1
|
Peng L, Chang L, Bai R, Sun Q, Zhang Y, Liu H, Ma C, Lin J, Han B. Zwitterion polymer-modified graphene oxides enhance antibacterial activity with improved biocompatibility and osteogenesis: An in vitro study. REACT FUNCT POLYM 2025; 212:106229. [DOI: 10.1016/j.reactfunctpolym.2025.106229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
2
|
Liu M, Wang J, Chen G, Wang L, Wang X, Xiang B, Deng Y, He C, Wang L. Neutrophils-mediated accelerated blood clearance phenomenon in beagles and rats based on the cross-injection of non-PEGylated and PEGylated nanoemulsions. Int J Pharm X 2025; 9:100318. [PMID: 40070371 PMCID: PMC11894323 DOI: 10.1016/j.ijpx.2025.100318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
The initial injection of PEGylated nanoparticles can activate antibodies and the complement system, leading to the accelerated blood clearance (ABC) phenomenon, characterized by reduced circulation time and abnormal liver and spleen accumulation upon re-exposure. However, PEGylation is not essential for ABC induction, as non-PEGylated nanoparticles can also trigger the similar ABC phenomenon. In this study, we found non-PEGylated nanoemulsions (CE) could accelerate the blood clearance of subsequent injection of PEGylated nanoemulsions (PE) in beagles and rats, which was independent of antibodies and the complement system, but was associated with an increase in neutrophil numbers and phagocytic activity. We propose classifying this as a "general ABC phenomenon," broadening clinical relevance and highlighting potential immune risks of ABC phenomenon. The intensity of the ABC phenomenon correlated with the initial CE phospholipid dose in both species. Notably, larger CE particles (∼ 300 nm) induced the ABC phenomenon in beagles, while smaller particles (∼ 80 nm) with higher immunogenicity were required in rats. This suggested that beagles are more susceptible to CE-induced ABC phenomenon. The higher neutrophil proportion in beagles likely contributed to species differences in ABC phenomenon. This is the first study to report neutrophil involvement in ABC induction by non-PEGylated nanoparticles, more importantly, underscoring potential immune risks in the cross-injection of non-PEGylated and PEGylated nanoparticles during the developments and clinical applications of nano-drug delivery systems.
Collapse
Affiliation(s)
- Mengyang Liu
- Postdoctoral Research Station in Clinical Medicine of Hebei Medical University, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jia Wang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ge Chen
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, PR China
| | - Lirong Wang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xuling Wang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Bai Xiang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yihui Deng
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Chaoxing He
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Lei Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, PR China
| |
Collapse
|
3
|
Sindeeva OA, Kozyreva ZV, Abdurashitov AS, Sukhorukov GB. Engineering colloidal systems for cell manipulation, delivery, and tracking. Adv Colloid Interface Sci 2025; 340:103462. [PMID: 40037017 DOI: 10.1016/j.cis.2025.103462] [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: 10/14/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Men-made colloidal systems are widely presented across various aspects of biomedical science. There is a strong demand for engineering colloids to tailor their functions and properties to meet the requirements of biological and medical tasks. These requirements are not only related to size, shape, capacity to carry bioactive compounds as drug delivery systems, and the ability to navigate via chemical and physical targeting. Today, the more challenging aspects of colloid design are how the colloidal particles interact with biological cells, undergo internalization by cells, how they reside in the cell interior, and whether we can explore cells with colloids, intervene with biochemical processes, and alter cell functionality. Cell tracking, exploitation of cells as natural transporters of internalized colloidal carriers loaded with drugs, and exploring physical methods as external triggers of cell functions are ongoing topics in the research agenda. In this review, we summarize recent advances in these areas, focusing on how colloidal particles interact and are taken up by mesenchymal stem cells, dendritic cells, neurons, macrophages, neutrophils and lymphocytes, red blood cells, and platelets. The engineering of colloidal vesicles with cell membrane fragments and exosomes facilitates their application. The perspectives of different approaches in colloid design, their limitations, and obstacles on the biological side are discussed.
Collapse
Affiliation(s)
- Olga A Sindeeva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| | - Zhanna V Kozyreva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Arkady S Abdurashitov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia; Life Improvement by Future Technologies (LIFT) Center, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Gleb B Sukhorukov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| |
Collapse
|
4
|
Yao B, Zhang J, Chen Z, Qiu H, Xu D, Li Y, Yin S. Macrophage-membrane-engineered NIR II biomimetic nanomaterials for enhanced synergistic chemo-photothermal immunotherapy in cancer treatment. Colloids Surf B Biointerfaces 2025; 253:114759. [PMID: 40349454 DOI: 10.1016/j.colsurfb.2025.114759] [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: 02/01/2025] [Revised: 04/18/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Nanoparticle encapsulated with PEG-based polymers face limitations in their circulation stability and tumor tissue accumulation during blood transport due to the production of anti-PEG antibodies and their inherent nature as foreign substances, which leads to immune surveillance and clearance by the body. The design of biomimetic nanomaterials based on cell membranes offers a solution to these issues. In this context, we have successfully developed a biomimetic nanomaterials designed for the near-infrared region II (NIR II), which leverage the combined power of chemotherapy and photothermal therapy to activate an immune response against tumors. We synthesized nanoparticle loaded with IR1061 and doxorubicin (DOX) using microemulsion and nano-precipitation techniques, and then coated them with the pluronic (F127) polymer to enhance their stability and biocompatibility within biological systems. To further extend their circulation time and minimize the risk of immune detection, we encapsulated the nanoparticle within macrophage membranes. These customized nanoparticle, termed CIN and CDN, are capable of precisely targeting tumors through the bloodstream and effectively eliminating cancer cells under the dual onslaught of photothermal and chemotherapeutic actions. Throughout the treatment, the destruction of tumor cells triggers the release of antigens, which in turn activate CD4+ and CD8+ T cells, stimulating an immune response. Our findings indicate that the integration of chemotherapy with immunotherapy can significantly amplify the immune response by facilitating the demise of tumor cells, representing a highly promising synergistic strategy in the fight against cancer.
Collapse
Affiliation(s)
- Bo Yao
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China
| | - Jingpei Zhang
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China
| | - Zhenghui Chen
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China
| | - Huayu Qiu
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China
| | - Dongdong Xu
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China
| | - Yang Li
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China.
| | - Shouchun Yin
- Key Laboratory of Organosilicon Chemistry andMaterials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, PR China.
| |
Collapse
|
5
|
Long X, Cheng S, Lan X, Wei W, Jiang D. Trends in nanobody radiotheranostics. Eur J Nucl Med Mol Imaging 2025; 52:2225-2238. [PMID: 39800806 DOI: 10.1007/s00259-025-07077-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/04/2025] [Indexed: 04/23/2025]
Abstract
As the smallest antibody fragment with specific binding affinity, nanobody-based nuclear medicine has demonstrated significant potential to revolutionize the field of precision medicine, supported by burgeoning preclinical investigations and accumulating clinical evidence. However, the visualization of nanobodies has also exposed their suboptimal biodistribution patterns, which has spurred collaborative efforts to refine their pharmacokinetic and pharmacodynamic profiles for improved therapeutic efficacy. In this review, we present clinical results that exemplify the benefits of nanobody-based molecular imaging in cancer diagnosis. Moreover, we emphasize the indispensable role of molecular imaging as a tool for evaluating and optimizing nanobodies, thereby expanding their therapeutic potential in cancer treatment in the foreseeable future.
Collapse
Affiliation(s)
- Xingru Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
| | - Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200233, China.
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
| |
Collapse
|
6
|
Zhang L, Seow BYL, Bae KH, Zhang Y, Liao KC, Wan Y, Yang YY. Role of PEGylated lipid in lipid nanoparticle formulation for in vitro and in vivo delivery of mRNA vaccines. J Control Release 2025; 380:108-124. [PMID: 39875076 DOI: 10.1016/j.jconrel.2025.01.071] [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: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
mRNA-loaded lipid nanoparticles (mRNA-LNPs) hold great potential for disease treatment and prevention. LNPs are normally made from four lipids including ionizable lipid, helper lipid, cholesterol, and PEGylated lipid (PEG-lipid). Although PEG-lipid has the lowest content, it plays a crucial role in the effective delivery of mRNA-LNPs. However, previous studies have yet to elucidate the key factors of PEG-lipid that influence the properties of LNPs. This study reported how PEG-lipid content, lipid tail length, and chemical linkage between PEG and lipid affected in vitro and in vivo properties of mRNA-LNPs. Forty-eight LNP formulations were prepared and characterized. The results revealed that a PEG-lipid molar content exceeding 3.0 % significantly reduced the encapsulation efficiency of mRNA in LNPs via manual mixing. An increased PEG-lipid content also significantly decreased mRNA translation efficiency. Although the chemical linkage had minimal impact, the lipid tail length of PEG-lipid significantly affected the properties of mRNA-LNPs, irrespective of whether the LNPs were prepared using manual or microfluidic mixing. mRNA-LNPs made from ALC-0159 with C14 lipid tails, which is used in Pfizer/BioNTech COVID-19 mRNA vaccines, or C16-Ceramide-PEG preferably accumulated in the liver, while mRNA-LNPs prepared from C8-Ceramide-PEG were largely found in the lymph nodes. In a mouse SARS-CoV-2 Delta variant spike protein-encoded mRNA vaccine model, mRNA-LNPs made from either C8-Ceramide-PEG or C16-Ceramide-PEG yielded comparable vaccination efficacy to mRNA-LNPs made from ALC-0159, while mRNA-LNPs formulated with DSPE-PEG with C18 lipid tails mediated lower vaccination efficacy. C16-Ceramide-PEG LNPs and DSPE-PEG LNPs induced higher anti-PEG antibody response than C8-Ceramide-PEG and ALC-0159 LNPs. All the LNPs tested did not cause significant toxicity in mice. These results offer valuable insights into the use of PEG-lipid in LNP formulations and suggest that C8-Ceramide-PEG holds potential for use in the formulation of mRNA vaccine-loaded LNPs.
Collapse
Affiliation(s)
- Li Zhang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Brandon Yi Loong Seow
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Yue Zhang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Kuo-Chieh Liao
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Yue Wan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore.
| |
Collapse
|
7
|
Li X, Xu Z. Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases. Pharmaceutics 2025; 17:479. [PMID: 40284474 PMCID: PMC12030376 DOI: 10.3390/pharmaceutics17040479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) is implicated in tumor progression and vascular diseases, contributing to angiogenesis, metastasis, and extracellular matrix degradation. This review comprehensively examines the relationship between MMP-9 and these pathologies, exploring the underlying molecular mechanisms and signaling pathways involved. Specifically, we discuss the contribution of MMP-9 to tumor epithelial-mesenchymal transition, angiogenesis, and metastasis, as well as its involvement in a spectrum of vascular diseases, including macrovascular, cerebrovascular, and ocular vascular diseases. This review focuses on recent advances in MMP-9-targeted nanomedicine strategies, highlighting the design and application of responsive nanoparticles for enhanced drug delivery. These nanotherapeutic strategies leverage MMP-9 overexpression to achieve targeted drug release, improved tumor penetration, and reduced systemic toxicity. We explore various nanoparticle platforms, such as liposomes and polymer nanoparticles, and discuss their mechanisms of action, including degradation, drug release, and targeting specificity. Finally, we address the challenges posed by the heterogeneity of MMP-9 expression and their implications for personalized therapies. Ultimately, this review underscores the diagnostic and therapeutic potential of MMP-9-targeted nanomedicines against tumors and vascular diseases.
Collapse
Affiliation(s)
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
8
|
Tian J, Feng C, Xue H, You J, Shi M, Yin L. Development of a UPLC-MS/MS assay for determination of PA-PEG 8-PA polymers in rat plasma coupled with [M - H] - to enhance sensitivity. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:2676-2685. [PMID: 40099544 DOI: 10.1039/d4ay02050b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Propionic acid-polyethylene glycol-propionic acid (PA-PEG-PA) is a commonly used biocompatible polymer in drug delivery systems. Unraveling the in vivo pharmacokinetic behavior of PA-PEG-PA polymer is important for the safety evaluation of PA-PEG-PA related drug delivery systems. In this research, a highly sensitive ultra-high performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) assay was developed for detection of PA-PEG8-PA polymers in complex biological matrices. Methoxy-polyethylene glycol propionic acid polymers with 6 subunits (mPEG6-PA) were used as the internal standard (IS). The multiple reaction monitoring (MRM) transitions at m/z 513.4 ([M - H]- precursor ions) → 441.2 (fragment ions) and m/z 367.3 ([M - H]- precursor ions) → 118.8 (fragment ions) were chosen to determine PA-PEG8-PA and mPEG6-PA, respectively. The analysis time was only 5 min for each sample. Numerous parameters like specificity, sensitivity, accuracy, precision, recoveries, matrix effects and dilution effect were validated for the developed assay. The UPLC-MS/MS assay showed excellent linearity over the range of 30-1500 ng mL-1(r > 0.995). The assay was successfully applied to quantify the concentration of PA-PEG8-PA polymers in rat plasma.
Collapse
Affiliation(s)
- Jiye Tian
- Cancer Hospital of Dalian University of Technology, Dalian University of Technology, Shenyang, 110042, China
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, 124221, China
| | - Chunpeng Feng
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, 124221, China
| | - Hongyu Xue
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, 124221, China
| | - Jiansong You
- Aim Honesty Biopharmaceutical Co., Ltd, Dalian, Liaoning, 116600, China
| | - Meiyun Shi
- Cancer Hospital of Dalian University of Technology, Dalian University of Technology, Shenyang, 110042, China
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, 124221, China
- Aim Honesty Biopharmaceutical Co., Ltd, Dalian, Liaoning, 116600, China
| | - Lei Yin
- Cancer Hospital of Dalian University of Technology, Dalian University of Technology, Shenyang, 110042, China
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, 124221, China
| |
Collapse
|
9
|
Gu X, Du L, Lin R, Ding Z, Guo Z, Wei J, Li Y. How Advanced Is Nanomedicine for Atherosclerosis? Int J Nanomedicine 2025; 20:3445-3470. [PMID: 40125442 PMCID: PMC11928726 DOI: 10.2147/ijn.s508757] [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: 11/26/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in nanotechnology have opened new avenues for precision therapy, personalized medicine, and multifunctional theranostics in atherosclerosis (AS). This review provides a comprehensive overview of the role of nanoparticles (NPs) in precision medicine for AS, discussing their applications, challenges, and future prospects. The review first analyzes the current treatment landscape of AS and outlines potential biological targets for therapy. Various nanocarriers, including organic, inorganic, and hybrid systems, are evaluated for their therapeutic potential, with a focus on targeted drug delivery, anti-inflammatory therapy, vascular repair, plaque stabilization, and lipid clearance. Additionally, the review explores NP preparation methods, emphasizing strategies to enhance drug loading, stability, and controlled release. Finally, the translational challenges of NP-based therapies, including biocompatibility, large-scale production, regulatory hurdles, and clinical implementation, are critically analyzed. Future directions highlight the importance of interdisciplinary collaboration and technological innovation in advancing nanoparticle-based precision medicine for AS.
Collapse
Affiliation(s)
- Xiang Gu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lixin Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruifang Lin
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zehui Ding
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
10
|
Fang C, Cai Y, He C, Li Y, He L, Wang X, Lu Y. Endogenous Protein-Modified Gold Nanorods as Immune-Inert Biomodulators for Tumor-Specific Imaging and Therapy. Adv Healthc Mater 2025; 14:e2404548. [PMID: 39846276 PMCID: PMC11912115 DOI: 10.1002/adhm.202404548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Engineered modifications of nanomaterials inspired by nature hold great promise for disease-specific imaging and therapies. However, conventional polyethylene glycol modification is limited by immune system rejection. The manipulation of gold nanorods (Au NRs) modified by endogenous proteins (eP@Au) is reported as an engineered biomodulator for enhanced breast tumor therapy. The results show that eP@Au NRs neither activate inflammatory factors in vitro nor elicit rejection of immune responses in vivo. Tumor-specific eP@Au NRs exhibit a dual-modal imaging capability and trigger a mild photothermal effect under near-infrared light irradiation, enabling highly efficient imaging and therapy of tumors. Transcriptome sequencing and confirmatory experiments reveal that the antitumor effect is mainly attributed to the repression of PI3K-Akt/MAPK signaling pathways at the molecular level. This powerful and surprising in situ eP-regulated biomodulation demonstrates the advantages of convenient fabrication, inert immunogenicity, and biocompatibility, providing an alternative strategy for biomedical imaging and therapy.
Collapse
Affiliation(s)
- Chunyan Fang
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesInstitute of Translational MedicineShanghai Jiao Tong UniversityShanghai200240P.R. China
| | - Yueming Cai
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesInstitute of Translational MedicineShanghai Jiao Tong UniversityShanghai200240P.R. China
| | - Cui He
- Department of Basic MedicineShanxi Medical UniversityJinzhong030000P.R. China
| | - Ying Li
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesInstitute of Translational MedicineShanghai Jiao Tong UniversityShanghai200240P.R. China
| | - Lei He
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesInstitute of Translational MedicineShanghai Jiao Tong UniversityShanghai200240P.R. China
| | - Xiaoyan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200240P.R. China
| | - Yong Lu
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesInstitute of Translational MedicineShanghai Jiao Tong UniversityShanghai200240P.R. China
- School of Laboratory MedicineWannan Medical CollegeWuhu241002P.R. China
| |
Collapse
|
11
|
Rennie C, Morshed N, Faria M, Collins-Praino L, Care A. Nanoparticle Association with Brain Cells Is Augmented by Protein Coronas Formed in Cerebrospinal Fluid. Mol Pharm 2025; 22:940-957. [PMID: 39805033 DOI: 10.1021/acs.molpharmaceut.4c01179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Neuronanomedicine harnesses nanoparticle technology for the treatment of neurological disorders. An unavoidable consequence of nanoparticle delivery to biological systems is the formation of a protein corona on the nanoparticle surface. Despite the well-established influence of the protein corona on nanoparticle behavior and fate, as well as FDA approval of neuro-targeted nanotherapeutics, the effect of a physiologically relevant protein corona on nanoparticle-brain cell interactions is insufficiently explored. Indeed, less than 1% of protein corona studies have investigated protein coronas formed in cerebrospinal fluid (CSF), the fluid surrounding the brain. Herein, we utilize two clinically relevant polymeric nanoparticles (PLGA and PLGA-PEG) to evaluate the formation of serum and CSF protein coronas. LC-MS analysis revealed distinct protein compositions, with selective enrichment/depletion profiles. Enhanced association of CSF precoated particles with brain cells demonstrates the importance of selecting physiologically relevant biological fluids to more accurately study protein corona formation and subsequent nanoparticle-cell interactions, paving the way for improved nanoparticle engineering for in vivo applications.
Collapse
Affiliation(s)
- Claire Rennie
- School of Life Sciences, University of Technology Sydney, Sydney 2007, New South Wales, Australia
- Australian Institute for Microbiology and Infection, Sydney 2007, New South Wales, Australia
| | - Nabila Morshed
- School of Life Sciences, University of Technology Sydney, Sydney 2007, New South Wales, Australia
| | - Matthew Faria
- Department of Biomedical Engineering, The University of Melbourne, Melbourne 3010, Victoria, Australia
| | - Lyndsey Collins-Praino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney 2007, New South Wales, Australia
| |
Collapse
|
12
|
Tian Y, Lv H, Ju Y, Hao J, Cui J. Zwitterionic Poly(ethylene glycol) Nanoparticles Minimize Protein Adsorption and Immunogenicity for Improved Biological Fate. ACS APPLIED MATERIALS & INTERFACES 2025; 17:6125-6133. [PMID: 39824773 DOI: 10.1021/acsami.4c20890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
We report the assembly of poly(ethylene glycol) nanoparticles (PEG NPs) and optimize their surface chemistry to minimize the formation of protein coronas and immunogenicity for improved biodistribution. PEG NPs cross-linked with disulfide bonds are synthesized utilizing zeolitic imidazolate framework-8 NPs as the templates, which are subsequently modified with PEG molecules with different end groups (carboxyl, methoxy, or amino) to vary the surface chemistry. Among the modifications, the amino and residual carboxyl groups form a pair of zwitterionic structures on the surface of PEG NPs, which minimize the adsorption of proteins (e.g., immunoglobulin, complement proteins) and maximize the blood circulation time. The influence of preexisting PEG antibodies in mice on the pharmacokinetics of zwitterionic PEG NPs is negligible, which demonstrates the resistance of anti-PEG antibodies and inhibition of the accelerated blood clearance phenomenon. This research highlights the importance of the surface chemistry of PEGylated NPs in the design of delivery systems and reveals their translational potential for cancer therapy.
Collapse
Affiliation(s)
- Yuan Tian
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Huiyuan Lv
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Yi Ju
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
13
|
Kojima C, Hirata R, Dei N, He H, Ikemoto Y, Matsumoto A. Hydration and Biodistribution of Zwitterionic Dendrimers Conjugating a Sulfobetaine Monomer and Polymers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:1411-1417. [PMID: 39778908 PMCID: PMC11755784 DOI: 10.1021/acs.langmuir.4c04276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Zwitterionic polymers exhibit strong hydration, high biocompatibility, and antifouling properties. Dendrimers are regularly branched polymers, which are used in the drug delivery system (DDS). In this study, we synthesized zwitterionic monomer- and polymer-conjugated dendrimers as a biocompatible nanoparticle to investigate the relation between the hydration property and biodistribution. A sulfobetaine monomer (SBM) was conjugated at the termini of the polyamidoamine (PAMAM) dendrimer. Polysulfobetaines (PSBs) were produced by reversible addition-fragmentation chain transfer polymerization and were also conjugated at the termini. Intermediate water, that is, water molecules loosely bound to the material, can be estimated from the melting peaks at less than 0 °C in differential scanning calorimetry (DSC) measurement. Our DSC results showed that the PSB-conjugated dendrimers (PSM-dens) contained more intermediate water than the SBM-conjugated dendrimer (SBM-den). PSB-dens accumulated in the tumor after intravenous administration, but SBM-den did not. These suggested that the amount of intermediate water, that is, the hydration property, was related to the biodistribution of the zwitterionic dendrimers. This relation is a possible design criterion for drug carriers. PSB-dens accumulated in the tumor even after the second injection, possibly overcoming the accelerated blood clearance observed with poly(ethylene glycol)-modified nanoparticles. Thus, this kind of zwitterionic polymer-conjugated dendrimer is useful for the DDS in cancer treatment.
Collapse
Affiliation(s)
- Chie Kojima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Rikuto Hirata
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Nanako Dei
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Hao He
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yuka Ikemoto
- Spectroscopy Division, Japan Synchrotron Radiation Research Institute, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5198, Japan
| | - Akikazu Matsumoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
14
|
Morbidelli M, Romio M, Chandorkar Y, Gogos A, Hirsch C, Kolrosova B, Trachsel L, Lorandi F, Badocco D, Pastore P, Arrigoni G, Franchin C, Tavano R, Hoogenboom R, Papini E, Benetti EM. The Topology of Poly(2-methyl-2-oxazine) Shells on Nanoparticles Determines Their Interaction with Serum and Uptake by Immune Cells. Biomacromolecules 2025; 26:556-566. [PMID: 39725524 DOI: 10.1021/acs.biomac.4c01340] [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: 12/28/2024]
Abstract
Cyclic poly(2-methyl-2-oxazine) (c-PMOZI) brush shells on Au nanoparticles (NPs) exhibit enhanced stealth properties toward serum and different cell lines compared to their linear PMOZI (l-PMOZI) counterparts. While selectively recruiting immunoglobulins, c-PMOZI shells reduce overall human serum (HS) protein binding and alter the processing of complement factor 3 (C3) compared to chemically identical linear shells. Polymer cyclization significantly decreases NP uptake by nonphagocytic cells and macrophages in both complement-deficient fetal bovine serum (FBS) and complement-expressing HS, indicating ineffective functional opsonization. Even in serum-free media, c-PMOZI-coated NPs show reduced internalization by macrophages compared to l-PMOZI-coated NPs, suggesting lower opsonin-independent cell surface affinity. This study demonstrates that cyclic PMOZI suppresses interactions of NPs with proteins and cells, highlighting how control over chain topology expands the polymer chemistry toolbox for modulating the behavior of core-shell NPs within physiological environments.
Collapse
Affiliation(s)
- Maria Morbidelli
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | - Matteo Romio
- Biointerfaces Lab, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5 St. Gallen 9014, Switzerland
| | - Yashoda Chandorkar
- Biointerfaces Lab, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5 St. Gallen 9014, Switzerland
| | - Alexander Gogos
- Particles-Biology Interactions Lab, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Cordula Hirsch
- Particles-Biology Interactions Lab, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Barbora Kolrosova
- Biointerfaces Lab, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5 St. Gallen 9014, Switzerland
| | - Lucca Trachsel
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Francesca Lorandi
- Laboratory for Macromolecular and Organic Chemistry, Department of Chemical Sciences, University of Padova, Padova 35131, Italy
| | - Denis Badocco
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, Padua 35131, Italy
| | - Paolo Pastore
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, Padua 35131, Italy
| | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | - Cinzia Franchin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, Padova 35131, Italy
| | - Regina Tavano
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Department of Organic Chemistry and Macromolecular Chemistry, Ghent University, Krijgslaan 281, Gent S4 B-9000, Belgium
| | - Emanuele Papini
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | - Edmondo M Benetti
- Laboratory for Macromolecular and Organic Chemistry, Department of Chemical Sciences, University of Padova, Padova 35131, Italy
| |
Collapse
|
15
|
Tan YF, Hii LW, Lim WM, Cheong SK, Leong CO, Yee MSL, Mai CW. Polyethylene glycol-phospholipid functionalized single-walled carbon nanotubes for enhanced siRNA systemic delivery. Sci Rep 2024; 14:30098. [PMID: 39627280 PMCID: PMC11615393 DOI: 10.1038/s41598-024-80646-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
Small interfering RNAs (siRNA) technology has emerged as a promising therapeutic tool for human health conditions like cancer due to its ability to regulate gene silencing. Despite FDA-approved, their delivery remains localized and limiting their systemic use. This study used single-walled carbon nanotubes (SWNTs) functionalized with polyethylene glycolated (PEGylated) phospholipids (PL-PEG) derivatives for systemic siRNA delivery. We developed an siRNA systemic delivery vehicle (SWNT-siRNA) by conjugating SWNT functionalized with PL-PEG containing either amine (PA) or maleimide (MA). The functionalized SWNT with a lower molecular weight of PA produced the SWNT-siRNA conjugate system with the highest stability and high siRNA loading quantity. The system delivered siRNA to a panel of tumour cell lines of different organs (i.e. HeLa, H1299 and MCF-7) and a non-cancerous human embryonic kidney 293 cells (HEK293T) with high biocompatibility and low toxicity. The cellular uptake of SWNT-siRNA conjugates by epithelial cells was found to be energy dependent. Importantly, the presence of P-glycoprotein, a marker for drug resistance, did not inhibit SWNT-mediated siRNA delivery. Mouse xenograft model further confirmed the potential of SWNT-siRNA conjugates with a significant gene knock-down without signs of acute toxicity. These findings pave the way for potential gene therapy applications using SWNTs as delivery vehicles.
Collapse
Affiliation(s)
- Yuen-Fen Tan
- Centre for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), IMU University, Kuala Lumpur, 57000, Malaysia
- School of Postgraduate Studies, IMU University, Kuala Lumpur, 57000, Malaysia
- Cytovision Sdn. Bhd, Kuala Lumpur, 57000, Malaysia
| | - Ling-Wei Hii
- Centre for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), IMU University, Kuala Lumpur, 57000, Malaysia
| | - Wei-Meng Lim
- Centre for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), IMU University, Kuala Lumpur, 57000, Malaysia
- School of Pharmacy, Monash University Malaysia, Shah Alam, 47500, Selangor, Malaysia
| | - Soon-Keng Cheong
- Centre for Stem Cell Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Shah Alam, 43000, Selangor, Malaysia
| | - Chee-Onn Leong
- Centre for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), IMU University, Kuala Lumpur, 57000, Malaysia
- School of Pharmacy, IMU University, Kuala Lumpur, 57000, Malaysia
- AGTC Genomics Sdn. Bhd, Kuala Lumpur, 57000, Malaysia
| | - Maxine Swee-Li Yee
- Nanotechnology Research Group, Center for Nanotechnology and Advanced Materials, University of Nottingham Malaysia, Semenyih, 43500, Selangor, Malaysia.
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), IMU University, Kuala Lumpur, 57000, Malaysia.
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
16
|
Barros AS, Pinto S, Viegas J, Martins C, Almeida H, Alves I, Pinho S, Nunes R, Harris A, Sarmento B. Orally Delivered Stimulus-Sensitive Nanomedicine to Harness Teduglutide Efficacy in Inflammatory Bowel Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402502. [PMID: 39007246 DOI: 10.1002/smll.202402502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/03/2024] [Indexed: 07/16/2024]
Abstract
Inflammatory Bowel Disease (IBD) is a chronic inflammatory condition affecting the gastrointestinal tract (GIT). Glucagon-like peptide-2 (GLP-2) analogs possess high potential in the treatment of IBD by enhancing intestinal repair and attenuating inflammation. Due to the enzymatic degradation and poor intestinal absorption, GLP-2 analogs are administered parenterally, which leads to poor patient compliance. This work aims to develop IBD-targeted nanoparticles (NPs) for the oral delivery of the GLP-2 analog, Teduglutide (TED). Leveraging the overproduction of Reactive Oxygen Species (ROS) in the IBD environment, ROS-sensitive NPs are developed to target the intestinal epithelium, bypassing the mucus barrier. PEGylation of NPs facilitates mucus transposition, but subsequent PEG removal is crucial for cellular internalization. This de-PEGylation is possible by including a ROS-sensitive thioketal linker within the system. ROS-sensitive NPs are established, with the ability to fully de-PEGylate via ROS-mediated cleavage. Encapsulation of TED into NPs resulted in the absence of absorption in 3D in vitro models, potentially promoting a localized action, and avoiding adverse effects due to systemic absorption. Upon oral administration to colitis-induced mice, ROS-sensitive NPs are located in the colon, displaying healing capacity and reducing inflammation. Cleavable PEGylated NPs demonstrate effective potential in managing IBD symptoms and modulating the disease's progression.
Collapse
Affiliation(s)
- Andreia S Barros
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
| | - Soraia Pinto
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
| | - Juliana Viegas
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
| | - Claúdia Martins
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
| | - Helena Almeida
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
| | - Inês Alves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
| | - Salomé Pinho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
- FMUP- Faculty of Medicine, University of Porto, Porto, 4200-319, Portugal
| | - Rute Nunes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- IUCS-CESPU-Instituto Universitário de Ciências das Saúde, Porto, 4585-116, Portugal
| | - Alan Harris
- Ferring Pharmaceuticals, 1162-Saint-Prex, SA Chemin de la Vergognausaz 50, Switzerland
| | - Bruno Sarmento
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- IUCS-CESPU-Instituto Universitário de Ciências das Saúde, Porto, 4585-116, Portugal
| |
Collapse
|
17
|
Jain M, Yu X, Schneck JP, Green JJ. Nanoparticle Targeting Strategies for Lipid and Polymer-Based Gene Delivery to Immune Cells In Vivo. SMALL SCIENCE 2024; 4:2400248. [PMID: 40212067 PMCID: PMC11935263 DOI: 10.1002/smsc.202400248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/15/2024] [Indexed: 04/13/2025] Open
Abstract
Lipid nanoparticles and polymeric nanoparticles are promising biomaterial platforms for robust intracellular DNA and mRNA delivery, highlighted by the widespread use of nanoparticle- (NP) based mRNA vaccines to help end the COVID-19 pandemic. Recent research has sought to adapt this nanotechnology to transfect and engineer immune cells in vivo. The immune system is an especially appealing target due to its involvement in many different diseases, and ex vivo-engineered immune cell therapies like chimeric antigen receptor (CAR) T therapy have already demonstrated remarkable clinical success in certain blood cancers. Although gene delivery can potentially address some of the cost and manufacturing concerns associated with current autologous immune cell therapies, transfecting immune cells in vivo is challenging. Not only is extrahepatic NP delivery to lymphoid organs difficult, but immune cells like T cells have demonstrated particular resistance to transfection. Despite these challenges, the modular nature of NPs allows researchers to examine critical structure-function relationships between a particle's properties and its ability to specifically engineer immune cells in vivo. Herein, several nanomaterial components are outlined, including targeting ligands, nucleic acid cargo, chemical properties, physical properties, and the route of administration to specifically target NPs to immune cells for optimal in vivo transfection.
Collapse
Affiliation(s)
- Manav Jain
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Xinjie Yu
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jonathan P. Schneck
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Pathology and MedicineJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of OncologyThe Sidney Kimmel Comprehensive Cancer CenterThe Bloomberg∼Kimmel Institute for Cancer ImmunotherapyJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Ophthalmology, Neurosurgery, and Materials Science & EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
18
|
Jarak I, Ramos S, Caldeira B, Domingues C, Veiga F, Figueiras A. The Many Faces of Cyclodextrins within Self-Assembling Polymer Nanovehicles: From Inclusion Complexes to Valuable Structural and Functional Elements. Int J Mol Sci 2024; 25:9516. [PMID: 39273469 PMCID: PMC11395033 DOI: 10.3390/ijms25179516] [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: 06/12/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 09/15/2024] Open
Abstract
Most chemotherapeutic agents are poorly soluble in water, have low selectivity, and cannot reach the tumor in the desired therapeutic concentration. On the other hand, sensitive hydrophilic therapeutics like nucleic acids and proteins suffer from poor bioavailability and cell internalization. To solve this problem, new types of controlled release systems based on nano-sized self-assemblies of cyclodextrins able to control the speed, timing, and location of therapeutic release are being developed. Cyclodextrins are macrocyclic oligosaccharides characterized by a high synthetic plasticity and potential for derivatization. Introduction of new hydrophobic and/or hydrophilic domains and/or formation of nano-assemblies with therapeutic load extends the use of CDs beyond the tried-and-tested CD-drug host-guest inclusion complexes. The recent advances in nano drug delivery have indicated the benefits of the hybrid amphiphilic CD nanosystems over individual CD and polymer components. This review provides a comprehensive overview of the most recent advances in the design of CDs self-assemblies and their use for delivery of a wide range of therapeutic molecules. It aims to offer a valuable insight into the many roles of CDs within this class of drug nanocarriers as well as current challenges and future perspectives.
Collapse
Affiliation(s)
- Ivana Jarak
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Sara Ramos
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Beatriz Caldeira
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Cátia Domingues
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
19
|
Xie M, Meng F, Wang P, Díaz-García AM, Parkhats M, Santos-Oliveira R, Asim MH, Bostan N, Gu H, Yang L, Li Q, Yang Z, Lai H, Cai Y. Surface Engineering of Magnetic Iron Oxide Nanoparticles for Breast Cancer Diagnostics and Drug Delivery. Int J Nanomedicine 2024; 19:8437-8461. [PMID: 39170101 PMCID: PMC11338174 DOI: 10.2147/ijn.s477652] [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: 05/10/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Data published in 2020 by the International Agency for Research on Cancer (IARC) of the World Health Organization show that breast cancer (BC) has become the most common cancer globally, affecting more than 2 million women each year. The complex tumor microenvironment, drug resistance, metastasis, and poor prognosis constitute the primary challenges in the current diagnosis and treatment of BC. Magnetic iron oxide nanoparticles (MIONPs) have emerged as a promising nanoplatform for diagnostic tumor imaging as well as therapeutic drug-targeted delivery due to their unique physicochemical properties. The extensive surface engineering has given rise to multifunctionalized MIONPs. In this review, the latest advancements in surface modification strategies of MIONPs over the past five years are summarized and categorized as constrast agents and drug delivery platforms. Additionally, the remaining challenges and future prospects of MIONPs-based targeted delivery are discussed.
Collapse
Affiliation(s)
- Mengjie Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China / Guangdong Key Laboratory of Traditional Chinese Medicine Informatization / International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
| | - Fansu Meng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, 528400, People’s Republic of China
| | - Panpan Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
| | | | - Marina Parkhats
- B. I. Stepanov Institute of Physics, National Academy of Sciences of Belarus, Minsk, 220072, Belarus
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro, RJ, 21941906, Brazil
| | | | - Nazish Bostan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Honghui Gu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Lina Yang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Qi Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Zhenjiang Yang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Haibiao Lai
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, 528400, People’s Republic of China
| | - Yu Cai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China / Guangdong Key Laboratory of Traditional Chinese Medicine Informatization / International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
| |
Collapse
|
20
|
Kojima C, Yao J, Nakajima K, Suzuki M, Tsujimoto A, Kuge Y, Ogawa M, Matsumoto A. Attenuated polyethylene glycol immunogenicity and overcoming accelerated blood clearance of a fully PEGylated dendrimer. Int J Pharm 2024; 659:124193. [PMID: 38703934 DOI: 10.1016/j.ijpharm.2024.124193] [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: 01/18/2024] [Revised: 04/20/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Polyethylene glycol (PEG) is a popular biocompatible polymer and PEGylated nanoparticles passively accumulate in tumor tissues because of their enhanced permeability and retention effects. Recently, the anti-PEG immunity of PEGylated nanoparticles has become an issue that needs to be solved for their clinical applications. Dendrimers are highly branched and well-defined polymers with many terminal groups, which act as potent drug carriers. In this study, we examined the pharmacokinetics, biodistribution, anti-PEG immunity, and tumor accumulation of a fully PEGylated polyamidoamine (PAMAM) dendrimer after the first and second injections and compared them to those of a PEGylated liposome with the same lipid component as Doxil®. The PEGylated dendrimer showed greater blood circulation than that of the PEGylated liposome after the first and second injections in rats. In mice injected with the PEGylated dendrimer, much less anti-PEG immunoglobulin M (IgM) was generated than that in mice injected with the PEGylated liposome. The PEGylated dendrimer accumulated in the tumor after both the first and second injections. Our results indicated that the PEGylated dendrimer with a small size and high PEG density showed attenuated anti-PEG immunity and overcame the accelerated blood clearance phenomenon, which is useful for drug delivery systems for cancer treatment.
Collapse
Affiliation(s)
- Chie Kojima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan.
| | - Junjie Yao
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan
| | - Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Motofumi Suzuki
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Ayako Tsujimoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan
| | - Yuji Kuge
- Central Institutes of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-0815, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Akikazu Matsumoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan
| |
Collapse
|
21
|
Lin W, Li A, Qiu L, Huang H, Cui P, Wang J. Albumin Nanoparticles Increase the Efficacy of Doxorubicin Hydrochloride Liposome Injection Based on Threshold Theory. Mol Pharm 2024; 21:2970-2980. [PMID: 38742943 DOI: 10.1021/acs.molpharmaceut.4c00097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
One of the most significant reasons hindering the clinical translation of nanomedicines is the rapid clearance of intravenously injected nanoparticles by the mononuclear phagocyte system, particularly by Kupffer cells in the liver, leading to an inefficient delivery of nanomedicines for tumor treatment. The threshold theory suggests that the liver's capacity to clear nanoparticles is limited, and a single high dose of nanoparticles can reduce the hepatic clearance efficiency, allowing more nanomedicines to reach tumor tissues and enhance therapeutic efficacy. Building upon this theory, researchers have conducted numerous validation studies based on the same nanoparticle carrier systems. These studies involve the use of albumin nanoparticles to improve the therapeutic efficacy of albumin nanomedicines as well as polyethylene glycol (PEG)-modified liposomal nanoparticles to enhance the efficacy of PEGylated liposomal nanomedicines. However, there is no research indicating the feasibility of the threshold theory when blank nanoparticles and nanomedicine belong to different nanoparticle carrier systems currently. In this study, we prepared two different sizes of albumin nanoparticles by using bovine serum albumin. We used the marketed nanomedicine liposomal doxorubicin hydrochloride injection (trade name: LIBOD, manufacturer: Shanghai Fudan-zhangjiang Biopharmaceutical Co., Ltd.), as the representative nanomedicine. Through in vivo experiments, we found that using threshold doses of albumin nanoparticles still can reduce the clearance rate of LIBOD, prolong its time in vivo, increase the area under the plasma concentration-time curve (AUC), and also lead to an increased accumulation of the drug at the tumor site. Furthermore, evaluation of in vivo efficacy and safety further indicates that threshold doses of 100 nm albumin nanoparticles can enhance the antitumor effect of LIBOD without causing harm to the animals. During the study, we found that the particle size of albumin nanoparticles influenced the in vivo distribution of the nanomedicine at the same threshold dose. Compared with 200 nm albumin nanoparticles, 100 nm albumin nanoparticles more effectively reduce the clearance efficiency of LIBOD and enhance nanomedicine accumulation at the tumor site, warranting further investigation. This study utilized albumin nanoparticles to reduce hepatic clearance efficiency and enhance the delivery efficiency of nonalbumin nanocarrier liposomal nanomedicine, providing a new avenue to improve the efficacy and clinical translation of nanomedicines with different carrier systems.
Collapse
Affiliation(s)
- Wei Lin
- School of Petroleum and Chemical Engineering, Changzhou University, Changzhou 213164, P. R. China
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Anyin Li
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Hai Huang
- School of Petroleum and Chemical Engineering, Changzhou University, Changzhou 213164, P. R. China
| | - Pengfei Cui
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| |
Collapse
|
22
|
Liu H, Ji M, Xiao P, Gou J, Yin T, He H, Tang X, Zhang Y. Glucocorticoids-based prodrug design: Current strategies and research progress. Asian J Pharm Sci 2024; 19:100922. [PMID: 38966286 PMCID: PMC11222810 DOI: 10.1016/j.ajps.2024.100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/04/2024] [Accepted: 03/06/2024] [Indexed: 07/06/2024] Open
Abstract
Attributing to their broad pharmacological effects encompassing anti-inflammation, antitoxin, and immunosuppression, glucocorticoids (GCs) are extensively utilized in the clinic for the treatment of diverse diseases such as lupus erythematosus, nephritis, arthritis, ulcerative colitis, asthma, keratitis, macular edema, and leukemia. However, long-term use often causes undesirable side effects, including metabolic disorders-induced Cushing's syndrome (buffalo back, full moon face, hyperglycemia, etc.), osteoporosis, aggravated infection, psychosis, glaucoma, and cataract. These notorious side effects seriously compromise patients' quality of life, especially in patients with chronic diseases. Therefore, glucocorticoid-based advanced drug delivery systems for reducing adverse effects have received extensive attention. Among them, prodrugs have the advantages of low investment, low risk, and high success rate, making them a promising strategy. In this review, we propose the strategies for the design and summarize current research progress of glucocorticoid-based prodrugs in recent decades, including polymer-based prodrugs, dendrimer-based prodrugs, antibody-drug conjugates, peptide-drug conjugates, carbohydrate-based prodrugs, aliphatic acid-based prodrugs and so on. Besides, we also raise issues that need to be focused on during the development of glucocorticoid-based prodrugs. This review is expected to be helpful for the research and development of novel GCs and prodrugs.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Muse Ji
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peifu Xiao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingxin Gou
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Yin
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haibing He
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xing Tang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
23
|
He W, Gao H, Wu W. Nanomedicine biointeractions during body trafficking. Adv Drug Deliv Rev 2024; 209:115324. [PMID: 38663551 DOI: 10.1016/j.addr.2024.115324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Affiliation(s)
- Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Wei Wu
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
24
|
Mahmud MM, Pandey N, Winkles JA, Woodworth GF, Kim AJ. Toward the scale-up production of polymeric nanotherapeutics for cancer clinical trials. NANO TODAY 2024; 56:102314. [PMID: 38854931 PMCID: PMC11155436 DOI: 10.1016/j.nantod.2024.102314] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Nanotherapeutics have gained significant attention for the treatment of numerous cancers, primarily because they can accumulate in and/or selectively target tumors leading to improved pharmacodynamics of encapsulated drugs. The flexibility to engineer the nanotherapeutic characteristics including size, morphology, drug release profiles, and surface properties make nanotherapeutics a unique platform for cancer drug formulation. Polymeric nanotherapeutics including micelles and dendrimers represent a large number of formulation strategies developed over the last decade. However, compared to liposomes and lipid-based nanotherapeutics, polymeric nanotherapeutics have had limited clinical translation from the laboratory. One of the key limitations of polymeric nanotherapeutics formulations for clinical translation has been the reproducibility in preparing consistent and homogeneous large-scale batches. In this review, we describe polymeric nanotherapeutics and discuss the most common laboratory and scale-up formulation methods, specifically those proposed for clinical cancer therapies. We also provide an overview of the major challenges and opportunities for scaling polymeric nanotherapeutics to clinical-grade formulations. Finally, we will review the regulatory requirements and challenges in advancing nanotherapeutics to the clinic.
Collapse
Affiliation(s)
- Md Musavvir Mahmud
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey A. Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Graeme F. Woodworth
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Anthony J. Kim
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| |
Collapse
|
25
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [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: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
26
|
Gatto MS, Johnson MP, Najahi-Missaoui W. Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life (Basel) 2024; 14:672. [PMID: 38929656 PMCID: PMC11204409 DOI: 10.3390/life14060672] [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: 03/30/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
In drug development, it is not uncommon that an active substance exhibits efficacy in vitro but lacks the ability to specifically reach its target in vivo. As a result, targeted drug delivery has become a primary focus in the pharmaceutical sciences. Since the approval of Doxil® in 1995, liposomes have emerged as a leading nanoparticle in targeted drug delivery. Their low immunogenicity, high versatility, and well-documented efficacy have led to their clinical use against a wide variety of diseases. That being said, every disease is accompanied by a unique set of physiological conditions, and each liposomal product must be formulated with this consideration. There are a multitude of different targeting techniques for liposomes that can be employed depending on the application. Passive techniques such as PEGylation or the enhanced permeation and retention effect can improve general pharmacokinetics, while active techniques such as conjugating targeting molecules to the liposome surface may bring even further specificity. This review aims to summarize the current strategies for targeted liposomes in the treatment of diseases.
Collapse
Affiliation(s)
| | | | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA; (M.S.G.); (M.P.J.)
| |
Collapse
|
27
|
Li J, Li W, Zhuang L. Natural biomimetic nano-system for drug delivery in the treatment of rheumatoid arthritis: a literature review of the last 5 years. Front Med (Lausanne) 2024; 11:1385123. [PMID: 38784236 PMCID: PMC11114446 DOI: 10.3389/fmed.2024.1385123] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized primarily by synovitis, leading to the destruction of articular cartilage and bone and ultimately resulting in joint deformity, loss of function, and a significant impact on patients' quality of life. Currently, a combination of anti-rheumatic drugs, hormonal drugs, and biologics is used to mitigate disease progression. However, conventional drug therapy has limited bioavailability, and long-term use often leads to drug resistance and toxic side effects. Therefore, exploring new therapeutic approaches for RA is of great clinical importance. Nanodrug delivery systems offer promising solutions to overcome the limitations of conventional drugs. Among them, liposomes, the first nanodrug delivery system to be approved for clinical application and still widely studied, demonstrate the ability to enhance therapeutic efficacy with fewer adverse effects through passive or active targeting mechanisms. In this review, we provide a review of the research progress on the targeting mechanisms of various natural biomimetic nano-delivery systems in RA therapy. Additionally, we predict the development trends and application prospects of these systems, offering new directions for precision treatment of RA.
Collapse
Affiliation(s)
| | | | - Liping Zhuang
- Beidahuang Group Mudanjiang Hospital, Mudanjiang, Heilongjiang, China
| |
Collapse
|
28
|
Pan F, Liu M, Li G, Chen B, Chu Y, Yang Y, Wu E, Yu Y, Lin S, Ding T, Wei X, Zhan C, Qian J. Phospholipid Type Regulates Protein Corona Composition and In Vivo Performance of Lipid Nanodiscs. Mol Pharm 2024; 21:2272-2283. [PMID: 38607681 DOI: 10.1021/acs.molpharmaceut.3c01084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Over the years, there has been significant interest in PEGylated lipid-based nanocarriers within the drug delivery field. The inevitable interplay between the nanocarriers and plasma protein plays a pivotal role in their in vivo biological fate. Understanding the factors influencing lipid-based nanocarrier and protein corona interactions is of paramount importance in the design and clinical translation of these nanocarriers. Herein, discoid-shaped lipid nanodiscs (sNDs) composed of different phospholipids with varied lipid tails and head groups were fabricated. We investigated the impact of phospholipid components on the interaction between sNDs and serum proteins, particle stability, and biodistribution. The results showed that all of these lipid nanodiscs remained stable over a 15 day storage period, while their stability in the blood serum demonstrated significant differences. The sND composed of POPG exhibited the least stability due to its potent complement activation capability, resulting in rapid blood clearance. Furthermore, a negative correlation between the complement activation capability and serum stability was identified. Pharmacokinetic and biodistribution experiments indicated that phospholipid composition did not influence the capability of sNDs to evade the accelerated blood clearance phenomenon. Complement deposition on the sND was inversely associated with the area under the curve. Additionally, all lipid nanodiscs exhibited dominant adsorption of apolipoprotein. Remarkably, the POPC-based lipid nanodisc displayed a significantly higher deposition of apolipoprotein E, contributing to an obvious brain distribution, which provides a promising tool for brain-targeted drug delivery.
Collapse
Affiliation(s)
- Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| | - Mengyuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| | - Guanghui Li
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| | - Boqian Chen
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Yuxiu Chu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Ercan Wu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Yifei Yu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Shiqi Lin
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Tianhao Ding
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoli Wei
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Jun Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
29
|
Liu J, Yang F, Hu J, Zhang X. Nanoparticles for efficient drug delivery and drug resistance in glioma: New perspectives. CNS Neurosci Ther 2024; 30:e14715. [PMID: 38708806 PMCID: PMC11071172 DOI: 10.1111/cns.14715] [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: 12/30/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/07/2024] Open
Abstract
Gliomas are the most common primary tumors of the central nervous system, with glioblastoma multiforme (GBM) having the highest incidence, and their therapeutic efficacy depends primarily on the extent of surgical resection and the efficacy of postoperative chemotherapy. The role of the intracranial blood-brain barrier and the occurrence of the drug-resistant gene O6-methylguanine-DNA methyltransferase have greatly limited the efficacy of chemotherapeutic agents in patients with GBM and made it difficult to achieve the expected clinical response. In recent years, the rapid development of nanotechnology has brought new hope for the treatment of tumors. Nanoparticles (NPs) have shown great potential in tumor therapy due to their unique properties such as light, heat, electromagnetic effects, and passive targeting. Furthermore, NPs can effectively load chemotherapeutic drugs, significantly reduce the side effects of chemotherapeutic drugs, and improve chemotherapeutic efficacy, showing great potential in the chemotherapy of glioma. In this article, we reviewed the mechanisms of glioma drug resistance, the physicochemical properties of NPs, and recent advances in NPs in glioma chemotherapy resistance. We aimed to provide new perspectives on the clinical treatment of glioma.
Collapse
Affiliation(s)
- Jiyuan Liu
- Department of Neurosurgerythe First Hospital of China Medical UniversityShenyangChina
| | - Fan Yang
- Department of Cardiologythe Fourth Affiliated Hospital of China Medical UniversityShenyangChina
| | - Jinqu Hu
- Department of Neurosurgerythe First Hospital of China Medical UniversityShenyangChina
| | - Xiuchun Zhang
- Department of Neurologythe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
30
|
Huang L, Luo S, Tong S, Lv Z, Wu J. The development of nanocarriers for natural products. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1967. [PMID: 38757428 DOI: 10.1002/wnan.1967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/01/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
Natural bioactive compounds from plants exhibit substantial pharmacological potency and therapeutic value. However, the development of most plant bioactive compounds is hindered by low solubility and instability. Conventional pharmaceutical forms, such as tablets and capsules, only partially overcome these limitations, restricting their efficacy. With the recent development of nanotechnology, nanocarriers can enhance the bioavailability, stability, and precise intracellular transport of plant bioactive compounds. Researchers are increasingly integrating nanocarrier-based drug delivery systems (NDDS) into the development of natural plant compounds with significant success. Moreover, natural products benefit from nanotechnological enhancement and contribute to the innovation and optimization of nanocarriers via self-assembly, grafting modifications, and biomimetic designs. This review aims to elucidate the collaborative and reciprocal advancement achieved by integrating nanocarriers with botanical products, such as bioactive compounds, polysaccharides, proteins, and extracellular vesicles. This review underscores the salient challenges in nanomedicine, encompassing long-term safety evaluations of nanomedicine formulations, precise targeting mechanisms, biodistribution complexities, and hurdles in clinical translation. Further, this study provides new perspectives to leverage nanotechnology in promoting the development and optimization of natural plant products for nanomedical applications and guiding the progression of NDDS toward enhanced efficiency, precision, and safety. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Liying Huang
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Shicui Luo
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Sen Tong
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhuo Lv
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Junzi Wu
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Clinical Medical Research Center for Geriatric Diseases, Yunnan First People's Hospital, Kunming, Yunnan, China
| |
Collapse
|
31
|
Meng Y, Gao J, Zhou P, Qin X, Tian M, Wang X, Zhou C, Li K, Huang F, Cao Y. NIR-II Conjugated Electrolytes as Biomimetics of Lipid Bilayers for In Vivo Liposome Tracking. Angew Chem Int Ed Engl 2024; 63:e202318632. [PMID: 38327029 DOI: 10.1002/anie.202318632] [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: 12/05/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/09/2024]
Abstract
Liposomes serve as promising and versatile vehicles for drug delivery. Tracking these nanosized vesicles, particularly in vivo, is crucial for understanding their pharmacokinetics. This study introduces the design and synthesis of three new conjugated electrolyte (CE) molecules, which emit in the second near-infrared window (NIR-II), facilitating deeper tissue penetration. Additionally, these CEs, acting as biomimetics of lipid bilayers, demonstrate superior compatibility with lipid membranes compared to commonly used carbocyanine dyes like DiR. To counteract the aggregation-caused quenching effect, CEs employ a twisted backbone, as such their fluorescence intensities can effectively enhance after a fluorophore multimerization strategy. Notably, a "passive" method was employed to integrate CEs into liposomes during the liposome formation, and membrane incorporation efficiency was significantly promoted to nearly 100%. To validate the in vivo tracking capability, the CE-containing liposomes were functionalized with cyclic arginine-glycine-aspartic acid (cRGD) peptides, serving as tumor-targeting ligands. Clear fluorescent images visualizing tumor site in living mice were captured by collecting the NIR-II emission. Uniquely, these CEs exhibit additional emission peak in visible region, enabling in vitro subcellular analysis using routine confocal microscopy. These results underscore the potential of CEs as a new-generation of membrane-targeting probes to facilitate the liposome-based medicine research.
Collapse
Affiliation(s)
- Yingying Meng
- State Key Laboratory of Luminescent Materials and Devices, Institute of Polymer Optoelectronic Materials and Devices, School of Materials Science and Engineering, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, 510640, Guangzhou, P. R. China
| | - Ji Gao
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Peirong Zhou
- State Key Laboratory of Luminescent Materials and Devices, Institute of Polymer Optoelectronic Materials and Devices, School of Materials Science and Engineering, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, 510640, Guangzhou, P. R. China
| | - Xudong Qin
- State Key Laboratory of Luminescent Materials and Devices, Institute of Polymer Optoelectronic Materials and Devices, School of Materials Science and Engineering, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, 510640, Guangzhou, P. R. China
| | - Miao Tian
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 510640, Guangzhou, China
| | - Xiaohui Wang
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 510640, Guangzhou, China
| | - Cheng Zhou
- State Key Laboratory of Luminescent Materials and Devices, Institute of Polymer Optoelectronic Materials and Devices, School of Materials Science and Engineering, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, 510640, Guangzhou, P. R. China
| | - Kai Li
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Fei Huang
- State Key Laboratory of Luminescent Materials and Devices, Institute of Polymer Optoelectronic Materials and Devices, School of Materials Science and Engineering, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, 510640, Guangzhou, P. R. China
| | - Yong Cao
- State Key Laboratory of Luminescent Materials and Devices, Institute of Polymer Optoelectronic Materials and Devices, School of Materials Science and Engineering, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, 510640, Guangzhou, P. R. China
| |
Collapse
|
32
|
Zhang H, Wei S, Hu Y, Zhang Y, Yao H, Qi G, Adu-Frimpong M, Sun C. Influence of Different Ratios of DSPE-PEG2k on Ester Prodrug Self-Assembly Nanoparticles for Cell Migration and Proliferation Suppression. Int J Nanomedicine 2024; 19:2807-2821. [PMID: 38525014 PMCID: PMC10959298 DOI: 10.2147/ijn.s446741] [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: 11/21/2023] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
Background Bufalin (BFL, an active anti-tumor compound derived from toad venom) is limited in its application due to high toxicity and rapid metabolism of the cardiotonic steroid. Ester prodrug self-assembly nanoparticles have shown significant improved effects in addressing the above-mentioned issues. Methods An ester bond was formed between linoleic acid and bufalin to synthesize linoleic acid-bufalin prodrug (LeB). The self-assembly nanoparticles (LeB-PSNs) containing different mass ratios of DSPE-PEG2k and prodrug (6:4, 7:3, 8:2, 9:1 and 10:0) were prepared via co-precipitation method and defined as 6:4-PSNs, 7:3-PSNs, 8:2-PSNs, 9:1-PSNs and LeB-PSNs, respectively. Further, the characterization (particle size, zeta potential, surface morphology and stability) of the nanoparticles was carried out. Finally, we evaluated the impact of different ratios of DSPE-PEG2k on the hydrolysis rate, cytotoxicity, cellular uptake, cell migration and proliferation suppression potential of the prodrug nanoparticles. Results The linoleic acid-bufalin prodrug (LeB) was successfully synthesized. Upon the addition of DSPE-PEG2k at different weight ratios, both particle size and polydispersity index (PDI) significantly decreased, while the zeta potential increased remarkably. No significant differences in particle size, PDI and Zeta potential were observed among the 9:1, 8:2 and 7:3 PSNs. Notably, the 8:2 (w/w) DSPE-PEG2k nanoparticles exhibited superior stability, hydrolysis and cellular uptake rates, along with efficient cell cytotoxicity, cell migration and proliferation suppression. Conclusion These findings indicate that DSPE-PEG2k could improve the performance of BFL prodrug nanoparticles, namely enhancing stability and achieving adaptive drug release by modulating the hydrolysis rate of esterase. This study therefore provides more opportunities for the development of BFL application.
Collapse
Affiliation(s)
- Huiyun Zhang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, 224003, People’s Republic of China
| | - Shunru Wei
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, 224003, People’s Republic of China
| | - Yunfei Hu
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, 224003, People’s Republic of China
| | - Yu Zhang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, 224003, People’s Republic of China
| | - Hao Yao
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, 224003, People’s Republic of China
| | - Gang Qi
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, 224003, People’s Republic of China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, UK-0215-5321, Ghana
| | - Congyong Sun
- Department of Central Laboratory, The Affiliated Huaian No.1 People’s Hospital, Nanjing Medical University, Huai’an, Jiangsu, 223300, People’s Republic of China
| |
Collapse
|
33
|
Alshangiti DM, El-Damhougy TK, Zaher A, Madani M, Mohamady Ghobashy M. Revolutionizing biomedicine: advancements, applications, and prospects of nanocomposite macromolecular carbohydrate-based hydrogel biomaterials: a review. RSC Adv 2023; 13:35251-35291. [PMID: 38053691 PMCID: PMC10694639 DOI: 10.1039/d3ra07391b] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
Nanocomposite hydrogel biomaterials represent an exciting Frontier in biomedicine, offering solutions to longstanding challenges. These hydrogels are derived from various biopolymers, including fibrin, silk fibroin, collagen, keratin, gelatin, chitosan, hyaluronic acid, alginate, carrageenan, and cellulose. While these biopolymers possess inherent biocompatibility and renewability, they often suffer from poor mechanical properties and rapid degradation. Researchers have integrated biopolymers such as cellulose, starch, and chitosan into hydrogel matrices to overcome these limitations, resulting in nanocomposite hydrogels. These innovative materials exhibit enhanced mechanical strength, improved biocompatibility, and the ability to finely tune drug release profiles. The marriage of nanotechnology and hydrogel chemistry empowers precise control over these materials' physical and chemical properties, making them ideal for tissue engineering, drug delivery, wound healing, and biosensing applications. Recent advancements in the design, fabrication, and characterization of biopolymer-based nanocomposite hydrogels have showcased their potential to transform biomedicine. Researchers are employing strategic approaches for integrating biopolymer nanoparticles, exploring how nanoparticle properties impact hydrogel performance, and utilizing various characterization techniques to evaluate structure and functionality. Moreover, the diverse biomedical applications of these nanocomposite hydrogels hold promise for improving patient outcomes and addressing unmet clinical needs.
Collapse
Affiliation(s)
| | - Tasneam K El-Damhougy
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University P.O. Box: 11754, Yousef Abbas Str. Nasr City Cairo Egypt
| | - Ahmed Zaher
- Chemistry Department, Faculty of Science, El-Mansoura University Egypt
| | - Mohamed Madani
- College of Science and Humanities, Imam Abdulrahman Bin Faisal University Jubail Saudi Arabia
| | - Mohamed Mohamady Ghobashy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority P.O. Box 29 Nasr City Cairo Egypt
| |
Collapse
|