1
|
Li SR, Tao SY, Li Q, Hu CY, Sun ZJ. Harnessing nanomaterials for copper-induced cell death. Biomaterials 2025; 313:122805. [PMID: 39250865 DOI: 10.1016/j.biomaterials.2024.122805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024]
Abstract
Copper (Cu), an essential micronutrient with redox properties, plays a pivotal role in a wide array of pathological and physiological processes across virtually all cell types. Maintaining an optimal copper concentration is critical for cellular survival: insufficient copper levels disrupt respiration and metabolism, while excess copper compromises cell viability, potentially leading to cell death. Similarly, in the context of cancer, copper exhibits a dual role: appropriate amount of copper can promote tumor progression and be an accomplice, yet beyond befitting level, copper can bring about multiple types of cell death, including autophagy, apoptosis, ferroptosis, immunogenic cell death, pyroptosis, and cuproptosis. These forms of cell death are beneficial against cancer progression; however, achieving precise copper regulation within tumors remains a significant challenge in the pursuit of effective cancer therapies. The emergence of nanodrug delivery systems, distinguished by their precise targeting, controlled release, high payload capacity, and the ability to co-deliver multiple agents, has revitalized interest in exploiting copper's precise regulatory capabilities. Nevertheless, there remains a dearth of comprehensive review of copper's bidirectional effects on tumorigenesis and the role of copper-based nanomaterials in modulating tumor progression. This paper aims to address this gap by elucidating the complex role in cancer biology and highlighting its potential as a therapeutic target. Through an exploration of copper's dualistic nature and the application of nanotechnology, this review seeks to offer novel insights and guide future research in advancing cancer treatment.
Collapse
Affiliation(s)
- Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Shi-Yue Tao
- Bathune School of Stomatology, Jilin University, Changchun, 130021, Jilin, PR China
| | - Qian Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Chuan-Yu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China.
| |
Collapse
|
2
|
Nkosi NC, Basson AK, Ntombela ZG, Dlamini NG, Pullabhotla RVSR. A Review on Bioflocculant-Synthesized Copper Nanoparticles: Characterization and Application in Wastewater Treatment. Bioengineering (Basel) 2024; 11:1007. [PMID: 39451384 PMCID: PMC11504074 DOI: 10.3390/bioengineering11101007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Copper nanoparticles (CuNPs) are tiny materials with special features such as high electric conductivity, catalytic activity, antimicrobial activity, and optical activity. Published reports demonstrate their utilization in various fields, including biomedical, agricultural, environmental, wastewater treatment, and sensor fields. CuNPs can be produced utilizing traditional procedures; nevertheless, such procedures have restrictions like excessive consumption of energy, low production yields, and the utilization of detrimental substances. Thus, the adoption of environmentally approachable "green" approaches for copper nanoparticle synthesis is gaining popularity. These approaches involve employing plants, bacteria, and fungi. Nonetheless, there is a scarcity of data regarding the application of microbial bioflocculants in the synthesis of copper NPs. Therefore, this review emphasizes copper NP production using microbial flocculants, which offer economic benefits and are sustainable and harmless. The review also provides a characterization of the synthesized copper nanoparticles, employing numerous analytical tools to determine their compositional, morphological, and topographical features. It focuses on scientific advances from January 2015 to December 2023 and emphasizes the use of synthesized copper NPs in wastewater treatment.
Collapse
Affiliation(s)
- Nkanyiso C. Nkosi
- Biochemistry and Microbiology Department, Faculty of Science, Agriculture, and Engineering, University of Zululand, P/Bag X1001, KwaDlangezwa 3886, South Africa
| | - Albertus K. Basson
- Biochemistry and Microbiology Department, Faculty of Science, Agriculture, and Engineering, University of Zululand, P/Bag X1001, KwaDlangezwa 3886, South Africa
| | - Zuzingcebo G. Ntombela
- Biochemistry and Microbiology Department, Faculty of Science, Agriculture, and Engineering, University of Zululand, P/Bag X1001, KwaDlangezwa 3886, South Africa
| | - Nkosinathi G. Dlamini
- Biochemistry and Microbiology Department, Faculty of Science, Agriculture, and Engineering, University of Zululand, P/Bag X1001, KwaDlangezwa 3886, South Africa
| | - Rajasekhar V. S. R. Pullabhotla
- Chemistry Department, Faculty of Science, Agriculture, and Engineering, University of Zululand, P/Bag X1001, KwaDlangezwa 3886, South Africa
| |
Collapse
|
3
|
Mohammadi A, Bagheri F, Abutalebi Y, Aghaei A, Danafar H. Platinum nanoparticles-embedded single-walled carbon nanotubes as a new carrier for curcumin delivery and investigating its anticancer effect on cell line 4T1. Heliyon 2024; 10:e33703. [PMID: 39027555 PMCID: PMC11255493 DOI: 10.1016/j.heliyon.2024.e33703] [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: 03/27/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
Cancer, a prevalent disease across various societies, presents a significant challenge in treatment research. Studies show that combination therapies are one of the methods that can help in the effective treatment of cancer. Chemotherapy and radiation therapy are among the main cancer treatments and in this project, for combined chemoradiotherapy treatment, carbon nanotubes were used as improved carriers of chemotherapy in tumors, as well as a substrate for the preparation of radiation sensitizers for local radiation therapy. Following the synthesis of CNT-Platinum-Curcumin nanoparticles (CNT-Pt-CUR), a series of analyses were conducted to verify the successful production of these nanoparticles. Techniques such as Transmission Electron Microscopy (TEM), Dynamic Light Scattering (DLS), UV-Vis spectroscopy, Fourier Transform Infrared Spectroscopy (FTIR), and X-Ray Diffraction (XRD) were employed. The characterization data revealed a spherical shape Pt nanoparticle morphology with an 8.5 nm diameter on rod-shape CNT, as observed through TEM. Furthermore, FTIR analysis confirmed the successful loaded of the drug into the nanoparticles, highlighting the potential of this approach in cancer treatment. Then, hemolysis and (3(-4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) tests on normal cells were used to assess the biocompatibility of CNT-Pt-CUR nanoparticles. It also explored the anticancer efficacy of these nanoparticles at varying concentrations against cancer cells, both with and without exposure to X-rays. The research confirmed the successful synthesis of these nanoparticles and demonstrated their potential impact on cell viability. Specifically, breast cancer cells exhibited heightened susceptibility to toxicity when exposed to nanoparticles and X-rays. Further analysis revealed that the toxicity of nanoparticles is dose-dependent, and modifying the surface of carbon nanotube (CNT) nanoparticles with CUR significantly reduced blood toxicity. Interestingly, nanoparticle toxicity was significantly amplified in the presence of X-rays, suggesting mechanisms such as DNA damage and increased reactive oxygen species (ROS) levels within cells.
Collapse
Affiliation(s)
- Ali Mohammadi
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fariba Bagheri
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Yasamin Abutalebi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Afsoon Aghaei
- Department of Chemistry, University of Zanjan, Zanjan, Iran
| | - Hossein Danafar
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
4
|
Florance I, Cordani M, Pashootan P, Moosavi MA, Zarrabi A, Chandrasekaran N. The impact of nanomaterials on autophagy across health and disease conditions. Cell Mol Life Sci 2024; 81:184. [PMID: 38630152 PMCID: PMC11024050 DOI: 10.1007/s00018-024-05199-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 04/19/2024]
Abstract
Autophagy, a catabolic process integral to cellular homeostasis, is constitutively active under physiological and stress conditions. The role of autophagy as a cellular defense response becomes particularly evident upon exposure to nanomaterials (NMs), especially environmental nanoparticles (NPs) and nanoplastics (nPs). This has positioned autophagy modulation at the forefront of nanotechnology-based therapeutic interventions. While NMs can exploit autophagy to enhance therapeutic outcomes, they can also trigger it as a pro-survival response against NP-induced toxicity. Conversely, a heightened autophagy response may also lead to regulated cell death (RCD), in particular autophagic cell death, upon NP exposure. Thus, the relationship between NMs and autophagy exhibits a dual nature with therapeutic and environmental interventions. Recognizing and decoding these intricate patterns are essential for pioneering next-generation autophagy-regulating NMs. This review delves into the present-day therapeutic potential of autophagy-modulating NMs, shedding light on their status in clinical trials, intervention of autophagy in the therapeutic applications of NMs, discusses the potency of autophagy for application as early indicator of NM toxicity.
Collapse
Affiliation(s)
- Ida Florance
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Parya Pashootan
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O Box 14965/161, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O Box 14965/161, Tehran, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, Taiwan
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
5
|
Shao L, Wang X, Du X, Yin S, Qian Y, Yao Y, Yang L. Application of Multifunctional Nanozymes in Tumor Therapy. ACS OMEGA 2024; 9:15753-15767. [PMID: 38617672 PMCID: PMC11007812 DOI: 10.1021/acsomega.4c00258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/25/2024] [Accepted: 03/13/2024] [Indexed: 04/16/2024]
Abstract
Tumors are one of the main diseases threatening human life and health. The emergence of nanotechnology in recent years has introduced a novel therapeutic avenue for addressing tumors. Through the amalgamation of nanotechnology's inherent attributes with those of natural enzymes, nanozymes have demonstrated the ability to initiate catalytic reactions, modulate the biological microenvironment, and facilitate the adoption of multifaceted therapeutic approaches, thereby exhibiting considerable promise in the realm of cancer treatment. In this Review, the application of nanozymes in chemodynamic therapy, radiotherapy, photodynamic therapy, photothermal therapy, and starvation therapy are summarized. Moreover, a detailed discussion regarding the mechanism of conferring physiotherapeutic functionality upon catalytic nanosystems is provided. It is posited that this innovative catalytic treatment holds significant potential to play a crucial role within the domain of nanomedicine.
Collapse
Affiliation(s)
- Lihua Shao
- Department
of Colorectal Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Xueyuan Wang
- College of
Life Science, Nanjing Normal University, Nanjing, Jiangsu 210046, China
| | - Xiao Du
- Department
of Pharmacy, Nanjing Medical Center for Clinical Pharmacy, Nanjing
Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Shaoping Yin
- School of
Pharmacy, Nanjing University of Chinese
Medicine, Nanjing, Jiangsu 210023, China
| | - Yun Qian
- Dermatologic
Surgery Department, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing, Jiangsu 210042, China
| | - Yawen Yao
- Department
of Pharmaceutics, School of Pharmacy, China
Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Lin Yang
- College of
Science, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| |
Collapse
|
6
|
Golchin A, Maleki M, Alemi F, Malakoti F, Yousefi B. Autophagy-targeted nanoparticles in breast carcinoma: A systematic review. Cell Biol Int 2023; 47:1767-1781. [PMID: 37671447 DOI: 10.1002/cbin.12081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/10/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023]
Abstract
Breast cancer is a commonly known cancer type and the leading cause of cancer death among females. One of the unresolved problems in cancer treatment is the increased resistance of the tumor to existing treatments, which is a direct result of apoptotic defects. Calculating an alternative to cell death (autophagy) may be the ultimate solution to maximizing cancer cell death. Our aim in this study was to investigate the potential of free nanoparticles (un-drug-loaded) in the induction or inhibition of autophagy and consider this effect on the therapy process. When the studies met the inclusion criteria, the full texts of all relevant articles were carefully examined and classified. Of the 25 articles included in the analysis, carried out on MCF-7, MDA-MB-231, MDA-MB-231-TXSA, MDA-MB-468, SUM1315, and 4T1 cell lines. Twenty in vitro studies and five in vivo/in vitro studies applied five different autophagy tests: Acridine orange, western blot, Cyto-ID Autophagy Detection Kit, confocal microscope, and quantitative polymerase chain reaction. Nanoparticles (NPs) in the basic format, including Ag, Au, Y2 O3 , Se, ZnO, CuO, Al, Fe, vanadium pentoxide, and liposomes, were prepared in the included articles. Three behaviors of NPs related to autophagy were seen: induction, inhibition, and no action. Screened and presented data suggest that most of the involved free NPs (metallic NPs) in this systematic review had reactive oxygen species-mediated pathways with autophagy induction (36%). Also, PI3K/Akt/mTOR and MAPK/ERK signaling pathways were mentioned in just four studies (16%). An impressive percentage of studies (31%) did not examine the NP-related autophagy pathway.
Collapse
Affiliation(s)
- Asal Golchin
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoumeh Maleki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
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: 1] [Impact Index Per Article: 1.0] [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
|
8
|
Buglewicz DJ, Su C, Banks AB, Stenger-Smith J, Elmegerhi S, Hirakawa H, Fujimori A, Kato TA. Metal Ions Modify In Vitro DNA Damage Yields with High-LET Radiation. TOXICS 2023; 11:773. [PMID: 37755783 PMCID: PMC10537317 DOI: 10.3390/toxics11090773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023]
Abstract
Cu2+ and Co2+ are metals known to increase DNA damage in the presence of hydrogen peroxide through a Fenton-type reaction. We hypothesized that these metals could increase DNA damage following irradiations of increasing LET values as hydrogen peroxide is a product of the radiolysis of water. The reaction mixtures contain either double- or single-stranded DNA in solution with Cu2+ or Co2+ and were irradiated either with X-ray, carbon-ion or iron-ion beams, or they were treated with hydrogen peroxide or bleomycin at increasing radiation dosages or chemical concentrations. DNA damage was then assessed via gel electrophoresis followed with a band intensity analysis. DNA damage was the greatest when DNA in the solution with either metal was treated with only hydrogen peroxide followed by the DNA damage of DNA in the solution with either metal post irradiation of low-LET (X-Ray) or high-LET (carbon-ion and iron-ion), respectively, and demonstrated the least damage after treatment with bleomycin. Cu2+ portrayed greater DNA damage than Co2+ following all experimental conditions. The metals' effect caused more DNA damage and was observed to be LET-dependent for single-strand break formation but inversely dependent for double-strand break formation. These results suggest that Cu2+ is more efficient than Co2+ at inducing both DNA single-strand and double-strand breaks following all irradiations and chemical treatments.
Collapse
Affiliation(s)
- Dylan J. Buglewicz
- National Institute of Radiological Sciences, National Institutes of Quantum Science and Technology, Chiba 263-8555, Japan; (D.J.B.); (H.H.); (A.F.)
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Cathy Su
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Austin B. Banks
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Jazmine Stenger-Smith
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Suad Elmegerhi
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Hirokazu Hirakawa
- National Institute of Radiological Sciences, National Institutes of Quantum Science and Technology, Chiba 263-8555, Japan; (D.J.B.); (H.H.); (A.F.)
| | - Akira Fujimori
- National Institute of Radiological Sciences, National Institutes of Quantum Science and Technology, Chiba 263-8555, Japan; (D.J.B.); (H.H.); (A.F.)
| | - Takamitsu A. Kato
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| |
Collapse
|
9
|
Baranwal J, Barse B, Di Petrillo A, Gatto G, Pilia L, Kumar A. Nanoparticles in Cancer Diagnosis and Treatment. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5354. [PMID: 37570057 PMCID: PMC10420054 DOI: 10.3390/ma16155354] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
The use of tailored medication delivery in cancer treatment has the potential to increase efficacy while decreasing unfavourable side effects. For researchers looking to improve clinical outcomes, chemotherapy for cancer continues to be the most challenging topic. Cancer is one of the worst illnesses despite the limits of current cancer therapies. New anticancer medications are therefore required to treat cancer. Nanotechnology has revolutionized medical research with new and improved materials for biomedical applications, with a particular focus on therapy and diagnostics. In cancer research, the application of metal nanoparticles as substitute chemotherapy drugs is growing. Metals exhibit inherent or surface-induced anticancer properties, making metallic nanoparticles extremely useful. The development of metal nanoparticles is proceeding rapidly and in many directions, offering alternative therapeutic strategies and improving outcomes for many cancer treatments. This review aimed to present the most commonly used nanoparticles for cancer applications.
Collapse
Affiliation(s)
- Jaya Baranwal
- DBT-ICGEB Centre for Advanced Bioenergy Research, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Brajesh Barse
- US India Business Council|US Chamber of Commerce, DLF Centre, Sansad Marg, New Delhi 110001, India
| | - Amalia Di Petrillo
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042 Cagliari, Italy;
| | - Gianluca Gatto
- Department of Electrical and Electronic Engineering, University of Cagliari, Via Marengo 2, 09123 Cagliari, Italy;
| | - Luca Pilia
- Department of Mechanical, Chemical and Material Engineering, University of Cagliari, Via Marengo 2, 09123 Cagliari, Italy
| | - Amit Kumar
- Department of Electrical and Electronic Engineering, University of Cagliari, Via Marengo 2, 09123 Cagliari, Italy;
| |
Collapse
|
10
|
Zhou Q, Li Y, Li L, Sun N, Zhang H, Jiang J, Du T, Mo Y, Aldeen A, Xiao R, Chen Y, Wang S, Liu M, Li C, Feng X. Radiosensitization of Nasopharyngeal Carcinoma by Graphene Oxide Nanosheets to Reduce Bcl-2 Level. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:4245-4256. [PMID: 36913208 DOI: 10.1021/acs.langmuir.2c03106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
There are many treatments for nasopharyngeal carcinoma (NPC), but none of them are very effective. Radiotherapy is used extensively in NPC treatment, but radioresistance is a major problem. Graphene oxide (GO) has been previously studied in cancer treatment, and this study is aimed to explore its role in radiosensitization of NPC. Therefore, graphene oxide nanosheets were prepared, and the relationship between GO and radioresistance was explored. The GO nanosheets were synthesized by a modified Hummers' method. The morphologies of the GO nanosheets were characterized by field-emission environmental scanning electron microscopy (SEM) and transmission electron microscopy (TEM). The morphological changes and radiosensitivity of C666-1 and HK-1 cells with or without the GO nanosheets were observed by an inverted fluorescence microscopy and laser scanning confocal microscopy (LSCM). Colony formation assay and Western Blot were applied for analysis of NPC radiosensitivity. The as-synthesized GO nanosheets have lateral dimensions (sizes ∼1 μm) and exhibit a thin wrinkled two-dimensional lamellar structure with slight folds and crimped edges (thickness values ∼1 nm). C666-1 cells with the GO was significantly changed the morphology of cells postirradiation. The full field of view visualized by a microscope showed the shadow of dead cells or cell debris. The synthesized graphene oxide nanosheets inhibited cell proliferation, promoted cell apoptosis, and inhibited the expression of Bcl-2 in C666-1 and HK-1 cells but increased the level of Bax. The GO nanosheets could affect the cell apoptosis and reduce the pro-survival protein Bcl-2 related to the intrinsic mitochondrial pathway. The GO nanosheets could enhance radiosensitivity, which might be a radioactive material in NPC cells.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Yadong Li
- Chenzhou Maternal and Child Health Hospital, Chenzhou 423000, China
| | - Liya Li
- Powder Metallurgy Research Institute, Central South University, Changsha 410083, China
| | - Nianzhe Sun
- Department of Orthopedics, Hand & Microsurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Hanghao Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Jiahui Jiang
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Tao Du
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Yan Mo
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Alaa Aldeen
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Runsha Xiao
- Department of Gastrointestinal, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yiting Chen
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Shuanglian Wang
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Mian Liu
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Chengmin Li
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
- Department of Pathology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Xueping Feng
- Department of Otolaryngology-head and Neck Surgery, Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| |
Collapse
|
11
|
Długosz O, Matyjasik W, Hodacka G, Szostak K, Matysik J, Krawczyk P, Piasek A, Pulit-Prociak J, Banach M. Inorganic Nanomaterials Used in Anti-Cancer Therapies:Further Developments. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13061130. [PMID: 36986024 PMCID: PMC10051539 DOI: 10.3390/nano13061130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/14/2023]
Abstract
In this article, we provide an overview of the progress of scientists working to improve the quality of life of cancer patients. Among the known methods, cancer treatment methods focusing on the synergistic action of nanoparticles and nanocomposites have been proposed and described. The application of composite systems will allow precise delivery of therapeutic agents to cancer cells without systemic toxicity. The nanosystems described could be used as a high-efficiency photothermal therapy system by exploiting the properties of the individual nanoparticle components, including their magnetic, photothermal, complex, and bioactive properties. By combining the advantages of the individual components, it is possible to obtain a product that would be effective in cancer treatment. The use of nanomaterials to produce both drug carriers and those active substances with a direct anti-cancer effect has been extensively discussed. In this section, attention is paid to metallic nanoparticles, metal oxides, magnetic nanoparticles, and others. The use of complex compounds in biomedicine is also described. A group of compounds showing significant potential in anti-cancer therapies are natural compounds, which have also been discussed.
Collapse
|
12
|
Chuang YC, Wu PH, Shen YA, Kuo CC, Wang WJ, Chen YC, Lee HL, Chiou JF. Recent Advances in Metal-Based NanoEnhancers for Particle Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1011. [PMID: 36985905 PMCID: PMC10056155 DOI: 10.3390/nano13061011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Radiotherapy is one of the most common therapeutic regimens for cancer treatment. Over the past decade, proton therapy (PT) has emerged as an advanced type of radiotherapy (RT) that uses proton beams instead of conventional photon RT. Both PT and carbon-ion beam therapy (CIBT) exhibit excellent therapeutic results because of the physical characteristics of the resulting Bragg peaks, which has been exploited for cancer treatment in medical centers worldwide. Although particle therapies show significant advantages to photon RT by minimizing the radiation damage to normal tissue after the tumors, they still cause damage to normal tissue before the tumor. Since the physical mechanisms are different from particle therapy and photon RT, efforts have been made to ameliorate these effects by combining nanomaterials and particle therapies to improve tumor targeting by concentrating the radiation effects. Metallic nanoparticles (MNPs) exhibit many unique properties, such as strong X-ray absorption cross-sections and catalytic activity, and they are considered nano-radioenhancers (NREs) for RT. In this review, we systematically summarize the putative mechanisms involved in NRE-induced radioenhancement in particle therapy and the experimental results in in vitro and in vivo models. We also discuss the potential of translating preclinical metal-based NP-enhanced particle therapy studies into clinical practice using examples of several metal-based NREs, such as SPION, Abraxane, AGuIX, and NBTXR3. Furthermore, the future challenges and development of NREs for PT are presented for clinical translation. Finally, we propose a roadmap to pursue future studies to strengthen the interplay of particle therapy and nanomedicine.
Collapse
Affiliation(s)
- Yao-Chen Chuang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
| | - Ping-Hsiu Wu
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Chia-Chun Kuo
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei 110301, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jun Wang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yu-Chen Chen
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Hsin-Lun Lee
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
13
|
Salehiabar M, Ghaffarlou M, Mohammadi A, Mousazadeh N, Rahimi H, Abhari F, Rashidzadeh H, Nasehi L, Rezaeejam H, Barsbay M, Ertas YN, Nosrati H, Kavetskyy T, Danafar H. Targeted CuFe 2O 4 hybrid nanoradiosensitizers for synchronous chemoradiotherapy. J Control Release 2023; 353:850-863. [PMID: 36493951 DOI: 10.1016/j.jconrel.2022.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/08/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
Multifunctional nanoplatforms based on novel bimetallic nanoparticles have emerged as effective radiosensitizers owing to their potential capability in cancer cells radiosensitization. Implementation of chemotherapy along with radiotherapy, known as synchronous chemoradiotherapy, can augment the treatment efficacy. Herein, a tumor targeted nanoradiosensitizer with synchronous chemoradiotion properties, termed as CuFe2O4@BSA-FA-CUR, loaded with curcumin (CUR) and modified by bovine serum albumin (BSA) and folic acid (FA) was developed to enhance tumor accumulation and promote the anti-cancer activity while attenuating adverse effects. Both copper (Cu) and iron (Fe) were utilized in the construction of these submicron scale entities, therefore strong radiosensitization effect is anticipated by implementation of these two metals. The structure-function relationships between constituents of nanomaterials and their function led to the development of nanoscale materials with great radiosensitizing capacity and biosafety. BSA was used to anchor Fe and Cu ions but also to improve colloidal stability, blood circulation time, biocompatibility, and further functionalization. Moreover, to specifically target tumor sites and enhance cellular uptake, FA was conjugated onto the surface of hybrid bimetallic nanoparticles. Finally, CUR as a natural chemotherapeutic agent was encapsulated into the developed bimetallic nanoparticles. With incorporation of all abovementioned stages into one multifunctional nanoplatform, CuFe2O4@BSA-FA-CUR is produced for synergistic chemoradiotherapy with positive outcomes. In vitro investigation revealed that these nanoplatforms bear excellent biosafety, great tumor cell killing ability and radiosensitizing capacity. In addition, high cancer-suppression efficiency was observed through in vivo studies. It is worth mentioning that co-use of CuFe2O4@BSA-FA-CUR nanoplatforms and X-ray radiation led to complete tumor ablation in almost all of the treated mice. No mortality or radiation-induced normal tissue toxicity were observed following administration of CuFe2O4@BSA-FA-CUR nanoparticles which highlights the biosafety of these submicron scale entities. These results offer powerful evidence for the potential capability of CuFe2O4@BSA-FA-CUR in radiosensitization of malignant tumors and opens up a new avenue of research in this area.
Collapse
Affiliation(s)
- Marziyeh Salehiabar
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine
| | | | - Ali Mohammadi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Navid Mousazadeh
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Rahimi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Abhari
- Department of Radiology, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan 45139- 56184, Iran
| | - Hamid Rashidzadeh
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Leila Nasehi
- Department of Medical Laboratory, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamed Rezaeejam
- Department of Radiology, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan 45139- 56184, Iran
| | - Murat Barsbay
- Hacettepe University, Department of Chemistry, Beytepe, Ankara 06800, Türkiye
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Türkiye; Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Türkiye
| | - Hamed Nosrati
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine.
| | - Taras Kavetskyy
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine; Department of Materials Engineering, The John Paul II Catholic University of Lublin, 20-950 Lublin, Poland; Drohobych Ivan Franko State Pedagogical University, 82100 Drohobych, Ukraine.
| | - Hossein Danafar
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine.
| |
Collapse
|
14
|
Improve the cytotoxic effects of megavoltage radiation treatment by Fe3O4@Cus–PEG nanoparticles as a novel radiosensitizer in colorectal cancer cells. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00131-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
To enhance the performance of radiotherapy, emerging nanoparticles that can professionally enhance X-ray irradiation to destruct cancer cells are extremely necessary. Here, we examined the potential of PEG-coated magnetite copper sulfide hetero-nanoparticles (Fe3O4@Cus–PEG) as a radiosensitizer agent.
Methods
Fe3O4@Cus–PEG nanoparticles were synthesized and characterized. The toxicity of nanoparticles on HT-29 colorectal cancer cells was assessed by the MTT assay. The radio-sensitizing effects of Fe3O4@Cus–PEG nanoparticles on HT-29 cancer cells were investigated by the MTT and colony formation assays. Moreover, the underlying mechanisms for Fe3O4@Cus–PEG nanoparticles to improve the radiation sensitivity of cells were evaluated.
Results
The results demonstrated that nanoparticles enhanced the effects of X-ray irradiation in a dose-dependent manner. The effects of combined treatments (nanoparticles and X-ray radiation) were strongly synergistic. The sensitizing enhancement ratio (SER) of nanoparticles was 2.02. Our in vitro assays demonstrated that the nitric oxide production, the intracellular hydrogen peroxide concentration, and the expression level of Bax and Caspase-3 genes significantly increased in the cells treated with the combination of nanoparticles and radiation. Whereas, the Glutathione peroxidase enzyme activity and the expression level of the Bcl-2 gene in the combined treatment significantly decreased compared to the radiation alone.
Conclusions
Our study suggests that Fe3O4@Cus–PEG nanoparticles are the promising nano radio-sensitizing agents for the treatment of cancer cells to enhance the efficacy of radiation therapy through increasing the reactive oxygen species generation, nitric oxide production, and inducing apoptosis.
Graphical Abstract
Collapse
|
15
|
Negi S, Chaudhuri A, Kumar DN, Dehari D, Singh S, Agrawal AK. Nanotherapeutics in autophagy: a paradigm shift in cancer treatment. Drug Deliv Transl Res 2022; 12:2589-2612. [PMID: 35149969 DOI: 10.1007/s13346-022-01125-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 12/15/2022]
Abstract
Autophagy is a catabolic process in which an organism responds to its nutrient or metabolic emergencies. It involves the degradation of cytoplasmic proteins and organelles by forming double-membrane vesicles called "autophagosomes." They sequester cargoes, leading them to degradation in the lysosomes. Although autophagy acts as a protective mechanism for maintaining homeostasis through cellular recycling, it is ostensibly a cause of certain cancers, but a cure for others. In other words, insufficient autophagy, due to genetic or cellular dysfunctions, can lead to tumorigenesis. However, many autophagy modulators are developed for cancer therapy. Diverse nanoparticles have been documented to induce autophagy. Also, the highly stable nanoparticles show blockage to autophagic flux. In this review, we revealed a general mechanism by which autophagy can be induced or blocked via nanoparticles as well as several studies recently performed to prove the stated fact. In addition, we have also elucidated the paradoxical roles of autophagy in cancer and how their differential role at different stages of various cancers can affect its treatment outcomes. And finally, we summarize the breakthroughs in cancer disease treatments by using metallic, polymeric, and liposomal nanoparticles as potent autophagy modulators.
Collapse
Affiliation(s)
- Shloka Negi
- Department of Pharmaceutical Eng. & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India
| | - Aiswarya Chaudhuri
- Department of Pharmaceutical Eng. & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Eng. & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India
| | - Deepa Dehari
- Department of Pharmaceutical Eng. & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India
| | - Sanjay Singh
- Department of Pharmaceutical Eng. & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Eng. & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India.
| |
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: 4] [Impact Index Per Article: 2.0] [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
|
Mundekkad D, Cho WC. Mitophagy Induced by Metal Nanoparticles for Cancer Treatment. Pharmaceutics 2022; 14:2275. [PMID: 36365094 PMCID: PMC9699542 DOI: 10.3390/pharmaceutics14112275] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022] Open
Abstract
Research on nanoparticles, especially metal nanoparticles, in cancer therapy is gaining momentum. The versatility and biocompatibility of metal nanoparticles make them ideal for various applications in cancer therapy. They can bring about apoptotic cell death in cancer cells. In addition to apoptosis, nanoparticles mediate a special type of autophagy facilitated through mitochondria called mitophagy. Interestingly, nanoparticles with antioxidant properties are capable of inducing mitophagy by altering the levels of reactive oxygen species and by influencing signaling pathways like PINK/Parkin pathway and P13K/Akt/mTOR pathway. The current review presents various roles of metal nanoparticles in inducing mitophagy in cancer cells. We envision this review sheds some light on the blind spots in the research related to mitophagy induced by nanoparticles for cancer treatment.
Collapse
Affiliation(s)
- Deepa Mundekkad
- Centre for NanoBioTechnology (CNBT), Vellore Institute of Technology, Vellore 632014, India
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| |
Collapse
|
18
|
Li N, Du H, Mao L, Xu G, Zhang M, Fan Y, Dong X, Zheng L, Wang B, Qin X, Jiang X, Chen C, Zou Z, Zhang J. Reciprocal regulation of NRF2 by autophagy and ubiquitin-proteasome modulates vascular endothelial injury induced by copper oxide nanoparticles. J Nanobiotechnology 2022; 20:270. [PMID: 35690781 PMCID: PMC9188091 DOI: 10.1186/s12951-022-01486-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/01/2022] [Indexed: 12/30/2022] Open
Abstract
NRF2 is the key antioxidant molecule to maintain redox homeostasis, however the intrinsic mechanisms of NRF2 activation in the context of nanoparticles (NPs) exposure remain unclear. In this study, we revealed that copper oxide NPs (CuONPs) exposure activated NRF2 pathway in vascular endothelial cells. NRF2 knockout remarkably aggravated oxidative stress, which were remarkably mitigated by ROS scavenger. We also demonstrated that KEAP1 (the negative regulator of NRF2) was not primarily involved in NRF2 activation in that KEAP1 knockdown did not significantly affect CuONPs-induced NRF2 activation. Notably, we demonstrated that autophagy promoted NRF2 activation as evidenced by that ATG5 knockout or autophagy inhibitors significantly blocked NRF2 pathway. Mechanically, CuONPs disturbed ubiquitin–proteasome pathway and consequently inhibited the proteasome-dependent degradation of NRF2. However, autophagy deficiency reciprocally promoted proteasome activity, leading to the acceleration of degradation of NRF2 via ubiquitin–proteasome pathway. In addition, the notion that the reciprocal regulation of NRF2 by autophagy and ubiquitin–proteasome was further proven in a CuONPs pulmonary exposure mice model. Together, this study uncovers a novel regulatory mechanism of NRF2 activation by protein degradation machineries in response to CuONPs exposure, which opens a novel intriguing scenario to uncover therapeutic strategies against NPs-induced vascular injury and disease. CuONPs exposure activates NRF2 signaling in vascular endothelial cells and mouse thoracic aorta. KEAP1 is dispensable for NRF2 activation in CuONPs-treated vascular endothelial cells. CuONPs-induced autophagy facilitates NRF2 activation in vascular endothelial cells and mouse thoracic aorta. Autophagy and ubiquitin–proteasome reciprocally regulate NRF2 activation in CuONPs-treated vascular endothelial cells and mouse thoracic aorta.
Collapse
Affiliation(s)
- Na Li
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hang Du
- Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing, 400060, People's Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Ge Xu
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Mengling Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yinzhen Fan
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaomei Dong
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lijun Zheng
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Bin Wang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China.,Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China. .,Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China. .,Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
19
|
Wang S, Gao CZ, Liu X, Wu FG, Han X. Long-Chain Poly-d-Lysines Interact with the Plasma Membrane and Induce Protective Autophagy and Intense Cell Necrosis. Bioconjug Chem 2022; 33:938-947. [PMID: 35442635 DOI: 10.1021/acs.bioconjchem.2c00153] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Polylysines have been frequently used in drug delivery and antimicrobial and cell adhesion studies. Because of steric hindrance, chirality plays a major role in the functional difference between poly-l-lysine (PLL) and poly-d-lysine (PDL), especially when they interact with the plasma membranes of mammalian cells. Therefore, it is speculated that the interaction between chiral polylysines and the plasma membrane may cause different cellular behaviors. Here, we carefully investigated the interaction pattern of PLL and PDL with plasma membranes. We found that PDL could be anchored onto the plasma membrane and interact with the membrane lipids, leading to the rapid morphological change and death of A549 cells (a human lung cancer cell line) and HPAEpiCs (a human pulmonary alveolar epithelial cell line). In contrast, PLL exhibited good cytocompatibility and was not anchored onto the plasma membranes of these cells. Unlike PLL, PDL could trigger protective autophagy to prevent cells in a certain degree, and the PDL-caused cell death occurred via intense necrosis (featured by increased intracellular Ca2+ content and plasma membrane disruption). In addition, it was found that the short-chain PDL with a repeat unit number of 9 (termed DL9) could locate in lysosomes and induce autophagy at high concentrations, but it could not elicit drastic cell death, which proved that the repeat unit number of polylysine could affect its cellular action. This research confirms that the interaction between chiral polylysines and the plasma membrane can induce autophagy and intense necrosis, which provides guidance for the future studies of chiral molecules/drugs.
Collapse
Affiliation(s)
- Shujing Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental, Biomedical Engineering Education, Southeast University, Nanjing 210096, China
| | - Cheng-Zhe Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental, Biomedical Engineering Education, Southeast University, Nanjing 210096, China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental, Biomedical Engineering Education, Southeast University, Nanjing 210096, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental, Biomedical Engineering Education, Southeast University, Nanjing 210096, China
| | - Xiaofeng Han
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental, Biomedical Engineering Education, Southeast University, Nanjing 210096, China
| |
Collapse
|
20
|
Paskeh MDA, Entezari M, Clark C, Zabolian A, Ranjbar E, Farahani MV, Saleki H, Sharifzadeh SO, Far FB, Ashrafizadeh M, Samarghandian S, Khan H, Ghavami S, Zarrabi A, Łos MJ. Targeted regulation of autophagy using nanoparticles: New insight into cancer therapy. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166326. [DOI: 10.1016/j.bbadis.2021.166326] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/31/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022]
|
21
|
Wang X, Li Y, Lu J, Deng X, Wu Y. Engineering Nanoplatform for Combined Cancer Therapeutics via Complementary Autophagy Inhibition. Int J Mol Sci 2022; 23:657. [PMID: 35054843 PMCID: PMC8776236 DOI: 10.3390/ijms23020657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 12/21/2022] Open
Abstract
Despite advances in the development of tumor treatments, mortality from cancer continues to increase. Nanotechnology is expected to provide an innovative anti-cancer therapy, to combat challenges such as multidrug resistance and tumor recurrence. Nevertheless, tumors can greatly rely on autophagy as an alternative source for metabolites, and which desensitizes cancer cells to therapeutic stress, hindering the success of any current treatment paradigm. Autophagy is a conserved process by which cells turn over their own constituents to maintain cellular homeostasis. The multistep autophagic pathway provides potentially druggable targets to inhibit pro-survival autophagy under various therapeutic stimuli. In this review, we focus on autophagy inhibition based on functional nanoplatforms, which may be a potential strategy to increase therapeutic sensitivity in combinational cancer therapies, including chemotherapy, radiotherapy, phototherapy, sonodynamic therapy, and immunotherapy.
Collapse
Affiliation(s)
- Xuan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunhao Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China;
| | - Jianqing Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
| | - Xiongwei Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Hu P, Hou X, Yu X, Wei X, Li Y, Yang D, Jiang X. Folic Acid-Conjugated Gold Nanostars for Computed Tomography Imaging and Photothermal/Radiation Combined Therapy. ACS APPLIED BIO MATERIALS 2021; 4:4862-4871. [PMID: 35007035 DOI: 10.1021/acsabm.1c00171] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Fabrication of multifunctional nanoprobes, which integrate tumor targeting, imaging, and effective treatment, has been widely explored in nanomedicine. In the present study, we fabricated tumor-targeting polymer folic acid-terminated polyethylene glycol thiol-modified gold nanostars (GNS-FA), which could realize X-ray computed tomography (CT) imaging and PTT/RT synergistic therapy. The synthesized GNS-FA exhibited good biocompatibility. GNS-FA could be used as a CT imaging contrast agent due to the strong X-ray attenuation of Au. GNS-FA exhibited good near-infrared (NIR) light absorption and excellent photothermal conversion performance, making them promising photothermal transduction agents (PTAs). Furthermore, GNS-FA could be used as an RT sensitizer to enhance the radio-mediated cell death due to the high atomic number (high Z) of gold. Hence, GNS-FA were used as the CT imaging agent, PTA, and radiosensitizer in this work. The in vitro antitumor experiments showed that the PTT/RT combined treatment had enhanced anticancer efficacy compared with the monotherapy (PTT or RT). Our results indicated that the bioconjugated GNS could offer an excellent nanoplatform for CT imaging-guided PTT/RT combined cancer therapy in the future.
Collapse
Affiliation(s)
- Ping Hu
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Xu Hou
- Department of Hepatobiliary Surgery, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Xiaojun Yu
- Department of Radiotherapy, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Xuguo Wei
- Department of Radiotherapy, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Yang Li
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Xiaohong Jiang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| |
Collapse
|
23
|
Autophagy deficiency exacerbates acute lung injury induced by copper oxide nanoparticles. J Nanobiotechnology 2021; 19:162. [PMID: 34059066 PMCID: PMC8166141 DOI: 10.1186/s12951-021-00909-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/24/2021] [Indexed: 12/31/2022] Open
Abstract
Copper oxide nanoparticles (CuONPs) are one of the widely used metal nanoparticles in the industrial and commercial fields. Autophagy is an intracellular degradation system that delivers cytoplasmic constituents to the lysosome and has been linked to nanoparticles-induced toxicity. In particular, the roles of autophagy in response to CuONPs have been explored in vitro, although the conclusions are controversial. To clarify the role of autophagy in CuONPs-induced acute lung injury, microtubule-associated protein 1 light chain 3 beta (Map1lc3b or lc3b) knockout mice and their corresponding wild type mice are applied. Our results showed that single-dose intratracheal instillation of CuONPs with dosages of 1.25, 2.5 or 5 mg/kg caused acute lung injury 3 days after treatment in a dose-dependent manner, as evidenced by deteriorative lung histopathology, more infiltration of macrophage cells, increased oxidative stress and copper ions. Loss of lc3b resulted in aggravated lung injury induced by CuONPs, which was probably due to the blockade of mitophagy and consequently the accumulation of aberrant mitochondria with overloaded copper ions. Our study provides the first in vivo evidence that autophagy deficiency exacerbates CuONPs-induced acute lung injury, and highlights that targeting autophagy is a meaningful strategy against CuONPs-associated respiratory toxicity.
Collapse
|
24
|
Jia S, Ge S, Fan X, Leong KW, Ruan J. Promoting reactive oxygen species generation: a key strategy in nanosensitizer-mediated radiotherapy. Nanomedicine (Lond) 2021; 16:759-778. [PMID: 33856241 DOI: 10.2217/nnm-2020-0448] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The radiotherapy enhancement effect of numerous nanosensitizers is based on the excessive production of reactive oxygen species (ROS), and only a few systematic reviews have focused on the key strategy in nanosensitizer-mediated radiotherapy. To clarify the mechanism underlying this effect, it is necessary to understand the role of ROS in radiosensitization before clinical application. Thus, the source of ROS and their principle of tumor inhibition are first introduced. Then, nanomaterial-mediated ROS generation in radiotherapy is reviewed. The double-edged sword effect of ROS and the potential dangers they may pose to cancer patients are subsequently addressed. Finally, future perspectives regarding ROS-regulated nanosensitizer applications and development are discussed.
Collapse
Affiliation(s)
- Shichong Jia
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Jing Ruan
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China.,Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
25
|
Mohapatra A, Uthaman S, Park IK. External and Internal Stimuli-Responsive Metallic Nanotherapeutics for Enhanced Anticancer Therapy. Front Mol Biosci 2021; 7:597634. [PMID: 33505987 PMCID: PMC7831291 DOI: 10.3389/fmolb.2020.597634] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Therapeutic, diagnostic, and imaging approaches based on nanotechnology offer distinct advantages in cancer treatment. Various nanotherapeutics have been presented as potential alternatives to traditional anticancer therapies such as chemotherapy, radiotherapy, and surgical intervention. Notably, the advantage of nanotherapeutics is mainly attributable to their accumulation and targeting ability toward cancer cells, multiple drug-carrying abilities, combined therapies, and imaging approaches. To date, numerous nanoparticle formulations have been developed for anticancer therapy and among them, metallic nanotherapeutics reportedly demonstrate promising cancer therapeutic and diagnostic efficiencies owing to their dense surface functionalization ability, uniform size distribution, and shape-dependent optical responses, easy and cost-effective synthesis procedure, and multiple anti-cancer effects. Metallic nanotherapeutics can remodel the tumor microenvironment by changing unfavorable therapeutic conditions into therapeutically accessible ones with the help of different stimuli, including light, heat, ultrasound, an alternative magnetic field, redox, and reactive oxygen species. The combination of metallic nanotherapeutics with both external and internal stimuli can be used to trigger the on-demand release of therapeutic molecules, augmenting the therapeutic efficacies of anticancer therapies such as photothermal therapy, photodynamic therapy, magnetic hyperthermia, sonodynamic therapy, chemodynamic therapy, and immunotherapy. In this review, we have summarized the role of different metallic nanotherapeutics in anti-cancer therapy, as well as their combinational effects with multiple stimuli for enhanced anticancer therapy.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeollanam-do, South Korea
| | - Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon, South Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeollanam-do, South Korea
| |
Collapse
|
26
|
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: 2.3] [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
|
27
|
Morita K, Nishimura Y, Nakamura S, Arai Y, Numako C, Sato K, Nakayama M, Akasaka H, Sasaki R, Ogino C, Kondo A. Titanium oxide nano-radiosensitizers for hydrogen peroxide delivery into cancer cells. Colloids Surf B Biointerfaces 2020; 198:111451. [PMID: 33223346 DOI: 10.1016/j.colsurfb.2020.111451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/16/2020] [Accepted: 11/03/2020] [Indexed: 11/18/2022]
Abstract
Polyacrylic acid-modified titanium peroxide nanoparticles (PAA-TiOx NPs) are promising radiosensitizers that enhance the therapeutic effect of X-ray irradiation after local injection into tumors. However, the mechanism for this reaction has remained unclear with the exception of the involvement of hydrogen peroxide (H2O2), which is released by PAA-TiOx NPs to a liquid phase during dispersion. In the present study, a clonogenic assay was used to compare PAA-TiOx NPs with free H2O2 molecules to investigate the effect exerted on the radiosensitivity of cancer cells in vitro. A cell-free dialysis method revealed that a portion of the H2O2 adsorbed onto the PAA-TiOx NPs during synthesis could be released during a treatment regimen. The H2O2 release lasted for 7 h, which was sufficient for one radiation treatment procedure. For in vitro experiments, cultured human pancreatic cancer cells took up PAA-TiOx NPs in 10 min after administration. Interestingly, when the cells were washed with a buffer after treatment with either a PAA-TiOx NP or H2O2 solution, the intracellular H2O2 levels remained higher with PAA-TiOx NP treatment compared with the H2O2 solution treatment. Furthermore, the effects of subsequent X-ray irradiation corresponded to the intracellular H2O2 levels. These results indicate that PAA-TiOx NPs are efficient carriers of H2O2 into cancer cells and thus enhance the radiosensitivity.
Collapse
Affiliation(s)
- Kenta Morita
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan; Research Facility Center for Science and Technology, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| | - Yuya Nishimura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| | - Satoko Nakamura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| | - Yuki Arai
- Graduate School of Science, Chiba University, 1-33, Yayoi, Inage, Chiba, 263-8522, Japan
| | - Chiya Numako
- Graduate School of Science, Chiba University, 1-33, Yayoi, Inage, Chiba, 263-8522, Japan
| | - Kazuyoshi Sato
- Division of Environmental Engineering Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjincho, Kiryu, Gunma, 376-8515, Japan
| | - Masao Nakayama
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-2, Chuou-ku, Kobe 650-0017, Japan
| | - Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-2, Chuou-ku, Kobe 650-0017, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-2, Chuou-ku, Kobe 650-0017, Japan
| | - Chiaki Ogino
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan.
| | - Akihiko Kondo
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan; Organization of Advanced Science and Technology, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
28
|
Meidanchi A. Mg (1-x)Cu xFe 2O 4 superparamagnetic nanoparticles as nano-radiosensitizer agents in radiotherapy of MCF-7 human breast cancer cells. NANOTECHNOLOGY 2020; 31:325706. [PMID: 32330910 DOI: 10.1088/1361-6528/ab8cf2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Magnesium-doped copper spinel ferrite superparamagnetic nanoparticles (Mg(1-x)CuxFe2O4 SPMNPs, 0.2 ≤ x ≤ 0.8) were successfully synthesized by a hydrothermal method. The cytotoxicity effects and cell viability of MCF-7 on human breast cancer cells pre and post exposure to the Mg1-xCuxFe2O4 SPMNPs at different concentrations of 0.1, 1, 10 and 100 μg ml-1 under radiotherapy were studied by MTT (3-[4,5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide) assay. Here, x-ray diffraction, scanning electron microscopy, atomic force microscopy, UV-visible spectrophotometry, Fourier transform infrared spectroscopy and vibrating-sample magnetometry were evaluated for the crystal structure, morphology, optical and magnetic property of the Mg(1-x)CuxFe2O4 SPMNPs. The results showed that the Mg(1-x)CuxFe2O4 SPMNPs all at x values had no significant cytotoxicity at concentrations of 0.1,1 and 10 μg ml-1, but were enhanced by increasing of Cu content. Furthermore, cell destruction of MCF-7 human breast cancer cells post exposure to Mg(1-x)CuxFe2O4 SPMNPs under x-ray irradiation was enhanced by increasing the Cu content and concentration. Superparamagnetic properties of the Mg(1-x)CuxFe2O4 SPMNPs cause their localization and elimination, by only an external magnetic field. In conclusion, the Mg(1-x)CuxFe2O4 SPMNPs with optimum values of x = 0.2 (10 μg ml-1) and x = 0.6 (1 μg ml-1) can be considered as a nano-radiosensitizer because of the synergistic treatment effect without cytotoxicity on the MCF-7 cells.
Collapse
Affiliation(s)
- Alireza Meidanchi
- Department of Physics, Payame Noor University (PNU), P.O. Box 19395-3697, Tehran, Iran
| |
Collapse
|
29
|
Meidanchi A, Ansari H. Copper Spinel Ferrite Superparamagnetic Nanoparticles as a Novel Radiotherapy Enhancer Effect in Cancer Treatment. J CLUST SCI 2020. [DOI: 10.1007/s10876-020-01832-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Sun H, Wang X, Zhai S. The Rational Design and Biological Mechanisms of Nanoradiosensitizers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E504. [PMID: 32168899 PMCID: PMC7153263 DOI: 10.3390/nano10030504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
Radiotherapy (RT) has been widely used for cancer treatment. However, the intrinsic drawbacks of RT, such as radiotoxicity in normal tissues and tumor radioresistance, promoted the development of radiosensitizers. To date, various kinds of nanoparticles have been found to act as radiosensitizers in cancer radiotherapy. This review focuses on the current state of nanoradiosensitizers, especially the related biological mechanisms, and the key design strategies for generating nanoradiosensitizers. The regulation of oxidative stress, DNA damage, the cell cycle, autophagy and apoptosis by nanoradiosensitizers in vitro and in vivo is highlighted, which may guide the rational design of therapeutics for tumor radiosensitization.
Collapse
Affiliation(s)
- Hainan Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
- Shandong Vocational College of Light Industry, Zibo 255300, Shandong, China
| | - Xiaoling Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| |
Collapse
|
31
|
Howard D, Sebastian S, Le QVC, Thierry B, Kempson I. Chemical Mechanisms of Nanoparticle Radiosensitization and Radioprotection: A Review of Structure-Function Relationships Influencing Reactive Oxygen Species. Int J Mol Sci 2020; 21:E579. [PMID: 31963205 PMCID: PMC7013516 DOI: 10.3390/ijms21020579] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 01/19/2023] Open
Abstract
Metal nanoparticles are of increasing interest with respect to radiosensitization. The physical mechanisms of dose enhancement from X-rays interacting with nanoparticles has been well described theoretically, however have been insufficient in adequately explaining radiobiological response. Further confounding experimental observations is examples of radioprotection. Consequently, other mechanisms have gained increasing attention, especially via enhanced production of reactive oxygen species (ROS) leading to chemical-based mechanisms. Despite the large number of variables differing between published studies, a consensus identifies ROS-related mechanisms as being of significant importance. Understanding the structure-function relationship in enhancing ROS generation will guide optimization of metal nanoparticle radiosensitisers with respect to maximizing oxidative damage to cancer cells. This review highlights the physico-chemical mechanisms involved in enhancing ROS, commonly used assays and experimental considerations, variables involved in enhancing ROS generation and damage to cells and identifies current gaps in the literature that deserve attention. ROS generation and the radiobiological effects are shown to be highly complex with respect to nanoparticle physico-chemical properties and their fate within cells. There are a number of potential biological targets impacted by enhancing, or scavenging, ROS which add significant complexity to directly linking specific nanoparticle properties to a macroscale radiobiological result.
Collapse
Affiliation(s)
| | | | | | | | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia; (D.H.); (B.T.)
| |
Collapse
|
32
|
Nosrati H, Charmi J, Abhari F, Attari E, Bochani S, Johari B, Rezaeejam H, Kheiri Manjili H, Davaran S, Danafar H. Improved synergic therapeutic effects of chemoradiation therapy with the aid of a co-drug-loaded nano-radiosensitizer under conventional-dose X-ray irradiation. Biomater Sci 2020; 8:4275-4286. [DOI: 10.1039/d0bm00353k] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The goal of this work is to harness the advantages of a targeted hybrid nanostructure, BSA-coated Fe3O4 (F)-Au heterodimer, as a radiosensitizer and co-delivery vehicle of chemotherapeutic drugs for enhanced synergic cancer therapy and protection of healthy tissues.
Collapse
Affiliation(s)
- Hamed Nosrati
- Zanjan Pharmaceutical Nanotechnology Research Center
- Zanjan University of Medical Sciences
- Zanjan
- Iran
- Department of pharmaceutical biomaterials
| | - Jalil Charmi
- Zanjan Pharmaceutical Nanotechnology Research Center
- Zanjan University of Medical Sciences
- Zanjan
- Iran
| | - Fatemeh Abhari
- Faculty of Medicine
- Department of Medical Physics
- Tabriz University of Medical Sciences
- Tabriz
- Iran
| | - Elahe Attari
- Zanjan Pharmaceutical Nanotechnology Research Center
- Zanjan University of Medical Sciences
- Zanjan
- Iran
| | - Shayesteh Bochani
- Zanjan Pharmaceutical Nanotechnology Research Center
- Zanjan University of Medical Sciences
- Zanjan
- Iran
| | - Behrooz Johari
- Department of Medical Biotechnology
- School of Medicine
- Zanjan University of Medical Sciences
- Zanjan
- Iran
| | - Hamed Rezaeejam
- Department of Radiology
- School of Paramedical and Health
- Zanjan University of Medical Sciences
- Zanjan
- Iran
| | - Hamidreza Kheiri Manjili
- Zanjan Pharmaceutical Nanotechnology Research Center
- Zanjan University of Medical Sciences
- Zanjan
- Iran
| | - Soodabeh Davaran
- Drug Applied Research Center
- Tabriz University of Medical Sciences
- Tabriz
- Iran
| | - Hossein Danafar
- Zanjan Pharmaceutical Nanotechnology Research Center
- Zanjan University of Medical Sciences
- Zanjan
- Iran
- Department of pharmaceutical biomaterials
| |
Collapse
|
33
|
Guerreiro A, Chatterton N, Crabb EM, Golding JP. A comparison of the radiosensitisation ability of 22 different element metal oxide nanoparticles using clinical megavoltage X-rays. Cancer Nanotechnol 2019. [DOI: 10.1186/s12645-019-0057-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
A wide range of nanoparticles (NPs), composed of different elements and their compounds, are being developed by several groups as possible radiosensitisers, with some already in clinical trials. However, no systematic experimental survey of the clinical X-ray radiosensitising potential of different element nanoparticles has been made. Here, we directly compare the irradiation-induced (10 Gy of 6-MV X-ray photon) production of hydroxyl radicals, superoxide anion radicals and singlet oxygen in aqueous solutions of the following metal oxide nanoparticles: Al2O3, SiO2, Sc2O3, TiO2, V2O5, Cr2O3, MnO2, Fe3O4, CoO, NiO, CuO, ZnO, ZrO2, MoO3, Nd2O3, Sm2O3, Eu2O3, Gd2O3, Tb4O7, Dy2O3, Er2O3 and HfO2. We also examine DNA damage due to these NPs in unirradiated and irradiated conditions.
Results
Without any X-rays, several NPs produced more radicals than water alone. Thus, V2O5 NPs produced around 5-times more hydroxyl radicals and superoxide radicals. MnO2 NPs produced around 10-times more superoxide anions and Tb4O7 produced around 3-times more singlet oxygen. Lanthanides produce fewer hydroxyl radicals than water. Following irradiation, V2O5 NPs produced nearly 10-times more hydroxyl radicals than water. Changes in radical concentrations were determined by subtracting unirradiated values from irradiated values. These were then compared with irradiation-induced changes in water only. Irradiation-specific increases in hydroxyl radical were seen with most NPs, but these were only significantly above the values of water for V2O5, while the Lanthanides showed irradiation-specific decreases in hydroxyl radical, compared to water. Only TiO2 showed a trend of irradiation-specific increase in superoxides, while V2O5, MnO2, CoO, CuO, MoO3 and Tb4O7 all demonstrated significant irradiation-specific decreases in superoxide, compared to water. No irradiation-specific increases in singlet oxygen were seen, but V2O5, NiO, CuO, MoO3 and the lanthanides demonstrated irradiation-specific decreases in singlet oxygen, compared to water. MoO3 and CuO produced DNA damage in the absence of radiation, while the highest irradiation-specific DNA damage was observed with CuO. In contrast, MnO2, Fe3O4 and CoO were slightly protective against irradiation-induced DNA damage.
Conclusions
Beyond identifying promising metal oxide NP radiosensitisers and radioprotectors, our broad comparisons reveal unexpected differences that suggest the surface chemistry of NP radiosensitisers is an important criterion for their success.
Collapse
|
34
|
Cheng X, Zhang X, Liu P, Xia LY, Jiang YW, Gao G, Wang HY, Li YH, Ma N, Ran HH, Wu FG. Sequential Treatment of Cell Cycle Regulator and Nanoradiosensitizer Achieves Enhanced Radiotherapeutic Outcome. ACS APPLIED BIO MATERIALS 2019; 2:2050-2059. [DOI: 10.1021/acsabm.9b00085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
35
|
Martínez-Rovira I, Seksek O, Puxeu J, Gómez J, Kreuzer M, Dučić T, Ferreres MJ, Artigues M, Yousef I. Synchrotron-based infrared microspectroscopy study on the radiosensitization effects of Gd nanoparticles at megavoltage radiation energies. Analyst 2019; 144:5511-5520. [DOI: 10.1039/c9an00792j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Synchrotron-based infrared microspectroscopy is a powerful technique for disentangling biochemical effects in nanoparticle-based radiotherapy approaches.
Collapse
Affiliation(s)
- Immaculada Martínez-Rovira
- ALBA-CELLS Synchrotron
- MIRAS Beamline
- 09290 Cerdanyola del Vallès
- Spain
- Ionizing Radiation Research Group (GRRI)
| | - Olivier Seksek
- Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC)
- CNRS, Univ Paris Sud
- Université Paris-Saclay
- F-91400 Orsay
- France
| | - Josep Puxeu
- Hospital Universitari Sant Joan de Reus
- Institut d'Investigació Sanitària Pere Virgili
- 43204 Reus
- Spain
| | - Joan Gómez
- Ionizing Radiation Research Group (GRRI)
- Physics Department
- Universitat Autònoma de Barcelona (UAB)
- 08193 Cerdanyola del Vallès
- Spain
| | - Martin Kreuzer
- ALBA-CELLS Synchrotron
- MIRAS Beamline
- 09290 Cerdanyola del Vallès
- Spain
| | - Tanja Dučić
- ALBA-CELLS Synchrotron
- MIRAS Beamline
- 09290 Cerdanyola del Vallès
- Spain
| | | | - Manel Artigues
- Hospital Universitari Sant Joan de Reus
- Institut d'Investigació Sanitària Pere Virgili
- 43204 Reus
- Spain
| | - Ibraheem Yousef
- ALBA-CELLS Synchrotron
- MIRAS Beamline
- 09290 Cerdanyola del Vallès
- Spain
| |
Collapse
|