1
|
Taheri A, Khandaker MU, Moradi F, Bradley DA. A simulation study on the radiosensitization properties of gold nanorods. Phys Med Biol 2024; 69:045029. [PMID: 38286017 DOI: 10.1088/1361-6560/ad2380] [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: 09/14/2023] [Accepted: 01/29/2024] [Indexed: 01/31/2024]
Abstract
Objective. Gold nanorods (GNRs) have emerged as versatile nanoparticles with unique properties, holding promise in various modalities of cancer treatment through drug delivery and photothermal therapy. In the rapidly evolving field of nanoparticle radiosensitization (NPRS) for cancer therapy, this study assessed the potential of gold nanorods as radiosensitizing agents by quantifying the key features of NPRS, such as secondary electron emission and dose enhancement, using Monte Carlo simulations.Approach. Employing the TOPAS track structure code, we conducted a comprehensive evaluation of the radiosensitization behavior of spherical gold nanoparticles and gold nanorods. We systematically explored the impact of nanorod geometry (in particular size and aspect ratio) and orientation on secondary electron emission and deposited energy ratio, providing validated results against previously published simulations.Main results. Our findings demonstrate that gold nanorods exhibit comparable secondary electron emission to their spherical counterparts. Notably, nanorods with smaller surface-area-to-volume ratios (SA:V) and alignment with the incident photon beam proved to be more efficient radiosensitizing agents, showing superiority in emitted electron fluence. However, in the microscale, the deposited energy ratio (DER) was not markedly influenced by the SA:V of the nanorod. Additionally, our findings revealed that the geometry of gold nanoparticles has a more significant impact on the emission of M-shell Auger electrons (with energies below 3.5 keV) than on higher-energy electrons.Significance. This research investigated the radiosensitization properties of gold nanorods, positioning them as promising alternatives to the more conventionally studied spherical gold nanoparticles in the context of cancer research. With increasing interest in multimodal cancer therapy, our findings have the potential to contribute valuable insights into the perspective of gold nanorods as effective multipurpose agents for synergistic photothermal therapy and radiotherapy. Future directions may involve exploring alternative metallic nanorods as well as further optimizing the geometry and coating materials, opening new possibilities for more effective cancer treatments.
Collapse
Affiliation(s)
- Ali Taheri
- Applied Physics and Radiation Technologies Group, CCDCU, School of Engineering and Technology, Sunway University, 47500 Bandar Sunway, Selangor, Malaysia
| | - Mayeen Uddin Khandaker
- Applied Physics and Radiation Technologies Group, CCDCU, School of Engineering and Technology, Sunway University, 47500 Bandar Sunway, Selangor, Malaysia
- Faculty of Graduate Studies, Daffodil International University, Daffodil Smart City, Birulia, Savar, Dhaka 1216, Bangladesh
| | - Farhad Moradi
- Fibre Optics Research Centre, Faculty of Engineering, Multimedia University, Jalan Multimedia 63100, Cyberjaya, Malaysia
| | - David Andrew Bradley
- Applied Physics and Radiation Technologies Group, CCDCU, School of Engineering and Technology, Sunway University, 47500 Bandar Sunway, Selangor, Malaysia
- School of Mathematics and Physics, University of Surrey, Guildford, GU2 7XH, United Kingdom
| |
Collapse
|
2
|
Volpini C, Bloise N, Dominoni M, Barra F, Vellone VG, Minzioni P, Gardella B, Ferrero S, Visai L. The nano-revolution in the diagnosis and treatment of endometriosis. NANOSCALE 2023; 15:17313-17325. [PMID: 37874212 DOI: 10.1039/d3nr03527a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Endometriosis is a painful gynecological disease with a high prevalence, affecting millions of women worldwide. Innovative, non-invasive treatments, and new patient follow-up strategies are needed to deal with the harmful social and economic effects. In this scenario, considering the recent, very promising results already reported in the literature, a commitment to new research in the field of nanomedicine is urgently needed. Study findings clearly show the potential of this approach in both the diagnostic and therapeutic phases of endometriosis. Here, we offer a brief review of the recent exciting and effective applications of nanomedicine in both the diagnosis and therapy of endometriosis. Special emphasis will be placed on the emerging theranostic application of nanoproducts, and the combination of phototherapy and nanotechnology as new therapeutic modalities for endometriosis. The review will also provide interested readers with a guide to the selection process and parameters to consider when designing research into this type of approach.
Collapse
Affiliation(s)
- Cristina Volpini
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy.
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, Pavia, Italy
- Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), University of Pavia Unit, Italy
| | - Nora Bloise
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy.
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, Pavia, Italy
- Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), University of Pavia Unit, Italy
| | - Mattia Dominoni
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy.
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fabio Barra
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Valerio Gaetano Vellone
- Anatomia Patologica Universitaria, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate (DISC), Università di Genova, Italy
| | - Paolo Minzioni
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100 Pavia, Italy
| | - Barbara Gardella
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy.
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Simone Ferrero
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
- DINOGMI, University of Genova, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy.
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, Pavia, Italy
- Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), University of Pavia Unit, Italy
| |
Collapse
|
3
|
Tavangari Z, Asadi M, Irajirad R, Sarikhani A, Alamzadeh Z, Ghaznavi H, Khoei S. 3D modeling of in vivo MRI-guided nano-photothermal therapy mediated by magneto-plasmonic nanohybrids. Biomed Eng Online 2023; 22:77. [PMID: 37528482 PMCID: PMC10394893 DOI: 10.1186/s12938-023-01131-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Nano-photothermal therapy (NPTT) has gained wide attention in cancer treatment due to its high efficiency and selective treatment strategy. The biggest challenges in the clinical application are the lack of (i) a reliable platform for mapping the thermal dose and (ii) efficient photothermal agents (PTAs). This study developed a 3D treatment planning for NPTT to reduce the uncertainty of treatment procedures, based on our synthesized nanohybrid. METHODS This study aimed to develop a three-dimensional finite element method (FEM) model for in vivo NPTT in mice using magneto-plasmonic nanohybrids, which are complex assemblies of superparamagnetic iron oxide nanoparticles and gold nanorods. The model was based on Pennes' bio-heat equation and utilized a geometrically correct mice whole-body. CT26 colon tumor-bearing BALB/c mice were injected with nanohybrids and imaged using MRI (3 Tesla) before and after injection. MR images were segmented, and STereoLithography (STL) files of mice bodies and nanohybrid distribution in the tumor were established to create a realistic geometry for the model. The accuracy of the temperature predictions was validated by using an infrared (IR) camera. RESULTS The photothermal conversion efficiency of the nanohybrids was experimentally determined to be approximately 30%. The intratumoral (IT) injection group showed the highest temperature increase, with a maximum of 17 °C observed at the hottest point on the surface of the tumor-bearing mice for 300 s of laser exposure at a power density of 1.4 W/cm2. Furthermore, the highest level of tissue damage, with a maximum value of Ω = 0.4, was observed in the IT injection group, as determined through a simulation study. CONCLUSIONS Our synthesized nanohybrid shows potential as an effective agent for MRI-guided NPTT. The developed model accurately predicted temperature distributions and tissue damage in the tumor. However, the current temperature validation method, which relies on limited 2D measurements, may be too lenient. Further refinement is necessary to improve validation. Nevertheless, the presented FEM model holds great promise for clinical NPTT treatment planning.
Collapse
Affiliation(s)
- Zahed Tavangari
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Asadi
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Irajirad
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Samideh Khoei
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Haque M, Shakil MS, Mahmud KM. The Promise of Nanoparticles-Based Radiotherapy in Cancer Treatment. Cancers (Basel) 2023; 15:cancers15061892. [PMID: 36980778 PMCID: PMC10047050 DOI: 10.3390/cancers15061892] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Radiation has been utilized for a long time for the treatment of cancer patients. However, radiotherapy (RT) has many constraints, among which non-selectivity is the primary one. The implementation of nanoparticles (NPs) with RT not only localizes radiation in targeted tissue but also provides significant tumoricidal effect(s) compared to radiation alone. NPs can be functionalized with both biomolecules and therapeutic agents, and their combination significantly reduces the side effects of RT. NP-based RT destroys cancer cells through multiple mechanisms, including ROS generation, which in turn damages DNA and other cellular organelles, inhibiting of the DNA double-strand damage-repair system, obstructing of the cell cycle, regulating of the tumor microenvironment, and killing of cancer stem cells. Furthermore, such combined treatments overcome radioresistance and drug resistance to chemotherapy. Additionally, NP-based RT in combined treatments have shown synergistic therapeutic benefit(s) and enhanced the therapeutic window. Furthermore, a combination of phototherapy, i.e., photodynamic therapy and photothermal therapy with NP-based RT, not only reduces phototoxicity but also offers excellent therapeutic benefits. Moreover, using NPs with RT has shown promise in cancer treatment and shown excellent therapeutic outcomes in clinical trials. Therefore, extensive research in this field will pave the way toward improved RT in cancer treatment.
Collapse
Affiliation(s)
- Munima Haque
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Md Salman Shakil
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Kazi Mustafa Mahmud
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| |
Collapse
|
5
|
Mousavi M, Koosha F, Neshastehriz A. Chemo-radiation therapy of U87-MG glioblastoma cells using SPIO@AuNP-Cisplatin-Alginate nanocomplex. Heliyon 2023; 9:e13847. [PMID: 36873545 PMCID: PMC9976303 DOI: 10.1016/j.heliyon.2023.e13847] [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: 06/01/2022] [Revised: 01/21/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Megavoltage radiotherapy and cisplatin-based chemotherapy are the primary glioblastoma treatments. Novel nanoparticles have been designed to reduce adverse effects and boost therapeutic effectiveness. In the present study, we synthesized the SPIO@AuNP-Cisplatin-Alginate (SACA) nanocomplex, composed of a SPIO core, a gold shell, and an alginate coating. SACA was characterized using transmission electron microscopy (TEM) and dynamic light scattering (DLS). U87-MG human glioblastoma cells and the HGF cell line (a healthy primary gingival fibroblast) were treated in multiple groups by a combination of SACA, cisplatin, and 6 MV X-ray. The MTT assay was used to assess the cytotoxicity of cisplatin and SACA (at various concentrations and for 4 h). Following the treatments, apoptosis and cell viability were evaluated in each treatment group using flow cytometry and the MTT assay, respectively. The findings demonstrated that the combination of SACA and 6 MV X-rays (at the doses of 2 and 4 Gy) drastically decreased the viability of U87MG cells, whereas the viability of HGF cells remained unchanged. Moreover, U87MG cells treated with SACA in combination with radiation exhibited a significant increase in apoptosis, demonstrating that this nanocomplex effectively boosted the radiosensitivity of cancer cells. Even though additional in vivo studies are needed, these findings suggest that SACA might be used as a radiosensitizer nanoparticle in the therapy of brain tumors.
Collapse
Affiliation(s)
- Mahdie Mousavi
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Radiation Science Department, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Fereshteh Koosha
- Department of Radiology Technology, school of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Neshastehriz
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Radiation Science Department, Iran University of Medical Science (IUMS), Tehran, Iran
| |
Collapse
|
6
|
Zheng W, Li X, Zou H, Xu Y, Li P, Zhou X, Wu M. Dual-Target Multifunctional Superparamagnetic Cationic Nanoliposomes for Multimodal Imaging-Guided Synergistic Photothermal/Photodynamic Therapy of Retinoblastoma. Int J Nanomedicine 2022; 17:3217-3237. [PMID: 35924259 PMCID: PMC9339948 DOI: 10.2147/ijn.s364264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/16/2022] [Indexed: 11/23/2022] Open
Abstract
Background With high malignancy, retinoblastoma (RB) commonly occurs in infants and has incredible difficulty with the early diagnosis. In recent years, the integrated theranostics of multimodal imaging-guided therapy has shown promising potential for oncotherapy. Purpose To prepare folate/magnetic dual-target theranostic nanoparticles integrating with US/PA/MR imaging and the synergistic photothermal treatment (PTT)/photodynamic treatment (PDT) for the early diagnosis and timely intervention of RB cancer. Methods Folate/magnetic dual-target cationic nanoliposomes (CN) encapsulating indocyanine green (ICG) and perfluorohexane(PFH)(FA-CN-PFH-ICG-Fe3O4, FCNPIFE) were synthesized and characterized. Then we evaluated their targeting ability, US/PA/MR imaging effects, and the efficacy of synergistic PTT/PDT in vitro and in vivo. Finally, we explored the mechanism of synergistic PTT/PDT in Y79 tumor-bearing mice. Results FCNPIFEs were stable and uniform in 7 days. They showed excellent in vitro targeting ability with a 95.29% cell uptake rate. The in vitro US/PA/MRI imaging results of FCNPIFEs showed a concentration-dependent manner, and in vitro therapy FCNPIFEs exhibited an enhanced anticancer efficacy against Y79 cells. In vivo analysis confirmed that FCNPIFEs enabled a targeted synergistic PTT/PDT under US/PA/MR imaging guidance in Y79 tumor-bearing mice, achieving almost complete tumor regression. Immunofluorescence results displayed weaker fluorescence intensity compared with other single treatment groups, confirming that PTT/PDT synergistic therapy effect was achieved by down-regulating the expression of HIF-1α and HSP70. Conclusion FCNPIFEs were verified as promising theranostic nanoliposomes for RB oncotherapy and showed great potential in clinical application.
Collapse
Affiliation(s)
- Wendi Zheng
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xing Li
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hongmi Zou
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yan Xu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiyuan Zhou
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Mingxing Wu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Correspondence: Mingxing Wu; Xiyuan Zhou, Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, People’s Republic of China, Tel +86 183 2342 5867; +86 139 9628 6679, Email ;
| |
Collapse
|
7
|
Ghaznavi H, Hajinezhad MR, Shirvaliloo M, Shahraki S, Shahraki K, Saravani R, Shirvalilou S, Shahraki O, Nazarlou Z, Sheervalilou R, Sargazi S. Effects of folate-conjugated Fe 2O 3@Au core-shell nanoparticles on oxidative stress markers, DNA damage, and histopathological characteristics: evidence from in vitro and in vivo studies. Med Oncol 2022; 39:122. [PMID: 35716197 DOI: 10.1007/s12032-022-01713-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/16/2022] [Indexed: 11/30/2022]
Abstract
The aim of this work was to assess the cytotoxicity, genotoxicity, and histopathological effects of Fe2O3@Au-FA NPs using in vitro and in vivo models. Cytotoxicity and cellular uptake of nanoparticles (NPs) by HUVECs were examined via 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and inductively coupled plasma-mass-spectrometry (ICP-MS). This safe dose was then used for cytotoxicity assays, including total protein, total antioxidant capacity, lipid peroxidation, cell membrane integrity, reactive oxygen species, enzyme activity, and DNA damage. In the animal model, 32 Wistar rats were randomly categorized into 4 groups and received intraperitoneal injections of NPs. Blood samples for biochemical properties and histopathological changes were investigated. MTT results indicated 20 μg/ml as the safe dose for NPs. According to ICP-MS, treated cells showed significantly higher levels of the intracellular content of Fe (p < 0.001) and Au (p < 0.01) compared with the control group. In vitro tests did not show any significant cytotoxicity or genotoxicity at the safe dose of NPs. We found no significant elevation in intracellular γ-H2AX levels after treatment of HUVEC cells with Fe2O3@Au core-shell NPs (P > 0.05). As for the in vivo analysis, we observed no marked difference in serum biochemical parameters of rats treated with 50 mg/kg and 100 mg/kg doses of our NPs. Histopathological assessments indicated that liver, kidney, and testis tissues were not significantly affected at 50 mg/kg (liver), 50 mg/kg, and 100 mg/kg (kidney and testis) on NPs administration. These findings imply that the nanotoxicity of Fe2O3@Au-FA NPs in HUVECs and animals depends largely on the administrated dose. Our study suggests that Fe2O3@Au-FA NPs at a safe dose could be considered as new candidates in nanobiomedicine.
Collapse
Affiliation(s)
- Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran
| | - Mohammad Reza Hajinezhad
- Basic Veterinary Science Department, Veterinary medicine Faculty, University of Zabol, Postal Code: 9861335856, Zabol, Iran
| | - Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Postal Code: 5166614766, Tabriz, Iran
| | - Sheida Shahraki
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran
| | - Kourosh Shahraki
- Noor Ophthalmology Research Center, Noor Eye Hospital, Tehran, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran
| | - Sakine Shirvalilou
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Postal Code: 1449614535, Tehran, Iran
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran
| | - Ziba Nazarlou
- Material Engineering Department, College of Science Koç University, Istanbul, 34450, 1449614535, Turkey
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran. .,Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Postal Code: 9816743463, Zahedan, Iran.
| |
Collapse
|
8
|
Lin Y, Qiu T, Lan Y, Li Z, Wang X, Zhou M, Li Q, Li Y, Liang J, Zhang J. Multi-Modal Optical Imaging and Combined Phototherapy of Nasopharyngeal Carcinoma Based on a Nanoplatform. Int J Nanomedicine 2022; 17:2435-2446. [PMID: 35656166 PMCID: PMC9151321 DOI: 10.2147/ijn.s357493] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/11/2022] [Indexed: 11/23/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignant tumor of the head and neck with a high incidence rate worldwide, especially in southern China. Phototheranostics in combination with nanoparticles is an integrated strategy for enabling simultaneous diagnosis, real-time monitoring, and administration of precision therapy for nasopharyngeal carcinoma (NPC). It has shown great potential in the field of cancer diagnosis and treatment owing to its unique noninvasive advantages. Many Chinese and international research teams have applied nano-targeted drugs to optical diagnosis and treatment technology to conduct multimodal imaging and collaborative treatment of NPC, which has become a hot research topic. In this review, we aimed to introduce the recent developments in phototheranostics of NPC based on a nanoplatform. This study aimed to elaborate on the applications of nanoplatform-based optical imaging strategies and treatment modalities, including fluorescence imaging, photoacoustic imaging, Raman spectroscopy imaging, photodynamic therapy, and photothermal therapy. This study is expected to provide a scientific basis for further research and development of NPC diagnosis and treatment.
Collapse
Affiliation(s)
- Yanping Lin
- Department of Radiology, DongGuan Tungwah Hospital, DongGuan, Guangdong, 523000, People's Republic of China
| | - Ting Qiu
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, People's Republic of China
| | - Yintao Lan
- Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China
| | - Zhaoyong Li
- Department of Radiology, DongGuan Tungwah Hospital, DongGuan, Guangdong, 523000, People's Republic of China
| | - Xin Wang
- Department of Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511500, People's Republic of China
| | - Mengyu Zhou
- Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China
| | - Qiuyu Li
- Department of Radiology, DongGuan Tungwah Hospital, DongGuan, Guangdong, 523000, People's Republic of China
| | - Yao Li
- Department of Radiology, DongGuan Tungwah Hospital, DongGuan, Guangdong, 523000, People's Republic of China
| | - Junsheng Liang
- Department of Radiology, DongGuan Tungwah Hospital, DongGuan, Guangdong, 523000, People's Republic of China
| | - Jian Zhang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China.,Department of Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511500, People's Republic of China
| |
Collapse
|
9
|
Hao Y, Li X. Deacetylated-poly-N-acetylglucosamine-folic Acid as a Nanocarrier for Delivering miR-196a Inhibitor to Anticancer Activity. Balkan Med J 2022; 39:55-65. [PMID: 35330543 PMCID: PMC8941238 DOI: 10.4274/balkanmedj.galenos.2021.2021-8-62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
10
|
Khani T, Alamzadeh Z, Sarikhani A, Mousavi M, Mirrahimi M, Tabei M, Irajirad R, Abed Z, Beik J. Fe 3O 4@Au core-shell hybrid nanocomposite for MRI-guided magnetic targeted photo-chemotherapy. Lasers Med Sci 2022; 37:2387-2395. [PMID: 35066676 DOI: 10.1007/s10103-021-03486-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022]
Abstract
The combination of multiple therapeutic and diagnostic functions is fast becoming a key feature in the area of clinical oncology. The advent of nanotechnology promises multifunctional nanoplatforms with the potential to deliver multiple therapeutics while providing diagnostic information simultaneously. In this study, novel iron oxide-gold core-shell hybrid nanocomposites (Fe3O4@Au HNCs) coated with alginate hydrogel carrying doxorubicin (DOX) were constructed for targeted photo-chemotherapy and magnetic resonance imaging (MRI). The magnetic core enables the HNCs to be detected through MRI and targeted towards the tumor using an external magnetic field, a method known as magnetic drug targeting (MDT). The Au shell could respond to light in the near-infrared (NIR) region, generating a localized heating for photothermal therapy (PTT) of the tumor. The cytotoxicity assay showed that the treatment of CT26 colon cancer cells with the DOX-loaded HNCs followed by laser irradiation induced a significantly higher cell death as opposed to PTT and chemotherapy alone. The in vivo MRI study proved MDT to be an effective strategy for targeting the HNCs to the tumor, thereby enhancing their intratumoral concentration. The antitumor study revealed that the HNCs can successfully combine chemotherapy and PTT, resulting in superior therapeutic outcome. Moreover, the use of MDT following the injection of HNCs caused a more extensive tumor shrinkage as compared to non-targeted group. Therefore, the as-prepared HNCs could be a promising nanoplatform for image-guided targeted combination therapy of cancer.
Collapse
Affiliation(s)
- Tahereh Khani
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdie Mousavi
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehri Mirrahimi
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mousa Tabei
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Irajirad
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ziaeddin Abed
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jaber Beik
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Bilynsky C, Millot N, Papa A. Radiation nanosensitizers in cancer therapy-From preclinical discoveries to the outcomes of early clinical trials. Bioeng Transl Med 2022; 7:e10256. [PMID: 35079631 PMCID: PMC8780058 DOI: 10.1002/btm2.10256] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/05/2021] [Accepted: 08/12/2021] [Indexed: 12/31/2022] Open
Abstract
Improving the efficacy and spatial targeting of radiation therapy while sparing surrounding normal tissues has been a guiding principle for its use in cancer therapy. Nanotechnologies have shown considerable growth in terms of innovation and the development of new therapeutic approaches, particularly as radiosensitizers. The aim of this study was to systematically review how nanoparticles (NPs) are used to enhance the radiotherapeutic effect, including preclinical and clinical studies. Clinicaltrials.gov was used to perform the search using the following terms: radiation, cancer, and NPs. In this review, we describe the various designs of nano-radioenhancers, the rationale for using such technology, as well as their chemical and biological effects. Human trials are then discussed with an emphasis on their design and detailed clinical outcomes.
Collapse
Affiliation(s)
- Colette Bilynsky
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Present address:
Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Nadine Millot
- Laboratoire Interdisciplinaire Carnot de BourgogneUMR 6303, CNRS, Université Bourgogne Franche‐ComtéDijon CedexFrance
| | - Anne‐Laure Papa
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
12
|
Xie L, Zhang X, Chu C, Dong Y, Zhang T, Li X, Liu G, Cai W, Han S. Preparation, toxicity reduction and radiation therapy application of gold nanorods. J Nanobiotechnology 2021; 19:454. [PMID: 34963479 PMCID: PMC8715590 DOI: 10.1186/s12951-021-01209-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/14/2021] [Indexed: 12/28/2022] Open
Abstract
Gold nanorods (GNRs) have a broad application prospect in biomedical fields because of their unique properties and controllable surface modification. The element aurum (Au) with high atomic number (high-Z) render GNRs ideal radiosensitive materials for radiation therapy and computed tomography (CT) imaging. Besides, GNRs have the capability of efficiently converting light energy to heat in the near-infrared (NIR) region for photothermal therapy. Although there are more and more researches on GNRs for radiation therapy, how to improve their biocompatibility and how to efficiently utilize them for radiation therapy should be further studied. This review will focuse on the research progress regarding the preparation and toxicity reduction of GNRs, as well as GNRs-mediated radiation therapy.
Collapse
Affiliation(s)
- Lina Xie
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xujia Zhang
- Institute of Medical Engineering, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Chengchao Chu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yingqi Dong
- Institute of Medical Engineering, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Tianzi Zhang
- Institute of Medical Engineering, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyue Li
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, Fujian, China
| | - Wen Cai
- Institute of Medical Engineering, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Suxia Han
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
13
|
Nonlinear optical response of cancer cells following conventional and nano-technology based treatment strategies: Results of chemo-, thermo- and radiation therapies. Photodiagnosis Photodyn Ther 2021; 37:102686. [PMID: 34915185 DOI: 10.1016/j.pdpdt.2021.102686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Although traditional treatments are able to increase cancer survival rate, undesirable impact on off-target tissues are considered a limitation of these approaches. Nanotechnology-based treatments have been proposed as a possible option to enhance targeting., Further,current methods for evaluating cellular damage, are time consuming, highly dependent on the operator skills, and expensive. The aim of this study was to evaluate the capability of nonlinear optical response of cells to determine cellular damages during conventional and nano-technology based treatments. METHODS Three different cancer cell lines, CT26, KB, and MCF-7 were used in this study. The alginate hydrogel co-loaded with cisplatin and Au nanoparticle (ACA) nanocomplex and gold-coated iron oxide nanoparticle (Au@IONP) were considered for chemo- and chemo-photothermal therapies, and thermo-radiation therapy, respectively. The sign and value of nonlinear optical absorption coefficient and imaginary part of the third-order nonlinear susceptibility of cells were computed. MTT assay was utilized as a reference method. RESULTS The value of nonlinear optical indices increased with increasing cellular damage and cell death. The linear regression analysis indicated high correlation between nonlinear optical indices and MTT results, in all treatments. CONCLUSION The nonlinear optical indices are robust from confounding factors, namely treatment approach (traditional and nano-technology based), treatment modality (chemotherapy, thermotherapy, photothermal therapy, and radiation therapy), and cell types. Nonlinear optical properties of cells can be used as a rapid estimation method for cell damage, at the nanoscale level.
Collapse
|
14
|
Yu F, Zhang F. A feasible strategy of fabricating hybrid drugs co-loaded polymer-lipid nanoparticles for the treatment of nasopharyngeal cancer therapy. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
15
|
Lima-Sousa R, Alves CG, Melo BL, Moreira AF, Mendonça AG, Correia IJ, de Melo-Diogo D. Poly(2-ethyl-2-oxazoline) functionalized reduced graphene oxide: Optimization of the reduction process using dopamine and application in cancer photothermal therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112468. [PMID: 34702543 DOI: 10.1016/j.msec.2021.112468] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/10/2021] [Accepted: 09/26/2021] [Indexed: 01/22/2023]
Abstract
The high near infrared (NIR) absorption displayed by reduced graphene oxide (rGO) nanostructures renders them a great potential for application in cancer photothermal therapy. However, the production of this material often relies on the use of hydrazine as a reductant, leading to poor biocompatibility and environmental-related issues. In addition, to improve rGO colloidal stability, this material has been functionalized with poly(ethylene glycol). However, recent studies have reported the immunogenicity of poly(ethylene glycol)-based coatings. In this work, the production of rGO, by using dopamine as the reducing agent, was optimized considering the size distribution and NIR absorption of the attained materials. The obtained results unveiled that the rGO produced by using a 1:5 graphene oxide:dopamine weight ratio and a reaction time of 4 h (termed as DOPA-rGO) displayed the highest NIR absorption while retaining its nanometric size distribution. Subsequently, the DOPA-rGO was functionalized with thiol-terminated poly(2-ethyl-2-oxazoline) (P-DOPA-rGO), revealing suitable physicochemical features, colloidal stability and cytocompatibility. When irradiated with NIR light, the P-DOPA-rGO could produce a temperature increase (ΔT) of 36 °C (75 μg/mL; 808 nm, 1.7 W/cm2, 5 min). The photothermal therapy mediated by P-DOPA-rGO was capable of ablating breast cancer cells monolayers (viability < 3%) and could reduce heterotypic breast cancer spheroids' viability to just 30%. Overall, P-DOPA-rGO holds a great potential for application in breast cancer photothermal therapy.
Collapse
Affiliation(s)
- Rita Lima-Sousa
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Cátia G Alves
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Bruna L Melo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - André F Moreira
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - António G Mendonça
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; Departamento de Química, Universidade da Beira Interior, 6201-001 Covilhã, Portugal
| | - Ilídio J Correia
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, 3030-790 Coimbra, Portugal.
| | - Duarte de Melo-Diogo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
16
|
A general in-situ reduction method to prepare core-shell liquid-metal / metal nanoparticles for photothermally enhanced catalytic cancer therapy. Biomaterials 2021; 277:121125. [PMID: 34534859 DOI: 10.1016/j.biomaterials.2021.121125] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/29/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Gallium indium (GaIn) alloy as a kind of liquid metal (LM) with unique chemical and physical properties has attracted increasing attention for its potential biomedical applications. Herein, a series of core-shell GaIn@Metal (Metal: Pt, Au, Ag, and Cu) heterogeneous nanoparticles (NPs) are obtained by a simple in-situ reduction method. Take core-shell GaIn@Pt NPs for example, the synthesized GaIn@Pt NPs after Pt growth on their surface showed significantly improved photothermal conversion efficiency (PCE) and thermal stability under near-infrared (NIR) II light irradiation. Moreover, the core-shell GaIn@Pt NPs also exhibited good Fenton-like catalytic effect due to the presence of Pt on their surface, and could convert tumor endogenous H2O2 to generate reactive oxygen species (ROS) for cancer cell killing. With biocompatible polyethylene glycol (PEG) modification, such GaIn@Pt-PEG NPs showed efficient tumor homing after intravenous injection, and could lead to effective NIR II triggered photothermal-chemodynamic synergistic therapy of tumors as evidenced in a mouse tumor model. Our work highlights the ingenious use of the chemical properties of metals, providing a rather simple route for the surface engineering of LM-based multifunctional nanoplatforms to achieve a variety of functionalities.
Collapse
|
17
|
Maleki S, Farhadi M, Kamrava SK, Asghari A, Daneshi A. Simulation and In Vitro Experimental Studies on Targeted Photothermal Therapy of Cancer using Folate-PEG-Gold Nanorods. J Biomed Phys Eng 2021; 11:435-446. [PMID: 34458191 PMCID: PMC8385224 DOI: 10.31661/jbpe.v0i0.1108] [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: 01/10/2019] [Accepted: 02/28/2019] [Indexed: 11/16/2022]
Abstract
Background: Selective targeting of malignant cells is the ultimate goal of anticancer studies around the world. There are some modalities for cancer therapy devastating tumor size and growth rate,
meanwhile attacking normal cells. Utilizing appropriate ligands, like folate, allow the delivery of therapeutic molecules to cancer cells selectively. There are a variety of photosensitizers,
like gold nanorods (GNRs), capable of absorbing the energy of light and converting it to heat, evidently build a photothermal procedure for cancer therapy. Objective: To develop a one-step approach for calculating the temperature distribution by solving the heat transfer equation with multiple heat sources originating from NIR laser-exposed GNRs. Material and Methods: In this experimental study, we simulated NIR laser heating process in a single cancer cell, with and without incubation with folate conjugated PEG-GNRs.
This simulation was based on a real TEM image from an experiment with the same setup. An in vitro experiment based on aforesaid scenario was performed to validate the simulated model in practice. Results: According to the simplifications due to computational resource limits, the resulting outcome of simulation showed significant compatibility to the supporting experiment.
Both simulation and experimental studies showed a similar trend for heating and cooling of the cells incubated with GNRs and irradiated by NIR laser (5 min, 1.8 W/cm2).
It was observed that temperature of the cells in microplate reached 53.6 °C when irradiated by laser. Conclusion: This new method can be of great application in developing a planning technique for treating tumors utilizing GNP-mediated thermal therapy.
Collapse
Affiliation(s)
- Shayan Maleki
- PhD, ENT and Head & Neck Research Center and Department, Hazrat Rasoul Hospital, the Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farhadi
- MD, ENT and Head & Neck Research Center and Department, Hazrat Rasoul Hospital, the Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Kamran Kamrava
- MD, ENT and Head & Neck Research Center and Department, Hazrat Rasoul Hospital, the Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Alimohamad Asghari
- MD, Skull Base Research Center, the Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Daneshi
- MD, ENT and Head & Neck Research Center and Department, Hazrat Rasoul Hospital, the Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Lu Y, Peng Z, Zhu D, Jia Y, Taledaohan A, Li Y, Liu J, Wang Y, Wang Y. RGD Peptide and PAD4 Inhibitor-Loaded Gold Nanorods for Chemo-Photothermal Combined Therapy to Inhibit Tumor Growth, Prevent Lung Metastasis and Improve Biosafety. Int J Nanomedicine 2021; 16:5565-5580. [PMID: 34429600 PMCID: PMC8379711 DOI: 10.2147/ijn.s319210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/02/2021] [Indexed: 01/21/2023] Open
Abstract
Purpose A targeted drug delivery system that combines protein-arginine deiminase type-4 (PAD4) inhibitors YW3-56 (356) with PTT of NPs is constructed to both decrease the accumulation of gold in metabolic organs and reduce the dose of chemotherapeutic agents. Patients and Methods In vitro cytotoxicity test and in vivo S180 tumor-bearing mice model were used to compare antitumor activity of 356-modified gold nanospheres and nanorods. The A549 tumor-bearing mice model was also exploited in antitumor assessment. In addition, ICP-MS, blood cell analyzer and blood biochemistry analyzer are applied for assessing the biosafety of NPs. Results Both 356-modified gold nanospheres and nanorods showed antitumor activity. However, 356-loaded gold nanorods are found to have better tumor inhibitory activity than 356-loaded gold nanospheres in the presence of laser and without laser irradiation. Thus, 356-loaded gold nanorods are selected to be applied for chemo-photothermal combined therapy on in vivo. We find that combination therapy could inhibit tumor growth and reduce lung tumor metastasis and inflammatory infiltration compared with individual therapy. It triggers apoptosis in tumor tissue observed by TUNEL assay and TEM pictures. Conclusion Thus, an RGD targeting and PAD4 inhibitor-loaded system are established based on chemo-photothermal combined therapy. It could inhibit tumor growth, prevent lung metastasis and improve biosafety.
Collapse
Affiliation(s)
- Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing, 100069, People's Republic of China
| | - Zidong Peng
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing, 100069, People's Republic of China
| | - Di Zhu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing, 100069, People's Republic of China
| | - Yijiang Jia
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing, 100069, People's Republic of China
| | - Ayijiang Taledaohan
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing, 100069, People's Republic of China
| | - Yuanming Li
- Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Beijing, 100730, People's Republic of China
| | - Jiawang Liu
- Medicinal Chemistry Core, The University of Tennessee Health Science Center, 579 College of Pharmacy Building, Memphis, TN, 38163, USA
| | - Yanming Wang
- School of Life Sciences, Henan University, Kaifeng, 475004, People's Republic of China
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing, 100069, People's Republic of China
| |
Collapse
|
19
|
Rezaeian A, Amini SM, Najafabadi MRH, Farsangi ZJ, Samadian H. Plasmonic hyperthermia or radiofrequency electric field hyperthermia of cancerous cells through green-synthesized curcumin-coated gold nanoparticles. Lasers Med Sci 2021; 37:1333-1341. [PMID: 34406533 DOI: 10.1007/s10103-021-03399-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/06/2021] [Indexed: 10/20/2022]
Abstract
Nanoparticle-mediated hyperthermia is one of the prominent adjuvant therapies which has been faced by many problematic challenges such as efficiency and safety. To compare the nanoparticle-mediated photothermal therapy and radiofrequency electric field hyperthermia, green-synthesized curcumin-coated gold nanoparticles (Cur@AuNPs) were applied in an in vitro study. Using recently published methodologies, each step of the study was performed. Through green chemistry, curcumin was applied as both a reducing and a capping agent in the gold nanoparticle synthesis process. Various techniques were applied for the characterization of the synthesized nanoparticles. The heating rate of Cur@AuNPs in the presence of RFEF or laser irradiation was recorded by using a non-contact thermometer. The cellular uptake of the Cur@AuNPs was studied by ICP-AES. The cellular viability and apoptosis rate of different treatment were measured to investigate the effect of two different nano-hyperthermia techniques on the murine colorectal cancer cell line. The average size of Cur@AuNPs was 7.2 ± 3.3 nm. The stability of the gold nanoparticles in the phosphate buffer saline with and without fetal bovine serum was verified by UV-Vis spectroscopy. FTIR, UV-Vis spectroscopy, and TEM indicate that the stability is a result of phenolic coating on the surface of nanoparticles. Cur@AuNPs can absorb both light and radiofrequency electric field exposure in a way that could kill cancerous cells in a significant number (30% in 64 μg/ml concentration). Green-synthesized Cur@AuNPs could induce apoptosis cell death in photothermal therapy and radiofrequency electric field hyperthermia.
Collapse
Affiliation(s)
- Abbas Rezaeian
- Department of Medical Physics, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Seyed Mohammad Amini
- Radiation Biology Research Center, Iran University of Medical Sciences (IUMS), 14003391769, Tehran, Iran.
| | - Mohammad Reza H Najafabadi
- Medical Nanotechnology Department, School of Advanced Technologies in Medicine, Tran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zohreh Jomeh Farsangi
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Hadi Samadian
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
20
|
Neha Desai, Momin M, Khan T, Gharat S, Ningthoujam RS, Omri A. Metallic nanoparticles as drug delivery system for the treatment of cancer. Expert Opin Drug Deliv 2021; 18:1261-1290. [PMID: 33793359 DOI: 10.1080/17425247.2021.1912008] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The targeted delivery of anticancer agents to tumor is a major challenge because most of the drugs show off-target effect resulting in nonspecific cell death. Multifunctionalized metallic nanoparticles (NPs) are explored as new carrier system in the era of cancer therapeutics. Researchers investigated the potential of metallic NPs to target tumor cells by active and passive mechanisms, thereby reducing off-target effects of anticancer agents. Moreover, photocatalytic activity of upconversion nanoparticles (UCNPs) and the enhanced permeation and retention (EPR) effect have also gained wide potential in cancer treatment. Recent advancement in the field of nanotechnology highlights their potency for cancer therapy. AREAS COVERED This review summarizes the types of gold and silver metallic NPs with targeting mechanisms and their potentiality in cancer therapy. EXPERT OPINION Recent advances in the field of nanotechnology for cancer therapy offer high specificity and targeting efficiency. Targeting tumor cells through mechanistic pathways using metallic NPs for the disruption/alteration of molecular profile and survival rate of the tumor cells has led to an effective approach for cancer therapeutics. This alteration in the survival rate of the tumor cells might decrease the proliferation thereby resulting in more efficient management in the treatment of cancer.
Collapse
Affiliation(s)
- Neha Desai
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Sankalp Gharat
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | | | - Abdelwahab Omri
- The Novel Drug and Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada
| |
Collapse
|
21
|
Huda S, Alam MA, Sharma PK. Smart nanocarriers-based drug delivery for cancer therapy: An innovative and developing strategy. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102018] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Safari A, Sarikhani A, Shahbazi-Gahrouei D, Alamzadeh Z, Beik J, Dezfuli AS, Mahabadi VP, Tohfeh M, Shakeri-Zadeh A. Optimal scheduling of the nanoparticle-mediated cancer photo-thermo-radiotherapy. Photodiagnosis Photodyn Ther 2020; 32:102061. [PMID: 33068822 DOI: 10.1016/j.pdpdt.2020.102061] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 01/18/2023]
Abstract
Maximal synergistic effect between photothermal therapy and radiotherapy (RT) may be achieved when the interval between these two modalities is optimal. In this study, we tried to determine the optimal schedule of the combined regime of RT and nano-photothermal therapy (NPTT), based on the cell cycle distribution and kinetics of cell death. To this end, alginate-coated iron oxide-gold core-shell nanoparticles (Fe3O4@Au/Alg NPs) were synthesized, characterized, and their photo-radio sensitization potency was evaluated on human nasopharyngeal cancer KB cells. Our results demonstrated that synthesized NPs have a good potential in radiotherapy and near-infrared (NIR) photothermal therapy. However, results from flow cytometry analysis indicated that a major portion of KB cells were accumulated in the most radiosensitive phases of cell cycle (G2/M) 24 h after NPTT. Moreover, the maximal synergistic anticancer efficacy (12.3% cell viability) was observed when RT was applied 24 h following the administration of NPTT (NPs [30 μg/mL, 4 h incubation time] + Laser [808 nm, 1 W/cm2, 5 min] + RT [6 Gy]). It is noteworthy that apoptosis was the dominant cell death pathway in the group of cells treated by combination of NPTT and RT. This highly synergistic anticancer efficacy provides a mechanistic basis for Fe3O4@Au/Alg NPs-mediated photothermal therapy combined with RT. Knowing such a basis is helpful to promote novel nanotechnology cancer treatment strategies.
Collapse
Affiliation(s)
- Arash Safari
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Daryoush Shahbazi-Gahrouei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Jaber Beik
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Vahid Pirhajati Mahabadi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Tohfeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ali Shakeri-Zadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Amani S, Mehdizadeh A, Movahedi MM, Keshavarz M, Koosha F. Investigation of the Dose-Enhancement Effects of Spherical and Rod-Shaped Gold Nanoparticles on the HeLa Cell Line. Galen Med J 2020; 9:e1581. [PMID: 34466556 PMCID: PMC8343815 DOI: 10.31661/gmj.v9i0.1581] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 05/31/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023] Open
Abstract
Background: Cervical cancer cells are known as radioresistant cells. Current treatment methods have not improved the patients’ survival efficiently; thus, new therapeutic strategies are needed to enhance the efficacy of radiotherapy. Gold nanomaterials with different shapes and sizes have been explored as radiosensitizers. The present study compared the radiosensitizing effects of gold nanorods (AuNRs) with spherical gold nanoparticles (AuNPs) on the HeLa cell line irradiated with megavoltage X-rays. Materials and Methods: The cytotoxicity of AuNRs and AuNPs on HeLa cells in the presence and absence of 6-MV X-ray was investigated using the MTT assay. For this aim, HeLa cells were incubated with and AuNPs and AuNRs at various concentrations (5, 10, and 15 µg/mL) for 6 hours. Afterward, HeLa cells were irradiated with 6-MV X-ray at a single dose of 2 Gy. Results: The results showed that the addition of AuNRs and AuNPs could enhance the radiosensitivity of HeLa cells. Both AuNRs and AuNPs showed low toxicity on HeLa cells, while AuNRs were more toxic than AuNPs at the examined concentrations. Moreover, it was found that AuNRs could enhance the radiosensitivity of HeLa cells more than spherical-shaped AuNPs. Conclusion: This study revealed that the shape of nanoparticles is an effective factor when they are used as radiosensitizing agents during radiotherapy.
Collapse
Affiliation(s)
- Samad Amani
- Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Mehdizadeh
- Department of Medical Physics and Medical Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Movahedi
- Department of Medical Physics and Medical Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Keshavarz
- Department of Medical Physics and Medical Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Koosha
- Department of Radiology Technology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Correspondence to: Fereshteh Koosha, Ph.D, Assistant Professor, Department of Radiology Technology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Darband St, Ghods Sq., Tehran, Iran Telephone Number: +98-2122717503 Email Address:
| |
Collapse
|
24
|
Gui X, Chen Y, Zhang Z, Lei L, Zhu F, Yang W, Guo Y, Chu M. Fluorescent hollow mesoporous carbon spheres for drug loading and tumor treatment through 980-nm laser and microwave co-irradiation. Biomaterials 2020; 248:120009. [PMID: 32299016 DOI: 10.1016/j.biomaterials.2020.120009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 01/06/2023]
Abstract
Hollow mesoporous particles for drug delivery and cancer therapy have attracted significant attention over recent decades. Here, we develop a simple and highly efficient strategy for preparing fluorescent hollow mesoporous carbon spheres (HMCSs). Compared with typical carbon materials such as fullerene C60, carbon nanotubes, reduced graphene oxide, and carbon nanohorns; HMCSs showed fewer effects on cell cycle distribution and lower toxicity to cells. Ten different drugs were incorporated into the HMCSs, and the maximum loading efficiency reached 42.79 ± 2.7%. Importantly, microwaves were found to improve the photothermal effect generated by HMCSs when combined with 980-nm laser irradiation. The cell killing and tumor growth inhibition efficiencies of HMCSs and drug-loaded HMCSs under co-irradiation with laser and microwaves were significantly improved compared with those under laser irradiation alone. After local administration HMCSs were only distributed in tissue at the injection site. HMCSs showed almost no toxicity in mice after local injection and could be completely removed from the injection site.
Collapse
Affiliation(s)
- Xin Gui
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Yang Chen
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China; Institute of Biophysics, Chinese Academy of Science, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zheyu Zhang
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Longfei Lei
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Fangliang Zhu
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Wenxuan Yang
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Yuliang Guo
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Maoquan Chu
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| |
Collapse
|
25
|
Gold nanoparticles promote a multimodal synergistic cancer therapy strategy by co-delivery of thermo-chemo-radio therapy. Eur J Pharm Sci 2020; 145:105235. [DOI: 10.1016/j.ejps.2020.105235] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022]
|
26
|
Zhao X, Ning Q, Mo Z, Tang S. A promising cancer diagnosis and treatment strategy: targeted cancer therapy and imaging based on antibody fragment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3621-3630. [PMID: 31468992 DOI: 10.1080/21691401.2019.1657875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the arrival of the precision medicine and personalized treatment era, targeted therapy that improves efficacy and reduces side effects has become the mainstream approach of cancer treatment. Antibody fragments that further enhance penetration and retain the most critical antigen-specific binding functions are considered the focus of research targeting cancer imaging and therapy. Thanks to the superior penetration and rapid blood clearance of antibody fragments, antibody fragment-based imaging agents enable efficient and sensitive imaging of tumour sites. In tumour-targeted therapy, antibody fragments can directly inhibit tumour proliferation and growth, serve as an ideal carrier for delivery of anti-tumour drugs, or manipulate the immune system to eliminate tumour cells. In this review, the excellent physicochemical properties and the basic structure of antibody fragments are expressly depicted depicted, the progress of antibody fragments in cancer therapy and imaging are thoroughly summarized, and the future development of antibody fragments is predicted.
Collapse
Affiliation(s)
- Xuhong Zhao
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China , Hengyang , China.,Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| | - Zhongcheng Mo
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China , Hengyang , China
| | - Shengsong Tang
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China , Hengyang , China.,Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| |
Collapse
|
27
|
Lungu II, Grumezescu AM, Volceanov A, Andronescu E. Nanobiomaterials Used in Cancer Therapy: An Up-To-Date Overview. Molecules 2019; 24:E3547. [PMID: 31574993 PMCID: PMC6804091 DOI: 10.3390/molecules24193547] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 01/09/2023] Open
Abstract
The disadvantages that come with traditional cancer treatments, such as chemotherapy and radiotherapy, generated a research shift toward nanotechnology. However, even with the important advancements regarding cancer therapy, there are still serious stepping stones that need to be addressed. The use of both nanotechnology and nanomedicine has generated significant improvements in nano-sized materials development and their use as therapeutic, diagnosis, and imaging agents. The biological barriers that come from the healthy body, as well from the tumorous sites, are important parameters that need to be taken into consideration when designing drug delivery systems. There are several aspects of extreme importance such as the tumor microenvironment and vasculature, the reticuloendothelial system, the blood-brain barrier, the blood-tumor barrier, and the renal system. In order to achieve an effective system for cancer therapy, several characteristics of the nanoparticles have been outlined. Moreover, this review has also focused on the different types of nanoparticles that have been studied over the years as potential candidates for cancer therapy.
Collapse
Affiliation(s)
- Iulia Ioana Lungu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania.
- National Institute of Laser, Plasma and Radiation Physics (NILPRP), Bucharest-Magurele, 077125 Magurele, Romania.
| | - Alexandru Mihai Grumezescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Adrian Volceanov
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Ecaterina Andronescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| |
Collapse
|
28
|
Yan S, Huang Q, Chen J, Song X, Chen Z, Huang M, Xu P, Zhang J. Tumor-targeting photodynamic therapy based on folate-modified polydopamine nanoparticles. Int J Nanomedicine 2019; 14:6799-6812. [PMID: 31692522 PMCID: PMC6711554 DOI: 10.2147/ijn.s216194] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/27/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Photodynamic therapy (PDT), a clinical anticancer therapeutic modality, has a long history in clinical cancer treatments since the 1970s. However, PDT has not been widely used largely because of metabolic problems and off-target phototoxicities of the current clinical photosensitizers. PURPOSE The objective of the study is to develop a high-efficiency and high-specificity carrier to precisely deliver photosensitizers to tumor sites, aiming at addressing metabolic problems, as well as the systemic damages current clinical photosensitizers are known to cause. METHODS We synthesized a polydopamine (PDA)-based carrier with the modification of folic acid (FA), which is to target the overexpressed folate receptors on tumor surfaces. We used this carrier to load a cationic phthalocyanine-type photosensitizer (Pc) and generated a PDA-FA-Pc nanomedicine. We determined the antitumor effects and the specificity to tumor cell lines in vitro. In addition, we established human cancer-xenografted mice models to evaluate the tumor-targeting property and anticancer efficacies in vivo. RESULTS Our PDA-FA-Pc nanomedicine demonstrated a high stability in normal physiological conditions, however, could specifically release photosensitizers in acidic conditions, eg, tumor microenvironment and lysosomes in cancer cells. Additionally, PDA-FA-Pc nanomedicine demonstrated a much higher cellular uptake and phototoxicity in cancer cell lines than in healthy cell lines. Moreover, the in vivo imaging data indicated excellent tumor-targeting properties of PDA-FA-Pc nanomedicine in human cancer-xenografted mice. Lastly, PDA-FA-Pc nanomedicine was found to significantly suppress tumor growth within two human cancer-xenografted mice models. CONCLUSION Our current study not only demonstrates PDA-FA-Pc nanomedicine as a highly potent and specific anticancer agent, but also suggests a strategy to address the metabolic and specificity problems of clinical photosensitizers.
Collapse
Affiliation(s)
- Shufeng Yan
- Medical Plant Exploitation and Utilization Engineering Research Center, Sanming University, Sanming, Fujian365004, People’s Republic of China
| | - Qingqing Huang
- Medical Plant Exploitation and Utilization Engineering Research Center, Sanming University, Sanming, Fujian365004, People’s Republic of China
| | - Jincan Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian350002, People’s Republic of China
| | - Xiaorong Song
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian350002, People’s Republic of China
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian350002, People’s Republic of China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, People’s Republic of China
| | - Peng Xu
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore138673, Singapore
| | - Juncheng Zhang
- Medical Plant Exploitation and Utilization Engineering Research Center, Sanming University, Sanming, Fujian365004, People’s Republic of China
| |
Collapse
|
29
|
Lopes TS, Alves GG, Pereira MR, Granjeiro JM, Leite PEC. Advances and potential application of gold nanoparticles in nanomedicine. J Cell Biochem 2019; 120:16370-16378. [DOI: 10.1002/jcb.29044] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Talíria Silva Lopes
- Graduate Program in Sciences and Biotechnology, Fluminense Federal University – UFF Niteroi RJ Brazil
| | - Gutemberg Gomes Alves
- Cell and Molecular Biology Department Biology Institute, Fluminense Federal University – UFF Niteroi RJ Brazil
| | | | - Jose Mauro Granjeiro
- Dental School – Fluminense Federal University – UFF Niteroi RJ Brazil
- Laboratory of Bioengineering and in Vitro Toxicology Directory of Metrology Applied to Life Sciences – Dimav, National Institute of Metrology Quality and Technology – INMETRO Duque de Caxias RJ Brazil
| | - Paulo Emílio Corrêa Leite
- Laboratory of Bioengineering and in Vitro Toxicology Directory of Metrology Applied to Life Sciences – Dimav, National Institute of Metrology Quality and Technology – INMETRO Duque de Caxias RJ Brazil
| |
Collapse
|
30
|
Mirrahimi M, Abed Z, Beik J, Shiri I, Shiralizadeh Dezfuli A, Mahabadi VP, Kamran Kamrava S, Ghaznavi H, Shakeri-Zadeh A. A thermo-responsive alginate nanogel platform co-loaded with gold nanoparticles and cisplatin for combined cancer chemo-photothermal therapy. Pharmacol Res 2019; 143:178-185. [DOI: 10.1016/j.phrs.2019.01.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/16/2018] [Accepted: 01/02/2019] [Indexed: 11/26/2022]
|
31
|
Hadi F, Tavakkol S, Laurent S, Pirhajati V, Mahdavi SR, Neshastehriz A, Shakeri-Zadeh A. Combinatorial effects of radiofrequency hyperthermia and radiotherapy in the presence of magneto-plasmonic nanoparticles on MCF-7 breast cancer cells. J Cell Physiol 2019; 234:20028-20035. [PMID: 30982979 DOI: 10.1002/jcp.28599] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 01/05/2023]
Abstract
Here, the effects of combinatorial cancer therapy including radiotherapy (RT) and radiofrequency (RF) hyperthermia in the presence of gold-coated iron oxide nanoparticles (Au@IONPs), as a thermo-radio-sensitizer, are reported. The level of cell death and the ratio of Bax/Bcl2 genes, involved in the pathway of apoptosis, were measured to evaluate the synergistic effect of Au@IONPs-mediated RF hyperthermia and RT. MCF-7 human breast adenocarcinoma cells were treated with different concentrations of Au@IONPs. After incubation with NPs, the cells were exposed to RF waves (13.56 MHz; 100 W; 15 min). At the same time, thermometry was performed with an infrared (IR) camera. Then, the cells were exposed to 6 MV X-ray at various doses of 2 and 4 Gy. MTT (3-[4,5-dimethylthiazol-2-y1]-2,5-diphenyltetrazolium bromide) assay was performed to evaluate cell viability and quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the expression ratio of Bax/Bcl2. Cellular uptake of nanoparticles was confirmed qualitatively and quantitatively. The results obtained from MTT assay and qRT-PCR studies showed that NPs and RF hyperthermia had no significant effect when applied separately, while their combination had synergistic effects on cell viability percentage and the level of apoptosis induction. A synergistic effect was also observed when the cancer cells were treated with a combination of NPs, RF hyperthermia, and RT. On the basis of the obtained results, it may be concluded that the use of magneto-plasmonic NPs in the process of hyperthermia and RT of cancer holds a great promise to develop a new combinatorial cancer therapy strategy.
Collapse
Affiliation(s)
- Fahimeh Hadi
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Radiation Science Department, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Shima Tavakkol
- Cellular and Molecular Research Center, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Vahid Pirhajati
- Cellular and Molecular Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Neuroscience Research Center, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Seied Rabi Mahdavi
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Finetech in Medicine Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Medical Physics Department, School of Medicine, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Ali Neshastehriz
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Radiation Science Department, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Ali Shakeri-Zadeh
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Finetech in Medicine Research Center, Iran University of Medical Science (IUMS), Tehran, Iran.,Medical Physics Department, School of Medicine, Iran University of Medical Science (IUMS), Tehran, Iran
| |
Collapse
|