1
|
Lin Z, Zhu T, Zhong X. NIR-triggered NO production combined with photodynamic therapy for tumor treatment. Photodiagnosis Photodyn Ther 2024; 49:104241. [PMID: 39168068 DOI: 10.1016/j.pdpdt.2024.104241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 08/23/2024]
Abstract
Photodynamic therapy (PDT), as one of the most promising cancer therapy methods, is still limited by several drawbacks, such as tissue hypoxia and shallow light penetration of blue-violet light (200-450 nm), and red light (750 nm) is more penetrating to tissues than blue-violet light, but still lower than near-red light (750-1350 nm). Therefore, we proposed a synergistic therapy system by combining the near-infrared light-triggered PDT with nitric oxide (NO)-based gas therapy to enhance the anti-tumor effects. Upconversion nanoparticles (UCNPs) were loaded with the photosensitizers of ZnPc and the NO donors of l-arginine (L-Arg) to obtain the nanocomposites of UCN@mSiO2@ZnPc@L-Arg. Under 980 nm laser irradiation, reactive oxygen species (ROS) could be produced for PDT and react with l-Arg to produce NO, which is previously reported to have a greater killing effect on tumor cells than ROS and also plays an important role in promoting PDT in our study. Both the in vitro and in vivo tests demonstrated that the combined therapy of PDT with NO therapy could enhance the tumor killing effect significantly compared with the unique application of PDT. The UCNPs-based nanocomposites are expected to be widely used in biomedicine for tumor inhibition.
Collapse
Affiliation(s)
- Zhiyuan Lin
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Tao Zhu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Xiaoqin Zhong
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
2
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
3
|
Ye Z, Liu J, Liu Y, Zhao Y, Li Z, Xu B, Chen D, Wang B, Wang Q, Shen Y. Hybrid nanopotentiators with dual cascade amplification for glioma combined interventional therapy. J Control Release 2024; 372:95-112. [PMID: 38851536 DOI: 10.1016/j.jconrel.2024.06.016] [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: 09/24/2023] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Glioma is an aggressive malignant brain tumor with a very poor prognosis for survival. The poor tumor targeting efficiency and tumor microenvironment penetration barrier also as troubles inhibited the effective glioma chemotherapy. Here, we design a core-shell structure cascade amplified hybrid catalytic nanopotentiators CFpAD with DM1 encapsulated to overcome the glioma therapeutic obstacles. NIR laser-based BBB penetrating enhances the tumor accumulation of CFpAD. When CFpAD, as the cascade amplified drug, is treated on the cancer cells, the bomb-like CFpAD releases gold nanoparticles as glucose oxidase (GOx) and ferric oxide nanoparticles (FNPs) as peroxides (POx) after blasting, producing ROS via a cascade amplification for tumor cell apoptosis. Gold nanoparticles can rest CAFs and reduce ECM secretion, achieving deep penetration of CFpAD. Moreover, CFpAD also cuts off the nutritional supply of the tumor, reduces the pH value, and releases free radicals to destroy the cancer. The glioma cell viability was significantly decreased through DNA damage and ROS aggregation due to the DM1-based chemotherapy synergistically combined with interventional photothermal therapy (IPTT) and radiotherapy (RT). This domino cascade amplified loop, combined with starvation therapy with IPTT and RT, has good tumor penetration and outstanding antitumor efficacy, and is a promising glioma treatment system.
Collapse
Affiliation(s)
- Zixuan Ye
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ji Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yanyan Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yan Zhao
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Zhen Li
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Bohui Xu
- School of Pharmacy, Nantong University, No.19 Qixiu Road, Nantong 226001,China
| | - Daquan Chen
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
| | - Buhai Wang
- Cancer Institute of Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, China.
| | - Qiyue Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China.
| | - Yan Shen
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
4
|
Lin Y, Yang Y, Ren X, Liu Z. NIR-Mediated Pyroelectric Catalysis for Sustained ROS/RNS Generation and Advanced Cancer Therapy In Vivo via Injectable Hydrogel. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38942-38955. [PMID: 39039973 DOI: 10.1021/acsami.4c05836] [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: 07/24/2024]
Abstract
Exogenous electrical stimulation has attracted considerable attention due to the advantages of microelectric induction and subsequent biological effects such as actin reorganization and reactive oxygen species (ROS) generation. Herein, an injectable hydrogel of BPR-ARG@Gel (BAG) with pyroelectric BPR nanoparticle loading and l-arginine (ARG) introduction was fabricated for advanced cancer therapy in vivo. Due to the photothermal effect, the holes and electrons in BPR nanoparticles were separated to produce an open-circuit voltage and consequently catalyze water H2O to generate toxic superoxide (•O2-) and hydroxyl radicals (•OH). These ROS substances further oxidize ARG to produce NO for synergistic tumor treatments. The mice experiments indicated that the employment of BAG hydrogel incorporation with a near-infrared laser downregulated the heat shock protein and recruited immune cells with 5-fold-enhanced expression of proinflammatory cytokines of interferon-γ. It was also noteworthy that the injectable hydrogel of BAG substantially induced the generation of reactive oxygen/nitrogen species (ROS/RNS) with reliable biosafety and strong tumor inhibition. Overall, these findings have provided potentially new inspirations and a feasible strategy to translate this multifunctional hydrogel toward tumor therapy in a pyroelectric stimulation manner.
Collapse
Affiliation(s)
- Yandai Lin
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China
| | - Yanxi Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China
| | - Xueli Ren
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China
| | - Zhe Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China
| |
Collapse
|
5
|
Hu X, Hu J, Pang Y, Wang M, Zhou W, Xie X, Zhu C, Wang X, Sun X. Application of nano-radiosensitizers in non-small cell lung cancer. Front Oncol 2024; 14:1372780. [PMID: 38646428 PMCID: PMC11027897 DOI: 10.3389/fonc.2024.1372780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 04/23/2024] Open
Abstract
Radiotherapy stands as a cornerstone in the treatment of numerous malignant tumors, including non-small cell lung cancer. However, the critical challenge of amplifying the tumoricidal effectiveness of radiotherapy while minimizing collateral damage to healthy tissues remains an area of significant research interest. Radiosensitizers, by methods such as amplifying DNA damage and fostering the creation of free radicals, play a pivotal role in enhancing the destructive impact of radiotherapy on tumors. Over recent decades, nano-dimensional radiosensitizers have emerged as a notable advancement. Their mechanisms include cell cycle arrest in the G2/M phase, combating tumor hypoxia, and others, thereby enhancing the efficacy of radiotherapy. This review delves into the evolving landscape of nanomaterials used for radiosensitization in non-small cell lung cancer. It provides insights into the current research progress and critically examines the challenges and future prospects within this burgeoning field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaonan Sun
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Xu X, Zhang J, Wang T, Li J, Rong Y, Wang Y, Bai C, Yan Q, Ran X, Wang Y, Zhang T, Sun J, Jiang Q. Emerging non-antibody‒drug conjugates (non-ADCs) therapeutics of toxins for cancer treatment. Acta Pharm Sin B 2024; 14:1542-1559. [PMID: 38572098 PMCID: PMC10985036 DOI: 10.1016/j.apsb.2023.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/23/2023] [Indexed: 04/05/2024] Open
Abstract
The non-selective cytotoxicity of toxins limits the clinical relevance of the toxins. In recent years, toxins have been widely used as warheads for antibody‒drug conjugates (ADCs) due to their efficient killing activity against various cancer cells. Although ADCs confer certain targeting properties to the toxins, low drug loading capacity, possible immunogenicity, and other drawbacks also limit the potential application of ADCs. Recently, non-ADC delivery strategies for toxins have been extensively investigated. To further understand the application of toxins in anti-tumor, this paper provided an overview of prodrugs, nanodrug delivery systems, and biomimetic drug delivery systems. In addition, toxins and their combination strategies with other therapies were discussed. Finally, the prospect and challenge of toxins in cancer treatment were also summarized.
Collapse
Affiliation(s)
- Xiaolan Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiaming Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tao Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yukang Rong
- School of Education, University of Nottingham, Nottingham NG7 2RD, UK
| | - Yanfang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenxia Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing Yan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaohua Ran
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingli Wang
- Department of Pharmacy, Linyi People's Hospital, Shandong University, Linyi 276000, China
| | - Tianhong Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qikun Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Wang R, Cheng L, He L, Du C, Wang H, Peng B, Yu X, Liu W, Luo W, Ran H, Yang L. Nitric oxide nano-reactor DNMF/PLGA enables tumor vascular microenvironment and chemo-hyperthermia synergetic therapy. J Nanobiotechnology 2024; 22:110. [PMID: 38481281 PMCID: PMC10938667 DOI: 10.1186/s12951-024-02366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/22/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Breast cancer ranks first among malignant tumors, of which triple-negative breast cancer (TNBC) is characterized by its highly invasive behavior and the worst prognosis. Timely diagnosis and precise treatment of TNBC are substantially challenging. Abnormal tumor vessels play a crucial role in TNBC progression and treatment. Nitric oxide (NO) regulates angiogenesis and maintains vascular homeostasis, while effective NO delivery can normalize the tumor vasculature. Accordingly, we have proposed here a tumor vascular microenvironment remodeling strategy based on NO-induced vessel normalization and extracellular matrix collagen degradation with multimodality imaging-guided nanoparticles against TNBC called DNMF/PLGA. RESULTS Nanoparticles were synthesized using a chemotherapeutic agent doxorubicin (DOX), a NO donor L-arginine (L-Arg), ultrasmall spinel ferrites (MnFe2O4), and a poly (lactic-co-glycolic acid) (PLGA) shell. Nanoparticle distribution in the tumor was accurately monitored in real-time through highly enhanced magnetic resonance imaging and photoacoustic imaging. Near-infrared irradiation of tumor cells revealed that MnFe2O4 catalyzes the production of a large amount of reactive oxygen species (ROS) from H2O2, resulting in a cascade catalysis of L-Arg to trigger NO production in the presence of ROS. In addition, DOX activates niacinamide adenine dinucleotide phosphate oxidase to generate and supply H2O2. The generated NO improves the vascular endothelial cell integrity and pericellular contractility to promote vessel normalization and induces the activation of endogenous matrix metalloproteinases (mainly MMP-1 and MMP-2) so as to promote extravascular collagen degradation, thereby providing an auxiliary mechanism for efficient nanoparticle delivery and DOX penetration. Moreover, the chemotherapeutic effect of DOX and the photothermal effect of MnFe2O4 served as a chemo-hyperthermia synergistic therapy against TNBC. CONCLUSION The two therapeutic mechanisms, along with an auxiliary mechanism, were perfectly combined to enhance the therapeutic effects. Briefly, multimodality image-guided nanoparticles provide a reliable strategy for the potential application in the fight against TNBC.
Collapse
Affiliation(s)
- Ruoyao Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Long Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Lingyun He
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Chier Du
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Haiyang Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Bohao Peng
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Xiaoqing Yu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Weiwei Liu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Wenpei Luo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Lu Yang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
8
|
Natangelo S, Trapani D, Koukoutzeli C, Boscolo Bielo L, Marvaso G, Jereczek-Fossa BA, Curigliano G. Radiation therapy, tissue radiosensitization, and potential synergism in the era of novel antibody-drug conjugates. Crit Rev Oncol Hematol 2024; 195:104270. [PMID: 38272150 DOI: 10.1016/j.critrevonc.2024.104270] [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: 09/14/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Antibody-drug conjugates (ADCs) represent a therapeutic class of agents designed to selectively deliver cytotoxic payloads to cancer cells. With the increasingly positioning of ADCs in the clinical practice, combinations with other treatment modalities, including radiation therapy (RT), will open new opportunities but also challenges. This review evaluates ADC-RT interactions, examining therapeutic synergies and potential caveats. ADC payloads can be radiosensitizing, enhancing cytotoxicity when used in combination with RT. Antigens targeted by ADCs can have various tissue expressions, resulting in possible off-target toxicities by tissue radiosensitization. Notably, the HER-2-directed ADC trastuzumab emtansine has appeared to increase the risk of radionecrosis when used concomitantly with brain RT, as glial cells can express HER2, too. Other possible organ-specific effects are discussed, such as pulmonary and cardiac toxicities. The lack of robust clinical data on the ADC-RT combination raises concerns regarding specific side effects and the ultimate trade-off of toxicity and safety of some combined approaches. Clinical studies are needed to assess ADC-RT combination safety and efficacy.
Collapse
Affiliation(s)
- Stefano Natangelo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Chrysanthi Koukoutzeli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giulia Marvaso
- Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy; Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
9
|
Sun M, Xie H, Zhang W, Li X, Jiang Z, Liang Y, Zhao G, Huang N, Mao J, Liu G, Zhang Z. Bioinspired Lipoproteins of Furoxans-Gemcitabine Preferentially Targets Glioblastoma and Overcomes Radiotherapy Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306190. [PMID: 38049204 PMCID: PMC10853724 DOI: 10.1002/advs.202306190] [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: 09/29/2023] [Revised: 10/31/2023] [Indexed: 12/06/2023]
Abstract
Radiotherapy (RT) resistance is an enormous challenge in glioblastoma multiforme (GBM) treatment, which is largely associated with DNA repair, poor distribution of reactive radicals in tumors, and limited delivery of radiosensitizers to the tumor sites. Inspired by the aberrant upregulation of RAD51 (a critical protein of DNA repair), scavenger receptor B type 1 (SR-B1), and C-C motif chemokine ligand 5 (CCL5) in GBM patients, a reduction-sensitive nitric oxide (NO) donor conjugate of gemcitabine (RAD51 inhibitor) (NG) is synthesized as radio-sensitizer and a CCL5 peptide-modified bioinspired lipoprotein system of NG (C-LNG) is rationally designed, aiming to preferentially target the tumor sites and overcome the RT resistance. C-LNG can preferentially accumulate at the orthotopic GBM tumor sites with considerable intratumor permeation, responsively release the gemcitabine and NO, and then generate abundant peroxynitrite (ONOO- ) upon X-ray radiation, thereby producing a 99.64% inhibition of tumor growth and a 71.44% survival rate at 120 days in GL261-induced orthotopic GBM tumor model. Therefore, the rationally designed bioinspired lipoprotein of NG provides an essential strategy to target GBM and overcome RT resistance.
Collapse
Affiliation(s)
- Maoyuan Sun
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Honglei Xie
- Institute of PharmacologySchool of Pharmaceutical SciencesShandong First Medical University & Shandong Academy of Medical Sciences619 Changcheng RoadTaian271016China
| | - Wenli Zhang
- Department of RadiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Xianlu Li
- School of Pharmacy & Key Laboratory of Smart Drug Delivery (Ministry of Education)Fudan UniversityShanghai201203China
| | - Zhan Jiang
- Department of OncologyThe Chongqing General HospitalChongqing400016China
| | - Yiyu Liang
- School of Pharmacy & Key Laboratory of Smart Drug Delivery (Ministry of Education)Fudan UniversityShanghai201203China
| | - Guanjian Zhao
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Ning Huang
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Jinning Mao
- Health Management CenterThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Guodong Liu
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Zhiwen Zhang
- School of Pharmacy & Key Laboratory of Smart Drug Delivery (Ministry of Education)Fudan UniversityShanghai201203China
| |
Collapse
|
10
|
Dai X, Xie Y, Feng W, Chen Y. Nanomedicine-Enabled Chemical Regulation of Reactive X Species for Versatile Disease Treatments. Angew Chem Int Ed Engl 2023; 62:e202309160. [PMID: 37653555 DOI: 10.1002/anie.202309160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/02/2023]
Abstract
Reactive X species (RXS), encompassing elements such as O, N, C, S, Se, Cl, Br, I, and H, play vital roles in cell biology and physiological function, impacting cellular signal transduction, metabolic regulation, and disease processes. The redox unbalance of RXS is firmly implicated in an assortment of physiological and pathological disorders, including cancer, diabetes, cardiovascular disease, and neurodegenerative diseases. However, the intricate nature and multifactorial dependence of RXS pose challenges in comprehending and precisely modulating their biological behavior. Nanomaterials with distinct characteristics and biofunctions offer promising avenues for generating or scavenging RXS to maintain redox homeostasis and advance disease therapy. This minireview provides a tutorial summary of the relevant chemistry and specific mechanisms governing different RXS, focusing on cellular metabolic regulation, stress responses, and the role of nanomedicine in RXS generation and elimination. The challenges associated with chemically regulating RXS for diverse disease treatments are further discussed along with the future prospects, aiming to facilitate the clinical translation of RXS-based nanomedicine and open new avenues for improved therapeutic interventions.
Collapse
Affiliation(s)
- Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
11
|
Zhou LL, Guan Q, Zhou W, Kan JL, Teng K, Hu M, Dong YB. A Multifunctional Covalent Organic Framework Nanozyme for Promoting Ferroptotic Radiotherapy against Esophageal Cancer. ACS NANO 2023; 17:20445-20461. [PMID: 37801392 DOI: 10.1021/acsnano.3c06967] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Radiotherapy is inevitably accompanied by some degree of radiation resistance, which leads to local recurrence and even therapeutic failure. To overcome this limitation, herein, we report the room-temperature synthesis of an iodine- and ferrocene-loaded covalent organic framework (COF) nanozyme, termed TADI-COF-Fc, for the enhancement of radiotherapeutic efficacy in the treatment of radioresistant esophageal cancer. The iodine atoms on the COF framework not only exerted a direct effect on radiotherapy, increasing its efficacy by increasing X-ray absorption, but also promoted the radiolysis of water, which increased the production of reactive oxygen species (ROS). In addition, the ferrocene surface decoration disrupted redox homeostasis by increasing the levels of hydroxyl and lipid peroxide radicals and depleting intracellular antioxidants. Both in vitro and in vivo experiments substantiated the excellent radiotherapeutic response of TADI-COF-Fc. This study demonstrates the potential of COF-based multinanozymes as radiosensitizers and suggests a possible treatment integration strategy for combination oncotherapy.
Collapse
Affiliation(s)
- Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| | - Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| | - Wei Zhou
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Jing-Lan Kan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| | - Kai Teng
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Man Hu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
12
|
Zhao Z, Shan X, Zhang H, Shi X, Huang P, Sun J, He Z, Luo C, Zhang S. Nitric oxide-driven nanotherapeutics for cancer treatment. J Control Release 2023; 362:151-169. [PMID: 37633361 DOI: 10.1016/j.jconrel.2023.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
Nitric oxide (NO) is a gaseous molecule endowed with diverse biological functions, offering vast potential in the realm of cancer treatment. Considerable efforts have been dedicated to NO-based cancer therapy owing to its good biosafety and high antitumor activity, as well as its efficient synergistic therapy with other antitumor modalities. However, delivering this gaseous molecule effectively into tumor tissues poses a significant challenge. To this end, nano drug delivery systems (nano-DDSs) have emerged as promising platforms for in vivo efficient NO delivery, with remarkable achievements in recent years. This review aims to provide a summary of the emerging NO-driven antitumor nanotherapeutics. Firstly, the antitumor mechanism and related clinical trials of NO therapy are detailed. Secondly, the latest research developments in the stimulation of endogenous NO synthesis are presented, including the regulation of nitric oxide synthases (NOS) and activation of endogenous NO precursors. Moreover, the emerging nanotherapeutics that rely on tumor-specific delivery of NO donors are outlined. Additionally, NO-driven combined nanotherapeutics for multimodal cancer theranostics are discussed. Finally, the future directions, application prospects, and challenges of NO-driven nanotherapeutics in clinical translation are highlighted.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinzhu Shan
- Department of State Key Laboratory of Natural and Biomimetic Drugs, College of Pharmaceutical Sciences, Peking University, Beijing 100871, PR China
| | - Hongyuan Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Peiqi Huang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shenwu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
13
|
Kumar A, Estes Bright LM, Garren MRS, Manuel J, Shome A, Handa H. Chemical Modification of Tiopronin for Dual Management of Cystinuria and Associated Bacterial Infections. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43332-43344. [PMID: 37671841 PMCID: PMC10520916 DOI: 10.1021/acsami.3c07160] [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: 05/18/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023]
Abstract
Cystinuria is an inherited autosomal recessive disease of the kidneys of recurring nature that contributes to frequent urinary tract infections due to bacterial growth and biofilm formation surrounding the stone microenvironment. In the past, commonly used strategies for managing cystinuria involved the use of (a) cystine crystal growth inhibitors such as l-cystine dimethyl ester and lipoic acid, and (b) thiol-based small molecules such as N-(2-mercaptopropionyl) glycine, commonly known as tiopronin, that reduce the formation of cystine crystals by reacting with excess cystine and generating more soluble disulfide compounds. However, there is a dearth of simplistic chemical approaches that have focused on the dual treatment of cystinuria and the associated microbial infections. This work strategically exploited a single chemical approach to develop a nitric oxide (NO)-releasing therapeutic compound, S-nitroso-2-mercaptopropionyl glycine (tiopronin-NO), for the dual management of cystine stone formation and the related bacterial infections. The results successfully demonstrated that (a) the antibacterial activity of NO rendered tiopronin-NO effective against the stone microenvironment inhabitants, Escherichia coli and Pseudomonas aeruginosa, and (b) tiopronin-NO retained the ability to undergo disulfide exchange with cystine while being reported to be safe against canine kidney and mouse fibroblast cells. Thus, the synthesis of such a facile molecule aimed at the dual management of cystinuria and related infections is unprecedented in the literature.
Collapse
Affiliation(s)
- Anil Kumar
- School
of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Lori M. Estes Bright
- School
of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Mark Richard Stephen Garren
- School
of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - James Manuel
- School
of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Arpita Shome
- School
of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Hitesh Handa
- School
of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
- Pharmaceutical
and Biomedical Sciences Department, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
14
|
Ghaffarlou M, Mohammadi A, Mousazadeh N, Salehiabar M, Kalantari Y, Charmi J, Barsbay M, Ertas YN, Danafar H, Rezaeejam H, Nosrati H, Javani S. Facile preparation of silver based radiosensitizers via biomineralization method for enhanced in vivo breast cancer radiotherapy. Sci Rep 2023; 13:15131. [PMID: 37704633 PMCID: PMC10499791 DOI: 10.1038/s41598-023-40763-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023] Open
Abstract
To solve the traditional radiotherapy obstacles, and also to enhance the radiation therapy efficacy various radiosensitizers have been developed. Radiosensitizers are promising agents that under X-ray irradiation enhance injury to tumor tissue by accelerating DNA damage. In this report, silver-silver sulfide nanoparticles (Ag-Ag2S NPs) were synthesized via a facile, one-pot and environmentally friendly biomineralization method. Ag-Ag2S was coated with bovine serum albumin (BSA) in situ and applied as an X-ray sensitizer to enhance the efficiency of radiotherapy. Also, folic acid (FA) was conjugated to Ag-Ag2S@BSA to impart active targeting capability to the final formulation (Ag-Ag2S@BSA-FA). Prepared NPs were characterized by transmission electron microscopes (TEM), scanning electron microscope (SEM), dynamic light scattering (DLS), ultraviolet-visible spectroscopy (UV-Vis), X-ray diffraction analysis (XRD), and X-ray photoelectron spectroscopy (XPS) techniques. Results show that most of the NPs have well-defined uniform Janus structures. The biocompatibility of the NPs was then evaluated both in vitro and in vivo. A series of in vitro assays were performed on 4T1 cancer cells to evaluate the therapeutic efficacy of the designed NPs. In addition, the radio-enhancing ability of the NPs was tested on the 4T1 breast cancer murine model. MTT, live and dead cell staining, apoptosis, ROS generation, and clonogenic in vitro assays demonstrated the efficacy of NPs as radiosensitizers in radiotherapy. In vivo results as well as H&E staining tumor tissues confirmed tumor destruction in the group that received Ag-Ag2S@BSA-FA NPs and exposed to X-ray. The results showed that prepared tumor-targeted Ag-Ag2S@BSA-FA NPs could be potential candidates as radiosensitizers for enhanced radiotherapy.
Collapse
Affiliation(s)
| | - Ali Mohammadi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Navid Mousazadeh
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marziyeh Salehiabar
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Yahya Kalantari
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jalil Charmi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Murat Barsbay
- Department of Chemistry, Hacettepe University, Beytepe, Ankara, 06800, Turkey
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Turkey
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38039, Turkey
| | - Hossein Danafar
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamed Rezaeejam
- Department of Radiology Technology, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan, 45139-56184, Iran.
| | - Hamed Nosrati
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Siamak Javani
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
- School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
15
|
Zafar S, Armaghan M, Khan K, Hassan N, Sharifi-Rad J, Habtemariam S, Kieliszek M, Butnariu M, Bagiu IC, Bagiu RV, Cho WC. New insights into the anticancer therapeutic potential of maytansine and its derivatives. Biomed Pharmacother 2023; 165:115039. [PMID: 37364476 DOI: 10.1016/j.biopha.2023.115039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Maytansine is a pharmacologically active 19-membered ansamacrolide derived from various medicinal plants and microorganisms. Among the most studied pharmacological activities of maytansine over the past few decades are anticancer and anti-bacterial effects. The anticancer mechanism of action is primarily mediated through interaction with the tubulin thereby inhibiting the assembly of microtubules. This ultimately leads to decreased stability of microtubule dynamics and cause cell cycle arrest, resulting in apoptosis. Despite its potent pharmacological effects, the therapeutic applications of maytansine in clinical medicine are quite limited due to its non-selective cytotoxicity. To overcome these limitations, several derivatives have been designed and developed mostly by modifying the parent structural skeleton of maytansine. These structural derivatives exhibit improved pharmacological activities as compared to maytansine. The present review provides a valuable insight into maytansine and its synthetic derivatives as anticancer agents.
Collapse
Affiliation(s)
- Sameen Zafar
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Khushbukhat Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan.
| | - Nazia Hassan
- Department of Biochemistry, University of Agriculture Faisalabad, Pakistan
| | | | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK.
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159 C, 02-776 Warsaw, Poland.
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, 300645, Calea Aradului 119, Timis, Romania.
| | - Iulia-Cristina Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Multidisciplinary Research Center on Antimicrobial Resistance, Timisoara, Romania
| | - Radu Vasile Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Preventive Medicine Study Center, Timisoara, Romania
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
16
|
Girigoswami A, Girigoswami K. Potential Applications of Nanoparticles in Improving the Outcome of Lung Cancer Treatment. Genes (Basel) 2023; 14:1370. [PMID: 37510275 PMCID: PMC10379962 DOI: 10.3390/genes14071370] [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: 03/10/2023] [Revised: 04/20/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Lung cancer is managed using conventional therapies, including chemotherapy, radiation therapy, or a combination of both. Each of these therapies has its own limitations, such as the indiscriminate killing of normal as well as cancer cells, the solubility of the chemotherapeutic drugs, rapid clearance of the drugs from circulation before reaching the tumor site, the resistance of cancer cells to radiation, and over-sensitization of normal cells to radiation. Other treatment modalities include gene therapy, immunological checkpoint inhibitors, drug repurposing, and in situ cryo-immune engineering (ICIE) strategy. Nanotechnology has come to the rescue to overcome many shortfalls of conventional therapies. Some of the nano-formulated chemotherapeutic drugs, as well as nanoparticles and nanostructures with surface modifications, have been used for effective cancer cell killing and radio sensitization, respectively. Nano-enabled drug delivery systems act as cargo to deliver the sensitizer molecules specifically to the tumor cells, thereby enabling the radiation therapy to be more effective. In this review, we have discussed the different conventional chemotherapies and radiation therapies used for inhibiting lung cancer. We have also discussed the improvement in chemotherapy and radiation sensitization using nanoparticles.
Collapse
Affiliation(s)
- Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India
| |
Collapse
|
17
|
Kumar A, Mondal A, Douglass ME, Francis DJ, Garren MR, Estes Bright LM, Ghalei S, Xie J, Brisbois EJ, Handa H. Nanoarchitectonics of nitric oxide releasing supramolecular structures for enhanced antibacterial efficacy under visible light irradiation. J Colloid Interface Sci 2023; 640:144-161. [PMID: 36842420 PMCID: PMC10081829 DOI: 10.1016/j.jcis.2023.02.083] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
Light-controlled therapies offer a promising strategy to prevent and suppress infections caused by numerous bacterial pathogens. Excitation of exogenously supplied photosensitizers (PS) at specific wavelengths elicits levels of reactive oxygen intermediates toxic to bacteria. Porphyrin-based supramolecular nanostructure frameworks (SNF) are effective PS with unique physicochemical properties that have led to their widespread use in photomedicine. Herein, we developed a nitric oxide (NO) releasing, biocompatible, and stable porphyrin-based SNF (SNF-NO), which was achieved through a confined noncovalent self-assembly process based on π-π stacking. Characterization of the SNFs via scanning electron microscopy (SEM) and transmission electron microscopy (TEM) analysis showed the formation of three-dimensional, well-defined octahedral structures. These SNF-NO were shown to exhibit a red shift due to the noncovalent self-assembly of porphyrins, which also show extended light absorption to broadly cover the entire visible light spectrum to enhance photodynamic therapy (PDT). Under visible light irradiation (46 J cm-2), the SNF generates high yields of singlet oxygen (1O2) radicals, hydroxyl radicals (HO), superoxide radicals (O2), and peroxynitrite (ONOO-) radicals that have shown potential to enhance antimicrobial photodynamic therapy (APDT) against Gram-positive methicillin-resistant Staphylococcus aureus (MRSA) and Gram-negative Escherichia coli (E. coli). The resulting SNFs also exhibit significant biofilm dispersion and a decrease in biomass production. The combination of robust photosensitizer SNFs with nitric oxide-releasing capabilities is dynamic in its ability to target pathogenic infections while remaining nontoxic to mammalian cells. The engineered SNFs have enormous potential for treating and managing microbial infections.
Collapse
Affiliation(s)
- Anil Kumar
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Arnab Mondal
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Megan E Douglass
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Divine J Francis
- Department of Chemistry, University of Georgia, Athens, GA 30602, United States
| | - Mark R Garren
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Lori M Estes Bright
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Sama Ghalei
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA 30602, United States
| | - Elizabeth J Brisbois
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States
| | - Hitesh Handa
- School of Chemical Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, United States.
| |
Collapse
|
18
|
Wang Z, Jin A, Yang Z, Huang W. Advanced Nitric Oxide Generating Nanomedicine for Therapeutic Applications. ACS NANO 2023; 17:8935-8965. [PMID: 37126728 PMCID: PMC10395262 DOI: 10.1021/acsnano.3c02303] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nitric oxide (NO), a gaseous transmitter extensively present in the human body, regulates vascular relaxation, immune response, inflammation, neurotransmission, and other crucial functions. Nitrite donors have been used clinically to treat angina, heart failure, pulmonary hypertension, and erectile dysfunction. Based on NO's vast biological functions, it further can treat tumors, bacteria/biofilms and other infections, wound healing, eye diseases, and osteoporosis. However, delivering NO is challenging due to uncontrolled blood circulation release and a half-life of under five seconds. With advanced biotechnology and the development of nanomedicine, NO donors packaged with multifunctional nanocarriers by physically embedding or chemically conjugating have been reported to show improved therapeutic efficacy and reduced side effects. Herein, we review and discuss recent applications of NO nanomedicines, their therapeutic mechanisms, and the challenges of NO nanomedicines for future scientific studies and clinical applications. As NO enables the inhibition of the replication of DNA and RNA in infectious microbes, including COVID-19 coronaviruses and malaria parasites, we highlight the potential of NO nanomedicines for antipandemic efforts. This review aims to provide deep insights and practical hints into design strategies and applications of NO nanomedicines.
Collapse
Affiliation(s)
- Zhixiong Wang
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| | - Wei Huang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| |
Collapse
|
19
|
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
|
20
|
Navale GR, Singh S, Ghosh K. NO donors as the wonder molecules with therapeutic potential: Recent trends and future perspectives. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
21
|
Wu Y, Zhu K, Zhang X, Du W, Song J, Yang H. Emerging plasmonic nanoparticles and their assemblies for cancer radiotherapy. Adv Drug Deliv Rev 2023; 194:114710. [PMID: 36708774 DOI: 10.1016/j.addr.2023.114710] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/07/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
Plasmonic nanoparticles and their assemblies have been widely used in biosensing, optical imaging, and biomedicine over the past few decades. Especially in the field of radiotherapy, the physicochemical properties of high-Z plasmonic nanomaterials endow them with the ability to sensitize radiotherapy. Compared with single particles, the assembled structure with tunable properties leads to versatile applications in drug delivery and cancer treatment. In this review, we focus on plasmonic nanoparticles and their assemblies for cancer radiotherapy. First, the sensitization mechanism of plasmonic radiosensitizers is briefly introduced. Subsequently, the recent progress in cancer radiotherapy is systematically discussed according to the structure and shape classification. Finally, the current challenges and future perspectives in this field are also discussed in detail.
Collapse
Affiliation(s)
- Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China
| | - Xuan Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Wei Du
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China.
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China.
| |
Collapse
|
22
|
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
|
23
|
Li J, Zhu L, Kwok HF. Nanotechnology-based approaches overcome lung cancer drug resistance through diagnosis and treatment. Drug Resist Updat 2023; 66:100904. [PMID: 36462375 DOI: 10.1016/j.drup.2022.100904] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lung cancer continues to be a malignant tumor with high mortality. Two obstacles interfere with curative therapy of lung cancer: (i) poor diagnosis at the early stages, as symptoms are not specific or asymptomatic; and (ii) invariably emerging drug resistance after treatment. Some factors contributing to drug resistance include preexisting genetic/genomic drug-resistant alteration(s); activation of adaptive drug resistance pathways; remodeling of the tumor microenvironment; and pharmacological mechanisms or activation of drug efflux pumps. Despite the mechanisms explored to better understand drug resistance, a gap remains between molecular understanding and clinical application. Therefore, facilitating the translation of basic science into the clinical setting is a great challenge. Nanomedicine has emerged as a promising tool for cancer treatment. Because of their excellent physicochemical properties and enhanced permeability and retention effects, nanoparticles have great potential to revolutionize conventional lung cancer diagnosis and combat drug resistance. Nanoplatforms can be designed as carriers to improve treatment efficacy and deliver multiple drugs in one system, facilitating combination treatment to overcome drug resistance. In this review, we describe the difficulties in lung cancer treatment and review recent research progress on nanoplatforms aimed at early diagnosis and lung cancer treatment. Finally, future perspectives and challenges of nanomedicine are also discussed.
Collapse
Affiliation(s)
- Junnan Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Lipeng Zhu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
24
|
Duan Y, Shen C, Zhang Y, Luo Y. Advanced diagnostic and therapeutic strategies in nanotechnology for lung cancer. Front Oncol 2022; 12:1031000. [PMID: 36568152 PMCID: PMC9767962 DOI: 10.3389/fonc.2022.1031000] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
As a highly invasive thoracic malignancy with increasing prevalence, lung cancer is also the most lethal cancer worldwide due to the failure of effective early detection and the limitations of conventional therapeutic strategies for advanced-stage patients. Over the past few decades, nanotechnology has emerged as an important technique to obtain desired features by modifying and manipulating different objects on a molecular level and gained a lot of attention in many fields of medical applications. Studies have shown that in lung cancer, nanotechnology may be more effective and specific than traditional methods for detecting extracellular cancer biomarkers and cancer cells in vitro, as well as imaging cancer in vivo; Nanoscale drug delivery systems have developed rapidly to overcome various forms of multi-drug resistance and reduce detrimental side effects to normal tissues by targeting cancerous tissue precisely. There is no doubt that nanotechnology has the potential to enhance healthcare systems by simplifying and improving cancer diagnostics and treatment. Throughout this review, we summarize and highlight recent developments in nanotechnology applications for lung cancer in diagnosis and therapy. Moreover, the prospects and challenges in the translation of nanotechnology-based diagnostic and therapeutic methods into clinical applications are also discussed.
Collapse
Affiliation(s)
- Yujuan Duan
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China,School of Chemical Science and Engineering, Tongji University, Shanghai, China,Department of Laboratory Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Shen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai, China,*Correspondence: Yao Luo, ; Yinan Zhang,
| | - Yao Luo
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Yao Luo, ; Yinan Zhang,
| |
Collapse
|
25
|
Li Y, Yoon B, Dey A, Nguyen VQ, Park JH. Recent progress in nitric oxide-generating nanomedicine for cancer therapy. J Control Release 2022; 352:179-198. [PMID: 36228954 DOI: 10.1016/j.jconrel.2022.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/26/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Nitric oxide (NO) is an endogenous, multipotent biological signaling molecule that participates in several physiological processes. Recently, exogenous supplementation of tumor tissues with NO has emerged as a potential anticancer therapy. In particular, it induces synergistic effects with other conventional therapies (such as chemo-, radio-, and photodynamic therapies) by regulating the activity of P-glycoprotein, acting as a vascular relaxant to relieve tumor hypoxia, and participating in the metabolism of reactive oxygen species. However, NO is highly reactive, and its half-life is relatively short after generation. Meanwhile, NO-induced anticancer activity is dose-dependent. Therefore, the targeted delivery of NO to the tumor is required for better therapeutic effects. In the past decade, NO-generating nanomedicines (NONs), which enable sustained and specific NO release in tumor tissues, have been developed for enhanced cancer therapy. This review describes the recent efforts and preclinical achievements in the development of NON-based cancer therapies. The chemical structures employed in the fabrication of NONs are summarized, and the strategies involved in NON-based cancer therapies are elaborated.
Collapse
Affiliation(s)
- Yuce Li
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Been Yoon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Anup Dey
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Quy Nguyen
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea.; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea.
| |
Collapse
|
26
|
Kim J, Thomas SN. Opportunities for Nitric Oxide in Potentiating Cancer Immunotherapy. Pharmacol Rev 2022; 74:1146-1175. [PMID: 36180108 PMCID: PMC9553106 DOI: 10.1124/pharmrev.121.000500] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/15/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
Despite nearly 30 years of development and recent highlights of nitric oxide (NO) donors and NO delivery systems in anticancer therapy, the limited understanding of exogenous NO's effects on the immune system has prevented their advancement into clinical use. In particular, the effects of exogenously delivered NO differing from that of endogenous NO has obscured how the potential and functions of NO in anticancer therapy may be estimated and exploited despite the accumulating evidence of NO's cancer therapy-potentiating effects on the immune system. After introducing their fundamentals and characteristics, this review discusses the current mechanistic understanding of NO donors and delivery systems in modulating the immunogenicity of cancer cells as well as the differentiation and functions of innate and adaptive immune cells. Lastly, the potential for the complex modulatory effects of NO with the immune system to be leveraged for therapeutic applications is discussed in the context of recent advancements in the implementation of NO delivery systems for anticancer immunotherapy applications. SIGNIFICANCE STATEMENT: Despite a 30-year history and recent highlights of nitric oxide (NO) donors and delivery systems as anticancer therapeutics, their clinical translation has been limited. Increasing evidence of the complex interactions between NO and the immune system has revealed both the potential and hurdles in their clinical translation. This review summarizes the effects of exogenous NO on cancer and immune cells in vitro and elaborates these effects in the context of recent reports exploiting NO delivery systems in vivo in cancer therapy applications.
Collapse
Affiliation(s)
- Jihoon Kim
- Parker H. Petit Institute for Bioengineering and Bioscience (J.K., S.N.T.), George W. Woodruff School of Mechanical Engineering (J.K., S.N.T.), and Wallace H. Coulter Department of Biomedical Engineering (S.N.T.), Georgia Institute of Technology, Atlanta, Georgia; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia (S.N.T.); and Division of Biological Science and Technology, Yonsei University, Wonju, South Korea (J.K.)
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience (J.K., S.N.T.), George W. Woodruff School of Mechanical Engineering (J.K., S.N.T.), and Wallace H. Coulter Department of Biomedical Engineering (S.N.T.), Georgia Institute of Technology, Atlanta, Georgia; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia (S.N.T.); and Division of Biological Science and Technology, Yonsei University, Wonju, South Korea (J.K.)
| |
Collapse
|
27
|
Xu XX, Chen SY, Yi NB, Li X, Chen SL, Lei Z, Cheng DB, Sun T. Research progress on tumor hypoxia-associative nanomedicine. J Control Release 2022; 350:829-840. [PMID: 36100192 DOI: 10.1016/j.jconrel.2022.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Hypoxia at the solid tumor site is generally related to the unrestricted proliferation and metabolism of cancerous cells, which can cause tumor metastasis and aggravate tumor progression. Besides, hypoxia plays a substantial role in tumor treatment, and it is one of the main reasons that malignant tumors are difficult to cure and have a poor prognosis. On account of the tumor specific hypoxic environment, many hypoxia-associative nanomedicine have been proposed for tumor treatment. Considering the enhanced targeting effect, designing hypoxia-associative nanomedicine can not only minimize the adverse effects of drugs on normal tissues, but also achieve targeted therapy at the lesion site. Mostly, there can be three strategies for the treatment of hypoxic tumor, including improvement of hypoxic environment, hypoxia responsive drug release and hypoxia activated prodrug. The review describes the design principle and applications of tumor hypoxia-associative nanomedicine in recent years, and also explores its development trends in solid tumor treatment. Moreover, this review presents the current limitations of tumor hypoxia-associative nanomedicine in chemotherapy, radiotherapy, photodynamic therapy, sonodynamic therapy and immunotherapy, which may provide a reference for clinic translation of tumor hypoxia-associative nanomedicine.
Collapse
Affiliation(s)
- Xiao-Xue Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Yi Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Ning-Bo Yi
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xin Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| |
Collapse
|
28
|
Yang C, Mu G, Zhang Y, Gao Y, Zhang W, Liu J, Zhang W, Li P, Yang L, Yang Z, Gao J, Liu J. Supramolecular Nitric Oxide Depot for Hypoxic Tumor Vessel Normalization and Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202625. [PMID: 35906003 DOI: 10.1002/adma.202202625] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/15/2022] [Indexed: 06/15/2023]
Abstract
In cancer radiotherapy, the lack of fixed DNA damage by oxygen in hypoxic microenvironment of solid tumors often leads to severe radioresistance. Nitric oxide (NO) is a potent radiosensitizer that acts in two ways. It can directly react with the radical DNA thus fixing the damage. It also normalizes the abnormal tumor vessels, thereby increasing blood perfusion and oxygen supply. To achieve these functions, the dosage and duration of NO treatment need to be carefully controlled, otherwise it will lead to the exact opposite outcomes. However, a delivery method that fulfills both requirements is still lacking. A NO depot is designed for the control of NO releasing both over quantity and duration for hypoxic tumor vessel normalization and radiosensitization. In B16-tumor-bearing mice, the depot can provide low dosage NO continuously and release large amount of NO immediately before irradiation for a short period of time. These two modes of treatment work in synergy to reverse the radioresistance of B16 tumors more efficiently than releasing at single dosage.
Collapse
Affiliation(s)
- Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Ganen Mu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Yang Gao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Wenxue Zhang
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Wenwen Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Paiyun Li
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
29
|
Minaei SE, Khoei S, Khoee S, Mahdavi SR. Sensitization of glioblastoma cancer cells to radiotherapy and magnetic hyperthermia by targeted temozolomide-loaded magnetite tri-block copolymer nanoparticles as a nanotheranostic agent. Life Sci 2022; 306:120729. [PMID: 35753439 DOI: 10.1016/j.lfs.2022.120729] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/09/2022] [Accepted: 06/20/2022] [Indexed: 11/27/2022]
Abstract
AIMS Recently, the development of new strategies in the treatment and diagnosis of cancer cells such as thermo-radiation-sensitizer and theranostic agents have received a great deal of attention. In this work, folic acid-conjugated temozolomide-loaded SPION@PEG-PBA-PEG nanoparticles (TMZ-MNP-FA NPs) were proposed for use as magnetic resonance imaging (MRI) contrast agents and to enhance the cytotoxic effects of hyperthermia and radiotherapy. MAIN METHODS Nanoparticles were synthesized by the Nano-precipitation method and their characteristics were determined by dynamic light scattering (DLS), scanning electron microscopy (SEM) and X-ray powder diffraction (XRD). To evaluate the thermo-radio-sensitization effects of NPs, C6 cells were treated with nanoparticles for 24 h and then exposed to 6-MV X-ray radiation. After radiotherapy, the cells were subjected to an alternating magnetic field (AMF) hyperthermia. The therapeutic potential was assessed using clonogenic assay, ROS generation measurement, flow cytometry assay, and qRT-PCR analysis. Also, the diagnostic properties of the nanoparticles were assessed by MRI. KEY FINDINGS MRI scanning indicated that nanoparticles accumulated in C6 cells could be tracked by T2-weighted MR imaging. Colony formation assay proved that TMZ-MNP-FA NPs enhanced the anti-proliferation effects of AMF by 1.94-fold compared to AMF alone (P < 0.0001). Moreover, these NPs improved the radiation effects with a dose enhancement factor of 1.65. All results showed that the combination of carrier-based chemotherapy with hyperthermia and radiotherapy caused a higher anticancer efficacy than single- or two-modality treatments. SIGNIFICANCE The nanoparticles advanced in this study can be proposed as the promising theranostic and thermo-radio-sensitizer platform for the diagnosis and tri-modal synergistic cancer therapy.
Collapse
Affiliation(s)
- Soraya Emamgholizadeh Minaei
- Department of Medical Physics and Imaging, School of Allied Medical Sciences, Urmia University of Medical Sciences, Urmia, Iran
| | - Samideh Khoei
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Khoee
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Seied Rabi Mahdavi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Lin J, Yin M, Liu X, Meng F, Luo L. Nanomaterials Based on Functional Polymers for Sensitizing Cancer Radiotherapy. Macromol Rapid Commun 2022; 43:e2200194. [PMID: 35578790 DOI: 10.1002/marc.202200194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/21/2022] [Indexed: 11/12/2022]
Abstract
Despite being the mainstay treatment for many types of cancer in clinic, radiotherapy is undertaking great challenges in overcoming a series of limitations. Radiosensitizers are promising agents capable of depositing irradiation energy and generating free radicals to enhance the radiosensitivity of tumor cells. Combining radiosensitizers with functional polymer-based nanomaterials holds great potential to improve biodistribution, circulation time, and stability in vivo. The derived polymeric nano-radiosensitizers can significantly improve the efficiency of tumor targeting and radiotherapy, and reduce the side effect to healthy tissues. In this review, we provide an overview of functional polymer-based nanomaterials for radiosensitization in recent years. Particular emphases are given to the action mechanisms, drug loading methods, targeting efficiencies, the impact on therapeutic effects and biocompatibility of various radiosensitizing polymers, which are classified as polymeric micelles, dendrimers, polymeric nanospheres, nanoscale coordination polymers, polymersomes, and nanogels. The challenges and outlooks of polymeric nano-radiosensitizers are also discussed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jinfeng Lin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mingming Yin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
31
|
Liu W, Semcheddine F, Guo Z, Jiang H, Wang X. Glucose-Responsive ZIF-8 Nanocomposites for Targeted Cancer Therapy through Combining Starvation with Stimulus-Responsive Nitric Oxide Synergistic Treatment. ACS APPLIED BIO MATERIALS 2022; 5:2902-2912. [PMID: 35533346 DOI: 10.1021/acsabm.2c00262] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
With the rapid development of nanomedicine, low side effects and high-efficiency green antitumor approaches have attracted great attention. Herein, we report a strategy for the in situ synthesis of graphene oxide@zeolitic imidazolate framework-8 (GOx@ZIF-8) composite nanoparticles with high catalytic efficiency, under mild conditions by adding GOx molecules to the precursor of ZIF-8, and use them as a carrier to achieve efficient loading of l-Arg. In addition. folic-acid-conjugated bovine serum albumin (FA-BSA) has been used to engineer the surface of GOx@ZIF-8-l-Arg composite nanoparticles to enhance their specific recognition of tumor cells. With the high glucose level and low pH in the tumor intracellular environment, FA-BSA/GOx@ZIF-8-l-Arg rapidly consumed the intracellular glucose and produced H2O2, which profusely deteriorated the intracellular environment. Subsequently, a large amount of l-Arg was continuously released from the nanoparticles, reacting with H2O2 to continuously produce a high concentration of nitric oxide (NO), which further damaged the tumor cells. The FA-BSA/GOx@ZIF-8-l-Arg composite nanoparticles were cleverly designed to kill cancer cells efficiently through a starvation-NO synergistic process. This emerging green antitumor method has a promising application prospect in targeted therapy for the efficient clearance of cancers.
Collapse
Affiliation(s)
- Weiwei Liu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.,School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Farouk Semcheddine
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zengchao Guo
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
32
|
Sodano F, Gazzano E, Fruttero R, Lazzarato L. NO in Viral Infections: Role and Development of Antiviral Therapies. Molecules 2022; 27:2337. [PMID: 35408735 PMCID: PMC9000700 DOI: 10.3390/molecules27072337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022] Open
Abstract
Nitric oxide is a ubiquitous signaling radical that influences critical body functions. Its importance in the cardiovascular system and the innate immune response to bacterial and viral infections has been extensively investigated. The overproduction of NO is an early component of viral infections, including those affecting the respiratory tract. The production of high levels of NO is due to the overexpression of NO biosynthesis by inducible NO synthase (iNOS), which is involved in viral clearance. The development of NO-based antiviral therapies, particularly gaseous NO inhalation and NO-donors, has proven to be an excellent antiviral therapeutic strategy. The aim of this review is to systematically examine the multiple research studies that have been carried out to elucidate the role of NO in viral infections and to comprehensively describe the NO-based antiviral strategies that have been developed thus far. Particular attention has been paid to the potential mechanisms of NO and its clinical use in the prevention and therapy of COVID-19.
Collapse
Affiliation(s)
- Federica Sodano
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy; (R.F.); (L.L.)
- Department of Pharmacy, “Federico II” University of Naples, 80131 Naples, Italy
| | - Elena Gazzano
- Department of Life Sciences and Systems Biology, University of Torino, 10123 Torino, Italy
| | - Roberta Fruttero
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy; (R.F.); (L.L.)
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy; (R.F.); (L.L.)
| |
Collapse
|
33
|
Li Y, Yang J, Gu G, Guo X, He C, Sun J, Zou H, Wang H, Liu S, Li X, Zhang S, Wang K, Yang L, Jiang Y, Wu L, Sun X. Pulmonary Delivery of Theranostic Nanoclusters for Lung Cancer Ferroptosis with Enhanced Chemodynamic/Radiation Synergistic Therapy. NANO LETTERS 2022; 22:963-972. [PMID: 35073699 DOI: 10.1021/acs.nanolett.1c03786] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Inefficient tumor accumulation and penetration remain as the main challenges to therapy efficacy of lung cancer. Local delivery of smart nanoclusters can increase drug penetration and provide superior antitumor effects than systemic routes. Here, we report self-assembled pH-sensitive superparamagnetic iron oxide nanoclusters (SPIONCs) that enhance in situ ferroptosis and apoptosis with radiotherapy and chemodynamic therapy. After pulmonary delivery in orthotopic lung cancer, SPIONCs disintegrate into smaller nanoparticles and release more iron ions in an acidic microenvironment. Under single-dose X-ray irradiation, endogenous superoxide dismutase converts superoxide radicals produced by mitochondria to hydrogen peroxide, which in turn generates hydroxyl radicals by the Fenton reaction from iron ions accumulated inside the tumor. Finally, irradiation and iron ions enhance tumor lipid peroxidation and induce cell apoptosis and ferroptosis. Thus, rationally designed pulmonary delivered nanoclusters provide a promising strategy for noninvasive imaging of lung cancer and synergistic therapy.
Collapse
Affiliation(s)
- Yingbo Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Jie Yang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Guangying Gu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150028, China
| | - Xu Guo
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Chunbo He
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Jiemei Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Hongyan Zou
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Hongbin Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Shuang Liu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Xiaona Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Shujun Zhang
- Department of Pathology, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Kai Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Lili Yang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Ying Jiang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Lina Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin 150028, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin 150028, China
| |
Collapse
|
34
|
Yu S, Wang Y, He P, Shao B, Liu F, Xiang Z, Yang T, Zeng Y, He T, Ma J, Wang X, Liu L. Effective Combinations of Immunotherapy and Radiotherapy for Cancer Treatment. Front Oncol 2022; 12:809304. [PMID: 35198442 PMCID: PMC8858950 DOI: 10.3389/fonc.2022.809304] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Though single tumor immunotherapy and radiotherapy have significantly improved the survival rate of tumor patients, there are certain limitations in overcoming tumor metastasis, recurrence, and reducing side effects. Therefore, it is urgent to explore new tumor treatment methods. The new combination of radiotherapy and immunotherapy shows promise in improving therapeutic efficacy and reducing recurrence by enhancing the ability of the immune system to recognize and eradicate tumor cells, to overcome tumor immune tolerance mechanisms. Nanomaterials, as new drug-delivery-system materials of the 21st century, can maintain the activity of drugs, improve drug targeting, and reduce side effects in tumor immunotherapy. Additionally, nanomaterials, as radiosensitizers, have shown great potential in tumor radiotherapy due to their unique properties, such as light, heat, electromagnetic effects. Here, we review the mechanisms of tumor immunotherapy and radiotherapy and the synergy of radiotherapy with multiple types of immunotherapies, including immune checkpoint inhibitors (ICIs), tumor vaccines, adoptive cell therapy, and cytokine therapy. Finally, we propose the potential for nanomaterials in tumor radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Siting Yu
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ping He
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Bianfei Shao
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Liu
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongzheng Xiang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Yang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zeng
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao He
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiachun Ma
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiran Wang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Liu
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Lei Liu,
| |
Collapse
|
35
|
Ma W, Yang Y, Zhu J, Jia W, Zhang T, Liu Z, Chen X, Lin Y. Biomimetic Nanoerythrosome-Coated Aptamer-DNA Tetrahedron/Maytansine Conjugates: pH-Responsive and Targeted Cytotoxicity for HER2-Positive Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109609. [PMID: 35064993 DOI: 10.1002/adma.202109609] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/19/2022] [Indexed: 02/05/2023]
Abstract
DNA materials have emerged as potential nanocarriers for targeted cancer therapy to precisely deliver cargos with specific purposes. The short half-life and low bioavailability of DNA materials due to their interception by the reticuloendothelial system and blood clearance further limit their clinical translation. This study employs an HER2-targeted DNA-aptamer-modified DNA tetrahedron (HApt-tFNA) as a drug delivery system, and combines maytansine (DM1) to develop the HApt-DNA tetrahedron/DM1 conjugate (HApt-tFNA@DM1, HTD, HApDC) for targeted therapy of HER2-positive cancer. To optimize the pharmacokinetics and tumor-aggregation of HTD, a biomimetic camouflage is applied to embed HTD. The biomimetic camouflage is constructed by merging the erythrocyte membrane with pH-responsive functionalized synthetic liposomes, thus with excellent performance of drug delivery and tumor-stimulated drug release. The hybrid erythrosome-based nanoparticles show better inhibition of HER2-positive cancer than other drug formulations and exhibit superior biosafety. With the strengths of precise delivery, increased drug loading, sensitive tumor probing, and prolonged circulation time, the HApDC represents a promising nanomedicine to treat HER2-positive tumors. Notably, this study developsa dual-targeting nanoparticle by combining pH-sensitive camouflage and HApDC, initiating an important step toward the development and application of DNA-based medicine and biomimetic cell membrane materials in cancer treatment and other potential biological applications.
Collapse
Affiliation(s)
- Wenjuan Ma
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yuting Yang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Jianwei Zhu
- Department of Neurosurgery Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu 611731 P. R. China
| | - Weiqiang Jia
- Department of Neurosurgery Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu 611731 P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Xingyu Chen
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
- College of Biomedical Engineering Sichuan University Chengdu 610041 P. R. 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
|
Yue S, Zhang Y, Wei Y, Haag R, Sun H, Zhong Z. Cetuximab-Polymersome-Mertansine Nanodrug for Potent and Targeted Therapy of EGFR-Positive Cancers. Biomacromolecules 2021; 23:100-111. [PMID: 34913340 DOI: 10.1021/acs.biomac.1c01065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Targeted nanomedicines particularly armed with monoclonal antibodies are considered to be the most promising advanced chemotherapy for malignant cancers; however, their development is hindered by their instability and drug leakage problems. Herein, we constructed a robust cetuximab-polymersome-mertansine nanodrug (C-P-DM1) for highly potent and targeted therapy of epidermal growth factor receptor (EGFR)-positive solid tumors. C-P-DM1 with a tailored cetuximab surface density of 2 per P-DM1 exhibited a size of ca. 60 nm, high stability with minimum DM1 leakage, glutathione-triggered release of native DM1, and 6.0-11.3-fold stronger cytotoxicity in EGFR-positive human breast (MDA-MB-231), lung (A549), and liver (SMMC-7721) cancer cells (IC50 = 27.1-135.5 nM) than P-DM1 control. Notably, intravenous injection of C-P-DM1 effectively repressed subcutaneous MDA-MB-231 breast cancer and orthotopic A549-Luc lung carcinoma in mice without inducing toxic effects. Strikingly, intratumoral injection of C-P-DM1 completely cured 60% of mice bearing breast tumor without recurrence. This robust cetuximab-polymersome-mertansine nanodrug provides a promising new strategy for targeted treatment of EGFR-positive solid malignancies.
Collapse
Affiliation(s)
- Shujing Yue
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Yaohua Wei
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Rainer Haag
- Department of Biology, Chemistry and Pharmacy, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Huanli Sun
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
38
|
An intelligent cell-selective polymersome-DM1 nanotoxin toward triple negative breast cancer. J Control Release 2021; 340:331-341. [PMID: 34774889 DOI: 10.1016/j.jconrel.2021.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022]
Abstract
Antibody-drug conjugates (ADCs) are among the most significant advances in clinical cancer treatments, however, they are haunted with fundamental issues like low drug/antibody ratio (DAR), need of large amount of antibody, and complex chemistry. Targeted nanomedicines while offering a promising alternative to ADCs are afflicted with drug leakage and inferior cancer-specificity. Herein, we developed an intelligent cell-selective nanotoxin based on anti-CD44 antibody-polymersome-DM1 conjugates (aCD44-AP-DM1) for potent treatment of solid tumors. DM1 was simultaneously coupled to vesicular membrane via disulfide bonds during self-assembly and anti-CD44 antibody was facilely clicked onto polymersome surface, tailor-making an optimal aCD44-AP-DM1 with a controlled antibody density of 5.0, extraordinary DAR of 275, zero drug leakage and rapid reduction-responsive DM1 release. aCD44-AP-DM1 displayed a high specificity and exceptional cytotoxicity toward MDA-MB-231 triple negative breast cancer, SMMC-7721 hepatocellular carcinoma and A549 non-small cell lung cancer cells with half-maximal inhibitory concentrations (IC50) of 21.4, 3.7 and 64.6 ng/mL, respectively, 3.6-47.2-fold exceeding non-targeted P-DM1. Intriguingly, the systemic administration of aCD44-AP-DM1 significantly suppressed subcutaneous MDA-MB-231 tumor xenografts in nude mice while intratumoral injection achieved complete tumor eradication in four out of five mice, without causing toxicity. This intelligent cell-selective nanotoxin has emerged as a better platform over ADCs for targeted cancer therapy.
Collapse
|
39
|
Zhao Y, Ouyang X, Peng Y, Peng S. Stimuli Responsive Nitric Oxide-Based Nanomedicine for Synergistic Therapy. Pharmaceutics 2021; 13:1917. [PMID: 34834332 PMCID: PMC8622285 DOI: 10.3390/pharmaceutics13111917] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022] Open
Abstract
Gas therapy has received widespread attention from the medical community as an emerging and promising therapeutic approach to cancer treatment. Among all gas molecules, nitric oxide (NO) was the first one to be applied in the biomedical field for its intriguing properties and unique anti-tumor mechanisms which have become a research hotspot in recent years. Despite the great progress of NO in cancer therapy, the non-specific distribution of NO in vivo and its side effects on normal tissue at high concentrations have impaired its clinical application. Therefore, it is important to develop facile NO-based nanomedicines to achieve the on-demand release of NO in tumor tissue while avoiding the leakage of NO in normal tissue, which could enhance therapeutic efficacy and reduce side effects at the same time. In recent years, numerous studies have reported the design and development of NO-based nanomedicines which were triggered by exogenous stimulus (light, ultrasound, X-ray) or tumor endogenous signals (glutathione, weak acid, glucose). In this review, we summarized the design principles and release behaviors of NO-based nanomedicines upon various stimuli and their applications in synergistic cancer therapy. We also discuss the anti-tumor mechanisms of NO-based nanomedicines in vivo for enhanced cancer therapy. Moreover, we discuss the existing challenges and further perspectives in this field in the aim of furthering its development.
Collapse
Affiliation(s)
- Yijun Zhao
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China; (Y.Z.); (X.O.)
| | - Xumei Ouyang
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China; (Y.Z.); (X.O.)
| | - Yongjun Peng
- The Department of Medical Imaging, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China; (Y.Z.); (X.O.)
| |
Collapse
|
40
|
Zeng W, Liu C, Wang S, Wang Z, Huang Q. SnFe 2O 4 Nanozyme Based TME Improvement System for Anti-Cancer Combination Thermoradiotherapy. Front Oncol 2021; 11:768829. [PMID: 34746011 PMCID: PMC8564484 DOI: 10.3389/fonc.2021.768829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022] Open
Abstract
High doses of radiotherapy (RT) are associated with resistance induction. Therefore, highly selective and controllable radiosensitizers are urgently needed. To address this issue, we developed a tin ferrite (SFO)-based tumor microenvironment (TME)-improved system (SIS) that can be used in combination with low-dose radiation. The SIS was delivered via intratumoral injection directly to the tumor site, where it was stored as a ration depot. Due to the photothermal properties of SFO, SIS steadily dissolved under near-infrared (NIR) laser irradiation. Simultaneously, the dual glutathione oxidase (GSH-OXD) and catalase (CAT) activities of the SFO nanozyme significantly lowered the content of GSH in tumor tissues and efficiently catalyzed the conversion of intracellular hydrogen peroxide to produce a large amount of oxygen (O2) for intracellular redox homeostasis disruption, thus reducing radiotherapy resistance. Our in vivo and in vitro studies suggested that combining the SIS and NIR irradiation with RT (2Gy) significantly reduced tumor proliferation without side effects such as inflammation. To conclude, this study revealed that SFO-based nanozymes show great promise as a catalytic, radiosensitizing anti-tumor therapy.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Molecular Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuntao Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziqi Wang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, China
| | - Qinqin Huang
- Department of Molecular Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
41
|
Liu W, Chen B, Zheng H, Xing Y, Chen G, Zhou P, Qian L, Min Y. Advances of Nanomedicine in Radiotherapy. Pharmaceutics 2021; 13:pharmaceutics13111757. [PMID: 34834172 PMCID: PMC8622383 DOI: 10.3390/pharmaceutics13111757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) remains one of the current main treatment strategies for many types of cancer. However, how to improve RT efficiency while reducing its side effects is still a large challenge to be overcome. Advancements in nanomedicine have provided many effective approaches for radiosensitization. Metal nanoparticles (NPs) such as platinum-based or hafnium-based NPs are proved to be ideal radiosensitizers because of their unique physicochemical properties and high X-ray absorption efficiency. With nanoparticles, such as liposomes, bovine serum albumin, and polymers, the radiosensitizing drugs can be promoted to reach the tumor sites, thereby enhancing anti-tumor responses. Nowadays, the combination of some NPs and RT have been applied to clinical treatment for many types of cancer, including breast cancer. Here, as well as reviewing recent studies on radiotherapy combined with inorganic, organic, and biomimetic nanomaterials for oncology, we analyzed the underlying mechanisms of NPs radiosensitization, which may contribute to exploring new directions for the clinical translation of nanoparticle-based radiosensitizers.
Collapse
Affiliation(s)
- Wei Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
| | - Bo Chen
- Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China; (B.C.); (Y.M.)
| | - Haocheng Zheng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Yun Xing
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Guiyuan Chen
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Peijie Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
| | - Liting Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
- Correspondence:
| | - Yuanzeng Min
- Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China; (B.C.); (Y.M.)
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
42
|
Sun X, Li L, Zhang H, Dong M, Wang J, Jia P, Bu T, Wang X, Wang L. Near-Infrared Light-Regulated Drug-Food Homologous Bioactive Molecules and Photothermal Collaborative Precise Antibacterial Therapy Nanoplatform with Controlled Release Property. Adv Healthc Mater 2021; 10:e2100546. [PMID: 34081401 DOI: 10.1002/adhm.202100546] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/30/2021] [Indexed: 12/28/2022]
Abstract
Herein, a collaborative precise antibacterial wound healing therapy nanoplatform integrating drug-food homologous bioactive molecule (cinnamaldehyde, CA) with photothermal therapy (PTT) is presented. Copper-gallic acids-cinnamaldehyde-polydopamine nanorods (Cu-GA-CA-PDA NRs) with near-infrared light (NIR)-controlled CA release property are fabricated, which also integrate CA and photothermal synergistic sterilization, as well as antioxidant, anti-inflammatory, and anti-infection capacities. The characteristics of NIR-mediated CA release and photothermal response of Cu-GA-CA-PDA NRs support their excellent sterilization performance in vitro/in vivo. In addition, under the guidance of NIR, Cu-GA-CA-PDA NRs can hinder the formation of inflammatory cells, reduce oxidative stress damage, accelerate the regeneration of skin tissues in S. aureus-infected wound sites, and achieve the goal of promoting wound healing. Therefore, NIR-mediated Cu-GA-CA-PDA NRs with multifunctional biological activities provide a highly competitive strategy for curing bacteria-infected wounds.
Collapse
Affiliation(s)
- Xinyu Sun
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Lihua Li
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Hui Zhang
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Mengna Dong
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Jiao Wang
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Pei Jia
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Tong Bu
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Xin Wang
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| | - Li Wang
- College of Food Science and Engineering Northwest A&F University Yangling Shaanxi 712100 P. R. China
| |
Collapse
|
43
|
Li S, Xu S, Liang X, Xue Y, Mei J, Ma Y, Liu Y, Liu Y. Nanotechnology: Breaking the Current Treatment Limits of Lung Cancer. Adv Healthc Mater 2021; 10:e2100078. [PMID: 34019739 DOI: 10.1002/adhm.202100078] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/03/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer is one of the most rapidly growing malignancies in terms of morbidity and mortality. Although traditional treatments have been used for more than 50 years, it is still far from solving the problems of postoperative risks and systemic toxicity. Emerging targeting and immunotherapy are developing continuously and are gaining recognition; eventually, they face the inevitable challenge of drug resistance. Nanotechnology has several important effects on lung cancer treatment, owing to its unique properties. Several nanoparticle-based treatments have successfully become cancer treatments. Good biocompatibility with higher specific surface area can carry substantial amounts of lung cancer treatment medications while avoiding medication toxicity; editable and modified characteristics give rise to multifunctional nanomedicines; excellent photoelectric effects make lung cancer a multimodal treatment. This article summarizes the breakthroughs achieved by nanotechnology, targeted therapy, and immunotherapy, reflecting the importance and necessity of nanotechnology in the treatment of lung cancer.
Collapse
Affiliation(s)
- Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Shanshan Xu
- Institute for Advanced Study Shenzhen University Shenzhen Guangdong 518060 P. R. China
| | - Xiaoyu Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing 100190 P. R. China
- School of Pharmaceutical Sciences of Zhengzhou University Zhengzhou Henan 450001 P. R. China
| | - Yueguang Xue
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing 100190 P. R. China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Yongfu Ma
- Department of Thoracic Surgery Chinese PLA General Hospital Beijing 100853 P. R. China
| | - Yang Liu
- Department of Thoracic Surgery Chinese PLA General Hospital Beijing 100853 P. R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing 100190 P. R. China
- The GBA National Institute for Nanotechnology Innovation Guangzhou Guangdong 510700 P. R. China
| |
Collapse
|
44
|
Garren MR, Ashcraft M, Qian Y, Douglass M, Brisbois EJ, Handa H. Nitric oxide and viral infection: Recent developments in antiviral therapies and platforms. APPLIED MATERIALS TODAY 2021; 22:100887. [PMID: 38620577 PMCID: PMC7718584 DOI: 10.1016/j.apmt.2020.100887] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/11/2020] [Accepted: 11/14/2020] [Indexed: 05/09/2023]
Abstract
Nitric oxide (NO) is a gasotransmitter of great significance to developing the innate immune response to many bacterial and viral infections, while also modulating vascular physiology. The generation of NO from the upregulation of endogenous nitric oxide synthases serves as an efficacious method for inhibiting viral replication in host defense and warrants investigation for the development of antiviral therapeutics. With increased incidence of global pandemics concerning several respiratory-based viral infections, it is necessary to develop broad therapeutic platforms for inhibiting viral replication and enabling more efficient host clearance, as well as to fabricate new materials for deterring viral transmission from medical devices. Recent developments in creating stabilized NO donor compounds and their incorporation into macromolecular scaffolds and polymeric substrates has created a new paradigm for developing NO-based therapeutics for long-term NO release in applications for bactericidal and blood-contacting surfaces. Despite this abundance of research, there has been little consideration of NO-releasing scaffolds and substrates for reducing passive transmission of viral infections or for treating several respiratory viral infections. The aim of this review is to highlight the recent advances in developing gaseous NO, NO prodrugs, and NO donor compounds for antiviral therapies; discuss the limitations of NO as an antiviral agent; and outline future prospects for guiding materials design of a next generation of NO-releasing antiviral platforms.
Collapse
Key Words
- ACE, angiotensin converting enzyme
- AP1, activator protein 1
- COVID-19
- COVID-19, coronavirus disease 2019
- ECMO, extracorporeal membrane oxygenation, FDA, United States Food and Drug Administration
- GNSO, S-nitrosoglutathione
- H1N1, influenza A virus subtype H1N1
- HI, Host Immunology
- HIV, human immunodeficiency virus
- HPV, human papillomavirus
- HSV, herpes simplex virus
- I/R, pulmonary ischemia-reperfusion
- IC50, inhibitory concentration 50
- IFN, interferon
- IFNγ, interferon gamma
- IKK, inhibitor of nuclear factor kappa B kinase
- IRF-1, interferon regulatory factor 1
- Inhalation therapy
- Medical Terminology: ARDS, acute respiratory distress syndrome
- NF-κB, nuclear factor kappa-light-chain enhancer of activated B cells
- NO, nitric oxide
- NOS, nitric oxide synthase
- Nitric Oxide and Related Compounds: eNOS/NOS 3, endothelial nitric oxide synthase
- Nitric oxide
- Other: DNA, deoxyribonucleic acid
- P38-MAPK, P38 mitogen-activated protein kinases
- PAMP, pathogen-associated molecular pattern
- PCV2, porcine circovirus type 2
- PHT, pulmonary hypertension
- PKR, protein kinase R
- RNA, ribonucleic acid
- RNI, reactive nitrogen intermediate
- RSNO, S-nitrosothiol
- SARS, severe acute respiratory syndrome
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SNAP, S-nitroso-N-acetyl-penicillamine
- STAT-1, signal transducer and activator of transcription 1
- Severe acute respiratory distress
- TAK1, transforming growth factor β-activated kinases-1
- TLR, toll-like receptor
- VAP, ventilator associated pneumonia
- Viral infection
- Viruses: CVB3, coxsackievirus
- dsRNA, double stranded (viral) ribonucleic acid
- gNO, gaseous nitric oxide
- iNOS/NOS 2, inducible nitric oxide synthase
- mtALDH, mitochondrial aldehyde dehydrogenase
- nNOS/NOS 1, neuronal nitric oxide synthase
Collapse
Affiliation(s)
- Mark R Garren
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Morgan Ashcraft
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Yun Qian
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Megan Douglass
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Elizabeth J Brisbois
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Hitesh Handa
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| |
Collapse
|
45
|
Yang H, Jiang F, Zhang L, Wang L, Luo Y, Li N, Guo Y, Wang Q, Zou J. Multifunctional l-arginine-based magnetic nanoparticles for multiple-synergistic tumor therapy. Biomater Sci 2021; 9:2230-2243. [PMID: 33507174 DOI: 10.1039/d0bm01932a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor therapy is facing the big challenge of insufficient treatment. Here, we report high-intensity focused ultrasound (HIFU)-responsive magnetic nanoparticles based on superparamagnetic iron oxide (SPIO, Fe3O4 NPs) as the shell and l-arginine (LA) as the core entrapped by poly-lactide-co-glycolide (PLGA) nanoparticles (Fe3O4@PLGA/LA NPs) for synergistic breast cancer therapy. These NPs can significantly enhance therapeutic performance due to their enhanced accumulation and prolonged retention at the tumor site under magnetic guidance. The Fe3O4@PLGA/LA NPs exhibited synergistic therapeutic effects by the rational combination of HIFU-based tumor ablation and nitric oxide (NO) assisted antitumor gas therapy. Both Fe3O4 NPs and LA could be released rapidly under HIFU irradiation, where Fe3O4 NPs can promote HIFU-based tumor ablation by changing the acoustic properties of the tumor tissues and LA can spontaneously react with hydrogen peroxide (H2O2) in the tumor microenvironment to generate NO for gas therapy. Moreover, Fe3O4 NPs can react with H2O2 to produce highly reactive oxygen-containing species (ROS) to accelerate the oxidation of LA and the release of NO. This novel strategy showed synergistic tumor growth suppression as compared with individual HIFU therapy or gas therapy. This can be attributed to the rational design of multifunctional NPs with magnetic targeting and multi-modality imaging properties.
Collapse
Affiliation(s)
- Haiyan Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
A hybrid semiconducting organosilica-based O 2 nanoeconomizer for on-demand synergistic photothermally boosted radiotherapy. Nat Commun 2021; 12:523. [PMID: 33483518 PMCID: PMC7822893 DOI: 10.1038/s41467-020-20860-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
The outcome of radiotherapy is significantly restricted by tumor hypoxia. To overcome this obstacle, one prevalent solution is to increase intratumoral oxygen supply. However, its effectiveness is often limited by the high metabolic demand for O2 by cancer cells. Herein, we develop a hybrid semiconducting organosilica-based O2 nanoeconomizer pHPFON-NO/O2 to combat tumor hypoxia. Our solution is twofold: first, the pHPFON-NO/O2 interacts with the acidic tumor microenvironment to release NO for endogenous O2 conservation; second, it releases O2 in response to mild photothermal effect to enable exogenous O2 infusion. Additionally, the photothermal effect can be increased to eradicate tumor residues with radioresistant properties due to other factors. This “reducing expenditure of O2 and broadening sources” strategy significantly alleviates tumor hypoxia in multiple ways, greatly enhances the efficacy of radiotherapy both in vitro and in vivo, and demonstrates the synergy between on-demand temperature-controlled photothermal and oxygen-elevated radiotherapy for complete tumor response. Tumor hypoxia is a major limitation in radiotherapy, and strategies to address this often fail due to high oxygen consumption. Here, the authors report a nanomaterial assembly for the simultaneous reduction in mitochondrial respiration and to supply oxygen to potentiate radiotherapy.
Collapse
|
47
|
A Single-wavelength NIR-triggered Polymer for in Situ Generation of Peroxynitrite (ONOO−) to Enhance Phototherapeutic Efficacy. CHINESE JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1007/s10118-021-2540-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
48
|
Wang Z, Zhan M, Li W, Chu C, Xing D, Lu S, Hu X. Photoacoustic Cavitation‐Ignited Reactive Oxygen Species to Amplify Peroxynitrite Burst by Photosensitization‐Free Polymeric Nanocapsules. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Zhixiong Wang
- Guangdong Provincial Key Laboratory of Laser Life Science MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 China
- College of Biophotonics South China Normal University Guangzhou 510631 China
| | - Meixiao Zhan
- Zhuhai Precision Medical Center, Zhuhai People's Hospital Zhuhai Hospital Affiliated with Jinan University Jinan University Zhuhai Guangdong 519000 China
| | - Weijie Li
- Guangdong Provincial Key Laboratory of Laser Life Science MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 China
- College of Biophotonics South China Normal University Guangzhou 510631 China
| | - Chengyan Chu
- Guangdong Provincial Key Laboratory of Laser Life Science MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 China
- College of Biophotonics South China Normal University Guangzhou 510631 China
| | - Da Xing
- Guangdong Provincial Key Laboratory of Laser Life Science MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 China
- College of Biophotonics South China Normal University Guangzhou 510631 China
| | - Siyu Lu
- Green Catalysis Center College of Chemistry Zhengzhou University Zhengzhou 450000 China
| | - Xianglong Hu
- Guangdong Provincial Key Laboratory of Laser Life Science MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 China
- College of Biophotonics South China Normal University Guangzhou 510631 China
| |
Collapse
|
49
|
Wang Z, Zhan M, Li W, Chu C, Xing D, Lu S, Hu X. Photoacoustic Cavitation-Ignited Reactive Oxygen Species to Amplify Peroxynitrite Burst by Photosensitization-Free Polymeric Nanocapsules. Angew Chem Int Ed Engl 2021; 60:4720-4731. [PMID: 33210779 DOI: 10.1002/anie.202013301] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/06/2020] [Indexed: 12/25/2022]
Abstract
Photoacoustic (PA) technology can transform light energy into acoustic wave, which can be used for either imaging or therapy that depends on the power density of pulsed laser. Here, we report photosensitizer-free polymeric nanocapsules loaded with nitric oxide (NO) donors, namely NO-NCPs, formulated from NIR light-absorbable amphiphilic polymers and a NO-releasing donor, DETA NONOate. Controlled NO release and nanocapsule dissociation are achieved in acidic lysosomes of cancer cells. More importantly, upon pulsed laser irradiation, the PA cavitation can excite water to generate significant reactive oxygen species (ROS) such as superoxide radical (O2 .- ), which further spontaneously reacts with the in situ released NO to burst highly cytotoxic peroxynitrite (ONOO- ) in cancer cells. The resultant ONOO- generation greatly promotes mitochondrial damage and DNA fragmentation to initiate programmed cancer cell death. Apart from PA imaging, PA cavitation can intrinsically amplify reactive species via photosensitization-free materials for promising disease theranostics.
Collapse
Affiliation(s)
- Zhixiong Wang
- Guangdong Provincial Key Laboratory of Laser Life Science, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Meixiao Zhan
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Weijie Li
- Guangdong Provincial Key Laboratory of Laser Life Science, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Chengyan Chu
- Guangdong Provincial Key Laboratory of Laser Life Science, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Da Xing
- Guangdong Provincial Key Laboratory of Laser Life Science, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Siyu Lu
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, 450000, China
| | - Xianglong Hu
- Guangdong Provincial Key Laboratory of Laser Life Science, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
50
|
Xia D, Hang D, Li Y, Jiang W, Zhu J, Ding Y, Gu H, Hu Y. Au-Hemoglobin Loaded Platelet Alleviating Tumor Hypoxia and Enhancing the Radiotherapy Effect with Low-Dose X-ray. ACS NANO 2020; 14:15654-15668. [PMID: 33108152 DOI: 10.1021/acsnano.0c06541] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Radiotherapy (RT) is a widely explored clinical modality to combat cancer. However, its therapeutic efficacy is not always satisfied because of the severe hypoxic microenvironment in solid tumors and the high dosage of radiation harmful to the adjacent healthy tissue. Herein, Au nanoparticle-hemoglobin complex nanoparticle loaded platelets (Au-Hb@PLT) were fabricated. These Au-Hb@PLT would be activated by tumor cells, and the formed platelet-derivate particles (PM) could deliver Au nanoparticle-hemoglobin complex deeply into tumor tissue because of their small size and tumor homing ability. Hemoglobin acts as an oxygen carrier to relieve the hypoxia and gold nanoparticles work as radiosensitizers to potentiate the sensitivity of tumor cells to X-ray, thus, enhancing the in vivo therapeutic outcome even under a low-dose RT in tumor bearing mice. The enhanced antitumor effect and survival benefits endowed by the Au-Hb@PLT were confirmed in vitro and in vivo. These results demonstrate that these Au-Hb@PLT can work as an oxygen vehicle, offer a promising approach to mitigate hypoxia and improve RT efficacy with a low RT dosage.
Collapse
Affiliation(s)
- Donglin Xia
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, P. R. China
- School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Daming Hang
- Nantong Tumor Hospital, Nantong, Jiangsu 226362, P.R. China
| | - Yuanyuan Li
- School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Wei Jiang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, P. R. China
| | - Jianfeng Zhu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, P. R. China
| | - Yin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P. R. China
| | - Haiying Gu
- School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, P. R. China
| |
Collapse
|