1
|
Song Y, Tan KB, Zhou SF, Zhan G. Biocompatible Copper-Based Nanocomposites for Combined Cancer Therapy. ACS Biomater Sci Eng 2024; 10:3673-3692. [PMID: 38717176 DOI: 10.1021/acsbiomaterials.4c00586] [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] [Indexed: 06/11/2024]
Abstract
Copper (Cu) and Cu-based nanomaterials have received tremendous attention in recent years because of their unique physicochemical properties and good biocompatibility in the treatment of various diseases, especially cancer. To date, researchers have designed and fabricated a variety of integrated Cu-based nanocomplexes with distinctive nanostructures and applied them in cancer therapy, mainly including chemotherapy, radiotherapy (RT), photothermal therapy (PTT), chemodynamic therapy (CDT), photodynamic therapy (PDT), cuproptosis-mediated therapy, etc. Due to the limited effect of a single treatment method, the development of composite diagnostic nanosystems that integrate chemotherapy, PTT, CDT, PDT, and other treatments is of great significance and offers great potential for the development of the next generation of anticancer nanomedicines. In view of the rapid development of Cu-based nanocomplexes in the field of cancer therapy, this review focuses on the current state of research on Cu-based nanomaterials, followed by a discussion of Cu-based nanocomplexes for combined cancer therapy. Moreover, the current challenges and future prospects of Cu-based nanocomplexes in clinical translation are proposed to provide some insights into the design of integrated Cu-based nanotherapeutic platforms.
Collapse
Affiliation(s)
- Yibo Song
- College of Chemical Engineering, Academy of Advanced Carbon Conversion Technology, Huaqiao University, 668 Jimei Avenue, Xiamen, 361021 Fujian, P. R. China
| | - Kok Bing Tan
- College of Chemical Engineering, Academy of Advanced Carbon Conversion Technology, Huaqiao University, 668 Jimei Avenue, Xiamen, 361021 Fujian, P. R. China
| | - Shu-Feng Zhou
- College of Chemical Engineering, Academy of Advanced Carbon Conversion Technology, Huaqiao University, 668 Jimei Avenue, Xiamen, 361021 Fujian, P. R. China
| | - Guowu Zhan
- College of Chemical Engineering, Academy of Advanced Carbon Conversion Technology, Huaqiao University, 668 Jimei Avenue, Xiamen, 361021 Fujian, P. R. China
| |
Collapse
|
2
|
Akbarzadeh F, Khoshgard K. Enhancement of the effect of novel targeted 5-aminolevulinic acid conjugated bismuth oxide nanoparticles-based photodynamic therapy by simultaneous radiotherapy on KB cells. Photodiagnosis Photodyn Ther 2024; 46:104025. [PMID: 38403143 DOI: 10.1016/j.pdpdt.2024.104025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Selective accumulation of photosensitizers into cancerous cells is one of the most important factors affecting photodynamic therapy (PDT) efficacy. 5-aminolevulinic acid (5-ALA) is the precursor of a strong photosensitizer, protoporphyrin-IX; but it has poor permeability into the cells. Folate receptors are overexpressed on the surface of many tumor cells. In the present study, folic acid (FA) and 5-ALA conjugated bismuth oxide nanoparticles were synthesized; and used in PDT, radiotherapy (RT), and concurrent PDT & RT against nasopharyngeal carcinoma (KB cell line). METHODS The KB cells were incubated with the synthesized nanoparticles (NPs) for 2 h; then illuminated using a custom-made LED lamp at the light dose of 26 J/cm2. Irradiation of the cells was carried out using X-ray 6 MV (2 Gy); and synergistic effect of the simultaneous RT and PDT treatments was evaluated using fractional product values. Efficacy of the treatments was determined using MTT and Caspase-3 enzyme activity assays. RESULTS Targeting of folic acid receptors enables the selective endocytosis of the conjugated NPs. RT results in the presence of Bi2O3 NPs showed a significant radiosensitizer potential of these NPs. Fractional product values of 1.49±0.05, 1.36±0.06, and 1.05±0.06 obtained in the presence of FA-5-ALA conjugated NPs, 5-ALA conjugated NPs, and in the absence of the NPs, respectively. Therefore, simultaneous RT and PDT in the presence of these conjugated NPs is superior to RT in the presence of the NPs. CONCLUSION Simultaneous PDT and RT in the presence of FA-5-ALA conjugated bismuth oxide NPs can be introduced as a promising therapeutic approach in controlling KB cancer cells.
Collapse
Affiliation(s)
- Fatemeh Akbarzadeh
- Students Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Karim Khoshgard
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Sorkheh-Lizhe Blvd, Kermanshah, P.O.Box:1568, Iran.
| |
Collapse
|
3
|
Xu Y, Lai H, Pan S, Pan L, Liu T, Yang Z, Chen T, Zhu X. Selenium promotes immunogenic radiotherapy against cervical cancer metastasis through evoking P53 activation. Biomaterials 2024; 305:122452. [PMID: 38154440 DOI: 10.1016/j.biomaterials.2023.122452] [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: 10/14/2023] [Revised: 12/06/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
Radiotherapy is still the recommended treatment for cervical cancer. However, radioresistance and radiation-induced side effects remain one of the biggest clinical problems. Selenium (Se) has been confirmed to exhibit radiation-enhancing effects for cancer treatment. However, Se species dominate the biological activities and which form of Se possesses better radiosensitizing properties and radiation safety remains elusive. Here, different Se species (the valence state of Se ranged from - 2, 0, +4 to + 6) synergy screen was carried out to identify the potential radiosensitizing effects and radiation safety of Se against cervical cancer. We found that the therapeutic effects varied with the changes in the Se valence state. Sodium selenite (+4) displayed strong cancer-killing effects but also possessed severe cytotoxicity. Sodium selenate (+6) neither enhanced the killing effects of X-ray nor possessed anticancer activity by its alone treatment. Although nano-selenium (0), especially Let-SeNPs, has better radiosensitizing activity, the - 2 organic Se, such as selenadiazole derivative SeD (-2) exhibited more potent anticancer effects and possessed a higher safe index. Overall, the selected Se drugs were able to synergize with X-ray to inhibit cell growth, clone formation, and cell migration by triggering G2/M phase arrest and apoptosis, and SeD (-2) was found to exhibit more potent enhancing capacity. Further mechanism studies showed that SeD mediated p53 pathway activation by inducing DNA damage through promoting ROS production. Additionally, SeD combined with X-ray therapy can induce an anti-tumor immune response in vivo. More importantly, SeD combined with X-ray significantly inhibited the liver metastasis of tumor cells and alleviated the side effects caused by radiation therapy in tumor-bearing mice. Taken together, this study demonstrates the radiosensitization and radiation safety effects of different Se species, which may shed light on the application of such Se-containing drugs serving as side effects-reducing agents for cervical cancer radiation treatment.
Collapse
Affiliation(s)
- Yanchao Xu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China; Department of Chemistry, Jinan University, China
| | - Haoqiang Lai
- Department of Chemistry, Jinan University, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Liuliu Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Ting Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Ziyi Yang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Tianfeng Chen
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China; Department of Chemistry, Jinan University, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China.
| |
Collapse
|
4
|
Singh N, Kulkarni PP, Tripathi P, Agarwal V, Dash D. Nanogold-coated stent facilitated non-invasive photothermal ablation of stent thrombosis and restoration of blood flow. NANOSCALE ADVANCES 2024; 6:1497-1506. [PMID: 38419863 PMCID: PMC10898437 DOI: 10.1039/d3na00751k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
In-stent restenosis (ISR) and stent thrombosis (ST) are the most serious complications of coronary angioplasty and stenting. Although the evolution of drug-eluting stents (DES) has significantly restricted the incidence of ISR, they are associated with an enhanced risk of ST. In the present study, we explore the photothermal ablation of a thrombus using a nano-enhanced thermogenic stent (NETS) as a modality for revascularization following ST. The photothermal activity of NETS, fabricated by coating bare metal stents with gold nanorods generating a thin plasmonic film of gold, was found to be effective in rarefying clots formed within the stent lumen in various in vitro assays including those under conditions mimicking blood flow. NETS implanted in the rat common carotid artery generated heat following exposure to a NIR-laser that led to effective restoration of blood flow within the occluded vessel in a model of ferric chloride-induced thrombosis. Our results present a proof-of-concept for a novel photothermal ablation approach by employing coated stents in the non-invasive management of ST.
Collapse
Affiliation(s)
- Nitesh Singh
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| | - Paresh P Kulkarni
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| | - Prashant Tripathi
- School of Physical Sciences, Jawaharlal Nehru University New Mehrauli Road New Delhi Delhi-110067 India
| | - Vikas Agarwal
- Department of Cardiology, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| | - Debabrata Dash
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| |
Collapse
|
5
|
Mamatha M, Ansari MA, Begum MY, Prasad B. D, Al Fatease A, Hani U, Alomary MN, Sultana S, Punekar SM, M.B. N, Lakshmeesha TR, Ravikiran T. Green Synthesis of Cerium Oxide Nanoparticles, Characterization, and Their Neuroprotective Effect on Hydrogen Peroxide-Induced Oxidative Injury in Human Neuroblastoma (SH-SY5Y) Cell Line. ACS OMEGA 2024; 9:2639-2649. [PMID: 38250384 PMCID: PMC10795031 DOI: 10.1021/acsomega.3c07505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
Cerium oxide nanoparticles (CeO2NPs) have a broad scale of applications in the biomedical field due to their excellent physicochemical and catalytic properties. The present study aims to synthesize the CeO2NPs from Centella asiatica (C. asiatica) leaf extract, which has been used in Indian traditional medicine for its neuroprotective properties. The CeO2NPs were characterized by ultraviolet-visible, X-ray diffraction, Fourier transform infrared, Raman spectroscopy, scanning electron microscopy- energy dispersive X-ray spectroscopy, and high-resolution transmission electron microscopy. The antioxidant property was evaluated by 2,2-di (4-tert-octyl phenyl)-1-picrylhydrazyl and OH radical assays. The neuroprotective potential was assessed against the oxidative stress (OS) induced by H2O2 in the human neuroblastoma (SH-SY5Y) cell line. CeO2NPs exhibited significant DPPH and OH radical scavenging activity. Our results revealed that CeO2NPs significantly increased H2O2-induced cell viability, decreased lactate dehydrogenase, protein carbonyls, reactive oxygen species generation, apoptosis, and upregulated antioxidant enzyme activity. Our findings suggest that the CeO2NPs protect the SH-SY5Y cells from OS and apoptosis, which could potentially counter OS-related neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Mohammad Azam Ansari
- Department
of Epidemic Disease Research, Institutes for Research and Medical
Consultations (IRMC), Imam, Abdulrahman
Bin Faisal University, Dammam 31441, Saudi Arabia
| | - M Yasmin Begum
- Department
of Pharmaceutics, College of Pharmacy, King
Khalid University, Abha 61421, Saudi Arabia
| | - Daruka Prasad B.
- Department
of Physics, B.M.S. Institute of Technology, Bengaluru 560064, Karnataka, India
| | - Adel Al Fatease
- Department
of Pharmaceutics, College of Pharmacy, King
Khalid University, Abha 61421, Saudi Arabia
| | - Umme Hani
- Department
of Pharmaceutics, College of Pharmacy, King
Khalid University, Abha 61421, Saudi Arabia
| | - Mohammad N. Alomary
- Advanced
Diagnostic and Therapeutic Institute, King
Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Sumreen Sultana
- Department
of Microbiology and Biotechnology, Bangalore
University, Bengaluru 560056, Karnataka, India
| | - Shital Manohar Punekar
- Department
of Microbiology and Biotechnology, Bangalore
University, Bengaluru 560056, Karnataka, India
| | - Nivedika M.B.
- Department
of Microbiology and Biotechnology, Bangalore
University, Bengaluru 560056, Karnataka, India
| | | | - Tekupalli Ravikiran
- Department
of Microbiology and Biotechnology, Bangalore
University, Bengaluru 560056, Karnataka, India
| |
Collapse
|
6
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
7
|
Ni Y, Deng P, Yin R, Zhu Z, Ling C, Ma M, Wang J, Li S, Liu R. Effect and mechanism of paclitaxel loaded on magnetic Fe 3O 4@mSiO 2-NH 2-FA nanocomposites to MCF-7 cells. Drug Deliv 2023; 30:64-82. [PMID: 36474448 PMCID: PMC9744220 DOI: 10.1080/10717544.2022.2154411] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Magnetic Fe3O4 nanoparticles were prepared via a simple hydrothermal method and utilized to load paclitaxel. The average particle size of Fe3O4 nanoparticles was found to be 20.2 ± 3.0 nm, and the calculated saturation magnetization reached 129.38 emu/g, verifying superparamagnetism of nanomaterials. The specific surface area and pore volume were 84.756 m2/g and 0.265 cm3/g, respectively. Subsequently, Fe3O4@mSiO2 nanoparticles were successfully fabricated using the Fe3O4 nanoparticles as precursors with an average size of 27.81 nm. The relevant saturation magnetization, zeta potential, and specific surface area of Fe3O4@mSiO2-NH2-FA were respectively 76.3 emu/g, -14.1 mV, and 324.410 m2/g. The pore volume and average adsorption pore size were 0.369 cm3/g and 4.548 nm, respectively. Compared to free paclitaxel, the solubility and stability of nanoparticles loaded with paclitaxel were improved. The drug loading efficiency and drug load of the nanoformulation were 44.26 and 11.38%, respectively. The Fe3O4@mSiO2-NH2-FA nanocomposites were easy to construct with excellent active targeting performance, pH sensitivity, and sustained-release effect. The nanoformulation also showed good biocompatibility, where the cell viability remained at 73.8% when the concentration reached 1200 μg/mL. The nanoformulation induced cell death through apoptosis, as confirmed by AO/EB staining and flow cytometry. Western blotting results suggested that the nanoformulation could induce iron death by inhibiting Glutathione Peroxidase 4 (GPX4) activity or decreasing Ferritin Heavy Chain 1 (FTH1) expression. Subsequently, the expression of HIF-1α was upregulated owing to the accumulation of reactive oxygen species (ROS), thus affecting the expression of apoptosis-related proteins regulated by p53, inducing cell apoptosis.
Collapse
Affiliation(s)
- Yun Ni
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Peng Deng
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, P.R. China
| | - Ruitong Yin
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Ziye Zhu
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Chen Ling
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Mingyi Ma
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Jie Wang
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Shasha Li
- Affiliated Kunshan Hospital, Jiangsu University, Suzhou, P.R. China,CONTACT Shasha Li
| | - Ruijiang Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China,Ruijiang Liu
| |
Collapse
|
8
|
Zhang J, Li L, Shang H, Feng Z, Chao T. A molecular classification system for estimating radiotherapy response and anticancer immunity for individual breast cancer patients. Front Oncol 2023; 13:1288698. [PMID: 37927478 PMCID: PMC10623135 DOI: 10.3389/fonc.2023.1288698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
Objective Radiotherapy is a cornerstone of breast cancer therapy, but radiotherapy resistance is a major clinical challenge. Herein, we show a molecular classification approach for estimating individual responses to radiotherapy. Methods Consensus clustering was adopted to classify radiotherapy-sensitive and -resistant clusters in the TCGA-BRCA cohort based upon prognostic differentially expressed radiotherapy response-related genes (DERRGs). The stability of the classification was proven in the GSE58812 cohort via NTP method and the reliability was further verified by quantitative RT-PCR analyses of DERRGs. A Riskscore system was generated through Least absolute shrinkage and selection operator (LASSO) analysis, and verified in the GSE58812 and GSE17705. Treatment response and anticancer immunity were evaluated via multiple well-established computational approaches. Results We classified breast cancer patients as radiotherapy-sensitive and -resistant clusters, namely C1 and C2, also verified by quantitative RT-PCR analyses of DERRGs. Two clusters presented heterogeneous clinical traits, with poorer prognosis, older age, more advanced T, and more dead status in the C2. The C1 tumors had higher activity of reactive oxygen species and response to X-ray, proving better radiotherapeutic response. Stronger anticancer immunity was found in the C1 tumors that had rich immune cell infiltration, similar expression profiling to patients who responded to anti-PD-1, and activated immunogenic cell death and ferroptosis. The Riskscore was proposed for improving patient prognosis. High Riskscore samples had lower radiotherapeutic response and stronger DNA damage repair as well as poor anticancer immunity, while low Riskscore samples were more sensitive to docetaxel, doxorubicin, and paclitaxel. Conclusion Our findings propose a novel radiotherapy response classification system based upon molecular profiles for estimating radiosensitivity for individual breast cancer patients, and elucidate a methodological advancement for synergy of radiotherapy with ICB.
Collapse
Affiliation(s)
- Jiaxuan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Long Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haotian Shang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhaoyan Feng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
9
|
Biagioni A, Peri S, Versienti G, Fiorillo C, Becatti M, Magnelli L, Papucci L. Gastric Cancer Vascularization and the Contribution of Reactive Oxygen Species. Biomolecules 2023; 13:886. [PMID: 37371466 DOI: 10.3390/biom13060886] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Blood vessels are the most important way for cancer cells to survive and diffuse in the body, metastasizing distant organs. During the process of tumor expansion, the neoplastic mass progressively induces modifications in the microenvironment due to its uncontrolled growth, generating a hypoxic and low pH milieu with high fluid pressure and low nutrients concentration. In such a particular condition, reactive oxygen species play a fundamental role, enhancing tumor proliferation and migration, inducing a glycolytic phenotype and promoting angiogenesis. Indeed, to reach new sources of oxygen and metabolites, highly aggressive cancer cells might produce a new abnormal network of vessels independently from endothelial cells, a process called vasculogenic mimicry. Even though many molecular markers and mechanisms, especially in gastric cancer, are still unclear, the formation of such intricate, leaky and abnormal vessel networks is closely associated with patients' poor prognosis, and therefore finding new pharmaceutical solutions to be applied along with canonical chemotherapies in order to control and normalize the formation of such networks is urgent.
Collapse
Affiliation(s)
- Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Sara Peri
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
10
|
Size-Controllable Nanosystem with Double Responsive for Deep Photodynamic Therapy. Pharmaceutics 2023; 15:pharmaceutics15030940. [PMID: 36986801 PMCID: PMC10056800 DOI: 10.3390/pharmaceutics15030940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/16/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising strategy for cancer treatment. However, a poor tissue penetration of activation light and low target specificity seriously hindered the clinical application of PDT. Here, we designed and constructed a size-controllable nanosystem (UPH) with inside-out responsive for deep PDT with enhanced biosafety. To obtain nanoparticles with the best quantum yield, a series of core-shell nanoparticles (UCNP@nPCN) with different thicknesses were synthesized by a layer-by-layer self-assembly method to incorporate a porphyritic porous coordination network (PCN) onto the surface of upconverting nanoparticles (UCNPs), followed by coating with hyaluronic acid (HA) on the surface of nanoparticles with optimized thickness to form the UPH nanoparticles. With the aid of HA, the UPH nanoparticles were capable of preferentially enriching in tumor sites and specific endocytosis by CD44 receptors as well as responsive degradation by hyaluronidase in cancer cells after intravenous administration. Subsequently, after being activated by strong penetrating 980 nm near-infrared light (NIR), the UPH nanoparticles efficiently converted oxygen into strongly oxidizing reactive oxygen species based on the fluorescence resonance energy transfer (FRET) effect, thereby significantly inhibiting tumor growth. Experimental results in vitro and in vivo indicated that such dual-responsive nanoparticles successfully realize the photodynamic therapy of deep-seated cancer with negligible side effects, which showed great potential for potential clinical translational research.
Collapse
|
11
|
Wu H, Du X, Xu J, Kong X, Li Y, Liu D, Yang X, Ye L, Ji J, Xi Y, Zhai G. Multifunctional biomimetic nanoplatform based on photodynamic therapy and DNA repair intervention for the synergistic treatment of breast cancer. Acta Biomater 2023; 157:551-565. [PMID: 36513248 DOI: 10.1016/j.actbio.2022.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/27/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Photodynamic therapy (PDT) is a minimally invasive and locally effective treatment method, which has been used in the clinical treatment of a variety of superficial tumors. In recent years, PDT has received extensive attention due to its induction of immunogenic cell death (ICD). However, the repair mechanism of tumor cells and low immune response limit the further development of PDT. To this end, a multifunctional biomimetic nanoplatform 4T1Mem@PGA-Ce6/Ola (MPCO) is developed to co-deliver the photosensitizer Chlorin e6 (Ce6) and Olaparib (Ola) with the function of preventing DNA repair. The nanoplatform shows efficient tumor targeting and cellular internalization properties due to cell membrane camouflage, and Ce6 and Ola produce a significant synergistic anti-tumor effect under laser irradiation. Meanwhile, the nanoplatform can also activate the cyclic guanosine monophosphate-adenosine monophosphate synthase-interferon gene stimulator signaling (cGAS-STING) pathway to produce cytokines. The damage-associated molecular patterns induced by ICD can work with these cytokines to recruit and stimulate the maturation of dendritic cells and induce the systemic anti-tumor immune response. Overall, this multifunctional biomimetic nanoplatform integrating PDT, chemotherapy, and immunotherapy is highlighted here to boost anti-tumor therapy. STATEMENT OF SIGNIFICANCE: Self-repair of DNA damage is the most important reason for the failure of primary tumor eradication and the formation of secondary and metastatic tumors. To address this issue, a multifunctional biomimetic nanoplatform 4T1Mem@PGA-Ce6/Ola (MPCO) was developed to integrate a photosensitizer Chlorine a6 and a poly (ADP-ribose) polymerase inhibitor Olaparib. With tumor targeting ability and controlled release of drugs, the MPCO was expected to enhance tumor immunogenicity and facilitate antitumor immunity through the induction of immunogenic cell death as well as the activation of the cGAS-STING pathway. This study develops a promising combination strategy against tumors and has substantial implications for the prognosis of patients with breast cancer.
Collapse
Affiliation(s)
- Hang Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xiyou Du
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Jiangkang Xu
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xinru Kong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Dongzhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yanwei Xi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| |
Collapse
|
12
|
Zhu L, Luo M, Zhang Y, Fang F, Li M, An F, Zhao D, Zhang J. Free radical as a double-edged sword in disease: Deriving strategic opportunities for nanotherapeutics. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
Wang J, Hao L, Li X, Liu S. W18O49@EP nanoparticles improve the anti-tumor effect of radiotherapy and photodynamic therapy by avoiding the limitation of hypoxia. Front Bioeng Biotechnol 2022; 10:1060467. [DOI: 10.3389/fbioe.2022.1060467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
Insufficient oxygen supply at the tumor site and hypoxia caused during tumor treatment lead to a poor therapeutic effect and poor prognosis. Therefore, effectively overcoming the problem of hypoxia in tumors and avoiding hypoxia that compromises the efficacy of the treatment could improve the anti-tumor therapeutic effect. Thus, this study reports the ability of W18O49@EP nanoparticles to release reactive oxygen species (ROS) during the combined tumor radiotherapy (RT) and photodynamic therapy (PDT). The release of ROS by the nanoparticles during near infrared light (NIR) irradiation was demonstrated by in vitro and in vivo experiments, realizing an effective PDT without inducing hypoxia. Indeed, the ROS did not derive from the oxygen in the tumor microenvironment but they were released by the nanoparticles. Thus, ROS could improve the therapeutic effect of RT avoiding the problem of hypoxia after RT. Hence, W18O49@EP nanoparticles greatly improved the anti-tumor effect due to their effectiveness despite the insufficient oxygen supply and hypoxia caused by traditional RT and PDT.
Collapse
|
14
|
Zhou HY, Chen Y, Li P, He X, Zhong J, Hu Z, Liu L, Chen Y, Cui G, Sun D, Zheng T. Sonodynamic therapy for breast cancer: A literature review. OPEN CHEM 2022. [DOI: 10.1515/chem-2022-0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Abstract
Breast cancer (BC) is a malignant tumor with the highest incidence among women. Surgery, radiotherapy, and chemotherapy are currently used as the first-line methods for treating BC. Sonodynamic therapy (SDT) in combination with sonosensitizers exerts a synergistic effect. The therapeutic effects of SDT depend on factors, such as the intensity, frequency, and duration of ultrasound, and the type and the biological model of sonosensitizer. Current reviews have focused on the possibility of using tumor-seeking sonosensitizers, sometimes in combination with different therapies, such as immunotherapy. This study elucidates the therapeutic mechanism of interaction between SDT and tissue as well as the current progress in medical applications of SDT to BC.
Collapse
Affiliation(s)
- Hai-ying Zhou
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Yi Chen
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Ping Li
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Xiaoxin He
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Jieyu Zhong
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Zhengming Hu
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Li Liu
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Yun Chen
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Guanghui Cui
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Desheng Sun
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| | - Tingting Zheng
- Department of Ultrasonography, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Institute of Ultrasound Medicine, Shenzhen-PKU-HKUST Medical Center , Shenzhen , 518036 , China
| |
Collapse
|
15
|
Gao M, Huang X, Wu Z, Wang L, Yuan S, Du Z, Luo S, Li R, Wang W. Synthesis of a versatile mitochondria-targeting small molecule for cancer near-infrared fluorescent imaging and radio/photodynamic/photothermal synergistic therapies. Mater Today Bio 2022; 15:100316. [PMID: 35721281 PMCID: PMC9198388 DOI: 10.1016/j.mtbio.2022.100316] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 12/17/2022]
Abstract
Although as a mainstay modal for cancer treatment, the clinical effect of radiotherapy (RT) does not yet meet the need of cancer patients. Developing tumour-preferential radiosensitizers or combining RT with other treatments has been acknowledged highly necessary to enhance the efficacy of RT. The present study reported a multifunctional bioactive small-molecule (designated as IR-83) simultaneously exhibiting tumour-preferential accumulation, near-infrared imaging and radio/photodynamic/photothermal therapeutic effects. IR-83 was designed and synthesized by introducing 2-nitroimidazole as a radiosensitizer into the framework of heptamethine cyanine dyes inherently with tumour-targeting and photosensitizing effects. As results, IR-83 preferentially accumulated in tumours, suppressed tumour growth and metastasis by integrating radio/photodynamic/photothermal multimodal therapies. Mechanism studies showed that IR-83 accumulated in cancer cell mitochondria, induced excessive reactive oxygen species (ROS), and generated high heat after laser irradiation. On one hand, these phenomena led to mitochondrial dysfunction and a sharp decline in oxidative phosphorylation to lessen tissue oxygen consumption. On the other hand, excessive ROS in mitochondria destroyed the balance of antioxidants and oxidative stress balance by down-regulating the intracellular antioxidant system, and subsequently sensitized ionizing radiation-generated irreversible DNA double-strand breaks. Therefore, this study presented a promising radiosensitizer and a new alternative strategy to enhance RT efficacy via mitochondria-targeting multimodal synergistic treatment. IR-83 is chemically synthesized via introduction of a radiosensitizing moiety into a cancer-targeting heptamethine cyanine framework.. IR-83 exhibits multifunctional bioactivities of cancer-preferential accumulation, near infrared imaging-guided multimodal treatment. IR-83 exerts a synergistic therapeutic effect of RT/PDT/PTT by targeting cancer cell mitochondria. Cancer radiotherapy is significantly sensitized by mitochondria-targeting delivery of a radiosensitizing moiety, PTT-triggered increase of O2 level and PDT-induced irreversible DNA double-strand breaks.
Collapse
Affiliation(s)
- Mingquan Gao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Xie Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zifei Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shaolong Yuan
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zaizhi Du
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shenglin Luo
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Rong Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Corresponding author. No. 30, Gaotanyan Zheng Street, Shapingba District, Chongqing, China.
| | - Weidong Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
- Corresponding author. No. 55, section 4, Renmin South Road, Chengdu, Sichuan Province, China.
| |
Collapse
|
16
|
Ouyang J, Xie A, Zhou J, Liu R, Wang L, Liu H, Kong N, Tao W. Minimally invasive nanomedicine: nanotechnology in photo-/ultrasound-/radiation-/magnetism-mediated therapy and imaging. Chem Soc Rev 2022; 51:4996-5041. [PMID: 35616098 DOI: 10.1039/d1cs01148k] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Traditional treatments such as chemotherapy and surgery usually cause severe side effects and excruciating pain. The emergence of nanomedicines and minimally invasive therapies (MITs) has brought hope to patients with malignant diseases. Especially, minimally invasive nanomedicines (MINs), which combine the advantages of nanomedicines and MITs, can effectively target pathological cells/tissues/organs to improve the bioavailability of drugs, minimize side effects and achieve painless treatment with a small incision or no incision, thereby acquiring good therapeutic effects. In this review, we provide a comprehensive review of the research status and challenges of MINs, which generally refers to the medical applications of nanotechnology in photo-/ultrasound-/radiation-/magnetism-mediated therapy and imaging. Additionally, we also discuss their combined application in various fields including cancers, cardiovascular diseases, tissue engineering, neuro-functional diseases, and infectious diseases. The prospects, and potential bench-to-bedside translation of MINs are also presented in this review. We expect that this review can inspire the broad interest for a wide range of readers working in the fields of interdisciplinary subjects including (but not limited to) chemistry, nanomedicine, bioengineering, nanotechnology, materials science, pharmacology, and biomedicine.
Collapse
Affiliation(s)
- Jiang Ouyang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Angel Xie
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Jun Zhou
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Runcong Liu
- Zhuhai Precision Medical Center, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong 519000, China
| | - Liqiang Wang
- Henan Province Industrial Technology Research Institute of Resources and Materials, School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Haijun Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Yu XT, Sui SY, He YX, Yu CH, Peng Q. Nanomaterials-based photosensitizers and delivery systems for photodynamic cancer therapy. BIOMATERIALS ADVANCES 2022; 135:212725. [PMID: 35929205 DOI: 10.1016/j.bioadv.2022.212725] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
The increasing cancer morbidity and mortality requires the development of high-efficiency and low-toxicity anticancer approaches. In recent years, photodynamic therapy (PDT) has attracted much attention in cancer therapy due to its non-invasive features and low side effects. Photosensitizer (PS) is one of the key factors of PDT, and its successful delivery largely determines the outcome of PDT. Although a few PS molecules have been approved for clinical use, PDT is still limited by the low stability and poor tumor targeting capacity of PSs. Various nanomaterial systems have shown great potentials in improving PDT, such as metal nanoparticles, graphene-based nanomaterials, liposomes, ROS-sensitive nanocarriers and supramolecular nanomaterials. The small molecular PSs can be loaded in functional nanomaterials to enhance the PS stability and tumor targeted delivery, and some functionalized nanomaterials themselves can be directly used as PSs. Herein, we aim to provide a comprehensive understanding of PDT, and summarize the recent progress of nanomaterials-based PSs and delivery systems in anticancer PDT. In addition, the concerns of nanomaterials-based PDT including low tumor targeting capacity, limited light penetration, hypoxia and nonspecific protein corona formation are discussed. The possible solutions to these concerns are also discussed.
Collapse
Affiliation(s)
- Xiao-Tong Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shang-Yan Sui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yu-Xuan He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chen-Hao Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
18
|
Quiñones ED, Lu TY, Liu KT, Fan YJ, Chuang EY, Yu J. Glycol chitosan/iron oxide/polypyrrole nanoclusters for precise chemodynamic/photothermal synergistic therapy. Int J Biol Macromol 2022; 203:268-279. [PMID: 35051505 DOI: 10.1016/j.ijbiomac.2022.01.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 01/14/2023]
Abstract
Noninvasive photothermal therapy (PTT) represents a promising direction for more modern and precise medical applications. However, PTT efficacy is still not satisfactory due to the existence of heat shock proteins (HSPs) and poorly targeted delivery. Herein, the design of a nanosystem with improved delivery efficacy for anticancer treatment employing the synergetic effects of reactive oxygen species (ROS)-driven chemodynamic therapy (CDT) to inactivated HSPs with photothermal-hyperthermia was therefore achieved through the development of pH-targeting glycol chitosan/iron oxide enclosed core polypyrrole nanoclusters (GCPI NCs). The designed NCs effectively accumulated toward cancer cells due to their acidic microenvironment, initiating ROS generation via Fenton reaction at the outset and performing site-specific near infrared (NIR)-photothermal effect. A comprehensive analysis of both surface and bulk material properties of the CDT/PTT NCs as well as biointerface properties were ascertained via numerous surface specific analytical techniques by bringing together heightened accumulation of CDT/PTT NCs, which can significantly eradicate cancer cells thus minimizing the side effects of conventional chemotherapies. All of these attributes act in synergy over the cancer cells succeeding in fashioning NC's able to act as competent agents in the MRI-monitored enhanced CDT/PTT synergistic therapy. Findings in this study evoke attention in future oncological therapeutic strategies.
Collapse
Affiliation(s)
- Edgar Daniel Quiñones
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Ting-Yu Lu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Kuan-Ting Liu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Yu-Jui Fan
- School of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 11031, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jiashing Yu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
19
|
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
|
20
|
Hu H, Zheng S, Hou M, Zhu K, Chen C, Wu Z, Qi L, Ren Y, Wu B, Xu Y, Yan C, Zhao B. Functionalized Au@Cu-Sb-S Nanoparticles for Spectral CT/Photoacoustic Imaging-Guided Synergetic Photo-Radiotherapy in Breast Cancer. Int J Nanomedicine 2022; 17:395-407. [PMID: 35115774 PMCID: PMC8800589 DOI: 10.2147/ijn.s338085] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/06/2022] [Indexed: 11/23/2022] Open
Abstract
Background Radiotherapy (RT) is clinically well-established cancer treatment. However, radioresistance remains a significant issue associated with failure of RT. Phototherapy-induced radiosensitization has recently attracted attention in translational cancer research. Methods Cu-Sb-S nanoparticles (NPs) coated with ultra-small Au nanocrystals (Au@Cu-Sb-S) were synthesized and characterized. The biosafety profiles, absorption of near-infrared (NIR) laser and radiation-enhancing effect of the NPs were evaluated. In vitro and in vivo spectral computed tomography (CT) imaging and photoacoustic (PA) imaging were performed in 4T1 breast cancer-bearing mice. The synergetic radio-phototherapy was assessed by in vivo tumor inhibition studies. Results Au@Cu-Sb-S NPs were prepared by in situ growth of Au NCs on the surface of Cu-Sb-S NPs. The cell viability experiments showed that the combination of Au@Cu-Sb-S+NIR+RT was significantly more cytotoxic to tumor cells than the other treatments at concentrations above 25 ppm Sb. In vitro and in vivo spectral CT imaging demonstrated that the X-ray attenuation ability of Au@Cu-Sb-S NPs was superior to that of the clinically used Iodine, particularly at lower KeV levels. Au@Cu-Sb-S NPs showed a concentration-dependent and remarkable PA signal brightening effect. In vivo tumor inhibition studies showed that the prepared Au@Cu-Sb-S NPs significantly suppressed tumor growth in 4T1 breast cancer-bearing mice treated with NIR laser irradiation and an intermediate X-ray dose (4 Gy). Conclusion These results indicate that Au@Cu-Sb-S integrated with spectral CT, PA imaging, and phototherapy-enhanced radiosensitization is a promising multifunctional theranostic nanoplatform for clinical applications.
Collapse
Affiliation(s)
- Honglei Hu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Shuting Zheng
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Meirong Hou
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Kai Zhu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Chuyao Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Zede Wu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Li Qi
- Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yunyan Ren
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Bin Wu
- Institute of Respiratory Diseases, Respiratory Department, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Chenggong Yan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Bingxia Zhao
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Correspondence: Bingxia Zhao; Yikai Xu, Tel +86 20 61647272; +86 20 62787333, Email ;
| |
Collapse
|
21
|
Jiang S, Liu Z, Tian Y, Zhuang M, Piao S, Gao Y, Tam A, Hu H, Cheng W. A Comprehensive Evaluation of ZrC Nanoparticle in Combined Photothermal and Radiation Therapy for Treatment of Triple-Negative Breast Cancer. Front Oncol 2021; 11:801352. [PMID: 34993150 PMCID: PMC8724783 DOI: 10.3389/fonc.2021.801352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 01/18/2023] Open
Abstract
Because of the difficulty in treating triple-negative breast cancer (TNBC), the search for treatments has never stopped. Treatment opinions remain limited for triple-negative breast cancer (TNBC). The current treatment approach of using photothermal therapy (PTT) is often imprecise and has limited penetration below the surface of the skin. On the other hand, radiation therapy (RT) has its unavoidable disadvantages, such as side effects or ineffectiveness against hypoxic tumor microenvironment (TME). In this study, we proposed the use of ZrC nanoparticles in conjunction with RT/PTT to enhance antitumor and antimetastatic effect. We modified the ZrC nanoparticle with bovine serum albumin (BSA) and folic acid (FA), sizing desirable about 100nm. The photothermal conversion efficiency was calculated to be 40.51% and sensitizer enhancement ration (SER) was 1.8. With addition of ZrC NPs, more DNA were damaged in γ-H2AX and more ROS were detected with immunofluorescence. In vitro and vivo, the combined therapy with ZrC NPS showed the best effect of tumor cell inhibition and safety. Our results provide evidence that the combination of ZrC NPs, PT, and RT is effective in of TNBC, making it a great potential application for cancer therapy in clinic.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ming Zhuang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shiqi Piao
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yan Gao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Andrew Tam
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Hongtao Hu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Wen Cheng, ; Hongtao Hu,
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Wen Cheng, ; Hongtao Hu,
| |
Collapse
|
22
|
Zhu Z, Wu M, Sun J, Huangfu Z, Yin L, Yong W, Sun J, Wang G, Meng F, Zhong Z. Redox-sensitive iodinated polymersomes carrying histone deacetylase inhibitor as a dual-functional nano-radiosensitizer for enhanced radiotherapy of breast cancer. Drug Deliv 2021; 28:2301-2309. [PMID: 34730060 PMCID: PMC8567935 DOI: 10.1080/10717544.2021.1995080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 01/14/2023] Open
Abstract
Radiotherapy (RT) is a frequently used means in clinical tumor treatment. The outcome of RT varies, however, to a great extent, due to RT resistance or intolerable dose, which might be resolved by the development of radio-sensitizing strategies. Here, we report redox-sensitive iodinated polymersomes (RIP) carrying histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA, vorinostat), as a new dual-functional nano-radiosensitizer for breast cancer radiotherapy. SAHA-loaded RIP (RIP-SAHA) with a size of about 101 nm exhibited good colloidal stability while the reduction-activated release of SAHA, giving rise to better antitumor effect to 4T1 breast carcinoma cells than free SAHA. Accordingly, RIP-SAHA combined with a 4 Gy dose of X-ray radiation led to significantly enhanced suppression of 4T1 cells compared with SAHA combined 4 Gy of X-ray radiation, as a result of enhanced DNA damage and impeded DNA damage repair. The pharmacokinetics and biodistribution studies by single-photon emission computed tomography (SPECT) with 125I-labeled SAHA (125I-SAHA) showed a 17.3-fold longer circulation and 237.7-fold better tumor accumulation of RIP-SAHA over SAHA. The systemic administration of RIP-SAHA greatly sensitized radiotherapy of subcutaneous 4T1 breast tumors and brought about significant inhibition of tumor growth, without causing damages to major organs, compared with radiotherapy alone. RIP not only enhanced SAHA delivery but also acted as a radiosensitizer. RIP-SAHA emerges as a smart dual-functional nano-radiosensitizer to effectively enhance tumor radiotherapy.
Collapse
Affiliation(s)
- Zhehong Zhu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Manran Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Juan Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Zhengyuan Huangfu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Lingling Yin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Weipeng Yong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jing Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Liang Y, Li M, Huang Y, Guo B. An Integrated Strategy for Rapid Hemostasis during Tumor Resection and Prevention of Postoperative Tumor Recurrence of Hepatocellular Carcinoma by Antibacterial Shape Memory Cryogel. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101356. [PMID: 34382336 DOI: 10.1002/smll.202101356] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/25/2021] [Indexed: 06/13/2023]
Abstract
The inevitable bleeding during tumor resection greatly increases the risk of tumor recurrence caused by metastasis of cancer cells with blood, and hemostasis and prevention of post-operation tumor recurrence is still a challenge. However, a biomaterials approach for rapid hemostasis during tumor resection and simultaneous prevention of tumor recurrence is rarely reported. Here, zeolitic imidazolate framework (ZIF-8) nanoparticle-enhanced multinetwork cryogels are proposed which provide an integrated treatment regimen for rapid hemostasis through intraoperative blood trigger shape recovery and enhanced coagulation, and prevention of postoperative cancer recurrence via sonodynamic anticancer in a hepatocellular carcinoma model. A series of antibacterial shape memory multifunctional cryogels are synthesized based on glycidyl methacrylate-functionalized quaternized chitosan (QCSG), dopamine-modified hyaluronic acid (HA-DA), and hematoporphyrin monomethyl ether (HMME)-loaded dopamine-modified ZIF-8 (ZDH). Blood loss in different bleeding models confirms good hemostasis of ZIF-8 loading cryogels. Besides, in vitro tests confirm that QCSG/HA-DA/ZDH (QH/ZDH) cryogels significantly killed cancer cells by generating reactive oxygen species under ultrasound. Finally, significantly reduced tumor recurrence after the resection of ectopic hepatocellular carcinoma further confirms the good effect of QH/ZDH cryogels in preventing recurrence by a coordinated strategy of intraoperative hemostasis and postoperative sonodynamic therapy by pH-responsive HMME release, showing great potential in clinical application.
Collapse
Affiliation(s)
- Yongping Liang
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Meng Li
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Huang
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Baolin Guo
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
24
|
Lu TY, Lu WF, Wang YH, Liao MY, Wei Y, Fan YJ, Chuang EY, Yu J. Keratin-Based Nanoparticles with Tumor-Targeting and Cascade Catalytic Capabilities for the Combinational Oxidation Phototherapy of Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:38074-38089. [PMID: 34351754 DOI: 10.1021/acsami.1c10160] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Photodynamic therapy (PDT) holds tantalizing prospects of a prominent cancer treatment strategy. However, its efficacy remains limited by virtue of the hypoxic tumor microenvironment and the inadequate tumor-targeted delivery of photosensitizers, and these can be further exacerbated by the lack of development of a well-controlled nitric oxide (NO) release system at the target site. Inspired by Chinese medicine, we propose a revealing new keratin application. Keratin has garnered attention as an NO generator; however, its oncological use has rarely been investigated. We hypothesized that the incorporation of a phenylboronic acid (PBA) targeting ligand/methylene blue (MB) photosensitizer with a keratin NO donor would facilitate precise tumor delivery, enhancing PDT. Herein, we demonstrated that MB@keratin/PBA/d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) nanoparticles (MB@KPTNPs) specifically targeted breast cancer cells and effectively suppressed their growth. Through MB-mediated biometabolism, the endocytic MB@KPTNPs produced a sufficient amount of intracellular NO that reduced the glutathione level while boosting the efficiency of PDT. A therapeutic combination of NO/PDT was therefore achieved, resulting in significant inhibition of both in vivo tumor growth and lung metastasis. These findings underscore the importance of utilizing keratin-based nanoparticles that simultaneously combine targeting of the tumor and self-generating NO with a cascading catalytic ability as a novel oxidation therapeutic strategy for enhancing PDT.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Wei-Fan Lu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Yin-Hsu Wang
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Mei-Yi Liao
- Department of Applied Chemistry, National Pingtung University, Pingtung 90003, Taiwan
| | - Yang Wei
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106, Taiwan
| | - Yu-Jui Fan
- School of Biomedical Engineering; and International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering; and International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Jiashing Yu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
25
|
Lu H, Wang W, Li X, Zhang M, Cheng X, Sun K, Ding Y, Li X, Hu A. A carrier-free nanoparticle with dual NIR/acid responsiveness by co-assembly of enediyne and IR820 for combined PTT/chemotherapy. J Mater Chem B 2021; 9:4056-4064. [PMID: 33949615 DOI: 10.1039/d1tb00279a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Combined photothermal therapy/chemotherapy by co-delivery of a photosensitizer (PS) and a chemotherapeutic drug has demonstrated great potential for cancer treatment. The intrinsic drawbacks of traditional drug delivery systems (DDSs), such as tedious synthetic procedures, side effects originated from the carrier materials, low loading efficiency, and uncontrolled drug release, however, have impaired their further advancement. On the other hand, enediyne antibiotics are highly cytotoxic toward cancer cells through the generation of lethal carbon radicals via thermal-induced cyclization, endowing them with great potential to achieve enhanced synergistic anticancer performance by incorporation with the photothermal effect of PS. To this end, a carrier-free and NIR/acid dual-responsive DDS was constructed for combined photothermal therapy/chemotherapy. The facile co-assembly of maleimide-based enediyne and PS IR820 was achieved in aqueous solution to give nanoparticles (EICN) with a hydrodynamic diameter of 90 nm and high stability. In vitro study confirmed the acid/NIR dual-responsive degradation and drug release, free radical generation and DNA-cleaving ability of EICN, which was accomplished by the corporation of enediyne and IR820 moieties. Further tests on HeLa cells verified the excellent synergistic anticancer performance of EICN including the improved cellular uptake, NIR-enhanced drug release, DNA damage and histone deacetylase inhibitor capacity. Overall, this carrier-free DDS with dual acid/NIR-responsivity would potentially provide new insights for the development of combined photothermal/chemotherapy.
Collapse
Affiliation(s)
- Haotian Lu
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Wenbo Wang
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Xiaoxuan Li
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Mengsi Zhang
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Xiaoyu Cheng
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Ke Sun
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Yun Ding
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Xinxin Li
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Aiguo Hu
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
26
|
Antitumor Effects in Gas Plasma-Treated Patient-Derived Microtissues—An Adjuvant Therapy for Ulcerating Breast Cancer? APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11104527] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite global research and continuous improvement in therapy, cancer remains a challenging disease globally, substantiating the need for new treatment avenues. Medical gas plasma technology has emerged as a promising approach in oncology in the last years. Several investigations have provided evidence of an antitumor action in vitro and in vivo, including our recent work on plasma-mediated reduction of breast cancer in mice. However, studies of gas plasma exposure on patient-derived tumors with their distinct microenvironment (TME) are scarce. To this end, we here investigated patient-derived breast cancer tissue after gas plasma-treated ex vivo. The tissues were disjoint to pieces smaller than 100 µm, embedded in collagen, and incubated for several days. The viability of the breast cancer tissue clusters and their outgrowth into their gel microenvironment declined with plasma treatment. This was associated with caspase 3-dependent apoptotic cell death, paralleled by an increased expression of the anti-metastatic adhesion molecule epithelial (E)-cadherin. Multiplex chemokine/cytokine analysis revealed a marked decline in the release of the interleukins 6 and 8 (IL-6, IL-8) and monocyte-chemoattractant-protein 1 (MCP) known to promote a cancer-promoting milieu in the TME. In summary, we provide here, for the first time, evidence of a beneficial activity of gas plasma exposure on human patient-derived breast cancer tissue.
Collapse
|
27
|
Zielińska A, Szalata M, Gorczyński A, Karczewski J, Eder P, Severino P, Cabeda JM, Souto EB, Słomski R. Cancer Nanopharmaceuticals: Physicochemical Characterization and In Vitro/In Vivo Applications. Cancers (Basel) 2021; 13:1896. [PMID: 33920840 PMCID: PMC8071188 DOI: 10.3390/cancers13081896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Physicochemical, pharmacokinetic, and biopharmaceutical characterization tools play a key role in the assessment of nanopharmaceuticals' potential imaging analysis and for site-specific delivery of anti-cancers to neoplastic cells/tissues. If diagnostic tools and therapeutic approaches are combined in one single nanoparticle, a new platform called nanotheragnostics is generated. Several analytical technologies allow us to characterize nanopharmaceuticals and nanoparticles and their properties so that they can be properly used in cancer therapy. This paper describes the role of multifunctional nanoparticles in cancer diagnosis and treatment, describing how nanotheragnostics can be useful in modern chemotherapy, and finally, the challenges associated with the commercialization of nanoparticles for cancer therapy.
Collapse
Affiliation(s)
- Aleksandra Zielińska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (M.S.); (R.S.)
- Department of Pharmaceutical Echnology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Marlena Szalata
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (M.S.); (R.S.)
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland
| | - Adam Gorczyński
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
| | - Jacek Karczewski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznań, Poland;
| | - Piotr Eder
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznań, Poland;
| | - Patrícia Severino
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women & Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA;
- Biotechnological Postgraduate Program, Institute of Technology and Research (ITP), Nanomedicine and Nanotechnology Laboratory (LNMed), University of Tiradentes (Unit), Av. Murilo Dantas 300, Aracaju 49010-390, Brazil
- Tiradentes Institute, 150 Mt Vernon St, Dorchester, MA 02125, USA
| | - José M. Cabeda
- ESS-FP, Escola Superior de Saúde Fernando Pessoa, Rua Delfim Maia 334, 4200-253 Porto, Portugal;
- FP-ENAS-Fernando Pessoa Energy, Environment and Health Research Unit, Universidade Fernando Pessoa, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
| | - Eliana B. Souto
- Department of Pharmaceutical Echnology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- CEB–Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Ryszard Słomski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (M.S.); (R.S.)
| |
Collapse
|
28
|
Lu TY, Chiang CY, Fan YJ, Jheng PR, Quiñones ED, Liu KT, Kuo SH, Hsieh HY, Tseng CL, Yu J, Chuang EY. Dual-Targeting Glycol Chitosan/Heparin-Decorated Polypyrrole Nanoparticle for Augmented Photothermal Thrombolytic Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10287-10300. [PMID: 33615773 DOI: 10.1021/acsami.0c20940] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Near-infrared (NIR)-light-modulated photothermal thrombolysis has been investigated to overcome the hemorrhage danger posed by clinical clot-busting substances. A long-standing issue in thrombosis fibrinolytics is the lack of lesion-specific therapy, which should not be ignored. Herein, a novel thrombolysis therapy using photothermal disintegration of a fibrin clot was explored through dual-targeting glycol chitosan/heparin-decorated polypyrrole nanoparticles (GCS-PPY-H NPs) to enhance thrombus delivery and thrombolytic therapeutic efficacy. GCS-PPY-H NPs can target acidic/P-selectin high-expression inflammatory endothelial cells/thrombus sites for initiating lesion-site-specific thrombolysis by hyperthermia using NIR irradiation. A significant fibrin clot-clearance rate was achieved with thrombolysis using dual-targeting/modality photothermal clot disintegration in vivo. The molecular level mechanisms of the developed nanoformulations and interface properties were determined using multiple surface specific analytical techniques, such as particle size distribution, zeta potential, electron microscopy, Fourier-transform infrared spectroscopy (FTIR), wavelength absorbance, photothermal, immunofluorescence, and histology. Owing to the augmented thrombus delivery of GCS-PPY-H NPs and swift treatment time, dual-targeting photothermal clot disintegration as a systematic treatment using GCS-PPY-H NPs can be effectively applied in thrombolysis. This novel approach possesses a promising future for thrombolytic treatment.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chih-Yu Chiang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Yu-Jui Fan
- School of Biomedical Engineering; and International Ph. D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Ru Jheng
- Graduate Institute of Biomedical Materials and Tissue Engineering; and International Ph. D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Edgar Daniel Quiñones
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Kuan-Ting Liu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Shuo-Hsiu Kuo
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Han Yun Hsieh
- School of Biomedical Engineering; and International Ph. D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering; and International Ph. D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Jiashing Yu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering; and International Ph. D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
29
|
|