1
|
Sang R, Nixdorf S, Hung T, Power C, Deng F, Bui TA, Engel A, Goldys EM, Deng W. Unlocking the in vivo therapeutic potential of radiation-activated photodynamic therapy for locally advanced rectal cancer with lymph node involvement. EBioMedicine 2025; 116:105724. [PMID: 40359628 DOI: 10.1016/j.ebiom.2025.105724] [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: 10/31/2024] [Revised: 03/14/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Rectal cancer is a leading cause of cancer-related mortality worldwide. The recurrence of locally advanced rectal cancer (LARC), particularly in cases involving lymph node-positive tumours, remains a critical challenge in rectal cancer management. In this study, a therapeutic strategy, radiation-activated photodynamic therapy (RA-PDT), for the treatment of LARC with lymph node-positive tumours was developed and evaluated. METHODS RA-PDT was achieved by using a lipid-polymer hybrid nanoplatform loaded with verteporfin (VP) and functionalised with folic acid (FA) as a targeting molecule. Upon receiving a single 4 Gy fraction of radiation, VP was effectively activated, generating sufficient reactive oxygen species (ROS) to induce cancer cell death-however surrounding tissue was less affected and was spared. The efficacy of this strategy was assessed through in vitro cytotoxicity studies in HCT116 cells, as well as in orthotopic and subcutaneous mouse models. In vivo lymph node tumour progression was also evaluated. FINDINGS RA-PDT effectively generated ROS following 4 Gy irradiation and exhibited significant cytotoxicity in HCT116 cells. In vivo, this strategy largely inhibited primary tumour growth in both orthotopic and subcutaneous mouse models while also suppressing lymph node tumour progression. Surrounding tissues were minimally affected, highlighting the precision and safety of this approach. INTERPRETATION RA-PDT demonstrates potential as a safe therapeutic strategy for LARC, paving the way for its clinical translation. FUNDING This study was supported by the Australian National Health and Medical Research Council (GNT1181889), fellowship award (2019/CDF1013) from Cancer Institute NSW, Australia, the Australian Research Council Centre of Excellence for Nanoscale Biophotonics (CE140100003), UNSW SHARP funding, project grant from National Foundation for Medical Research and Innovation, Australia, International Research Training Program Scholarship (IRTP) from Australian Government, PhD Research Scholar Award from Sydney Vital Translational Cancer Research, and Translational Cancer Research Network PhD Scholarship Top-up award.
Collapse
Affiliation(s)
- Rui Sang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Sheri Nixdorf
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Tzongtyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Carl Power
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thuy Anh Bui
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Alexander Engel
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia; Department of Colorectal Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| | - Ewa M Goldys
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
2
|
Zhang Z, Li L, Ge Y, Chen A, Diao S, Yang Y, Chen Q, Zhou Y, Shao J, Meng F, Yu L, Tian M, Qian X, Lin Z, Xie C, Liu B, Li R. Verteporfin-Mediated In Situ Nanovaccine Based on Local Conventional-Dose Hypofractionated Radiotherapy Enhances Antitumor and Immunomodulatory Effect. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413387. [PMID: 40231790 PMCID: PMC12120762 DOI: 10.1002/advs.202413387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/06/2025] [Indexed: 04/16/2025]
Abstract
In situ radiotherapy is the most successful cytotoxic therapy available for the treatment of solid tumors, while high-dose radiotherapy per fraction is not yet widely and reliably used. To some extent, the major considerations of the disappointing results are on the risk of high-dose irradiation-induced damage to the surrounding normal tissues and the difficulty in distant metastasis control. To break these restraints, a gelatinase-responsive amphiphilic methoxypolyethyleneglycol-PVGLIG-polycaprolactone (mPEG-PVGLIG-PCL) nanoparticles' loading verteporfin (N@VP), a special photosensitizer that can also be excited by X-rays to produce cytotoxic singlet oxygen and greatly enhance radiotherapy efficacy, is prepared in this study. Herein, it is shown that the formed N@VP combined with conventional-dose radiation therapy (RT, 2 Gy (gray, a radiation dose unit)) can realize an antitumor effect no less than high-dose RT (8 Gy) and minimize radiation dose necessary to achieve local tumor control. Moreover, this radiosensitive nanosystem can exert excellent systemic antitumor immunity and abscopal effect, providing a preferable "in situ vaccine" strategy based on conventional-dose RT to achieve efficient systemic management of distant tumor metastasis. When combined with immunotherapy, this novel strategy for radiosensitization results in better immunotherapy sensitivity by stimulating significant immunogenic tumor cell death and synergistic antitumor immune responses.
Collapse
Affiliation(s)
- Zhifan Zhang
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Lin Li
- Department of PathologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Department of OncologyNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210008China
| | - Yuchen Ge
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Anni Chen
- Nanjing International HospitalMedical School of Nanjing UniversityNanjing210019China
| | - Shanchao Diao
- State Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM)Jiangsu Key Laboratory for BiosensorsNanjing University of Posts & TelecommunicationsNanjing210023China
| | - Yueling Yang
- Department of OncologyNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210008China
| | - Qianyue Chen
- State Key Laboratory of Pharmaceutical BiotechnologyMinistry of Education Key Laboratory of Model Animal for Disease StudyJiangsu Key Laboratory of Molecular MedicineModel Animal Research CenterNational Resource Center for Mutant Mice of ChinaNanjing Drum Tower HospitalSchool of MedicineNanjing UniversityNanjing210061China
| | - Yingling Zhou
- Department of OncologyNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210008China
| | - Jie Shao
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Fanyan Meng
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Lixia Yu
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Manman Tian
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Xiaoping Qian
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Zhaoyu Lin
- State Key Laboratory of Pharmaceutical BiotechnologyMinistry of Education Key Laboratory of Model Animal for Disease StudyJiangsu Key Laboratory of Molecular MedicineModel Animal Research CenterNational Resource Center for Mutant Mice of ChinaNanjing Drum Tower HospitalSchool of MedicineNanjing UniversityNanjing210061China
| | - Chen Xie
- State Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM)Jiangsu Key Laboratory for BiosensorsNanjing University of Posts & TelecommunicationsNanjing210023China
| | - Baorui Liu
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| | - Rutian Li
- The Comprehensive Cancer Center of Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Clinical Cancer InstituteNanjing UniversityNanjing210008China
| |
Collapse
|
3
|
Appidi T, China D, Ștefan GR, Moreau M, Mao S, Velarde E, Toyang N, Lowe H, Rengan AK, Ding K, Ngwa W. Engineered multifunctional nanoparticles for enhanced radiation therapy: three-in-one approach for cancer treatment. Mol Cancer 2025; 24:68. [PMID: 40050802 PMCID: PMC11883980 DOI: 10.1186/s12943-025-02266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/07/2025] [Indexed: 03/10/2025] Open
Abstract
Clinical cancer treatment modalities include radiation as one of the first-line therapies used for treating almost two-thirds of cancer patients. Combinational therapy for cancer is becoming extremely popular, with multiple therapies and their pharmacological effects expected to provide a synergistic outcome. The nanotechnology-based combinational therapeutic approach is emerging as a more effective strategy, for its advantages include simultaneous loading of multiple drugs, on-demand drug delivery controlled by external or internal stimulus, targeting a particular site, and the potential to combine physical treatment modalities (like radiation, thermal therapies, etc.) with chemical interventions (like chemotherapy, immunotherapy, etc.). We report a combination of radiotherapy and chemotherapy mediated by a multifunctional lipo-polymeric hybrid nanosystem coated with gold, demonstrating the three different functionalities using a single nanosystem: a) radio sensitization, b) radiation-triggered delivery of drugs, and c) application as an X-ray/CT contrast agent. The lipo-polymeric hybrid nanoparticles, synthesized using a modified hydrogel isolation method, were loaded with a natural plant-derived anti-cancer agent "Caflanone." These nanoparticles were further subjected to in-situ reduction for a surface coating of gold, which provided enhanced radiosensitivity, radiation triggered drug delivery and X-ray/CT imaging. This approach using a multifunctional nanosystem leverages the biocompatibility of the lipo-polymeric hybrid system for the loading of drugs, precise spatiotemporal controllability of radiation for drug release, and the cytotoxicity of the plant-derived anti-cancer agent "Caflanone." A significant therapeutic efficacy in vitro against breast cancer (p = 0.0002), pancreatic cancer (p < 0.0001), and glioblastoma (p < 0.0001) was demonstrated with the combinational approach. The application of the nanosystem as an X-ray/CT contrast agent has been shown in vivo in tumor-bearing mice and the safety profile and histopathology evaluated in healthy mice showed no adverse effects. A significant increase (p = 0.01) in the survival of breast tumor-bearing mice treated with a combinational approach was also demonstrated. The engineered multifunctional nanoparticles enhanced the radiation therapy and triggered the drug release at the tumor site, triggering the action of encapsulated chemotherapeutic agents while providing image guidance.
Collapse
Affiliation(s)
- Tejaswini Appidi
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Debarghya China
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Michele Moreau
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Serena Mao
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ngeh Toyang
- Flavocure Biotech Inc, Baltimore, Maryland, USA
| | - Henry Lowe
- Flavocure Biotech Inc, Baltimore, Maryland, USA
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, India
| | - Kai Ding
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wilfred Ngwa
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
4
|
Zhou L, Du Y, Shang Y, Xiang D, Xia X. A Novel Triptolide Nano-Liposome with Mitochondrial Targeting for Treatment of Hepatocellular Carcinoma. Int J Nanomedicine 2024; 19:12975-12998. [PMID: 39654802 PMCID: PMC11626209 DOI: 10.2147/ijn.s498099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
Background Modern pharmacological studies have demonstrated that although triptolide (TP) is effective against hepatocellular carcinoma, it has poor water solubility and more toxic side effects. In this study, we used triptolide (TP), a bioactive constituent in Tripterygium wilfordii Hook F, as a model drug to develop a novel nano-liposome drug delivery system for the treatment of liver tumours. Methods We constructed a functionally-modified triptolide liposome (FA+TPP-TP-Lips) using the film-dispersion method and investigated its physicochemical properties, mitochondrial targeting of hepatic tumour cells, in vitro and in vivo anti-hepatic tumour activity and its mechanism. Results The prepared FA+TPP-TP-Lips had a particle size of 99.28 ± 5.7 nm, a PDI of 0.20 ± 0.02, a zeta potential of 1.2 ± 0.08 mV, and an encapsulation rate of 74.37% ± 1.07%.FA+TPP-TP-Lips facilitates the cellular uptake of drug delivery systems and improves their targeted delivery to mitochondria. The results of cell efficacy showed that FA+TPP-TP-Lips significantly inhibited the growth of liver cancer cells, decreased mitochondrial membrane potential, and increased intracellular ROS, thus enhancing the highest apoptosis rate of liver cancer cells. The targeted liposomes (FA-TP-Lips, TPP-TP-Lips, and FA+TPP-TP-Lips) had some degree of inhibitory migration effect on Huh-7 cells relative to the unmodified TP-Lips. Studies on tumor-bearing mice demonstrated that FA+ TPP-TP-Lips effectively accumulated in tumor tissues and significantly inhibited the growth of subcutaneous tumors, achieving a tumor inhibition rate of 79.37%. FA+ TPP-TP-Lips demonstrated an enhanced anti-liver tumor effect and significantly mitigated the hepatotoxicity and systemic toxicity associated with TP. Conclusion In summary, the results of this study can provide a feasible solution for improving the mitochondrial targeting of nano-liposomes, and lay a foundation for further developing a novel nano targeting preparation of triptolide for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Yang Du
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Yating Shang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Debiao Xiang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, 410208, People’s Republic of China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| |
Collapse
|
5
|
Ismail M, Wang Y, Li Y, Liu J, Zheng M, Zou Y. Stimuli-Responsive Polymeric Nanocarriers Accelerate On-Demand Drug Release to Combat Glioblastoma. Biomacromolecules 2024; 25:6250-6282. [PMID: 39259212 DOI: 10.1021/acs.biomac.4c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with a poor prognosis and limited treatment options. Drug delivery by stimuli-responsive nanocarriers holds great promise for improving the treatment modalities of GBM. At the beginning of the review, we highlighted the stimuli-active polymeric nanocarriers carrying therapies that potentially boost anti-GBM responses by employing endogenous (pH, redox, hypoxia, enzyme) or exogenous stimuli (light, ultrasonic, magnetic, temperature, radiation) as triggers for controlled drug release mainly via hydrophobic/hydrophilic transition, degradability, ionizability, etc. Modifying these nanocarriers with target ligands further enhanced their capacity to traverse the blood-brain barrier (BBB) and preferentially accumulate in glioma cells. These unique features potentially lead to more effective brain cancer treatment with minimal adverse reactions and superior therapeutic outcomes. Finally, the review summarizes the existing difficulties and future prospects in stimuli-responsive nanocarriers for treating GBM. Overall, this review offers theoretical guidelines for developing intelligent and versatile stimuli-responsive nanocarriers to facilitate precise drug delivery and treatment of GBM in clinical settings.
Collapse
Affiliation(s)
- Muhammad Ismail
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan Zou
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
6
|
Borisenkova AA, Bolshakova OI, Titova AV, Ryabokon IS, Markova MA, Lyutova ZB, Sedov VP, Varfolomeeva EY, Bakhmetyev VV, Arutyunyan AV, Burdakov VS, Sarantseva SV. Fullerene C 60 Conjugate with Folic Acid and Polyvinylpyrrolidone for Targeted Delivery to Tumor Cells. Int J Mol Sci 2024; 25:5350. [PMID: 38791388 PMCID: PMC11120752 DOI: 10.3390/ijms25105350] [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: 04/18/2024] [Revised: 05/04/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
The use of targeted drug delivery systems, including those based on selective absorption by certain receptors on the surface of the target cell, can lead to a decrease in the minimum effective dose and the accompanying toxicity of the drug, as well as an increase in therapeutic efficacy. A fullerene C60 conjugate (FA-PVP-C60) with polyvinylpyrrolidone (PVP) as a biocompatible spacer and folic acid (FA) as a targeting ligand for tumor cells with increased expression of folate receptors (FR) was obtained. Using 13C NMR spectroscopy, FT-IR, UV-Vis spectrometry, fluorometry and thermal analysis, the formation of the conjugate was confirmed and the nature of the binding of its components was established. The average particle sizes of the conjugate in aqueous solutions and cell culture medium were determined using dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA). The FA-PVP-C60 showed antiradical activity against •DPPH, •OH and O2•-, but at the same time, it was shown to generate 1O2. It was found that the conjugate in the studied concentration range (up to 200 μg/mL) is non-toxic in vitro and does not affect the cell cycle. To confirm the ability of the conjugate to selectively accumulate through folate-mediated endocytosis, its uptake into cells was analyzed by flow cytometry and confocal microscopy. It was shown that the conjugate is less absorbed by A549 cells with low FR expression than by HeLa, which has a high level of expression of this receptor.
Collapse
Affiliation(s)
- Alina A. Borisenkova
- Radiation Technology Department, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Olga I. Bolshakova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Anna V. Titova
- Radiation Technology Department, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Irina S. Ryabokon
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Maria A. Markova
- Radiation Technology Department, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Zhanna B. Lyutova
- Radiation Technology Department, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Victor P. Sedov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Elena Yu. Varfolomeeva
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Vadim V. Bakhmetyev
- Radiation Technology Department, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
| | - Alexandr V. Arutyunyan
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Vladimir S. Burdakov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Svetlana V. Sarantseva
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| |
Collapse
|
7
|
Hübinger L, Wetzig K, Runge R, Hartmann H, Tillner F, Tietze K, Pretze M, Kästner D, Freudenberg R, Brogsitter C, Kotzerke J. Investigation of Photodynamic Therapy Promoted by Cherenkov Light Activated Photosensitizers-New Aspects and Revelations. Pharmaceutics 2024; 16:534. [PMID: 38675195 PMCID: PMC11054706 DOI: 10.3390/pharmaceutics16040534] [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: 03/08/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
This work investigates the proposed enhanced efficacy of photodynamic therapy (PDT) by activating photosensitizers (PSs) with Cherenkov light (CL). The approaches of Yoon et al. to test the effect of CL with external radiation were taken up and refined. The results were used to transfer the applied scheme from external radiation therapy to radionuclide therapy in nuclear medicine. Here, the CL for the activation of the PSs (psoralen and trioxsalen) is generated by the ionizing radiation from rhenium-188 (a high-energy beta-emitter, Re-188). In vitro cell survival studies were performed on FaDu, B16 and 4T1 cells. A characterization of the PSs (absorbance measurement and gel electrophoresis) and the CL produced by Re-188 (luminescence measurement) was performed as well as a comparison of clonogenic assays with and without PSs. The methods of Yoon et al. were reproduced with a beam line at our facility to validate their results. In our studies with different concentrations of PS and considering the negative controls without PS, the statements of Yoon et al. regarding the positive effect of CL could not be confirmed. There are slight differences in survival fractions, but they are not significant when considering the differences in the controls. Gel electrophoresis showed a dominance of trioxsalen over psoralen in conclusion of single and double strand breaks in plasmid DNA, suggesting a superiority of trioxsalen as a PS (when irradiated with UVA). In addition, absorption measurements showed that these PSs do not need to be shielded from ambient light during the experiment. An observational test setup for a PDT nuclear medicine approach was found. The CL spectrum of Re-188 was measured. Fluctuating inconclusive results from clonogenic assays were found.
Collapse
Affiliation(s)
- Lisa Hübinger
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Kerstin Wetzig
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Roswitha Runge
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Holger Hartmann
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Falk Tillner
- Department of Radiation Therapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | - Katja Tietze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Kästner
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Robert Freudenberg
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Claudia Brogsitter
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
8
|
Shin HJ, Kim IS, Choi SG, Lee K, Park H, Shin J, Kim D, Beom J, Yi YY, Gupta DP, Song GJ, Chung WS, Lee CJ, Kim DW. Rejuvenating aged microglia by p16 ink4a-siRNA-loaded nanoparticles increases amyloid-β clearance in animal models of Alzheimer's disease. Mol Neurodegener 2024; 19:25. [PMID: 38493185 PMCID: PMC10943801 DOI: 10.1186/s13024-024-00715-x] [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: 09/25/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
Age-dependent accumulation of amyloid plaques in patients with sporadic Alzheimer's disease (AD) is associated with reduced amyloid clearance. Older microglia have a reduced ability to phagocytose amyloid, so phagocytosis of amyloid plaques by microglia could be regulated to prevent amyloid accumulation. Furthermore, considering the aging-related disruption of cell cycle machinery in old microglia, we hypothesize that regulating their cell cycle could rejuvenate them and enhance their ability to promote more efficient amyloid clearance. First, we used gene ontology analysis of microglia from young and old mice to identify differential expression of cyclin-dependent kinase inhibitor 2A (p16ink4a), a cell cycle factor related to aging. We found that p16ink4a expression was increased in microglia near amyloid plaques in brain tissue from patients with AD and 5XFAD mice, a model of AD. In BV2 microglia, small interfering RNA (siRNA)-mediated p16ink4a downregulation transformed microglia with enhanced amyloid phagocytic capacity through regulated the cell cycle and increased cell proliferation. To regulate microglial phagocytosis by gene transduction, we used poly (D,L-lactic-co-glycolic acid) (PLGA) nanoparticles, which predominantly target microglia, to deliver the siRNA and to control microglial reactivity. Nanoparticle-based delivery of p16ink4a siRNA reduced amyloid plaque formation and the number of aged microglia surrounding the plaque and reversed learning deterioration and spatial memory deficits. We propose that downregulation of p16ink4a in microglia is a promising strategy for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Hyo Jung Shin
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Seung Gyu Choi
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Kayoung Lee
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyewon Park
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Dayoung Kim
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jaewon Beom
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yoon Young Yi
- Department of Pediatrics, College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Deepak Prasad Gupta
- Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-Do, Republic of Korea
| | - Gyun Jee Song
- Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-Do, Republic of Korea
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Oral Anatomy and Developmental Biology, College of Dentistry Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Jiang X, Jiang X, Wu D, Xie W, Liu X, Zheng J. A pH-Sensitive Nanoparticle as Reactive Oxygen Species Amplifier to Regulate Tumor Microenvironment and Potentiate Tumor Radiotherapy. Int J Nanomedicine 2024; 19:709-725. [PMID: 38283195 PMCID: PMC10812755 DOI: 10.2147/ijn.s436160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024] Open
Abstract
Background Radiotherapy is a widely used clinical tool for tumor treatment but can cause systemic toxicity if excessive radiation is administered. Although numerous nanoparticles have been developed as radiosensitizers to reduce the required dose of X-ray irradiation, they often have limitations, such as passive reliance on radiation-induced apoptosis in tumors, and little consider the unique tumor microenvironment that contributes radiotherapy resistance. Methods In this study, we developed and characterized a novel self-assembled nanoparticle containing dysprosium ion and manganese ion (Dy/Mn-P). We systematically investigated the potential of Dy/Mn-P nanoparticles (NPs) as a reactive oxygen species (ROS) amplifier and radiosensitizer to enhance radiation therapy and modulate the tumor microenvironment at the cellular level. Additionally, we evaluated the effect of Dy/Mn-P on the stimulator of interferon genes (STING), an innate immune signaling pathway. Results Physicochemical analysis demonstrated the prepared Dy/Mn-P NPs exhibited excellent dispersibility and stability, and degraded rapidly at lower pH values. Furthermore, Dy/Mn-P was internalized by cells and exhibited selective toxicity towards tumor cells compared to normal cells. Our findings also revealed that Dy/Mn-P NPs improved the tumor microenvironment and significantly increased ROS generation under ionizing radiation, resulting in a ~70% increase in ROS levels compared to radiation therapy alone. This enhanced ROS generation inhibited ~92% of cell clone formation and greatly contributed to cytoplasmic DNA exposure. Subsequently, the activation of the STING pathway was observed, leading to the secretion of pro-inflammatory immune factors and maturation of dendritic cells (DCs). Conclusion Our study demonstrates that Dy/Mn-P NPs can potentiate tumor radiotherapy by improving the tumor microenvironment and increasing endogenous ROS levels within the tumor. Furthermore, Dy/Mn-P can amplify the activation of the STING pathway during radiotherapy, thereby triggering an anti-tumor immune response. This novel approach has the potential to expand the application of radiotherapy in tumor treatment.
Collapse
Affiliation(s)
- Xiaomei Jiang
- Department of Dermatology, Liuzhou Traditional Chinese Medicine Hospital, Liuzhou, 545001, People’s Republic of China
| | - Xiaohong Jiang
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, People’s Republic of China
| | - Dongjie Wu
- Department of Dermatology, Liuzhou Traditional Chinese Medicine Hospital, Liuzhou, 545001, People’s Republic of China
| | - Wanzhu Xie
- Department of Rehabilitation, Liuzhou Worker’s Hospital, Liuzhou, 545001, People’s Republic of China
| | - Xiong Liu
- Department of Dermatology, Liuzhou Traditional Chinese Medicine Hospital, Liuzhou, 545001, People’s Republic of China
| | - Jintao Zheng
- Department of Biotechnology and Food Engineering, Guangdong Technology Israel Institute of Technology, Shantou, 515063, People’s Republic of China
| |
Collapse
|
10
|
Dinakaran D, Wilson BC. The use of nanomaterials in advancing photodynamic therapy (PDT) for deep-seated tumors and synergy with radiotherapy. Front Bioeng Biotechnol 2023; 11:1250804. [PMID: 37849983 PMCID: PMC10577272 DOI: 10.3389/fbioe.2023.1250804] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023] Open
Abstract
Photodynamic therapy (PDT) has been under development for at least 40 years. Multiple studies have demonstrated significant anti-tumor efficacy with limited toxicity concerns. PDT was expected to become a major new therapeutic option in treating localized cancer. However, despite a shifting focus in oncology to aggressive local therapies, PDT has not to date gained widespread acceptance as a standard-of-care option. A major factor is the technical challenge of treating deep-seated and large tumors, due to the limited penetration and variability of the activating light in tissue. Poor tumor selectivity of PDT sensitizers has been problematic for many applications. Attempts to mitigate these limitations with the use of multiple interstitial fiberoptic catheters to deliver the light, new generations of photosensitizer with longer-wavelength activation, oxygen independence and better tumor specificity, as well as improved dosimetry and treatment planning are starting to show encouraging results. Nanomaterials used either as photosensitizers per se or to improve delivery of molecular photosensitizers is an emerging area of research. PDT can also benefit radiotherapy patients due to its complementary and potentially synergistic mechanisms-of-action, ability to treat radioresistant tumors and upregulation of anti-tumoral immune effects. Furthermore, recent advances may allow ionizing radiation energy, including high-energy X-rays, to replace external light sources, opening a novel therapeutic strategy (radioPDT), which is facilitated by novel nanomaterials. This may provide the best of both worlds by combining the precise targeting and treatment depth/volume capabilities of radiation therapy with the high therapeutic index and biological advantages of PDT, without increasing toxicities. Achieving this, however, will require novel agents, primarily developed with nanomaterials. This is under active investigation by many research groups using different approaches.
Collapse
Affiliation(s)
- Deepak Dinakaran
- National Cancer Institute, National Institute of Health, Bethesda, MD, United States
- Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Brian C. Wilson
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Zhang G, Guo M, Ma H, Wang J, Zhang XD. Catalytic nanotechnology of X-ray photodynamics for cancer treatments. Biomater Sci 2023; 11:1153-1181. [PMID: 36602259 DOI: 10.1039/d2bm01698b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Photodynamic therapy (PDT) has been applied in cancer treatment because of its high selectivity, low toxicity, and non-invasiveness. However, the limited penetration depth of the light still hampers from reaching deep-seated tumors. Considering the penetrating ability of high-energy radiotherapy, X-ray-induced photodynamic therapy (X-PDT) has evolved as an alternative to overcome tissue blocks. As the basic principle of X-PDT, X-rays stimulate the nanoparticles to emit scintillating or persistent luminescence and further activate the photosensitizers to generate reactive oxygen species (ROS), which would cause a series of molecular and cellular damages, immune response, and eventually break down the tumor tissue. In recent years, catalytic nanosystems with unique structures and functions have emerged that can enhance X-PDT therapeutic effects via an immune response. The anti-cancer effect of X-PDT is closely related to the following factors: energy conversion efficiency of the material, the radiation dose of X-rays, quantum yield of the material, tumor resistance, and biocompatibility. Based on the latest research in this field and the classical theories of nanoscience, this paper systematically elucidates the current development of the X-PDT and related immunotherapy, and highlights its broad prospects in medical applications, discussing the connection between fundamental science and clinical translation.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China.
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China.
| | - Huizhen Ma
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China.
| | - Junying Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiao-Dong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China. .,Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
12
|
Psoralen as a Photosensitizers for Photodynamic Therapy by Means of In Vitro Cherenkov Light. Int J Mol Sci 2022; 23:ijms232315233. [PMID: 36499568 PMCID: PMC9735954 DOI: 10.3390/ijms232315233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
Possible enhancements of DNA damage with light of different wavelengths and ionizing radiation (Rhenium-188-a high energy beta emitter (Re-188)) on plasmid DNA and FaDu cells via psoralen were investigated. The biophysical experimental setup could also be used to investigate additional DNA damage due to photodynamic effects, resulting from Cherenkov light. Conformational changes of plasmid DNA due to DNA damage were detected and quantified by gel electrophoresis and fluorescent staining. The clonogene survival of the FaDu cells was analyzed with colony formation assays. Dimethyl sulfoxide was chosen as a chemical modulator, and Re-188 was used to evaluate the radiotoxicity and light (UVC: λ = 254 nm and UVA: λ = 366 nm) to determine the phototoxicity. Psoralen did not show chemotoxic effects on the plasmid DNA or FaDu cells. After additional treatment with light (only 366 nm-not seen with 254 nm), a concentration-dependent increase in single strand breaks (SSBs) was visible, resulting in a decrease in the survival fraction due to the photochemical activation of psoralen. Whilst UVC light was phototoxic, UVA light did not conclude in DNA strand breaks. Re-188 showed typical radiotoxic effects with SSBs, double strand breaks, and an overall reduced cell survival for both the plasmid DNA and FaDu cells. While psoralen and UVA light showed an increased toxicity on plasmid DNA and human cancer cells, Re-188, in combination with psoralen, did not provoke additional DNA damage via Cherenkov light.
Collapse
|
13
|
Sang R, Deng F, Engel A, Goldys E, Deng W. Lipid-polymer nanocarrier platform enables X-ray induced photodynamic therapy against human colorectal cancer cells. Biomed Pharmacother 2022; 155:113837. [PMID: 36271586 DOI: 10.1016/j.biopha.2022.113837] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we brought together X-ray induced photodynamic therapy (X-PDT) and chemo-drug (5-FU) for the treatment on colorectal cancer cells. This was achieved by developing a lipid-polymer hybrid nanoparticle delivery system (FA-LPNPs-VP-5-FU). It was prepared by incorporating a photosensitizer (verteporfin), chemotherapy drug (5-FU) and a targeting moiety (folic acid) into one platform. The average size of these nanoparticles was around 100 nm with low polydispersity. When exposed to clinical doses of 4 Gy X-ray radiation, FA-LPNPs-VP-5-FU generated sufficient amounts of reactive oxygen species, triggering the apoptosis and necrosis pathway of cancer cells. Our combined X-PDT and chemo-drug strategy was effective in inhibiting cancer cells' growth and proliferation. Cell cycle analyses revealed that our treatment induced G2/M and S phase arrest in HCT116 cells. Our results indicate that this combined treatment provides better antitumour effect in colorectal cancer cells than each of these modalities alone. This may offer a novel approach for effective colorectal cancer treatment with reduced off-target effect and drug toxicity.
Collapse
Affiliation(s)
- Rui Sang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW 2052, Australia
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW 2052, Australia
| | - Alexander Engel
- Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia; Department of Colorectal Surgery, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Ewa Goldys
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW 2052, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
| |
Collapse
|
14
|
Bianfei S, Fang L, Zhongzheng X, Yuanyuan Z, Tian Y, Tao H, Jiachun M, Xiran W, Siting Y, Lei L. Application of Cherenkov radiation in tumor imaging and treatment. Future Oncol 2022; 18:3101-3118. [PMID: 36065976 DOI: 10.2217/fon-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cherenkov radiation (CR) is the characteristic blue glow that is generated during radiotherapy or radioisotope decay. Its distribution and intensity naturally reflect the actual dose and field of radiotherapy and the location of radioisotope imaging agents in vivo. Therefore, CR can represent a potential in situ light source for radiotherapy monitoring and radioisotope-based tumor imaging. When used in combination with new imaging techniques, molecular probes or nanomedicine, CR imaging exhibits unique advantages (accuracy, low cost, convenience and fast) in tumor radiotherapy monitoring and imaging. Furthermore, photosensitive nanomaterials can be used for CR photodynamic therapy, providing new approaches for integrating tumor imaging and treatment. Here the authors review the latest developments in the use of CR in tumor research and discuss current challenges and new directions for future studies.
Collapse
Affiliation(s)
- Shao Bianfei
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Liu Fang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang Zhongzheng
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Zeng Yuanyuan
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Tian
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - He Tao
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Ma Jiachun
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Wang Xiran
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Siting
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Liu Lei
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Secchi V, Monguzzi A, Villa I. Design Principles of Hybrid Nanomaterials for Radiotherapy Enhanced by Photodynamic Therapy. Int J Mol Sci 2022; 23:8736. [PMID: 35955867 PMCID: PMC9369190 DOI: 10.3390/ijms23158736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/24/2022] Open
Abstract
Radiation (RT) remains the most frequently used treatment against cancer. The main limitation of RT is its lack of specificity for cancer tissues and the limited maximum radiation dose that can be safely delivered without damaging the surrounding healthy tissues. A step forward in the development of better RT is achieved by coupling it with other treatments, such as photodynamic therapy (PDT). PDT is an anti-cancer therapy that relies on the light activation of non-toxic molecules-called photosensitizers-to generate ROS such as singlet oxygen. By conjugating photosensitizers to dense nanoscintillators in hybrid architectures, the PDT could be activated during RT, leading to cell death through an additional pathway with respect to the one activated by RT alone. Therefore, combining RT and PDT can lead to a synergistic enhancement of the overall efficacy of RT. However, the involvement of hybrids in combination with ionizing radiation is not trivial: the comprehension of the relationship among RT, scintillation emission of the nanoscintillator, and therapeutic effects of the locally excited photosensitizers is desirable to optimize the design of the hybrid nanoparticles for improved effects in radio-oncology. Here, we discuss the working principles of the PDT-activated RT methods, pointing out the guidelines for the development of effective coadjutants to be tested in clinics.
Collapse
Affiliation(s)
- Valeria Secchi
- Department of Materials Science, University of Milano-Bicocca, Via R. Cozzi 55, 20125 Milan, Italy
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
| | - Angelo Monguzzi
- Department of Materials Science, University of Milano-Bicocca, Via R. Cozzi 55, 20125 Milan, Italy
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
| | - Irene Villa
- Institute of Physics of the Czech Academy of Sciences, FZU, Cukrovarnická 10/112, 16200 Prague, Czech Republic
| |
Collapse
|
16
|
Chen C, Wu C, Yu J, Zhu X, Wu Y, Liu J, Zhang Y. Photodynamic-based combinatorial cancer therapy strategies: Tuning the properties of nanoplatform according to oncotherapy needs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214495] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Farrer NJ, Higgins GS, Kunkler IH. Radiation-induced prodrug activation: extending combined modality therapy for some solid tumours. Br J Cancer 2022; 126:1241-1243. [PMID: 35217798 PMCID: PMC8873346 DOI: 10.1038/s41416-022-01746-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/26/2022] [Accepted: 02/04/2022] [Indexed: 11/30/2022] Open
Abstract
Combined chemoradiotherapy is the standard of care for locally advanced solid tumours. However, systemic toxicity may limit the delivery of planned chemotherapy. New approaches such as radiation-induced prodrug activation might diminish systemic toxicity, while retaining anticancer benefit. Organic azides have recently been shown to be reduced and activated under hypoxic conditions with clinically relevant doses of radiotherapy, uncaging pazopanib and doxorubicin in preclinical models with similar efficacy as the drug, but lower systemic toxicity. This approach may be relevant to the chemoradiation of glioblastoma and other solid tumours and offers potential for switching on drug delivery from implanted devices. The inclusion of reporters to confirm drug activation, avoidance of off-target effects and synchronisation of irradiation with optimal intratumoral drug concentration will be critical. Further preclinical validation studies of this approach should be encouraged.
Collapse
Affiliation(s)
- Nicola J Farrer
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK.
| | - Geoff S Higgins
- Department of Oncology, MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| |
Collapse
|
18
|
Gong L, Zhang Y, Zhao J, Zhang Y, Tu K, Jiao L, Xu Q, Zhang M, Han S. All-In-One Biomimetic Nanoplatform Based on Hollow Polydopamine Nanoparticles for Synergistically Enhanced Radiotherapy of Colon Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107656. [PMID: 35150039 DOI: 10.1002/smll.202107656] [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] [Received: 12/09/2021] [Revised: 01/04/2022] [Indexed: 06/14/2023]
Abstract
Even though radiotherapy is the most important therapeutic strategy for colon cancer treatment, there is an enormous demand to improve radiosensitivity in solid tumor destruction. For this purpose, a biomimetic nanoplatform based on hollow polydopamine nanoparticles (HP) with homologous targeting and pH-responsive drug release properties is designed. In this work, HP is constructed by using a chelation competition-induced polymerization strategy and then modified with the cancer cell membrane. Hollow polydopamine integrated with Pt nanoparticles (Pt@HP) has a catalase-like activity, which can be used to trigger endogenous H2 O2 into O2 , relieving hypoxia of the tumor microenvironment (TME). With mesoporous shells and large cavities, Pt@HP shows efficient apoptin100-109 (AP) and verteporfin (VP) loading to form AVPt@HP@M. Under X-ray irradiation, AVPt@HP@M exerts a radiosensitization effect via multiple strategies, including relieving hypoxia (Pt NPs), enhancing tumor apoptosis (AP), and X-ray-induced photodynamic therapy (X-PDT) (VP). Further metabonomics analysis shows that the specific mechanism of the AVPt@HP@M is through influencing purine metabolism. Without appreciable systemic toxicity, this nanoplatform highlights a new strategy for effective radiosensitization and provides a reference for treating malignant tumors.
Collapse
Affiliation(s)
- Liuyun Gong
- Department of Radiotherapy, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Jing Zhao
- Department of Radiotherapy, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215000, China
| | - Yilei Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lianying Jiao
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Suxia Han
- Department of Radiotherapy, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
19
|
Zhang B, Xue R, Sun C. Rational design of ROS-responsive nanocarriers for targeted X-ray-induced photodynamic therapy and cascaded chemotherapy of intracranial glioblastoma. NANOSCALE 2022; 14:5054-5067. [PMID: 35293920 DOI: 10.1039/d2nr00436d] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Glioblastoma (GBM) is the most lethal primary intracranial tumor because of its high invasiveness and recurrence. Therefore, nanocarriers with blood-brain barrier (BBB) penetration and transcranial-controlled drug release and activation are rather attractive options for glioblastoma treatment. Herein, we designed a multifunctional nanocarrier (T-TKNPVP) that combined targeted X-ray-induced photodynamic therapy (X-PDT) and cascaded reactive oxygen species (ROS)-boosted chemotherapy. The T-TKNPVP loaded with verteporfin (VP) and paclitaxel (PTX) was self-assembled from an angiopep-2 (Ang) peptide, functionalized Ang-PEG-DSPE and ROS-sensitive PEG-TK-PTX conjugate. After systemic injection, the T-TKNPVP efficiently crossed the BBB and targeted the GBM cells via receptor-mediated transcytosis. Upon X-ray irradiation, they can generate a certain amount of ROS, which not only induces X-PDT but also locoregionally activates PTX release and action by cleaving the TK bridged bonds. As evidenced by 9.4 T MRI and other experiments, such nanocarriers offer significant growth inhibition of GBM in situ and prolong the survival times of U87-MG tumor-bearing mice. Taken together, the designed T-TKNPVP provided an alternative avenue for realizing transcranial X-PDT and X-ray-activated chemotherapy for targeted and locoregional GBM treatment in vivo.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Rui Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Chunyang Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Multimodality Preclinical Molecular Imaging Center, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.
| |
Collapse
|
20
|
Patel M, Prabhu A. Smart nanocomposite assemblies for multimodal cancer theranostics. Int J Pharm 2022; 618:121697. [PMID: 35337903 DOI: 10.1016/j.ijpharm.2022.121697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/28/2022]
Abstract
Despite great strides in anticancer research, performance statistics of current treatment modalities remain dismal, highlighting the need for safe, efficacious strategies for tumour mitigation. Non-invasive fusion technology platforms combining photodynamic, photothermal and hyperthermia therapies have emerged as alternate strategies with potential to meet many of the unmet clinical demands in the domain of cancer. These therapies make use of metallic and magnetic nanoparticles with light absorbing properties, which are manipulated to generate either reactive cytotoxic oxygen species or heat for tumour ablation. Combination therapies integrating light, heat and magnetism-mediated nanoplatforms with the conventional approaches of chemotherapy, radiotherapy and surgery are emerging as precision medicine for targeted interventions against cancer. This article aims to compile recent developments of advanced nanocomposite assemblies that integrate multimodal therapeutics for cancer treatment. Amalgamation of various effective, non-invasive technological platforms such as photodynamic therapy (PDT), photothermal therapy (PTT), magnetic hyperthermia (MHT), and chemodynamic therapy (CDT) have tremendous potential in presenting safe and efficacious solutions to the formidable challenges in cancer therapeutics.
Collapse
Affiliation(s)
- Manshi Patel
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
21
|
Drzewiecka-Matuszek A, Rutkowska-Zbik D. Application of TD-DFT Theory to Studying Porphyrinoid-Based Photosensitizers for Photodynamic Therapy: A Review. Molecules 2021; 26:7176. [PMID: 34885763 PMCID: PMC8658767 DOI: 10.3390/molecules26237176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022] Open
Abstract
An important focus for innovation in photodynamic therapy (PDT) is theoretical investigations. They employ mostly methods based on Time-Dependent Density Functional Theory (TD-DFT) to study the photochemical properties of photosensitizers. In the current article we review the existing state-of-the-art TD-DFT methods (and beyond) which are employed to study the properties of porphyrinoid-based systems. The review is organized in such a way that each paragraph is devoted to a separate aspect of the PDT mechanism, e.g., correct prediction of the absorption spectra, determination of the singlet-triplet intersystem crossing, and interaction with molecular oxygen. Aspects of the calculation schemes are discussed, such as the choice of the most suitable functional and inclusion of a solvent. Finally, quantitative structure-activity relationship (QSAR) methods used to explore the photochemistry of porphyrinoid-based systems are discussed.
Collapse
Affiliation(s)
| | - Dorota Rutkowska-Zbik
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland;
| |
Collapse
|
22
|
Yang YL, Lin K, Yang L. Progress in Nanocarriers Codelivery System to Enhance the Anticancer Effect of Photodynamic Therapy. Pharmaceutics 2021; 13:1951. [PMID: 34834367 PMCID: PMC8617654 DOI: 10.3390/pharmaceutics13111951] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising anticancer noninvasive method and has great potential for clinical applications. Unfortunately, PDT still has many limitations, such as metastatic tumor at unknown sites, inadequate light delivery and a lack of sufficient oxygen. Recent studies have demonstrated that photodynamic therapy in combination with other therapies can enhance anticancer effects. The development of new nanomaterials provides a platform for the codelivery of two or more therapeutic drugs, which is a promising cancer treatment method. The use of multifunctional nanocarriers for the codelivery of two or more drugs can improve physical and chemical properties, increase tumor site aggregation, and enhance the antitumor effect through synergistic actions, which is worthy of further study. This review focuses on the latest research progress on the synergistic enhancement of PDT by simultaneous multidrug administration using codelivery nanocarriers. We introduce the design of codelivery nanocarriers and discuss the mechanism of PDT combined with other antitumor methods. The combination of PDT and chemotherapy, gene therapy, immunotherapy, photothermal therapy, hyperthermia, radiotherapy, sonodynamic therapy and even multidrug therapy are discussed to provide a comprehensive understanding.
Collapse
Affiliation(s)
| | | | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.-L.Y.); (K.L.)
| |
Collapse
|
23
|
Greco A, Garoffolo G, Chiesa E, Riva F, Dorati R, Modena T, Conti B, Pesce M, Genta I. Nanotechnology, a booster for the multitarget drug verteporfin. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Kirsanova DY, Gadzhimagomedova ZM, Maksimov AY, Soldatov AV. Nanomaterials for Deep Tumor Treatment. Mini Rev Med Chem 2021; 21:677-688. [PMID: 33176645 DOI: 10.2174/1389557520666201111161705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/25/2020] [Accepted: 08/20/2020] [Indexed: 11/22/2022]
Abstract
According to statistics, cancer is the second leading cause of death in the world. Thus, it is important to solve this medical and social problem by developing new effective methods for cancer treatment. An alternative to more well-known approaches, such as radiotherapy and chemotherapy, is photodynamic therapy (PDT), which is limited to the shallow tissue penetration (< 1 cm) of visible light. Since the PDT process can be initiated in deep tissues by X-ray irradiation (X-ray induced PDT, or XPDT), it has a great potential to treat tumors in internal organs. The article discusses the principles of therapies. The main focus is on various nanoparticles used with or without photosensitizers, which allow the conversion of X-ray irradiation into UV-visible light. Much attention is given to the synthesis of nanoparticles and analysis of their characteristics, such as size and spectral features. The results of in vitro and in vivo experiments are also discussed.
Collapse
Affiliation(s)
- Daria Yu Kirsanova
- The Smart Materials Research Institute, Southern Federal University, Sladkova 178/24, 344090, Rostov-on-Don, Russian Federation
| | - Zaira M Gadzhimagomedova
- The Smart Materials Research Institute, Southern Federal University, Sladkova 178/24, 344090, Rostov-on-Don, Russian Federation
| | - Aleksey Yu Maksimov
- National Medical Research Centre for Oncology, 14 liniya str. 63, 344037, Rostov-on-Don, Russian Federation
| | - Alexander V Soldatov
- The Smart Materials Research Institute, Southern Federal University, Sladkova 178/24, 344090, Rostov-on-Don, Russian Federation
| |
Collapse
|
25
|
Clement S, Anwer AG, Pires L, Campbell J, Wilson BC, Goldys EM. Radiodynamic Therapy Using TAT Peptide-Targeted Verteporfin-Encapsulated PLGA Nanoparticles. Int J Mol Sci 2021; 22:ijms22126425. [PMID: 34204001 PMCID: PMC8232618 DOI: 10.3390/ijms22126425] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022] Open
Abstract
Radiodynamic therapy (RDT) is a recent extension of conventional photodynamic therapy, in which visible/near infrared light irradiation is replaced by a well-tolerated dose of high-energy X-rays. This enables greater tissue penetration to allow non-invasive treatment of large, deep-seated tumors. We report here the design and testing of a drug delivery system for RDT that is intended to enhance intra- or peri-nuclear localization of the photosensitizer, leading to DNA damage and resulting clonogenic cell kill. This comprises a photosensitizer (Verteporfin, VP) incorporated into poly (lactic-co-glycolic acid) nanoparticles (PLGA NPs) that are surface-functionalized with a cell-penetrating HIV trans-activator of transcription (TAT) peptide. In addition to a series of physical and photophysical characterization studies, cytotoxicity tests in pancreatic (PANC-1) cancer cells in vitro under 4 Gy X-ray exposure from a clinical 6 MV linear accelerator (LINAC) showed that TAT targeting of the nanoparticles markedly enhances the effectiveness of RDT treatment, particularly when assessed by a clonogenic, i.e., DNA damage-mediated, cell kill.
Collapse
Affiliation(s)
- Sandhya Clement
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
- Correspondence:
| | - Ayad G. Anwer
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| | - Layla Pires
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada; (L.P.); (B.C.W.)
| | - Jared Campbell
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| | - Brian C. Wilson
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada; (L.P.); (B.C.W.)
| | - Ewa M. Goldys
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
26
|
Clement S, Guller A, Mahbub SB, Goldys EM. Oxygen-Carrying Polymer Nanoconstructs for Radiodynamic Therapy of Deep Hypoxic Malignant Tumors. Biomedicines 2021; 9:322. [PMID: 33810115 PMCID: PMC8005177 DOI: 10.3390/biomedicines9030322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 02/07/2023] Open
Abstract
Radiodynamic therapy (RDT) is an emerging non-invasive anti-cancer treatment based on the generation of the reactive oxygen species (ROS) at the lesion site following the interaction between X-rays and a photosensitizer drug (PS). The broader application of RDT is impeded by the tumor-associated hypoxia that results in low availability of oxygen for the generation of sufficient amounts of ROS. Herein, a novel nanoparticle drug formulation for RDT, which addresses the problem of low oxygen availability, is reported. It consists of poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) co-loaded with a PS drug verteporfin (VP), and the clinically approved oxygen-carrying molecule, perfluorooctylbromide (PFOB). When triggered by X-rays (4 Gy), under both normoxic and hypoxic conditions, PLGA-VP-PFOB nanoconstructs (NCs) induced a significant increase of the ROS production compared with matching PLGA-VP nanoparticles. The RDT with NCs effectively killed ~60% of human pancreatic cancer cells in monolayer cultures, and almost completely suppressed the outgrowth of tumor cells in 2-weeks clonogenic assay. In a 3D engineered model of pancreatic cancer metastasis to the liver, RDT with NCs destroyed ~35% of tumor cells, demonstrating an exceptional efficiency at a tissue level. These results show that PLGA-VP-PFOB is a promising agent for RDT of deep-seated hypoxic tumors.
Collapse
Affiliation(s)
- Sandhya Clement
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney 2052, Australia; (S.B.M.); (E.M.G.)
| | - Anna Guller
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney 2052, Australia; (S.B.M.); (E.M.G.)
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Saabah B. Mahbub
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney 2052, Australia; (S.B.M.); (E.M.G.)
| | - Ewa M. Goldys
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney 2052, Australia; (S.B.M.); (E.M.G.)
| |
Collapse
|
27
|
Xiao J, Cong H, Wang S, Yu B, Shen Y. Recent research progress in the construction of active free radical nanoreactors and their applications in photodynamic therapy. Biomater Sci 2021; 9:2384-2412. [PMID: 33576752 DOI: 10.1039/d0bm02013c] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy is the most important treatment strategy in free radical therapy. However, tumor microenvironment hypoxia is a key obstacle in PDT. In order to overcome this obstacle, the strategy of in situ production of O2/radicals by catalytic reaction in solid tumors was proposed. In recent years, it has been found that there are many oxygen-independent carbon-based free radicals that can generate toxic active free radicals under laser irradiation and lead to tumor cell death. Based on the rational design of multifunctional nano-medicine, the active free radical nano-generator has opened up a new way for the highly developed nanotechnology and tumor cooperative therapy to improve the therapeutic effect. In this paper, the research status of active free radical nano-generators, especially reactive oxygen species, including the construction mechanism of active free radical nanomaterials, is reviewed and the application of free radical nano-generators in tumor therapy is emphasized.
Collapse
Affiliation(s)
- Jingyuan Xiao
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| | | | | | | | | |
Collapse
|
28
|
Clement S, Campbell JM, Deng W, Guller A, Nisar S, Liu G, Wilson BC, Goldys EM. Mechanisms for Tuning Engineered Nanomaterials to Enhance Radiation Therapy of Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2003584. [PMID: 33344143 PMCID: PMC7740107 DOI: 10.1002/advs.202003584] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Indexed: 05/12/2023]
Abstract
Engineered nanomaterials that produce reactive oxygen species on exposure to X- and gamma-rays used in radiation therapy offer promise of novel cancer treatment strategies. Similar to photodynamic therapy but suitable for large and deep tumors, this new approach where nanomaterials acting as sensitizing agents are combined with clinical radiation can be effective at well-tolerated low radiation doses. Suitably engineered nanomaterials can enhance cancer radiotherapy by increasing the tumor selectivity and decreasing side effects. Additionally, the nanomaterial platform offers therapeutically valuable functionalities, including molecular targeting, drug/gene delivery, and adaptive responses to trigger drug release. The potential of such nanomaterials to be combined with radiotherapy is widely recognized. In order for further breakthroughs to be made, and to facilitate clinical translation, the applicable principles and fundamentals should be articulated. This review focuses on mechanisms underpinning rational nanomaterial design to enhance radiation therapy, the understanding of which will enable novel ways to optimize its therapeutic efficacy. A roadmap for designing nanomaterials with optimized anticancer performance is also shown and the potential clinical significance and future translation are discussed.
Collapse
Affiliation(s)
- Sandhya Clement
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Jared M. Campbell
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Wei Deng
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Anna Guller
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
- Institute for Regenerative MedicineSechenov First Moscow State Medical University (Sechenov University)Trubetskaya StreetMoscow119991Russia
| | - Saadia Nisar
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Guozhen Liu
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Brian C. Wilson
- Department of Medical BiophysicsUniversity of Toronto/Princess Margaret Cancer CentreUniversity Health NetworkColledge StreetTorontoOntarioON M5G 2C1Canada
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| |
Collapse
|
29
|
Gierlich P, Mata AI, Donohoe C, Brito RMM, Senge MO, Gomes-da-Silva LC. Ligand-Targeted Delivery of Photosensitizers for Cancer Treatment. Molecules 2020; 25:E5317. [PMID: 33202648 PMCID: PMC7698280 DOI: 10.3390/molecules25225317] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising cancer treatment which involves a photosensitizer (PS), light at a specific wavelength for PS activation and oxygen, which combine to elicit cell death. While the illumination required to activate a PS imparts a certain amount of selectivity to PDT treatments, poor tumor accumulation and cell internalization are still inherent properties of most intravenously administered PSs. As a result, common consequences of PDT include skin photosensitivity. To overcome the mentioned issues, PSs may be tailored to specifically target overexpressed biomarkers of tumors. This active targeting can be achieved by direct conjugation of the PS to a ligand with enhanced affinity for a target overexpressed on cancer cells and/or other cells of the tumor microenvironment. Alternatively, PSs may be incorporated into ligand-targeted nanocarriers, which may also encompass multi-functionalities, including diagnosis and therapy. In this review, we highlight the major advances in active targeting of PSs, either by means of ligand-derived bioconjugates or by exploiting ligand-targeting nanocarriers.
Collapse
Affiliation(s)
- Piotr Gierlich
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Ana I. Mata
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| | - Claire Donohoe
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Rui M. M. Brito
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- BSIM Therapeutics, Instituto Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Mathias O. Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Lígia C. Gomes-da-Silva
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| |
Collapse
|
30
|
Dinakaran D, Sengupta J, Pink D, Raturi A, Chen H, Usmani N, Kumar P, Lewis JD, Narain R, Moore RB. PEG-PLGA nanospheres loaded with nanoscintillators and photosensitizers for radiation-activated photodynamic therapy. Acta Biomater 2020; 117:335-348. [PMID: 32956872 DOI: 10.1016/j.actbio.2020.09.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022]
Abstract
Photodynamic Therapy (PDT) is an effective treatment modality for cancers, with Protoporphyrin IX (PPIX)-based PDT being the most widely used to treat cancers in patients. However, PDT is limited to superficial, thin (few mm in depth) lesions that can be accessed by visible wavelength light. Interstitial light-delivery strategies have been developed to treat deep-seated lesions (i.e. prostate cancer). The most promising of these are X-ray-induced scintillation nanoparticles, which have shown potential benefits for PDT of deep-seated tumors. Herein, the design and use of a new nanoscintillator-based radiation-activated PDT (radioPDT) system is investigated in the treatment of deep-seated tumors. Poly(ethylene glycol) methyl ether-block-poly(lactide-co-glycolide) (PEG-PLGA) nanospheres were loaded with a scintillator (LaF3:Ce3+) and photosensitizer (PPIX) to effect radioPDT. UV-Vis spectroscopy and electron microscopy studies demonstrated efficient encapsulation of nanoscintillators and PPIX (>90% efficiency) into the PEG-PLGA nanospheres. The nanoparticles were uniform in size and approximately 100 nm in diameter. They were highly stable and functional for up to 24 h under physiological conditions and demonstrated slow release kinetics. In vitro and in vivo toxicity studies showed no appreciable drug toxicity to human skin fibroblast (GM38), prostate cancer cells (PC3), and to C57/BL mice. Cell uptake studies demonstrated accumulation of the nanoparticles in the cytoplasm of PC3 cells. When activated, fluorescent resonant energy transfer (FRET) was evident via fluorescent spectroscopy and singlet oxygen yield. Determination of stability revealed that the nanoparticles were stable for up to 4 weeks. The nanoparticle production was scaled-up with no change in properties. This nanoparticle represents a unique, optimally designed therapeutic and diagnostic agent (theranostic) agent for radioPDT with characteristics capable of potentially augmenting radiotherapy for deep-seated tumors and integrating into current cancer radiotherapy.
Collapse
Affiliation(s)
- Deepak Dinakaran
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada.
| | - Jayeeta Sengupta
- Department of Chemical and Materials Engineering, University of Alberta, 9211-116 Street NW, Edmonton, AB T6G 2G6, Canada
| | - Desmond Pink
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada; Nanostics Precision Health, Edmonton, AB, Canada
| | - Arun Raturi
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada; Entos Pharmaceuticals, Edmonton, AB. Canada
| | - Hua Chen
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada
| | - Nawaid Usmani
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada
| | - Piyush Kumar
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada
| | - John D Lewis
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, 9211-116 Street NW, Edmonton, AB T6G 2G6, Canada.
| | - Ronald B Moore
- Department of Oncology, University of Alberta, 11560 University Avenue NW, Edmonton, AB T6G 1Z2, Canada; Department of Surgery, University of Alberta, 8440 - 112 Street NW, Edmonton, AB T6G 2B7, Canada
| |
Collapse
|
31
|
Gomez S, Tsung A, Hu Z. Current Targets and Bioconjugation Strategies in Photodynamic Diagnosis and Therapy of Cancer. Molecules 2020; 25:E4964. [PMID: 33121022 PMCID: PMC7662882 DOI: 10.3390/molecules25214964] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/18/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
Photodynamic diagnosis (PDD) and therapy (PDT) are emerging, non/minimally invasive techniques for cancer diagnosis and treatment. Both techniques require a photosensitizer and light to visualize or destroy cancer cells. However, a limitation of conventional, non-targeted PDT is poor selectivity, causing side effects. The bioconjugation of a photosensitizer to a tumor-targeting molecule, such as an antibody or a ligand peptide, is a way to improve selectivity. The bioconjugation strategy can generate a tumor-targeting photosensitizer conjugate specific for cancer cells, or ideally, for multiple tumor compartments to improve selectivity and efficacy, such as cancer stem cells and tumor neovasculature within the tumor microenvironment. If successful, such targeted photosensitizer conjugates can also be used for specific visualization and detection of cancer cells and/or tumor angiogenesis (an early event in tumorigenesis) with the hope of an early diagnosis of cancer. The purpose of this review is to summarize some current promising target molecules, e.g., tissue factor (also known as CD142), and the currently used bioconjugation strategies in PDT and PDD, with a focus on newly developed protein photosensitizers. These are genetically engineered photosensitizers, with the possibility of generating a fusion protein photosensitizer by recombinant DNA technology for both PDT and PDD without the need of chemical conjugation. We believe that providing an overview of promising targets and bioconjugation strategies will aid in driving research in this field forward towards more effective, less toxic, and non- or minimally invasive treatment and diagnosis options for cancer patients.
Collapse
Affiliation(s)
- Salvador Gomez
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
- College of Medicine, The Ohio State University, 370 W 9th Ave, Columbus, OH 43210, USA
| | - Allan Tsung
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
| | - Zhiwei Hu
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
| |
Collapse
|
32
|
Hou YJ, Yang XX, Liu RQ, Zhao D, Guo CX, Zhu AC, Wen MN, Liu Z, Qu GF, Meng HX. Pathological Mechanism of Photodynamic Therapy and Photothermal Therapy Based on Nanoparticles. Int J Nanomedicine 2020; 15:6827-6838. [PMID: 32982235 PMCID: PMC7501968 DOI: 10.2147/ijn.s269321] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
The ultimate goal of phototherapy based on nanoparticles, such as photothermal therapy (PTT) which generates heat and photodynamic therapy (PDT) which not only generates reactive oxygen species (ROS) but also induces a variety of anti-tumor immunity, is to kill tumors. In addition, due to strong efficacy in clinical treatment with minimal invasion and negligible side effects, it has received extensive attention and research in recent years. In this paper, the generations of nanomaterials in PTT and PDT are described separately. In clinical application, according to the different combination pathway of nanoparticles, it can be used to treat different diseases such as tumors, melanoma, rheumatoid and so on. In this paper, the mechanism of pathological treatment is described in detail in terms of inducing apoptosis of cancer cells by ROS produced by PDT, immunogenic cell death to provoke the maturation of dendritic cells, which in turn activate production of CD4+ T cells, CD8+T cells and memory T cells, as well as inhibiting heat shock protein (HSPs), STAT3 signal pathway and so on.
Collapse
Affiliation(s)
- Yun-Jing Hou
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Xin-Xin Yang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Rui-Qi Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Hospital, Guangzhou, People's Republic of China
| | - Di Zhao
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Chen-Xu Guo
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - An-Chao Zhu
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Mei-Na Wen
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University, Harbin, People's Republic of China
| | - Guo-Fan Qu
- Department of Orthopedics, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Hong-Xue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| |
Collapse
|
33
|
Gu X, Shen C, Li H, Goldys EM, Deng W. X-ray induced photodynamic therapy (PDT) with a mitochondria-targeted liposome delivery system. J Nanobiotechnology 2020; 18:87. [PMID: 32522291 PMCID: PMC7288491 DOI: 10.1186/s12951-020-00644-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 06/01/2020] [Indexed: 01/19/2023] Open
Abstract
In this study, we constructed multifunctional liposomes with preferentially mitochondria-targeted feature and gold nanoparticles-assisted synergistic photodynamic therapy. We systemically investigated the in vitro X-ray triggered PDT effect of these liposomes on HCT 116 cells including the levels of singlet oxygen, mitochondrial membrane potential, cell apoptosis/necrosis and the expression of apoptosis-related proteins. The results corroborated that synchronous action of PDT and X-ray radiation enhance the generation of cytotoxic reactive oxygen species produced from the engineered liposomes, causing mitochondrial dysfunction and increasing the levels of apoptosis.
Collapse
Affiliation(s)
- Xuefan Gu
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.,ARC Centre of Excellence for Nanoscale Biophotonics, Graduate School of Biomedical Engineering, University of New South Wales, Kensington, NSW, 2052, Australia.,Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Chao Shen
- Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Hua Li
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Ewa M Goldys
- ARC Centre of Excellence for Nanoscale Biophotonics, Graduate School of Biomedical Engineering, University of New South Wales, Kensington, NSW, 2052, Australia.
| | - Wei Deng
- ARC Centre of Excellence for Nanoscale Biophotonics, Graduate School of Biomedical Engineering, University of New South Wales, Kensington, NSW, 2052, Australia.
| |
Collapse
|
34
|
Deng W, McKelvey KJ, Guller A, Fayzullin A, Campbell JM, Clement S, Habibalahi A, Wargocka Z, Liang L, Shen C, Howell VM, Engel AF, Goldys EM. Application of Mitochondrially Targeted Nanoconstructs to Neoadjuvant X-ray-Induced Photodynamic Therapy for Rectal Cancer. ACS CENTRAL SCIENCE 2020; 6:715-726. [PMID: 32490188 PMCID: PMC7256935 DOI: 10.1021/acscentsci.9b01121] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 05/06/2023]
Abstract
In this work, we brought together two existing clinical techniques used in cancer treatment-X-ray radiation and photodynamic therapy (PDT), whose combination termed X-PDT uniquely allows PDT to be therapeutically effective in deep tissue. To this end, we developed mitochondrially targeted biodegradable polymer poly(lactic-co-glycolic acid) nanocarriers incorporating a photosensitizer verteporfin, ultrasmall (2-5 nm) gold nanoparticles as radiation enhancers, and triphenylphosphonium acting as the mitochondrial targeting moiety. The average size of the nanocarriers was about 160 nm. Upon X-ray radiation our nanocarriers generated cytotoxic amounts of singlet oxygen within the mitochondria, triggering the loss of membrane potential and mitochondria-related apoptosis of cancer cells. Our X-PDT strategy effectively controlled tumor growth with only a fraction of radiotherapy dose (4 Gy) and improved the survival rate of a mouse model bearing colorectal cancer cells. In vivo data indicate that our X-PDT treatment is cytoreductive, antiproliferative, and profibrotic. The nanocarriers induce radiosensitization effectively, which makes it possible to amplify the effects of radiation. A radiation dose of 4 Gy combined with our nanocarriers allows equivalent control of tumor growth as 12 Gy of radiation, but with greatly reduced radiation side effects (significant weight loss and resultant death).
Collapse
Affiliation(s)
- Wei Deng
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
| | - Kelly J. McKelvey
- Bill
Walsh Translational Cancer Research Laboratory, The Northern Clinical
School, Faculty of Medicine and Health, The University of Sydney and Northern Sydney Local Health District
Research (Kolling Institute), St
Leonards, New South Wales 2065, Australia
| | - Anna Guller
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
- Institute
for Regenerative Medicine, Sechenov First Moscow State Medical University
(Sechenov University), Moscow 119991, Russia
| | - Alexey Fayzullin
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
- Institute
for Regenerative Medicine, Sechenov First Moscow State Medical University
(Sechenov University), Moscow 119991, Russia
| | - Jared M. Campbell
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
| | - Sandhya Clement
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
| | - Abbas Habibalahi
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
| | - Zofia Wargocka
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
| | - Liuen Liang
- Department
of Physics and Astronomy, Faculty of Science and Engineering, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Chao Shen
- Faculty
of Science and Engineering, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Viive Maarika Howell
- Bill
Walsh Translational Cancer Research Laboratory, The Northern Clinical
School, Faculty of Medicine and Health, The University of Sydney and Northern Sydney Local Health District
Research (Kolling Institute), St
Leonards, New South Wales 2065, Australia
| | - Alexander Frank Engel
- Sydney Medical
School, University of Sydney, Sydney, New South Wales 2006, Australia
- Department
of Colorectal Surgery, Royal North Shore
Hospital, St Leonards, New South Wales 2065, Australia
| | - Ewa M. Goldys
- ARC
Centre of Excellence for Nanoscale Biophotonics, Graduate School of
Biomedical Engineering, University of New
South Wales, Kensington, New South Wales 2052, Australia
| |
Collapse
|
35
|
Sorrin AJ, Ruhi MK, Ferlic NA, Karimnia V, Polacheck WJ, Celli JP, Huang HC, Rizvi I. Photodynamic Therapy and the Biophysics of the Tumor Microenvironment. Photochem Photobiol 2020; 96:232-259. [PMID: 31895481 PMCID: PMC7138751 DOI: 10.1111/php.13209] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Targeting the tumor microenvironment (TME) provides opportunities to modulate tumor physiology, enhance the delivery of therapeutic agents, impact immune response and overcome resistance. Photodynamic therapy (PDT) is a photochemistry-based, nonthermal modality that produces reactive molecular species at the site of light activation and is in the clinic for nononcologic and oncologic applications. The unique mechanisms and exquisite spatiotemporal control inherent to PDT enable selective modulation or destruction of the TME and cancer cells. Mechanical stress plays an important role in tumor growth and survival, with increasing implications for therapy design and drug delivery, but remains understudied in the context of PDT and PDT-based combinations. This review describes pharmacoengineering and bioengineering approaches in PDT to target cellular and noncellular components of the TME, as well as molecular targets on tumor and tumor-associated cells. Particular emphasis is placed on the role of mechanical stress in the context of targeted PDT regimens, and combinations, for primary and metastatic tumors.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Mustafa Kemal Ruhi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
| | - Nathaniel A. Ferlic
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Vida Karimnia
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA, 02125, USA
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jonathan P. Celli
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA, 02125, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
36
|
Chen J, Fan T, Xie Z, Zeng Q, Xue P, Zheng T, Chen Y, Luo X, Zhang H. Advances in nanomaterials for photodynamic therapy applications: Status and challenges. Biomaterials 2020; 237:119827. [PMID: 32036302 DOI: 10.1016/j.biomaterials.2020.119827] [Citation(s) in RCA: 423] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/13/2020] [Accepted: 01/25/2020] [Indexed: 12/24/2022]
Abstract
Photodynamic therapy (PDT), as a non-invasive therapeutic modality that is alternative to radiotherapy and chemotherapy, is extensively investigated for cancer treatments. Although conventional organic photosensitizers (PSs) are still widely used and have achieved great progresses in PDT, the disadvantages such as hydrophobicity, poor stability within PDT environment and low cell/tissue specificity largely limit their clinical applications. Consequently, nano-agents with promising physicochemical and optical properties have emerged as an attractive alternative to overcome these drawbacks of traditional PSs. Herein, the up-to-date advances in the fabrication and fascinating applications of various nanomaterials in PDT have been summarized, including various types of nanoparticles, carbon-based nanomaterials, and two-dimensional nanomaterials, etc. In addition, the current challenges for the clinical use of PDT, and the corresponding strategies to address these issues, as well as future perspectives on further improvement of PDT have also been discussed.
Collapse
Affiliation(s)
- Jianming Chen
- Institute of Microscale Optoelectronics, Collaborative Innovation Centre for Optoelectronic Science & Technology, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Micro-Nano Photonic Information Technology, Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen University, Shenzhen, 518060, PR China
| | - Taojian Fan
- Institute of Microscale Optoelectronics, Collaborative Innovation Centre for Optoelectronic Science & Technology, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Micro-Nano Photonic Information Technology, Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen University, Shenzhen, 518060, PR China
| | - Zhongjian Xie
- Institute of Microscale Optoelectronics, Collaborative Innovation Centre for Optoelectronic Science & Technology, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Micro-Nano Photonic Information Technology, Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen University, Shenzhen, 518060, PR China
| | - Qiqiao Zeng
- Department of Ophthalmology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen City, Guangdong Province, 518020, PR China
| | - Ping Xue
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Tingting Zheng
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Yun Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Xiaoling Luo
- Department of Ophthalmology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen City, Guangdong Province, 518020, PR China.
| | - Han Zhang
- Institute of Microscale Optoelectronics, Collaborative Innovation Centre for Optoelectronic Science & Technology, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Micro-Nano Photonic Information Technology, Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen University, Shenzhen, 518060, PR China.
| |
Collapse
|
37
|
Ma BA, Sun CY. Tumor pH-triggered “charge conversion” nanocarriers with on-demand drug release for precise cancer therapy. J Mater Chem B 2020; 8:9351-9361. [DOI: 10.1039/d0tb01692f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The pHe-triggered “charge conversion” nanocarriers were developed for combined X-ray-induced photodynamic therapy (X-PDT) and hypoxia-activated chemotherapy.
Collapse
Affiliation(s)
- Bo-Ai Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging
- Tianjin Medical University General Hospital
- Tianjin 300052
- P. R. China
- School of Food and Biological Engineering
| | - Chun-Yang Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging
- Tianjin Medical University General Hospital
- Tianjin 300052
- P. R. China
| |
Collapse
|
38
|
Mohsenian NB, Shanei A, Alavi SJ, Kheirollahi M, Nia AH, Tavakoli MB. Mn-doped ZnS quantum dots-chlorin e6 shows potential as a treatment for chondrosarcoma: an in vitro study. IET Nanobiotechnol 2019; 13:387-391. [PMID: 31171743 DOI: 10.1049/iet-nbt.2018.5387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Chondrosarcoma is the second-most malignant cancer of the bone and routine treatments such as chemotherapy and radiotherapy have not responded to the treatment of this cancer. Due to the resistance of chondrosarcoma to radiotherapy, the combination of therapeutic methods has been considered in recent years. In this study, a novel combination approach is used that allows photodynamic therapy to be activated by X-rays. The synthesis of Mn-doped zinc sulphide (ZnS) quantum dots was carried out and chlorin e6 photosensitiser attached by covalent and non-covalent methods and their application as an intracellular light source for photodynamic activation was investigated. The toxicity of each nanoparticles was evaluated on chondrosarcoma cancer cells (SW1353) before and after radiation. Also, the effect nanoparticle-photosensitiser conjugated type was investigated in the therapeutic efficacy. The characterisation test (SEM, TEM, EDS, TGA, XRD and ICP analyses) was shown successful synthesis of Mn-doped ZnS quantum dots. Chondrosarcoma cancer cell viability was significantly reduced when cells were treated with MPA-capped Mn-doped ZnS quantum dots-chlorin e6 with spermine linker and with covalent attachment (P ≤ 0.001). These results indicate that X-ray can activate the quantum dot complexes for cancer treatment, which can be a novel method for treatment of chondrosarcoma.
Collapse
Affiliation(s)
- Neda Baradaran Mohsenian
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Shanei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Jamal Alavi
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Kheirollahi
- Department of Genetic and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azadeh Hashem Nia
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohamad Bagher Tavakoli
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
39
|
Yang M, Yang T, Mao C. Enhancement of Photodynamic Cancer Therapy by Physical and Chemical Factors. Angew Chem Int Ed Engl 2019; 58:14066-14080. [PMID: 30663185 PMCID: PMC6800243 DOI: 10.1002/anie.201814098] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Indexed: 12/25/2022]
Abstract
The viable use of photodynamic therapy (PDT) in cancer therapy has never been fully realized because of its undesirable effects on healthy tissues. Herein we summarize some physicochemical factors that can make PDT a more viable and effective option to provide future oncological patients with better-quality treatment options. These physicochemical factors include light sources, photosensitizer (PS) carriers, microwaves, electric fields, magnetic fields, and ultrasound. This Review is meant to provide current information pertaining to PDT use, including a discussion of in vitro and in vivo studies. Emphasis is placed on the physicochemical factors and their potential benefits in overcoming the difficulty in transitioning PDT into the medical field. Many advanced techniques, such as employing X-rays as a light source, using nanoparticle-loaded stem cells and bacteriophage bio-nanowires as a photosensitizer carrier, as well as integration with immunotherapy, are among the future directions.
Collapse
Affiliation(s)
- Mingying Yang
- College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Chuanbin Mao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK, 73019, USA
| |
Collapse
|
40
|
Yang M, Yang T, Mao C. Optimierung photodynamischer Krebstherapien auf der Grundlage physikalisch‐chemischer Faktoren. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201814098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mingying Yang
- College of Animal Science Zhejiang University Hangzhou Zhejiang 310058 China
| | - Tao Yang
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 China
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center Institute for Biomedical Engineering, Science and Technology University of Oklahoma 101 Stephenson Parkway Norman OK 73019 USA
| |
Collapse
|
41
|
Liu H, Carter PJH, Laan AC, Eelkema R, Denkova AG. Singlet Oxygen Sensor Green is not a Suitable Probe for 1O 2 in the Presence of Ionizing Radiation. Sci Rep 2019; 9:8393. [PMID: 31182744 PMCID: PMC6557857 DOI: 10.1038/s41598-019-44880-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022] Open
Abstract
A great number of fluorescent probes have been developed for detecting singlet oxygen (1O2), which is considered to be one of the most effective reactive oxygen species (ROS), especially in clinical applications. The commercially available fluorescent probe Singlet Oxygen Sensor Green (SOSG) is widely used due to its reported high selectivity to 1O2. In this study, we carried out systemic experiments to determine the activation of SOSG in the presence of ionizing radiation. The results show that the SOSG probe exhibits a pronounced fluorescence increase as a function of radiation dose delivered by gamma-rays as well as X-rays, in conditions where the formation of singlet oxygen is not expected. Furthermore, scavenger tests indicate that hydroxyl radicals may be involved directly or indirectly in the activation process of SOSG although the exact mechanism remains unknown.
Collapse
Affiliation(s)
- Huanhuan Liu
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629, JB, Delft, The Netherlands.,Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629, HZ, Delft, The Netherlands
| | - Philippe J H Carter
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629, JB, Delft, The Netherlands
| | - Adrianus C Laan
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629, JB, Delft, The Netherlands
| | - Rienk Eelkema
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629, HZ, Delft, The Netherlands.
| | - Antonia G Denkova
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629, JB, Delft, The Netherlands.
| |
Collapse
|
42
|
Shi L, Liu P, Wu J, Ma L, Zheng H, Antosh MP, Zhang H, Wang B, Chen W, Wang X. The effectiveness and safety of X-PDT for cutaneous squamous cell carcinoma and melanoma. Nanomedicine (Lond) 2019; 14:2027-2043. [PMID: 31165659 DOI: 10.2217/nnm-2019-0094] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: To clarify the effectiveness and safety of x-ray-activated photodynamic therapy (X-PDT) for cutaneous squamous cell carcinoma (SCC) and melanoma. Materials & methods: Copper-cysteamine nanoparticles were used as a photosensitizer of X-PDT. The dark toxicity and cytotoxicity were studied in vitro. Tumor volume, microvessel density and acute toxicity of mice were evaluated in vivo. Results: Without x-ray irradiation, copper-cysteamine nanoparticles were nontoxic for keratinocyte cells. XL50 cells (SCC) were more sensitive to X-PDT than B16F10 cells (melanoma). X-PDT successfully inhibited the growth of SCC in vivo (p < 0.05), while the B16F10 melanoma was resistant. Microvessel density in SCC tissue was remarkably reduced (p < 0.05). No obvious acute toxicity reaction was observed. Conclusion: X-PDT is a safe and effective treatment for SCC.
Collapse
Affiliation(s)
- Lei Shi
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, PR China
| | - Pei Liu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, PR China
| | - Jing Wu
- Department of Computer Science & Statistics, University of Rhode Island, 9 Greenhouse Rd, Kingston, RI 02881, USA
| | - Lun Ma
- Department of Physics, the University of Texas at Arlington, Arlington, TX 76019-0059, USA
| | - Han Zheng
- Department of Physics, the University of Texas at Arlington, Arlington, TX 76019-0059, USA
| | - Michael P Antosh
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI 02881, USA.,Institute for Brain & Neural Systems, Brown University, 184 Hope St, Providence, RI 02912, USA
| | - Haiyan Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, PR China
| | - Bo Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, PR China
| | - Wei Chen
- Department of Physics, the University of Texas at Arlington, Arlington, TX 76019-0059, USA
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, PR China
| |
Collapse
|
43
|
Chen X, Song J, Chen X, Yang H. X-ray-activated nanosystems for theranostic applications. Chem Soc Rev 2019; 48:3073-3101. [PMID: 31106315 DOI: 10.1039/c8cs00921j] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
X-rays are widely applied in clinical medical facilities for radiotherapy (RT) and biomedical imaging. However, the sole use of X-rays for cancer treatment leads to insufficient radiation energy deposition due to the low X-ray attenuation coefficients of living tissues and organs, producing unavoidable excessive radiation doses with serious side effects to healthy body parts. Over the past decade, developments in materials science and nanotechnology have led to rapid progress in the field of X-ray-activated tumor-targeting nanosystems, which are able to tackle even systemic tumors and relieve the burden of exposure to large radiation doses. Additionally, novel imaging contrast agents and techniques have also been developed. In comparison with conventional external light sources (e.g., near infrared), the X-ray technique is ideal for the activation of nanosystems for cancer treatment and biomedical imaging applications due to its nearly unlimited penetration depth in living tissues and organisms. In this review, we systematically describe the interaction mechanisms between X-rays and nanosystems, and provide an overview of X-ray-sensitive materials and the recent progress on X-ray-activated nanosystems for cancer-associated theranostic applications.
Collapse
Affiliation(s)
- Xiaofeng Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.
| | | | | | | |
Collapse
|