1
|
Zeng Y, Pan Z, Yuan J, Song Y, Feng Z, Chen Z, Ye Z, Li Y, Bao Y, Ran Z, Li X, Ye H, Zhang K, Liu X, He Y. Inhibiting Osteolytic Breast Cancer Bone Metastasis by Bone-Targeted Nanoagent via Remodeling the Bone Tumor Microenvironment Combined with NIR-II Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301003. [PMID: 37211708 DOI: 10.1002/smll.202301003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/08/2023] [Indexed: 05/23/2023]
Abstract
Bone is one of the prone metastatic sites of patients with advanced breast cancer. The "vicious cycle" between osteoclasts and breast cancer cells plays an essential role in osteolytic bone metastasis from breast cancer. In order to inhibit bone metastasis from breast cancer, NIR-II photoresponsive bone-targeting nanosystems (CuP@PPy-ZOL NPs) are designed and synthesized. CuP@PPy-ZOL NPs can trigger the photothermal-enhanced Fenton response and photodynamic effect to enhance the photothermal treatment (PTT) effect and thus achieve synergistic anti-tumor effect. Meanwhile, they exhibit a photothermal enhanced ability to inhibit osteoclast differentiation and promote osteoblast differentiation, which reshaped the bone microenvironment. CuP@PPy-ZOL NPs effectively inhibited the proliferation of tumor cells and bone resorption in the in vitro 3D bone metastases model of breast cancer. In a mouse model of breast cancer bone metastasis, CuP@PPy-ZOL NPs combined with PTT with NIR-II significantly inhibited the tumor growth of breast cancer bone metastases and osteolysis while promoting bone repair to achieve the reversal of osteolytic breast cancer bone metastases. Furthermore, the potential biological mechanisms of synergistic treatment are identified by conditioned culture experiments and mRNA transcriptome analysis. The design of this nanosystem provides a promising strategy for treating osteolytic bone metastases.
Collapse
Affiliation(s)
- Yaoxun Zeng
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhenxing Pan
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Jiongpeng Yuan
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yuqiong Song
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P. R. China
| | - Zhenzhen Feng
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zefeng Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, SAR, 999077, P. R. China
| | - Zhaoyi Ye
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yushan Li
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Ying Bao
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhili Ran
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Xinyi Li
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Huiling Ye
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Kun Zhang
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Xujie Liu
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yan He
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| |
Collapse
|
2
|
Domingo-Diez J, Souiade L, Manzaneda-González V, Sánchez-Díez M, Megias D, Guerrero-Martínez A, Ramírez-Castillejo C, Serrano-Olmedo J, Ramos-Gómez M. Effectiveness of Gold Nanorods of Different Sizes in Photothermal Therapy to Eliminate Melanoma and Glioblastoma Cells. Int J Mol Sci 2023; 24:13306. [PMID: 37686114 PMCID: PMC10488215 DOI: 10.3390/ijms241713306] [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: 08/03/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Gold nanorods are the most commonly used nanoparticles in photothermal therapy for cancer treatment due to their high efficiency in converting light into heat. This study aimed to investigate the efficacy of gold nanorods of different sizes (large and small) in eliminating two types of cancer cell: melanoma and glioblastoma cells. After establishing the optimal concentration of nanoparticles and determining the appropriate time and power of laser irradiation, photothermal therapy was applied to melanoma and glioblastoma cells, resulting in the highly efficient elimination of both cell types. The efficiency of the PTT was evaluated using several methods, including biochemical analysis, fluorescence microscopy, and flow cytometry. The dehydrogenase activity, as well as calcein-propidium iodide and Annexin V staining, were employed to determine the cell viability and the type of cell death triggered by the PTT. The melanoma cells exhibited greater resistance to photothermal therapy, but this resistance was overcome by irradiating cells at physiological temperatures. Our findings revealed that the predominant cell-death pathway activated by the photothermal therapy mediated by gold nanorods was apoptosis. This is advantageous as the presence of apoptotic cells can stimulate antitumoral immunity in vivo. Considering the high efficacy of these gold nanorods in photothermal therapy, large nanoparticles could be useful for biofunctionalization purposes. Large nanorods offer a greater surface area for attaching biomolecules, thereby promoting high sensitivity and specificity in recognizing target cancer cells. Additionally, large nanoparticles could also be beneficial for theranostic applications, involving both therapy and diagnosis, due to their superior detection sensitivity.
Collapse
Affiliation(s)
- Javier Domingo-Diez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
| | - Lilia Souiade
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
| | - Vanesa Manzaneda-González
- Departamento de Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain (A.G.-M.)
| | - Marta Sánchez-Díez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Grupo de Sistemas Complejos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Diego Megias
- Advanced Optical Microscopy Unit, UCCTs, Instituto de Salud Carlos III (ISCIII), 28222 Madrid, Spain
| | - Andrés Guerrero-Martínez
- Departamento de Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain (A.G.-M.)
| | - Carmen Ramírez-Castillejo
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Grupo de Sistemas Complejos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Javier Serrano-Olmedo
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Centro de Investigación Biomédica en Red para Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Milagros Ramos-Gómez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Centro de Investigación Biomédica en Red para Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Experimental Neurology Unit, Center for Biomedical Technology, Universidad Politécnica de Madrid, Campus de Montegancedo s/n, Pozuelo de Alarcón, 28223 Madrid, Spain
| |
Collapse
|
3
|
Li W, Zhang X, Shi Y, Hu X, Wang X, Liang N, Shen T, Zou X, Shi J. A dual-modal biosensor coupling cooperative catalysis strategy for sensitive detection of AFB 1 in agri-products. Food Chem 2023; 426:136553. [PMID: 37354581 DOI: 10.1016/j.foodchem.2023.136553] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/23/2023] [Accepted: 06/04/2023] [Indexed: 06/26/2023]
Abstract
Herein, the cooperative catalysis effect between nanocomposite (AgPd NPs/POD-M/PEI-rGO) and horseradish peroxidase (HRP) was applied for the fast and sensitive detection of aflatoxin B1 (AFB1). Upon specific and competitive binding of HRP@DNA and AFB1 to cDNA, the working electrode presented different catalytic capacities for supporting electrolytes (TMB and H2O2). In the redox mechanism of TMB and H2O2, HRP and nanocomposite effectively catalyzed the oxidization of TMB to form the one-electron oxidation intermediate TMB+, and contributed the electrical signals and absorbance signals. Electrochemistry and colorimetric analyses were successfully realized for AFB1 detection with 0.2 pg/mL and 8 pg/mL of detection limits, respectively, which is much lower than that of traditional HPLC methods. Overall, this method had significant reliability and sensitivity, offering a promising potential for conveniently evaluating the quality of agri-products polluted with AFB1. Moreover, this approach provides a new idea for fast and accurate detection of mycotoxin.
Collapse
Affiliation(s)
- Wenting Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xinai Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yongqiang Shi
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xuetao Hu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xin Wang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Nini Liang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tingting Shen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xiaobo Zou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Joint Laboratory of China-UK on Food Nondestructive Sensing, Jiangsu University, Zhenjiang 212013, China; International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang 212013, China
| | - Jiyong Shi
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Joint Laboratory of China-UK on Food Nondestructive Sensing, Jiangsu University, Zhenjiang 212013, China; International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
4
|
Wang X, Xuan L, Pan Y. Photothermal ablation of murine melanomas by Fe 3O 4 nanoparticle clusters. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2022; 13:255-264. [PMID: 35281633 PMCID: PMC8895026 DOI: 10.3762/bjnano.13.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/10/2022] [Indexed: 06/14/2023]
Abstract
Melanoma is one of the deadliest forms of cancer, for which therapeutic regimens are usually limited by the development of resistance. Here, we fabricated Fe3O4 nanoparticle clusters (NPCs), which have drawn widespread attention, and investigated their role in the treatment of melanoma by photothermal therapy (PTT). Scanning electron microscopy imaging shows that our synthesized NPCs are spherical with an average diameter of 329.2 nm. They are highly absorptive at the near-infrared wavelength of 808 nm and efficient at locally converting light into heat. In vitro experiments using light-field microscopy and cell viability assay showed that Fe3O4 NPCs, in conjunction with near-infrared irradiation, effectively ablated A375 melanoma cells by inducing overt apoptosis. Consistently, in vivo studies using BALB/c mice found that intratumoral administration of Fe3O4 NPCs and concomitant in situ exposure to near-infrared light significantly inhibited the growth of implanted tumor xenografts. Finally, we revealed, by experimental approaches including semi-quantitative PCR, western blot and immunohistochemistry, the heat shock protein HSP70 to be upregulated in response to PTT, suggesting this chaperone protein could be a plausible underlying mechanism for the observed therapeutic outcome. Altogether, our results highlight the promise of Fe3O4 NPCs as a new PTT option to treat melanoma.
Collapse
Affiliation(s)
- Xue Wang
- Department of Obstetrics & Gynecology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Lili Xuan
- Department of Obstetrics & Gynecology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Ying Pan
- Department of Obstetrics & Gynecology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
5
|
Phenotypic Switching of B16F10 Melanoma Cells as a Stress Adaptation Response to Fe3O4/Salicylic Acid Nanoparticle Therapy. Pharmaceuticals (Basel) 2021; 14:ph14101007. [PMID: 34681232 PMCID: PMC8537856 DOI: 10.3390/ph14101007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a melanocyte-derived skin cancer that has a high heterogeneity due to its phenotypic plasticity, a trait that may explain its ability to survive in the case of physical or molecular aggression and to develop resistance to therapy. Therefore, the therapy modulation of phenotypic switching in combination with other treatment modalities could become a common approach in any future therapeutic strategy. In this paper, we used the syngeneic model of B16F10 melanoma implanted in C57BL/6 mice to evaluate the phenotypic changes in melanoma induced by therapy with iron oxide nanoparticles functionalized with salicylic acid (SaIONs). The results of this study showed that the oral administration of the SaIONs aqueous dispersion was followed by phenotypic switching to highly pigmented cells in B16F10 melanoma through a cytotoxicity-induced cell selection mechanism. The hyperpigmentation of melanoma cells by the intra- or extracellular accumulation of melanic pigment deposits was another consequence of the SaIONs therapy. Additional studies are needed to assess the reversibility of SaIONs-induced phenotypic switching and the impact of tumor hyperpigmentation on B16F10 melanoma’s progression and metastasis abilities.
Collapse
|
6
|
Kang K, Zhou X, Zhang Y, Zhu N, Li G, Yi Q, Wu Y. Cell-Released Magnetic Vesicles Capturing Metabolic Labeled Rare Circulating Tumor Cells Based on Bioorthogonal Chemistry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007796. [PMID: 33749110 DOI: 10.1002/smll.202007796] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/24/2021] [Indexed: 06/12/2023]
Abstract
Capture of circulating tumor cells (CTCs) with high efficiency and high purity holds great value for potential clinical applications. Besides the existing problems of contamination from blood cells and plasma proteins, unknown/down-regulated expression of targeting markers (e.g., antigen, receptor, etc.) of CTCs have questioned the reliability and general applicability of current CTCs capture methodologies based on immune/aptamer-affinity. Herein, a cell-engineered strategy is designed to break down such barriers by employing the cell metabolism as the leading force to solve key problems. Generally, through an extracellular vesicle generation way, the cell-released magnetic vesicles inherited parent cellular membrane characteristics are produced, and then functionalized with dibenzoazacyclooctyne to target and isolate the metabolic labeled rare CTCs. This strategy offers good reliability and broader possibilities to capture different types of tumor cells, as proven by the capture efficiency above 84% and 82% for A549 and HepG2 cell lines as well as an extremely low detection limitation of 5 cells. Moreover, it enabled high purity enrichment of CTCs from 1 mL blood samples of tumor-bearing mice, only ≈5-757 white blood cells are non-specific caught, ignoring the potential phenotypic fluctuation associated with the cancer progression.
Collapse
Affiliation(s)
- Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiaoxi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yujia Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Guohao Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
7
|
Cheng Y, Chen Q, Guo Z, Li M, Yang X, Wan G, Chen H, Zhang Q, Wang Y. An Intelligent Biomimetic Nanoplatform for Holistic Treatment of Metastatic Triple-Negative Breast Cancer via Photothermal Ablation and Immune Remodeling. ACS NANO 2020; 14:15161-15181. [PMID: 33143424 DOI: 10.1021/acsnano.0c05392] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Metastasis is one of the main causes of failure in the treatment of triple-negative breast cancer (TNBC). Immunotherapy brings hope and opportunity to solve this challenge, while its clinical applications are greatly inhibited by the tumor immunosuppressive environment. Here, an intelligent biomimetic nanoplatform was designed based on dendritic large-pore mesoporous silica nanoparticles (DLMSNs) for suppressing metastatic TNBC by combining photothermal ablation and immune remodeling. Taking advantage of the ordered large-pore structure and easily chemically modified property of DLMSNs, the copper sulfide (CuS) nanoparticles with high photothermal conversion efficiency were in situ deposited inside the large pores of DLMSNs, and the immune adjuvant resiquimod (R848) was loaded controllably. A homogenous cancer cell membrane was coated on the surfaces of these DLMSNs, followed by conjugation with the anti-PD-1 peptide AUNP-12 through a polyethylene glycol linker with an acid-labile benzoic-imine bond. The thus-obtained AM@DLMSN@CuS/R848 was applied to holistically treat metastatic TNBC in vitro and in vivo. The data showed that AM@DLMSN@CuS/R848 had a high TNBC-targeting ability and induced efficient photothermal ablation on primary TNBC tumors under 980 nm laser irradiation. Tumor antigens thus generated and increasingly released R848 by response to the photothermal effect, combined with AUNP-12 detached from AM@DLMSN@CuS/R848 in the weakly acidic tumor microenvironment, synergistically exerted tumor vaccination, and T lymphocyte activation functions on immune remodeling to prevent TNBC recurrence and metastasis. Taken together, this study provides an intelligent biomimetic nanoplatform to enhance therapeutic outcomes in metastatic TNBC.
Collapse
Affiliation(s)
- Yuanyuan Cheng
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Qian Chen
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Zhaoyang Guo
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Mengwen Li
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xiaoying Yang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Guoyun Wan
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Hongli Chen
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Qiqing Zhang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Yinsong Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
8
|
Abstract
Melanoma is an aggressive form of skin cancer with a very high mortality rate. Early diagnosis of the disease, the utilization of more potent pharmacological agents, and more effective drug delivery systems are essential to achieve an optimal treatment plan. The applications of nanotechnology to improve therapeutic efficacy and early diagnosis for melanoma treatment have received great interest among researchers and clinicians. In this review, we summarize the recent progress of utilizing various nanomaterials for theranostics of melanoma. The key importance of using nanomaterials for theranostics of melanoma is to improve efficacy and reduce side effects, ensuring safe implementation in clinical use. As opposed to conventional in vitro diagnostic methods, in vivo medical imaging technologies have the advantages of being a type of non-invasive, real-time monitoring. Several common nanoparticles, including ultrasmall superparamagnetic iron oxide nanoparticles, silica nanoparticles, and carbon-based nanoparticles, have been applied to deliver chemotherapeutic agents for the theranostics of melanoma. The application of nanomaterials for theranostics in molecular imaging (MRI, PET, US, OI, etc.) plays an important role in targeting drug delivery of melanoma, by monitoring the distribution site of the molecular imaging probe and the therapeutic drug in the body in real-time. Hence, it is worthwhile to anticipate the approval of these nanomaterials for theranostics in molecular imaging by the US Food and Drug Administration in clinical trials.
Collapse
|
9
|
Wang X, Li H, Li F, Han X, Chen G. Prussian blue-coated lanthanide-doped core/shell/shell nanocrystals for NIR-II image-guided photothermal therapy. NANOSCALE 2019; 11:22079-22088. [PMID: 31720650 DOI: 10.1039/c9nr07973d] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Lanthanide-doped nanoparticles have long been stereotyped for optical luminescence bioimaging. However, they are known to be unable to produce therapeutic abilities. Here, we describe a lanthanide-based theranostic agent, namely, prussian blue (PB)-coated NaErF4@NaYF4@NaNdF4 core/shell/shell nanocrystals encapsulated in a phospholipid PEG micelle (PEG-CSS@PB), which showed switched imaging and hyperthermia abilities under distinct near infrared (NIR) light activation. The erbium (Er3+)-enriched inner core nanocrystals (NaErF4) enabled the emission of tissue-penetrating luminescence (1525 nm) in the second biological window (NIR-II, 1000-1700 nm), which endowed high-resolution optical imaging of the blood vessels and tumors under ∼980 nm excitation. High neodymium (Nd3+) concentrations in the epitaxial outer NaNdF4 shell introduced maximum cross relaxation processes that converted the absorbed NIR light (∼808 nm) into heat at high efficiencies, thus providing abilities for photothermal therapy (PTT). Importantly, the coated Prussian blue (PB) increased light absorption by about 10-fold compared to the composite free of PB, thus entailing a high light-to-heat conversion efficiency of ∼50.5%. This commensurated with that of well-established gold nanorods. As a result, the PEG-CSS@PB nanoparticles with MTT-determined low toxicities resulted in ∼80% death of HeLa cells at a dose of 600 μg mL-1 under 808 nm laser irradiance (1 W cm-2) for 10 min. Moreover, utilizing the same light dose, a single PTT treatment in tumor-bearing BALB/c mice shrunk the tumor size by ∼12-fold compared to the tumors without treatment. Our results, here, constituted a solid step forward to entitle lanthanide-based nanoparticles as theranostic agents in nanomedicine studies.
Collapse
Affiliation(s)
- Xin Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, People's Republic of China.
| | | | | | | | | |
Collapse
|