1
|
Guo R, Zhang R, Xin Y, Wang Z, Xu Z, Qiu J. Recent developments in photothermal therapy: a bibliometric and visual analysis. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:892-912. [PMID: 39645603 DOI: 10.1080/09205063.2024.2434308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Photothermal therapy (PTT) has recently garnered significant attention as a prominent noninvasive treatment modality for a broad spectrum of diseases. Despite the increasing volume of scholarly output over the last 20 years, a holistic synthesis that delineates worldwide research trajectories remains elusive. We undertook a bibliometric analysis of the literature from 2004 to 2023, aiming to delineate the prevailing focal points and illuminate prospective research avenues. Research articles on PTT were retrieved from the Web of Science Core Collection. Using tools such as CiteSpace, VOSviewer, and Bibliometrix, we comprehensively analyzed and visualized 11,184 published academic PTT papers. China has the highest number of publications. Journals related to PTT are primarily comprised of interdisciplinary and comprehensive journals. Research associated with PTT has focused primarily on its antitumor properties. Current focal areas in this domain include the synergistic combination of PTT with photodynamic therapy, immunological mechanisms of PTT to enhance its therapeutic efficacy, integrated use of PTT with nanoenzyme catalysis, and the role of PTT in antimicrobial applications. This bibliometric analysis provides an initial comprehensive examination of the medical applications of PTT, offering insights into the global research landscape, key areas of interest, and emerging trends, thereby serving as a valuable reference for future studies in this field.
Collapse
Affiliation(s)
- Runying Guo
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Medical College, Nanchang University, Nanchang, China
| | - Rongrong Zhang
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Medical College, Nanchang University, Nanchang, China
| | - Yuqi Xin
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Medical College, Nanchang University, Nanchang, China
| | - Zhonghao Wang
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Medical College, Nanchang University, Nanchang, China
| | - Zichen Xu
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Medical College, Nanchang University, Nanchang, China
| | - Jiaxuan Qiu
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Fu Y, Sun J, Yang C, Li W, Wang Y. Diversified nanocarrier design to optimize glucose oxidase-mediated anti-tumor therapy: Strategy and progress. Int J Biol Macromol 2025; 306:141581. [PMID: 40023419 DOI: 10.1016/j.ijbiomac.2025.141581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Given the inherent complexity and heterogeneity of tumors, current therapeutic approaches often fall short in meeting prognostic requirements. Starvation therapy (ST) utilizing glucose oxidase (GOx) has emerged as a promising strategy, specifically targeting tumor glucose consumption to disrupt nutrient supply. However, the therapeutic potential of GOx is significantly hampered by its inherent limitations as a protein, particularly its poor stability and short in vivo half-life. In recent years, the development of nanocarriors has provided an effective platform for intravenous and local tumor delivery of GOx. This review systematically examines three key strategies in GOx delivery: stimulus-response, biofilm modification, and local delivery. The progress in various carrier systems for GOx-mediated tumor therapy is comprehensively summarized, providing valuable insights for nanocarrier design. Furthermore, the existing challenges and future directions to advance the development of GOx-based tumor therapies are critically analyzed.
Collapse
Affiliation(s)
- Yuhan Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jialin Sun
- Department of medicine, Heilongjiang Minzu College, Harbin, Heilongjiang Province, China
| | - Chunyu Yang
- Department of Pathology, Harbin 242 Hospital, Harbin, Heilongjiang Province, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
3
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
4
|
Fu J, Chen H, Zhao Y, Xi H, Huang Y, Liu C, Wu Y, Song W, Liu X, Du B, Sun G. Self-assembled injectable Icariin@ Ti 3C 2Tx/doxorubicin hydrogel preserving osteogenesis while synergizing photodynamic and chemodynamic therapy for osteosarcoma. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2025; 36:28. [PMID: 40088393 PMCID: PMC11910421 DOI: 10.1007/s10856-025-06874-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/20/2025] [Indexed: 03/17/2025]
Abstract
Local therapy involving injectable hydrogel systems loaded with doxorubicin (DOX) has garnered significant attention in the realm of osteosarcoma (OS) research. Nevertheless, it has been noted that the local delivery of high-dose DOX exerts a pronounced inhibitory impact on osteogenesis, which is detrimental to the restoration of functional capabilities after OS treatment. To address this challenge, we have designed a self-assembled injectable hydrogel system that integrates photodynamic and chemodynamic therapy, aiming to enhance efficacy while mitigating adverse effects on osteogenic differentiation. In this study, an injectable sodium alginate (SA) hydrogel was fabricated by encapsulating titanium carbide powder (Ti3C2Tx) and osteoprotegerin Icariin (ICA) along with DOX. This hydrogel system demonstrated remarkable drug-loading capacity and sustained drug release. Furthermore, under near-infrared (NIR) irradiation, the hydrogel displayed outstanding photothermal effects, which, in conjunction with chemotherapy and phototherapy, effectively eradicated UMR-106 tumor cells in vitro. The incorporation of ICA not only enhanced the anti-tumor effect but also alleviated the adverse effects of DOX on the osteogenic differentiation inhibition of bone marrow mesenchymal stem cells (BMSCs). In vivo, findings further confirmed that injectable ITD/SA hydrogels can synergistically heighten anti-osteosarcoma effectiveness while mitigating local osteogenic toxicity. Given these benefits, this hydrogel holds extensive application prospects in the local therapy of OS.
Collapse
Affiliation(s)
- Jiahao Fu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Hao Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Yiqiao Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Hongzhong Xi
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Yixuan Huang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Chenglin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Yaokun Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Wei Song
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Xin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China.
| | - Bin Du
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China.
| | - Guangquan Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
- Department of Orthopaedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Wang L, Xiong M, Li S, Ma S, Jiang S, Wang H, Zhang J, Li X. Beneficial effects of EGCG on boar sperm quality during liquid storage at 4 °C are mediated by DRD2 receptor. Theriogenology 2025; 234:174-185. [PMID: 39718310 DOI: 10.1016/j.theriogenology.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
Epigallocatechin gallate (EGCG), a natural antioxidant, plays a vital role in modulating sperm function, yet its protective impact on boar spermatozoa during liquid preservation at 4 °C remains elusive. This study aimed to investigate the beneficial effects of EGCG on boar semen preservation, and elucidate the potential mechanism. Multiple parameters including sperm quality, anti-oxidative status, protein phosphorylation levels, membrane receptor and cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathways were analyzed using computer-assisted semen analysis system, Western blot and molecular docking techniques. Results revealed that supplementation with EGCG, particularly with 10 μg/mL, significantly increased sperm motility, acrosome integrity, mitochondrial membrane potential and intracellular ATP content. Moreover, EGCG enhanced the antioxidant defenses of sperm through eliminating excessive reactive oxygen species. Intriguingly, the antioxidant property of EGCG partly prevented protein dephosphorylation, thereby indirectly enhancing protein phosphorylation. Additionally, the dopamine receptor (DRD2) was detected in boar spermatozoa and inhibition of DRD2 greatly prevented EGCG-caused enhancement of protein phosphorylation levels and sperm motility, suggesting the role of DRD2 in regulation of the beneficial effects of EGCG. Molecular docking results indicated that EGCG has favorable binding interactions with the active sites of DRD2, involving crucial hydrogen bonding and hydrophobic interactions, further suggesting that EGCG might directly interact with DRD2, mediate protein phosphorylation via activating the DRD2/cAMP/PKA pathway and thus boost sperm motility. The present study is the first to explore the interacting cell-surface receptor of EGCG on boar sperm and provides comprehensive insights into the protective mechanism of EGCG during hypothermic liquid storage.
Collapse
Affiliation(s)
- Lirui Wang
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Manyi Xiong
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Sisi Li
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Sheng Ma
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shengyao Jiang
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haolei Wang
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jian Zhang
- Department of Agriculture, Hetao College, Bayannur, 015000, China
| | - Xinhong Li
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
6
|
Li Y, Miao W, Yuan C, Tang J, Zhong N, Jin Y, Hu Y, Tang Y, Wang S. PARP inhibitor boost the efficacy of photothermal therapy to TNBC through enhanced DNA damage and inhibited homologous recombination repair. Drug Deliv Transl Res 2025; 15:955-967. [PMID: 38954244 DOI: 10.1007/s13346-024-01650-6] [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] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Triple-negative breast cancer (TNBC) could benefit from PARP inhibitors (PARPi) for their frequent defective homologous recombination repair (HR). However, the efficacy of PARPi is limited by their lower bioavailability and high susceptibility to drug resistance, so it often needs to be combined with other treatments. Herein, polydopamine nanoparticles (PDMN) were constructed to load Olaparib (AZD) as two-channel therapeutic nanoplatforms. The PDMN has a homogeneous spherical structure around 100 nm and exhibits a good photothermal conversion efficiency of 62.4%. The obtained AZD-loaded nanoplatform (PDMN-AZD) showed enhanced antitumor effects through the combination of photothermal therapy (PTT) and PARPi. By western blot and flow cytometry, we found that PTT and PARPi could exert synergistic antitumor effects by further increasing DNA double-strand damage (DSBs) and enhancing HR defects. The strongest therapeutic effect of PDMN-AZD was observed in a BRCA-deficient mouse tumor model. In conclusion, the PDMN-AZD nanoplatform designed in this study demonstrated the effectiveness of PTT and PARPi for synergistic treatment of TNBC and preliminarily explained the mechanism.
Collapse
Affiliation(s)
- Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Wenfang Miao
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Chen Yuan
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Jiajia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Nan Zhong
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Yingying Jin
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Yongzhi Hu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300, Guangzhoulu, Nanjing, Jiangsu, China.
| |
Collapse
|
7
|
Wang L, Xiong M, Zhang J, Li S, Ma S, Jiang S, Jiang Y, Li X. Polydopamine-based nano-protectant for prolonged boar semen preservation by eliminating ROS and regulating protein phosphorylation via D2DR-mediated cAMP/PKA signaling pathway. J Nanobiotechnology 2025; 23:151. [PMID: 40016742 PMCID: PMC11869443 DOI: 10.1186/s12951-025-03215-2] [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: 09/03/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Preservation of porcine semen is essential for artificial insemination and genetic improvement in pig breeding programs. However, the overproduction of reactive oxygen species (ROS) and lower levels of protein phosphorylation emerge as two challenges during semen preservation. Inspired by the innate ligand-receptor binding biofunction of dopamine, herein, a dual-task nano-protectant that combines ROS-scavenging and protein phosphorylation-regulating properties via incorporating the natural antioxidant epigallocatechin gallate (EGCG) into polydopamine nanoparticles (EGCG@PDA NPs) was proposed to enhance the quality of pig semen during storage at 4 ℃. The results suggested that EGCG@PDA NPs significantly maintained sperm motility, acrosome integrity and mitochondrial membrane potential, extending semen storage time from 3 days to 10 days. Furthermore, EGCG@PDA NPs effectively scavenged excess ROS and inhibited ROS-mediated sperm apoptosis through the extracellular regulated protein kinases (ERK) signaling pathway. Intriguingly, EGCG@PDA NPs could degrade into ultrasmall particles (< 10 nm) in the semen or H2O2 systems. These particles could target and activate the dopamine D2 receptor (D2DR) on membrane surface of sperm midpiece, thereby enhancing protein phosphorylation via the downstream cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathway, ultimately improving sperm motility parameters. This study presents a novel nano-strategy to boost the quality of pig semen, offering significant implications for the pig industry.
Collapse
Affiliation(s)
- Lirui Wang
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Manyi Xiong
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jian Zhang
- Department of Agriculture, Hetao College, Bayannur, 015000, China
| | - Sisi Li
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Sheng Ma
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shengyao Jiang
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanping Jiang
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinhong Li
- Shanghai Key Laboratory for Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
8
|
Hu Y, Zhou Y, Li K, Zhou D. Recent advances in near-infrared stimulated nanohybrid hydrogels for cancer photothermal therapy. Biomater Sci 2024; 12:4590-4606. [PMID: 39136645 DOI: 10.1039/d4bm00662c] [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: 09/11/2024]
Abstract
Nanomedicine has emerged as a promising avenue for advancing cancer treatment, but the challenge of mitigating its in vivo side effects necessitates the development of innovative structures and materials. Recent investigation has unveiled nanogels as particularly compelling candidates, characterized by a porous, three-dimensional network architecture that exhibits exceptional drug loading capacity. Beyond this, nanogels boast a substantial specific surface area and can be tailored with specific chemical functionalities. Consequently, nanogels are frequently engineered as a multi-modal synergistic platform for combating cancer, wherein photothermal therapy stands out due to its capacity to penetrate deep tissues and achieve localized tumor eradication through the application of elevated temperatures. In this review, we delve into the synthesis of diverse varieties of photothermal nanogels capable of controlled drug release triggered by either chemical or physical stimuli. It also summarizes their potential for synergistic integration with photothermal therapy alongside other therapeutic modalities to realize effective tumor ablation. Moreover, we analyze the primary mechanisms underlying the contribution of photothermal nanogels to cancer treatment while underscoring their adeptness in regulating therapeutic temperatures for repairing bone defects resulting from tumor-associated trauma. Envisioned as an auspicious strategy in the realm of cancer therapy, photothermal nanogels hold promise for furnishing controlled drug delivery and precise thermal ablation capabilities.
Collapse
Affiliation(s)
- Yongjun Hu
- Department of Oncology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yi Zhou
- Huanggang Central Hospital of Yangtze University, Huanggang, 438000, China
| | - Kaichun Li
- Department of Oncology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Dong Zhou
- Engineering Research Centre for Biomedical Materials of Ministry of Education, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
9
|
Zhang M, Mi M, Hu Z, Li L, Chen Z, Gao X, Liu D, Xu B, Liu Y. Polydopamine-Based Biomaterials in Orthopedic Therapeutics: Properties, Applications, and Future Perspectives. Drug Des Devel Ther 2024; 18:3765-3790. [PMID: 39219693 PMCID: PMC11363944 DOI: 10.2147/dddt.s473007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Polydopamine is a versatile and modifiable polymer, known for its excellent biocompatibility and adhesiveness. It can also be engineered into a variety of nanoparticles and biomaterials for drug delivery, functional modification, making it an excellent choice to enhance the prevention and treatment of orthopedic diseases. Currently, the application of polydopamine biomaterials in orthopedic disease prevention and treatment is in its early stages, despite some initial achievements. This article aims to review these applications to encourage further development of polydopamine for orthopedic therapeutic needs. We detail the properties of polydopamine and its biomaterial types, highlighting its superior performance in functional modification on nanoparticles and materials. Additionally, we also explore the challenges and future prospects in developing optimal polydopamine biomaterials for clinical use in orthopedic disease prevention and treatment.
Collapse
Affiliation(s)
- Min Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Man Mi
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Zilong Hu
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Lixian Li
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Zhiping Chen
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People’s Republic of China
| | - Di Liu
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Bilian Xu
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Yanzhi Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| |
Collapse
|
10
|
Alimohammadvand S, Kaveh Zenjanab M, Mashinchian M, Shayegh J, Jahanban-Esfahlan R. Recent advances in biomimetic cell membrane-camouflaged nanoparticles for cancer therapy. Biomed Pharmacother 2024; 177:116951. [PMID: 38901207 DOI: 10.1016/j.biopha.2024.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
The emerging strategy of biomimetic nanoparticles (NPs) via cellular membrane camouflage holds great promise in cancer therapy. This scholarly review explores the utilization of cellular membranes derived from diverse cellular entities; blood cells, immune cells, cancer cells, stem cells, and bacterial cells as examples of NP coatings. The camouflaging strategy endows NPs with nuanced tumor-targeting abilities such as self-recognition, homotypic targeting, and long-lasting circulation, thus also improving tumor therapy efficacy overall. The comprehensive examination encompasses a variety of cell membrane camouflaged NPs (CMCNPs), elucidating their underlying targeted therapy mechanisms and delineating diverse strategies for anti-cancer applications. Furthermore, the review systematically presents the synthesis of source materials and methodologies employed in order to construct and characterize these CMCNPs, with a specific emphasis on their use in cancer treatment.
Collapse
Affiliation(s)
- Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Mashinchian
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Shayegh
- Department of Microbiology, Faculty of Veterinary and Agriculture, Islamic Azad University, Shabestar branch, Shabestar, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Hu K, Zhang D, Ma W, Gu Y, Zhao J, Mu X. Polydopamine-Based Nanoparticles for Synergistic Chemotherapy of Prostate Cancer. Int J Nanomedicine 2024; 19:6717-6730. [PMID: 38979530 PMCID: PMC11230127 DOI: 10.2147/ijn.s468946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction Immune regulatory small molecule JQ1 can block its downstream effector PD-L1 pathway and effectively reverse the PD-L1 upregulation induced by doxorubicin (DOX). So the synergistic administration of chemotherapeutic drug DOX and JQ1 is expected to increase the sensitivity of tumors to immune checkpoint therapy and jointly enhance the body's own immunity, thus effectively killing tumor cells. Therefore, a drug delivery system loaded with DOX and JQ1 was devised in this study. Methods Polydopamine nanoparticles (PDA NPs) were synthesized through spontaneous polymerization. Under appropriate pH conditions, DOX and JQ1 were loaded onto the surface of PDA NPs, and the release of DOX and JQ1 were measured using UV-Vis or high performance liquid chromatography (HPLC). The mechanism of fabricated nanocomplex in vitro was investigated by cell uptake experiment, cell viability assays, apoptosis assays, and Western blot analysis. Finally, the tumor-bearing mouse model was used to evaluate the tumor-inhibiting efficacy and the biosafety in vivo. Results JQ1 and DOX were successfully loaded onto PDA NPs. PDA-DOX/JQ1 NPs inhibited the growth of prostate cancer cells, reduced the expression of apoptosis related proteins and induced apoptosis in vitro. The in vivo biodistribution indicated that PDA-DOX/JQ1 NPs could accumulated at the tumor sites through the EPR effect. In tumor-bearing mice, JQ1 delivered with PDA-DOX/JQ1 NPs reduced PD-L1 expression at tumor sites, generating significant tumor suppression. Furthermore, PDA-DOX/JQ1 NPs could reduce the side effects, and produce good synergistic treatment effect in vivo. Conclusion We have successfully prepared a multifunctional platform for synergistic prostate cancer therapy.
Collapse
Affiliation(s)
- Kebang Hu
- Department of Urology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, People’s Republic of China
| | - Dongqi Zhang
- Department of Urology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, People’s Republic of China
| | - Weiran Ma
- College of Pharmacy, Jilin University, Changchun, 130021, People’s Republic of China
| | - Yanzhi Gu
- College of Pharmacy, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiang Zhao
- Department of Urology, Xi’an First Hospital, Xi’an, 710002, People’s Republic of China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
12
|
Chheda D, Shete S, Tanisha T, Devrao Bahadure S, Sampathi S, Junnuthula V, Dyawanapelly S. Multifaceted therapeutic applications of biomimetic nanovaccines. Drug Discov Today 2024; 29:103991. [PMID: 38663578 DOI: 10.1016/j.drudis.2024.103991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
The development of vaccines has had a crucial role in preventing and controlling infectious diseases on a global scale. Innovative formulations of biomimetic vaccines inspired by natural defense mechanisms combine long-term antigen stability, immunogenicity, and targeted delivery with sustained release. Types of biomimetic nanoparticle (NP) include bacterial outer membrane vesicles (OMVs), cell membrane-decorated NPs, liposomes, and exosomes. These approaches have shown potential for cancer immunotherapy, and in antibacterial and antiviral applications. Despite current challenges, nanovaccines have immense potential to transform disease prevention and treatment, promising therapeutic approaches for the future. In this review, we highlight recent advances in biomimetic vaccine design, mechanisms of action, and clinical applications, emphasizing their role in personalized medicine, targeted drug delivery, and immunomodulation.
Collapse
Affiliation(s)
- Dev Chheda
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Sukhen Shete
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Tanisha Tanisha
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Sumedh Devrao Bahadure
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Sunitha Sampathi
- Department of Pharmacy, School of Pharmacy, Vishwakarma University, Pune, Maharashtra, India.
| | | | - Sathish Dyawanapelly
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India.
| |
Collapse
|
13
|
Briffault E, Garcia-Garcia P, Martinez-Borrajo R, Evora C, Delgado A, Diaz-Rodriguez P. Harnessing extracellular vesicle membrane for gene therapy: EVs-biomimetic nanoparticles. Colloids Surf B Biointerfaces 2024; 239:113951. [PMID: 38759295 DOI: 10.1016/j.colsurfb.2024.113951] [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: 01/07/2024] [Revised: 04/28/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024]
Abstract
One of the main concerns in oligonucleotide-based therapeutics is achieving a successful cell targeting while avoiding drug degradation and clearance. Nanoparticulated drug delivery systems have emerged as a way of overcoming these issues. Among them, membrane-coated nanoparticles are of increasing relevance mainly due to their enhanced cellular uptake, immune evasion and biocompatibility. In this study, we designed and elaborated a simple and highly tuneable biomimetic drug delivery nanosystem based on a polymeric core surrounded by extracellular vesicles (EVs)-derived membranes. This strategy should allow the nanosystems to benefit from the properties conferred by the membrane proteins present in EVs membrane, key paracrine mediators. The developed systems were able to successfully encapsulate the required oligonucleotides. Also, their characterisation through already well standardised methods (dynamic light scattering, transmission electron microscopy and nanoparticle tracking analysis) and by fluorescence cross-correlation spectroscopy (FCCS) showed the desired core-shell structure. The cellular uptake using different cell types further confirmed the coating though an enhancement in cell internalisation of the developed biomimetic nanoparticles. This study brings up new possibilities for GapmeR delivery as it might be a base for the development of new delivery systems for gene therapy.
Collapse
Affiliation(s)
- Erik Briffault
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna, La Laguna 38206, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna, La Laguna 38320, Spain
| | - Patricia Garcia-Garcia
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna, La Laguna 38206, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna, La Laguna 38320, Spain
| | - Rebeca Martinez-Borrajo
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Carmen Evora
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna, La Laguna 38206, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna, La Laguna 38320, Spain
| | - Araceli Delgado
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna, La Laguna 38206, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna, La Laguna 38320, Spain.
| | - Patricia Diaz-Rodriguez
- Institute of Biomedical Technologies (ITB), Universidad de La Laguna, La Laguna 38320, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| |
Collapse
|
14
|
Zhang S, Zhang X, Gao H, Zhang X, Sun L, Huang Y, Zhang J, Ding B. Cell Membrane-Coated Biomimetic Nanoparticles in Cancer Treatment. Pharmaceutics 2024; 16:531. [PMID: 38675192 PMCID: PMC11055162 DOI: 10.3390/pharmaceutics16040531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Nanoparticle-based drug delivery systems hold promise for cancer treatment by enhancing the solubility and stability of anti-tumor drugs. Nonetheless, the challenges of inadequate targeting and limited biocompatibility persist. In recent years, cell membrane nano-biomimetic drug delivery systems have emerged as a focal point of research and development, due to their exceptional traits, including precise targeting, low toxicity, and good biocompatibility. This review outlines the categorization and advantages of cell membrane bionic nano-delivery systems, provides an introduction to preparation methods, and assesses their applications in cancer treatment, including chemotherapy, gene therapy, immunotherapy, photodynamic therapy, photothermal therapy, and combination therapy. Notably, the review delves into the challenges in the application of various cell membrane bionic nano-delivery systems and identifies opportunities for future advancement. Embracing cell membrane-coated biomimetic nanoparticles presents a novel and unparalleled avenue for personalized tumor therapy.
Collapse
Affiliation(s)
- Shu Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 214122, China;
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Xiaojuan Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Huan Gao
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Xiaoqin Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Lidan Sun
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Yueyan Huang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Jie Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Baoyue Ding
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| |
Collapse
|
15
|
Menichetti A, Mordini D, Montalti M. Polydopamine Nanosystems in Drug Delivery: Effect of Size, Morphology, and Surface Charge. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:303. [PMID: 38334574 PMCID: PMC10856634 DOI: 10.3390/nano14030303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
Recently, drug delivery strategies based on nanomaterials have attracted a lot of interest in different kinds of therapies because of their superior properties. Polydopamine (PDA), one of the most interesting materials in nanomedicine because of its versatility and biocompatibility, has been widely investigated in the drug delivery field. It can be easily functionalized to favor processes like cellular uptake and blood circulation, and it can also induce drug release through two kinds of stimuli: NIR light irradiation and pH. In this review, we describe PDA nanomaterials' performance on drug delivery, based on their size, morphology, and surface charge. Indeed, these characteristics strongly influence the main mechanisms involved in a drug delivery system: blood circulation, cellular uptake, drug loading, and drug release. The understanding of the connections between PDA nanosystems' properties and these phenomena is pivotal to obtain a controlled design of new nanocarriers based on the specific drug delivery applications.
Collapse
Affiliation(s)
| | | | - Marco Montalti
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (A.M.); (D.M.)
| |
Collapse
|
16
|
Han X, Gong C, Yang Q, Zheng K, Wang Z, Zhang W. Biomimetic Nano-Drug Delivery System: An Emerging Platform for Promoting Tumor Treatment. Int J Nanomedicine 2024; 19:571-608. [PMID: 38260239 PMCID: PMC10802790 DOI: 10.2147/ijn.s442877] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
With the development of nanotechnology, nanoparticles (NPs) have shown broad prospects as drug delivery vehicles. However, they exhibit certain limitations, including low biocompatibility, poor physiological stability, rapid clearance from the body, and nonspecific targeting, which have hampered their clinical application. Therefore, the development of novel drug delivery systems with improved biocompatibility and high target specificity remains a major challenge. In recent years, biofilm mediated biomimetic nano-drug delivery system (BNDDS) has become a research hotspot focus in the field of life sciences. This new biomimetic platform uses bio-nanotechnology to encapsulate synthetic NPswithin biomimetic membrane, organically integrating the low immunogenicity, low toxicity, high tumor targeting, good biocompatibility of the biofilm with the adjustability and versatility of the nanocarrier, and shows promising applications in the field of precision tumor therapy. In this review, we systematically summarize the new progress in BNDDS used for optimizing drug delivery, providing a theoretical reference for optimizing drug delivery and designing safe and efficient treatment strategies to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Xiujuan Han
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Qingru Yang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Kaile Zheng
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
| | - Zhuo Wang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
17
|
Li S, Meng X, Peng B, Huang J, Liu J, Xiao H, Ma L, Liu Y, Tang J. Cell membrane-based biomimetic technology for cancer phototherapy: Mechanisms, recent advances and perspectives. Acta Biomater 2024; 174:26-48. [PMID: 38008198 DOI: 10.1016/j.actbio.2023.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/04/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Despite significant advances in medical technology and antitumour treatments, the diagnosis and treatment of tumours have undergone remarkable transformations. Noninvasive phototherapy methods, such as photodynamic therapy (PDT) and photothermal therapy (PTT), have gained significant interest in antitumour medicine. However, traditional photosensitisers or photothermal agents face challenges like immune system recognition, rapid clearance from the bloodstream, limited tumour accumulation, and phototoxicity concerns. Researchers combine photosensitisers or photothermal agents with natural cell membranes to overcome these obstacles to create a nano biomimetic therapeutic platform. When used to coat nanoparticles, red blood cells, platelets, cancer cells, macrophages, lymphocytes, and bacterial outer membranes could provide prolonged circulation, tumour targeting, immune stimulation, or antigenicity. This article covers the principles of cellular membrane biomimetic nanotechnology and phototherapy, along with recent advancements in applying nano biomimetic technology to PDT, PTT, PCT, and combined diagnosis and treatment. Furthermore, the challenges and issues of using nano biomimetic nanoparticles in phototherapy are discussed. STATEMENT OF SIGNIFICANCE: Currently, there has been significant progress in the field of cell membrane biomimetic technology. Researchers are exploring its potential application in tumor diagnosis and treatment through phototherapy. Scholars have conducted extensive research on combining cell membrane technology and phototherapy in anticancer diagnosis and treatment. This review aims to highlight the mechanisms of phototherapy and the latest advancements in single phototherapy (PTT, PDT) and combination phototherapy (PCT, PRT, and PIT), as well as diagnostic approaches. The review provides an overview of various cell membrane technologies, including RBC membranes, platelet membranes, macrophage cell membranes, tumour cell membranes, bacterial membranes, hybrid membranes, and their potential for anticancer applications under phototherapy. Lastly, the review discusses the challenges and future directions in this field.
Collapse
Affiliation(s)
- Songtao Li
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xiangrui Meng
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Bo Peng
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Ju Huang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jingwen Liu
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Hang Xiao
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, PR China
| | - Li Ma
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, PR China
| | - Yiyao Liu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, PR China.
| | - Jianyuan Tang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
18
|
Lin Y, Guan X, Su J, Chen S, Fu X, Xu X, Deng X, Chang J, Qin A, Shen A, Zhang L. Cell Membrane-Camouflaged Nanoparticles Mediated Nucleic Acids Delivery. Int J Nanomedicine 2023; 18:8001-8021. [PMID: 38164266 PMCID: PMC10758188 DOI: 10.2147/ijn.s433737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024] Open
Abstract
Nucleic acids have emerged as promising therapeutic agents for many diseases because of their potential in modulating gene expression. However, the delivery of nucleic acids remains a significant challenge in gene therapy. Although viral vectors have shown high transfection efficiency, concerns regarding teratogenicity or carcinogenicity have been raised. Non-viral vehicles, including cationic polymers, liposomes, and inorganic materials possess advantages in terms of safety, ease of preparation, and low cost. Nevertheless, they also face limitations related to immunogenicity, quick clearance in vivo, and lack of targeting specificity. On the other hand, bioinspired strategies have shown increasing potential in the field of drug delivery, yet there is a lack of comprehensive reviews summarizing the rapid development of bioinspired nanoparticles based on the cell membrane camouflage to construct the nucleic acids vehicles. Herein, we enumerated the current difficulties in nucleic acid delivery with various non-viral vehicles and provided an overview of bioinspired strategies for nucleic acid delivery.
Collapse
Affiliation(s)
- Yinshan Lin
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xiaoling Guan
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Jianfen Su
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Sheng Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xihua Fu
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Xiaowei Xu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xiaohua Deng
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Jishuo Chang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Aiping Qin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Ao Shen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Lingmin Zhang
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| |
Collapse
|
19
|
Yu Z, Wang H, Ying B, Mei X, Zeng D, Liu S, Qu W, Pan X, Pu S, Li R, Qin Y. Mild photothermal therapy assist in promoting bone repair: Related mechanism and materials. Mater Today Bio 2023; 23:100834. [PMID: 38024841 PMCID: PMC10643361 DOI: 10.1016/j.mtbio.2023.100834] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/21/2023] [Accepted: 10/14/2023] [Indexed: 12/01/2023] Open
Abstract
Achieving precision treatment in bone tissue engineering (BTE) remains a challenge. Photothermal therapy (PTT), as a form of precision therapy, has been extensively investigated for its safety and efficacy. It has demonstrated significant potential in the treatment of orthopedic diseases such as bone tumors, postoperative infections and osteoarthritis. However, the high temperatures associated with PTT can lead to certain limitations and drawbacks. In recent years, researchers have explored the use of biomaterials for mild photothermal therapy (MPT), which offers a promising approach for addressing these limitations. This review provides a comprehensive overview of the mechanisms underlying MPT and presents a compilation of photothermal agents and their utilization strategies for bone tissue repair. Additionally, the paper discusses the future prospects of MPT-assisted bone tissue regeneration, aiming to provide insights and recommendations for optimizing material design in this field.
Collapse
Affiliation(s)
- Zehao Yu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Hao Wang
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Boda Ying
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Xiaohan Mei
- National & Local Joint Engineering Laboratory for Synthesis Technology of High-Performance Polymer, College of Chemistry, Jilin University, Changchun, 130012, People’s Republic of China
| | - Dapeng Zeng
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Shibo Liu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Wenrui Qu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Xiangjun Pan
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Si Pu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Ruiyan Li
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Yanguo Qin
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| |
Collapse
|
20
|
Wu K, Zhou Z, Liu T, Liu C, Mu X, Jiang J. Co-delivery of curcumin and si-STAT3 with a bioinspired tumor homing for polydopamine nanoparticles for synergistic osteosarcoma therapy. Cancer Nanotechnol 2023; 14:66. [DOI: 10.1186/s12645-023-00215-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/02/2023] [Indexed: 01/06/2025] Open
Abstract
Abstract
Purpose
Owing to the complexity of cancer, a synergistic combination of chemotherapy and gene therapy can be a promising therapeutic strategy. This study aimed to use stem cell membrane (SCM)-camouflaged polydopamine nanoparticles for simultaneous delivery of curcumin (CUR) and siRNA-targeting STAT3 (CPDA/siSTAT3@SCM NPs) for osteosarcoma (OS).
Methods
Transmission electron microscopy, UV–Vis absorbance spectra, zeta potential, cell co-localization, and Coomassie bright blue staining were used to characterize CPDA/siSTAT3@SCM NPs constructed by the self-assembly method. Drug release, cellular uptake, cell proliferation, apoptosis, wound healing, and transwell assays were evaluated in vitro. The expression levels of epithelial–mesenchymal transition (EMT)- and apoptosis-related proteins were measured by western blotting. Furthermore, the biodistribution, antitumor efficacy, and biosafety of CPDA/siSTAT3@SCM NPs in an MG63 xenograft mouse model were evaluated.
Results
CPDA/siSTAT3@SCM NPs were successfully synthesized to deliver CUR and siRNA simultaneously, and they showed osteosarcoma-targeting ability. Furthermore, it showed high cellular uptake and excellent synergistic antitumor effects in vitro. CPDA/siSTAT3@SCM NPs suppressed OS cell proliferation, migration, invasion, and EMT progression, and promoted the apoptotic process. In tumor-bearing mice, the treatment with CPDA/siSTAT3@SCM NPs showed an excellent antitumor effect with no side effects in major organs.
Conclusion
This study revealed that CPDA/siSTAT3@SCM NPs can target drug delivery by biomimetic multifunctional nanoparticles to treat OS through chemo-gene combined therapy.
Collapse
|
21
|
Chao B, Jiao J, Yang L, Wang Y, Jiang W, Yu T, Wang L, Liu H, Zhang H, Wang Z, Wu M. Application of advanced biomaterials in photothermal therapy for malignant bone tumors. Biomater Res 2023; 27:116. [PMID: 37968707 PMCID: PMC10652612 DOI: 10.1186/s40824-023-00453-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/21/2023] [Indexed: 11/17/2023] Open
Abstract
Malignant bone tumors are characterized by severe disability rate, mortality rate, and heavy recurrence rate owing to the complex pathogenesis and insidious disease progression, which seriously affect the terminal quality of patients' lives. Photothermal therapy (PTT) has emerged as an attractive adjunctive treatment offering prominent hyperthermal therapeutic effects to enhance the effectiveness of surgical treatment and avoid recurrence. Simultaneously, various advanced biomaterials with photothermal capacity are currently created to address malignant bone tumors, performing distinctive biological functions, including nanomaterials, bioceramics (BC), polymers, and hydrogels et al. Furthermore, PTT-related combination therapeutic strategies can provide more significant curative benefits by reducing drug toxicity, improving tumor-killing efficiency, stimulating anti-cancer immunity, and improving immune sensitivity relative to monotherapy, even in complex tumor microenvironments (TME). This review summarizes the current advanced biomaterials applicable in PTT and relevant combination therapies on malignant bone tumors for the first time. The multiple choices of advanced biomaterials, treatment methods, and new prospects for future research in treating malignant bone tumors with PTT are generalized to provide guidance. Malignant bone tumors seriously affect the terminal quality of patients' lives. Photothermal therapy (PTT) has emerged as an attractive adjunctive treatment enhancing the effectiveness of surgical treatment and avoiding recurrence. In this review, advanced biomaterials applicable in the PTT of malignant bone tumors and their distinctive biological functions are comprehensively summarized for the first time. Simultaneously, multiple PTT-related combination therapeutic strategies are classified to optimize practical clinical issues, contributing to the selection of biomaterials, therapeutic alternatives, and research perspectives for the adjuvant treatment of malignant bone tumors with PTT in the future.
Collapse
Affiliation(s)
- Bo Chao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Lili Yang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Weibo Jiang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Tong Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Linfeng Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Han Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China.
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China.
| |
Collapse
|
22
|
Gallo J, Villasante A. Recent Advances in Biomimetic Nanocarrier-Based Photothermal Therapy for Cancer Treatment. Int J Mol Sci 2023; 24:15484. [PMID: 37895165 PMCID: PMC10607206 DOI: 10.3390/ijms242015484] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Nanomedicine presents innovative solutions for cancer treatment, including photothermal therapy (PTT). PTT centers on the design of photoactivatable nanoparticles capable of absorbing non-toxic near-infrared light, generating heat within target cells to induce cell death. The successful transition from benchside to bedside application of PTT critically depends on the core properties of nanoparticles responsible for converting light into heat and the surface properties for precise cell-specific targeting. Precisely targeting the intended cells remains a primary challenge in PTT. In recent years, a groundbreaking approach has emerged to address this challenge by functionalizing nanocarriers and enhancing cell targeting. This strategy involves the creation of biomimetic nanoparticles that combine desired biocompatibility properties with the immune evasion mechanisms of natural materials. This review comprehensively outlines various strategies for designing biomimetic photoactivatable nanocarriers for PTT, with a primary focus on its application in cancer therapy. Additionally, we shed light on the hurdles involved in translating PTT from research to clinical practice, along with an overview of current clinical applications.
Collapse
Affiliation(s)
- Juan Gallo
- Advanced Magnetic Theranostic Nanostructures Lab, International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal;
| | - Aranzazu Villasante
- Nanobioengineering Lab, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Electronic and Biomedical Engineering, Faculty of Physics, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
23
|
Zhang H, Luo P, Huang X. Engineered nanomaterials enhance drug delivery strategies for the treatment of osteosarcoma. Front Pharmacol 2023; 14:1269224. [PMID: 37670948 PMCID: PMC10475588 DOI: 10.3389/fphar.2023.1269224] [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: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in adolescents, and the clinical treatment of OS mainly includes surgery, radiotherapy, and chemotherapy. However, the side effects of chemotherapy drugs are an issue that clinicians cannot ignore. Nanomedicine and drug delivery technologies play an important role in modern medicine. The development of nanomedicine has ushered in a new turning point in tumor treatment. With the emergence and development of nanoparticles, nanoparticle energy surfaces can be designed with different targeting effects. Not only that, nanoparticles have unique advantages in drug delivery. Nanoparticle delivery drugs can not only reduce the toxic side effects of chemotherapy drugs, but due to the enhanced permeability retention (EPR) properties of tumor cells, nanoparticles can survive longer in the tumor microenvironment and continuously release carriers to tumor cells. Preclinical studies have confirmed that nanoparticles can effectively delay tumor growth and improve the survival rate of OS patients. In this manuscript, we present the role of nanoparticles with different functions in the treatment of OS and look forward to the future treatment of improved nanoparticles in OS.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Spine, Trauma Surgery, The First People’s Hospital of Guangyuan, Guangyuan, China
| | - Ping Luo
- Science and Technology Education Section, The First People’s Hospital of Guangyuan, Guangyuan, China
| | - Xiaojun Huang
- Department of Spine, Trauma Surgery, The First People’s Hospital of Guangyuan, Guangyuan, China
| |
Collapse
|
24
|
Desai N, Rana D, Pande S, Salave S, Giri J, Benival D, Kommineni N. "Bioinspired" Membrane-Coated Nanosystems in Cancer Theranostics: A Comprehensive Review. Pharmaceutics 2023; 15:1677. [PMID: 37376125 DOI: 10.3390/pharmaceutics15061677] [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: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Achieving precise cancer theranostics necessitates the rational design of smart nanosystems that ensure high biological safety and minimize non-specific interactions with normal tissues. In this regard, "bioinspired" membrane-coated nanosystems have emerged as a promising approach, providing a versatile platform for the development of next-generation smart nanosystems. This review article presents an in-depth investigation into the potential of these nanosystems for targeted cancer theranostics, encompassing key aspects such as cell membrane sources, isolation techniques, nanoparticle core selection, approaches for coating nanoparticle cores with the cell membrane, and characterization methods. Moreover, this review underscores strategies employed to enhance the multi-functionality of these nanosystems, including lipid insertion, membrane hybridization, metabolic engineering, and genetic modification. Additionally, the applications of these bioinspired nanosystems in cancer diagnosis and therapeutics are discussed, along with the recent advances in this field. Through a comprehensive exploration of membrane-coated nanosystems, this review provides valuable insights into their potential for precise cancer theranostics.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | | |
Collapse
|
25
|
Gao S, Lv R, Hao N, Wang H, Lv Y, Li Y, Ji Y, Liu Y. Fabrication of pH/photothermal-responsive ZIF-8 nanocarriers loaded with baicalein for effective drug delivery and synergistic chem-photothermal effects. Colloids Surf A Physicochem Eng Asp 2023. [DOI: 10.1016/j.colsurfa.2023.131401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
26
|
Fan L, Wei A, Gao Z, Mu X. Current progress of mesenchymal stem cell membrane-camouflaged nanoparticles for targeted therapy. Biomed Pharmacother 2023; 161:114451. [PMID: 36870279 DOI: 10.1016/j.biopha.2023.114451] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Nanodrug delivery systems have been widely used in disease treatment. However, weak drug targeting, easy to be cleared by the immune system, and low biocompatibility are great obstacles for drug delivery. As an important part of cell information transmission and behavior regulation, cell membrane can be used as drug coating material which represents a promising strategy and can overcome these limitations. Mesenchymal stem cell (MSC) membrane, as a new carrier, has the characteristics of active targeting and immune escape of MSC, and has broad application potential in tumor treatment, inflammatory disease, tissue regeneration and other fields. Here, we review recent progress on the use of MSC membrane-coated nanoparticles for therapy and drug delivery, aiming to provide guidance for the design and clinical application of membrane carrier in the future.
Collapse
Affiliation(s)
- Lianlian Fan
- Department of Pharmacy, China-Japan Union Hospital, Jilin University, Changchun130033, China
| | - Anhui Wei
- Department of Regenerative Medicine, College of Pharmacy, Jilin University, Changchun130021, China
| | - Zihui Gao
- Changchun City Experimental High School, Changchun130117, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun130033, China.
| |
Collapse
|
27
|
Bao M, Wang K, Li J, Li Y, Zhu H, Lu M, Zhang Y, Fan Q, Han L, Wang K, Wang D, Gao Y, Peng B, Ming Z, Liu W. ROS Scavenging and inflammation-directed polydopamine nanoparticles regulate gut immunity and flora therapy in inflammatory bowel disease. Acta Biomater 2023; 161:250-264. [PMID: 36863680 DOI: 10.1016/j.actbio.2023.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Dysfunction of the intestinal mucosal immune system and dysbiosis of the intestinal microflora can induce inflammatory bowel disease. However, drug-mediated clinical treatment remains a challenge due to its poor therapeutic efficacy and severe side effects. Herein, a ROS scavenging and inflammation-directed nanomedicine is designed and fabricated by coupling polydopamine nanoparticles with mCRAMP, an antimicrobial peptide, while wrapping macrophage membrane in the outer layer. The designed nanomedicine reduced the secretion of pro-inflammatory cytokines and elevate the expression of anti-inflammatory cytokine in vivo and in vitro inflammation models, demonstrating its significant ability of improving inflammatory responses. Importantly, the macrophage membrane encapsulated nanoparticles exhibit the obviously enhanced targeting performance in local inflamed tissues. Furthermore, the 16S rRNA sequencing of fecal microorganisms showed that probiotics increased and pathogenic bacteria were inhibited after oral delivery the nanomedicine, indicating that the designed nano platform played a significant role in optimizing intestinal microbiome. Taken together, the designed nanomedicine are not only easy to prepare and exhibit high biocompatibility, but also show the inflammatory targeting property, anti-inflammatory function and positive regulation of intestinal flora, thus providing a new idea for the intervention and treatment of colitis. STATEMENT OF SIGNIFICANCE: Inflammatory bowel disease (IBD), a chronic and intractable disease, may lead to colon cancer in severe cases without effective treatment. However, clinical drugs are largely ineffective owing to insufficient therapeutic efficacies and side effects. Herein, we constructed a biomimetic polydopamine nanoparticle for oral administration to treat the IBD by modulating mucosal immune homeostasis and optimizing intestinal microorganisms. In vitro and in vivo experiments showed that the designed nanomedicine not only exhibits the anti-inflammatory function and inflammatory targeting property but also positively regulate the gut microflora. Taken together, the designed nanomedicine combined immunoregulation and intestinal microecology modulation to significantly enhance the therapeutic effect on colitis in mice, thus providing a new approach for the clinical treatment of colitis.
Collapse
Affiliation(s)
- Meiyu Bao
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Keyi Wang
- Department of Urology Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Jingqiang Li
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Yueying Li
- Department of Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200070, China
| | - Huanhuan Zhu
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Meiling Lu
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Yue Zhang
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Qiangyuan Fan
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Lin Han
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Kesheng Wang
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Dongyan Wang
- Department of Gastroenterology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, China
| | - Yan Gao
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China
| | - Bo Peng
- Department of Urology Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China.
| | - Zunzhen Ming
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301# Yanchang Middle Road, Shanghai, 200072, China.
| | - Weiwei Liu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China.
| |
Collapse
|
28
|
Zhang Y, Liu X, Geng C, Shen H, Zhang Q, Miao Y, Wu J, Ouyang R, Zhou S. Two Hawks with One Arrow: A Review on Bifunctional Scaffolds for Photothermal Therapy and Bone Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13030551. [PMID: 36770512 PMCID: PMC9920372 DOI: 10.3390/nano13030551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 05/21/2023]
Abstract
Despite the significant improvement in the survival rate of cancer patients, the total cure of bone cancer is still a knotty clinical challenge. Traditional surgical resectionof bone tumors is less than satisfactory, which inevitably results in bone defects and the inevitable residual tumor cells. For the purpose of realizing minimal invasiveness and local curative effects, photothermal therapy (PTT) under the irradiation of near-infrared light has made extensive progress in ablating tumors, and various photothermal therapeutic agents (PTAs) for the treatment of bone tumors have thus been reported in the past few years, has and have tended to focus on osteogenic bio-scaffolds modified with PTAs in order to break through the limitation that PTT lacks, osteogenic capacity. These so-called bifunctional scaffolds simultaneously ablate bone tumors and generate new tissues at the bone defects. This review summarizes the recent application progress of various bifunctional scaffolds and puts forward some practical constraints and future perspectives on bifunctional scaffolds for tumor therapy and bone regeneration: two hawks with one arrow.
Collapse
Affiliation(s)
- Yulong Zhang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xueyu Liu
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Chongrui Geng
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Hongyu Shen
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qiupeng Zhang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yuqing Miao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
- Correspondence: (Y.M.); (J.W.); (R.O.)
| | - Jingxiang Wu
- Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (Y.M.); (J.W.); (R.O.)
| | - Ruizhuo Ouyang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
- Correspondence: (Y.M.); (J.W.); (R.O.)
| | - Shuang Zhou
- Cancer Institute, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
29
|
Chen Y, Zhu M, Huang B, Jiang Y, Su J. Advances in cell membrane-coated nanoparticles and their applications for bone therapy. BIOMATERIALS ADVANCES 2023; 144:213232. [PMID: 36502750 DOI: 10.1016/j.bioadv.2022.213232] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Due to the specific structure of natural bone, most of the therapeutics are incapable to be delivered into the targeted site with effective concentrations. Nanotechnology has provided a good way to improve this issue, cell membrane mimetic nanoparticles (NPs) have been emerging as an ideal nanomaterial which integrates the advantages of natural cell membranes with synthetic NPs to significantly improve the biocompatibility as well as achieving long-lasting circulation and targeted delivery. In addition, functionalized modifications of the cell membrane facilitate more precise targeting and therapy. Here, an overview of the preparation of cell membrane-coated NPs and the properties of cell membranes from different cell sources has been given to expatiate their function and potential applications. Strategies for functionalized modification of cell membranes are also briefly described. The application of cell membrane-coated NPs for bone therapy is then presented according to the function of cell membranes. Moreover, the prospects and challenges of cell membrane-coated NPs for translational medicine have also been discussed.
Collapse
Affiliation(s)
- Yutong Chen
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China; School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Mengru Zhu
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Biaotong Huang
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; Wenzhou Institute of Shanghai University, Wenzhou 325000, PR China.
| | - Yingying Jiang
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jiacan Su
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
30
|
Tian L, Li X, Ji H, Yu Q, Yang M, Guo L, Huang L, Gao W. Melanin-like nanoparticles: advances in surface modification and tumour photothermal therapy. J Nanobiotechnology 2022; 20:485. [PMCID: PMC9675272 DOI: 10.1186/s12951-022-01698-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022] Open
Abstract
Currently, tumor treatments are characterized by intelligence, diversity and personalization, but the therapeutic reagents used are often limited in clinical efficacy due to problems with water solubility, targeting, stability and multidrug resistance. To remedy these shortcomings, the application of multifunctional nanotechnology in the biomedical field has been widely studied. Synthetic melanin nanoparticles (MNPs) surfaces which contain highly reactive chemical groups such as carboxyl, hydroxyl and amine groups, can be used as a reaction platform on which to graft different functional components. In addition, MNPs easily adhere to substrate surface, and serve as a secondary reaction platform to modify it. The multifunctionality and intrinsic biocompatibility make melanin-like nanoparticles promising as a multifunctional and powerful nanoplatform for oncological applications. This paper first reviews the preparation methods, polymerization mechanisms and physicochemical properties of melanin including natural melanin and chemically synthesized melanin to guide scholars in MNP-based design. Then, recent advances in MNPs especially synthetic polydopamine (PDA) melanin for various medical oncological applications are systematically and thoroughly described, mainly focusing on bioimaging, photothermal therapy (PTT), and drug delivery for tumor therapy. Finally, based on the investigated literature, the current challenges and future directions for clinical translation are reasonably discussed, focusing on the innovative design of MNPs and further elucidation of pharmacokinetics. This paper is a timely and comprehensive and detailed study of the progress of MNPs in tumor therapy, especially PTT, and provides ideas for the design of personalized and customizable oncology nanomedicines to address the heterogeneity of the tumor microenvironment.
Collapse
Affiliation(s)
- Luyao Tian
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Xia Li
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Haixia Ji
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Qing Yu
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Mingjuan Yang
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Lanping Guo
- grid.410318.f0000 0004 0632 3409National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Luqi Huang
- grid.410318.f0000 0004 0632 3409National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Wenyuan Gao
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| |
Collapse
|
31
|
Tang X, Li D, Gu Y, Zhao Y, Li A, Qi F, Liu J. Natural cell based biomimetic cellular transformers for targeted therapy of digestive system cancer. Theranostics 2022; 12:7080-7107. [PMID: 36276645 PMCID: PMC9576611 DOI: 10.7150/thno.75937] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022] Open
Abstract
Digestive system cancer is the most common cause of cancer death in the world. Although cancer treatment options are increasingly diversified, the mortality rate of malignant cancer of the digestive system remains high. Therefore, it is necessary to explore effective cancer treatment methods. Recently, biomimetic nanoparticle delivery systems based on natural cells that organically integrate the low immunogenicity, high biocompatibility, cancer targeting, and controllable, versatile functionality of smart nanocarrier design with natural cells have been expected to break through the bottleneck of tumor targeted therapy. In this review, we focus on the dynamic changes and complex cellular communications that occur in vivo in natural cells based vehicles. Recent studies on the development of advanced targeted drug delivery systems using the dynamic behaviors such as specific surface protein affinity, morphological changes, and phenotypic polarization of natural cells are summarized. In addition to drug delivery mediated by dynamic behavior, functional "delivery" based on the natural cell themselves is also involved. Aiming to make the best use of the functions of cells, providing clues for the development of advanced drug delivery platforms.
Collapse
Affiliation(s)
- Xiaomeng Tang
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yunan Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Aixue Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Fu Qi
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pharmacy, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| |
Collapse
|
32
|
Stem cell membrane-coated abiotic nanomaterials for biomedical applications. J Control Release 2022; 351:174-197. [PMID: 36103910 DOI: 10.1016/j.jconrel.2022.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 12/15/2022]
Abstract
Nanoscale materials have been extensively employed for diagnostic and therapeutic purposes. However, the developed nanosystems still suffer from some limitations, namely the rapid elimination by the immune system, lack of targeting to specific cells, and insufficient biocompatibility. Therefore, novel strategies based upon a biomimetic approach have received attention to improving the pharmacokinetics and safety profile of nanosystems. One promising strategy is the application of a biomimetic coating consisting of cell membranes derived from different cell types onto nanoparticle cores. Stem cells have been investigated to develop targeted nanodevices owing to their excellent intrinsic tissue-specific homing features, protecting them from the immune system to reach the sites of inflammation. This targeting ability is conferred by a surface repertoire of stem cell-associated biomolecules. Such nanoscopical materials offer sustained circulation and boosted drug accumulation at target sites, augmenting therapeutic efficacy and safety. Additionally, the coating of nanoparticles with cell membranes acts as a camouflage mechanism to increase their circulation time. The current review explores the particular features of stem cell membrane coating as multifunctional biomimetic surface functionalization agents to camouflage nanoparticle cores. Biomedical applications of engineered stem cell membrane-coated nanoparticles, challenges in clinical translation, and their future prospects are addressed.
Collapse
|
33
|
Aboeleneen SB, Scully MA, Harris JC, Sterin EH, Day ES. Membrane-wrapped nanoparticles for photothermal cancer therapy. NANO CONVERGENCE 2022; 9:37. [PMID: 35960404 PMCID: PMC9373884 DOI: 10.1186/s40580-022-00328-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/27/2022] [Indexed: 05/31/2023]
Abstract
Cancer is a global health problem that needs effective treatment strategies. Conventional treatments for solid-tumor cancers are unsatisfactory because they cause unintended harm to healthy tissues and are susceptible to cancer cell resistance. Nanoparticle-mediated photothermal therapy is a minimally invasive treatment for solid-tumor cancers that has immense promise as a standalone therapy or adjuvant to other treatments like chemotherapy, immunotherapy, or radiotherapy. To maximize the success of photothermal therapy, light-responsive nanoparticles can be camouflaged with cell membranes to endow them with unique biointerfacing capabilities that reduce opsonization, prolong systemic circulation, and improve tumor delivery through enhanced passive accumulation or homotypic targeting. This ensures a sufficient dose of photoresponsive nanoparticles arrives at tumor sites to enable their complete thermal ablation. This review summarizes the state-of-the-art in cell membrane camouflaged nanoparticles for photothermal cancer therapy and provides insights to the path forward for clinical translation.
Collapse
Affiliation(s)
| | | | - Jenna C Harris
- Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - Eric H Sterin
- Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Emily S Day
- Biomedical Engineering, University of Delaware, Newark, DE, USA.
- Materials Science and Engineering, University of Delaware, Newark, DE, USA.
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA.
| |
Collapse
|
34
|
Abstract
There is an unmet need for carriers that can deliver nucleic acids (NAs) to cancer cells and tumors to perpetuate gene regulation and manage disease progression. Membrane-wrapped nanoparticles (NPs) can be loaded with exogenously designed nucleic acid cargoes, such as plasmid deoxyribonucleic acid (pDNA), messenger ribonucleic acid (mRNA), small interfering RNA (siRNA), microRNA (miRNA), and immunostimulatory CpG oligodeoxynucleotides (CpG ODNs), to mitigate challenges presented by NAs' undesirable negative charge, hydrophilicity, and relatively large size. By conjugating or encapsulating NAs within membrane-wrapped NPs, various physiological barriers can be overcome so that NAs experience increased blood circulation half-lives and enhanced accumulation in intended sites. This review discusses the status of membrane-wrapped NPs as NA delivery vehicles and their advancement in gene regulation for cancer management in vitro and in vivo. With continued development, membrane-wrapped NPs have great potential as future clinical tools to treat cancer and other diseases with a known genetic basis.
Collapse
Affiliation(s)
| | - Eric H Sterin
- Biomedical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Emily S Day
- Biomedical Engineering, University of Delaware, Newark, DE 19716, USA.
- Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE 19713, USA
| |
Collapse
|
35
|
|
36
|
Zhang W, Huang X. Stem cell membrane-camouflaged targeted delivery system in tumor. Mater Today Bio 2022; 16:100377. [PMID: 35967738 PMCID: PMC9364095 DOI: 10.1016/j.mtbio.2022.100377] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023] Open
|
37
|
Khosravi N, Pishavar E, Baradaran B, Oroojalian F, Mokhtarzadeh A. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: A promising biomimetic nanoplatforms for cancer theranostics. J Control Release 2022; 348:706-722. [PMID: 35732250 DOI: 10.1016/j.jconrel.2022.06.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Nanomedicine research has advanced dramatically in recent decades. Nonetheless, traditional nanomedicine faces significant obstacles such as the low concentration of the drug at target sites and accelerated removal of the drug from blood circulation. Various techniques of nanotechnology, including cell membrane coating, have been developed to address these challenges and to improve targeted distribution and redcue cell membrane-mediated immunogenicity. Recently, stem cell (SC) membranes, owing to their immunosuppressive and regenerative properties, have grabbed attention as attractive therapeutic carriers for targeting specific tissues or organs. Bioengineering strategies that combine synthetic nanoparticles (NPs) with SC membranes, because of their homing potential and tumor tropism, have recently received a lot of publicity. Several laboratory experiments and clinical trials have indicated that the benefits of SC-based technologies are mostly related to the effects of SC-derived exosomes (SC-Exos). Exosomes are known as nano-sized extracellular vehicles (EVs) that deliver particular bioactive molecules for cell-to-cell communication. In this regard, SC-derived exosome membranes have recently been employed to improve the therapeutic capability of engineered drug delivery vehicles. Most recently, for further enhancing NPs' functionality, a new coating approach has been offered that combines membranes from two separate cells. These hybrid membrane delivery vehicles have paved the way for the development of biocompatible, high-efficiency, biomimetic NPs with varying hybrid capabilities that can overcome the drawbacks of present NP-based treatment techniques. This review explores stem cell membranes, SC-Exos, and hybrid SC-camouflaged NPs preparation methods and their importance in cancer therapy.
Collapse
Affiliation(s)
- Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Pishavar
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
38
|
Mesenchymal stem cells: A living carrier for active tumor-targeted delivery. Adv Drug Deliv Rev 2022; 185:114300. [PMID: 35447165 DOI: 10.1016/j.addr.2022.114300] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022]
Abstract
The strategy of using mesenchymal stem cells (MSCs) as a living carrier for active delivery of therapeutic agents targeting tumor sites has been attempted in a wide range of studies to validate the feasibility and efficacy for tumor treatment. This approach reveals powerful tumor targeting and tumor penetration. In addition, MSCs have been confirmed to actively participate in immunomodulation of the tumor microenvironment. Thus, MSCs are not inert delivery vehicles but have a strong impact on the fate of tumor cells. In this review, these active properties of MSCs are addressed to highlight the advantages and challenges of using MSCs for tumor-targeted delivery. In addition, some of the latest examples of using MSCs to carry a variety of anti-tumor agents for tumor-targeted therapy are summarized. Recent technologies to improve the performance and safety of this delivery strategy will be introduced. The advances, applications, and challenges summarized in this review will provide a general understanding of this promising strategy for actively delivering drugs to tumor tissues.
Collapse
|
39
|
Heptamethine Cyanine-Loaded Nanomaterials for Cancer Immuno-Photothermal/Photodynamic Therapy: A Review. Pharmaceutics 2022; 14:pharmaceutics14051015. [PMID: 35631600 PMCID: PMC9144181 DOI: 10.3390/pharmaceutics14051015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
The development of strategies capable of eliminating metastasized cancer cells and preventing tumor recurrence is an exciting and extremely important area of research. In this regard, therapeutic approaches that explore the synergies between nanomaterial-mediated phototherapies and immunostimulants/immune checkpoint inhibitors have been yielding remarkable results in pre-clinical cancer models. These nanomaterials can accumulate in tumors and trigger, after irradiation of the primary tumor with near infrared light, a localized temperature increase and/or reactive oxygen species. These effects caused damage in cancer cells at the primary site and can also (i) relieve tumor hypoxia, (ii) release tumor-associated antigens and danger-associated molecular patterns, and (iii) induced a pro-inflammatory response. Such events will then synergize with the activity of immunostimulants and immune checkpoint inhibitors, paving the way for strong T cell responses against metastasized cancer cells and the creation of immune memory. Among the different nanomaterials aimed for cancer immuno-phototherapy, those incorporating near infrared-absorbing heptamethine cyanines (Indocyanine Green, IR775, IR780, IR797, IR820) have been showing promising results due to their multifunctionality, safety, and straightforward formulation. In this review, combined approaches based on phototherapies mediated by heptamethine cyanine-loaded nanomaterials and immunostimulants/immune checkpoint inhibitor actions are analyzed, focusing on their ability to modulate the action of the different immune system cells, eliminate metastasized cancer cells, and prevent tumor recurrence.
Collapse
|
40
|
Liang S, Wang M, Wang J, Chen G. Red-Blood-Cell-Membrane-Coated Metal-Drug Nanoparticles for Enhanced Chemotherapy. Chembiochem 2021; 22:3184-3189. [PMID: 34468067 DOI: 10.1002/cbic.202100313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/12/2021] [Indexed: 12/11/2022]
Abstract
To overcome high toxicity, low bioavailability and poor water solubility of chemotherapeutics, a variety of drug carriers have been designed. However, most carriers are severely limited by low drug loading capacity and adverse side effects. Here, a new type of metal-drug nanoparticles (MDNs) was designed and synthesized. The MDNs self-assembled with Fe(III) ions and drug molecules through coordination, resulting in nanoparticles with high drug loading. To assist systemic delivery and prolong circulation time, the obtained MDNs were camouflaged with red blood cell (RBCs) membranes (RBCs@Fe-DOX MDNs) to improve their stability and dispersity. The RBCs@Fe-DOX MDNs presented pH-responsive release functionalities, resulting in drug release accelerated in acidic tumor microenvironments. The outstanding in vitro and in vivo antitumor therapeutic outcome was realized by RBCs@Fe-DOX MDNs. This study provides an innovative design guideline for chemotherapy and demonstrates the great capacity of nanomaterials in anticancer treatments.
Collapse
Affiliation(s)
- Shuya Liang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| | - Miaomiao Wang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| | - Jun Wang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| | - Guanzhi Chen
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| |
Collapse
|
41
|
Chen H, Deng J, Yao X, He Y, Li H, Jian Z, Tang Y, Zhang X, Zhang J, Dai H. Bone-targeted erythrocyte-cancer hybrid membrane-camouflaged nanoparticles for enhancing photothermal and hypoxia-activated chemotherapy of bone invasion by OSCC. J Nanobiotechnology 2021; 19:342. [PMID: 34702291 PMCID: PMC8549398 DOI: 10.1186/s12951-021-01088-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background Jaw bones are the most common organs to be invaded by oral malignancies, such as oral squamous cell carcinoma (OSCC), because of their special anatomical relationship. Various serious complications, such as pathological fractures and bone pain can significantly decrease the quality of life or even survival outcomes for a patient. Although chemotherapy is a promising strategy for bone invasion treatment, its clinical applications are limited by the lack of tumor-specific targeting and poor permeability in bone tissue. Therefore, it is necessary to develop a smart bone and cancer dual targeting drug delivery platform. Results We designed a dual targeting nano-biomimetic drug delivery vehicle Asp8[H40-TPZ/IR780@(RBC-H)] that has excellent bone and cancer targeting as well as immune escape abilities to treat malignancies in jaw bones. These nanoparticles were camouflaged with a head and neck squamous cell carcinoma WSU-HN6 cell (H) and red blood cell (RBC) hybrid membrane, which were modified by an oligopeptide of eight aspartate acid (Asp8). The spherical morphology and typical core-shell structure of biomimetic nanoparticles were observed by transmission electron microscopy. These nanoparticles exhibited the same surface proteins as those of WSU-HN6 and RBC. Flow cytometry and confocal microscopy showed a greater uptake of the biomimetic nanoparticles when compared to bare H40-PEG nanoparticles. Biodistribution of the nanoparticles in vivo revealed that they were mainly localized in the area of bone invasion by WSU-HN6 cells. Moreover, the Asp8[H40-TPZ/IR780@(RBC-H)] nanoparticles exhibited effective cancer growth inhibition properties when compared to other TPZ or IR780 formulations. Conclusions Asp8[H40-TPZ/IR780@(RBC-H)] has bone targeting, tumor-homing and immune escape abilities, therefore, it is an efficient multi-targeting drug delivery platform for achieving precise anti-cancer therapy during bone invasion. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01088-9.
Collapse
Affiliation(s)
- Hongying Chen
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jiang Deng
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xintong Yao
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Yungang He
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Hanyue Li
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Zhixiang Jian
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Yi Tang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xiaoqing Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| | - Hongwei Dai
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China. .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China. .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| |
Collapse
|
42
|
Sun J, Xing F, Braun J, Traub F, Rommens PM, Xiang Z, Ritz U. Progress of Phototherapy Applications in the Treatment of Bone Cancer. Int J Mol Sci 2021; 22:ijms222111354. [PMID: 34768789 PMCID: PMC8584114 DOI: 10.3390/ijms222111354] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023] Open
Abstract
Bone cancer including primary bone cancer and metastatic bone cancer, remains a challenge claiming millions of lives and affecting the life quality of survivors. Conventional treatments of bone cancer include wide surgical resection, radiotherapy, and chemotherapy. However, some bone cancer cells may remain or recur in the local area after resection, some are highly resistant to chemotherapy, and some are insensitive to radiotherapy. Phototherapy (PT) including photodynamic therapy (PDT) and photothermal therapy (PTT), is a clinically approved, minimally invasive, and highly selective treatment, and has been widely reported for cancer therapy. Under the irradiation of light of a specific wavelength, the photosensitizer (PS) in PDT can cause the increase of intracellular ROS and the photothermal agent (PTA) in PTT can induce photothermal conversion, leading to the tumoricidal effects. In this review, the progress of PT applications in the treatment of bone cancer has been outlined and summarized, and some envisioned challenges and future perspectives have been mentioned. This review provides the current state of the art regarding PDT and PTT in bone cancer and inspiration for future studies on PT.
Collapse
Affiliation(s)
- Jiachen Sun
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (J.S.); (J.B.); (F.T.); (P.M.R.)
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
| | - Fei Xing
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
| | - Joy Braun
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (J.S.); (J.B.); (F.T.); (P.M.R.)
| | - Frank Traub
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (J.S.); (J.B.); (F.T.); (P.M.R.)
| | - Pol Maria Rommens
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (J.S.); (J.B.); (F.T.); (P.M.R.)
| | - Zhou Xiang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
- Correspondence: (Z.X.); (U.R.)
| | - Ulrike Ritz
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (J.S.); (J.B.); (F.T.); (P.M.R.)
- Correspondence: (Z.X.); (U.R.)
| |
Collapse
|
43
|
Zhang Y, Guo L, Kong F, Duan L, Li H, Fang C, Zhang K. Nanobiotechnology-enabled energy utilization elevation for augmenting minimally-invasive and noninvasive oncology thermal ablation. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1733. [PMID: 34137183 DOI: 10.1002/wnan.1733] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/15/2021] [Accepted: 05/26/2021] [Indexed: 12/19/2022]
Abstract
Depending on the local or targeted treatment, independence on tumor type and minimally-invasive and noninvasive feature, various thermal ablation technologies have been established, but they still suffer from the intractable paradox between safety and efficacy. It has been extensively accepted that improving energy utilization efficiency is the primary means of decreasing thermal ablation power and shortening ablation time, which is beneficial for concurrently improving both treatment safety and treatment efficiency. Recent efforts have been made to receive a significant advance in various thermal methods including non-invasive high-intensity focused ultrasound, minimally-invasive radiofrequency and microwave, and non-invasive and minimally-invasive photothermal ablation, and so on. Especially, various nanobiotechnologies and design methodologies were employed to elevate the energy utilization efficiency for acquiring unexpected ablation outcomes accompanied with tremendously reduced power and time. More significantly, some combined technologies, for example, chemotherapy, photodynamic therapy (PDT), gaseous therapy, sonodynamic therapy (SDT), immunotherapy, chemodynamic therapy (CDT), or catalytic nanomedicine, were used to assist these ablation means to repress or completely remove tumors. We discussed and summarized the ablation principles and energy transformation pathways of the four ablation means, and reviewed and commented the progress in this field including newly developed technology or new material types with a highlight on nanobiotechnology-inspired design principles, and provided the deep insights into the existing problems and development direction. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medical Ultrasound, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Lehang Guo
- Department of Medical Ultrasound, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Fanlei Kong
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Hongyan Li
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Chao Fang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Kun Zhang
- Department of Medical Ultrasound, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China.,Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
44
|
Mu X, Zhang M, Wei A, Yin F, Wang Y, Hu K, Jiang J. Doxorubicin and PD-L1 siRNA co-delivery with stem cell membrane-coated polydopamine nanoparticles for the targeted chemoimmunotherapy of PCa bone metastases. NANOSCALE 2021; 13:8998-9008. [PMID: 33973580 DOI: 10.1039/d0nr08024a] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Programmed cell death ligand 1 (PD-L1) blockade has achieved great success in cancer immunotherapy. PD-L1 siRNA can restore the immune anti-tumor activity of T cells by downregulating the level of PD-L1 on tumor cells, but the efficiency of PD-1/PD-L1 monotherapy is relatively low. Doxorubicin (DOX) can induce tumor cell apoptosis, and then increase the release of tumor antigen. But the expression of PD-L1 in tumor tissues treated with DOX will be enhanced adaptively. Therefore, DOX combination with PD-L1 siRNA can produce a good synergistic anti-tumor effect. In this study, stem cell membrane (SCM) camouflaged polydopamine nanoparticles carrying DOX and PD-L1 siRNA (PDA-DOX/siPD-L1@SCM) were constructed for targeting prostate cancer (PCa) bone metastases. PDA-DOX/siPD-L1@SCM NPs could effectively enhance blood retention and improve accumulation at tumor sites. In vitro and in vivo studies demonstrated that PDA-DOX/siPD-L1@SCM NPs showed excellent performance in synergistic chemoimmunotherapy for PCa bone metastases. Hence, this study provided an effective strategy for developing biomimetic multifunctional nanoparticles for PCa bone metastasis treatment.
Collapse
Affiliation(s)
- Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, China.
| | | | | | | | | | | | | |
Collapse
|
45
|
Su Y, Zhang T, Huang T, Gao J. Current advances and challenges of mesenchymal stem cells-based drug delivery system and their improvements. Int J Pharm 2021; 600:120477. [PMID: 33737099 DOI: 10.1016/j.ijpharm.2021.120477] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as a promising living carrier for targeted drug delivery. A wealth of literature has shown evidence for great advances in MSCs-based drug delivery system (MSCs-DDS) in the treatment of various diseases. Nevertheless, as this field of study rapidly advances, several challenges associated with this delivery strategy have arisen, mainly due to the inherent limitations of MSCs. To this end, several novel technologies are being developed in parallel to improve the efficiency or safety of this system. In this review, we introduce recent advances and summarize the present challenges of MSCs-DDS. We also highlight some potential technologies to improve MSCs-DDS, including nanotechnology, genome engineering technology, and biomimetic technology. Finally, prospects for application of artificially improved MSCs-DDS are addressed. The technologies summarized in this review provide a general guideline for the improvement of MSCs-DDS.
Collapse
Affiliation(s)
- Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|