1
|
Wang L, Zhang X, He L, Wei Y, Zhang Y, Wu A, Li J. Iron-Based Nanomaterials for Modulating Tumor Microenvironment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70001. [PMID: 39788569 DOI: 10.1002/wnan.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Iron-based nanomaterials (IBNMs) have been widely applied in biomedicine applications including magnetic resonance imaging, targeted drug delivery, tumor therapy, and so forth, due to their unique magnetism, excellent biocompatibility, and diverse modalities. Further research on its enormous biomedical potential is still ongoing, and its new features are constantly being tapped and demonstrated. Among them, various types of IBNMs have demonstrated significant cancer therapy capabilities by regulating the tumor microenvironment (TME). In this review, a variety of IBNMs including iron oxide-based nanomaterials (IONMs), iron-based complex conjugates (ICCs), and iron-based single iron atom nanomaterials (ISANMs) will be introduced, and their advantages in regulating TME would also be emphasized. Besides, the recent progress of IBNMs for cancer diagnosis and treatment through the strategy of modulating TME will be summarized, including overcoming hypoxia, modulating acidity, decreasing redox species, and immunoregulation. Finally, the challenges and opportunities in this field are briefly discussed. This review is expected to contribute to the future design and development of next-generation TME-modulate IBNMs for cancer treatment.
Collapse
Affiliation(s)
- Le Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Xiaoting Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Lulu He
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yuanyuan Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yujie Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Aiguo Wu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Juan Li
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| |
Collapse
|
2
|
Su Y, Huang L, Zhang D, Zeng Z, Hong S, Lin X. Recent Advancements in Ultrasound Contrast Agents Based on Nanomaterials for Imaging. ACS Biomater Sci Eng 2024; 10:5496-5512. [PMID: 39246058 DOI: 10.1021/acsbiomaterials.4c00890] [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] [Indexed: 09/10/2024]
Abstract
Ultrasound (US) is a type of mechanical wave that is capable of transmitting energy through biological tissues. By utilization of various frequencies and intensities, it can elicit specific biological effects. US imaging (USI) technology has been continuously developed with the advantages of safety and the absence of radiation. The advancement of nanotechnology has led to the utilization of various nanomaterials composed of both organic and inorganic compounds as ultrasound contrast agents (UCAs). These UCAs enhance USI, enabling real-time monitoring, diagnosis, and treatment of diseases, thereby facilitating the widespread adoption of UCAs in precision medicine. In this review, we introduce various UCAs based on nanomaterials for USI. Their principles can be roughly divided into the following categories: carrying and transporting gases, endogenous gas production, and the structural characteristics of the nanomaterial itself. Furthermore, the synergistic benefits of US in conjunction with various imaging modalities and their combined application in disease monitoring and diagnosis are introduced. In addition, the challenges and prospects for the development of UCAs are also discussed.
Collapse
Affiliation(s)
- Yina Su
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Linjie Huang
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Dongdong Zhang
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Zheng Zeng
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Shanni Hong
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Xiahui Lin
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| |
Collapse
|
3
|
Kim C, Mai DK, Kim WJ, Badon IW, Jo J, Kang D, Kim SJ, Kim HJ, Yang J. Red fluorescent BODIPY-based nanoparticles for targeted cancer imaging-guided photodynamic therapy. Biomater Sci 2024; 12:1536-1548. [PMID: 38299265 DOI: 10.1039/d3bm01520c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Imaging-guided diagnosis and treatment of cancer hold potential to significantly improve therapeutic accuracies and efficacies. Central to this theragnostic approach has been the use of multicomponent-based multimodal nanoparticles (NPs). Apart from this conventional approach, here we propose a design strategy for the simple and straightforward formulation of NPs based on boron dipyrromethene (BODIPY) derivatives, LaB-X (X = H, Et, and Br). Specifically, the conjugation of lactose to the inherently hydrophobic BODIPY promoted the formation of LaB-X NPs in water. Furthermore, the BODIPY backbone was subjected to distyrylation, dibromination, and diethylation to tailor the optical window and the balance between fluorescence and singlet oxygen generation capabilities. We demonstrate that while the photoinduced anticancer activities of LaB-H and LaB-Et NPs were trivial, LaB-Br NPs effectively induced the apoptotic death of hepatocellular carcinoma cells under red light irradiation while allowing fluorescence cell imaging in the phototherapeutic window. This dual fluorescence photosensitizing activity of LaB-Br NPs could be switched off and on, so that both fluorescence and singlet oxygen generation were paused during NP formation in an aqueous solution, while both processes resumed after cellular uptake, likely due to NP disassembly.
Collapse
Affiliation(s)
- Chanwoo Kim
- Department of Chemistry, Yonsei University, Wonju, Gangwon 26493, Korea.
| | - Duy Khuong Mai
- Department of Chemistry, Chosun University, Gwangju 61452, Korea.
| | - Won-Jin Kim
- Department of Integrative Biological Sciences, BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
- Institute of Well-Aging Medicare, Chosun University, Gwangju 61452, Korea
| | - Isabel Wen Badon
- Department of Chemistry, Chosun University, Gwangju 61452, Korea.
| | - Jinwoong Jo
- Department of Chemistry, Yonsei University, Wonju, Gangwon 26493, Korea.
| | - Dongho Kang
- Department of Chemistry, Yonsei University, Wonju, Gangwon 26493, Korea.
| | - Seok-Jun Kim
- Department of Integrative Biological Sciences, BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
- Institute of Well-Aging Medicare, Chosun University, Gwangju 61452, Korea
| | - Ho-Joong Kim
- Department of Chemistry, Chosun University, Gwangju 61452, Korea.
| | - Jaesung Yang
- Department of Chemistry, Yonsei University, Wonju, Gangwon 26493, Korea.
| |
Collapse
|
4
|
Guo B, Sofias AM, Lammers T, Xu J. Image-guided drug delivery: Nanoparticle and probe advances. Adv Drug Deliv Rev 2024; 206:115188. [PMID: 38272185 DOI: 10.1016/j.addr.2024.115188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Affiliation(s)
- Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
| | - Jian Xu
- Institute of Low-Dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
5
|
Pewklang T, Saiyasombat W, Chueakwon P, Ouengwanarat B, Chansaenpak K, Kampaengsri S, Lai RY, Kamkaew A. Revolutionary Pyrazole-based Aza-BODIPY: Harnessing Photothermal Power Against Cancer Cells and Bacteria. Chembiochem 2024; 25:e202300653. [PMID: 38095754 DOI: 10.1002/cbic.202300653] [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: 09/25/2023] [Revised: 11/25/2023] [Indexed: 01/11/2024]
Abstract
In the realm of cancer therapy and treatment of bacterial infection, photothermal therapy (PTT) stands out as a potential strategy. The challenge, however, is to create photothermal agents that can perform both imaging and PTT, a so-called theranostic agent. Photothermal agents that absorb and emit in the near-infrared region (750-900 nm) have recently received a lot of attention due to the extensive penetration of NIR light in biological tissues. In this study, we combined pyrazole with aza-BODIPY (PY-AZB) to develop a novel photothermal agent. PY-AZB demonstrated great photostability with a photothermal conversion efficiency (PCE) of up to 33 %. Additionally, PY-AZB can permeate cancer cells at a fast accumulation rate in less than 6 hours, according to the confocal images. Furthermore, in vitro photothermal therapy results showed that PY-AZB effectively eliminated cancer cells by up to 70 %. Interestingly, PY-AZB exhibited antibacterial activities against both gram-negative bacteria, Escherichia coli 780, and gram-positive bacteria, Staphylococcus aureus 1466. The results exhibit a satisfactory bactericidal effect against bacteria, with a killing efficiency of up to 100 % upon laser irradiation. As a result, PY-AZB may provide a viable option for photothermal treatment.
Collapse
Affiliation(s)
- Thitima Pewklang
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| | - Worakrit Saiyasombat
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| | - Piyasiri Chueakwon
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| | - Bongkot Ouengwanarat
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| | - Kantapat Chansaenpak
- National Nanotechnology Center, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand, 12120
| | - Sastiya Kampaengsri
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| | - Rung-Yi Lai
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| | - Anyanee Kamkaew
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand, 30000
| |
Collapse
|
6
|
Chen X, Mendes B, Zhuang Y, Conniot J, Mercado Argandona S, Melle F, Sousa DP, Perl D, Chivu A, Patra HK, Shepard W, Conde J, Fairen-Jimenez D. A Fluorinated BODIPY-Based Zirconium Metal-Organic Framework for In Vivo Enhanced Photodynamic Therapy. J Am Chem Soc 2024; 146:1644-1656. [PMID: 38174960 PMCID: PMC10797627 DOI: 10.1021/jacs.3c12416] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
Photodynamic therapy (PDT), an emergent noninvasive cancer treatment, is largely dependent on the presence of efficient photosensitizers (PSs) and a sufficient oxygen supply. However, the therapeutic efficacy of PSs is greatly compromised by poor solubility, aggregation tendency, and oxygen depletion within solid tumors during PDT in hypoxic microenvironments. Despite the potential of PS-based metal-organic frameworks (MOFs), addressing hypoxia remains challenging. Boron dipyrromethene (BODIPY) chromophores, with excellent photostability, have exhibited great potential in PDT and bioimaging. However, their practical application suffers from limited chemical stability under harsh MOF synthesis conditions. Herein, we report the synthesis of the first example of a Zr-based MOF, namely, 69-L2, exclusively constructed from the BODIPY-derived ligands via a single-crystal to single-crystal post-synthetic exchange, where a direct solvothermal method is not applicable. To increase the PDT performance in hypoxia, we modify 69-L2 with fluorinated phosphate-functionalized methoxy poly(ethylene glycol). The resulting 69-L2@F is an oxygen carrier, enabling tumor oxygenation and simultaneously acting as a PS for reactive oxygen species (ROS) generation under LED irradiation. We demonstrate that 69-L2@F has an enhanced PDT effect in triple-negative breast cancer MDA-MB-231 cells under both normoxia and hypoxia. Following positive results, we evaluated the in vivo activity of 69-L2@F with a hydrogel, enabling local therapy in a triple-negative breast cancer mice model and achieving exceptional antitumor efficacy in only 2 days. We envision BODIPY-based Zr-MOFs to provide a solution for hypoxia relief and maximize efficacy during in vivo PDT, offering new insights into the design of promising MOF-based PSs for hypoxic tumors.
Collapse
Affiliation(s)
- Xu Chen
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Bárbara
B. Mendes
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - Yunhui Zhuang
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - João Conniot
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - Sergio Mercado Argandona
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Francesca Melle
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Diana P. Sousa
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - David Perl
- Synchrotron
SOLEIL-UR1, L’Orme des Merisiers, Départementale 128, 91190 Saint-Aubin, France
| | - Alexandru Chivu
- Department
of Surgical Biotechnology, University College
London, London NW3 2PF, U.K.
| | - Hirak K. Patra
- Department
of Surgical Biotechnology, University College
London, London NW3 2PF, U.K.
| | - William Shepard
- Synchrotron
SOLEIL-UR1, L’Orme des Merisiers, Départementale 128, 91190 Saint-Aubin, France
| | - João Conde
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - David Fairen-Jimenez
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| |
Collapse
|
7
|
Bala VM, Lampropoulou DI, Grammatikaki S, Kouloulias V, Lagopati N, Aravantinos G, Gazouli M. Nanoparticle-Mediated Hyperthermia and Cytotoxicity Mechanisms in Cancer. Int J Mol Sci 2023; 25:296. [PMID: 38203467 PMCID: PMC10779099 DOI: 10.3390/ijms25010296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Hyperthermia has the potential to damage cancerous tissue by increasing the body temperature. However, targeting cancer cells whilst protecting the surrounding tissues is often challenging, especially when implemented in clinical practice. In this direction, there are data showing that the combination of nanotechnology and hyperthermia offers more successful penetration of nanoparticles in the tumor environment, thus allowing targeted hyperthermia in the region of interest. At the same time, unlike radiotherapy, the use of non-ionizing radiation makes hyperthermia an attractive therapeutic option. This review summarizes the existing literature regarding the use of hyperthermia and nanoparticles in cancer, with a focus on nanoparticle-induced cytotoxicity mechanisms.
Collapse
Affiliation(s)
| | | | - Stamatiki Grammatikaki
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (N.L.)
| | - Vassilios Kouloulias
- Radiation Oncology Unit, 2nd Department of Radiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nefeli Lagopati
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (N.L.)
| | | | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (N.L.)
| |
Collapse
|
8
|
Shen J, Chen G, Zhao L, Huang G, Liu H, Liu B, Miao Y, Li Y. Recent Advances in Nanoplatform Construction Strategy for Alleviating Tumor Hypoxia. Adv Healthc Mater 2023; 12:e2300089. [PMID: 37055912 DOI: 10.1002/adhm.202300089] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Hypoxia is a typical feature of most solid tumors and has important effects on tumor cells' proliferation, invasion, and metastasis. This is the key factor that leads to poor efficacy of different kinds of therapy including chemotherapy, radiotherapy, photodynamic therapy, etc. In recent years, the construction of hypoxia-relieving functional nanoplatforms through nanotechnology has become a new strategy to reverse the current situation of tumor microenvironment hypoxia and improve the effectiveness of tumor treatment. Here, the main strategies and recent progress in constructing nanoplatforms are focused on to directly carry oxygen, generate oxygen in situ, inhibit mitochondrial respiration, and enhance blood perfusion to alleviate tumor hypoxia. The advantages and disadvantages of these nanoplatforms are compared. Meanwhile, nanoplatforms based on organic and inorganic substances are also summarized and classified. Through the comprehensive overview, it is hoped that the summary of these nanoplatforms for alleviating hypoxia could provide new enlightenment and prospects for the construction of nanomaterials in this field.
Collapse
Affiliation(s)
- Jing Shen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Guobo Chen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Linghao Zhao
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Guoyang Huang
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Hui Liu
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuhao Li
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
9
|
Ge Y, Zhang J, Jin K, Ye Z, Wang W, Zhou Z, Ye J. Multifunctional Nanoparticles Precisely Reprogram the Tumor Microenvironment and Potentiate Antitumor Immunotherapy after Near-Infrared-II Light-Mediated Photothermal Therapy. Acta Biomater 2023:S1742-7061(23)00316-1. [PMID: 37302731 DOI: 10.1016/j.actbio.2023.05.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Mild-temperature photothermal therapy (mild PTT) is a safe and efficient antitumor therapy. However, mild PTT alone usually fails to activate the immune response and prevent tumor metastasis. Herein, a photothermal agent, copper sulfide@ovalbumin (CuS@OVA), with an effective PTT effect in the second near-infrared (NIR-II) window, is developed. CuS@OVA can optimize the tumor microenvironment (TME) and evoke an adaptive immune response. Copper ions are released in the acidic TME to promote the M1 polarization of tumor-associated macrophages. The model antigen OVA not only acts as a scaffold for nanoparticle growth but also promotes the maturation of dendritic cells, which primes naive T cells to stimulate adaptive immunity. CuS@OVA augments the antitumor efficiency of the immune checkpoint blockade (ICB) in vivo, which suppresses tumor growth and metastasis in a mouse melanoma model. The proposed therapeutic platform, CuS@OVA nanoparticles, may be a potential adjuvant for optimizing the TME and improving the efficiency of ICB as well as other antitumor immunotherapies. STATEMENT OF SIGNIFICANCE: Mild-temperature photothermal therapy (mild PTT) is a safe and efficient antitumor therapy, but usually fails to activate the immune response and prevent tumor metastasis. Herein, we develop a photothermal agent, copper sulfide@ovalbumin (CuS@OVA), with an excellent PTT effect in the second near-infrared (NIR-II) window. CuS@OVA can optimize the tumor microenvironment (TME) and evoke an adaptive immune response by promoting the M1 polarization of tumor-associated macrophages and the maturation of dendritic cells. CuS@OVA augments the antitumor efficiency of the immune checkpoint blockade (ICB) in vivo, suppressing tumor growth and metastasis. The platform may be a potential adjuvant for optimizing the TME and improving the efficiency of ICB as well as other antitumor immunotherapies.
Collapse
Affiliation(s)
- Yanni Ge
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Jiaojiao Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Kai Jin
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Ziqiang Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Wei Wang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Zhuxian Zhou
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Karanlık CC, Karanlık G, Gok B, Budama-Kilinc Y, Kecel-Gunduz S, Erdoğmuş A. Exploring anticancer properties of novel Nano-Formulation of BODIPY Compound, Photophysicochemical, in vitro and in silico evaluations. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 301:122964. [PMID: 37302199 DOI: 10.1016/j.saa.2023.122964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
A new BODIPY complex (C4) composed of meso- thienyl-pyridine substituted core unit diiodinated from 2- and 6- positions and distyryl moieties at 3- and 5- positions is synthesized. Nano-sized formulation of C4 is prepared by single emulsion method using poly(ε-caprolactone)(PCL) polymer. Encapsulation efficiency and loading capacity values of C4 loaded PCL nanoparticles (C4@PCL-NPs) are calculated and in vitro release profile of C4 is determined. The cytotoxicity and anti-cancer activity are conducted on the L929 and MCF-7 cell lines. Cellular uptake study is performed and interaction between C4@PCL-NPs and MCF-7 cell line is investigated. Anti-cancer activity of C4 is predicted with molecular docking studies and the inhibition property on EGFR, ERα, PR and mTOR are investigated for its anticancer properties. Molecular interactions, binding positions and docking score energies between C4 and EGFR, ERα, PR and mTOR targets are revealed using in silico methods. The druglikeness and pharmacokinetic properties of C4 are evaluated using the SwissADME and its bioavailability and toxicity profiles are assessed using the SwissADME, preADMET and pkCSM servers. In conclusion, the potential use of C4 as an anti-cancer agent is evaluated in vitro and in silico methods. Also, photophysicochemical properties are studied to investigate the potential of using Photodynamic Therapy (PDT). In photochemical studies, the calculated singlet oxygen quantum yield (ΦΔ) value was 0.73 for C4 and in photopysical studies, the calculated fluorescence quantum yield ΦF value was 0.19 for C4.
Collapse
Affiliation(s)
- Ceren Can Karanlık
- Department of Chemistry, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey.
| | - Gürkan Karanlık
- Department of Chemistry, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey.
| | - Bahar Gok
- Graduate School of Natural and Applied Science, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey.
| | - Yasemin Budama-Kilinc
- Department of Bioengineering, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey; Health Biotechnology Joint Research and Application Center of Excellence, 34220, Istanbul, Turkey.
| | | | - Ali Erdoğmuş
- Department of Chemistry, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey; Health Biotechnology Joint Research and Application Center of Excellence, 34220, Istanbul, Turkey.
| |
Collapse
|
11
|
Mao Z, Kim JH, Lee J, Xiong H, Zhang F, Kim JS. Engineering of BODIPY-based theranostics for cancer therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
12
|
Stimuli-Responsive Boron-Based Materials in Drug Delivery. Int J Mol Sci 2023; 24:ijms24032757. [PMID: 36769081 PMCID: PMC9917063 DOI: 10.3390/ijms24032757] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Drug delivery systems, which use components at the nanoscale level as diagnostic tools or to release therapeutic drugs to particular target areas in a regulated manner, are a fast-evolving field of science. The active pharmaceutical substance can be released via the drug delivery system to produce the desired therapeutic effect. The poor bioavailability and irregular plasma drug levels of conventional drug delivery systems (tablets, capsules, syrups, etc.) prevent them from achieving sustained delivery. The entire therapy process may be ineffective without a reliable delivery system. To achieve optimal safety and effectiveness, the drug must also be administered at a precision-controlled rate and the targeted spot. The issues with traditional drug delivery are overcome by the development of stimuli-responsive controlled drug release. Over the past decades, regulated drug delivery has evolved considerably, progressing from large- and nanoscale to smart-controlled drug delivery for several diseases. The current review provides an updated overview of recent developments in the field of stimuli-responsive boron-based materials in drug delivery for various diseases. Boron-containing compounds such as boron nitride, boronic acid, and boron dipyrromethene have been developed as a moving field of research in drug delivery. Due to their ability to achieve precise control over drug release through the response to particular stimuli (pH, light, glutathione, glucose or temperature), stimuli-responsive nanoscale drug delivery systems are attracting a lot of attention. The potential of developing their capabilities to a wide range of nanoscale systems, such as nanoparticles, nanosheets/nanospheres, nanotubes, nanocarriers, microneedles, nanocapsules, hydrogel, nanoassembly, etc., is also addressed and examined. This review also provides overall design principles to include stimuli-responsive boron nanomaterial-based drug delivery systems, which might inspire new concepts and applications.
Collapse
|
13
|
Sonkaya Ö, Soylukan C, Pamuk Algi M, Algi F. Aza-BODIPY-based Fluorescent and Colorimetric Sensors and Probes. Curr Org Synth 2023; 20:20-60. [PMID: 35170414 DOI: 10.2174/1570179419666220216123033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/30/2021] [Accepted: 12/18/2021] [Indexed: 12/16/2022]
Abstract
Aza-boron-dipyrromethenes (Aza-BODIPYs) represent an important class of chromophores absorbing and emitting in the near-infrared (NIR) region. They have unique optical and electronic features and higher physiological and photo stability than other NIR dyes. Especially after the development of facile synthetic routes, Aza-BODIPYs have become indispensable fluors that can find various applications ranging from chemosensors, bioimaging, phototherapy, solar energy materials, photocatalysis, photon upconversion, lasers, and optoelectronics. Herein, we review Aza-BODIPY based fluorescent and colorimetric chemosensors. We show the potential and untapped toolbox of Aza-BODIPY based fluorescent and colorimetric chemosensors. Hence, we divide the fluorescent and colorimetric chemosensors and probes into five sections according to the target analytes. The first section begins with the chemosensors developed for pH. Next, we discuss Aza-BODIPY based ion sensors, including metal ions and anions. Finally, we present the chemosensors and probes concerning reactive oxygen (ROS) and nitrogen species (RNS) along with biologically relevant species in the last two sections. We believe that Aza-BODIPYs are still in their infancy, and they have a promising future for translation from the bench to real biomedical and materials science applications. After two decades of intensive research, it seems that there are many more to come in this already fertile field. Overall, we hope that future work will further expand the applications of Aza-BODIPY in many areas.
Collapse
Affiliation(s)
- Ömer Sonkaya
- Department of Chemistry, Aksaray University, TR-68100 Aksaray, Turkey
- ASUBTAM Memduh Bilmez BioNanoTech Lab., Aksaray University, TR-68100 Aksaray, Turkey
| | - Caner Soylukan
- ASUBTAM Memduh Bilmez BioNanoTech Lab., Aksaray University, TR-68100 Aksaray, Turkey
- Department of Biotechnology & ASUBTAM Memduh Bilmez BioNanoTech Lab., Aksaray University, TR-68100 Aksaray, Turkey
| | - Melek Pamuk Algi
- Department of Chemistry, Aksaray University, TR-68100 Aksaray, Turkey
- ASUBTAM Memduh Bilmez BioNanoTech Lab., Aksaray University, TR-68100 Aksaray, Turkey
| | - Fatih Algi
- ASUBTAM Memduh Bilmez BioNanoTech Lab., Aksaray University, TR-68100 Aksaray, Turkey
- Department of Biotechnology & ASUBTAM Memduh Bilmez BioNanoTech Lab., Aksaray University, TR-68100 Aksaray, Turkey
| |
Collapse
|
14
|
Li R, Ren J, Zhang D, Lv M, Wang Z, Wang H, Zhang S, Du J, Jiang XD, Wang G. Attachment of −tBu groups to aza-BODIPY core at 3,5-sites with ultra-large Stokes shift to enhance photothermal therapy through apoptosis mechanism. Mater Today Bio 2022; 16:100446. [PMID: 36199559 PMCID: PMC9527945 DOI: 10.1016/j.mtbio.2022.100446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022]
Abstract
By the introduction of the −tBu groups into aza-BODIPY core, di-tert-butyl-substituted aza-BODIPYs at 3,5-sites (tBuazaBDPs) were prepared for the first time. Based on the X-ray analysis of CN-tBuazaBDP, this molecular structure is twisted. Near-infrared dye SMe-tBuazaBDP has the ultra-large Stokes shift (152 nm) in aza-BODIPY system, combining with the twisted intramolecular charge transfer and the free rotation of the −tBu groups at 3,5-sites. Although the barrier-free rotors of the distal −tBu groups in SMe-tBuazaBDP result in low fluorescence quantum yield, the photothermal conversion efficiency is markedly enhanced. SMe-tBuazaBDP nanoparticles with low power laser irradiation were proven to block cancer cell cycle, inhibit cancer cell proliferation, and induce cancer cell apoptosis in photothermal therapy (PTT). The strategy of “direct attachment of −tBu groups to aza-BODIPY core” gives a new design platform for a photothermal therapy agent. Di-tert-butyl-substituted aza-BODIPYs at 3,5-sites (tBuazaBDPs) were prepared for the first time. Near-infrared dye SMe-tBuazaBDP has the ultra-large Stokes shift (152 nm) in aza-BODIPY system. SMe-tBuazaBDP nanoparticles can photothermally induce apoptosis as a potential photothermal therapy agent.
Collapse
|
15
|
Wang Y, Wang D, Zhang Y, Xu H, Shen L, Cheng J, Xu X, Tan H, Chen X, Li J. Tumor Microenvironment-Adaptive Nanoplatform Synergistically Enhances Cascaded Chemodynamic Therapy. Bioact Mater 2022; 22:239-253. [PMID: 36254272 PMCID: PMC9550605 DOI: 10.1016/j.bioactmat.2022.09.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
Chemodynamic therapy (CDT), a noninvasive strategy, has emerged as a promising alternative to conventional chemotherapy for treating tumors. However, its therapeutic effect is limited by the amount of H2O2, pH value, the hypoxic environment of tumors, and it has suboptimal tumor-targeting ability. In this study, tumor cell membrane-camouflaged mesoporous Fe3O4 nanoparticles loaded with perfluoropentane (PFP) and glucose oxidase (GOx) are used as a tumor microenvironment-adaptive nanoplatform (M-mFeP@O2-G), which synergistically enhances the antitumor effect of CDT. Mesoporous Fe3O4 nanoparticles are selected as inducers for photothermal and Fenton reactions and as nanocarriers. GOx depletes glucose within tumor cells for starving the cells, while producing H2O2 for subsequent ·OH generation. Moreover, PFP, which can carry O2, relieves hypoxia in tumor cells and provides O2 for the cascade reaction. Finally, the nanoparticles are camouflaged with osteosarcoma cell membranes, endowing the nanoparticles with homologous targeting and immune escape abilities. Both in vivo and in vitro evaluations reveal high synergistic therapeutic efficacy of M-mFeP@O2-G, with a desirable tumor-inhibition rate (90.50%), which indicates the great potential of this platform for clinical treating cancer. GOx and PFP were loaded in mFe3O4 to form a TME-adaptive nanoplatform and synergistically enhance the cascaded reactions. Tumor cell membranes, endowing the nanoparticles with homologous targeting and immune escape abilities. The nanoparticles had excellent combined chemodynamic therapy, starvation therapy and photothermal tumor therapy effect. Tumor cell membranes coated nanoparticles improved cell uptake and had a desirable tumor-inhibition rate.
Collapse
Affiliation(s)
- Yuemin Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Duan Wang
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyue Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Hong Xu
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Luxuan Shen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jing Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xinyuan Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xingyu Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- College of Medicine, Southwest Jiaotong University, Chengdu, 610003, China
- Corresponding author. College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
- Corresponding author. College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
16
|
Yang DC, Wen LF, Du L, Luo CM, Lu ZY, Liu JY, Lin Z. A Hypoxia-Activated Prodrug Conjugated with a BODIPY-Based Photothermal Agent for Imaging-Guided Chemo-Photothermal Combination Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:40546-40558. [PMID: 36059107 DOI: 10.1021/acsami.2c09071] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hypoxia-activated prodrugs (HAPs) have drawn increasing attention for improving the antitumor effects while minimizing side effects. However, the heterogeneous distribution of the hypoxic region in tumors severely impedes the curative effect of HAPs. Additionally, most HAPs are not amenable to optical imaging, and it is difficult to precisely trace them in tissues. Herein, we carefully designed and synthesized a multifunctional therapeutic BAC prodrug by connecting the chemotherapeutic drug camptothecin (CPT) and the fluorescent photothermal agent boron dipyrromethene (BODIPY) via hypoxia-responsive azobenzene linkers. To enhance the solubility and tumor accumulation, the prepared BAC was further encapsulated into a human serum albumin (HSA)-based drug delivery system to form HSA@BAC nanoparticles. Since the CPT was caged by a BODIPY-based molecule at the active site, the BAC exhibited excellent biosafety. Importantly, the activated CPT could be quickly released from BAC and could perform chemotherapy in hypoxic cancer cells, which was ascribed to the cleavage of the azobenzene linker by overexpressed azoreductase. After irradiation with a 730 nm laser, HSA@BAC can efficiently generate hyperthermia to achieve irreversible cancer cell death by oxygen-independent photothermal therapy. Under fluorescence imaging-guided local irradiation, both in vitro and in vivo studies demonstrated that HSA@BAC exhibited superior antitumor effects with minimal side effects.
Collapse
Affiliation(s)
- De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Lin-Feng Wen
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Liyang Du
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Cheng-Miao Luo
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zi-Yao Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zhonghui Lin
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
17
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
18
|
Gao M, Han Z, Wang Z, Zou X, Peng L, Zhao Y, Sun L. Fabrication of a smart drug delivery system based on hollow Ag 2S@mSiO 2 nanoparticles for fluorescence-guided synergistic photothermal chemotherapy. Mikrochim Acta 2022; 189:376. [PMID: 36074274 DOI: 10.1007/s00604-022-05468-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
Abstract
A novel near-infrared (NIR) light-triggered smart nanoplatform has been developed for cancer targeting and imaging-guided combined photothermal-chemo treatment. Notably, Ag2S has a dual function of photothermal therapy and fluorescence imaging, which greatly simplifies the structure of the system. It can emit fluorescence at 820 nm under an excitation wavelength of 560 nm. The phase-change molecule of 1-tetradecanol (TD) is introduced as a temperature-sensitive gatekeeper to provide the nanocarrier with controlled release capability of doxorubicin (DOX). The nanocarrier (HAg2S@mSiO2-TD/DOX) shows a high drug loading capacity of 26.3% and exhibits an apparent NIR-responsive DOX release property. Under NIR irradiation, the photothermal effect of HAg2S nanocores facilitated the release of DOX through the melting of TD. The cytotoxicity test shows that the nanocarriers have good biocompatibility. As the same time, the synergistic combination leads to a better cancer inhibition effect than individual therapy alone in vitro. Cell uptake tests indicate that the carriers have excellent fluorescence imaging ability and high cellular uptake for HepG2 cells. This work provides a new strategy for the fabrication of smart nanocarriers with simple structures for fluorescence-mediated combination cancer therapy. Fabrication of a smart drug delivery system based on hollow Ag2S@mSiO2 nanoparticles for fluorescence-guided synergistic photothermal chemotherapy.
Collapse
Affiliation(s)
- Minjie Gao
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Zehua Han
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Zhihua Wang
- Henan Engineering Research Center of Industrial Circulating Water Treatment, College of Chemistry and Chemical Engineering, Henan University, Kaifeng, 475004, China.
| | - Xueyan Zou
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Lichao Peng
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Yanbao Zhao
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Lei Sun
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
19
|
Nanomaterials in cancer: Reviewing the combination of hyperthermia and triggered chemotherapy. J Control Release 2022; 347:89-103. [PMID: 35513211 DOI: 10.1016/j.jconrel.2022.04.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 01/10/2023]
Abstract
Nanoparticle mediated hyperthermia has been explored as a method to increase cancer treatment efficacy by heating tumours inside-out. With that purpose, nanoparticles have been designed and their properties tailored to respond to external stimuli and convert the supplied energy into heat, therefore inducing damage to tumour cells. Moreover, the combination of hyperthermia with chemotherapy has been described as a more effective strategy due to the synergy between the high temperature and the drug's effects, also associated with a remote controlled and on-demand drug release. In this review, the methods behind nanoparticle mediated hyperthermia, namely material design, external stimuli response and energy conversion will be discussed and critically analysed. We will address the most relevant studies on hyperthermia and temperature triggered drug release for cancer treatment. Finally, the advantages, difficulties and challenges of this therapeutic strategy will be discussed, while giving insight for future developments.
Collapse
|
20
|
Zhang C, Yan K, Fu C, Peng H, Hawker CJ, Whittaker AK. Biological Utility of Fluorinated Compounds: from Materials Design to Molecular Imaging, Therapeutics and Environmental Remediation. Chem Rev 2022; 122:167-208. [PMID: 34609131 DOI: 10.1021/acs.chemrev.1c00632] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The applications of fluorinated molecules in bioengineering and nanotechnology are expanding rapidly with the controlled introduction of fluorine being broadly studied due to the unique properties of C-F bonds. This review will focus on the design and utility of C-F containing materials in imaging, therapeutics, and environmental applications with a central theme being the importance of controlling fluorine-fluorine interactions and understanding how such interactions impact biological behavior. Low natural abundance of fluorine is shown to provide sensitivity and background advantages for imaging and detection of a variety of diseases with 19F magnetic resonance imaging, 18F positron emission tomography and ultrasound discussed as illustrative examples. The presence of C-F bonds can also be used to tailor membrane permeability and pharmacokinetic properties of drugs and delivery agents for enhanced cell uptake and therapeutics. A key message of this review is that while the promise of C-F containing materials is significant, a subset of highly fluorinated compounds such as per- and polyfluoroalkyl substances (PFAS), have been identified as posing a potential risk to human health. The unique properties of the C-F bond and the significant potential for fluorine-fluorine interactions in PFAS structures necessitate the development of new strategies for facile and efficient environmental removal and remediation. Recent progress in the development of fluorine-containing compounds as molecular imaging and therapeutic agents will be reviewed and their design features contrasted with environmental and health risks for PFAS systems. Finally, present challenges and future directions in the exploitation of the biological aspects of fluorinated systems will be described.
Collapse
Affiliation(s)
- Cheng Zhang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Kai Yan
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
- National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an 710021, China
- Xi'an Key Laboratory of Green Chemicals and Functional Materials, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Changkui Fu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hui Peng
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig J Hawker
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry & Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
21
|
Yang YL, Lin K, Yang L. Progress in Nanocarriers Codelivery System to Enhance the Anticancer Effect of Photodynamic Therapy. Pharmaceutics 2021; 13:1951. [PMID: 34834367 PMCID: PMC8617654 DOI: 10.3390/pharmaceutics13111951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising anticancer noninvasive method and has great potential for clinical applications. Unfortunately, PDT still has many limitations, such as metastatic tumor at unknown sites, inadequate light delivery and a lack of sufficient oxygen. Recent studies have demonstrated that photodynamic therapy in combination with other therapies can enhance anticancer effects. The development of new nanomaterials provides a platform for the codelivery of two or more therapeutic drugs, which is a promising cancer treatment method. The use of multifunctional nanocarriers for the codelivery of two or more drugs can improve physical and chemical properties, increase tumor site aggregation, and enhance the antitumor effect through synergistic actions, which is worthy of further study. This review focuses on the latest research progress on the synergistic enhancement of PDT by simultaneous multidrug administration using codelivery nanocarriers. We introduce the design of codelivery nanocarriers and discuss the mechanism of PDT combined with other antitumor methods. The combination of PDT and chemotherapy, gene therapy, immunotherapy, photothermal therapy, hyperthermia, radiotherapy, sonodynamic therapy and even multidrug therapy are discussed to provide a comprehensive understanding.
Collapse
Affiliation(s)
| | | | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.-L.Y.); (K.L.)
| |
Collapse
|
22
|
Hybridized double-shell periodic mesoporous organosilica nanotheranostics for ultrasound imaging guided photothermal therapy. J Colloid Interface Sci 2021; 608:2964-2972. [PMID: 34799047 DOI: 10.1016/j.jcis.2021.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/29/2021] [Accepted: 11/06/2021] [Indexed: 01/27/2023]
Abstract
Hybridized periodic mesoporous organosilica (PMO) nanoparticles are expected to provide a multifunctional theranostic platform for precision medicine by combining the advantages of different organic and inorganic components. In this work, double-shell-structured PMO nanotheranostics composed of ethane- and thioether-bridged organosilica shells were synthesized. Gold colloids were generated in situ by the thioether groups on the inner shell. The obtained double-shell PMO@Au (DSPA) has uniform size, large surface areas, ordered mesochannels and photothermal conversion capability. After being encapsulated with perfluorohexacene (PFH), DSPA-PFH produced a strong ultrasound signal upon laser irradiation due to the phase transit of PFH during hyperthermia. DSPA-PFH showed enhanced photothermal therapeutic efficacy, great ultrasound contrast, and minimal toxicity both in vitro and in vivo. These results demonstrated the distribution of different organosilica could be delicately adjusted in hybridized PMO nanoparticles. Furthermore, it showed the potential of using hybridized PMO nanoparticles as a theranostic platform for biomedical applications by combining unique characteristics of different organosilica through rational design.
Collapse
|
23
|
Lin L, Song X, Dong X, Li B. Nano-photosensitizers for enhanced photodynamic therapy. Photodiagnosis Photodyn Ther 2021; 36:102597. [PMID: 34699982 DOI: 10.1016/j.pdpdt.2021.102597] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 12/22/2022]
Abstract
Photodynamic therapy (PDT) utilizes photosensitizers (PSs) together with irradiation light of specific wavelength interacting with oxygen to generate cytotoxic reactive oxygen species (ROS), which could trigger apoptosis and/or necrosis-induced cell death in target tissues. During the past two decades, multifunctional nano-PSs employing nanotechnology and nanomedicine developed, which present not only photosensitizing properties but additionally accurate drug release abilities, efficient response to optical stimuli and hypoxia resistance. Further, nano-PSs have been developed to enhance PDT efficacy by improving the ROS yield. In addition, nano-PSs with additive or synergistic therapies are significant for both currently preclinical study and future clinical practice, given their capability of considerable higher therapeutic efficacy under safer systemic drug dosage. In this review, nano-PSs that allow precise drug delivery for efficient absorption by target cells are introduced. Nano-PSs boosting sensitivity and conversion efficiency to PDT-activating stimuli are highlighted. Nano-PSs developed to address the challenging hypoxia conditions during PDT of deep-sited tumors are summarized. Specifically, PSs capable of synergistic therapy and the emerging novel types with higher ROS yield that further enhance PDT efficacy are presented. Finally, future demands for ideal nano-PSs, emphasizing clinical translation and application are discussed.
Collapse
Affiliation(s)
- Li Lin
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics and Institute of Advanced Materials, Nanjing Technology University, Nanjing 211800, China
| | - Xiaocheng Dong
- Key Laboratory of Flexible Electronics and Institute of Advanced Materials, Nanjing Technology University, Nanjing 211800, China
| | - Buhong Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China.
| |
Collapse
|
24
|
Zhu J, Wang X, Yang D, Song X, Li B, Wang W, Dong X. Ultrasound‐Triggered In Situ Gelation to Overcome Tumor Hypoxia for Enhanced Photodynamic and Sustained Chemotherapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jiawei Zhu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) School of Physical and Mathematical Sciences Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Xiaorui Wang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) School of Physical and Mathematical Sciences Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) School of Physical and Mathematical Sciences Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) School of Physical and Mathematical Sciences Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Buhong Li
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education Fujian Provincial Key Laboratory of Photonics Technology Fujian Normal University Fuzhou 350007 China
| | - Wenjun Wang
- School of Physical Science and Information Technology Liaocheng University Liaocheng 252059 China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) School of Physical and Mathematical Sciences Nanjing Tech University (NanjingTech) Nanjing 211800 China
- School of Chemistry and Materials Science Nanjing University of Information Science & Technology Nanjing 210044 China
| |
Collapse
|
25
|
Zong J, Peng H, Qing X, Fan Z, Xu W, Du X, Shi R, Zhang Y. pH-Responsive Pluronic F127-Lenvatinib-Encapsulated Halogenated Boron-Dipyrromethene Nanoparticles for Combined Photodynamic Therapy and Chemotherapy of Liver Cancer. ACS OMEGA 2021; 6:12331-12342. [PMID: 34056385 PMCID: PMC8154152 DOI: 10.1021/acsomega.1c01346] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/23/2021] [Indexed: 06/12/2023]
Abstract
Combination therapy such as photodynamic therapy (PDT)-enhanced chemotherapy is regarded as a promising strategy for cancer treatment. Boron-dipyrromethene (BODIPY), as close relatives of porphyrins, was widely used in PDT. However, poor water solubility, rapid metabolism by the body and lack of targeting limits its clinical application. Lenvatinib, as the first-line drug for molecular-targeted therapy of liver cancer, restricted its clinical application for its side effects. Herein, to achieve the synergy between PDT and chemotherapy, we synthesized two halogenated BODIPY, BDPBr2 and BDPCl2, which were prepared into self-assembly nanoparticles with lenvatinib, and were encapsulated with Pluronic F127 through the nanoprecipitation method, namely, LBPNPs (LBBr2 NPs and LBCl2 NPs). The fluorescence quantum yields of LBPNPs were 0.73 and 0.71, respectively. The calculated loading rates of lenvatinib for LBBr2 NPs and LBCl2 NPs were 11.8 and 10.2%, respectively. LBPNPs can be hydrolyzed under weakly acidic conditions (pH 5.0) to generate reactive oxygen species (ROS), and the release rate of lenvatinib reached 88.5 and 82.4%. Additionally, LBPNPs can be effectively taken up by Hep3B and Huh7 liver cancer cells, releasing halogenated BODIPY and lenvatinib in the acidic environment of tumor cells to enhance the targeting performance of chemotherapeutics. Compared with free lenvatinib and separate halogenated BODIPY, LBPNPs can inhibit tumor growth more effectively through pH-responsive chemo/photodynamic synergistic therapy and significantly promote the cascade of caspase apoptotic protease. This study shows that LBPNPs can be a promising nanotheranostic agent for synergetic chemo/photodynamic liver cancer therapy.
Collapse
Affiliation(s)
- Jingjing Zong
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hao Peng
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xin Qing
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhe Fan
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
- The
Third People’s Hospital of Dalian, Dalian Medical University, Dalian 116033, China
| | - Wenjing Xu
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xuanlong Du
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ruihua Shi
- Department
of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao, Nanjing 210009, China
| | - Yewei Zhang
- Department
of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
26
|
Qin D, Zhang L, Zhu H, Chen J, Wu D, Bouakaz A, Wan M, Feng Y. A Highly Efficient One-for-All Nanodroplet for Ultrasound Imaging-Guided and Cavitation-Enhanced Photothermal Therapy. Int J Nanomedicine 2021; 16:3105-3119. [PMID: 33967577 PMCID: PMC8096805 DOI: 10.2147/ijn.s301734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Photothermal therapy (PTT) has attracted considerable attention for cancer treatment as it is highly controllable and minimally invasive. Various multifunctional nanosystems have been fabricated in an "all-in-one" form to guide and enhance PTT by integrating imaging and therapeutic functions. However, the complex fabrication of nanosystems and their high cost limit its clinical translation. MATERIALS AND METHODS Herein, a high efficient "one-for-all" nanodroplet with a simple composition but owning multiple capabilities was developed to achieve ultrasound (US) imaging-guided and cavitation-enhanced PTT. Perfluoropentane (PFP) nanodroplet with a polypyrrole (PPy) shell (PFP@PPy nanodroplet) was synthesized via ultrasonic emulsification and in situ oxidative polymerization. After characterization of the morphology, its photothermal effect, phase transition performance, as well as its capabilities of enhancing US imaging and acoustic cavitation were examined. Moreover, the antitumor efficacy of the combined therapy with PTT and acoustic cavitation via the PFP@PPy nanodroplets was studied both in vitro and in vivo. RESULTS The nanodroplets exhibited good stability, high biocompatibility, broad optical absorption over the visible and near-infrared (NIR) range, excellent photothermal conversion with an efficiency of 60.1% and activatable liquid-gas phase transition performance. Upon NIR laser and US irradiation, the phase transition of PFP cores into microbubbles significantly enhanced US imaging and acoustic cavitation both in vitro and in vivo. More importantly, the acoustic cavitation enhanced significantly the antitumor efficacy of PTT as compared to PTT alone thanks to the cavitation-mediated cell destruction, which demonstrated a substantial increase in cell detachment, 81.1% cell death in vitro and 99.5% tumor inhibition in vivo. CONCLUSION The PFP@PPy nanodroplet as a "one-for-all" theranostic agent achieved highly efficient US imaging-guided and cavitation-enhanced cancer therapy, and has considerable potential to provide cancer theranostics in the future.
Collapse
Affiliation(s)
- Dui Qin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, People’s Republic of China
| | - Lei Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Hongrui Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Junjie Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, F-37032, France
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yi Feng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
27
|
Plasma membrane targeted photodynamic O 2 economizer for hypoxic tumor therapy. Biomaterials 2021; 273:120854. [PMID: 33932703 DOI: 10.1016/j.biomaterials.2021.120854] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/31/2022]
Abstract
The development of photodynamic therapy (PDT) is severely limited by short half-life of singlet oxygen (1O2) and the hypoxic microenvironment. In this work, a plasma membrane targeted photodynamic O2 economizer (designated as P-POE) is developed to improve the subcellular delivery of photosensitizers and alleviate the tumor hypoxia for enhanced PDT effect. After self-assembly into nanomicelles, P-POE has a relatively high stability and a favorable photochemical performance, which are conducive to boosting the 1O2 production. Besides, the plasma membrane anchoring of P-POE contributes to enhancing the preferential retention and cellular accumulation of photosensitizers on tumor tissues and cells. More importantly, P-POE-induced mitochondrial respiratory depression is demonstrated to reduce the O2 consumption of tumor cells to relieve the hypoxia. Consequently, P-POE still exhibits a robust PDT effect against hypoxic tumors, which greatly inhibits the proliferation of breast cancer with low adverse reactions. This innovative combination of subcellular targeting and hypoxic alleviation would advance the development of individualized drug delivery systems for photodynamic therapy against hypoxic tumors.
Collapse
|
28
|
Cao C, Yang N, Dai H, Huang H, Song X, Zhang Q, Dong X. Recent advances in phase change material based nanoplatforms for cancer therapy. NANOSCALE ADVANCES 2021; 3:106-122. [PMID: 36131875 PMCID: PMC9419072 DOI: 10.1039/d0na00622j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/12/2020] [Indexed: 05/06/2023]
Abstract
Cancer has become a severe threat to human life due to its high mortality and metastatic rate. Effective inhibition and killing of cancer cells using chemotherapeutic drugs have been a promising means in clinical cancer therapy. However, the low selectivity, drug-resistance, uncontrollability and serious side effects of chemotherapy significantly limit its further development. There is an urgent need for new treatment strategies to compensate for deficiencies inherent in chemotherapy alone. A growing body of research shows that combined treatment strategies have the potential to overcome this dilemma by achieving significantly enhanced synergistic effects and reduced side effects. Emerging phase change materials (PCMs) create an ideal nanoplatform for cancer combination therapy due to their universal loading properties, stable and temperature-responsive phase transition capability, and excellent natural biocompatibility/biodegradability. The release of therapeutic agents can be precisely controlled through external, non-intrusive stimuli (such as NIR light and ultrasound), avoiding systemic toxicity associated with conventional chemotherapy. Herein, the construction methods and design principles of PCM-based nanoplatforms serving as strict gatekeeper and smart payload delivery systems are discussed in detail. Moreover, the advantages and disadvantages of these nanoplatforms are provided. A suitable discussion and perspective of the remaining challenges and future opportunities for PCM-based nanoplatforms in cancer treatment are also given in conclusion.
Collapse
Affiliation(s)
- Changyu Cao
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Nan Yang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Hanming Dai
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Han Huang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech) Nanjing 211800 China
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology Nanjing 210044 China
| |
Collapse
|
29
|
Depletion of glioma stem cells by synergistic inhibition of mTOR and c-Myc with a biological camouflaged cascade brain-targeting nanosystem. Biomaterials 2020; 268:120564. [PMID: 33296794 DOI: 10.1016/j.biomaterials.2020.120564] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022]
Abstract
Glioma stem cells (GSCs), as a subpopulation of stem cell-like cells, have been proposed to play a crucial role in the progression of drug-resistance in glioblastoma (GBM). Therefore, the targeted eradication of GSCs can serve as a promising therapeutic strategy for the reversal of drug-resistance in GBM. Herein, the effects of silencing c-Myc and m-TOR on primary GBM cells extracted from patients were investigated. Results confirmed that dual inhibition treatment significantly (p < 0.05) and synergistically suppressed GSCs, and consequently reversed TMZ-resistance when compared with the single treatment group. Subsequently, to facilitate effective crossing of the BBB, a biological camouflaged cascade brain-targeting nanosystem (PMRT) was created. The PMRT significantly inhibited tumor growth and extended the lifespan of orthotopic transplantation TMZ-resistant GBM-grafted mice. Our data demonstrated that PMRT could precisely facilitate drug release at the tumor site across the BBB. Simultaneously, c-Myc and m-TOR could serve as synergistic targets to eradicate the GSCs and reverse GBM resistance to TMZ.
Collapse
|
30
|
Zhang Y, Dong Y, Fu H, Huang H, Wu Z, Zhao M, Yang X, Guo Q, Duan Y, Sun Y. Multifunctional tumor-targeted PLGA nanoparticles delivering Pt(IV)/siBIRC5 for US/MRI imaging and overcoming ovarian cancer resistance. Biomaterials 2020; 269:120478. [PMID: 33213862 DOI: 10.1016/j.biomaterials.2020.120478] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (Pt(II)) resistance is an important factor in the high mortality rates of ovarian cancer. Herein, we synthesized multifunctional tumor-targeted poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs-cRGD) for monitoring therapeutic effects by dual-mode imaging and overcoming cisplatin resistance. Uniformly sized NPs-cRGD demonstrated controlled and sustained release of drugs and genes, excellent gene loading and gene protection capacity, good storage stability and no serum-induced aggregation in vitro. NPs-cRGD demonstrated clear, targeting and prolonged ultrasound imaging and magnetic resonance imaging (MRI) in vivo. The targeting of NPs-cRGD combined with ultrasound facilitated nanoparticle penetrattion into cells; entry was time-dependent. NPs-cRGD escaped from lysosomes, thereby preventing siBIRC5 degradation, which enabled siBIRC5 to efficiently inhibit the antiapoptosis effects of BIRC5 in SKO3-DDP to overcome the antiapoptosis properties of resistant cells. Furthermore, Pt(IV) in NPs-cRGD exhausted glutathione (GSH), thereby increasing drug accumulation to effectively increase Pt(II) levels. The subsequent combination of Pt(II) with DNA prevented the expressions of genes and upregulated the expression of p53 to induce the mitochondria apoptosis pathway. The reduced GSH activity and the generation of Pt(II) further promoted high levels of reactive oxygen species (ROS) to induce cell apoptosis. Therefore, NPs-cRGD with ultrasound promoted the apoptosis of resistant ovarian cancer cells by multiple mechanisms, including increased cellular drug accumulation, reversed antiapoptotic effects by siBIRC5, and enhanced ROS levels. In a tumor-bearing nude mice model, NPs-cRGD with US demonstrated excellent tumor-targeting, high efficiency tumor inhibition and low systemic toxicity. Therefore, NPs-cRGD provides a means to monitor treatment processes and can be combined with ultrasound treatment to overcome ovarian cancer resistance in vitro and in vivo.
Collapse
Affiliation(s)
- Yanhua Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Zhihua Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Meng Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xupeng Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
31
|
Zhao Z, Wijerathne H, Godwin AK, Soper SA. Isolation and analysis methods of extracellular vesicles (EVs). EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2020; 2:80-103. [PMID: 34414401 PMCID: PMC8372011 DOI: 10.20517/evcna.2021.07] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) have been recognized as an evolving biomarker within the liquid biopsy family. While carrying both host cell proteins and different types of RNAs, EVs are also present in sufficient quantities in biological samples to be tested using many molecular analysis platforms to interrogate their content. However, because EVs in biological samples are comprised of both disease and non-disease related EVs, enrichment is often required to remove potential interferences from the downstream molecular assay. Most benchtop isolation/enrichment methods require > milliliter levels of sample and can cause varying degrees of damage to the EVs. In addition, some of the common EV benchtop isolation methods do not sort the diseased from the non-diseased related EVs. Simultaneously, the detection of the overall concentration and size distribution of the EVs is highly dependent on techniques such as electron microscopy and Nanoparticle Tracking Analysis, which can include unexpected variations and biases as well as complexity in the analysis. This review discusses the importance of EVs as a biomarker secured from a liquid biopsy and covers some of the traditional and non-traditional, including microfluidics and resistive pulse sensing, technologies for EV isolation and detection, respectively.
Collapse
Affiliation(s)
- Zheng Zhao
- Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA
| | - Harshani Wijerathne
- Department of Mechanical Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Andrew K. Godwin
- KU Cancer Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Steven A. Soper
- Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, University of Kansas, Lawrence, KS 66045, USA
- KU Cancer Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Ulsan National Institute of Science & Technology, Ulju-gun, Ulsan, 44919, South Korea
| |
Collapse
|