therapeutic outcomes, especially for overcoming radiotherapy (RT) tolerance. Here, two-dimensional intermetallic PtBi/Pt nanoplates (PtBi NPs) were designed as a therapeutic platform to in situ generate oxygen, and thereby overcome tumor hypoxia for boosting photothermal/radiotherapy (PTT/RT). With high X-ray attenuation coefficient, PtBi NPs exhibited outstanding radiotherapy sensitization characteristics. Moreover, the high photothermal effect of PtBi NPs could promote the catalytic activity of PtBi NPs to achieve a synergistic PTT/RT effect. PEGylated PtBi NPs (PtBi-PEG) exhibited excellent biocompatibility, prolonged blood circulation time and enhanced tumor accumulation. Finally, PtBi-PEG showed excellent trimodal contrast enhancement for infrared (IR) imaging, photoacoustic (PA) imaging and X-ray imaging, facilitating imaging-guided cancer therapy. Thus, our work highlights PtBi-PEG as a novel multifunctional theranostic nanoplatform with great potential for future multimodal imaging-guided synergistic cancer therapy.
Collapse
Affiliation(s)
- Yongchun Liu
- College of Materials Science and Engineering, Hunan University, Changsha 410082, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yu Y, Feng Z, Liu J, Hou X, Zhou X, Gao J, Wang W, Zhang Y, Li G, Liu J. γ-Ray-Triggered Drug Release of Reactive Oxygen Species-Sensitive Nanomedicine for Enhanced Concurrent Chemoradiation Therapy. ACS OMEGA 2021; 6:19445-19457. [PMID: 34368532 PMCID: PMC8340104 DOI: 10.1021/acsomega.1c01500] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Radiotherapy (RT) combined with chemotherapy remains a dominant therapeutic manner in clinical tumor treatment, which is irreplaceable in a short term. To seek an intrinsic connection of combined chemoradiation therapy and maximize the antitumor efficacy, we developed a reactive oxygen species (ROS)-sensitive nanomicelle drug delivery system based on a self-assembled amphiphilic polymer, hyaluronic acid-graft-poly-(propylene sulfide) (HA-PPS). A chemical radiosensitizer, doxorubicin (DOX), was encapsulated into the core of HA-PPS nanomicelles, constituting the DOX-loaded nanomicelles (HA-PPS@DOX NMs) with a spherical structure of around 205.10 ± 11.33 nm diameter with a narrow polydispersity index (PDI) of 0.135 ± 0.01. When combined with RT, the ROS-sensitive HA-PPS@DOX NMs disintegrated and released great drug cargos, which further enhanced cytotoxicity. Meanwhile, as a radiosensitizer, the released DOX sensitized cancer cells to radiotherapy, which has been confirmed by an enhanced sensitizer enhancement ratio (SER) value of 1.78 contributing to the increased cytotoxicity of concurrent chemoradiation tumor therapy, as evidenced by the improvement of half maximal inhibitory concentration (IC50 value) of DOX from 2.316 to 0.8235 μg/mL. Moreover, in vivo studies revealed that HA-PPS@DOX NMs exhibited prolonged circulation time and improved tumor accumulation. Particularly, the released DOX triggered by radiation strengthened radiotherapy sensitization in return. Consequently, these superiorities of HA-PPS@DOX NMs shown by the concurrent chemoradiation tumor therapy resulted in an ideal tumor inhibition rate of 70.4%, thus providing a promising ROS-sensitive nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Ying Yu
- Lab
of Functional and Biomedical Nanomaterials, College of Materials Science
and Engineering, Qingdao University of Science
and Technology, Qingdao 266042, China
| | - Zujian Feng
- Department
of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering
(Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Jinjian Liu
- Tianjin
Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine,
Institute of Radiation Medicine, Chinese
Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Xiaoxue Hou
- Tianjin
Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine,
Institute of Radiation Medicine, Chinese
Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Xiaoqian Zhou
- Lab
of Functional and Biomedical Nanomaterials, College of Materials Science
and Engineering, Qingdao University of Science
and Technology, Qingdao 266042, China
| | - Jie Gao
- Tianjin
Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine,
Institute of Radiation Medicine, Chinese
Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wei Wang
- Lab
of Functional and Biomedical Nanomaterials, College of Materials Science
and Engineering, Qingdao University of Science
and Technology, Qingdao 266042, China
| | - Yumin Zhang
- Tianjin
Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine,
Institute of Radiation Medicine, Chinese
Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Guoliang Li
- Lab
of Functional and Biomedical Nanomaterials, College of Materials Science
and Engineering, Qingdao University of Science
and Technology, Qingdao 266042, China
| | - Jianfeng Liu
- Lab
of Functional and Biomedical Nanomaterials, College of Materials Science
and Engineering, Qingdao University of Science
and Technology, Qingdao 266042, China
- Tianjin
Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine,
Institute of Radiation Medicine, Chinese
Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
48
|
Ma YC, Tang XF, Xu YC, Jiang W, Xin YJ, Zhao W, He X, Lu LG, Zhan MX. Nano-enabled coordination platform of bismuth nitrate and cisplatin prodrug potentiates cancer chemoradiotherapy via DNA damage enhancement. Biomater Sci 2021; 9:3401-3409. [PMID: 33949448 DOI: 10.1039/d1bm00157d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The combination of chemotherapy and radiotherapy (chemoradiotherapy) is a promising strategy, extensively studied and applied clinically. Meanwhile, radiosensitizers play an important role in improving clinical radiotherapy therapeutic efficacy. There are still some disadvantages in practical applications, because radiosensitizers and drugs are difficult to deliver spatio-temporally to tumor sites and work simultaneously with low efficiency for DNA damage and repair inhibition, leading to an inferior synergistic effect. Herein, a suitable radiosensitizer of nano-enabled coordination platform (NP@PVP) with bismuth nitrate and cisplatin prodrug is developed by a simple synthetic route to improve the effectiveness of chemo-radiation synergistic therapy. When NP@PVP is internalized by a tumor cell, the bismuth in NP@PVP can sensitize radiation therapy (RT) by increasing the amount of reactive oxygen species generation to enhance DNA damage after X-ray radiation; meanwhile, the cisplatin in NP@PVP can inhibit DNA damage repair with spatio-temporal synchronization. NP@PVP is demonstrated to exhibit higher sensitization enhancement ratio (SER) of 2.29 and excellent tumor ablation capability upon irradiation in vivo in comparison with cisplatin (SER of 1.78). Our strategy demonstrates that the RT sensitization effect of bismuth and cisplatin based NP@PVP has great anticancer potential in chemo-radiation synergistic therapy, which is promising for clinical application.
Collapse
Affiliation(s)
- Yin-Chu Ma
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| | - Xin-Feng Tang
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - You-Cui Xu
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Wei Jiang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| | - Yong-Jie Xin
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| | - Wei Zhao
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| | - Xu He
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| | - Li-Gong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| | - Mei-Xiao Zhan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai 519000, China.
| |
Collapse
|
49
|
Yun B, Zhu H, Yuan J, Sun Q, Li Z. Synthesis, modification and bioapplications of nanoscale copper chalcogenides. J Mater Chem B 2021; 8:4778-4812. [PMID: 32226981 DOI: 10.1039/d0tb00182a] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Copper chalcogenides have a simple general formula, variable atomic ratios, and complicated crystal structures, which lead to their wealth of optical, electrical, and magnetic properties with great potential for wide applications ranging from energy conversion to the biomedical field. Herein, we summarize the recent advances in (1) the synthesis of size- and morphology tunable nanostructures by different methods; (2) surface modification and functionalization for different purposes; and (3) bioapplications for diagnosis and treatment of tumors by different imaging and therapy methods, as well as antibacterial applications. We also briefly discuss the future directions and challenges of copper chalcogenide nanoparticles in the biomedical field.
Collapse
Affiliation(s)
- Baofeng Yun
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China.
| | - Hongqin Zhu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China.
| | - Jiaxin Yuan
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China.
| | - Qiao Sun
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China.
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China.
| |
Collapse
|
50
|
Chen Q, Sun T, Jiang C. Recent Advancements in Nanomedicine for 'Cold' Tumor Immunotherapy. NANO-MICRO LETTERS 2021; 13:92. [PMID: 34138315 PMCID: PMC8006526 DOI: 10.1007/s40820-021-00622-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/31/2021] [Indexed: 05/02/2023]
Abstract
Although current anticancer immunotherapies using immune checkpoint inhibitors (ICIs) have been reported with a high clinical success rate, numerous patients still bear 'cold' tumors with insufficient T cell infiltration and low immunogenicity, responding poorly to ICI therapy. Considering the advancements in precision medicine, in-depth mechanism studies on the tumor immune microenvironment (TIME) among cold tumors are required to improve the treatment for these patients. Nanomedicine has emerged as a promising drug delivery system in anticancer immunotherapy, activates immune function, modulates the TIME, and has been applied in combination with other anticancer therapeutic strategies. This review initially summarizes the mechanisms underlying immunosuppressive TIME in cold tumors and addresses the recent advancements in nanotechnology for cold TIME reversal-based therapies, as well as a brief talk about the feasibility of clinical translation.
Collapse
Affiliation(s)
- Qinjun Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education), State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmaceutics, and School of Pharmacy, Research Center on Aging and Medicine, Fudan University, Shanghai, 201203, People's Republic of China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education), State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmaceutics, and School of Pharmacy, Research Center on Aging and Medicine, Fudan University, Shanghai, 201203, People's Republic of China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmaceutics, and School of Pharmacy, Research Center on Aging and Medicine, Fudan University, Shanghai, 201203, People's Republic of China.
| |
Collapse
|