1
|
Cammarata A, Marino J, Atia MN, Durán H, Glisoni RJ. Novel doxycycline gold nanoparticles via green synthesis using PEO-PPO block copolymers for enhanced radiosensitization of melanoma. Biomater Sci 2025. [PMID: 40261332 DOI: 10.1039/d5bm00253b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
This study focuses on a green and sustainable nanoplatform for the delivery of therapeutic agents, based on gold nanoparticles (AuNPs) synthesized using PEO-PPO block copolymers (F127, F68, P85, and their F127:P85 combination) as dual-function reducing and stabilizing agents. This eco-friendly approach eliminates the need for toxic chemical reductants, adheres to green chemistry principles, and yields highly stable, biocompatible nanosystems. The resulting polymer-stabilized AuNPs were associated with doxycycline (DOXY), a mitochondrial biogenesis inhibitor with radiosensitizing properties, and characterized using UV-Vis spectroscopy, dynamic light scattering (DLS), transmission electron microscopy (TEM), and X-ray fluorescence (XRF). The nanoparticles exhibited high colloidal stability, with tunable hydrodynamic diameters modulated by the copolymer composition. In vitro studies on A-375 and IIB-MEL-J melanoma cell lines revealed that DOXY-associated AuNPs, combined with gamma radiation (2 Gy, 137Cs), significantly enhanced radiosensitivity, reducing both cell viability and clonogenic survival. The physicochemical features of the nanosystems, particularly particle size and surface composition, influenced cellular uptake and therapeutic response. Notably, AuNPs stabilized with F127:P85 copolymer combination (∼19 nm) outperformed those with F127 (∼30 nm), despite displaying slightly higher polydispersity. Compared to Turkevich AuNPs, our copolymer-coated nanosystems demonstrated superior colloidal stability and cellular internalization. These findings highlight the potential of green-synthesized AuNPs as multifunctional, biocompatible platforms for therapeutic delivery, supporting the development of effective and environmentally responsible multimodal cancer therapies. Moreover, the simplicity, scalability, and cost-effectiveness of the synthesis process support its potential for future translational applications.
Collapse
Affiliation(s)
- Agostina Cammarata
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Biotecnología, Junín 956, C1113AAD Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Nanobiotecnología (NANOBIOTEC), Buenos Aires, Argentina.
| | - Julieta Marino
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Mariel N Atia
- Comisión Nacional de Energía Atómica (CNEA), Gerencia de Investigación y Aplicaciones, Subgerencia de Tecnología y Aplicaciones de Aceleradores, San Martín, Buenos Aires, Argentina
- Instituto de Nanociencia y Nanotecnología (INN-CNEA-CONICET), San Martín, Buenos Aires, Argentina
| | - Hebe Durán
- Comisión Nacional de Energía Atómica (CNEA), Gerencia de Investigación y Aplicaciones, Subgerencia de Tecnología y Aplicaciones de Aceleradores, San Martín, Buenos Aires, Argentina
- Instituto de Nanociencia y Nanotecnología (INN-CNEA-CONICET), San Martín, Buenos Aires, Argentina
- Universidad Nacional de San Martín, Escuela de Ciencia y Tecnología, San Martín, Buenos Aires, Argentina
| | - Romina J Glisoni
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Biotecnología, Junín 956, C1113AAD Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Nanobiotecnología (NANOBIOTEC), Buenos Aires, Argentina.
| |
Collapse
|
2
|
Jackson N, Cecchi D, Beckham W, Chithrani DB. Application of High-Z Nanoparticles to Enhance Current Radiotherapy Treatment. Molecules 2024; 29:2438. [PMID: 38893315 PMCID: PMC11173748 DOI: 10.3390/molecules29112438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Radiotherapy is an essential component of the treatment regimens for many cancer patients. Despite recent technological advancements to improve dose delivery techniques, the dose escalation required to enhance tumor control is limited due to the inevitable toxicity to the surrounding healthy tissue. Therefore, the local enhancement of dosing in tumor sites can provide the necessary means to improve the treatment modality. In recent years, the emergence of nanotechnology has facilitated a unique opportunity to increase the efficacy of radiotherapy treatment. The application of high-atomic-number (Z) nanoparticles (NPs) can augment the effects of radiotherapy by increasing the sensitivity of cells to radiation. High-Z NPs can inherently act as radiosensitizers as well as serve as targeted delivery vehicles for radiosensitizing agents. In this work, the therapeutic benefits of high-Z NPs as radiosensitizers, such as their tumor-targeting capabilities and their mechanisms of sensitization, are discussed. Preclinical data supporting their application in radiotherapy treatment as well as the status of their clinical translation will be presented.
Collapse
Affiliation(s)
- Nolan Jackson
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Daniel Cecchi
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Wayne Beckham
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada
- British Columbia Cancer-Victoria, Victoria, BC V8R 6V5, Canada
| | - Devika B. Chithrani
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada
- Centre for Advanced Materials and Related Technologies, Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- Department of Computer Science, Mathematics, Physics and Statistics, Okanagan Campus, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
3
|
Nandakumar A, Tang H, Andrikopoulos N, Quinn JF, Ding F, Ke PC, Li Y. Controlling nanoparticle-induced endothelial leakiness with the protein corona. NANOSCALE 2024; 16:9348-9360. [PMID: 38651870 PMCID: PMC11098680 DOI: 10.1039/d4nr01311e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Understanding nanoparticle-cell interaction is essential for advancing research in nanomedicine and nanotoxicology. Apart from the transcytotic pathway mediated by cellular recognition and energetics, nanoparticles (including nanomedicines) may harness the paracellular route for their transport by inducing endothelial leakiness at cadherin junctions. This phenomenon, termed as NanoEL, is correlated with the physicochemical properties of the nanoparticles in close association with cellular signalling, membrane mechanics, as well as cytoskeletal remodelling. However, nanoparticles in biological systems are transformed by the ubiquitous protein corona and yet the potential effect of the protein corona on NanoEL remains unclear. Using confocal fluorescence microscopy, biolayer interferometry, transwell, toxicity, and molecular inhibition assays, complemented by molecular docking, here we reveal the minimal to significant effects of the anionic human serum albumin and fibrinogen, the charge neutral immunoglobulin G as well as the cationic lysozyme on negating gold nanoparticle-induced endothelial leakiness in vitro and in vivo. This study suggests that nanoparticle-cadherin interaction and hence the extent of NanoEL may be partially controlled by pre-exposing the nanoparticles to plasma proteins of specific charge and topology to facilitate their biomedical applications.
Collapse
Affiliation(s)
- Aparna Nandakumar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia.
| | - Huayuan Tang
- Department of Engineering Mechanics, Hohai University, Nanjing 211100, China
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA.
| | - Nicholas Andrikopoulos
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia.
- Nanomedicine Centre, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - John F Quinn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia.
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA.
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia.
- Nanomedicine Centre, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia.
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| |
Collapse
|
4
|
Uritu CM, Al-Matarneh CM, Bostiog DI, Coroaba A, Ghizdovat V, Filipiuc SI, Simionescu N, Stefanescu C, Jalloul W, Nastasa V, Tamba BI, Maier SS, Pinteala M. Radiolabeled multi-layered coated gold nanoparticles as potential biocompatible PET/SPECT tracers. J Mater Chem B 2024; 12:3659-3675. [PMID: 38530751 DOI: 10.1039/d3tb02654j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
The demand for tailored, disease-adapted, and easily accessible radiopharmaceuticals is one of the most persistent challenges in nuclear imaging precision medicine. The aim of this work was to develop two multimodal radiotracers applicable for both SPECT and PET techniques, which consist of a gold nanoparticle core, a shell involved in radioisotope entrapment, peripherally placed targeting molecules, and biocompatibilizing polymeric sequences. Shell decoration with glucosamine units located in sterically hindered molecular environments is expected to result in nanoparticle accumulation in high-glucose-consuming areas. Gold cores were synthesized using the Turkevich method, followed by citrate substitution with linear PEG α,ω-functionalized with thiol and amine groups. The free amine groups facilitated the binding of branched polyethyleneimine through an epoxy ring-opening reaction by using PEG α,ω-diglycidyl ether as a linker. Afterwards, the glucose-PEG-epoxy prepolymer has been grafted onto the surface of AuPEG-PEI conjugates. Finally, the AuPEG-PEI-GA conjugates were radiolabeled with 99mTc or 68Ga. Instant thin-layer chromatography was used to evaluate the radiolabeling yield. The biocompatibility of non-labeled and 99mTc or 68Ga labeled nanoparticles was assessed on normal fibroblasts. The 99mTc complexes remained stable for over 22 hours, while the 68Ga containing ones revealed a slight decrease in stability after 1 hour.
Collapse
Affiliation(s)
- Cristina M Uritu
- Advanced Center for Research and Development in Experimental Medicine "Prof. Ostin C. Mungiu", "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Cristina M Al-Matarneh
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi, Romania.
| | - Denisse I Bostiog
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi, Romania.
| | - Adina Coroaba
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi, Romania.
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics, Nuclear medicine, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Silviu I Filipiuc
- Advanced Center for Research and Development in Experimental Medicine "Prof. Ostin C. Mungiu", "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Natalia Simionescu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi, Romania.
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics, Nuclear medicine, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Wael Jalloul
- Department of Biophysics and Medical Physics, Nuclear medicine, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Valentin Nastasa
- Faculty of Veterinary Medicine, "Ion Ionescu de la Brad" Iasi University of Life Science, Iasi, Romania.
| | - Bogdan I Tamba
- Advanced Center for Research and Development in Experimental Medicine "Prof. Ostin C. Mungiu", "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Stelian S Maier
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi, Romania.
- Polymers Research Center, "Gheorghe Asachi" Technical University of Iasi, Romania
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi, Romania.
| |
Collapse
|
5
|
Skrodzki D, Molinaro M, Brown R, Moitra P, Pan D. Synthesis and Bioapplication of Emerging Nanomaterials of Hafnium. ACS NANO 2024; 18:1289-1324. [PMID: 38166377 DOI: 10.1021/acsnano.3c08917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
A significant amount of progress in nanotechnology has been made due to the development of engineered nanoparticles. The use of metallic nanoparticles for various biomedical applications has been extensively investigated. Biomedical research is highly focused on them because of their inert nature, nanoscale structure, and similar size to many biological molecules. The intrinsic characteristics of these particles, including electronic, optical, physicochemical, and surface plasmon resonance, that can be altered by altering their size, shape, environment, aspect ratio, ease of synthesis, and functionalization properties, have led to numerous biomedical applications. Targeted drug delivery, sensing, photothermal and photodynamic therapy, and imaging are some of these. The promising clinical results of NBTXR3, a high-Z radiosensitizing nanomaterial derived from hafnium, have demonstrated translational potential of this metal. This radiosensitization approach leverages the dependence of energy attenuation on atomic number to enhance energy-matter interactions conducive to radiation therapy. High-Z nanoparticle localization in tumor issue differentially increases the effect of ionizing radiation on cancer cells versus nearby healthy ones and mitigates adverse effects by reducing the overall radiation burden. This principle enables material multifunctionality as contrast agents in X-ray-based imaging. The physiochemical properties of hafnium (Z = 72) are particularly advantageous for these applications. A well-placed K-edge absorption energy and high mass attenuation coefficient compared to elements in human tissue across clinical energy ranges leads to significant attenuation. Chemical reactivity allows for variety in nanoparticle synthesis, composition, and functionalization. Nanoparticles such as hafnium oxide exhibit excellent biocompatibility due to physiochemical inertness prior to incidence with ionizing radiation. Additionally, the optical and electronic properties are applicable in biosensing, optical component coatings, and semiconductors. The wide interest has prompted extensive research in design and synthesis to facilitate property fine-tuning. This review summarizes synthetic methods for hafnium-based nanomaterials and applications in therapy, imaging, and biosensing with a mechanistic focus. A discussion and future perspective section highlights clinical progress and elaborates on current challenges. By focusing on factors impacting applicational effectiveness and examining limitations this review aims to support researchers and expedite clinical translation of future hafnium-based nanomedicine.
Collapse
Affiliation(s)
- David Skrodzki
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Matthew Molinaro
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Richard Brown
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Parikshit Moitra
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Dipanjan Pan
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Huck Institutes of the Life Sciences, 101 Huck Life Sciences Building, University Park, Pennsylvania 16802, United States
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
6
|
Jenkins SV, Jung S, Jamshidi-Parsian A, Borrelli MJ, Dings RPM, Griffin RJ. Morphological Effects and In Vitro Biological Mechanisms of Radiation-Induced Cell Killing by Gold Nanomaterials. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58241-58250. [PMID: 38059477 DOI: 10.1021/acsami.3c15358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Gold nanomaterials have been shown to augment radiation therapy both in vitro and in vivo. However, studies on these materials are mostly phenomenological due to nanoparticle heterogeneity and the complexity of biological systems. Even accurate quantification of the particle dose still results in bulk average biases; the effect on individual cells is not measured but rather the effect on the overall population. To perform quantitative nanobiology, we coated glass coverslips uniformly at varying densities with Au nanoparticle preparations with different morphologies (45 nm cages, 25 nm spheres, and 30 nm rods). Consequently, the effect of a specific number of particles per unit area in contact with breast cancer cells growing on the coated surfaces was ascertained. Gold nanocages showed the highest degree of radiosensitization on a per particle basis, followed by gold nanospheres and gold nanorods, respectively. All three materials showed little cytotoxic effect at 0 Gy, but clonogenic survival decreased proportionally with the radiation dose and particle coverage density. A similar trend was seen in vivo in the combined treatment antitumor response in 4T1 tumor-bearing animals. The presence of gold affected the type and quantity of reactive oxygen species generated, specifically superoxide and hydroxyl radicals, and the concentration of nanocages correlated with the development of more numerous double-stranded DNA breaks and increased protein oxidation as measured by carbonylation. This work demonstrates the dependence on morphology and concentration of radiation enhancement by gold nanomaterials and may lead to a novel method to differentiate intra- and extracellular functionalities of gold nanomedicine treatment strategies. It further provides insights that can guide the rational development of gold nanomaterial-based radiosensitizers for clinical use.
Collapse
Affiliation(s)
- Samir V Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Seunghyun Jung
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Azemat Jamshidi-Parsian
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Michael J Borrelli
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
7
|
Mansouri E, Mesbahi A, Hamishehkar H, Montazersaheb S, Hosseini V, Rajabpour S. The effect of nanoparticle coating on biological, chemical and biophysical parameters influencing radiosensitization in nanoparticle-aided radiation therapy. BMC Chem 2023; 17:180. [PMID: 38082361 PMCID: PMC10712124 DOI: 10.1186/s13065-023-01099-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 12/04/2023] [Indexed: 04/25/2025] Open
Abstract
Nanoparticle-based composites have the potential to meet requirements for radiosensitization in both therapeutic and diagnostic applications. The radiosensitizing properties of nanoparticles could be reliant on the nature of their coating layer. Any gains in reduced toxicity and aggregation or improved delivery to tumor cells for coated nanoparticles must be weighed against the loss of dose enhancement. The radiosensitization potential of coated NPs is confirmed by numerous studies but in most of them, the coating layer is mostly applied to reduce toxicity of the NPs and for stability and biocompatibility aims. While the direct effects of the coating layer in radiosensitization-were ignored and not considered. This review provides an overview of double-edged impact of nanoparticle coating on the radiosensitization potential of nanostructures and discusses the challenges in choosing appropriate coating material in the aim of achieving improved radioenhancement. Coating layer could affect the radiosensitization processes and thereby the biological outcomes of nanoparticle-based radiation therapy. The physicochemical properties of the coating layer can be altered by the type of the coating material and its thickness. Under low-energy photon irradiation, the coating layer could act as a shield for nanoparticles capable of absorb produced low-energy electrons which are important levers for local and nanoscopic dose enhancement. Also, it seems that the coating layer could mostly affect the chemical process of ROS production rather than the physicochemical process. Based on the reviewed literature, for the irradiated coated nanoparticles, the cell survival and viability of cancer cells are decreased more than normal cells. Also, cell cycle arrest, inhibition of cell proliferation, DNA damage, cell death and apoptosis were shown to be affected by coated metallic nanoparticles under irradiation.
Collapse
Affiliation(s)
- Elham Mansouri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Institute of Biomedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghar Mesbahi
- Radiation Oncology Department, Olivia Newton-John Cancer, Wellness and Research center, Austin Health, Melbourne, Australia.
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Institute of Biomedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Molecular Medicine Research Center, Institute of Biomedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Rajabpour
- Medical Physics Department, Medical School, Tabriz University of Medical Sciences, Tabriz, Iran
| |
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
|
Gerken LRH, Gerdes ME, Pruschy M, Herrmann IK. Prospects of nanoparticle-based radioenhancement for radiotherapy. MATERIALS HORIZONS 2023; 10:4059-4082. [PMID: 37555747 PMCID: PMC10544071 DOI: 10.1039/d3mh00265a] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
Radiotherapy is a key pillar of solid cancer treatment. Despite a high level of conformal dose deposition, radiotherapy is limited due to co-irradiation of organs at risk and subsequent normal tissue toxicities. Nanotechnology offers an attractive opportunity for increasing the efficacy and safety of cancer radiotherapy. Leveraging the freedom of design and the growing synthetic capabilities of the nanomaterial-community, a variety of engineered nanomaterials have been designed and investigated as radiosensitizers or radioenhancers. While research so far has been primarily focused on gold nanoparticles and other high atomic number materials to increase the absorption cross section of tumor tissue, recent studies are challenging the traditional concept of high-Z nanoparticle radioenhancers and highlight the importance of catalytic activity. This review provides a concise overview on the knowledge of nanoparticle radioenhancement mechanisms and their quantification. It critically discusses potential radioenhancer candidate materials and general design criteria for different radiation therapy modalities, and concludes with research priorities in order to advance the development of nanomaterials, to enhance the efficacy of radiotherapy and to increase at the same time the therapeutic window.
Collapse
Affiliation(s)
- Lukas R H Gerken
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Maren E Gerdes
- Karolinska Institutet, Solnavägen 1, 171 77 Stockholm, Sweden
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Inge K Herrmann
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
10
|
Susnik E, Bazzoni A, Taladriz-Blanco P, Balog S, Moreno-Echeverri AM, Glaubitz C, Oliveira BB, Ferreira D, Baptista PV, Petri-Fink A, Rothen-Rutishauser B. Epidermal growth factor alters silica nanoparticle uptake and improves gold-nanoparticle-mediated gene silencing in A549 cells. FRONTIERS IN NANOTECHNOLOGY 2023; 5:1220514. [PMID: 37954478 PMCID: PMC7615298 DOI: 10.3389/fnano.2023.1220514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023] Open
Abstract
Introduction Delivery of therapeutic nanoparticles (NPs) to cancer cells represents a promising approach for biomedical applications. A key challenge for nanotechnology translation from the bench to the bedside is the low amount of administered NPs dose that effectively enters target cells. To improve NPs delivery, several studies proposed NPs conjugation with ligands, which specifically deliver NPs to target cells via receptor binding. One such example is epidermal growth factor (EGF), a peptide involved in cell signaling pathways that control cell division by binding to epidermal growth factor receptor (EGFR). However, very few studies assessed the influence of EGF present in the cell environment, on the cellular uptake of NPs. Methods We tested if the stimulation of EGFR-expressing lung carcinomacells A549 with EGF affects the uptake of 59 nm and 422 nm silica (SiO2) NPs. Additionally, we investigated whether the uptake enhancement can be achieved with gold NPs, suitable to downregulate the expression of cancer oncogene c-MYC. Results Our findings show that EGF binding to its receptor results in receptor autophosphorylation and initiate signaling pathways, leading to enhanced endocytosis of 59 nm SiO2 NPs, but not 422 nm SiO2 NPs. Additionally, we demonstrated an enhanced gold (Au) NPs endocytosis and subsequently a higher downregulation of c-MYC. Discussion These findings contribute to a better understanding of NPs uptake in the presence of EGF and that is a promising approach for improved NPs delivery.
Collapse
Affiliation(s)
- Eva Susnik
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Amelie Bazzoni
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | | | - Sandor Balog
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | | | | | - Beatriz Brito Oliveira
- i4HB, UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Daniela Ferreira
- i4HB, UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro Viana Baptista
- i4HB, UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
- Department of Chemistry, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
11
|
Musielak M, Boś-Liedke A, Piwocka O, Kowalska K, Markiewicz R, Lorenz A, Bakun P, Suchorska W. Methodological and Cellular Factors Affecting the Magnitude of Breast Cancer and Normal Cell Radiosensitization Using Gold Nanoparticles. Int J Nanomedicine 2023; 18:3825-3850. [PMID: 37457801 PMCID: PMC10349585 DOI: 10.2147/ijn.s412458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/18/2023] [Indexed: 07/18/2023] Open
Abstract
Purpose Breast cancer (BC) is the most common malignant tumor in women, which most often originates from the epithelial tissue of the breast gland. One of the most recommended kinds of treatment is radiotherapy (RT), but irradiation (IR) can affect not only the cancer tumor but also the healthy tissue around it. Au nanoparticles (AuNPs) were proposed as a radiosensitizing agent for RT which would allow for lower radiation doses, reducing the negative radiation effects on healthy tissues. The main objective of the study is to assess the dependence on the radiosensitivity of BC (MDA-MB-231) and normal mammary gland epithelial cells (MCF12A) to ionizing radiation, caused by functionalized AuNPs under diverse conditions. Methods The viability, uptake, reactive oxygen species induction, and mitochondrial membrane potential in cells were analyzed applying a time and concentration-dependent manner. After different incubation times with AuNPs, cells were exposed to 2 Gy. The determination of radiation effect in combination with AuNPs was investigated using the clonogenic assay, p53, and γH2AX level, as well as, Annexin V staining. Results Our results highlighted the strong need for assessing the experimental conditions' optimization before the AuNPs will be implemented with IR. Moreover, results indicated that AuNPs did not act universally in cells. Conclusion AuNPs could be a promising tool as a radiotherapy sensitizing agent, but it should be specified and deeply investigated under what conditions it will be applied taking into consideration not only AuNPs modifications but also the model and experimental conditions.
Collapse
Affiliation(s)
- Marika Musielak
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan, Poland
| | - Agnieszka Boś-Liedke
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Poznan, Poland
| | - Oliwia Piwocka
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan, Poland
| | - Katarzyna Kowalska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Aleksandra Lorenz
- Faculty of Agronomy, Horticulture and Bioengineering, Poznan University of Life Sciences, Poznan, Poland
| | - Paweł Bakun
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Poznan, Poland
| | - Wiktoria Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
12
|
Musielak M, Boś-Liedke A, Piwocka O, Kowalska K, Markiewicz R, Szymkowiak B, Bakun P, Suchorska WM. The Role of Functionalization and Size of Gold Nanoparticles in the Response of MCF-7 Breast Cancer Cells to Ionizing Radiation Comparing 2D and 3D In Vitro Models. Pharmaceutics 2023; 15:pharmaceutics15030862. [PMID: 36986725 PMCID: PMC10057027 DOI: 10.3390/pharmaceutics15030862] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/22/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Gold nanoparticles (AuNPs), as an agent enhancing radiosensitivity, play a key role in the potential treatment of breast cancer (BC). Assessing and understanding the kinetics of modern drug delivery systems is a crucial element that allows the implementation of AuNPs in clinical treatment. The main objective of the study was to assess the role of the properties of gold nanoparticles in the response of BC cells to ionizing radiation by comparing 2D and 3D models. In this research, four kinds of AuNPs, different in size and PEG length, were used to sensitize cells to ionizing radiation. The in vitro viability, uptake, and reactive oxygen species generation in cells were investigated in a time- and concentration-dependent manner using 2D and 3D models. Next, after the previous incubation with AuNPs, cells were irradiated with 2 Gy. The assessment of the radiation effect in combination with AuNPs was analyzed using the clonogenic assay and γH2AX level. The study highlights the role of the PEG chain in the efficiency of AuNPs in the process of sensitizing cells to ionizing radiation. The results obtained imply that AuNPs are a promising solution for combined treatment with radiotherapy.
Collapse
Affiliation(s)
- Marika Musielak
- Department of Electroradiology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Agnieszka Boś-Liedke
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, 61-614 Poznan, Poland
| | - Oliwia Piwocka
- Department of Electroradiology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Katarzyna Kowalska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 61-781 Poznan, Poland
| | - Roksana Markiewicz
- NanoBioMedical Centre, Adam Mickiewicz University, 61-614 Poznan, Poland
| | | | - Paweł Bakun
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, 61-781 Poznan, Poland
| | - Wiktoria M Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
13
|
Babaye Abdollahi B, Ghorbani M, Hamishehkar H, Malekzadeh R, Farajollahi A. Synthesis and characterization of actively HER-2 Targeted Fe 3O 4@Au nanoparticles for molecular radiosensitization of breast cancer. BIOIMPACTS : BI 2023; 13:17-29. [PMID: 36816996 PMCID: PMC9923814 DOI: 10.34172/bi.2022.23682] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 11/09/2022]
Abstract
Introduction: The present study was done to assess the effect of molecularly-targeted core/shell of iron oxide/gold nanoparticles (Fe3O4@AuNPs) on tumor radiosensitization of SKBr-3 breast cancer cells. Methods: Human epidermal growth factor receptor-2 (HER-2)-targeted Fe3O4@AuNPs were synthesized by conjugating trastuzumab (TZ, Herceptin) to PEGylated (PEG)-Fe3O4@AuNPs (41.5 nm). First, the Fe3O4@Au core-shell NPs were decorated with PEG-SH to synthesize PEG-Fe3O4@AuNPs. Then, the TZ was reacted to OPSS-PEG-SVA to conjugate with the PEG-Fe3O4@AuNPs. As a result, structure, size and morphology of the developed NPs were assessed using Fourier-transform infrared (FT-IR) spectroscopy, dynamic light scattering (DLS) and transmission electron microscopy (TEM), and ultraviolet-visible spectroscopy. The SKBr-3 cells were treated with different concentrations of TZ, Fe3O4@Au, and TZ-PEG-Fe3O4@AuNPs for irradiation at doses of 2, 4, and 8 Gy (from X-ray energy of 6 and 18 MV). Cytotoxicity was assessed by MTT assay, BrdU assay, and flow cytometry. Results: Results showed that the targeted TZ-PEG-Fe3O4@AuNPs significantly improved cell uptake. The cytotoxic effects of all the studied groups were increased in a higher concentration, radiation dose and energy-dependent manner. A combination of TZ, Fe3O4@Au, and TZ-PEG-Fe3O4@AuNPs with radiation reduced cell viability by 1.35 (P=0.021), 1.95 (P=0.024), and 1.15 (P=0.013) in comparison with 8 Gy dose of 18 MV radiation alone, respectively. These amounts were obtained as 1.27, 1.58, and 1.10 for 8 Gy dose of 6 MV irradiation, respectively. Conclusion: Radiosensitization of breast cancer to mega-voltage radiation therapy with TZ-PEG-Fe3O4@AuNPs was successfully obtained through an optimized therapeutic approach for molecular targeting of HER-2.
Collapse
Affiliation(s)
- Behnaz Babaye Abdollahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Ghorbani
- Nutration Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Malekzadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Farajollahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran,Imam Reza Educational Hospital, Radiotherapy Department, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding author: Ali Reza Farajollahi,
| |
Collapse
|
14
|
Hara D, Tao W, Schmidt RM, Yang YP, Daunert S, Dogan N, Ford JC, Pollack A, Shi J. Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4440. [PMID: 36558293 PMCID: PMC9784958 DOI: 10.3390/nano12244440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Metal nanoparticles are effective radiosensitizers that locally enhance radiation doses in targeted cancer cells. Compared with other metal nanoparticles, gold nanoparticles (GNPs) exhibit high biocompatibility, low toxicity, and they increase secondary electron scatter. Herein, we investigated the effects of active-targeting GNPs on the radiation-induced bystander effect (RIBE) in prostate cancer cells. The impact of GNPs on the RIBE presents implications for secondary cancers or spatially fractionated radiotherapy treatments. Anti-prostate-specific membrane antigen (PSMA) antibodies were conjugated with PEGylated GNPs through EDC-NHS chemistry. The media transfer technique was performed to induce the RIBE on the non-irradiated bystander cells. This study focused on the LNCaP cell line, because it can model a wide range of stages relating to prostate cancer progression, including the transition from androgen dependence to castration resistance and bone metastasis. First, LNCaP cells were pretreated with phosphate buffered saline (PBS) or PSMA-targeted GNPs (PGNPs) for 24 h and irradiated with 160 kVp X-rays (0-8 Gy). Following that, the collected culture media were filtered (sterile 0.45 µm polyethersulfone) in order to acquire PBS- and PGNP- conditioned media (CM). Then, PBS- and PGNP-CM were transferred to the bystander cells that were loaded with/without PGNPs. MTT, γ-H2AX, clonogenic assays and reactive oxygen species assessments were performed to compare RIBE responses under different treatments. Compared with 2 Gy-PBS-CM, 8 Gy-PBS-CM demonstrated a much higher RIBE response, thus validating the dose dependence of RIBE in LNCaP cells. Compared with PBS-CM, PGNP-CM exhibited lower cell viability, higher DNA damage, and a smaller survival fraction. In the presence of PBS-CM, bystander cells loaded with PGNPs showed increased cell death compared with cells that did not have PGNPs. These results demonstrate the PGNP-boosted expression and sensitivity of RIBE in prostate cancer cells.
Collapse
Affiliation(s)
- Daiki Hara
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - Wensi Tao
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ryder M. Schmidt
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sylvia Daunert
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Nesrin Dogan
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - John Chetley Ford
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - Alan Pollack
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Junwei Shi
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
15
|
Youden B, Jiang R, Carrier AJ, Servos MR, Zhang X. A Nanomedicine Structure-Activity Framework for Research, Development, and Regulation of Future Cancer Therapies. ACS NANO 2022; 16:17497-17551. [PMID: 36322785 DOI: 10.1021/acsnano.2c06337] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Despite their clinical success in drug delivery applications, the potential of theranostic nanomedicines is hampered by mechanistic uncertainty and a lack of science-informed regulatory guidance. Both the therapeutic efficacy and the toxicity of nanoformulations are tightly controlled by the complex interplay of the nanoparticle's physicochemical properties and the individual patient/tumor biology; however, it can be difficult to correlate such information with observed outcomes. Additionally, as nanomedicine research attempts to gradually move away from large-scale animal testing, the need for computer-assisted solutions for evaluation will increase. Such models will depend on a clear understanding of structure-activity relationships. This review provides a comprehensive overview of the field of cancer nanomedicine and provides a knowledge framework and foundational interaction maps that can facilitate future research, assessments, and regulation. By forming three complementary maps profiling nanobio interactions and pathways at different levels of biological complexity, a clear picture of a nanoparticle's journey through the body and the therapeutic and adverse consequences of each potential interaction are presented.
Collapse
Affiliation(s)
- Brian Youden
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Runqing Jiang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, Ontario N2G 1G3, Canada
| | - Andrew J Carrier
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Xu Zhang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Secondary Electrons in Gold Nanoparticle Clusters and Their Role in Therapeutic Ratio: The Outcome of a Monte Carlo Simulation Study. Molecules 2022; 27:molecules27165290. [PMID: 36014528 PMCID: PMC9415459 DOI: 10.3390/molecules27165290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Gold nanoparticles (GNPs) are used in proton therapy radio-sensitizers to help increase the dose of radiation to targeted tumors by the emission of secondary electrons. Thus, this study aimed to investigate the link between secondary electron yields produced from a nanoshell of GNPs and dose absorption according to the distance from the center of the nanoparticles by using a Monte Carlo model. Microscopic evaluation was performed by modeling the interactions of secondary electrons in a phase-space file (PSF), where the number of emitted electrons was calculated within a spherical GNP of 15 nm along with the absorbed dose near it. Then, the Geant4-DNA physics list was used to facilitate the tracking of low-energy electrons down to an energy below 50 eV in water. The results show a remarkable change in the number of secondary electrons, which can be compared at concentrations less than and greater than 5 mg/mL, with increased secondary electron production exhibited around NPs within a distance of 10–100 nm from the surface of all nanospheres. It was found that there was a steep dose enhancement drop-off up to a factor of dose enhancement factor (DFE) ≤ 1 within a short distance of 100 nm from the surface of the GNPs, which revealed that the dose enhancement existed locally at nanometer distances from the GNPs. Overall, our results indicate that the physical interactions of protons with GNP clusters should not be considered as being directly responsible for the radio-sensitization effect, but should be regarded as playing a major role in NP properties and concentrations, which has a subsequent impact on local dose enhancement.
Collapse
|
19
|
Guerra DB, Oliveira EMN, Sonntag AR, Sbaraine P, Fay AP, Morrone FB, Papaléo RM. Intercomparison of radiosensitization induced by gold and iron oxide nanoparticles in human glioblastoma cells irradiated by 6 MV photons. Sci Rep 2022; 12:9602. [PMID: 35688846 PMCID: PMC9187689 DOI: 10.1038/s41598-022-13368-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/22/2022] [Indexed: 12/04/2022] Open
Abstract
In this work, an intercomparison of sensitization effects produced by gold (GNP) and dextran-coated iron oxide (SPION-DX) nanoparticles in M059J and U87 human glioblastoma cells was performed using 6 MV-photons. Three variables were mapped: the nanoparticle material, treatment concentration, and cell radiosensitivity. For U87, GNP treatments resulted in high sensitization enhancement ratios (SER[Formula: see text] up to 2.04). More modest effects were induced by SPION-DX, but still significant reductions in survival were achieved (maximum SER[Formula: see text] ). For the radiosensitive M059J, sensitization by both NPs was poor. SER[Formula: see text] increased with the degree of elemental uptake in the cells, but not necessarily with treatment concentration. For GNP, where exposure concentration and elemental uptake were found to be proportional, SER[Formula: see text] increased linearly with concentration in both cell lines. For SPION-DX, saturation of sensitization enhancement and metal uptake occurred at high exposures. Fold change in the [Formula: see text] ratios extracted from survival curves are reduced by the presence of SPION-DX but strongly increased by GNPs , suggesting that sensitization by GNPs occurs mainly via promotion of lethal damage, while for SPION-DX repairable damage dominates. The NPs were more effective in eliminating the radioresistant glioblastoma cells, an interesting finding, as resistant cells are key targets to improve treatment outcome.
Collapse
Affiliation(s)
- Danieli B Guerra
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil.
| | - Elisa M N Oliveira
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Amanda R Sonntag
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Patricia Sbaraine
- Division of Radiotherapy, São Lucas Hospital of PUCRS, Porto Alegre, 90610-000, Brazil
| | - Andre P Fay
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Fernanda B Morrone
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Ricardo M Papaléo
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| |
Collapse
|
20
|
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
|
21
|
Cui X, Zhao Y, Zhang C, Meng Q. Nitro rotation tuned dissociative electron attachment upon targeted radiosensitizer 4-substituted Z bases. Phys Chem Chem Phys 2022; 24:10356-10364. [PMID: 35438101 DOI: 10.1039/d2cp00351a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this work, a set of new potential radiation sensitizers (4-substituted Z-bases: 4XZ, X = F, Cl, Br, and I) are designed based on the artificial 6-amino-5-nitro-3-(1'-β-D-2'-deoxyribofuranosyl)-2(1H)-pyridone (Z), which can selectively bind to breast cancer cells. The calculated electron affinities in water solution show that the halogenated Z-bases are efficient electron acceptors which possess significant electron-withdrawing characters following the order of 4XZ > Z ≫ U. To ensure the effective electron attachment induced dissociation, we constructed the energy profiles related to the X-C bond cleavage of neutral and anionic bases. The results show that the X-C bond becomes relatively weak after the electron attachment. In particular, the electron induced dehalogenations of (4BrZ)- and (4IZ)- are low-barrier and exothermic, which support a high radiosensitivity. Furthermore, we characterized the vibrational excitation effect on the dissociative electron attachment, which demonstrates that the charge distribution can be regulated by the rotation-induced structural distortion accompanied by the electron localization on the nitro group. Also examined is the influence of base pairing on the dehalogenation, which is not only conducive to the electron-driven dissociation but is also beneficial to the stabilization of related products. The current study suggests 4BrZ and 4IZ can be regarded as potential targeted radiosensitizers with possible applications in reducing the side effects in radiotherapy.
Collapse
Affiliation(s)
- Xixi Cui
- College of Physics and Electronics, Shandong Normal University, Jinan 250358, Shandong, China.
| | - Yu Zhao
- College of Physics and Electronics, Shandong Normal University, Jinan 250358, Shandong, China.
| | - Changzhe Zhang
- College of Physics and Electronics, Shandong Normal University, Jinan 250358, Shandong, China.
| | - Qingtian Meng
- College of Physics and Electronics, Shandong Normal University, Jinan 250358, Shandong, China.
| |
Collapse
|
22
|
Radiosensitization Effect of Gold Nanoparticles on Plasmid DNA Damage Induced by Therapeutic MV X-rays. NANOMATERIALS 2022; 12:nano12050771. [PMID: 35269259 PMCID: PMC8911739 DOI: 10.3390/nano12050771] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 01/30/2023]
Abstract
Gold nanoparticles (AuNPs) can be used with megavolt (MV) X-rays to exert radiosensitization effects, as demonstrated in cell survival assays and mouse experiments. However, the detailed mechanisms are not clear; besides physical dose enhancement, several chemical and biological processes have been proposed. Reducing the AuNP concentration while achieving sufficient enhancement is necessary for the clinical application of AuNPs. Here, we used positively charged (+) AuNPs to determine the radiosensitization effects of AuNPs combined with MV X-rays on DNA damage in vitro. We examined the effect of low concentrations of AuNPs on DNA damage and reactive oxygen species (ROS) generation. DNA damage was promoted by 1.4 nm +AuNP with dose enhancement factors of 1.4 ± 0.2 for single-strand breaks and 1.2 ± 0.1 for double-strand breaks. +AuNPs combined with MV X-rays induced radiosensitization at the DNA level, indicating that the effects were physical and/or chemical. Although −AuNPs induced similar ROS levels, they did not cause considerable DNA damage. Thus, dose enhancement by low concentrations of +AuNPs may have occurred with the increase in the local +AuNP concentration around DNA or via DNA binding. +AuNPs showed stronger radiosensitization effects than −AuNPs. Combining +AuNPs with MV X-rays in radiation therapy may improve clinical outcomes.
Collapse
|
23
|
Li R, Wang H, Liang Q, Chen L, Ren J. Radiotherapy for glioblastoma: clinical issues and nanotechnology strategies. Biomater Sci 2022; 10:892-908. [PMID: 34989724 DOI: 10.1039/d1bm01401c] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer in adults with poor prognosis. Despite the current state of knowledge on its genetic characteristics, relatively little progress has been made in improving the treatment of patients with this fatal disease. Radiotherapy (RT) has been identified as a crucial treatment for GBM following surgical resection to improve both local control and survival. Unfortunately, radiotherapy resistance is frequently observed in GBM patients, which is the major reason for the high mortality rate of cancer patients. Radioresistance of GBM is often multifactorial and heterogeneous, and associated with the recurrence of GBM after surgery. Nanotechnology has gained increasing attention and has already been investigated for optimization of radiosensitization due to the unique properties of nanobiomaterials, such as photoelectric decay characteristics or potential as carriers for drug delivery to the central nervous system. A large body of preclinical data has accumulated over the past several years, in which nanotechnology-based strategies exhibit promising potential to enhance the radiosensitivity of GBM, both in cellular and animal models. In this review, we summarize the mechanisms of GBM radioresistance, including tumor cell-intrinsic factors as well as tumor microenvironment (TME). We further discuss current nano-biotechnology-based radiosensitizer in the treatment of GBM, summarize the latest findings, highlight challenges, and put forward prospects for the future of nano-radiosensitizers. These data suggest that nanotechnology has the potential to address many of the clinical challenges and nanobiomaterials would become promising next-generation radiotherapy sensitizers for GBM treatment.
Collapse
Affiliation(s)
- Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| |
Collapse
|
24
|
Bartoli F, Eckelman WC, Boyd M, Mairs RJ, Erba PA. Principles of Molecular Targeting for Radionuclide Therapy. NUCLEAR ONCOLOGY 2022:41-93. [DOI: 10.1007/978-3-031-05494-5_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
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
|
26
|
Yan H, Carlson DJ, Abolfath R, Liu W. Microdosimetric Investigation and a Novel Model of Radiosensitization in the Presence of Metallic Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13122191. [PMID: 34959471 PMCID: PMC8709133 DOI: 10.3390/pharmaceutics13122191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/02/2022] Open
Abstract
Auger cascades generated in high atomic number nanoparticles (NPs) following ionization were considered a potential mechanism for NP radiosensitization. In this work, we investigated the microdosimetric consequences of the Auger cascades using the theory of dual radiation action (TDRA), and we propose the novel Bomb model as a general framework for describing NP-related radiosensitization. When triggered by an ionization event, the Bomb model considers the NPs that are close to a radiation sensitive cellular target, generates dense secondary electrons and kills the cells according to a probability distribution, acting like a “bomb.” TDRA plus a distance model were used as the theoretical basis for calculating the change in α of the linear-quadratic survival model and the relative biological effectiveness (RBE). We calculated these quantities for SQ20B and Hela human cancer cells under 250 kVp X-ray irradiation with the presence of gadolinium-based NPs (AGuIXTM), and 220 kVp X-ray irradiation with the presence of 50 nm gold NPs (AuNPs), respectively, and compared with existing experimental data. Geant4-based Monte Carlo (MC) simulations were used to (1) generate the electron spectrum and the phase space data of photons entering the NPs and (2) calculate the proximity functions and other related parameters for the TDRA and the Bomb model. The Auger cascade electrons had a greater proximity function than photoelectric and Compton electrons in water by up to 30%, but the resulting increases in α were smaller than those derived from experimental data. The calculated RBEs cannot explain the experimental findings. The relative increase in α predicted by TDRA was lower than the experimental result by a factor of at least 45 for SQ20B cells with AGuIX under 250 kVp X-ray irradiation, and at least four for Hela cells with AuNPs under 220 kVp X-ray irradiation. The application of the Bomb model to Hela cells with AuNPs under 220 kVp X-ray irradiation indicated that a single ionization event for NPs caused by higher energy photons has a higher probability of killing a cell. NPs that are closer to the cell nucleus are more effective for radiosensitization. Microdosimetric calculations of the RBE for cell death of the Auger electron cascade cannot explain the experimentally observed radiosensitization by AGuIX or AuNP, while the proposed Bomb model is a potential candidate for describing NP-related radiosensitization at low NP concentrations.
Collapse
Affiliation(s)
- Huagang Yan
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China;
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing 100069, China
| | - David J. Carlson
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Ramin Abolfath
- Department of Radiation Physics and Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 75031, USA;
- Department of Radiation Oncology, New Jersey Urology, West Orange, NJ 07052, USA
| | - Wu Liu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
27
|
Nandakumar A, Wei W, Siddiqui G, Li Y, Kakinen A, Wan X, Koppel K, Lin S, Davis TP, Leong DT, Creek DJ, Song Y, Ke PC. Dynamic Protein Corona of Gold Nanoparticles with an Evolving Morphology. ACS APPLIED MATERIALS & INTERFACES 2021; 13:58238-58251. [PMID: 34797630 PMCID: PMC8692073 DOI: 10.1021/acsami.1c19824] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Much has been learned about the protein coronae and their biological implications within the context of nanomedicine and nanotoxicology. However, no data is available about the protein coronae associated with nanoparticles undergoing spontaneous surface-energy minimization, a common phenomenon during the synthesis and shelf life of nanomaterials. Accordingly, here we employed gold nanoparticles (AuNPs) possessing the three initial states of spiky, midspiky, and spherical shapes and determined their acquisition of human plasma protein coronae with label-free mass spectrometry. The AuNPs collected coronal proteins that were different in abundance, physicochemical parameters, and interactive biological network. The size and structure of the coronal proteins matched the morphology of the AuNPs, where small globular proteins and large fibrillar proteins were enriched on spiky AuNPs, while large proteins were abundant on spherical AuNPs. Furthermore, the AuNPs induced endothelial leakiness to different degrees, which was partially negated by their protein coronae as revealed by confocal fluorescence microscopy, in vitro and ex vivo transwell assays, and signaling pathway assays. This study has filled a knowledge void concerning the dynamic protein corona of nanoparticles possessing an evolving morphology and shed light on their implication for future nanomedicine harnessing the paracellular pathway.
Collapse
Affiliation(s)
- Aparna Nandakumar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Wei Wei
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Xulin Wan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Kairi Koppel
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Sijie Lin
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Thomas P. Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - David T. Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| |
Collapse
|
28
|
Korman DB, Ostrovskaya LA, Bluhterova NV, Rykova VA, Fomina MM. Gold Nanoparticles as Potential Radiosensitizing and Cytotoxic Agents. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s0006350921060063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
29
|
Bennie LA, Feng J, Emmerson C, Hyland WB, Matchett KB, McCarthy HO, Coulter JA. Formulating RALA/Au nanocomplexes to enhance nanoparticle internalisation efficiency, sensitising prostate tumour models to radiation treatment. J Nanobiotechnology 2021; 19:279. [PMID: 34538237 PMCID: PMC8451112 DOI: 10.1186/s12951-021-01019-8] [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: 07/20/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Gold nanoparticles (AuNP) are effective radiosensitisers, however, successful clinical translation has been impeded by short systemic circulation times and poor internalisation efficiency. This work examines the potential of RALA, a short amphipathic peptide, to enhance the uptake efficiency of negatively charged AuNPs in tumour cells, detailing the subsequent impact of AuNP internalisation on tumour cell radiation sensitivity. RESULTS RALA/Au nanoparticles were formed by optimising the ratio of RALA to citrate capped AuNPs, with assembly occurring through electrostatic interactions. Physical nanoparticle characteristics were determined by UV-vis spectroscopy and dynamic light scattering. Nano-complexes successfully formed at w:w ratios > 20:1 (20 µg RALA:1 µg AuNP) yielding positively charged nanoparticles, sized < 110 nm with PDI values < 0.52. ICP-MS demonstrated that RALA enhanced AuNP internalisation by more than threefold in both PC-3 and DU145 prostate cancer cell models, without causing significant toxicity. Importantly, all RALA-AuNP formulations significantly increased prostate cancer cell radiosensitivity. This effect was greatest using the 25:1 RALA-AuNP formulation, producing a dose enhancement effect (DEF) of 1.54 in PC3 cells. Using clinical radiation energies (6 MV) RALA-AuNP also significantly augmented radiation sensitivity. Mechanistic studies support RALA-AuNP nuclear accumulation resulting in increased DNA damage yields. CONCLUSIONS This is the first study to demonstrate meaningful radiosensitisation using low microgram AuNP treatment concentrations. This effect was achieved using RALA, providing functional evidence to support our previous imaging study indicating RALA-AuNP nuclear accumulation.
Collapse
Affiliation(s)
- Lindsey A Bennie
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Jie Feng
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Christopher Emmerson
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Wendy B Hyland
- Western Health & Social Care Trust, North West Cancer Centre, Altnagelvin Hospital, Derry/Londonderry, BT47 6SB, Northern Ireland, UK
| | - Kyle B Matchett
- Northern Ireland Centre for Stratified Medicine, C-TRIC, Altnagelvin Hospital Campus, Derry/Londonderry, BT47 6SB, Northern Ireland, UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Jonathan A Coulter
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
30
|
Daems N, Michiels C, Lucas S, Baatout S, Aerts A. Gold nanoparticles meet medical radionuclides. Nucl Med Biol 2021; 100-101:61-90. [PMID: 34237502 DOI: 10.1016/j.nucmedbio.2021.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022]
Abstract
Thanks to their unique optical and physicochemical properties, gold nanoparticles have gained increased interest as radiosensitizing, photothermal therapy and optical imaging agents to enhance the effectiveness of cancer detection and therapy. Furthermore, their ability to carry multiple medically relevant radionuclides broadens their use to nuclear medicine SPECT and PET imaging as well as targeted radionuclide therapy. In this review, we discuss the radiolabeling process of gold nanoparticles and their use in (multimodal) nuclear medicine imaging to better understand their specific distribution, uptake and retention in different in vivo cancer models. In addition, radiolabeled gold nanoparticles enable image-guided therapy is reviewed as well as the enhancement of targeted radionuclide therapy and nanobrachytherapy through an increased dose deposition and radiosensitization, as demonstrated by multiple Monte Carlo studies and experimental in vitro and in vivo studies.
Collapse
Affiliation(s)
- Noami Daems
- Radiobiology Research Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium.
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire-NARILIS, University of Namur, Rue de Bruxelles 61, 5000 Namur, Belgium
| | - Stéphane Lucas
- Laboratory of Analysis by Nuclear Reaction (LARN)-NARILIS, University of Namur, Rue de Bruxelles 61, 5000 Namur, Belgium
| | - Sarah Baatout
- Radiobiology Research Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium
| | - An Aerts
- Radiobiology Research Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium
| |
Collapse
|
31
|
Apilan AG, Mothersill C. Targeted and Non-Targeted Mechanisms for Killing Hypoxic Tumour Cells-Are There New Avenues for Treatment? Int J Mol Sci 2021; 22:8651. [PMID: 34445354 PMCID: PMC8395506 DOI: 10.3390/ijms22168651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE A major issue in radiotherapy is the relative resistance of hypoxic cells to radiation. Historic approaches to this problem include the use of oxygen mimetic compounds to sensitize tumour cells, which were unsuccessful. This review looks at modern approaches aimed at increasing the efficacy of targeting and radiosensitizing hypoxic tumour microenvironments relative to normal tissues and asks the question of whether non-targeted effects in radiobiology may provide a new "target". Novel techniques involve the integration of recent technological advancements such as nanotechnology, cell manipulation, and medical imaging. Particularly, the major areas of research discussed in this review include tumour hypoxia imaging through PET imaging to guide carbogen breathing, gold nanoparticles, macrophage-mediated drug delivery systems used for hypoxia-activate prodrugs, and autophagy inhibitors. Furthermore, this review outlines several features of these methods, including the mechanisms of action to induce radiosensitization, the increased accuracy in targeting hypoxic tumour microenvironments relative to normal tissue, preclinical/clinical trials, and future considerations. CONCLUSIONS This review suggests that the four novel tumour hypoxia therapeutics demonstrate compelling evidence that these techniques can serve as powerful tools to increase targeting efficacy and radiosensitizing hypoxic tumour microenvironments relative to normal tissue. Each technique uses a different way to manipulate the therapeutic ratio, which we have labelled "oxygenate, target, use, and digest". In addition, by focusing on emerging non-targeted and out-of-field effects, new umbrella targets are identified, which instead of sensitizing hypoxic cells, seek to reduce the radiosensitivity of normal tissues.
Collapse
|
32
|
Pullambhatla M, Rowe SP, Lisok A, Wang Y, Todd GP, Danilkovitch A, Pomper MG. Enhancement of Radiotherapy with Human Mesenchymal Stem Cells Containing Gold Nanoparticles. ACTA ACUST UNITED AC 2021; 6:373-378. [PMID: 33364427 PMCID: PMC7744188 DOI: 10.18383/j.tom.2020.00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/07/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Radiotherapy is a common approach for the treatment of a wide variety of cancer types. Available data indicate that nanoparticles can enhance the effect of radiotherapy. We report the use of human mesenchymal stem cells to selectively deliver gold nanoparticles (GNPs) to MDA-MB-231 breast tumor xenografts in mice for the purpose of enhancing the effect of radiation therapy. Targeted delivery of GNPs to the tumor site, followed by irradiation of the tumor, enabled control of tumor growth. The results indicate that tumor-selective GNP delivery by human mesenchymal stem cells may represent a viable way to enhance the effectiveness of radiotherapy.
Collapse
Affiliation(s)
- Mrudula Pullambhatla
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Ala Lisok
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Yuchuan Wang
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | | | | | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD; and
| |
Collapse
|
33
|
Russell E, Dunne V, Russell B, Mohamud H, Ghita M, McMahon SJ, Butterworth KT, Schettino G, McGarry CK, Prise KM. Impact of superparamagnetic iron oxide nanoparticles on in vitro and in vivo radiosensitisation of cancer cells. Radiat Oncol 2021; 16:104. [PMID: 34118963 PMCID: PMC8199842 DOI: 10.1186/s13014-021-01829-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The recent implementation of MR-Linacs has highlighted theranostic opportunities of contrast agents in both imaging and radiotherapy. There is a lack of data exploring the potential of superparamagnetic iron oxide nanoparticles (SPIONs) as radiosensitisers. Through preclinical 225 kVp exposures, this study aimed to characterise the uptake and radiobiological effects of SPIONs in tumour cell models in vitro and to provide proof-of-principle application in a xenograft tumour model. METHODS SPIONs were also characterised to determine their hydrodynamic radius using dynamic light scattering and uptake was measured using ICP-MS in 6 cancer cell lines; H460, MiaPaCa2, DU145, MCF7, U87 and HEPG2. The impact of SPIONs on radiobiological response was determined by measuring DNA damage using 53BP1 immunofluorescence and cell survival. Sensitisation Enhancement Ratios (SERs) were compared with the predicted Dose Enhancement Ratios (DEFs) based on physical absorption estimations. In vivo efficacy was demonstrated using a subcutaneous H460 xenograft tumour model in SCID mice by following intra-tumoural injection of SPIONs. RESULTS The hydrodynamic radius was found to be between 110 and 130 nm, with evidence of being monodisperse in nature. SPIONs significantly increased DNA damage in all cell lines with the exception of U87 cells at a dose of 1 Gy, 1 h post-irradiation. Levels of DNA damage correlated with the cell survival, in which all cell lines except U87 cells showed an increased sensitivity (P < 0.05) in the linear quadratic curve fit for 1 h exposure to 23.5 μg/ml SPIONs. There was also a 30.1% increase in the number of DNA damage foci found for HEPG2 cells at 2 Gy. No strong correlation was found between SPION uptake and DNA damage at any dose, yet the biological consequences of SPIONs on radiosensitisation were found to be much greater, with SERs up to 1.28 ± 0.03, compared with predicted physical dose enhancement levels of 1.0001. In vivo, intra-tumoural injection of SPIONs combined with radiation showed significant tumour growth delay compared to animals treated with radiation or SPIONs alone (P < 0.05). CONCLUSIONS SPIONs showed radiosensitising effects in 5 out of 6 cancer cell lines. No correlation was found between the cell-specific uptake of SPIONs into the cells and DNA damage levels. The in vivo study found a significant decrease in the tumour growth rate.
Collapse
Affiliation(s)
- Emily Russell
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK.
- National Physical Laboratory, London, UK.
- Department of Medical Physics and Engineering, Leeds Teaching Hospitals, NHS Trust, Leeds, UK.
| | - Victoria Dunne
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | | | | | - Mihaela Ghita
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Stephen J McMahon
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Giuseppe Schettino
- National Physical Laboratory, London, UK
- Department of Physics, University of Surrey, Guildford, UK
| | - Conor K McGarry
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
- Northern Ireland Cancer Centre, Belfast, UK
| | - Kevin M Prise
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| |
Collapse
|
34
|
Rajendran I, Ponrasu T, Rajaram R, Suguna L. The apoptotic effect of Ferulic acid-synthesized gold nanoparticles against human epidermoid carcinoma (A431) cells via activation of caspase-3 pathway. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
35
|
Jindal M, Nagpal M, Singh M, Aggarwal G, Dhingra GA. Gold Nanoparticles- Boon in Cancer Theranostics. Curr Pharm Des 2021; 26:5134-5151. [PMID: 32611300 DOI: 10.2174/1381612826666200701151403] [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] [Received: 12/24/2019] [Accepted: 06/23/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cancer is the world's second-largest cause of death, with an estimated 9.6 million fatalities in 2018. Malignant tumour (cancer) is caused by a mixture of genetic modifications due to the environmental variables that tend to activate or inactivate different genes, ultimately resulting in neoplastic transformations. Cancer is a multi-stage process that results from the conversion of the ordinary cells to tumour cells and progresses from a pre-cancer lesion to abnormal growth. METHODS Chemotherapy inhibits the ability of the cells to divide rapidly in an abnormal manner, but this treatment simultaneously affects the entire cellular network in the human body leading to cytotoxic effects. In this review article, the same issue has been addressed by discussing various aspects of the newer class of drugs in cancer therapeutics, i.e., Gold Nanoparticles (AuNPs) from metal nanoparticle (NP) class. RESULTS Metal NPs are advantageous over conventional chemotherapy as the adverse drug reactions are lesser. Additionally, ease of drug delivery, targeting and gene silencing are salient features of this treatment. Functionalized ligand-targeting metal NPs provide better energy deposition control in tumour. AuNPs are promising agents in the field of cancer treatment and are comprehensively studied as contrast agents, carriers of medicinal products, radiosensitizers and photothermal agents. For the targeted delivery of chemotherapeutic agents, AuNPs are used and also tend to enhance tumour imaging in vivo for a variety of cancer types and diseased organs. CONCLUSION The first part of the review focuses on various nano-carriers that are used for cancer therapy and deals with the progression of metal NPs in cancer therapy. The second part emphasizes the use of nanotechnology by considering the latest studies for diagnostic and therapeutic properties of AuNPs. AuNPs present the latest studies in the field of nanotechnology, which leads to the development of early-stage clinical trials. The next part of the review discusses the major features of five principal types of AuNPs: gold nanorods, gold nanoshells, gold nanospheres, gold nanocages, and gold nanostars that have their application in photothermal therapy (PTT).
Collapse
Affiliation(s)
- Mehak Jindal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Geeta Aggarwal
- Delhi Pharmaceutical Sciences and Research University, New Delhi-110017, India
| | | |
Collapse
|
36
|
Yogo K, Misawa M, Shimizu M, Shimizu H, Kitagawa T, Hirayama R, Ishiyama H, Furukawa T, Yasuda H. Effect of Gold Nanoparticle Radiosensitization on Plasmid DNA Damage Induced by High-Dose-Rate Brachytherapy. Int J Nanomedicine 2021; 16:359-370. [PMID: 33469290 PMCID: PMC7813456 DOI: 10.2147/ijn.s292105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/19/2020] [Indexed: 01/20/2023] Open
Abstract
Purpose Gold nanoparticles (AuNPs) are candidate radiosensitizers for medium-energy photon treatment, such as γ-ray radiation in high-dose-rate (HDR) brachytherapy. However, high AuNP concentrations are required for sufficient dose enhancement for clinical applications. Here, we investigated the effect of positively (+) charged AuNP radiosensitization of plasmid DNA damage induced by 192Ir γ-rays, and compared it with that of negatively (−) charged AuNPs. Methods We observed DNA breaks and reactive oxygen species (ROS) generation in the presence of AuNPs at low concentrations. pBR322 plasmid DNA exposed to 64 ng/mL AuNPs was irradiated with 192Ir γ-rays via HDR brachytherapy. DNA breaks were detected by observing the changes in the form of the plasmid and quantified by agarose gel electrophoresis. The ROS generated by the AuNPs were measured with the fluorescent probe sensitive to ROS. The effects of positively (+) and negatively (−) charged AuNPs were compared to study the effect of surface charge on dose enhancement. Results +AuNPs at lower concentrations promoted a comparable level of radiosensitization by producing both single-stranded breaks (SSBs) and double-stranded breaks (DSBs) than those used in cell assays and Monte Carlo simulation experiments. The dose enhancement factor (DEF) for +AuNPs was 1.3 ± 0.2 for SSBs and 1.5 ± 0.4 for DSBs. The ability of +AuNPs to augment plasmid DNA damage is due to enhanced ROS generation. While −AuNPs generated similar ROS levels, they did not cause significant DNA damage. Thus, dose enhancement using low concentrations of +AuNPs presumably occurred via DNA binding or increasing local +AuNP concentration around the DNA. Conclusion +AuNPs at low concentrations displayed stronger radiosensitization compared to −AuNPs. Combining +AuNPs with 192Ir γ-rays in HDR brachytherapy is a candidate method for improving clinical outcomes. Future development of cancer cell-specific +AuNPs would allow their wider application for HDR brachytherapy.
Collapse
Affiliation(s)
- Katsunori Yogo
- Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masaki Misawa
- Health and Medical Research Institute, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Morihito Shimizu
- Health and Medical Research Institute, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Hidetoshi Shimizu
- Department of Radiation Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Tomoki Kitagawa
- Department of Radiation Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Ryoichi Hirayama
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba-shi, Chiba, Japan
| | - Hiromichi Ishiyama
- Graduate School of Medical Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Takako Furukawa
- Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hiroshi Yasuda
- Department of Radiation Biophysics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
37
|
Yang S, Han G, Chen Q, Yu L, Wang P, Zhang Q, Dong J, Zhang W, Huang J. Au-Pt Nanoparticle Formulation as a Radiosensitizer for Radiotherapy with Dual Effects. Int J Nanomedicine 2021; 16:239-248. [PMID: 33469284 PMCID: PMC7811476 DOI: 10.2147/ijn.s287523] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Background Radiotherapy occupies an essential position as one of the most significant approaches for the clinical treatment of cancer. However, we cannot overcome the shortcoming of X-rays which is the high value of the oxygen enhancement ratio (OER). Radiosensitizers with the ability to enhance the radiosensitivity of tumor cells provide an alternative to changing X-rays to protons and heavy ion radiotherapy. Materials and Methods We prepared the Au-Pt nanoparticles (Au-Pt NPs) using a one-step method. The characteristics of the Au-Pt NPs were determined using TEM, HAADF-STEM, elemental mapping images, and DLS. The enhanced radiotherapy was demonstrated in vitro using MTT assays, colony formation assays, fluorescence imaging, and flow cytometric analyses of the apoptosis. The biodistribution of the Au-Pt NPs was analyzed using ICP-OES, and thermal images. The enhanced radiotherapy was demonstrated in vitro using immunofluorescence images, tumor volume and weigh, and hematoxylin & eosin (H&E) staining. Results Polyethylene glycol (PEG) functionalized nanoparticles composed of the metallic elements Au and Pt were designed to increase synergistic radiosensitivity. The mechanism demonstrated that heavy metal NPs possess a high X-ray photon capture cross-section and Compton scattering effect which increased DNA damage. Furthermore, the Au-Pt NPs exhibited enzyme-mimicking activities by catalyzing the decomposition of endogenous H2O2 to O2 in the solid tumor microenvironment (TME). Conclusion Our work provides a systematically administered radiosensitizer that can selectively reside in a tumor via the EPR effect and enhances the efficiency of treating cancer with radiotherapy.
Collapse
Affiliation(s)
- Song Yang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Gaohua Han
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Quan Chen
- Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China
| | - Lei Yu
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Peng Wang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qi Zhang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jiang Dong
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Wei Zhang
- Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Infectious Disease, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China
| | - Junxing Huang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
38
|
Nosrati H, Baghdadchi Y, Abbasi R, Barsbay M, Ghaffarlou M, Abhari F, Mohammadi A, Kavetskyy T, Bochani S, Rezaeejam H, Davaran S, Danafar H. Iron oxide and gold bimetallic radiosensitizers for synchronous tumor chemoradiation therapy in 4T1 breast cancer murine model. J Mater Chem B 2021; 9:4510-4522. [PMID: 34027529 DOI: 10.1039/d0tb02561e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The development of highly integrated multifunctional nanomaterials with a superadditive therapeutic effect and good safety is an urgent but challenging task in cancer therapy research. The present study aims to design a nanoplatform that offers the opportunity to enhance antitumor activity while minimizing side effects. Given the Au-mediated X-ray radiation enhancement and the ability of Fe-based nanomaterials to create reactive oxygen species (ROS) and DNA damage, we anticipated that bimetallic Fe3O4-Au heterodimer would bring strong radiosensitizing capacity. Fe3O4-Au heterodimer surface was covered with bovine serum albumin (BSA) to achieve good surface functionality, stability and prolonged blood circulation. Folic acid (FA) moieties were added to the nanoformulation to increase tumor-homing, specificity and uptake. Finally, curcumin (CUR) was incorporated into the nanoparticle to function as a natural anticancer agent. The integration of all these components has yielded a single nanoplatform, Fe3O4-Au-BSA-FA-CUR, capable of successfully fulfilling the mission of superadditive cancer therapy to avoid the risks of organ removal surgery. The efficacy of the proposed nanoplatform was investigated in vitro and in vivo. High radiosensitizing ability, X-ray-induced ROS generation and DNA damage, and good biocompatibility were demonstrated through in vitro experiments. Also, the administration of Fe3O4-Au-BSA-FA-CUR with X-ray irradiation completely eradicated the tumor without any mortality and toxicity in healthy tissues in vivo. Our results highlight the potential of CUR-loaded Fe3O4-Au-BSA-FA heteronanostructure to enable synergistic localized radiochemotherapy and open up a new door to attractive possibilities that warrant further exploration.
Collapse
Affiliation(s)
- Hamed Nosrati
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran. and Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine, Baku, Azerbaijan and Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan 45139-56184, Iran
| | - Yasamin Baghdadchi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Reza Abbasi
- Department of Physics, Faculty of Science, University of Zanjan, Zanjan, 45371-38791, Iran
| | - Murat Barsbay
- Hacettepe University, Department of Chemistry, Beytepe, Ankara 06800, Turkey
| | | | - Fatemeh Abhari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Ali Mohammadi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Taras Kavetskyy
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine, Baku, Azerbaijan and Department of Surface Engineering, The John Paul II Catholic University of Lublin, 20-950 Lublin, Poland and Drohobych Ivan Franko State Pedagogical University, 82100 Drohobych, Ukraine
| | - Shayesteh Bochani
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Hamed Rezaeejam
- Department of Radiology, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Soodabeh Davaran
- Drug Applied Research Center, Tabriz University of Medical Sciences, P.O. Box: 51656-65811, Tabriz, Iran
| | - Hossein Danafar
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran. and Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine, Baku, Azerbaijan and Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan 45139-56184, Iran
| |
Collapse
|
39
|
Martínez-Rovira I, Seksek O, Dokic I, Brons S, Abdollahi A, Yousef I. Study of the intracellular nanoparticle-based radiosensitization mechanisms in F98 glioma cells treated with charged particle therapy through synchrotron-based infrared microspectroscopy. Analyst 2020; 145:2345-2356. [PMID: 31993615 DOI: 10.1039/c9an02350j] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The use of nanoparticles (NP) as dose enhancers in radiotherapy (RT) is a growing research field. Recently, the use of NP has been extended to charged particle therapy in order to improve the performance in radioresistant tumors. However, the biological mechanisms underlying the synergistic effects involved in NP-RT approaches are not clearly understood. Here, we used the capabilities of synchrotron-based Fourier Transform Infrared Microspectroscopy (SR-FTIRM) as a bio-analytical tool to elucidate the NP-induced cellular damage at the molecular level and at a single-cell scale. F98 glioma cells doped with AuNP and GdNP were irradiated using several types of medical ion beams (proton, helium, carbon and oxygen). Differences in cell composition were analyzed in the nucleic acids, protein and lipid spectral regions using multivariate methods (Principal Component Analysis, PCA). Several NP-induced cellular modifications were detected, such as conformational changes in secondary protein structures, intensity variations in the lipid CHx stretching bands, as well as complex DNA rearrangements following charged particle therapy irradiations. These spectral features seem to be correlated with the already shown enhancement both in the DNA damage response and in the reactive oxygen species (ROS) production by the NP, which causes cell damage in the form of protein, lipid, and/or DNA oxidations. Vibrational features were NP-dependent due to the NP heterogeneous radiosensitization capability. Our results provided new insights into the molecular changes in response to NP-based RT treatments using ion beams, and highlighted the relevance of SR-FTIRM as a useful and precise technique for assessing cell response to innovative radiotherapy approaches.
Collapse
Affiliation(s)
- I Martínez-Rovira
- MIRAS beamline BL01, ALBA-CELLS Synchrotron, Carrer de la Llum 2-26, 08290 Cerdanyola del Vallès, Spain.
| | - O Seksek
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, 91405 Orsay, France and Université de Paris, IJCLab, 91405 Orsay, France
| | - I Dokic
- Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany and Clinical Cooperation Unite Translational Radiation Oncology, German Cancer Consortium (DKTK) Core Center, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - S Brons
- Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - A Abdollahi
- Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany and Clinical Cooperation Unite Translational Radiation Oncology, German Cancer Consortium (DKTK) Core Center, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - I Yousef
- MIRAS beamline BL01, ALBA-CELLS Synchrotron, Carrer de la Llum 2-26, 08290 Cerdanyola del Vallès, Spain.
| |
Collapse
|
40
|
Clement S, Campbell JM, Deng W, Guller A, Nisar S, Liu G, Wilson BC, Goldys EM. Mechanisms for Tuning Engineered Nanomaterials to Enhance Radiation Therapy of Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2003584. [PMID: 33344143 PMCID: PMC7740107 DOI: 10.1002/advs.202003584] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Indexed: 05/12/2023]
Abstract
Engineered nanomaterials that produce reactive oxygen species on exposure to X- and gamma-rays used in radiation therapy offer promise of novel cancer treatment strategies. Similar to photodynamic therapy but suitable for large and deep tumors, this new approach where nanomaterials acting as sensitizing agents are combined with clinical radiation can be effective at well-tolerated low radiation doses. Suitably engineered nanomaterials can enhance cancer radiotherapy by increasing the tumor selectivity and decreasing side effects. Additionally, the nanomaterial platform offers therapeutically valuable functionalities, including molecular targeting, drug/gene delivery, and adaptive responses to trigger drug release. The potential of such nanomaterials to be combined with radiotherapy is widely recognized. In order for further breakthroughs to be made, and to facilitate clinical translation, the applicable principles and fundamentals should be articulated. This review focuses on mechanisms underpinning rational nanomaterial design to enhance radiation therapy, the understanding of which will enable novel ways to optimize its therapeutic efficacy. A roadmap for designing nanomaterials with optimized anticancer performance is also shown and the potential clinical significance and future translation are discussed.
Collapse
Affiliation(s)
- Sandhya Clement
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Jared M. Campbell
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Wei Deng
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Anna Guller
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
- Institute for Regenerative MedicineSechenov First Moscow State Medical University (Sechenov University)Trubetskaya StreetMoscow119991Russia
| | - Saadia Nisar
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Guozhen Liu
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Brian C. Wilson
- Department of Medical BiophysicsUniversity of Toronto/Princess Margaret Cancer CentreUniversity Health NetworkColledge StreetTorontoOntarioON M5G 2C1Canada
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| |
Collapse
|
41
|
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: 150] [Impact Index Per Article: 30.0] [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
|
42
|
Schuemann J, Bagley AF, Berbeco R, Bromma K, Butterworth KT, Byrne HL, Chithrani BD, Cho SH, Cook JR, Favaudon V, Gholami YH, Gargioni E, Hainfeld JF, Hespeels F, Heuskin AC, Ibeh UM, Kuncic Z, Kunjachan S, Lacombe S, Lucas S, Lux F, McMahon S, Nevozhay D, Ngwa W, Payne JD, Penninckx S, Porcel E, Prise KM, Rabus H, Ridwan SM, Rudek B, Sanche L, Singh B, Smilowitz HM, Sokolov KV, Sridhar S, Stanishevskiy Y, Sung W, Tillement O, Virani N, Yantasee W, Krishnan S. Roadmap for metal nanoparticles in radiation therapy: current status, translational challenges, and future directions. Phys Med Biol 2020; 65:21RM02. [PMID: 32380492 DOI: 10.1088/1361-6560/ab9159] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This roadmap outlines the potential roles of metallic nanoparticles (MNPs) in the field of radiation therapy. MNPs made up of a wide range of materials (from Titanium, Z = 22, to Bismuth, Z = 83) and a similarly wide spectrum of potential clinical applications, including diagnostic, therapeutic (radiation dose enhancers, hyperthermia inducers, drug delivery vehicles, vaccine adjuvants, photosensitizers, enhancers of immunotherapy) and theranostic (combining both diagnostic and therapeutic), are being fabricated and evaluated. This roadmap covers contributions from experts in these topics summarizing their view of the current status and challenges, as well as expected advancements in technology to address these challenges.
Collapse
Affiliation(s)
- Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Özçelik S, Pratx G. Nuclear-targeted gold nanoparticles enhance cancer cell radiosensitization. NANOTECHNOLOGY 2020; 31:415102. [PMID: 32585647 DOI: 10.1088/1361-6528/aba02b] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Radiation therapy aims to kill or inhibit proliferation of cancer cells while sparing normal cells. To enhance radiosensitization, we developed 40 nm-sized gold nanoparticles targeting the nucleus. We exploited a strategy that combined RGD and NLS peptides respectively targeting cancer cell and the nucleus to initiate cell-death activated by x-ray irradiation. We observed that the modified gold nanoparticles were either translocated in the nuclei or accumulated in the vicinity of the nuclei. We demonstrated that x-ray irradiation at 225 kVp energy reduced cell proliferation by 3.8-fold when the nuclear targeted gold nanoparticles were used. We determined that the radiation dose to have a 10% survival fraction was reduced from 11.0 Gy to 7.1 Gy when 10.0 µg ml-1 of the NLS/RGD/PEG-AuNP was incubated with A549 cancer cells. We conclude that the peptide-modified gold nanoparticles targeting the nucleus significantly enhance radiosensitization.
Collapse
Affiliation(s)
- Serdar Özçelik
- İzmir Institute of Technology, Department of Chemistry, Gülbahçe-Urla 35430, İzmir, Turkey. Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, United States of America
| | | |
Collapse
|
44
|
Advances in Gold Nanoparticle-Based Combined Cancer Therapy. NANOMATERIALS 2020; 10:nano10091671. [PMID: 32858957 PMCID: PMC7557687 DOI: 10.3390/nano10091671] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
According to the global cancer observatory (GLOBOCAN), there are approximately 18 million new cancer cases per year worldwide. Cancer therapies are largely limited to surgery, radiotherapy, and chemotherapy. In radiotherapy and chemotherapy, the maximum tolerated dose is presently being used to treat cancer patients. The integrated development of innovative nanoparticle (NP) based approaches will be a key to address one of the main issues in both radiotherapy and chemotherapy: normal tissue toxicity. Among other inorganic NP systems, gold nanoparticle (GNP) based systems offer the means to further improve chemotherapy through controlled delivery of chemotherapeutics, while local radiotherapy dose can be enhanced by targeting the GNPs to the tumor. There have been over 20 nanotechnology-based therapeutic products approved for clinical use in the past two decades. Hence, the goal of this review is to understand what we have achieved so far and what else we can do to accelerate clinical use of GNP-based therapeutic platforms to minimize normal tissue toxicity while increasing the efficacy of the treatment. Nanomedicine will revolutionize future cancer treatment options and our ultimate goal should be to develop treatments that have minimum side effects, for improving the quality of life of all cancer patients.
Collapse
|
45
|
Penninckx S, Heuskin AC, Michiels C, Lucas S. Gold Nanoparticles as a Potent Radiosensitizer: A Transdisciplinary Approach from Physics to Patient. Cancers (Basel) 2020; 12:E2021. [PMID: 32718058 PMCID: PMC7464732 DOI: 10.3390/cancers12082021] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, a growing interest in the improvement of radiation therapies has led to the development of gold-based nanomaterials as radiosensitizer. Although the radiosensitization effect was initially attributed to a dose enhancement mechanism, an increasing number of studies challenge this mechanistic hypothesis and evidence the importance of chemical and biological contributions. Despite extensive experimental validation, the debate regarding the mechanism(s) of gold nanoparticle radiosensitization is limiting its clinical translation. This article reviews the current state of knowledge by addressing how gold nanoparticles exert their radiosensitizing effects from a transdisciplinary perspective. We also discuss the current and future challenges to go towards a successful clinical translation of this promising therapeutic approach.
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; (S.P.); (A.-C.H.); (S.L.)
| | - 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; (S.P.); (A.-C.H.); (S.L.)
| | - 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; (S.P.); (A.-C.H.); (S.L.)
| |
Collapse
|
46
|
Neuer AL, Gerken LRH, Keevend K, Gogos A, Herrmann IK. Uptake, distribution and radio-enhancement effects of gold nanoparticles in tumor microtissues. NANOSCALE ADVANCES 2020; 2:2992-3001. [PMID: 36132396 PMCID: PMC9417636 DOI: 10.1039/d0na00256a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/28/2020] [Indexed: 05/27/2023]
Abstract
Radiotherapy is an integral and highly effective part of cancer therapy, applicable in over 50% of patients affected by cancer. Due to the low specificity of the X-ray irradiation, the maximal radiation dose is greatly limited in order to avoid damage to surrounding healthy tissue. The limitations in applicable dose oftentimes result in the survival of a subpopulation of radio-resistant cells that then cause cancer reoccurence. Approaches based on tumor-targeted high atomic number inorganic nanoparticles have been proposed to locally increase the photoelectric absorption cross-section of tumors relative to healthy tissue. However, the complex interplay between the nanoparticle radio-enhancers and the tumor tissue has led to poor translation of in vitro findings to (pre)clinics. Here, we report the development of a tumor microtissue model along with analytical imaging for the quantitative assessment of nanoparticle-based radio-enhancement as a function of nanoparticle size, uptake and intratissural distribution. The advanced in vitro model exhibits key features of cancerous tissues, including diminished susceptibility to drugs and attenuated response to nanoparticle treatment compared to corresponding conventional 2D cell cultures. Whereas radio-enhancement effects between 2D and 3D cell cultures were comparable for 5 nm gold particles, the limited penetration of 50 nm gold nanoparticles into 3D microtissues led to a significantly reduced radio-enhancement effect in 3D compared to 2D. Taken together, tumor microtissues, which in stark contrast to 2D cell culture exhibit tissue-like features, may provide a valuable high-throughput intermediate pre-selection step in the preclinical translation of nanoparticle-based radio-enhancement therapy designs.
Collapse
Affiliation(s)
- Anna L Neuer
- Laboratory for Particles Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa) Lerchenfeldstrasse 5 CH-9014 St. Gallen Switzerland +41 58 765 7153
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich Sonneggstrasse 3 CH-8092 Zurich Switzerland
| | - Lukas R H Gerken
- Laboratory for Particles Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa) Lerchenfeldstrasse 5 CH-9014 St. Gallen Switzerland +41 58 765 7153
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich Sonneggstrasse 3 CH-8092 Zurich Switzerland
| | - Kerda Keevend
- Laboratory for Particles Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa) Lerchenfeldstrasse 5 CH-9014 St. Gallen Switzerland +41 58 765 7153
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich Sonneggstrasse 3 CH-8092 Zurich Switzerland
| | - Alexander Gogos
- Laboratory for Particles Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa) Lerchenfeldstrasse 5 CH-9014 St. Gallen Switzerland +41 58 765 7153
| | - Inge K Herrmann
- Laboratory for Particles Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa) Lerchenfeldstrasse 5 CH-9014 St. Gallen Switzerland +41 58 765 7153
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich Sonneggstrasse 3 CH-8092 Zurich Switzerland
| |
Collapse
|
47
|
Babaye Abdollahi B, Malekzadeh R, Pournaghi Azar F, Salehnia F, Naseri AR, Ghorbani M, Hamishehkar H, Farajollahi AR. Main Approaches to Enhance Radiosensitization in Cancer Cells by Nanoparticles: A Systematic Review. Adv Pharm Bull 2020; 11:212-223. [PMID: 33880343 PMCID: PMC8046397 DOI: 10.34172/apb.2021.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/01/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
In recent years, high atomic number nanoparticles (NPs) have emerged as promising radio-enhancer agents for cancer radiation therapy due to their unique properties. Multi-disciplinary studies have demonstrated the potential of NPs-based radio-sensitizers to improve cancer therapy and tumor control at cellular and molecular levels. However, studies have shown that the dose enhancement effect of the NPs depends on the beam energy, NPs type, NPs size, NPs concentration, cell lines, and NPs delivery system. It has been believed that radiation dose enhancement of NPs is due to the three main mechanisms, but the results of some simulation studies failed to comply well with the experimental findings. Thus, this study aimed to quantitatively evaluate the physical, chemical, and biological factors of the NPs. An organized search of PubMed/Medline, Embase, ProQuest, Scopus, Cochrane and Google Scholar was performed. In total, 77 articles were thoroughly reviewed and analyzed. The studies investigated 44 different cell lines through 70 in-vitro and 4 in-vivo studies. A total of 32 different types of single or core-shell NPs in different sizes and concentrations have been used in the studies.
Collapse
Affiliation(s)
- Behnaz Babaye Abdollahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Malekzadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Pournaghi Azar
- Department of Operative Density, Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Salehnia
- Research Center for Evidence Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Reza Naseri
- Imam Reza Educational Hospital, Radiotherapy Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Ghorbani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Reza Farajollahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Imam Reza Educational Hospital, Radiotherapy Department, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
48
|
Spinelli AE, D'Agostino E, Broggi S, Claudio F, Boschi F. Small animal irradiator dose distribution verification using radioluminescence imaging. JOURNAL OF BIOPHOTONICS 2020; 13:e201960217. [PMID: 32163229 DOI: 10.1002/jbio.201960217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/05/2020] [Accepted: 03/08/2020] [Indexed: 06/10/2023]
Abstract
The main objective of this work was the development of a novel 2D dosimetry approach for small animal external radiotherapy using radioluminescence imaging (RLI) with a commercial complementary metal oxide semiconductor detector. Measurements of RLI were performed on the small animal image-guided platform SmART, RLI data were corrected for perspective distortion using Matlab. Four irradiation fields were tested and the planar 2D dose distributions and dose profiles were compared against dose calculations performed with a Monte Carlo based treatment planning system and gafchromic film. System linearity and RLI image noise against dose were also measured. The maximum difference between beam size measured with RLI and nominal beam size was less than 8% for all the tested beams. The image correction procedure was able to reduce perspective distortion. A novel RLI approach for quality assurance of a small animal irradiator was presented and tested. Results are in agreement with MC dose calculations and gafchromic film measurements.
Collapse
Affiliation(s)
| | | | - Sara Broggi
- Medical Physics Department, San Raffaele Scientific Institute, Italy
| | - Fiorino Claudio
- Medical Physics Department, San Raffaele Scientific Institute, Italy
| | | |
Collapse
|
49
|
Nakayama M, Akasaka H, Geso M, Morita K, Yada R, Uehara K, Sasaki R. Utilisation of the chemiluminescence method to measure the radiation dose enhancement caused by gold nanoparticles: A phantom-based study. RADIAT MEAS 2020. [DOI: 10.1016/j.radmeas.2020.106317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
50
|
Radiosensitization by Gold Nanoparticles: Impact of the Size, Dose Rate, and Photon Energy. NANOMATERIALS 2020; 10:nano10050952. [PMID: 32429500 PMCID: PMC7279506 DOI: 10.3390/nano10050952] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/06/2020] [Accepted: 05/09/2020] [Indexed: 01/09/2023]
Abstract
Gold nanoparticles (GNPs) emerged as promising antitumor radiosensitizers. However, the complex dependence of GNPs radiosensitization on the irradiation conditions remains unclear. In the present study, we investigated the impacts of the dose rate and photon energy on damage of the pBR322 plasmid DNA exposed to X-rays in the presence of 12 nm, 15 nm, 21 nm, and 26 nm GNPs. The greatest radiosensitization was observed for 26 nm GNPs. The sensitizer enhancement ratio (SER) 2.74 ± 0.61 was observed at 200 kVp with 2.4 mg/mL GNPs. Reduction of X-ray tube voltage to 150 and 100 kVp led to a smaller effect. We demonstrate for the first time that the change of the dose rate differentially influences on radiosensitization by GNPs of various sizes. For 12 nm, an increase in the dose rate from 0.2 to 2.1 Gy/min led to a ~1.13-fold increase in radiosensitization. No differences in the effect of 15 nm GNPs was found within the 0.85–2.1 Gy/min range. For 21 nm and 26 nm GNPs, an enhanced radiosensitization was observed along with the decreased dose rate from 2.1 to 0.2 Gy/min. Thus, GNPs are an effective tool for increasing the efficacy of orthovoltage X-ray exposure. However, careful selection of irradiation conditions is a key prerequisite for optimal radiosensitization efficacy.
Collapse
|