1
|
Yao XQ, Sabatinos SA, Da Silva E, Taggar A, Ha D, Khan R, Karshafian R, Gräfe J. Therapeutic enhancement effects using a lower energy 2.5 MV photon beam combined with gold nanoparticles on the BxPC-3 pancreatic cancer cell line, in vitro. Phys Med Biol 2025; 70:105017. [PMID: 40328291 DOI: 10.1088/1361-6560/add4b8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
Objective. This study investigates the feasibility of using a clinically relevant lower energy 2.5 megavoltage (MV) photon beam in combination with gold nanoparticles (GNPs).Approach.Pancreatic cancer cell line, BxPC-3 impregnated with GNPs were exposedin vitroto 2.5 MV photon beam and compared with orthovoltage 225 kV and clinical 6 MV photon beam. Bare, 50 nm diameter, spherical GNPs were introduced in the cell culture 24 h prior to irradiation at a concentration of either 10μg ml-1or 50μg ml-1. GNP uptake was determined using inductively coupled plasma optical emission spectroscopy. The cells were irradiated with doses between 0 Gy to 8 Gy. Cell survival curves were obtained via clonogenic assay using immediate or delayed plating (24 h) methods 12 d after irradiation. The terminal deoxynucleotidyl transferase dUTP nick end labeling assay was used to evaluate DNA damage at two time points post irradiation, immediate and 24 h for 1 Gy and 6 Gy.Main results. The enhancement factor (EF) in BxPC-3 cells was greatest for cells incubated with 50μg ml-1of GNPs analyzed immediately post irradiation. Cells irradiated with 225 kV showed greatest EF (1.57 ± 0.15), followed by 2.5 MV (1.51 ± 0.04). The lowest EF was seen for 6 MV, immediate plating (1.10 ± 0.04). A significant increase in the number of DNA double strand breaks (DSB) was observed in cells incubated with 50μg ml-1of GNPs irradiated at 6 Gy with 225 kV and 2.5 MV. There was no significant increase in DSBs for the cells irradiated with 6 MV.Significance.These results suggest that the 2.5 MV could be a compromise between an orthovoltage energy beam and a clinical 6 MV beam, showing comparable reduction in cell survival to the 225 kV beam. Future GNP radiation enhancement research may focus on intermediate energy beams.
Collapse
Affiliation(s)
- Xiao Qing Yao
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario M5B 1T8, Canada
| | - Sarah A Sabatinos
- Department of Chemistry & Biology, Toronto Metropolitan University, Toronto M5B 2K3, Canada
| | - Eric Da Silva
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Amandeep Taggar
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto M4N 3M5, Canada
| | - Diana Ha
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Rao Khan
- Department of Physics and Astronomy, Howard University, Washington, DC, United States of America
| | - Raffi Karshafian
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario M5B 1T8, Canada
| | - James Gräfe
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Department of Medical Physics, Dr H. Bliss Murphy Cancer Centre, NL Health Services, St. John's, Newfoundland, Canada
| |
Collapse
|
2
|
Zeinizade E, Yousefalizideh G, Aminfar P, Horn M, Ding L, Pires L, Jaglanian A, Malbeteau L, Harrington K, Calçada C, Dukuray M, Wilson BC, Koritzinsky M, Chen J, Stamplecoskie KG, Zheng G. Atomically-precise Au 22(Lys-Cys-Lys) 16 nanoclusters for radiation sensitization. J Nanobiotechnology 2025; 23:185. [PMID: 40050865 PMCID: PMC11887354 DOI: 10.1186/s12951-025-03256-7] [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: 01/18/2025] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
Radiotherapy is a leading method for cancer treatment, effectively eliminating cancer cells but often causing collateral damage to surrounding healthy tissue. Radiosensitizers aim to enhance the therapeutic effects of radiotherapy while minimizing harm to normal cells. We recently reported atomically-precise gold nanoclusters, Au22(Lys-Cys-Lys)16, synthesized via a photochemical method coupled with a novel accelerated size-focusing procedure. These nanoclusters exhibit a distinct luminescence emission profile, reflecting exceptional optical purity and the absence of contamination from other nanocluster species. They demonstrate efficient oxygen radicals generation under light irradiation. In this study, we comprehensively evaluated the radiosensitization potential of Au22(Lys-Cys-Lys)16 nanoclusters in vitro and in vivo, alongside their pharmacokinetics, biodistribution and toxicity. The nanoclusters demonstrated high stability under physiological conditions and efficient internalization in tumor cells, achieving dose enhancement factors of 2.0 and 1.6 in KB and 4T1 tumor cells, respectively, under 225 kVp X-ray irradiation. Mechanistic investigations revealed enhanced radiation-induced DNA damage and disruption of DNA repair pathways. The radiosensitizing effects were further validated in radioresistant pancreatic ductal adenocarcinoma cells using the clonogenic assay and γH2AX analysis of double-strand breaks, as well as in a duck chorioallantoic membrane model. With ultra small size (~ 1.7 nm) and favorable surface framework, the nanoclusters exhibited relevant pharmacokinetics (circulation half-life, t₁/₂ = 10.4 h) and renal clearance. In a KB tumor-bearing mouse model, Au22(Lys-Cys-Lys)16 significantly delayed tumor progression and prolonged survival under 8 Gy irradiation without observed side-effects. These findings establish Au22(Lys-Cys-Lys)16 nanoclusters as a potentially translatable radiosensitizer, advancing cancer radiotherapy strategies.
Collapse
Affiliation(s)
- Elham Zeinizade
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | | | - Parimah Aminfar
- Department of Chemistry, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Matthew Horn
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Lili Ding
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Layla Pires
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Alina Jaglanian
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Lucie Malbeteau
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Kristen Harrington
- Department of Chemistry, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Carla Calçada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Mohamad Dukuray
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Brian C Wilson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Juan Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada.
| | | | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
3
|
Vlastou E, Kougioumtzopoulou A, Platoni K, Georgakopoulos I, Lagopati N, Kouloulias V, Zygogianni A. The Emerging Role of Nanoparticles Combined with Either Radiotherapy or Hyperthermia in Head and Neck Cancer: A Current Review. Cancers (Basel) 2025; 17:899. [PMID: 40075746 PMCID: PMC11899074 DOI: 10.3390/cancers17050899] [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: 01/16/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Head and neck cancer (HNC) includes various malignancies and represents the seventh most common cancer worldwide. The early diagnosis of HNC results in a 70-90% five-year survival rate, which declines with locally advanced stages of disease. Current care employs a multimodal strategy encompassing surgery, radiation therapy (RT), chemotherapy, and immunotherapy, while treatment options vary according to the stage, tumor features, and patient characteristics. About 75% of patients with HNC will benefit from RT, either as a primary treatment or as adjuvant therapy following surgical resection. Technological improvements in RT, such as intensity-modulated RT (IMRT) and image-guided RT (IGRT), have enhanced tumor targeting and minimized adjacent healthy tissue irradiation while also expanding RT to the recurrent or metastatic setting. Innovative therapeutic strategies for HNC integrate RT with immunotherapy, gene therapy, molecular targeted therapy, photodynamic therapy, photothermal therapy, and nanoparticles (NPs), with the objective of optimizing tumor control while reducing damage to normal tissues. NPs are emerging as possible radiosensitizers in HNC treatment, enhancing the efficacy of RT, chemotherapy, and immunotherapy. In vivo and in vitro studies on the irradiation of tumors containing gold (Au), gadolinium (Gd), and hafnium oxide (HfO2) NPs show promising results in enhancing tumor destruction and survival rates, indicating their potential for clinical application. Hyperthermia, investigated as an adjunct treatment, potentially improves outcomes when combined with RT or chemotherapy, with advancements in nanotechnology renewing interest in this approach in HNC. At present, NBTXR3 is the sole NP that is being investigated in clinical trials for the enhancement of HNC RT.
Collapse
Affiliation(s)
- Elena Vlastou
- Radiotherapy Department, General Children’s Hospital ‘Pan. & Aglaia Kyriakou’, 11527 Athens, Greece;
| | - Andromachi Kougioumtzopoulou
- Department of Clinical Radiation Oncology, “ATTIKON” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.K.); (K.P.)
| | - Kalliopi Platoni
- Department of Clinical Radiation Oncology, “ATTIKON” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.K.); (K.P.)
| | - Ioannis Georgakopoulos
- Radiotherapy Unit, 1st Radiology Department, ‘Aretaieion’ University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.G.); (A.Z.)
| | - Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vasileios Kouloulias
- Department of Clinical Radiation Oncology, “ATTIKON” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.K.); (K.P.)
| | - Anna Zygogianni
- Radiotherapy Unit, 1st Radiology Department, ‘Aretaieion’ University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.G.); (A.Z.)
| |
Collapse
|
4
|
Kornienko AI, Teplonogova MA, Shevelyova MP, Popkov MA, Popov AL, Ivanov VE, Popova NR. Novel Flavin Mononucleotide-Functionalized Cerium Fluoride Nanoparticles for Selective Enhanced X-Ray-Induced Photodynamic Therapy. J Funct Biomater 2024; 15:373. [PMID: 39728173 DOI: 10.3390/jfb15120373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
X-ray-induced photodynamic therapy (X-PDT) represents a promising new method of cancer treatment. A novel type of nanoscintillator based on cerium fluoride (CeF3) nanoparticles (NPs) modified with flavin mononucleotide (FMN) has been proposed. A method for synthesizing CeF3-FMN NPs has been developed, enabling the production of colloidal, spherical NPs with an approximate diameter of 100 nm, low polydispersity, and a high fluorescence quantum yield of 0.42. It has been demonstrated that CeF3-FMN NPs exhibit pH-dependent radiation-induced redox activity when exposed to X-rays. This activity results in the generation of reactive oxygen species, which is associated with the scintillation properties of cerium and the transfer of electrons to FMN. The synthesized NPs have been demonstrated to exhibit minimal cytotoxicity towards normal cells (NCTC L929 fibroblasts) but are more toxic to tumor cells (epidermoid carcinoma A431). Concurrently, the synthesized NPs (CeF3 and CeF3-FMN NPs) demonstrate a pronounced selective radiosensitizing effect on tumor cells at concentrations of 10-7 and 10-3 M, resulting in a significant reduction in their clonogenic activity, increasing radiosensitivity for cancer cells by 1.9 times following X-ray irradiation at a dose of 3 to 6 Gy. In the context of normal cells, these nanoparticles serve the function of antioxidants, maintaining a high level of clonogenic activity. Functional nanoscintillators on the basis of cerium fluoride can be used as part of the latest technologies for the treatment of tumors within the framework of X-PDT.
Collapse
Affiliation(s)
- Anastasia I Kornienko
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Maria A Teplonogova
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, Moscow 119334, Russia
| | - Marina P Shevelyova
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Pushchino 142290, Russia
| | - Matvei A Popkov
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, Moscow 119334, Russia
| | - Anton L Popov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Vladimir E Ivanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Nelli R Popova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| |
Collapse
|
5
|
Tabbakh F. Significance of the proton energy loss mechanism to gold nanoparticles in proton therapy: a Geant4 simulation. Sci Rep 2024; 14:24978. [PMID: 39443616 PMCID: PMC11499847 DOI: 10.1038/s41598-024-76244-w] [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: 06/13/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
The biological effectiveness in proton therapy is only slightly greater than of the treatment by X-ray and hence, many researches have suggested the use of gold nanoparticles for increasing the ionization interactions to produce more secondary electrons and elevate the yield of DNA damage. But the ionization interactions also lead to protons energy loss inside the nanoparticles. The present study shows that, the protons slowed-down by High-Z nanoparticles are responsible for dose enhancement rather than the produced secondary electrons. To this purpose, using Geant4 Monte Carlo tool, one million nanoparticles distributed in a proton irradiated volume and variation of the proton's spectra and the dose related to this variation has been demonstrated. It was found that, the elevation in proton's LET values when passing through the gold nanoparticles will lead to a more significant dose enhancement than the increased dose due to the extra secondary electrons. Also, it was found that, the mechanism of protons slowing-down by gold nanoparticles has another useful aspect in proton therapy in which, the dose leakage to surrounding healthy tissues will be reduced which must be considered in future investigations more precisely.
Collapse
Affiliation(s)
- Farshid Tabbakh
- Plasma and Nuclear Fusion Research School, Nuclear Science and Technology Research Institute, Tehran, Iran, 14155-1339.
| |
Collapse
|
6
|
Wu L, He C, Zhao T, Li T, Xu H, Wen J, Xu X, Gao L. Diagnosis and treatment status of inoperable locally advanced breast cancer and the application value of inorganic nanomaterials. J Nanobiotechnology 2024; 22:366. [PMID: 38918821 PMCID: PMC11197354 DOI: 10.1186/s12951-024-02644-9] [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: 03/26/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
Locally advanced breast cancer (LABC) is a heterogeneous group of breast cancer that accounts for 10-30% of breast cancer cases. Despite the ongoing development of current treatment methods, LABC remains a severe and complex public health concern around the world, thus prompting the urgent requirement for innovative diagnosis and treatment strategies. The primary treatment challenges are inoperable clinical status and ineffective local control methods. With the rapid advancement of nanotechnology, inorganic nanoparticles (INPs) exhibit a potential application prospect in diagnosing and treating breast cancer. Due to the unique inherent characteristics of INPs, different functions can be performed via appropriate modifications and constructions, thus making them suitable for different imaging technology strategies and treatment schemes. INPs can improve the efficacy of conventional local radiotherapy treatment. In the face of inoperable LABC, INPs have proposed new local therapeutic methods and fostered the evolution of novel strategies such as photothermal and photodynamic therapy, magnetothermal therapy, sonodynamic therapy, and multifunctional inorganic nanoplatform. This article reviews the advances of INPs in local accurate imaging and breast cancer treatment and offers insights to overcome the existing clinical difficulties in LABC management.
Collapse
Affiliation(s)
- Linxuan Wu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Chuan He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Tingting Zhao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tianqi Li
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Hefeng Xu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Jian Wen
- Department of Breast Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xiaoqian Xu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China.
| | - Lin Gao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
7
|
Wang YL, Lee YH, Chou CL, Chang YS, Liu WC, Chiu HW. Oxidative stress and potential effects of metal nanoparticles: A review of biocompatibility and toxicity concerns. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123617. [PMID: 38395133 DOI: 10.1016/j.envpol.2024.123617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 02/17/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
Metal nanoparticles (M-NPs) have garnered significant attention due to their unique properties, driving diverse applications across packaging, biomedicine, electronics, and environmental remediation. However, the potential health risks associated with M-NPs must not be disregarded. M-NPs' ability to accumulate in organs and traverse the blood-brain barrier poses potential health threats to animals, humans, and the environment. The interaction between M-NPs and various cellular components, including DNA, multiple proteins, and mitochondria, triggers the production of reactive oxygen species (ROS), influencing several cellular activities. These interactions have been linked to various effects, such as protein alterations, the buildup of M-NPs in the Golgi apparatus, heightened lysosomal hydrolases, mitochondrial dysfunction, apoptosis, cell membrane impairment, cytoplasmic disruption, and fluctuations in ATP levels. Despite the evident advantages M-NPs offer in diverse applications, gaps in understanding their biocompatibility and toxicity necessitate further research. This review provides an updated assessment of M-NPs' pros and cons across different applications, emphasizing associated hazards and potential toxicity. To ensure the responsible and safe use of M-NPs, comprehensive research is conducted to fully grasp the potential impact of these nanoparticles on both human health and the environment. By delving into their intricate interactions with biological systems, we can navigate the delicate balance between harnessing the benefits of M-NPs and minimizing potential risks. Further exploration will pave the way for informed decision-making, leading to the conscientious development of these nanomaterials and safeguarding the well-being of society and the environment.
Collapse
Affiliation(s)
- Yung-Li Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, 406, Taiwan
| | - Chu-Lin Chou
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City, 320, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, 110, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 235, Taiwan
| | - Yu-Sheng Chang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 235, Taiwan; Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Wen-Chih Liu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, 114, Taiwan; Section of Nephrology, Department of Medicine, Antai Medical Care Corporation Antai Tian-Sheng Memorial Hospital, Pingtung, 928, Taiwan; Department of Nursing, Meiho University, Pingtung, 912, Taiwan
| | - Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, 110, Taiwan; Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 235, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
8
|
Zhang A, Gao L. The Refined Application and Evolution of Nanotechnology in Enhancing Radiosensitivity During Radiotherapy: Transitioning from Gold Nanoparticles to Multifunctional Nanomaterials. Int J Nanomedicine 2023; 18:6233-6256. [PMID: 37936951 PMCID: PMC10626338 DOI: 10.2147/ijn.s436268] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/21/2023] [Indexed: 11/09/2023] Open
Abstract
Radiotherapy is a pivotal method for treating malignant tumors, and enhancing the therapeutic gain ratio of radiotherapy through physical techniques is the direction of modern precision radiotherapy. Due to the inherent physical properties of high-energy radiation, enhancing the therapeutic gain ratio of radiotherapy through radiophysical techniques inevitably encounters challenges. The combination of hyperthermia and radiotherapy can enhance the radiosensitivity of tumor cells, reduce their radioresistance, and holds significant clinical utility in radiotherapy. Multifunctional nanomaterials with excellent biocompatibility and safety have garnered widespread attention in tumor hyperthermia research, demonstrating promising potential. Utilizing nanotechnology as a sensitizing carrier in conjunction with radiotherapy, and high atomic number nanomaterials can also serve independently as radiosensitizing carriers. This synergy between tumor hyperthermia and radiotherapy may overcome many challenges currently limiting tumor radiotherapy, offering new opportunities for its further advancement. In recent years, the continuous progress in the synthesis and design of novel nanomaterials will propel the future development of medical imaging and cancer treatment. This article summarizes the radiosensitizing mechanisms and effects based on gold nanotechnology and provides an overview of the advancements of other nanoparticles (such as bismuth-based nanomaterials, magnetic nanomaterials, selenium nanomaterials, etc.) in the process of radiation therapy.
Collapse
Affiliation(s)
- Anqi Zhang
- Oncology Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, Hebei, People’s Republic of China
| | - Lei Gao
- Medical Imaging Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, Hebei, People’s Republic of China
| |
Collapse
|
9
|
Ibáñez-Moragues M, Fernández-Barahona I, Santacruz R, Oteo M, Luján-Rodríguez VM, Muñoz-Hernando M, Magro N, Lagares JI, Romero E, España S, Espinosa-Rodríguez A, García-Díez M, Martínez-Nouvilas V, Sánchez-Tembleque V, Udías JM, Valladolid-Onecha V, Martín-Rey MÁ, Almeida-Cordon EI, Viñals i Onsès S, Pérez JM, Fraile LM, Herranz F, Morcillo MÁ. Zinc-Doped Iron Oxide Nanoparticles as a Proton-Activatable Agent for Dose Range Verification in Proton Therapy. Molecules 2023; 28:6874. [PMID: 37836718 PMCID: PMC10574368 DOI: 10.3390/molecules28196874] [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: 07/19/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Proton therapy allows the treatment of specific areas and avoids the surrounding tissues. However, this technique has uncertainties in terms of the distal dose fall-off. A promising approach to studying the proton range is the use of nanoparticles as proton-activatable agents that produce detectable signals. For this, we developed an iron oxide nanoparticle doped with Zn (IONP@Zn-cit) with a hydrodynamic size of 10 nm and stability in serum. Cytotoxicity, defined as half of the surveillance, was 100 μg Zn/mL in the U251 cell line. The effect on clonogenic cell death was tested after X-ray irradiation, which suggested a radioprotective effect of these nanoparticles at low concentrations (1-10 μg Zn/mL). To evaluate the production of positron emitters and prompt-gamma signals, IONP@Zn-cit was irradiated with protons, obtaining prompt-gamma signals at the lowest measured concentration (10 mg Zn/mL). Finally, 67Ga-IONP@Zn-cit showed accumulation in the liver and spleen and an accumulation in the tumor tissue of 0.95% ID/g in a mouse model of U251 cells. These results suggest the possibility of using Zn nanoparticles as proton-activatable agents to verify the range by prompt gamma detection and face the challenges of prompt gamma detection in a specific biological situation, opening different avenues to go forward in this field.
Collapse
Affiliation(s)
- Marta Ibáñez-Moragues
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Irene Fernández-Barahona
- Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Instituto de Química Médica—Consejo Superior de Investigaciones Científicas IQM-CSIC, Nanomedicine and Molecular Imaging Group, 28006 Madrid, Spain; (M.M.-H.)
| | - Rocío Santacruz
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Marta Oteo
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Víctor M. Luján-Rodríguez
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - María Muñoz-Hernando
- Instituto de Química Médica—Consejo Superior de Investigaciones Científicas IQM-CSIC, Nanomedicine and Molecular Imaging Group, 28006 Madrid, Spain; (M.M.-H.)
| | - Natalia Magro
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Juan I. Lagares
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Eduardo Romero
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Samuel España
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Andrea Espinosa-Rodríguez
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Miguel García-Díez
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Víctor Martínez-Nouvilas
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Víctor Sánchez-Tembleque
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - José Manuel Udías
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Víctor Valladolid-Onecha
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Miguel Á. Martín-Rey
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Hematopoietic Innovative Therapies Unit, 28040 Madrid, Spain;
| | - Edilia I. Almeida-Cordon
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Animal Facility Unit, 28040 Madrid, Spain;
| | - Sílvia Viñals i Onsès
- Center for Microanalysis of Materials (CMAM), Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - José Manuel Pérez
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| | - Luis Mario Fraile
- Nuclear Physics Group, Universidad Complutense de Madrid, IPARCOS &EMFTEL, CEI Moncloa, 28040 Madrid, Spain; (S.E.); (A.E.-R.); (M.G.-D.); (V.M.-N.); (V.S.-T.); (J.M.U.); (V.V.-O.); (L.M.F.)
- Instituto de Investigación del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Fernando Herranz
- Instituto de Química Médica—Consejo Superior de Investigaciones Científicas IQM-CSIC, Nanomedicine and Molecular Imaging Group, 28006 Madrid, Spain; (M.M.-H.)
| | - Miguel Ángel Morcillo
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas CIEMAT, Medical Applications of Ionizing Radiation Unit, 28040 Madrid, Spain; (R.S.); (M.O.); (V.M.L.-R.); (N.M.); (J.I.L.); (E.R.); (J.M.P.)
| |
Collapse
|
10
|
Wu Y, Zhu K, Zhang X, Du W, Song J, Yang H. Emerging plasmonic nanoparticles and their assemblies for cancer radiotherapy. Adv Drug Deliv Rev 2023; 194:114710. [PMID: 36708774 DOI: 10.1016/j.addr.2023.114710] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/07/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
Plasmonic nanoparticles and their assemblies have been widely used in biosensing, optical imaging, and biomedicine over the past few decades. Especially in the field of radiotherapy, the physicochemical properties of high-Z plasmonic nanomaterials endow them with the ability to sensitize radiotherapy. Compared with single particles, the assembled structure with tunable properties leads to versatile applications in drug delivery and cancer treatment. In this review, we focus on plasmonic nanoparticles and their assemblies for cancer radiotherapy. First, the sensitization mechanism of plasmonic radiosensitizers is briefly introduced. Subsequently, the recent progress in cancer radiotherapy is systematically discussed according to the structure and shape classification. Finally, the current challenges and future perspectives in this field are also discussed in detail.
Collapse
Affiliation(s)
- Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China
| | - Xuan Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Wei Du
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China.
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China.
| |
Collapse
|
11
|
Du TQ, Liu R, Zhang Q, Luo H, Chen Y, Tan M, Wang Q, Wu X, Liu Z, Sun S, Yang K, Tian J, Wang X. Does particle radiation have superior radiobiological advantages for prostate cancer cells? A systematic review of in vitro studies. Eur J Med Res 2022; 27:306. [PMID: 36572945 PMCID: PMC9793637 DOI: 10.1186/s40001-022-00942-2] [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: 09/21/2022] [Accepted: 12/07/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Charged particle beams from protons to carbon ions provide many significant physical benefits in radiation therapy. However, preclinical studies of charged particle therapy for prostate cancer are extremely limited. The aim of this study was to comprehensively investigate the biological effects of charged particles on prostate cancer from the perspective of in vitro studies. METHODS We conducted a systematic review by searching EMBASE (OVID), Medline (OVID), and Web of Science databases to identify the publications assessing the radiobiological effects of charged particle irradiation on prostate cancer cells. The data of relative biological effectiveness (RBE), surviving fraction (SF), standard enhancement ratio (SER) and oxygen enhancement ratio (OER) were extracted. RESULTS We found 12 studies met the eligible criteria. The relative biological effectiveness values of proton and carbon ion irradiation ranged from 0.94 to 1.52, and 1.67 to 3.7, respectively. Surviving fraction of 2 Gy were 0.17 ± 0.12, 0.55 ± 0.20 and 0.53 ± 0.16 in carbon ion, proton, and photon irradiation, respectively. PNKP inhibitor and gold nanoparticles were favorable sensitizing agents, while it was presented poorer performance in GANT61. The oxygen enhancement ratio values of photon and carbon ion irradiation were 2.32 ± 0.04, and 1.77 ± 0.13, respectively. Charged particle irradiation induced more G0-/G1- or G2-/M-phase arrest, more expression of γ-H2AX, more apoptosis, and lower motility and/or migration ability than photon irradiation. CONCLUSIONS Both carbon ion and proton irradiation have advantages over photon irradiation in radiobiological effects on prostate cancer cell lines. Carbon ion irradiation seems to have further advantages over proton irradiation.
Collapse
Affiliation(s)
- Tian-Qi Du
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Ruifeng Liu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Qiuning Zhang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Hongtao Luo
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Yanliang Chen
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Mingyu Tan
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Qian Wang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Xun Wu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Zhiqiang Liu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Shilong Sun
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Kehu Yang
- grid.32566.340000 0000 8571 0482Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Jinhui Tian
- grid.32566.340000 0000 8571 0482Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Xiaohu Wang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| |
Collapse
|
12
|
Varzandeh M, Labbaf S, Varshosaz J, Laurent S. An overview of the intracellular localization of high-Z nanoradiosensitizers. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:14-30. [PMID: 36029849 DOI: 10.1016/j.pbiomolbio.2022.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/17/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
Radiation therapy (RT) is a method commonly used for cancer treatment worldwide. Commonly, RT utilizes two routes for combating cancers: 1) high-energy radiation to generate toxic reactive oxygen species (ROS) (through the dissociation of water molecules) for damaging the deoxyribonucleic acid (DNA) inside the nucleus 2) direct degradation of the DNA. However, cancer cells have mechanisms to survive under intense RT, which can considerably decrease its therapeutic efficacy. Excessive radiation energy damages healthy tissues, and hence, low doses are applied for cancer treatment. Additionally, different radiosensitizers were used to sensitize cancer cells towards RT through individual mechanisms. Following this route, nanoparticle-based radiosensitizers (herein called nanoradiosensitizers) have recently gained attention owing to their ability to produce massive electrons which leads to the production of a huge amount of ROS. The success of the nanoradiosensitizer effect is closely correlated to its interaction with cells and its localization within the cells. In other words, tumor treatment is affected from the chain of events which is started from cell-nanoparticle interaction followed by the nanoparticles direction and homing inside the cell. Therefore, passive or active targeting of the nanoradiosensitizers in the subcellular level and the cell-nano interaction would determine the efficacy of the radiation therapy. The importance of the nanoradiosensitizer's targeting is increased while the organelles beyond nucleus are recently recognized as the mediators of the cancer cell death or resistance under RT. In this review, the principals of cell-nanomaterial interactions and which dominate nanoradiosensitizer efficiency in cancer therapy, are thoroughly discussed.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center and Department of Pharmaceutics, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sophie Laurent
- Laboratory of NMR and Molecular Imaging, Department of General, Organic Chemistry and Biomedical, University of Mons, Mons, Belgium.
| |
Collapse
|
13
|
Biological Response of Human Cancer Cells to Ionizing Radiation in Combination with Gold Nanoparticles. Cancers (Basel) 2022; 14:cancers14205086. [PMID: 36291870 PMCID: PMC9600885 DOI: 10.3390/cancers14205086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Various types of metallic nanoparticles and especially gold nanoparticles (AuNPs) have been utilized in radiation studies to enhance the radiosensitization of cancer cells while minimizing detrimental effects in normal tissue. The aim of our study was to investigate the biological responses of various human cancer cells to gold-nanoparticle-induced radiosensitization. This was accomplished by using different AuNPs and several techniques in order to provide valuable insights regarding the multiple adverse biological effects, following ionizing radiation (IR) in combination with AuNPs. Insightful methodologies such as transmission electron microscopy were employed to identify comprehensively the complexity of the biological damage occurrence. Our findings confirm that AuNP radiosensitization may occur due to extensive and/or complex DNA damage, cell death, or cellular senescence. This multiparameter study aims to further elucidate the biological mechanisms and at the same time provide new information regarding the use of AuNPs as radiosensitizers in cancer treatment. Abstract In the context of improving radiation therapy, high-atomic number (Z) metallic nanoparticles and, more importantly, gold-based nanostructures are developed as radiation enhancers/radiosensitizers. Due to the diversity of cell lines, nanoparticles, as well as radiation types or doses, the resulting biological effects may differ and remain obscure. In this multiparameter study, we aim to shed light on these effects and investigate them further by employing X-irradiation and three human cancer cell lines (PC3, A549, and U2OS cells) treated by multiple techniques. TEM experiments on PC3 cells showed that citrate-capped AuNPs were found to be located mostly in membranous structures/vesicles or autophagosomes, but also, in the case of PEG-capped AuNPs, inside the nucleus as well. The colony-forming capability of cancer cells radiosensitized by AuNPs decreased significantly and the DNA damage detected by cytogenetics, γH2AX immunostaining, and by single (γH2AX) or double (γH2AX and OGG1) immunolocalization via transmission electron microscopy (TEM) was in many cases higher and/or persistent after combination with AuNPs than upon individual exposure to ionizing radiation (IR). Moreover, different cell cycle distribution was evident in PC3 but not A549 cells after treatment with AuNPs and/or irradiation. Finally, cellular senescence was investigated by using a newly established staining procedure for lipofuscin, based on a Sudan Black-B analogue (GL13) which showed that based on the AuNPs’ concentration, an increased number of senescent cells might be observed after exposure to IR. Even though different cell lines or different types and concentrations of AuNPs may alter the levels of radiosensitization, our results imply that the complexity of damage might also be an important factor of AuNP-induced radiosensitization.
Collapse
|
14
|
Mochizuki C, Kayabe Y, Nakamura J, Igase M, Mizuno T, Nakamura M. Surface Functionalization of Organosilica Nanoparticles With Au Nanoparticles Inhibits Cell Proliferation and Induces Cell Death in 4T1 Mouse Mammary Tumor Cells for DNA and Mitochondrial-Synergized Damage in Radiotherapy. Front Chem 2022; 10:907642. [PMID: 35620651 PMCID: PMC9127317 DOI: 10.3389/fchem.2022.907642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is one of the most effective cancer treatments. Au nanoparticles (NPs) are one of the most used X-ray sensitizing materials however the effective small sub-nm size of Au NPs used for X-ray sensitizers is disadvantageous for cellular uptake. Here, we propose the surface functionalization of organosilica NPs (OS) with Au NPs (OS/Au), which combined the 100 nm size of OS and the sub-nm size of Au NPs, and synthesized effective Au materials as an X-ray sensitizer. The X-ray sensitizing potential for 4T1 mouse mammary tumor cells was revealed using a multifaceted evaluation combined with a fluorescence microscopic cell imaging assay. The number of polyethyleneimine (PEI)-modified OS (OS/PEI) and OS/Au (OS/Au/PEI) uptake per 4T1 mouse mammary tumor cell was the same; however, 4T1 cells treated with OS/Au/PEI exhibited significant inhibition of cell proliferation and increases in cell death by X-ray irradiation at 8Gy. The non-apoptotic death of OS/Au/PEI-treated 4T1 cells was increased by DNA and mitochondrial-synergized damage increase and showed potential applications in radiotherapy.
Collapse
Affiliation(s)
- Chihiro Mochizuki
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, Yamaguchi, Japan
| | - Yukihito Kayabe
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Junna Nakamura
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, Yamaguchi, Japan
| | - Masaya Igase
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takuya Mizuno
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Michihiro Nakamura
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
15
|
Roy I, Krishnan S, Kabashin AV, Zavestovskaya IN, Prasad PN. Transforming Nuclear Medicine with Nanoradiopharmaceuticals. ACS NANO 2022; 16:5036-5061. [PMID: 35294165 DOI: 10.1021/acsnano.1c10550] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nuclear medicine is expected to make major advances in cancer diagnosis and therapy; tumor-targeted radiopharmaceuticals preferentially eradicate tumors while causing minimal damage to healthy tissues. The current scope of nuclear medicine can be significantly expanded by integration with nanomedicine, which utilizes nanoparticles for cancer diagnosis and therapy by capitalizing on the increased surface area-to-volume ratio, the passive/active targeting ability and high loading capacity, the greater interaction cross section with biological tissues, the rich surface properties of nanomaterials, the facile decoration of nanomaterials with a plethora of functionalities, and the potential for multiplexing several functionalities within one construct. This review provides a comprehensive discussion of nuclear nanomedicine using tumor-targeted nanoparticles for cancer radiation therapy with either pre-embedded radionuclides or nonradioactive materials which can be extrinsically triggered using various external nuclear particle sources to produce in situ radioactivity. In addition, it describes the prospect of combining nuclear nanomedicine with other modalities to enable synergistically enhanced combination therapies. The review also discusses advances in the fabrication of radionuclides as well as describes laser ablation technologies for producing nanoradiopharmaceuticals, which combine the ease of production with exceptional purity and rapid biodegradability, along with additional imaging or therapeutic functionalities. From a practical standpoint, these attributes of nanoradiopharmaceuticals may provide distinct advantages in diagnostic/therapeutic sensitivity and specificity, imaging resolution, and scalability of turnkey platforms. Coupling image-guided targeted radiation therapy with the possibility of in situ activation of nanomaterials as well as combining with other therapeutic modalities using a multifunctional nanoplatform could herald an era of exciting technological and therapeutic advances to radically transform the landscape of nuclear medicine. The review concludes with a discussion of current challenges and presents the authors' views on future opportunities to stimulate further research in this rewarding field of high societal impact.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy - Case 917, 13288 Marseille, France
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
| | - Irina N Zavestovskaya
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Nuclear Physics and Astrophysics Department, LPI of RAS, 119991 Moscow, Russia
| | - Paras N Prasad
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Department of Chemistry and Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
16
|
GMT8 aptamer conjugated PEGylated Ag@Au core-shell nanoparticles as a novel radiosensitizer for targeted radiotherapy of glioma. Colloids Surf B Biointerfaces 2022; 211:112330. [PMID: 35032851 DOI: 10.1016/j.colsurfb.2022.112330] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/12/2021] [Accepted: 01/07/2022] [Indexed: 11/24/2022]
Abstract
Radiotherapy is one of the main treatment modalities for glioma, but the therapeutic efficacy is often limited by the radioresistance of tumor cells. The radiosensitization effects of silver nanoparticles (AgNPs) and gold nanoparticles (AuNPs) on tumors have been confirmed by previous studies. To enhance the specific killing effect of irradiation on tumor cells, targeted modification of radiosensitizers is urgently needed. Herein, we developed polyethylene glycol (PEG)-coated Ag@Au core-shell nanoparticles (PSGNPs) modified with GMT8 aptamer (GSGNPs) and evaluated their radiosensitization effects on glioma cells through in vivo and in vitro experiments. Transmission electron microscope image showed that the prepared PSGNPs had a spherical core-shell structure with an average size of 11 nm. The ultraviolet-visible absorption spectra and Fourier transform infrared spectra displayed that GMT8 was successfully conjugated to PSGNPs. The results of dark-field imaging revealed that the targeting ability of GSGNPs to U87 glioma cells was much better than that to normal human microvascular endothelial cells. Additionally, it was also found that the endocytic pathways of GSGNPs mainly involved clathrin-mediated endocytosis and macropinocytosis. The sensitization enhancement ratio of GSGNPs was calculated to be 1.62, which was higher than that of PSGNPs. In vivo imaging results showed that GSGNPs exhibited good tumor targeting and retention capabilities, and the fluorescence intensity ratio of Cy5-GSGNPs to Cy5-PSGNPs reached a peak at 4 h after injection. More importantly, the median survival time of mice bearing U87 glioma was significantly prolonged after intravenous administration of GSGNPs combined with radiotherapy. This work demonstrated that GSGNPs could be used as an effective nano-radiosensitizer for targeted radiotherapy of glioma.
Collapse
|
17
|
Pickford Scienti OLP, Darambara DG. An Overview of X-ray Photon Counting Spectral Imaging (x-CSI) with a Focus on Gold Nanoparticle Quantification in Oncology. J Imaging 2021; 8:4. [PMID: 35049845 PMCID: PMC8778032 DOI: 10.3390/jimaging8010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023] Open
Abstract
This review article offers an overview of the differences between traditional energy integrating (EI) X-ray imaging and the new technique of X-ray photon counting spectral imaging (x-CSI). The review is motivated by the need to image gold nanoparticles (AuNP) in vivo if they are to be used clinically to deliver a radiotherapy dose-enhancing effect (RDEE). The aim of this work is to familiarise the reader with x-CSI as a technique and to draw attention to how this technique will need to develop to be of clinical use for the described oncological applications. This article covers the conceptual differences between x-CSI and EI approaches, the advantages of x-CSI, constraints on x-CSI system design, and the achievements of x-CSI in AuNP quantification. The results of the review show there are still approximately two orders of magnitude between the AuNP concentrations used in RDEE applications and the demonstrated detection limits of x-CSI. Two approaches to overcome this were suggested: changing AuNP design or changing x-CSI system design. Optimal system parameters for AuNP detection and general spectral performance as determined by simulation studies were different to those used in the current x-CSI systems, indicating potential gains that may be made with this approach.
Collapse
Affiliation(s)
- Oliver L. P. Pickford Scienti
- Joint Department of Physics, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London SM2 5NG, UK;
| | | |
Collapse
|
18
|
Tudda A, Donzelli E, Nicolini G, Semperboni S, Bossi M, Cavaletti G, Castriconi R, Mangili P, Vecchio AD, Sarno A, Mettivier G, Russo P. Breast radiotherapy with kilovoltage photons and gold nanoparticles as radiosensitizer: An in vitro study. Med Phys 2021; 49:568-578. [PMID: 34778990 PMCID: PMC9299863 DOI: 10.1002/mp.15348] [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: 06/27/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/10/2022] Open
Abstract
Purpose We investigated the dose enhancement and internalization of gold nanoparticles (AuNPs) used as a radiosensitizer agent for rotational radiotherapy of breast cancer using a kilovoltage (kV) X‐ray beam. Methods Human breast cancer cells MDA‐MB‐231 were incubated with or without 100 μg/mL (4.87 nM) or 200 μg/mL (9.74 nM) 15 nm AuNPs and irradiated with 100 kV, 190 kV, or 6 MV X‐rays. To assess the toxicity of the AuNPs, we performed a Sulforhodamine B assay. Using atomic absorption spectroscopy, scanning electron microscopy, transmission electron microscopy, and time‐lapse optical microscopy (rate of 2 frames per minute), we carried out a quantitative assessment of the amount of gold internalized by MDA‐MB‐231 cells and a characterization of the static and dynamical aspects of this internalization process. Results No effect of AuNPs alone was shown on cell viability. Time‐lapse optical microscopy showed for the first time AuNPs cellular uptake and the dynamics of AuNPs internalization. Electron microscopy demonstrated AuNPs localization in endosomal vesicles, preferentially in the perinuclear region. After irradiation at doses up to 2 Gy, cell survival fraction curves showed increased mortality with AuNPs, with respect to irradiation without AuNPs. The highest effect of radioenhancement by AuNPs (at 9.74 nM AuNPs concentration) was observed at 190 kV showing a dose enhancement factor of 1.33 ± 0.06 (1.34 ± 0.02 at 100 kV), while at 6 MV it was 1.14 ± 0.06. Conclusions The observed radio‐sensitization effect is promising for future radio‐enhanced kV radiotherapy of breast cancer and quantitatively in the order of previous observations for 15 nm AuNPs. These results of a significant dose enhancement were obtained at 15 nm AuNPs concentration as low as several nanomolar units, at dose levels typical of a single dose fraction in a radiotherapy session. Dynamical behavior of the 3D spatial distribution of 15 nm AuNPs outside the nucleus of single breast cancer cell was observed, with possible implications for future models of AuNPs sensitization.
Collapse
Affiliation(s)
- Alessia Tudda
- Department of Physics "Ettore Pancini", University of Naples Federico II, Naples, Italy.,INFN Division of Naples, Naples, Italy.,Medical Physics Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Department of Physics, Specialty School of Medical Physics, University of Milan, Milan, Italy
| | - Elisabetta Donzelli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,INFN Division of Milano-Bicocca, Milan, Italy
| | - Gabriella Nicolini
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,INFN Division of Milano-Bicocca, Milan, Italy
| | - Sara Semperboni
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,INFN Division of Milano-Bicocca, Milan, Italy
| | - Mario Bossi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Roberta Castriconi
- Medical Physics Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,INFN Division of Milan, Milan, Italy
| | - Paola Mangili
- Medical Physics Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,INFN Division of Milan, Milan, Italy
| | - Antonella Del Vecchio
- Medical Physics Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,INFN Division of Milan, Milan, Italy
| | - Antonio Sarno
- Department of Physics "Ettore Pancini", University of Naples Federico II, Naples, Italy.,INFN Division of Naples, Naples, Italy
| | - Giovanni Mettivier
- Department of Physics "Ettore Pancini", University of Naples Federico II, Naples, Italy.,INFN Division of Naples, Naples, Italy
| | - Paolo Russo
- Department of Physics "Ettore Pancini", University of Naples Federico II, Naples, Italy.,INFN Division of Naples, Naples, Italy
| |
Collapse
|
19
|
Zhang P, Yu B, Jin X, Zhao T, Ye F, Liu X, Li P, Zheng X, Chen W, Li Q. Therapeutic Efficacy of Carbon Ion Irradiation Enhanced by 11-MUA-Capped Gold Nanoparticles: An in vitro and in vivo Study. Int J Nanomedicine 2021; 16:4661-4674. [PMID: 34262274 PMCID: PMC8275145 DOI: 10.2147/ijn.s313678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Gold nanoparticles (AuNPs) are widely studied as radiosensitizers, but their radiosensitization in carbon ion radiotherapy is unsatisfactory. There is a lack of in vivo data on the radiosensitization of AuNPs under carbon ion irradiation. This study focused on the radiosensitization effect of AuNPs in the mouse melanoma cell line B16-F10 in vitro and in vivo. MATERIALS AND METHODS 11-mercaptoundecanoic acid (11-MUA)-coated gold (Au) nanoparticles (mAuNPs) formulations were prepared and characterized. To verify the radiosensitization effect of mAuNPs, hydroxyl radicals were generated in aqueous solution, and the detection of intracellular reactive oxygen species (ROS) and clone survival were carried out in vitro. The tumor growth rate (TGR) and survival of mice were analyzed to verify the radiosensitization effect of mAuNPs in vivo. The apoptosis of tumor cells was detected, and the expression of key proteins in the apoptosis pathway was verified by immunohistochemistry. RESULTS The intracellular ROS level in B16-F10 cells was enhanced by mAuNPs under carbon ion irradiation. The sensitization rate of mAuNPs was 1.22 with a 10% cell survival rate. Compared with irradiation alone, the inhibitory effect of mAuNPs combined with carbon ion irradiation on tumor growth was 1.94-fold higher, the survival time of mice was prolonged by 1.75-fold, and the number of apoptotic cells was increased by 1.43-fold. The ratio of key proteins Bax and Bcl2 in the apoptosis pathway was up-regulated, and the expression of caspase-3, a key executor of the apoptosis pathway, was up-regulated. CONCLUSION In in vivo and in vitro experiments, mAuNPs showed radiosensitivity to carbon ion irradiation. The sensitization effect of mAuNPs on mice tumor may be achieved by activating the mitochondrial apoptosis pathway and increasing tumor tissue apoptosis. To our best knowledge, the present study is the first in vivo evidence for radiosensitization of mAuNPs in tumor-bearing mice exposed to carbon ion irradiation.
Collapse
Affiliation(s)
- Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, 730000, People’s Republic of China
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
| | - Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Xiaogang Zheng
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, 730000, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| |
Collapse
|
20
|
Wang X, Chen X, Li G, Han X, Gao T, Liu W, Tang X. Application of Carbon Ion and Its Sensitizing Agent in Cancer Therapy: A Systematic Review. Front Oncol 2021; 11:708724. [PMID: 34290989 PMCID: PMC8287631 DOI: 10.3389/fonc.2021.708724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Carbon ion radiation therapy (CIRT) is the most advanced radiation therapy (RT) available and offers new opportunities to improve cancer treatment and research. CIRT has a unique physical and biological advantage that allow them to kill tumor cells more accurately and intensively. So far, CIRT has been used in almost all types of malignant tumors, and showed good feasibility, safety and acceptable toxicity, indicating that CIRT has a wide range of development and application prospects. In addition, in order to improve the biological effect of CIRT, scientists are also trying to investigate related sensitizing agents to enhance the killing ability of tumor cells, which has attracted extensive attention. In this review, we tried to systematically review the rationale, advantages and problems, the clinical applications and the sensitizing agents of the CIRT. At the same time, the prospects of the CIRT in were prospected. We hope that this review will help researchers interested in CIRT, sensitizing agents, and radiotherapy to understand their magic more systematically and faster, and provide data reference and support for bioanalysis, clinical medicine, radiotherapy, heavy ion therapy, and nanoparticle diagnostics.
Collapse
Affiliation(s)
- Xiaolin Wang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Xiaojun Chen
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Guangfei Li
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Xiao Han
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Tianxin Gao
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Weifeng Liu
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Xiaoying Tang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
21
|
Popescu RC, Savu DI, Bierbaum M, Grbenicek A, Schneider F, Hosser H, Vasile BȘ, Andronescu E, Wenz F, Giordano FA, Herskind C, Veldwijk MR. Intracellular Delivery of Doxorubicin by Iron Oxide-Based Nano-Constructs Increases Clonogenic Inactivation of Ionizing Radiation in HeLa Cells. Int J Mol Sci 2021; 22:ijms22136778. [PMID: 34202550 PMCID: PMC8267614 DOI: 10.3390/ijms22136778] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, we determined the potential of polyethylene glycol-encapsulated iron oxide nanoparticles (IONPCO) for the intracellular delivery of the chemotherapeutic doxorubicin (IONPDOX) to enhance the cytotoxic effects of ionizing radiation. The biological effects of IONP and X-ray irradiation (50 kV and 6 MV) were determined in HeLa cells using the colony formation assay (CFA) and detection of γH2AX foci. Data are presented as mean ± SEM. IONP were efficiently internalized by HeLa cells. IONPCO radiomodulating effect was dependent on nanoparticle concentration and photon energy. IONPCO did not radiosensitize HeLa cells with 6 MV X-rays, yet moderately enhanced cellular radiosensitivity to 50 kV X-rays (DMFSF0.1 = 1.13 ± 0.05 (p = 0.01)). IONPDOX did enhance the cytotoxicity of 6 MV X-rays (DMFSF0.1 = 1.3 ± 0.1; p = 0.0005). IONP treatment significantly increased γH2AX foci induction without irradiation. Treatment of HeLa cells with IONPCO resulted in a radiosensitizing effect for low-energy X-rays, while exposure to IONPDOX induced radiosensitization compared to IONPCO in cells irradiated with 6 MV X-rays. The effect did not correlate with the induction of γH2AX foci. Given these results, IONP are promising candidates for the controlled delivery of DOX to enhance the cytotoxic effects of ionizing radiation.
Collapse
Affiliation(s)
- Roxana Cristina Popescu
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
- Department of Life and Environmental Physics, “Horia Hulubei” National Institute for Physics and Nuclear Engineering, 077125 Magurele, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania; (B.Ș.V.); (E.A.)
| | - Diana Iulia Savu
- Department of Life and Environmental Physics, “Horia Hulubei” National Institute for Physics and Nuclear Engineering, 077125 Magurele, Romania
- Correspondence: (D.I.S.); (M.R.V.); Tel.: +40214046134 (D.I.S.); +49-621-383-3750 (M.R.V.)
| | - Miriam Bierbaum
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
| | - Adriana Grbenicek
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
| | - Frank Schneider
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
| | - Hiltraud Hosser
- Department of Anatomy and Developmental Biology, Center for Biomedicine and Medical Technology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Bogdan Ștefan Vasile
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania; (B.Ș.V.); (E.A.)
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania; (B.Ș.V.); (E.A.)
| | - Frederik Wenz
- CEO, University Medical Center Freiburg, 79106 Freiburg, Germany;
| | - Frank A. Giordano
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
| | - Carsten Herskind
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
| | - Marlon R. Veldwijk
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (R.C.P.); (M.B.); (A.G.); (F.S.); (F.A.G.); (C.H.)
- Correspondence: (D.I.S.); (M.R.V.); Tel.: +40214046134 (D.I.S.); +49-621-383-3750 (M.R.V.)
| |
Collapse
|
22
|
Yu Z, Gao L, Chen K, Zhang W, Zhang Q, Li Q, Hu K. Nanoparticles: A New Approach to Upgrade Cancer Diagnosis and Treatment. NANOSCALE RESEARCH LETTERS 2021; 16:88. [PMID: 34014432 PMCID: PMC8137776 DOI: 10.1186/s11671-021-03489-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/27/2021] [Indexed: 05/07/2023]
Abstract
Traditional cancer therapeutics have been criticized due to various adverse effects and insufficient damage to targeted tumors. The breakthrough of nanoparticles provides a novel approach for upgrading traditional treatments and diagnosis. Actually, nanoparticles can not only solve the shortcomings of traditional cancer diagnosis and treatment, but also create brand-new perspectives and cutting-edge devices for tumor diagnosis and treatment. However, most of the research about nanoparticles stays in vivo and in vitro stage, and only few clinical researches about nanoparticles have been reported. In this review, we first summarize the current applications of nanoparticles in cancer diagnosis and treatment. After that, we propose the challenges that hinder the clinical applications of NPs and provide feasible solutions in combination with the updated literature in the last two years. At the end, we will provide our opinions on the future developments of NPs in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Zhongyang Yu
- Beijing University of Chinese Medicine, 11 North Third Ring East Road, Chaoyang District, Beijing, 100029, China
| | - Lei Gao
- Oncology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Fangguyuan Rd, Fengtai District, Beijing, 100078, China
| | - Kehan Chen
- College of Engineering, China Agricultural University, Tsinghua East Rd, Haidian District, Beijing, 100083, China
| | - Wenqiang Zhang
- College of Engineering, China Agricultural University, Tsinghua East Rd, Haidian District, Beijing, 100083, China
| | - Qihang Zhang
- Department of Management, Fredericton Campus, University of New Brunswick, 3 Bailey Drive, Fredericton, NB, E3B 5A3, Canada
| | - Quanwang Li
- Oncology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Fangguyuan Rd, Fengtai District, Beijing, 100078, China
| | - Kaiwen Hu
- Oncology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Fangguyuan Rd, Fengtai District, Beijing, 100078, China.
| |
Collapse
|
23
|
Huynh M, Kempson I, Bezak E, Phillips W. Predictive modeling of hypoxic head and neck cancers during fractionated radiotherapy with gold nanoparticle radiosensitization. Med Phys 2021; 48:3120-3133. [PMID: 33818799 DOI: 10.1002/mp.14872] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 11/05/2022] Open
Abstract
PURPOSE Intrinsic radioresistance and increased proliferation rates in head and neck cancers (HNCs) are associated with negative radiotherapy (RT) treatment responses. The use of gold nanoparticles (AuNPs) as radiosensitizers could enable total radiation dose reduction and lowered radiation toxicity. AuNP radiosensitization may overcome hypoxia-induced radioresistance and treatment-induced accelerated repopulation of cancer cells in HNCs, improving radiotherapy outcomes. METHODS Tumor control was determined by considering individual cancer cell responses in probabilistic computational simulations using HYP-RT software for clinical radiotherapy doses and fractionation schedules along with three different nanoparticle administration schedules. Antagonistic tumor hypoxia and rapid tumor regrowth due to accelerated repopulation of cancers cells were taken into consideration. RESULTS Simulations indicate that tumors that are conventionally uncontrollable can be controlled with AuNP radiosensitization. In simulations where the absence of AuNPs required radiotherapy doses above standard clinical prescriptions, reoccurring AuNP administration allowed for radiation dose reductions below standard clinical dose prescriptions. For example, considering a 2 Gy per fraction radiotherapy schedule, tumor control was achieved with 57.2 ± 5.1 Gy (P = <0.0001) for weekly AuNP administration and 53.0 ± 4.0 Gy (P = <0.0001) for biweekly AuNP administration compared to 69.9 ± 5.8 Gy with no radiosensitization. CONCLUSIONS AuNPs decreased the predicted RT total doses required to achieve tumor control via total stem cell elimination, offering an optimistic prediction and method for which hypoxia-induced and rapidly growing radioresistant tumors are treated more effectively. Outcomes are also shown to be sensitive to the RT schedule with data for hyperfractionated RT indicating the greatest benefits from radiosensitization.
Collapse
Affiliation(s)
- Myxuan Huynh
- Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Eva Bezak
- Cancer Research Institute, University of South Australia, Adelaide, SA, Australia.,Department of Physics, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Wendy Phillips
- Department of Physics, University of Adelaide, North Terrace, Adelaide, SA, Australia.,Department of Medical Physics, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
24
|
Shcherbakov V, Denisov SA, Mostafavi M. Selective Oxidation of Transient Organic Radicals in the Presence of Gold Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:727. [PMID: 33799351 PMCID: PMC7998999 DOI: 10.3390/nano11030727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 11/30/2022]
Abstract
The ability of gold nanoparticles (AuNPs) to catalyze reactions involving radicals is poorly studied. However, AuNPs are used in applications where chemical reactions involving transient radicals occur. Herein, we investigate AuNPs' catalytic effect on 2-propanol oxidation and acetanilide hydroxylation in aqueous solutions under ionizing radiation at room temperature. In both cases, the presence of AuNPs led to selective oxidation of organic radicals, significantly changing the products' composition and ratio. Based on these observations, we stress how AuNPs' catalytic activity can affect the correctness of reactive oxygen species concentration determination utilizing organic dyes. We also provide a discussion on the role of AuNPs' catalytic activity in the radiosensitization effect actively studied for radiotherapy.
Collapse
Affiliation(s)
| | - Sergey A. Denisov
- Institute de Chimie Physique (ICP), CNRS/Université Paris-Saclay, 91405 Orsay, France;
| | - Mehran Mostafavi
- Institute de Chimie Physique (ICP), CNRS/Université Paris-Saclay, 91405 Orsay, France;
| |
Collapse
|
25
|
Huynh M, Kempson I, Bezak E, Phillips W. In silico modeling of cellular probabilistic nanoparticle radiosensitization in head and neck cancers. Nanomedicine (Lond) 2020; 15:2837-2850. [DOI: 10.2217/nnm-2020-0301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: The use of gold nanoparticles (AuNPs) as radiosensitizers may offer a new approach in the treatment of head and neck cancers; minimizing treatment-associated toxicities and improving patient outcomes. AuNPs promote localized dose deposition; permitting improved local control and/or dose reduction. Aim: This work aimed to address the theoretical optimization of radiation doses, fractionation and nanoparticle injection schedules to maximize therapeutic benefits. Materials & methods: Probabilistic nanoparticle sensitization factors were incorporated into the individual cell-based HYP-RT computer model of tumor growth and radiotherapy. Results: Total dose outcomes across all radiation therapy treatment regimens were found to be significantly reduced with the presence of AuNPs, with bi-weekly injections showing the most decrease. Conclusion: Outcomes suggest the need for regular AuNP administration to permit effective radiosensitization.
Collapse
Affiliation(s)
- Myxuan Huynh
- Cancer Research Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, South Australia, Australia
| | - Eva Bezak
- Cancer Research Institute, University of South Australia, Adelaide, South Australia, Australia
- Department of Physics, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Wendy Phillips
- Department of Physics, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
- Department of Medical Physics, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
26
|
Chen Y, Yang J, Fu S, Wu J. Gold Nanoparticles as Radiosensitizers in Cancer Radiotherapy. Int J Nanomedicine 2020; 15:9407-9430. [PMID: 33262595 PMCID: PMC7699443 DOI: 10.2147/ijn.s272902] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022] Open
Abstract
The rapid development of nanotechnology offers a variety of potential therapeutic strategies for cancer treatment. High atomic element nanomaterials are often utilized as radiosensitizers due to their unique photoelectric decay characteristics. Among them, gold nanoparticles (GNPs) are one of the most widely investigated and are considered to be an ideal radiosensitizers for radiotherapy due to their high X-ray absorption and unique physicochemical properties. Over the last few decades, multi-disciplinary studies have focused on the design and optimization of GNPs to achieve greater dosing capability and higher therapeutic effects and highlight potential mechanisms for radiosensitization of GNPs. Although the radiosensitizing potential of GNPs has been widely recognized, its clinical translation still faces many challenges. This review analyses the different roles of GNPs as radiosensitizers in cancer radiotherapy and summarizes recent advances. In addition, the underlying mechanisms of GNP radiosensitization, including physical, chemical and biological mechanisms are discussed, which may provide new directions for the optimization and clinical transformation of next-generation GNPs.
Collapse
Affiliation(s)
- Yao Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Juan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| |
Collapse
|
27
|
Landry C, Morrison A, Ghandi K. Application of muon and other complementary radiation techniques to study interaction of radiation with nanostructures. Radiat Phys Chem Oxf Engl 1993 2020. [DOI: 10.1016/j.radphyschem.2020.108881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
28
|
Samani RK, Tavakoli MB, Maghsoudinia F, Motaghi H, Hejazi SH, Mehrgardi MA. Trastuzumab and folic acid functionalized gold nanoclusters as a dual-targeted radiosensitizer for megavoltage radiation therapy of human breast cancer. Eur J Pharm Sci 2020; 153:105487. [PMID: 32707173 DOI: 10.1016/j.ejps.2020.105487] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/06/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023]
Abstract
In the present study, the effect of functionalized gold nanoclusters (AuNCs) with trastuzumab (Herceptin®) and/or folic acid (FA) as a single and dual-targeted radiosensitizers for the enhancement of megavoltage radiation therapy efficacy was investigated. SK-BR3 breast cancer cells as human epidermal growth factor 2 (HER2) and folate overexpressing cell line and the murine fibroblast (L929) as a control cell line were selected. The cellular uptake was followed using inductively coupled plasma optical emission spectrometry (ICP-OES) that showed AuNCs-FA-HER uptake by SK-BR3 cells was 3 times more than the non-targeted AuNCs after 12 h incubation. MTT and clonogenic assays revealed that the viability and surviving fraction of cancer cells were significantly inhibited by treating with all AuNCs under radiation compared to treating with radiation alone. However, these effects in the dual-targeted AuNCs group (AuNCs-FA-HER) was significantly greater than non-targeted and single-targeted AuNCs groups. Also, apoptosis was evaluated using an Annexin V-FITC/propidium iodide (PI) kit in flow cytometry. All AuNCs, in combination with 4 Gy of photon beam, induced more apoptosis. By fitting the survival fraction data on the linear-quadratic model, the sensitization enhancement factor (SER) of AuNCs, AuNCs-FA, AuNCs-HER, and AuNCs-FA-HER, were obtained 1.17, 1.32, 1.48 and 1.77, respectively. SER for AuNCs-FA-HER was significantly higher than that non-targeted and single-targeted AuNCs (p-value < 0.05) that can be attributed to more internalization in the cancer cells. It was concluded that functionalized AuNCs with both folic acid and Herceptin could represent a promising strategy for increased cellular internalization that improved radiation therapy efficiency in SK-BR3 breast cancer cells.
Collapse
Affiliation(s)
- Roghayeh Kamran Samani
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Mohamad Bagher Tavakoli
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran.
| | - Fatemeh Maghsoudinia
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Hasan Motaghi
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud A Mehrgardi
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran.
| |
Collapse
|
29
|
Kempson I. Mechanisms of nanoparticle radiosensitization. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1656. [PMID: 32686321 DOI: 10.1002/wnan.1656] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023]
Abstract
Metal-based nanoparticles applied to potentiating the effects of radiotherapy have drawn significant attention from the research community and are now available clinically. By improving our mechanistic understanding, nanoparticles are likely to evolve to provide very significant improvements in radiotherapy outcomes with only incremental increase in cost. This review critically assesses the inconsistent observations surrounding physical, physicochemical, chemical and biological mechanisms of radiosensitization. In doing so, a number of needs are identified for continuing research and are highlighted. The large degree of variability from one nanoparticle to another emphasizes that it is a mistake to generalize nanoparticle radiosensitizer mechanisms. Nanoparticle formulations should be considered in an analogous way as pharmacological agents and as a broad class of therapeutic agents, needing to be considered with a high degree of individuality with respect to their interactions and ultimate impact on radiobiological response. In the same way that no universal anti-cancer drug exists, it is unlikely that a single nanoparticle formulation will lead to the best therapeutic outcomes for all cancers. The high degree of complexity and variability in mechanistic action provides notable opportunities for nanoparticle formulations to be optimized for specific indications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| |
Collapse
|
30
|
Popescu RC, Andronescu E, Vasile BS. Recent Advances in Magnetite Nanoparticle Functionalization for Nanomedicine. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1791. [PMID: 31888236 PMCID: PMC6956201 DOI: 10.3390/nano9121791] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Functionalization of nanomaterials can enhance and modulate their properties and behaviour, enabling characteristics suitable for medical applications. Magnetite (Fe3O4) nanoparticles are one of the most popular types of nanomaterials used in this field, and many technologies being already translated in clinical practice. This article makes a summary of the surface modification and functionalization approaches presented lately in the scientific literature for improving or modulating magnetite nanoparticles for their applications in nanomedicine.
Collapse
Affiliation(s)
- Roxana Cristina Popescu
- National Research Center for Micro and Nanomaterials, Department of Science and Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 060042 Bucharest, Romania; (R.C.P.); (E.A.)
- Department of Life and Environmental Physics, “Horia Hulubei” National Institute for Physics and Nuclear Engineering, 077125 Magurele, Romania
| | - Ecaterina Andronescu
- National Research Center for Micro and Nanomaterials, Department of Science and Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 060042 Bucharest, Romania; (R.C.P.); (E.A.)
| | - Bogdan Stefan Vasile
- National Research Center for Micro and Nanomaterials, Department of Science and Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 060042 Bucharest, Romania; (R.C.P.); (E.A.)
| |
Collapse
|
31
|
Li F, Li Z, Jin X, Liu Y, Li P, Shen Z, Wu A, Zheng X, Chen W, Li Q. Radiosensitizing Effect of Gadolinium Oxide Nanocrystals in NSCLC Cells Under Carbon Ion Irradiation. NANOSCALE RESEARCH LETTERS 2019; 14:328. [PMID: 31637533 PMCID: PMC6803611 DOI: 10.1186/s11671-019-3152-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/09/2019] [Indexed: 05/17/2023]
Abstract
Gadolinium-based nanomaterials can not only serve as contrast agents but also contribute to sensitization in the radiotherapy of cancers. Among radiotherapies, carbon ion irradiation is considered one of the superior approaches with unique physical and biological advantages. However, only a few metallic nanoparticles have been used to improve carbon ion irradiation. In this study, gadolinium oxide nanocrystals (GONs) were synthesized using a polyol method to decipher the radiosensitizing mechanisms in non-small cell lung cancer (NSCLC) cell lines irradiated by carbon ions. The sensitizer enhancement ratio at the 10% survival level was correlated with the concentration of Gd in NSCLC cells. GONs elicited an increase in hydroxyl radical production in a concentration-dependent manner, and the yield of reactive oxygen species increased obviously in irradiated cells, which led to DNA damage and cell cycle arrest. Apoptosis and cytostatic autophagy were also significantly induced by GONs under carbon ion irradiation. The GONs may serve as an effective theranostic material in carbon ion radiotherapy for NSCLC.
Collapse
Affiliation(s)
- Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zihou Li
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, 315201 Zhejiang China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Zheyu Shen
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, 315201 Zhejiang China
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, 315201 Zhejiang China
| | - Xiaogang Zheng
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| |
Collapse
|
32
|
Shanei A, Akbari-Zadeh H. Investigating the Sonodynamic-Radiosensitivity Effect of Gold Nanoparticles on HeLa Cervical Cancer Cells. J Korean Med Sci 2019; 34:e243. [PMID: 31559711 PMCID: PMC6763396 DOI: 10.3346/jkms.2019.34.e243] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In this article, we estimated the combined effect of radiotherapy (RT) with ultrasound (US) wave and the ability of gold nanoparticles (GNPs) to improve their combined therapeutic effects. METHODS At first, HeLa cells received the various treatment modalities: RT (6 MV; 0.5, 1, and 2 Gy), US irradiation (1 MHz; 0.5, 1, and 1.5 W/cm², 1 minute), and RT+US. Afterwards, the enhanced effect of US on RT was evaluated. Then, the effect of the synthesized GNPs at different concentrations (0.2, 1, and 5 μg/mL, 24 hours) was evaluated to assess the effect on HeLa cells combined with RT+US. Cell survival rates in the different treatment groups at 24, 48, and 72 hours post-treatment were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and trypan blue assays. RESULTS Our results show US irradiation could enhance the effect of RT at the same radiation dose and could be utilized as a sensitizer agent for RT. Moreover, our findings indicate RT+US in combination with different nanoparticle concentrations could enhance the effect of RT+US so that they can improve the treatment results up to 9.93 times and act as sonodynamic-radiosensitivity. These results also indicate that the combination of RT with US along with GNPs has synergistic effects compared to RT or US alone. Cell survival results show that combining the low US waves (1.5 W/cm²), GNPs (5 μ/mL), and X-rays (2 Gy) increase the cytotoxicity on HeLa cell up to 95.8%. CONCLUSION We concluded that GNPs could act as a good sensitizing agent in RT+US irradiation and could result in the synergistic effects.
Collapse
Affiliation(s)
- Ahmad Shanei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hadi Akbari-Zadeh
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
33
|
Rudek B, McNamara A, Ramos-Méndez J, Byrne H, Kuncic Z, Schuemann J. Radio-enhancement by gold nanoparticles and their impact on water radiolysis for x-ray, proton and carbon-ion beams. Phys Med Biol 2019; 64:175005. [PMID: 31295730 PMCID: PMC11222020 DOI: 10.1088/1361-6560/ab314c] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Gold nanoparticle (GNP) radio-enhancement is a promising technique to increase the dose deposition in a tumor while sparing neighboring healthy tissue. Previous experimental studies showed effects on cell survival and tumor control for keV x-rays but surprisingly also for MV-photons, proton and carbon-ion beams. In a systematic study, we use the Monte Carlo simulation tool TOPAS-nBio to model the GNP radio-enhancement within a cell as a function of GNP concentration, size and clustering for a wide range of energies for photons, protons and, for the first time, carbon-ions. Moreover, we include water radiolysis, which has been recognized as a major pathway of GNP mediated radio-enhancement. At a GNP concentration of 0.5% and a GNP diameter of 10 nm, the dose enhancement ratio was highest for 50 keV x-rays (1.36) and decreased in the orthovoltage (1.04 at 250 keV) and megavoltage range (1.01 at 1 MeV). The dose enhancement linearly increased with GNP concentration and decreased with GNP size and degree of clustering for all radiation modalities. While the highest physical dose enhancement at 5% concentrations was only 1.003 for 10 MeV protons and 1.004 for 100 MeV carbon-ions, we find the number of hydroxyl ([Formula: see text]) altered by 23% and 3% after 1 [Formula: see text]s at low, clinically-relevant concentrations. For the same concentration and proton-impact, the G-value is most sensitive to the nanoparticle size with 46 times more radical interactions at GNPs for 2 nm than for 50 nm GNP diameter within 1 [Formula: see text]s. Nanoparticle clustering was found to decrease the number of interactions at GNPs, e.g. for a cluster of 25 GNPs by a factor of 3.4. The changes in G-value correlate to the average distance between the chemical species and the GNPs. While the radiochemistry of GNP-loaded water has yet to be fully understood, this work offers a first relative quantification of radiolysis products for a broad parameter-set.
Collapse
Affiliation(s)
- Benedikt Rudek
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, MA, United States of America. Department of Physics, Boston University, Boston, Massachusetts, MA, United States of America. Department of Ionizing Radiation, Physikalisch-Technische Bundesanstalt, Braunschweig, Germany. Author to whom any correspondence should be addressed
| | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Turnbull T, Douglass M, Williamson NH, Howard D, Bhardwaj R, Lawrence M, Paterson DJ, Bezak E, Thierry B, Kempson IM. Cross-Correlative Single-Cell Analysis Reveals Biological Mechanisms of Nanoparticle Radiosensitization. ACS NANO 2019; 13:5077-5090. [PMID: 31009200 PMCID: PMC6546286 DOI: 10.1021/acsnano.8b07982] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Nanoparticle radiosensitization has been demonstrated well to enhance the effects of radiotherapy, motivate the improvement of therapeutic ratios, and decrease morbidity in cancer treatment. A significant challenge exists in optimizing formulations and translation due to insufficient knowledge of the associated mechanisms, which have historically been limited to physical concepts. Here, we investigated a concept for the role of biological mechanisms. The mere presence of gold nanoparticles led to a down-regulation of thymidylate synthase, important for DNA damage repair in the radioresistant S-phase cells. By developing a cross-correlative methodology to reveal probabilistic gold nanoparticle uptake by cell sub-populations and the associated sensitization as a function of the uptake, a number of revealing observations have been achieved. Surprisingly, for low numbers of nanoparticles, a desensitization action was observed. Sensitization was discovered to preferentially impact S-phase cells, in which impairment of the DNA damage response by the homologous recombination pathway dominates. This small but radioresistant cell population correlates with much greater proliferative ability. Thus, a paradigm is presented whereby enhanced DNA damage is not necessarily due to an increase in the number of DNA double-strand breaks (DSBs) created but can be from a nanoparticle-induced impairment of the damage response by down-regulating repair proteins such as thymidylate synthase.
Collapse
Affiliation(s)
- Tyron Turnbull
- Future Industries Institute , University of South Australia , Mawson Lakes , South Australia 5095 , Australia
| | - Michael Douglass
- Department of Medical Physics , Royal Adelaide Hospital , Adelaide , South Australia 5000 , Australia
- Department of Physics , University of Adelaide , Adelaide , South Australia 5005 , Australia
| | - Nathan H Williamson
- Future Industries Institute , University of South Australia , Mawson Lakes , South Australia 5095 , Australia
- Section on Quantitative Imaging and Tissue Sciences, NICHD , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Douglas Howard
- Future Industries Institute , University of South Australia , Mawson Lakes , South Australia 5095 , Australia
| | - Richa Bhardwaj
- Future Industries Institute , University of South Australia , Mawson Lakes , South Australia 5095 , Australia
| | - Mark Lawrence
- Department of Critical Care Medicine , Flinders University , Adelaide , South Australia 5042 , Australia
| | | | - Eva Bezak
- Department of Physics , University of Adelaide , Adelaide , South Australia 5005 , Australia
| | - Benjamin Thierry
- Future Industries Institute , University of South Australia , Mawson Lakes , South Australia 5095 , Australia
| | - Ivan M Kempson
- Future Industries Institute , University of South Australia , Mawson Lakes , South Australia 5095 , Australia
| |
Collapse
|
36
|
Li F, Li Z, Jin X, Liu Y, Zhang P, Li P, Shen Z, Wu A, Chen W, Li Q. Ultra-small gadolinium oxide nanocrystal sensitization of non-small-cell lung cancer cells toward X-ray irradiation by promoting cytostatic autophagy. Int J Nanomedicine 2019; 14:2415-2431. [PMID: 31040665 PMCID: PMC6455003 DOI: 10.2147/ijn.s193676] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gadolinium-based nanoparticles (GdNPs) have been used as theranostic sensitizers in clinical radiotherapy studies; however, the biomechanisms underlying the radio-sensitizing effects of GdNPs have yet to be determined. In this study, ultra-small gadolinium oxide nanocrystals (GONs) were employed to investigate their radiosensitizing effects and biological mechanisms in non-small-cell lung cancer (NSCLC) cells under X-ray irradiation. METHOD AND MATERIALS GONs were synthesized using polyol method. Hydroxyl radical production, oxidative stress, and clonogenic survival after X-ray irradiation were used to evaluate the radiosensitizing effects of GONs. DNA double-strand breakage, cell cycle phase, and apoptosis and autophagy incidences were investigated in vitro to determine the radiosensitizing biomechanism of GONs under X-ray irradiation. RESULTS GONs induced hydroxyl radical production and oxidative stress in a dose- and concentration-dependent manner in NSCLC cells after X-ray irradiation. The sensitizer enhancement ratios of GONs ranged between 19.3% and 26.3% for the NSCLC cells under investigation with a 10% survival rate compared with that of the cells treated with irradiation alone. Addition of 3-methyladenine to the cell medium decreased the incidence rate of autophagy and increased cell survival, supporting the idea that the GONs promoted cytostatic autophagy in NSCLC cells under X-ray irradiation. CONCLUSION This study examined the biological mechanisms underlying the radiosensitizing effects of GONs on NSCLC cells and presented the first evidence for the radiosensitizing effects of GONs via activation of cytostatic autophagy pathway following X-ray irradiation.
Collapse
Affiliation(s)
- Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zihou Li
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Zheyu Shen
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| |
Collapse
|
37
|
Radio-sensitization efficacy of gold nanoparticles in inhalational nanomedicine and the adverse effect of nano-detachment due to coating inactivation. Phys Med 2019; 60:7-13. [PMID: 31000089 DOI: 10.1016/j.ejmp.2019.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 01/21/2019] [Accepted: 02/15/2019] [Indexed: 12/25/2022] Open
Abstract
Gold nanoparticles (GNPs) are an emerging area of interest in radiation therapy due to their unique radio-sensitizing properties. In the literature, the enhancing capability of GNPs is usually quantified using the metric dose enhancement ratio (DER). Traditionally, the focus of the vast majority of studies has always been on intravenous administration of GNPs. However, recent work showed the potential of using GNP inhalation, rather than intravenous injection, to enhance the dose to the lung. Yet, some of these studies are employing simplistic analytical methods to calculate DER and, thus far, there are no detailed computations of the enhancement profiles therein. Moreover, the coating on the GNP surface can be adversely affected by the large gradient of the radiation dose in the immediate vicinity of GNPs, leading to the rupture of ligands and detachment of GNPs from the surface of the membrane, and hence the loss of its efficacy. In this study, a next-generation deterministic code was used to resolve the DER profile at the interface between the septum, air, and surface of GNPs when they are attached and detached. The results show that the large values of DER in conjunction with the developed hot spots are very effective in lung treatment; on the other hand, coating rupture can lead to significant reduction in DER that may reach 64%. Thus, GNPs can be beneficial in inhalational medicine to treat lung cancer, provided that more comprehensive studies on the characteristics of the coating are addressed to maximize the radio-therapeutic benefit of GNPs.
Collapse
|
38
|
Hespeels F, Lucas S, Tabarrant T, Scifoni E, Kraemer M, Chêne G, Strivay D, Tran HN, Heuskin AC. Experimental measurements validate the use of the binary encounter approximation model to accurately compute proton induced dose and radiolysis enhancement from gold nanoparticles. Phys Med Biol 2019; 64:065014. [PMID: 30731439 DOI: 10.1088/1361-6560/ab0516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In protontherapy, it has been suggested that nanoparticles of high-Z material like gold (GNP) could be used as radiosensitizers. The origin of this enhancement phenomenon for proton radiation is not yet well understood and additional mechanistic insights are required. Previous works have highlighted the good capabilities of TRAX to reproduce secondary electron emission from gold material. Therefore, TRAX cross sections obtained with the binary encounter approximation (BEA) model for proton ionization were implemented within Geant4 for gold material. Based on the TRAX cross sections, improved Geant4 simulations have been developed to investigate the energy deposition and radical species production around a spherical gold nanoparticle (5 and 10 nm in diameter) placed in a water volume during proton irradiation. Simulations were performed for incident 2 MeV proton. The dose enhancement factor and the radiolysis enhancement factor were quantified. Results obtained with the BEA model were compared with results obtained with condensed-history models. Experimental irradiation of 200 nm gold films were performed to validate the secondary electron emission reproduction capabilities of physical models used in Monte Carlo (MC) simulations. TRAX simulations reproduced the experimental backscattered electron energy spectrum from gold film with better agreement than Geant4. Results on gold film obtained with the BEA model enabled to estimate the electron emission from GNPs. Results obtained in our study tend to support that the use of the BEA discrete model leads to a significant increase of the dose in the near vicinity of GNPs (<20 nm), while condensed history models used in Geant4 seem to overestimate the dose and the number of chemical species for increasing distances from the GNP. Based on discrete BEA model results, no enhancement effect due to secondary electron emitted from the GNP is expected if the GNP is not in close proximity to key cellular functional elements (DNA, mitochondria…).
Collapse
Affiliation(s)
- F Hespeels
- University of Namur, PMR, 61 rue de Bruxelles, 5000 Namur, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Penninckx S, Heuskin AC, Michiels C, Lucas S. Thioredoxin Reductase Activity Predicts Gold Nanoparticle Radiosensitization Effect. NANOMATERIALS 2019; 9:nano9020295. [PMID: 30791480 PMCID: PMC6409576 DOI: 10.3390/nano9020295] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 02/06/2023]
Abstract
Gold nanoparticles (GNPs) have been shown to be effective contrast agents for imaging and emerge as powerful radiosensitizers, constituting a promising theranostic agent for cancer. Although the radiosensitization effect was initially attributed to a physical mechanism, an increasing number of studies challenge this mechanistic hypothesis and evidence the importance of oxidative stress in this process. This work evidences the central role played by thioredoxin reductase (TrxR) in the GNP-induced radiosensitization. A cell type-dependent reduction in TrxR activity was measured in five different cell lines incubated with GNPs leading to differences in cell response to X-ray irradiation. Correlation analyses demonstrated that GNP uptake and TrxR activity inhibition are associated to a GNP radiosensitization effect. Finally, Kaplan-Meier analyses suggested that high TrxR expression is correlated to low patient survival in four different types of cancer. Altogether, these results enable a better understanding of the GNP radiosensitization mechanism, which remains a mandatory step towards further use in clinic. Moreover, they highlight the potential application of this new treatment in a personalized medicine context.
Collapse
Affiliation(s)
- Sébastien Penninckx
- Research Center for the Physics of Matter and Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium.
| | - Anne-Catherine Heuskin
- Research Center for the Physics of Matter and Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium.
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium.
| | - Stéphane Lucas
- Research Center for the Physics of Matter and Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium.
| |
Collapse
|
40
|
Penninckx S, Heuskin AC, Michiels C, Lucas S. The role of thioredoxin reductase in gold nanoparticle radiosensitization effects. Nanomedicine (Lond) 2018; 13:2917-2937. [DOI: 10.2217/nnm-2018-0171] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To identify new mechanisms responsible for the radiosensitization effect of gold nanoparticles (GNPs). Materials & methods: A549 lung carcinoma cells were incubated with 10-nm GNPs during 6 or 24 h before to be exposed to 25 keV/μm protons or 225 kV x-rays. Results: GNP incubation led to a time-dependent mitochondria membrane depolarization, oxidative stress and to x-ray and proton radiosensitization. Moreover, a marked inhibition of thioredoxin reductase was observed. Irradiation of cells invalidated for thioredoxin reductase evidenced a radiosensitization effect, suggesting that this enzyme is a potential GNP target. Conclusion: We suggest that GNPs play a radiosensitizer role by weakening detoxification systems. Altogether, these results open up promising novel strategies for the development of nanotechnologies associated to radiotherapy.
Collapse
Affiliation(s)
- Sébastien Penninckx
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles, 61, B-5000 Namur, Belgium
| | - Anne-Catherine Heuskin
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles, 61, B-5000 Namur, Belgium
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Stéphane Lucas
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles, 61, B-5000 Namur, Belgium
| |
Collapse
|
41
|
Lee Y, Okayasu R. Strategies to Enhance Radiosensitivity to Heavy Ion Radiation Therapy. Int J Part Ther 2018; 5:114-121. [DOI: 10.14338/ijpt-18-00014.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/16/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Younghyun Lee
- Center for Radiological Research, Columbia University Medical Center, New York, NY, USA
| | - Ryuichi Okayasu
- Department of Basic Medical Sciences for Radiation Damages, National Institutes for Quantum and Radiological Science and Technology/National Institute of Radiological Sciences, Japan
| |
Collapse
|
42
|
Multifunctional Chitosan-Capped Gold Nanoparticles for enhanced cancer chemo-radiotherapy: An invitro study. Phys Med 2018; 48:76-83. [DOI: 10.1016/j.ejmp.2018.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/02/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022] Open
|
43
|
Peukert D, Kempson I, Douglass M, Bezak E. Metallic nanoparticle radiosensitisation of ion radiotherapy: A review. Phys Med 2018; 47:121-128. [DOI: 10.1016/j.ejmp.2018.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/12/2018] [Accepted: 03/05/2018] [Indexed: 01/19/2023] Open
|
44
|
Liu Y, Zhang P, Li F, Jin X, Li J, Chen W, Li Q. Metal-based NanoEnhancers for Future Radiotherapy: Radiosensitizing and Synergistic Effects on Tumor Cells. Theranostics 2018; 8:1824-1849. [PMID: 29556359 PMCID: PMC5858503 DOI: 10.7150/thno.22172] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is one of the major therapeutic strategies for cancer treatment. In the past decade, there has been growing interest in using high Z (atomic number) elements (materials) as radiosensitizers. New strategies in nanomedicine could help to improve cancer diagnosis and therapy at cellular and molecular levels. Metal-based nanoparticles usually exhibit chemical inertness in cellular and subcellular systems and may play a role in radiosensitization and synergistic cell-killing effects for radiation therapy. This review summarizes the efficacy of metal-based NanoEnhancers against cancers in both in vitro and in vivo systems for a range of ionizing radiations including gamma-rays, X-rays, and charged particles. The potential of translating preclinical studies on metal-based nanoparticles-enhanced radiation therapy into clinical practice is also discussed using examples of several metal-based NanoEnhancers (such as CYT-6091, AGuIX, and NBTXR3). Also, a few general examples of theranostic multimetallic nanocomposites are presented, and the related biological mechanisms are discussed.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| | - Jin Li
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| |
Collapse
|
45
|
Kuncic Z, Lacombe S. Nanoparticle radio-enhancement: principles, progress and application to cancer treatment. Phys Med Biol 2018; 63:02TR01. [PMID: 29125831 DOI: 10.1088/1361-6560/aa99ce] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Enhancement of radiation effects by high-atomic number nanoparticles (NPs) has been increasingly studied for its potential to improve radiotherapeutic efficacy. The underlying principle of NP radio-enhancement is the potential to release copious electrons into a nanoscale volume, thereby amplifying radiation-induced biological damage. While the vast majority of studies to date have focused on gold nanoparticles with photon radiation, an increasing number of experimental, theoretical and simulation studies have explored opportunities offered by other NPs (e.g. gadolinium, platinum, iron oxide, hafnium) and other therapeutic radiation sources such as ion beams. It is thus of interest to the research community to consolidate findings from the different studies and summarise progress to date, as well as to identify strategies that offer promising opportunities for clinical translation. This is the purpose of this Topical Review.
Collapse
Affiliation(s)
- Zdenka Kuncic
- School of Physics and Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | | |
Collapse
|
46
|
Wang H, Mu X, He H, Zhang XD. Cancer Radiosensitizers. Trends Pharmacol Sci 2017; 39:24-48. [PMID: 29224916 DOI: 10.1016/j.tips.2017.11.003] [Citation(s) in RCA: 362] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
Abstract
Radiotherapy (RT) is a mainstay treatment for many types of cancer, although it is still a large challenge to enhance radiation damage to tumor tissue and reduce side effects to healthy tissue. Radiosensitizers are promising agents that enhance injury to tumor tissue by accelerating DNA damage and producing free radicals. Several strategies have been exploited to develop highly effective and low-toxicity radiosensitizers. In this review, we highlight recent progress on radiosensitizers, including small molecules, macromolecules, and nanomaterials. First, small molecules are reviewed based on free radicals, pseudosubstrates, and other mechanisms. Second, nanomaterials, such as nanometallic materials, especially gold-based materials that have flexible surface engineering and favorable kinetic properties, have emerged as promising radiosensitizers. Finally, emerging macromolecules have shown significant advantages in RT because these molecules can be combined with biological therapy as well as drug delivery. Further research on the mechanisms of radioresistance and multidisciplinary approaches will accelerate the development of radiosensitizers.
Collapse
Affiliation(s)
- Hao Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Number 238, Baidi Road, Tianjin 300192, China; These authors have contributed equally
| | - Xiaoyu Mu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; These authors have contributed equally
| | - Hua He
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, China
| | - 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 Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, China.
| |
Collapse
|
47
|
Lacombe S, Porcel E, Scifoni E. Particle therapy and nanomedicine: state of art and research perspectives. Cancer Nanotechnol 2017; 8:9. [PMID: 29213338 PMCID: PMC5698390 DOI: 10.1186/s12645-017-0029-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
Cancer radiation therapy with charged particle beams, called particle therapy, is a new therapeutic treatment presenting major advantages when compared to conventional radiotherapy. Because ions have specific ballistic properties and a higher biological effectiveness, they are superior to x-rays. Numerous medical centres are starting in the world using mostly protons but also carbon ions as medical beams. Several investigations are attempting to reduce the cost/benefit ratio and enlarge the range of therapeutic indications. A major limitation of particle therapy is the presence of low but significant damage induced in healthy tissues located at the entrance of the ion track prior to reaching the tumour. It is thus a major challenge to improve the targeting of the tumours, concentrating radiation effects in the malignance. A novel strategy, based on the addition of nanoparticles targeting the tumour, was suggested over a decade ago to improve the performance of conventional photon therapy. Recently, similar developments have emerged for particle therapy and the amount of research is now exploding. In this paper, we review the experimental results, as well as theoretical and simulation studies that shed light in the promising outcomes of this strategy and in the underpinning mechanisms. Several experiments provide consistent evidence of significant enhancement of ion radiation effects in the presence of nanoparticles. In view of implementing this strategy for cancer treatment, simulation studies have begun to establish the rationale and the specificity of this effect. In addition, these studies will help to outline a list of possible mechanisms and to predict the impact of ion beams and nanoparticle characteristics. Many questions remain unsolved, but the findings of these first studies are encouraging and open new challenges. After summarizing the main results in the field, we propose a roadmap to pursue future research with the aim to strengthen the potential interplay between particle therapy and nanomedicine.
Collapse
Affiliation(s)
- Sandrine Lacombe
- Institut des Sciences Moléculaires d'Orsay (UMR 8214) Bât 351, University Paris Saclay, University of Paris Sud, CNRS, 91405 Orsay Cedex, France
| | - Erika Porcel
- Institut des Sciences Moléculaires d'Orsay (UMR 8214) Bât 351, University Paris Saclay, University of Paris Sud, CNRS, 91405 Orsay Cedex, France
| | - Emanuele Scifoni
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany.,TIFPA-INFN, Trento Institute for Fundamental Physics and Applications, University of Trento, 38121 Trento, Italy
| |
Collapse
|
48
|
Smith CL, Best SP, Gagliardi F, Tominaga T, Geso M. The effects of gold nanoparticles concentrations and beam quality/LET on dose enhancement when irradiated with X-rays and protons using alanine/EPR dosimetry. RADIAT MEAS 2017. [DOI: 10.1016/j.radmeas.2017.01.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Jung S, Sung W, Ye SJ. Pinhole X-ray fluorescence imaging of gadolinium and gold nanoparticles using polychromatic X-rays: a Monte Carlo study. Int J Nanomedicine 2017; 12:5805-5817. [PMID: 28860750 PMCID: PMC5565259 DOI: 10.2147/ijn.s141185] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
This work aims to develop a Monte Carlo (MC) model for pinhole K-shell X-ray fluorescence (XRF) imaging of metal nanoparticles using polychromatic X-rays. The MC model consisted of two-dimensional (2D) position-sensitive detectors and fan-beam X-rays used to stimulate the emission of XRF photons from gadolinium (Gd) or gold (Au) nanoparticles. Four cylindrical columns containing different concentrations of nanoparticles ranging from 0.01% to 0.09% by weight (wt%) were placed in a 5 cm diameter cylindrical water phantom. The images of the columns had detectable contrast-to-noise ratios (CNRs) of 5.7 and 4.3 for 0.01 wt% Gd and for 0.03 wt% Au, respectively. Higher concentrations of nanoparticles yielded higher CNR. For 1×1011 incident particles, the radiation dose to the phantom was 19.9 mGy for 110 kVp X-rays (Gd imaging) and 26.1 mGy for 140 kVp X-rays (Au imaging). The MC model of a pinhole XRF can acquire direct 2D slice images of the object without image reconstruction. The MC model demonstrated that the pinhole XRF imaging system could be a potential bioimaging modality for nanomedicine.
Collapse
Affiliation(s)
- Seongmoon Jung
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Wonmo Sung
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sung-Joon Ye
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea
| |
Collapse
|
50
|
Wozny AS, Aloy MT, Alphonse G, Magné N, Janier M, Tillement O, Lux F, Beuve M, Rodriguez-Lafrasse C. Gadolinium-based nanoparticles as sensitizing agents to carbon ions in head and neck tumor cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2655-2660. [PMID: 28779947 DOI: 10.1016/j.nano.2017.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/13/2017] [Accepted: 07/24/2017] [Indexed: 12/31/2022]
Abstract
Hadrontherapy presents the major advantage of improving tumor sterilization while sparing surrounding healthy tissues because of the particular ballistic (Bragg peak) of carbon ions. However, its efficacy is still limited in the most resistant cancers, such as grade III-IV head and neck squamous cell carcinoma (HNSCC), in which the association of carbon ions with gadolinium-based nanoparticles (AGuIX®) could be used as a Trojan horse. We report for the first time the radioenhancing effect of AGuIX® when combined with carbon ion irradiation in human tumor cells. An increase in relative biological effectiveness (1.7) in three HNSCC cell lines (SQ20B, FaDu, and Cal33) was associated with a significant reduction in the radiation dose needed for killing cells. Radiosensitization goes through a higher number of unrepaired DNA double-strand breaks. These results underline the strong potential of AGuIX® in sensitizing aggressive tumors to hadrontherapy and, therefore, improving local control while lowering acute/late toxicity.
Collapse
Affiliation(s)
- Anne-Sophie Wozny
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Marie-Thérèse Aloy
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France
| | - Gersende Alphonse
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Nicolas Magné
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Département de Radiothérapie, Institut de Cancérologie de la Loire Lucien Neuwirth, St Priest en Jarez, France
| | - Marc Janier
- Univ Lyon, Université Lyon1, CNRS, LAGEP UMR 5007, IMTHERNAT, Hôpital Edouard Herriot, Lyon, France
| | - Olivier Tillement
- Univ Lyon, Université Lyon 1, Institut Lumière Matière, UMR 5306 CNRS, Villeurbanne cedex, France
| | - François Lux
- Univ Lyon, Université Lyon 1, Institut Lumière Matière, UMR 5306 CNRS, Villeurbanne cedex, France
| | - Michael Beuve
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, PHABIO, Villeurbanne, France
| | - Claire Rodriguez-Lafrasse
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France.
| |
Collapse
|