1
|
Li Y, Lu Y, Zhu X, Wu T, Ali K, Zhang H, Ma Z, Liu H, Wang H, Zhu H, Bi Q, Wang J, Tuo Z, Tang L. An albumin-prodrug injectable formulation for synergistic cancer immunotherapy. J Colloid Interface Sci 2025; 686:1019-1032. [PMID: 39929010 DOI: 10.1016/j.jcis.2025.02.012] [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: 11/03/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/12/2025]
Abstract
The emergence of immunotherapy has significantly transformed cancer therapy, and achieved promising outcomes. However, low patient response rates and the potential immune-related adverse events remain critical challenges, warranting extensive research. In this study, we developed an albumin-prodrug injection formulation (PIF) to enhance cancer immunotherapy. The stimulator of interferon genes (STING) agonist MSA-2 was esterified with the Indoleamine 2,3-dioxygenase (IDO) inhibitor NLG919 to synthesize the prodrug MSA-NLG. Subsequently, bovine serum albumin (BSA) was employed to prepare the anti-tumor injection formulation MSA-NLG@BSA, which effectively prolonged the circulation time of the prodrug, improved pharmacokinetic properties, and promoted tumor aggregation and penetration. This strategy ensures that the drug remains inactive in normal tissues, thereby reducing immune activation-induced damage. The prodrug was dissociated into MSA-2 and NLG919 within tumor cells overexpressing esterase. The MSA-2 molecule stimulates immune cells to secrete cytotoxic cytokines, triggering an anti-tumor immune response and reshaping immune cell population. Meanwhile, NLG919 regulates the recognition and killing of tumor cells by immune cells, effectively blocking the immunosuppression caused by IDO overexpression. This study highlights the potential of a tumor environment-responsive prodrug strategy to enhance the efficacy of immunotherapy, demonstrating the promising clinical translation of the novel MSA-NLG@BSA (PIF) formulation.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Yuting Lu
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Xiaoling Zhu
- Department of Colorectal Surgery, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Tao Wu
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kamran Ali
- Department of Colorectal Surgery, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Huamiao Zhang
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Zhangqiang Ma
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Huiping Liu
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Hanxi Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Heping Zhu
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Qiuchen Bi
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jianwei Wang
- Department of Colorectal Surgery, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China.
| | - Zhan Tuo
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Longguang Tang
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
2
|
Mani R, Babu SV, Murugesan N, Duraisamy R, Thayumanavan P. A Comparative Study of Quercetin/Rutin Loaded PEG Polymeric Nanoparticles: Controlled Drug Release and Its Biological Activity. J Biochem Mol Toxicol 2025; 39:e70269. [PMID: 40269608 DOI: 10.1002/jbt.70269] [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: 01/03/2025] [Revised: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Flavonoids are natural polyphenolic compounds that primarily possess antioxidant properties and play a significant role in opposing various diseases. Current chemotherapeutic approaches are largely ineffective, thus calling for the development of alternative strategies to combat this disease. In this regard, numerous studies have reported the anticancer effect of flavonoids in different types of cancer. To enhance its therapeutic value, polymeric nanoparticles (PEG NPs) represent an ideal delivery system. Further, surface modification of NPs with PEG holds tremendous potential for improving the bioavailability and circulation time of native drugs in the blood. The present study aimed to develop Quercetin/Rutin-loaded PEG polymeric NPs (Qu-PEG/Ru-PEG NPs) with enhanced encapsulation efficiency and sustained drug release. The synthesized Qu-PEG NPs & Ru-PEG NPs were characterized by UV-Vis Spectroscopy, FTIR spectrum, NMR, and XRD and SEM analysis. In-vitro drug release study exhibited a cumulative release of Quercetin & rutin for 24 h at pH 7.4. Further, the polymeric nano-formulations of Quercetin & Rutin showed enhanced antioxidant activity, leading to defense against oxidative stress. In-vitro cellular studies demonstrated that Qu-PEG NPs and Ru-PEG NPs significantly inhibit KB cell proliferation compared to free drugs alone. The current study also showed that Qu-PEG NPs & Ru-PEG NPs enhance intracellular ROS generation compared to the drug alone. Hence, our research findings revealed that successful encapsulation of Quercetin & Rutin in PEG NPs targets the tumor microenvironment and enhances the efficacy of drugs. Based on these preliminary results, flavonoid-loaded polymeric-based NPs might be potential therapeutic molecules against cancer in the future.
Collapse
Affiliation(s)
- Renuka Mani
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Swethaa Viswaresh Babu
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Nishanth Murugesan
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Ramachandhiran Duraisamy
- Department of Biotechnology and Biochemistry, Faculty of Science, Annamalai University, Chidambaram, Annamalai nagar, Tamilnadu, India
| | - Palvannan Thayumanavan
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| |
Collapse
|
3
|
Sahane P, Puri N, Khairnar P, Phatale V, Shukla S, Priyadarshinee A, Srivastava S. Harnessing Folate Receptors: A Comprehensive Review on the Applications of Folate-Adorned Nanocarriers for the Management of Melanoma. ACS APPLIED BIO MATERIALS 2025. [PMID: 40275606 DOI: 10.1021/acsabm.5c00077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
The advancement in exclusively tailored therapeutic delivery systems has escalated a great deal of interest in targeted delivery to augment therapeutic efficacy and to lessen adverse effects. The targeted delivery approach promisingly helps to surmount the unmet clinical needs of conventional therapies, including chemoresistance, limited penetration, and side effects. In the case of melanoma, various receptors were overexpressed on the tumor site, among which folate receptor (FR) targeting is considered to be a progressive approach for managing melanoma. FRs are the macromolecules of the glycosyl phosphatidylinositol-attached protein that possess globular assembly with a greater affinity toward specific ligands. So, the functional ligands can be utilized to design targeted nanocarriers (NCs) that can effectively bind to overexpressed FRs. Hence, folate-adorned NCs (FNCs) offer various benefits such as site-specific targeting, cargo protection, and minimizing toxicity. This review focuses on the insights and implications of FRs, targeting FRs, and mechanisms, challenges, and advantages of FNCs. Further, the applications of various FNCs, such as liposomes, polymeric NCs, albumin nanoparticles, inorganic NCs, liquid crystalline nanoparticles, and nanogels, have been elaborated for melanoma therapy. Likewise, the potential of FNCs in immunotherapy, photodynamic therapy, chemotherapy, gene therapy, photothermal therapy, and tumor imaging has been exhaustively discussed. Furthermore, translational hurdles and potential solutions are discussed in detail. The present review is expected to give thoughtful ideas to researchers, industry stakeholders, and formulation scientists for the efficacious development of FNCs.
Collapse
Affiliation(s)
- Prajakta Sahane
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Niharika Puri
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Abhipsa Priyadarshinee
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| |
Collapse
|
4
|
Swain A, Das Anthuparambil N, Begam N, Chandran S, Basu JK. Harnessing interfacial entropic effects in polymer grafted nanoparticle composites for tailoring their thermo-mechanical and separation properties. SOFT MATTER 2025. [PMID: 40266282 DOI: 10.1039/d4sm01549e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Nanocomposites based on polymeric materials have been extensively studied to understand and control the thermodynamics, flow, and mechanical properties of the underlying matrix as well to create new materials with diverse optical, electrical, magnetic, separation, catalytic, and biomedical properties. In the form of thin films or membranes, such materials can impart remarkable improvements in various properties of the underlying substrates. Using nanoparticles with grafted polymer chains usually overcomes a major hurdle in achieving enhancements in various properties by enabling better dispersion in the matrix while at the same time introducing a new parameter - interfacial entropy - leading to the emergence of new parameter space for tuning dispersion, flow and thermal properties. In this article, we highlight how this interfacial entropic effect can be harnessed to control various properties in thin films and membranes of grafted nanoparticle composites, in particular their thermo-mechanical properties, viscosity, fragility, glass transition temperature (Tg), and dynamic heterogeneity as well as their ability to act as highly selective gas separation and water desalination membranes. We discuss the application of a range of experimental techniques as well as molecular dynamics simulation to extract these properties and obtain microscopic insight into how the interplay of various surface and interfacial effects lies at the centre of these significant property improvements and enhanced functionality. Finally, we provide an outlook on future opportunities for designing sustainable PNCs, emphasizing their potential in environmental, energy, and biomedical applications, with advanced experiments and modelling driving further innovations.
Collapse
Affiliation(s)
- Aparna Swain
- Department of Physics, Indian Institute of Science Bangalore, 560012, India.
| | - Nimmi Das Anthuparambil
- Department of Physics, Universität Siegen, Walter-Flex-Str. 3, 57072 Siegen, Germany
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Nafisa Begam
- Department of Physics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Sivasurender Chandran
- Soft and Biological Matter Laboratory, Department of Physics, Indian Institute of Technology, Kanpur-208016, India.
| | - J K Basu
- Department of Physics, Indian Institute of Science Bangalore, 560012, India.
| |
Collapse
|
5
|
Xiong Y, Yang S, Di Y, Yang J, Tu J, Yan L. GSH-Triggered Nitric Oxide Release from Polyurethane Nanocarriers for Gas-Photothermal Synergistic Therapy of Bacterial-Infected Wounds. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40232873 DOI: 10.1021/acsami.5c03249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Wound bacterial infections pose a significant challenge to global public health. Nitric oxide (NO), an endogenous gaseous molecule, shows great potential in antibacterial therapy. However, efficient NO delivery remains a critical challenge. Photothermal therapy (PTT) offers a noninvasive and highly localized treatment for bacterial infections. Leveraging the advantages of both approaches, this study developed a polymer-based nanocarrier to achieve synergistic NO delivery and PTT. For the first time, an amphiphilic polyurethane PEG-PNU-PEG containing dinitrate NO donors were synthesized, which further self-assembled into polymeric nanomicelles loaded with a NIR emitting conjugated polymer. In the bacterial microenvironment, elevated glutathione (GSH) triggered NO release. Simultaneously, the conjugated polymer enabled effective photothermal therapy, further promoting NO release for gas therapy. Experimental results demonstrated that the synergistic NO and PTT treatment effectively eradicated bacteria, eliminating 69.93% ± 2.79% of S. aureus biofilm, a > 4.5-fold improvement compared to NO treatment alone (14.8% ± 2.92%). The in vitro antibacterial assay showed an inhibition rate exceeding 99.0%, while in vivo studies on infected skin wounds revealed a bacterial clearance rate of 95.0% ± 1.95%. Furthermore, this system suppressed inflammatory cytokines, promoted collagen deposition, and accelerated skin regeneration, providing an effective strategy for broad-spectrum antibacterial therapy and wound healing.
Collapse
Affiliation(s)
- Yuyue Xiong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
- Shenzhen Institute of Wuhan University of Technology, Shenzhen 518000, China
| | - Shuai Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Yaodong Di
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Jinglong Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Jing Tu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
- Shenzhen Institute of Wuhan University of Technology, Shenzhen 518000, China
| |
Collapse
|
6
|
Samari M, Kashanian S, Zinadini S, Derakhshankhah H. Enhanced delivery of azithromycin using asymmetric polyethersulfone membrane modified with KIT-6 mesoporous material: Optimization and mechanistic studies. Eur J Pharm Sci 2025; 207:107038. [PMID: 39933630 DOI: 10.1016/j.ejps.2025.107038] [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: 11/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
This study presents the development of a novel drug delivery system designed for improving the release profile and sustained delivery of azithromycin (AZI), particularly aimed at applications requiring localized infection control and improved tissue compatibility. The system employs an asymmetric polyethersulfone (PES) membrane modified with KIT-6 mesoporous material, offering improved drug release performance and biocompatibility over conventional delivery platforms. Membrane optimization was achieved by systematically varying parameters such as thickness (150-600 µm), drug concentration (500-1500 mg/L), polymer content (13-21 % PES), pore maker percentage (0-4 % polyvinylpyrrolidone), and KIT-6 modifier percentage (0.5-2 %). Characterization included scanning electron microscopy, water contact angle measurements, porosity, tensile strength evaluation, and comprehensive bioactivity testing (cytotoxicity, antimicrobial efficacy, blood compatibility, and a novel tissue integrity assay). The optimized formulation (17 % PES, 2 % PVP, 1 % KIT-6) achieved a controlled and sustained release profile with improved drug availability (464 mg/L) compared to unmodified membranes (252 mg/L), with a sustained release profile governed by the Higuchi model. Additionally, the membrane demonstrated superior biocompatibility (-90 % cell viability, low hemolysis at 1.2 %) and preserved tissue integrity better than unmodified counterparts, as evidenced by in vitro and ex vivo studies. Notably, the system showed robust reusability over prolonged use, indicating its potential as an effective, sustainable, and biocompatible solution for localized AZI delivery. These advantages position this system as a promising alternative for medical applications requiring precise drug release and minimal tissue disruption.
Collapse
Affiliation(s)
- Mahya Samari
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| | - Soheila Kashanian
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran; Nanobiotechnology Department, Faculty of Innovative Science and Technology, Razi University, Kermanshah, Iran.
| | - Sirus Zinadini
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran; Environmental Research Center (ERC), Razi University, Kermanshah, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
7
|
Tu L, Xing B, Ma S, Zou Z, Wang S, Feng J, Cheng M, Jin Y. A review on polysaccharide-based tumor targeted drug nanodelivery systems. Int J Biol Macromol 2025; 304:140820. [PMID: 39933669 DOI: 10.1016/j.ijbiomac.2025.140820] [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: 10/25/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The tumor-targeted drug delivery system (TTDNS) uses nanocarriers to transport chemotherapeutic agents to target tumor cells or tissues precisely. This innovative approach considerably increases the effective concentration of these drugs at the tumor site, thereby enhancing their therapeutic efficacy. Many chemotherapeutic agents face challenges, such as low bioavailability, high cytotoxicity, and inadequate drug resistance. To address these obstacles, TTDNS comprising natural polysaccharides have gained increasing popularity in the field of nanotechnology owing to their ability to improve safety, bioavailability, and biocompatibility while reducing toxicity. In addition, it enhances permeability and allows for controlled drug delivery and release. This review focuses on the sources of natural polysaccharides and their direct and indirect mechanisms of anti-tumor activity. We also explored the preparation of various polysaccharide-based nanocarriers, including nanoparticles, nanoemulsions, nanohydrogels, nanoliposomes, nanocapsules, nanomicelles, nanocrystals, and nanofibers. Furthermore, this review delves into the versatile applications of polysaccharide-based nanocarriers, elucidating their capabilities for in vivo targeting, controlled release, and responsiveness to endogenous and exogenous stimuli, such as pH, reactive oxygen species, glutathione, light, ultrasound, and magnetic fields. This sophisticated design substantially enhances the chemotherapeutic efficacy of the encapsulated drugs at tumor sites and provides a basis for preclinical and clinical research. However, the in vivo stability, drug loading, and permeability of these preparations into tumor tissues still need to be improved. Most of the currently developed biomarker-sensitive polysaccharide nanocarriers are still in the laboratory stage, more innovative delivery mechanisms and clinical studies are needed to develop commercial nanocarriers for medical use.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Shufei Ma
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; Guangxi University of Chinese Medicine, Nanning 530200, PR China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| |
Collapse
|
8
|
Vreeker E, Grünewald F, van der Heide NJ, Bonini A, Marrink SJ, Tych K(K, Maglia G. Nanopore-Functionalized Hybrid Lipid-Block Copolymer Membranes Allow Efficient Single-Molecule Sampling and Stable Sensing of Human Serum. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418462. [PMID: 40033964 PMCID: PMC12004896 DOI: 10.1002/adma.202418462] [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: 11/26/2024] [Revised: 02/10/2025] [Indexed: 03/05/2025]
Abstract
Biological nanopores are powerful tools for single-molecule detection, with promising potential as next-generation biosensors. A major bottleneck in nanopore analysis is the fragility of the supporting lipid membranes, that easily rupture after exposure to biological samples. Membranes comprising PMOXA-PDMS-PMOXA (poly(2-methyloxazoline-b-dimethylsiloxane-b-2-methyloxazoline)) or PBD-PEO (poly(1,2-butadiene)-b-poly(ethylene oxide)) polymers may form robust alternatives, but their suitability for the reconstitution of a broad range of nanopores has not yet been investigated. Here, PBD-PEO membranes are found to be highly robust toward applied voltages and human serum, while providing a poor environment for nanopore reconstitution. However, hybrid membranes containing a similar molar ratio of PBD11PEO8 polymers and diphytanoyl phosphatidylcholine (DPhPC) lipids show the best of both worlds: highly robust membranes suitable for the reconstitution of a wide variety of nanopores. Molecular dynamics simulations reveal that lipids form ≈12 nm domains interspersed by a polymer matrix. Nanopores partition into these lipid nanodomains and sequester lipids, possibly offering the same binding strength as in a native bilayer. Nanopores reconstituted in hybrid membranes yield efficient sampling of biomolecules and enable sensing of high concentrations of human serum. This work thus shows that hybrid membranes functionalized with nanopores allow single-molecule sensing, while forming robust interfaces, resolving an important bottleneck for novel nanopore-based biosensors.
Collapse
Affiliation(s)
- Edo Vreeker
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
| | - Fabian Grünewald
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
- Heidelberg Institute for Theoretical Studies (HITS)Schloss‐Wolfsbrunnenweg 3569118HeidelbergGermany
| | - Nieck Jordy van der Heide
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
| | - Andrea Bonini
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
| | - Siewert J. Marrink
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
| | - Katarzyna (Kasia) Tych
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
| | - Giovanni Maglia
- Groningen Biomolecular Sciences and Biotechnology InstituteUniversity of GroningenNijenborg 7Groningen9747 AGThe Netherlands
| |
Collapse
|
9
|
Du J, Wang H, Zhong L, Wei S, Min X, Deng H, Zhang X, Zhong M, Huang Y. Bioactivity and biomedical applications of pomegranate peel extract: a comprehensive review. Front Pharmacol 2025; 16:1569141. [PMID: 40206073 PMCID: PMC11979244 DOI: 10.3389/fphar.2025.1569141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Pomegranate peel is a by-product generated during the processing of pomegranate (Punica granatum L.) fruit, accounting for approximately 50% of the total mass of the fruit. Although pomegranate peel is usually regarded as waste, it is rich in various bioactive metabolites such as polyphenols, tannins, and flavonoids, demonstrating significant medicinal and nutritional value. In recent years, Pomegranate peel extract (PPE) has shown broad application prospects in the biomedical field due to its multiple effects, including antioxidant, anti-inflammatory, antibacterial, anti-apoptotic properties, and promotion of cell regeneration. This review consolidates the major bioactive metabolites of PPE and explores its applications in biomedical materials, including nanodrug carriers, hydrogels, and tissue engineering scaffolds. By synthesizing the existing literature, we delve into the potential value of PPE in biomedicine, the challenges currently encountered, and the future directions for research. The aim of this review is to provide a scientific basis for optimizing the utilization of PPE and to facilitate its broader application in the biomedical field.
Collapse
Affiliation(s)
- Jinsong Du
- School of Health Management, Zaozhuang University, Zaozhuang, China
- Department of Teaching and Research, Shandong Coal Health School, Zaozhuang, China
| | - Heming Wang
- School of Nursing, Jilin University, Jilin, China
| | - Lingyun Zhong
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Shujie Wei
- Image Center, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Xiaoqiang Min
- Department of Teaching and Research, Shandong Coal Health School, Zaozhuang, China
- Department of Geriatics, Shandong Healthcare Group Xinwen Central Hospital, Taian, China
| | - Hongyan Deng
- School of Health Management, Zaozhuang University, Zaozhuang, China
| | - Xiaoyan Zhang
- Magnetic Resonance Imaging Department, Shandong Healthcare Group Zaozhuang Central Hospital, Zaozhuang, China
| | - Ming Zhong
- Lanshu Cosmetics Co., Ltd., Huzhou, Zhejiang, China
| | - Yi Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
López-Laguna H, Favaro MTP, Chellou-Bakkali S, Voltà-Durán E, Parladé E, Sánchez J, Corchero JL, Unzueta U, Villaverde A, Vázquez E. Citrate-Assisted Regulation of Protein Stability and Secretability from Synthetic Amyloids. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14940-14951. [PMID: 40009529 DOI: 10.1021/acsami.4c20784] [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: 02/28/2025]
Abstract
The mammalian endocrine system uses functional amyloids as dynamic depots to store and release protein hormones into the bloodstream. Such depots, acting as secretory granules within the microscale, are formed in specialized cells by the coordination between the ionic, divalent form of zinc (Zn2+) and the imidazole ring from accessible His residues. The reversibility of such cross-linking events allows for the release of monomeric or oligomeric forms of the functional protein for biological activity. In vitro, and mimicking such a natural coordination process, synthetic amyloidal granules with secretory properties can be fabricated using selected therapeutic proteins as building blocks. Then, these microparticles act as delivery systems for endocrine-like, sustained protein release, with proven applicability in vaccinology, cancer therapy, regenerative medicine, and as antimicrobial agents. While the temporal profile in which the protein is leaked from the material might be highly relevant to clinically oriented applications, the fine control of such parameters remains unclear. We have explored here how the kinetics of protein release can be regulated by intervening in the storage formulation of the granules, through the concentration of citrate not only as a buffer component and protein stabilizer but also as a chelating agent. The citrate-assisted, time-prolonged regulatable release of proteins, in their functional form, opens a spectrum of possibilities to adjust the preparation of synthetic secretory granules to specific clinical needs.
Collapse
Affiliation(s)
- Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Marianna T P Favaro
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Sara Chellou-Bakkali
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Julieta Sánchez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departamento de Química, Cátedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, ICTA, Universidad Nacional de Córdoba, Av. Vélez Sársfield 1611, Córdoba 5016, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - José Luis Corchero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| |
Collapse
|
11
|
Ren L, Wang B, Miao D, Xiang P, Zeng Z, Li Z, Chen X, Xu C, Gong Q, Luo K, Jing J. Topology-Oriented Lymph Node Drainage of Dendritic Polymer-TLR Agonist Conjugates to Enhance Vaccine Immunogenicity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417704. [PMID: 39962825 DOI: 10.1002/adma.202417704] [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: 11/15/2024] [Revised: 01/10/2025] [Indexed: 03/17/2025]
Abstract
Strategically targeting lymph nodes (LNs) to orchestrate the initiation and regulation of adaptive immune responses is one of the most pressing challenges in the context of vaccination. Herein, a series of polymer-TLR agonist conjugates (PTACs) is developed to investigate the impact of dendritic-topological characteristics on their LN targeting activity in vivo, and their molecular weight (MW) on their pharmacokinetics in support of their LN homing. Notably, the dendritic 6-arm PTAC with a MW of 60 kDa (6A-PTAC-60k) rapidly delivered cargo to draining LNs after administration to peripheral tissues. Specifically, this topologic structure ameliorated the targeting behavior within lymphatic vessels and LNs, including an elevated amount of TLR7/8 agonist delivered to the LNs, an improved distribution pattern among barrier cells and immune cells, increased permeability, and prolonged retention. Furthermore, the 6A-PTAC-60k formulation induced broad antibody and T cell responses, enhancing vaccine immunogenicity and suppressing tumor growth. The results revealed that both the topology and MW of polymers are crucial factors for immunoadjuvant distribution and their functional activity in the draining LNs, which, in turn, enhanced the immunogenicity of the vaccine formulation. This study may provide a chemical and structural basis for optimizing the design of immunoadjuvant delivery systems.
Collapse
Affiliation(s)
- Long Ren
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bing Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Di Miao
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pan Xiang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhen Zeng
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Zhiqian Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoting Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Jing Jing
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, Sichuan Engineering Research Center for Intelligent Diagnosis and Treatment of Breast Diseases, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
12
|
Pan J, Wang Y, Chen Y, Zhang C, Deng H, Lu J, Chen W. Emerging strategies against accelerated blood clearance phenomenon of nanocarrier drug delivery systems. J Nanobiotechnology 2025; 23:138. [PMID: 40001108 PMCID: PMC11853785 DOI: 10.1186/s12951-025-03209-0] [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: 10/31/2024] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Nanocarrier drug delivery systems (NDDS) have gained momentum in the field of anticancer or nucleic acid drug delivery due to their capacity to aggrandize the targeting efficacy and therapeutic outcomes of encapsulated drugs. A disadvantage of NDDS is that repeated administrations often encounter an obstacle known as the "accelerated blood clearance (ABC) phenomenon". This phenomenon results in the rapid clearance of the secondary dose from the bloodstream and markedly augmented liver accumulation, which substantially undermines the accurate delivery of drugs and the therapeutic effect of NDDS. Nevertheless, the underlying mechanism of this phenomenon has not been elucidated and there is currently no effective method for its eradication. In light of the above, the aim of this review is to provide a comprehensive summary of the underlying mechanism and potential countermeasures of the ABC phenomenon, with a view to rejuvenating both the slow-release property and expectation of NDDS in the clinic. In this paper, we innovatively introduce the pharmacokinetic mechanism of ABC phenomenon to further elucidate its occurrence mechanism after discussing its immunological mechanism, which provides a new direction for expanding the mechanistic study of ABC phenomenon. Whereafter, we conducted a critical conclusion of potential strategies for the suppression or prevention of the ABC phenomenon in terms of the physical and structural properties, PEG-lipid derivatives, dosage regimen and encapsulated substances of nanoformulations, particularly covering some novel high-performance nanomaterials and mixed modification methods. Alternatively, we innovatively propose a promising strategy of applying the characteristics of ABC phenomenon, as the significantly elevated hepatic accumulation and activated CYP3A1 profile associated with the ABC phenomenon are proved to be conducive to enhancing the efficacy of NDDS in the treatment of hepatocellular carcinoma. Collectively, this review is instructive for surmounting or wielding the ABC phenomenon and advancing the clinical applications and translations of NDDS.
Collapse
Affiliation(s)
- Jianquan Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yanyan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yunna Chen
- Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Cheng Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Huiya Deng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jinyuan Lu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, China.
| |
Collapse
|
13
|
Tang Z, Ye F, Ni N, Fan X, Lu L, Gu P. Frontier applications of retinal nanomedicine: progress, challenges and perspectives. J Nanobiotechnology 2025; 23:143. [PMID: 40001147 PMCID: PMC11863789 DOI: 10.1186/s12951-025-03095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/04/2025] [Indexed: 02/27/2025] Open
Abstract
The human retina is a fragile and sophisticated light-sensitive tissue in the central nervous system. Unhealthy retinas can cause irreversible visual deterioration and permanent vision loss. Effective therapeutic strategies are restricted to the treatment or reversal of these conditions. In recent years, nanoscience and nanotechnology have revolutionized targeted management of retinal diseases. Pharmaceuticals, theranostics, regenerative medicine, gene therapy, and retinal prostheses are indispensable for retinal interventions and have been significantly advanced by nanomedical innovations. Hence, this review presents novel insights into the use of versatile nanomaterial-based nanocomposites for frontier retinal applications, including non-invasive drug delivery, theranostic contrast agents, therapeutic nanoagents, gene therapy, stem cell-based therapy, retinal optogenetics and retinal prostheses, which have mainly been reported within the last 5 years. Furthermore, recent progress, potential challenges, and future perspectives in this field are highlighted and discussed in detail, which may shed light on future clinical translations and ultimately, benefit patients with retinal disorders.
Collapse
Affiliation(s)
- Zhimin Tang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Fuxiang Ye
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Ni Ni
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Linna Lu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
14
|
Cui Z, Brna EA, Crawford MA, Treerat P, Alimadad M, Hughes MA, Letteri RA. Presenting Antimicrobial Peptides on Poly(ethylene glycol): Star-Shaped vs Comb-Like Architectures. Macromolecules 2025; 58:2073-2084. [PMID: 40026451 PMCID: PMC11867009 DOI: 10.1021/acs.macromol.4c02762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/16/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
Conjugating antimicrobial peptides (AMPs) to nonlinear polymers is a promising strategy to overcome the translational challenges of AMPs toward treating infections caused by antibiotic-resistant bacteria. Nonlinear polymers, and therefore conjugates, can be prepared with various architectures (e.g., star-shaped, comb-like, hyperbranched, etc.), however, the effects of polymer architecture on antimicrobial performance and related properties, like size and morphology in solution and secondary structure, are not yet well-understood. Here, we compare conjugates of the human chemokine-derived AMP stapled P9 with poly(ethylene glycol) (PEG) prepared in two of the major nonlinear architectures: star-shaped and comb-like. At comparable molecular weights and compositions (peptide wt %), comb-like conjugates afford increased helicity, solubility, antimicrobial activity, and proteolytic stability compared to star-shaped analogs. We then leveraged the expansive design space of comb-like architectures to prepare conjugates with different backbone lengths and PEG side chain lengths, with shorter PEG side chains leading to increased helicity, yet potentially less shielding from proteolytic degradation and the longest backbone lengths furnishing the most potent antimicrobial activity. Both comb-like and star-shaped conjugates display high zeta potential, indicating that the cationic AMPs were accessible for electrostatic interactions with bacterial membranes. Yet, the comb-like conjugates showed a higher fraction of unimolecular structures indicative of a lower propensity for supramolecular assembly that could be encumbering the desired AMP-bacteria interactions in the star-shaped conjugates. Together, our work shows comb-like AMP-polymer conjugates to outperform analogous star-shaped conjugates, while adding design flexibility to access an expansive range of monomer chemistries, monomer distributions, and backbone lengths to modulate performance-determining properties and ultimately furnish an effective suite of AMP-polymer materials as alternatives to conventional antibiotics for combatting bacterial infections.
Collapse
Affiliation(s)
- Zixian Cui
- Department
of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Elliot A. Brna
- Department
of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Matthew A. Crawford
- Division
of Infectious Diseases & International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Puthayalai Treerat
- Division
of Infectious Diseases & International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Mobina Alimadad
- Department
of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Molly A. Hughes
- Division
of Infectious Diseases & International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Rachel A. Letteri
- Department
of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| |
Collapse
|
15
|
Pradhan A, Biswal S, Bhal S, Biswal BK, Kundu CN, Subuddhi U, Pati A, Hassan PA, Patel S. Amphiphilic Poly(ethylene glycol)-Cholesterol Conjugate: Stable Micellar Formulation for Efficient Loading and Effective Intracellular Delivery of Curcumin. ACS APPLIED BIO MATERIALS 2025; 8:1418-1436. [PMID: 39907519 DOI: 10.1021/acsabm.4c01657] [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: 02/06/2025]
Abstract
A biodegradable and biocompatible micellar-based drug delivery system was developed using amphiphilic methoxy-poly(ethylene glycol)-cholesterol (C1) and poly(ethylene glycol)-S-S-cholesterol (C2) conjugates and applied to the tumoral release of the water-insoluble drug curcumin. These synthesized surfactants C1 and C2 were found to form stable micelles (CMC ∼ 6 μM) and an average hydrodynamic size of around 20-25 nm. The curcumin-encapsulated C1 micelle was formulated by a solvent evaporation method. A very high drug encapsulation efficiency (EE) of ∼88% and a drug loading (DL) capacity of ∼9% were determined for both the micelles. From the reduced rate of curcumin degradation and differential scanning calorimetry (DSC) analysis, the stability of the curcumin-loaded C1 micelle was found to be higher than that of the unloaded micelle, which confirmed a more compact structural arrangement in the presence of hydrophobic curcumin. A pH-sensitive release of curcumin (faster release with decrease in pH) was observed for the curcumin-loaded C1 micelle, attributed to the diffusion and relaxation/erosion of micellar aggregates. To achieve reduction environment-sensitive drug release, a disulfide (S-S) chemical linkage-incorporated mPEG-cholesterol conjugate (C2) was synthesized, which was found to show glutathione-responsive faster release of curcumin. The in vitro experiments carried out in SCC9 oral cancer cell lines showed that the blank C1 and C2 micelles were noncytotoxic at lower concentrations (<50 μM), while curcumin-loaded C1 and C2 micelles inhibited the proliferation and promoted the apoptosis. An increased in vitro cytotoxicity was observed for curcumin-loaded micelles compared to that of curcumin itself, demonstrating a better cell penetration efficacy of the micelle. These results were further supplemented by the in vivo anticancer analysis of the curcumin-loaded C1 and C2 micellar formulations using the mice xenograft model. Notably, curcumin-loaded C2 micelles showed a significantly stronger apoptotic effect in xenograft mice compared to curcumin-loaded C1 micelles, indicating the GSH environment-sensitive drug release and improved bioavailability. In conclusion, the mPEG-cholesterol C1 and C2 micellar system with the advantages of small size, high encapsulation efficiency, high drug loading, simple preparing technique, biocompatibility, and good in vitro and in vivo performance may have the potential to be used as a drug carrier for sustained and stimuli-responsive release of the hydrophobic drug curcumin.
Collapse
Affiliation(s)
- Aiswarya Pradhan
- Department of Chemistry, National Institute of Technology, Rourkela 769 008, India
| | - Stuti Biswal
- Department of Life Sciences, National Institute of Technology, Rourkela 769 008, India
| | - Subhasmita Bhal
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, India
| | - Bijesh K Biswal
- Department of Life Sciences, National Institute of Technology, Rourkela 769 008, India
| | - Chanakya Nath Kundu
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, India
| | - Usharani Subuddhi
- Department of Chemistry, National Institute of Technology, Rourkela 769 008, India
| | - Anita Pati
- School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, India
| | - P A Hassan
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Sabita Patel
- Department of Chemistry, National Institute of Technology, Rourkela 769 008, India
| |
Collapse
|
16
|
Morsy HM, Zaky MY, Yassin NYS, Khalifa AYZ. Nanoparticle-based flavonoid therapeutics: Pioneering biomedical applications in antioxidants, cancer treatment, cardiovascular health, neuroprotection, and cosmeceuticals. Int J Pharm 2025; 670:125135. [PMID: 39732216 DOI: 10.1016/j.ijpharm.2024.125135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Flavonoids, a type of natural polyphenolic molecule, have garnered significant research interest due to their ubiquitous nature and diverse biological activities, including antioxidant, anti-inflammatory, and anticancer effects, making them appealing to various scientific disciplines. In this regard, the use of a flavonoid nanoparticle delivery system is to overcome low bioavailability, bioactivity, poor aqueous solubility, systemic absorption, and intensive metabolism. Therefore, this review summarizes the classification of nanoparticles (liposomes, polymeric, and solid lipid nanoparticles) and the advantages of using nanoparticle-flavonoid formulations to boost flavonoid bioavailability. Moreover, this review illustrated the pioneering biomedical applications of nanoparticle-based flavonoid therapeutics, as well as safety and toxicity considerations of using a flavonoid nanoparticle delivery system.
Collapse
Affiliation(s)
- Hadeer M Morsy
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Mohamed Y Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt.
| | - Nour Y S Yassin
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Ashraf Y Z Khalifa
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
17
|
Sriram A, Ithape H, Singh PK. Deep-insights: Nanoengineered gel-based localized drug delivery for arthritis management. Asian J Pharm Sci 2025; 20:101012. [PMID: 39995751 PMCID: PMC11848107 DOI: 10.1016/j.ajps.2024.101012] [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: 07/17/2023] [Revised: 05/15/2024] [Accepted: 07/03/2024] [Indexed: 02/26/2025] Open
Abstract
Arthritis is an inflammatory joint disorder that progressively impairs function and diminishes quality of life. Conventional therapies often prove ineffective, as oral administration lacks specificity, resulting in off-target side effects like hepatotoxicity and GIT-related issues. Intravenous administration causes systemic side effects. The characteristic joint-localized symptoms such as pain, stiffness, and inflammation make the localized drug delivery suitable for managing arthritis. Topical/transdermal/intra-articular routes have become viable options for drug delivery in treating arthritis. However, challenges with those localized drug delivery routes include skin barrier and cartilage impermeability. Additionally, conventional intra-articular drug delivery also leads to rapid clearance of drugs from the synovial joint tissue. To circumvent these limitations, researchers have developed nanocarriers that enhance drug permeability through skin and cartilage, influencing localized action. Gel-based nanoengineered therapy employs a gel matrix to incorporate the drug-encapsulated nanocarriers. This approach combines the benefits of gels and nanocarriers to enhance therapeutic effects and improve patient compliance. This review emphasizes deep insights into drug delivery using diverse gel-based novel nanocarriers, exploring their various applications embedded in hyaluronic acid (biopolymer)-based gels, carbopol-based gels, and others. Furthermore, this review discusses the influence of nanocarrier pharmacokinetics on the localization and therapeutic manipulation of macrophages mediated by nanocarriers. The ELVIS (extravasation through leaky vasculature and inflammatory cell-mediated sequestration) effect associated with arthritis is advantageous in drug delivery. Simply put, the ELVIS effect refers to the extravasation of nanocarriers through leaky vasculatures, which finally results in the accumulation of nanocarriers in the joint cavity.
Collapse
Affiliation(s)
| | | | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Telangana 500037, India
| |
Collapse
|
18
|
Austria E, Bilek M, Varamini P, Akhavan B. Breaking biological barriers: Engineering polymeric nanoparticles for cancer therapy. NANO TODAY 2025; 60:102552. [DOI: 10.1016/j.nantod.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
19
|
Vogelaar TD, Torjusen H, Lund R. Size-controlled antimicrobial peptide drug delivery vehicles through complex coacervation. SOFT MATTER 2025; 21:903-913. [PMID: 39801473 DOI: 10.1039/d4sm01157k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Due to the escalating threat of the pathogens' capability of quick adaptation to antibiotics, finding new alternatives is crucial. Although antimicrobial peptides (AMPs) are highly potent and effective, their therapeutic use is limited' as they are prone to enzymatic degradation, are cytotoxic and have low retention. To overcome these challenges, we investigate the complexation of the cationic AMP colistin with diblock copolymers poly(ethylene oxide)-b-poly(methacrylic acid) (PEO-b-PMAA) forming colistin-complex coacervate core micelles (colistin-C3Ms). We present long-term stable kinetically controlled colistin-C3Ms that can be prepared from several block lengths of PEO-b-PMAA polymers, where the polymerisation degree governs the overall micellar size. To achieve precise control over size and polydispersity, which are crucial for drug delivery applications, we investigate the hybridisation of PEO-b-PMAA polymers with varying chain lengths or PMAA homopolymers in ternary complex coacervation systems with colistin. This results in size-tunable colistin-C3Ms, ranging, depending on the mixing ratios, from micellar sizes of 26 nm to 100 nm. With size tunability at rather narrow size distributions and high stability, ternary colistin-C3Ms offer potential advancements in C3M drug delivery, paving the way for more effective and targeted treatments for bacterial infections in precision medicine.
Collapse
Affiliation(s)
- Thomas Daniel Vogelaar
- Department of Chemistry, University of Oslo, P.O. Box 1033 Blindern, NO-0315 Oslo, Norway.
| | - Henrik Torjusen
- Department of Chemistry, University of Oslo, P.O. Box 1033 Blindern, NO-0315 Oslo, Norway.
| | - Reidar Lund
- Department of Chemistry, University of Oslo, P.O. Box 1033 Blindern, NO-0315 Oslo, Norway.
- Hylleraas Centre for Quantum Molecular Sciences, University of Oslo, NO-0315 Oslo, Norway
| |
Collapse
|
20
|
Zhang D, Meng Y, Hao M, Xia Y. Nanocarriers Made of Natural Fatty Acids: Modulation of Their Release Profiles through Photo-Crosslinking. Angew Chem Int Ed Engl 2025; 64:e202415671. [PMID: 39609104 PMCID: PMC11735881 DOI: 10.1002/anie.202415671] [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: 08/16/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024]
Abstract
Natural fatty acids are attractive carrier materials for drug delivery, but their rapid dissolution and degradation in vivo calls for new strategies to enhance their stability. Here we report a simple and versatile method capable of photo-crosslinking carriers made of natural fatty acids for drug delivery under controlled release. By optimizing the crosslinking density, the nanoscale carriers show a high drug loading efficiency, together with a stable network structure for minimal degradation in a body fluid mimic. Fluorescence microscopy analysis also reveals the exceptional intracellular stability of the crosslinked network, resulting in negligible cytotoxicity toward A549 cells up to 24 h when loaded with a potent anticancer drug. We further extend this strategy to microscale carriers fabricated using electrospray. Upon photo-crosslinking, the carriers show a retarded release of nerve growth factor, resulting in slower neurite outgrowth from dorsal root ganglion. This work holds promise for addressing the efficacy and safety issues critical to nanomedicine and related applications.
Collapse
Affiliation(s)
- Dong Zhang
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA 30332USA
| | - Yuxuan Meng
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA 30332USA
| | - Min Hao
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA 30332USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA 30332USA
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA 30332USA
| |
Collapse
|
21
|
Chen J, Dong S. Polymer-based antimicrobial strategies for periodontitis. Front Pharmacol 2025; 15:1533964. [PMID: 39834832 PMCID: PMC11743519 DOI: 10.3389/fphar.2024.1533964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Periodontitis is a chronic inflammatory condition driven by plaque-associated microorganisms, where uncontrolled bacterial invasion and proliferation impair host immune responses, leading to localized periodontal tissue inflammation and bone destruction. Conventional periodontal therapies face challenges, including incomplete microbial clearance and the rise of antibiotic resistance, limiting their precision and effectiveness in managing periodontitis. Recently, nanotherapies based on polymeric materials have introduced advanced approaches to periodontal antimicrobial therapy through diverse antimicrobial mechanisms. This review explored specific mechanisms, emphasizing the design of polymer-based agents that employ individual or synergistic antimicrobial actions, and evaluated the innovations and limitations of current strategies while forecasting future trends in antimicrobial development for periodontitis.
Collapse
Affiliation(s)
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
22
|
Uriostegui-Pena AG, Torres-Copado A, Ochoa-Sanchez A, Luna-Bárcenas G, Sahare P, Paul S. Nanoformulated phytochemicals in skin anti-aging research: an updated mini review. 3 Biotech 2025; 15:31. [PMID: 39760004 PMCID: PMC11699038 DOI: 10.1007/s13205-024-04197-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025] Open
Abstract
Skin aging is characterized by progressive loss of functionality and regenerative potential of the skin, resulting in the appearance of wrinkles, irregular pigmentation, a decrease of elasticity, dryness, and rough texture. Damage to the skin caused by oxidative stress could substantially be slowed down by the use of phytochemicals that function as natural antioxidants. Although phytochemicals have immense potential as anti-aging medicines, their effectiveness as therapeutic agents is restricted by their poor solubility, biodistribution, stability, and hydrophilicity. Given their improved stability, solubility, efficacy, and occlusive properties, nanoformulations have emerged as promising drug delivery platforms for phytochemicals to achieve anti-aging effects. The efficacy of these nanoformulated phytochemicals in suppressing enzymes that accelerate skin aging, such as collagenase, tyrosinase and hyaluronidase, as well as enhancing superoxide dismutase, catalase, and collagen levels to improve skin appearance during aging has been demonstrated.
Collapse
Affiliation(s)
- Andrea G. Uriostegui-Pena
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Andrea Torres-Copado
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Adriana Ochoa-Sanchez
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Gabriel Luna-Bárcenas
- Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Padmavati Sahare
- Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| |
Collapse
|
23
|
Liu F, Wang X, Ren M, He P, Li Y, Cui J, Yang S. A shielded cascade of targeted nanocarriers spanning multiple microenvironmental barriers for inflammatory disease therapy. J Nanobiotechnology 2024; 22:789. [PMID: 39710698 DOI: 10.1186/s12951-024-03075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND The multi-biological barriers present in the inflammatory microenvironment severely limit the targeted aggregation of anti-inflammatory drugs in the lesion area. However, conventional responsive drug carriers inevitably come into contact with several pro-responsive stimulatory mediators simultaneously, leading to premature drug release and loss of most therapeutic effects. Breaking through the multi-level barriers of the inflammatory microenvironment is essential to improve the enrichment and bioavailability of drugs. RESULTS In this study, we propose a novel two-stage structural strategy to build shielded cascades of targeted nanocarriers (FA-PTP@Que) through inflammatory mediators, using cascade structures to cross multiple environmental barriers. The cascade structure of FA-PTP@Que is responsive to inflammatory mediators and exhibits ideal pathological microenvironmental response and drug release properties. FA-PTP@Que has shown good macrophage regulation and anti-inflammatory effects by efficiently targeting macrophages, scavenging intracellular reactive oxygen species (ROS), and down-regulating the secretion of pro-inflammatory factors. Significantly, in mice with arthritis and colitis, FA-PTP@Que enriches and targets macrophages at the sites of arthritis and colitis, showing significant anti-inflammatory effects. CONCLUSION FA-PTP@Que combines active chemotaxis of nanocarriers to inflammatory tissues and active targeting of effector cells, acting precisely at each barrier level in different microenvironments by responding to inflammatory mediators and overcoming the multiple barriers in the inflammatory microenvironment. This innovative strategy can effectively break through various inflammatory microenvironments and has the potential application to other inflammatory diseases.
Collapse
Affiliation(s)
- Fengyi Liu
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xu Wang
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
| | - Mingxing Ren
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
| | - Ping He
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuzhou Li
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jing Cui
- Chongqing Medical University, Chongqing, China
| | - Sheng Yang
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China.
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
24
|
Lorenzo-Anota HY, Gómez-Cantú JM, Vázquez-Garza E, Bernal-Ramirez J, Chapoy-Villanueva H, Mayolo-Deloisa K, Benavides J, Rito-Palomares M, Lozano O. Disulfiram-Loaded Nanoparticles Inhibit Long-Term Proliferation on Preadipocytes. Int J Nanomedicine 2024; 19:13301-13318. [PMID: 39679252 PMCID: PMC11645963 DOI: 10.2147/ijn.s467909] [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: 03/09/2024] [Accepted: 11/03/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Disulfiram (DSF) reduces insulin resistance and weight gain in obese mice. However, the effect on adipose tissue is unexplored due to their high instability under physiological conditions, limiting clinical applications. Thus, it is meaningful to develop a DSF carrier for sustained release to adipose tissue. We optimized the synthesis of poly-ε-caprolactone (PCL) nanoparticles (NPs) loaded with DSF and analyzed their effect on adipose tissue cells in vitro. Methods The NPs were synthesized by nanoprecipitation method, varying its solvent, either acetone or acetone/dichloromethane (60:40) (v/v), and ratio PCL:DSF (w/w) 1:2, 1:1, 2:1 and, 1:0; finding the best condition was obtained with acetone/dichloromethane solvent mixture and 2:1 PCL:DSF. Then, NPs toxicity was analyzed on adipose cells (preadipocytes, white-like adipocytes, and macrophages) assessing association and internalization, cell viability, and cell death mechanism. Results NPs were spherical with a particle size distribution of 203.2 ± 29.33 nm, a ζ-potential of -20.7 ± 4.58 mV, a PDI of 0.296 ± 0.084, and a physical drug loading of 18.6 ± 5.80%. Sustained release was observed from 0.5 h (10.94 ± 2.38%) up to 96 h (91.20 ± 6.03%) under physiological conditions. NPs internalize into macrophages, white-like adipocytes and preadipocytes without modifying cell viability on white-like adipocytes and macrophages. Preadipocytes reduce cell viability, inducing mitochondrial damage, increased mitochondrial reactive oxygen species production and loss of mitochondrial membrane potential, leading to effector caspases 3/7 cleaved, resulting in apoptosis. Finally, long-term proliferation inhibition was observed, highlighting the bioequivalent effect of PCL-DSF NPs compared to free DSF. Conclusion Our data demonstrated the biological interaction of PCL NPs with adipose cells in vitro. The selective cytotoxicity of DSF towards preadipocytes resulted in milder effects when it was delivered nanoencapsulated compared to the free drug. These results suggest promising pharmacological alternatives for DSF long-term delivery on adipose tissue.
Collapse
Affiliation(s)
- Helen Yarimet Lorenzo-Anota
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | | | | | | | - Héctor Chapoy-Villanueva
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | - Karla Mayolo-Deloisa
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, México
| | - Jorge Benavides
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, México
| | - Marco Rito-Palomares
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | - Omar Lozano
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| |
Collapse
|
25
|
Nair ST, Abhi C, Kamalasanan K, Pavithran K, Unni AR, Sithara MS, Sarma M, Mangalanandan TS. Pathophysiology-Driven Approaches for Overcoming Nanomedicine Resistance in Pancreatic Cancer. Mol Pharm 2024; 21:5960-5988. [PMID: 39561094 DOI: 10.1021/acs.molpharmaceut.4c00801] [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: 11/21/2024]
Abstract
Tumor heterogeneity poses a significant challenge in cancer therapy. To address this, we analyze pharmacotherapeutic challenges by categorizing them into static and dynamic barriers, reframing these challenges to improve drug delivery, efficacy, and the development of controlled-release nanomedicines (CRNMs). This pathophysiology-driven approach facilitates the design of novel therapeutics tailored to overcome obstacles in pancreatic ductal adenocarcinoma (PDAC) using nanotechnology. Advanced biomaterials in nanodrug delivery systems offer innovative solutions by combining controlled release, stimuli sensitivity, and smart design strategies. CRNMs are engineered to modulate spatiotemporal signaling and control drug release in PDAC, where resistance to conventional therapies is particularly high. This review explores pharmacokinetic considerations for nanomedicine design, RNA interference (RNAi) for stromal modulation, and the development of targeted nanomedicine strategies. Additionally, we highlight the limitations of current animal models in capturing the complexities of PDAC and discuss notable clinical failures, such as PEGylated hyaluronidase (Phase III HALO 109-301 trial) and evofosfamide (TH-302) with gemcitabine (MAESTRO trial), underscoring the need for improved models and treatment strategies. By targeting pathways like Notch and Hedgehog and incorporating stimuli-sensitive and pathway-modulating agents, CRNMs offer a promising avenue to enhance drug penetration and efficacy, reshaping the paradigm of pancreatic cancer treatment.
Collapse
Affiliation(s)
- Sreejith Thrivikraman Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - C Abhi
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Kaladhar Kamalasanan
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - K Pavithran
- Department of Medical Oncology and Hematology, School of Medicine, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Ashok R Unni
- Department of Veterinary Medicine, Central Animal Facility, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - M S Sithara
- Department of Veterinary Medicine, Central Animal Facility, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Manjit Sarma
- Department of Nuclear Medicine, Amrita School of Medicine, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - T S Mangalanandan
- Department of Endocrinology, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| |
Collapse
|
26
|
Agrohia DK, Goswami R, Çiçek YA, Tabaru NE, Brady TW, Rotello VM, Vachet RW. Multiplexed Quantitative Screening of the Cellular Uptake of Proteins Delivered by Polymeric Nanocarriers. Anal Chem 2024. [PMID: 39555943 DOI: 10.1021/acs.analchem.4c05191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Proteins are important therapeutic agents, yet better methods are needed to deliver them inside of cells. Polymeric nanocarriers (PNCs) are versatile materials for this purpose, and to enhance their development, it is necessary to quantify protein delivery efficiency into cells by numerous PNC designs at the same time. Current strategies for screening PNC systems are qualitative and mostly serial. Here, we describe a multiplexed approach that uses metal-coded mass tags (MMTs) to quantify the delivery of protein into cells by several different PNC designs simultaneously. Our approach will facilitate the development of more potent delivery systems by improving precision and reducing costs, effort, and time.
Collapse
Affiliation(s)
- Dheeraj K Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Yağız Anıl Çiçek
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Neslihan E Tabaru
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Trisha W Brady
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery - Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery - Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
27
|
Sheng Y, Zheng X, Li L, He H, Wu W, Lu Y. Ionic co-aggregates based intravenous drug delivery: Evaluation on kinetics and distribution of the drug payloads and nanocarriers. Int J Pharm 2024; 665:124657. [PMID: 39226987 DOI: 10.1016/j.ijpharm.2024.124657] [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: 06/05/2024] [Revised: 08/12/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
Surfactants are crucial in formulating poorly soluble drugs but lead to serious side effects due to PEG chains. Novel supra-amphiphiles consisting of fatty acids and choline are developed, which spontaneously form ionic co-aggregates (ICAs) in water and exhibit strong solubilizing capacity. Paclitaxel (PTX) is adopted as a model drug here to evaluate the feasibility of choline oleate-based ICAs in the intravenous delivery of poorly soluble drugs by comparing the kinetics and distribution of payloads and nanocarriers. Choline oleate presents a maximum 10-fold enhancement in solubilizing capacity to PTX than Cremophor EL (CreEL), enabling a one-tenth use level in the formulation. Aggregation-caused quenching probes are utilized to evaluate the kinetics and biodistribution of ICAs or CreEL-based micelles (MCs). A huge gap is found between the pharmacokinetic and particokinetic curves of either nanocarrier, indicating fast leakage. ICAs lead to faster PTX leakage in blood circulation but higher PTX distribution to organs than MCs. MCs present a longer circulation in blood but a slower distribution to organs than ICAs. ICAs do not arise adverse reactions in rats following repeated injections, while MCs cause pathological changes in varying degrees. In conclusion, choline oleate-based ICAs provide an alternative to surfactants in formulating poorly soluble drugs.
Collapse
Affiliation(s)
- Yuze Sheng
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xianzi Zheng
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lu Li
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200433, China; Fudan Zhangjiang Institute, Shanghai 201203, China.
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200433, China; Fudan Zhangjiang Institute, Shanghai 201203, China.
| |
Collapse
|
28
|
Wang L, Tkhilaishvili T, Jiang Z, Pirlar RF, Ning Y, Millán Laleona A, Wang J, Tang J, Wang Q, Trampuz A, Gonzalez Moreno M, Zhang X. Phage-liposome nanoconjugates for orthopedic biofilm eradication. J Control Release 2024; 376:949-960. [PMID: 39384150 DOI: 10.1016/j.jconrel.2024.09.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/18/2024] [Accepted: 09/29/2024] [Indexed: 10/11/2024]
Abstract
Infection by multidrug-resistant (MDR) bacteria has become one of the biggest threats to public health worldwide. One reason for the difficulty in treatment is the lack of proper delivery strategies into MDR bacterial biofilms, where the thick extracellular polymeric substance (EPS) layer impedes the penetration of antibiotics and nanoparticles. Here, we propose a novel bioactive nanoconjugate of drug-loaded liposomes and bacteriophages for targeted eradication of the MDR biofilms in orthopedic infections. Phage Sb-1, which has the ability to degrade EPS, was conjugated with antibiotic-loaded liposomes. Upon encountering the biofilm, phage Sb-1 degrades the EPS structure, thereby increasing the sensitivity of bacteria to antibiotics and allowing the antibiotics to penetrate deeply into the biofilm. As a result, effective removal of MDR bacterial biofilm was achieved with low dose of antibiotics, which was proved in this study by both in vitro and in vivo investigations. Notably, in the rat prosthetic joint infection (PJI) model, we found that the liposome-phage nanoconjugates could effectively decrease the bacterial load in the infected area and significantly promote osteomyelitis recovery. It is therefore believed that the conjugation of bacteriophage and liposomes could open new possibilities for the treatment of orthopedic infections, possibly other infections in the deep tissues.
Collapse
Affiliation(s)
- Lei Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233 Shanghai, China
| | - Tamta Tkhilaishvili
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), 13353 Berlin, Germany
| | - Zheng Jiang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233 Shanghai, China
| | - Rima Fanaei Pirlar
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), 13353 Berlin, Germany
| | - Yu Ning
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), 13353 Berlin, Germany
| | - Adrián Millán Laleona
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Zaragoza, Spain
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233 Shanghai, China
| | - Jin Tang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233 Shanghai, China
| | - Qiaojie Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233 Shanghai, China
| | - Andrej Trampuz
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), 13353 Berlin, Germany.
| | - Mercedes Gonzalez Moreno
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), 13353 Berlin, Germany.
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233 Shanghai, China.
| |
Collapse
|
29
|
Ye H, Franco C, Aboouf MA, Thiersch M, Sevim S, Llacer‐Wintle J, Veciana A, Llauradó‐Capdevila G, Wang K, Chen X, Tang Q, Matheu R, Wendel‐Garcia PD, Sánchez‐Murcia PA, Nelson BJ, Luo C, Puigmartí‐Luis J, Pané S. Insights into the Biological Activity and Bio-Interaction Properties of Nanoscale Imine-Based 2D and 3D Covalent Organic Frameworks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407391. [PMID: 39387248 PMCID: PMC11600295 DOI: 10.1002/advs.202407391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/16/2024] [Indexed: 10/15/2024]
Abstract
Covalent Organic Frameworks (COFs) emerged as versatile materials with promising potential in biomedicine. Their customizable functionalities and tunable pore structures make them valuable for various biomedical applications such as biosensing, bioimaging, antimicrobial activity, and targeted drug delivery. Despite efforts made to create nanoscale COFs (nCOFs) to enhance their interaction with biological systems, a comprehensive understanding of their inherent biological activities remains a significant challenge. In this study, a thorough investigation is conducted into the biocompatibility and anti-neoplastic properties of two distinct imine-based nCOFs. The approach involved an in-depth analysis of these nCOFs through in vitro experiments with various cell types and in vivo assessments using murine models. These findings revealed significant cytotoxic effects on tumor cells. Moreover, the activation of multiple cellular death pathways, including apoptosis, necroptosis, and ferroptosis is determined, supported by evidence at the molecular level. In vivo evaluations exhibited marked inhibition of tumor growth, associated with the elevated spontaneous accumulation of nCOFs in tumor tissues and the modulation of cell death-related protein expression. The research contributes to developing a roadmap for the characterization of the intricate interactions between nCOFs and biological systems and opens new avenues for exploiting their therapeutic potential in advanced biomedical applications.
Collapse
Affiliation(s)
- Hao Ye
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Carlos Franco
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Mostafa A. Aboouf
- Institute of Veterinary PhysiologyVetsuisse FacultyUniversity of ZurichWinterthurerstrasse 260Zurich8057Switzerland
- Department of BiochemistryFaculty of PharmacyAin Shams UniversityCairo11566Egypt
| | - Markus Thiersch
- Institute of Veterinary PhysiologyVetsuisse FacultyUniversity of ZurichWinterthurerstrasse 260Zurich8057Switzerland
| | - Semih Sevim
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Joaquin Llacer‐Wintle
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Andrea Veciana
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Gemma Llauradó‐Capdevila
- Departament de Ciència dels Materials i Química Física Institut de Química Teòrica i ComputacionalUniversity of BarcelonaBarcelona08028Spain
| | - Kaiyuan Wang
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical University103 Wenhua RoadShenyang Liaoning110016P. R. China
| | - Xiang‐Zhong Chen
- State Key Laboratory of Photovoltaic Science and TechnologyShanghai Frontiers Science Research Base of Intelligent Optoelectronics and PerceptionInstitute of OptoelectronicsInternational Institute of Intelligent Nanorobots and NanosystemsFudan UniversitySonghu Road 2005Shanghai200438China
- Yiwu Research Intitute of Fudan UniversityYiwu322000China
| | - Qiao Tang
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Roc Matheu
- Departament de Química Inorgànica i OrgànicaInstitut de Química Teòrica i ComputacionalBarcelona08028Spain
| | - Pedro D. Wendel‐Garcia
- Institute of Intensive Care MedicineUniversity Hospital ZurichRämistrasse 100Zurich8091Switzerland
| | - Pedro A. Sánchez‐Murcia
- Laboratory of Computer‐Aided Molecular DesignDivision of Medicinal ChemistryOtto‐Loewi Research CenterMedical University of GrazNeue Stiftingstalstraße 6/IIIGrazA‐8010Austria
| | - Bradley J. Nelson
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| | - Cong Luo
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical University103 Wenhua RoadShenyang Liaoning110016P. R. China
| | - Josep Puigmartí‐Luis
- Departament de Ciència dels Materials i Química Física Institut de Química Teòrica i ComputacionalUniversity of BarcelonaBarcelona08028Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Pg. Lluís Companys 23Barcelona08010Spain
| | - Salvador Pané
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZurichZurich8092Switzerland
| |
Collapse
|
30
|
Xie B, Liu Y, Li X, Yang P, He W. Solubilization techniques used for poorly water-soluble drugs. Acta Pharm Sin B 2024; 14:4683-4716. [PMID: 39664427 PMCID: PMC11628819 DOI: 10.1016/j.apsb.2024.08.027] [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: 05/18/2024] [Revised: 07/28/2024] [Accepted: 08/14/2024] [Indexed: 12/13/2024] Open
Abstract
About 40% of approved drugs and nearly 90% of drug candidates are poorly water-soluble drugs. Low solubility reduces the drugability. Effectively improving the solubility and bioavailability of poorly water-soluble drugs is a critical issue that needs to be urgently addressed in drug development and application. This review briefly introduces the conventional solubilization techniques such as solubilizers, hydrotropes, cosolvents, prodrugs, salt modification, micronization, cyclodextrin inclusion, solid dispersions, and details the crystallization strategies, ionic liquids, and polymer-based, lipid-based, and inorganic-based carriers in improving solubility and bioavailability. Some of the most commonly used approved carrier materials for solubilization techniques are presented. Several approved poorly water-soluble drugs using solubilization techniques are summarized. Furthermore, this review summarizes the solubilization mechanism of each solubilization technique, reviews the latest research advances and challenges, and evaluates the potential for clinical translation. This review could guide the selection of a solubilization approach, dosage form, and administration route for poorly water-soluble drugs. Moreover, we discuss several promising solubilization techniques attracting increasing attention worldwide.
Collapse
Affiliation(s)
- Bing Xie
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
31
|
Li X, Zuo Y, Lin X, Guo B, Jiang H, Guan N, Zheng H, Huang Y, Gu X, Yu B, Wang X. Develop Targeted Protein Drug Carriers through a High-Throughput Screening Platform and Rational Design. Adv Healthc Mater 2024; 13:e2401793. [PMID: 38804201 DOI: 10.1002/adhm.202401793] [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: 05/16/2024] [Revised: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Protein-based drugs offer advantages, such as high specificity, low toxicity, and minimal side effects compared to small molecule drugs. However, delivery of proteins to target tissues or cells remains challenging due to the instability, diverse structures, charges, and molecular weights of proteins. Polymers have emerged as a leading choice for designing effective protein delivery systems, but identifying a suitable polymer for a given protein is complicated by the complexity of both proteins and polymers. To address this challenge, a fluorescence-based high-throughput screening platform called ProMatch to efficiently collect data on protein-polymer interactions, followed by in vivo and in vitro experiments with rational design is developed. Using this approach to streamline polymer selection for targeted protein delivery, candidate polymers from commercially available options are identified and a polyhexamethylene biguanide (PHMB)-based system for delivering proteins to white adipose tissue as a treatment for obesity is developed. A branched polyethylenimine (bPEI)-based system for neuron-specific protein delivery to stimulate optic nerve regeneration is also developed. The high-throughput screening methodology expedites identification of promising polymer candidates for tissue-specific protein delivery systems, thereby providing a platform to develop innovative protein-based therapeutics.
Collapse
Affiliation(s)
- Xiaodan Li
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Nanhu Brain-Computer Interface Institute, Hangzhou, 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Yanming Zuo
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Nanhu Brain-Computer Interface Institute, Hangzhou, 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Xurong Lin
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Lingang Laboratory, Shanghai, 200031, China
| | - Binjie Guo
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Lingang Laboratory, Shanghai, 200031, China
| | - Haohan Jiang
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Lingang Laboratory, Shanghai, 200031, China
| | - Naiyu Guan
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Nanhu Brain-Computer Interface Institute, Hangzhou, 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Hanyu Zheng
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Lingang Laboratory, Shanghai, 200031, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, Jiangsu, 226001, P. R. China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, Jiangsu, 226001, P. R. China
| | - Xuhua Wang
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
- Nanhu Brain-Computer Interface Institute, Hangzhou, 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
- Lingang Laboratory, Shanghai, 200031, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, P. R. China
| |
Collapse
|
32
|
Bhirud D, Bhattacharya S, Prajapati BG. Bioengineered carbohydrate polymers for colon-specific drug release: Current trends and future prospects. J Biomed Mater Res A 2024; 112:1860-1872. [PMID: 38721841 DOI: 10.1002/jbm.a.37732] [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: 03/10/2024] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 09/03/2024]
Abstract
The worldwide health burden of colorectal cancer is still substantial, and traditional chemotherapeutic drugs sometimes have poor selectivity, which can result in systemic toxicity and unfavorable side effects. For colon-specific medication delivery, bioengineered carbohydrate polymers have shown promise as carriers. They may enhance treatment effectiveness while minimizing systemic exposure and associated side effects. The unique properties of these manufactured or naturally occurring biopolymers, such as hyaluronic acid, chitosan, alginate, and pectin, enable targeted medicine release. These qualities can be changed to meet the physiological needs of the colon. In the context of colorectal cancer therapy, this article provides a comprehensive overview of current developments and prospective future directions in the field of bioengineered carbohydrate polymer synthesis for colon-specific drug delivery. We discuss numerous techniques for achieving colon-targeted drug release, including enzyme-sensitive polymers, pH-responsive devices, and microbiota-activated processes. To increase tumor selectivity and cellular uptake, we also examine the inclusion of active targeting approaches, such as conjugating specific ligands. Furthermore, we discuss the potential of combination treatment strategies, which use the coadministration of numerous therapeutic medications to target multiple pathways implicated in cancer growth and address drug resistance mechanisms. We address recent biomimetic advances that potentially improve the biocompatibility, cellular uptake, and tumor penetration of carbohydrate polymer-based nanocarriers. These methods involve protein corona engineering and cell membrane coating. Furthermore, we look at the possibility of intelligent and sensitive systems that may adjust their behaviors in response to certain inputs or feedback loops, allowing for precise and regulated drug distribution.
Collapse
Affiliation(s)
- Darshan Bhirud
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Shree S.K. Patel College of Pharmaceutical Education & Research, Mehsana, Gujarat, India
| |
Collapse
|
33
|
Maurya R, Vikal A, Patel P, Narang RK, Kurmi BD. "Enhancing Oral Drug Absorption: Overcoming Physiological and Pharmaceutical Barriers for Improved Bioavailability". AAPS PharmSciTech 2024; 25:228. [PMID: 39354282 DOI: 10.1208/s12249-024-02940-5] [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: 05/18/2024] [Accepted: 09/11/2024] [Indexed: 10/03/2024] Open
Abstract
The oral route stands out as the most commonly used method for drug administration, prized for its non-invasive nature, patient compliance, and easy administration. Several elements influence the absorption of oral medications, including their solubility, permeability across mucosal membranes, and stability within the gastrointestinal (GI) environment. Research has delved into comprehending physicochemical, biochemical, metabolic, and biological obstacles that impact the bioavailability of a drug. To improve oral drug absorption, several pharmaceutical technologies and delivery methods have been studied, including cyclodextrins, micelles, nanocarriers, and lipid-based carriers. This review examines both traditional and innovative drug delivery methods, as well as the physiological and pharmacological barriers influencing medication bioavailability when taken orally. Additionally, it describes the challenges and advancements in developing formulations suitable for oral use.
Collapse
Affiliation(s)
- Rashmi Maurya
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Akash Vikal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Raj Kumar Narang
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
- ISF College of Pharmacy and Research, Rattian Road, Moga, 142048, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
34
|
Liu X, Guo Y, Pan J, Wu T, Zhao B, Wei S, Jiang W, Liu Y. Nanoparticles constructed from natural polyphenols are used in acute kidney injury. J Mater Chem B 2024; 12:8883-8896. [PMID: 39177039 DOI: 10.1039/d4tb00837e] [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: 08/24/2024]
Abstract
Acute kidney injury (AKI) is a severe clinical syndrome characterized by rapid deterioration of renal function caused by a variety of pathogeneses. Natural polyphenols have been considered to have potential in the treatment of AKI due to their powerful antioxidant and anti-inflammatory activities, but their low bioavailability in vivo limits their efficacy. Polyphenol nanoparticles based on a nano-delivery system show good effects in reducing kidney injury, improving renal function and promoting renal tissue repair, and brings new hope and possibility for the treatment of AKI. This review provides an overview of the common characteristics, treatments, and associated adverse effects of AKI. The classification and bioavailability of polyphenols as well as their therapeutic role in AKI and potential possible effects are outlined. The potential therapeutic effects of polyphenol-based nanoparticles on AKI and the underlying mechanisms are discussed.
Collapse
Affiliation(s)
- Xiaohua Liu
- Henan Science and Technology Innovation Promotion Center, Zhengzhou 450046, China
| | - Yike Guo
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiangpeng Pan
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Tingting Wu
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Bing Zhao
- Henan Finance University, Zhengzhou 450046, China
| | - Shuyi Wei
- Plastic Surgery Department, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.
| | - Wei Jiang
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Liu
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| |
Collapse
|
35
|
Yang J, Zeng H, Luo Y, Chen Y, Wang M, Wu C, Hu P. Recent Applications of PLGA in Drug Delivery Systems. Polymers (Basel) 2024; 16:2606. [PMID: 39339068 PMCID: PMC11435547 DOI: 10.3390/polym16182606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/18/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) is a widely used biodegradable and biocompatible copolymer in drug delivery systems (DDSs). In this article, we highlight the critical physicochemical properties of PLGA, including its molecular weight, intrinsic viscosity, monomer ratio, blockiness, and end caps, that significantly influence drug release profiles and degradation times. This review also covers the extensive literature on the application of PLGA in delivering small-molecule drugs, proteins, peptides, antibiotics, and antiviral drugs. Furthermore, we discuss the role of PLGA-based DDSs in the treating various diseases, including cancer, neurological disorders, pain, and inflammation. The incorporation of drugs into PLGA nanoparticles and microspheres has been shown to enhance their therapeutic efficacy, reduce toxicity, and improve patient compliance. Overall, PLGA-based DDSs holds great promise for the advancement of the treatment and management of multiple chronic conditions.
Collapse
Affiliation(s)
- Jie Yang
- Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Huiying Zeng
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Yusheng Luo
- International School, Jinan University, Guangzhou 510006, China
| | - Ying Chen
- Guangdong Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510660, China
| | - Miao Wang
- Guangdong Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510660, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Ping Hu
- Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| |
Collapse
|
36
|
Nascimento Júnior JAC, Santos AM, Oliveira AMS, Santos AB, de Souza Araújo AA, Aragón DM, Frank LA, Serafini MR. The Tiny Big Difference: Nanotechnology in Photoprotective Innovations - A Systematic Review. AAPS PharmSciTech 2024; 25:212. [PMID: 39242428 DOI: 10.1208/s12249-024-02925-4] [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: 07/07/2024] [Accepted: 08/18/2024] [Indexed: 09/09/2024] Open
Abstract
UV radiation causes long- and short-term skin damage, such as erythema and skin cancer. Therefore, the use of sunscreens is extremely important. However, concerns about UV filter safety have prompted exploration into alternative solutions, with nanotechnology emerging as a promising avenue. This systematic review identified 23 experimental studies utilizing nanocarriers to encapsulate sunscreens with the aim of enhancing their efficacy and safety. Polymeric and lipid nanoparticles are frequently employed to encapsulate both organic and inorganic UV filters along with natural antioxidants. Nanocarriers have demonstrated benefits including reduced active ingredient usage, increased sun protection factor, and mitigated photoinstability. Notably, they also decreased the skin absorption of UV filters. In summary, nanocarriers represent a viable strategy for improving sunscreen formulations, offering enhanced physicochemical properties and bolstered photoprotective effects, thereby addressing concerns regarding UV filter safety and efficacy in cosmetic applications.
Collapse
Affiliation(s)
| | | | - Ana Maria Santos Oliveira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| | | | - Adriano Antunes de Souza Araújo
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Brazil
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Brazil
| | - Diana Marcela Aragón
- Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional da Colombia, Bogotá, D.C, Colombia
| | - Luiza Abrahão Frank
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
- Núcleo de Terapias Nanotecnológicas (NTnano), Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Mairim Russo Serafini
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil.
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Brazil.
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Brazil.
| |
Collapse
|
37
|
Esnaashari F, Zahmatkesh H. Antivirulence activities of Rutin-loaded chitosan nanoparticles against pathogenic Staphylococcus aureus. BMC Microbiol 2024; 24:328. [PMID: 39244527 PMCID: PMC11380343 DOI: 10.1186/s12866-024-03446-7] [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: 10/09/2023] [Accepted: 07/26/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Staphylococcus aureus is an infectious bacterium that is frequently found in healthcare settings and the community. This study aimed to prepare rutin-loaded chitosan nanoparticles (Rut-CS NPs) and assess their antibacterial activity against pathogenic strains of S. aureus. RESULTS The synthesized Rut-CS NPs exhibited an amorphous morphology with a size ranging from 160 to 240 nm and a zeta potential of 37.3 mV. Rut-CS NPs demonstrated significant antibacterial activity against S. aureus strains. Following exposure to Rut-CS NPs, the production of staphyloxanthin pigment decreased by 43.31-89.63%, leading to increased susceptibility of S. aureus to hydrogen peroxide. Additionally, visual inspection of cell morphology indicated changes in membrane integrity and permeability upon Rut-CS NPs exposure, leading to a substantial increase (107.07-191.08%) in cytoplasmic DNA leakage in the strains. Furthermore, ½ MIC of Rut-CS NPs effectively inhibited the biofilm formation (22.5-37.5%) and hemolytic activity (69-82.59%) in the S. aureus strains. CONCLUSIONS Our study showcases that Rut-CS NPs can serve as a novel treatment agent to combat S. aureus infections by altering cell morphology and inhibiting virulence factors of S. aureus.
Collapse
Affiliation(s)
- Fatemeh Esnaashari
- Department of Biology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Hossein Zahmatkesh
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran.
| |
Collapse
|
38
|
Keum H, Cevik E, Kim J, Demirlenk YM, Atar D, Saini G, Sheth RA, Deipolyi AR, Oklu R. Tissue Ablation: Applications and Perspectives. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310856. [PMID: 38771628 PMCID: PMC11309902 DOI: 10.1002/adma.202310856] [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: 10/17/2023] [Revised: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Tissue ablation techniques have emerged as a critical component of modern medical practice and biomedical research, offering versatile solutions for treating various diseases and disorders. Percutaneous ablation is minimally invasive and offers numerous advantages over traditional surgery, such as shorter recovery times, reduced hospital stays, and decreased healthcare costs. Intra-procedural imaging during ablation also allows precise visualization of the treated tissue while minimizing injury to the surrounding normal tissues, reducing the risk of complications. Here, the mechanisms of tissue ablation and innovative energy delivery systems are explored, highlighting recent advancements that have reshaped the landscape of clinical practice. Current clinical challenges related to tissue ablation are also discussed, underlining unmet clinical needs for more advanced material-based approaches to improve the delivery of energy and pharmacology-based therapeutics.
Collapse
Affiliation(s)
- Hyeongseop Keum
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Enes Cevik
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Jinjoo Kim
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Yusuf M Demirlenk
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Dila Atar
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Gia Saini
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Rahul A Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Amy R Deipolyi
- Interventional Radiology, Department of Surgery, West Virginia University, Charleston Area Medical Center, Charleston, WV 25304, USA
| | - Rahmi Oklu
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
- Division of Vascular & Interventional Radiology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, Arizona 85054, USA
| |
Collapse
|
39
|
Kamenova K, Prancheva A, Radeva L, Yoncheva K, Zaharieva MM, Najdenski HM, Petrov PD. Nanosized Complexes of the Proteolytic Enzyme Serratiopeptidase with Cationic Block Copolymer Micelles Enhance the Proliferation and Migration of Human Cells. Pharmaceutics 2024; 16:988. [PMID: 39204333 PMCID: PMC11358905 DOI: 10.3390/pharmaceutics16080988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
In this study, we describe the preparation of the cationic block copolymer nanocarriers of the proteolytic enzyme serratiopeptidase (SER). Firstly, an amphiphilic poly(2-(dimethylamino)ethyl methacrylate)-b-poly(ε-caprolactone)-b-poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA9-b-PCL35-b-PDMAEMA9) triblock copolymer was synthesized by reversible addition-fragmentation chain-transfer (RAFT) polymerization. Then, cationic micellar nanocarriers consisting of a PCL hydrophobic core and a PDMAEMA hydrophilic shell were formed by the solvent evaporation method. SER was loaded into the polymeric micelles by electrostatic interaction between the positively charged micellar shell and the negatively charged enzyme molecules. The particle size, zeta potential, and colloid stability of complexes as a function of SER concentration were investigated by dynamic and electrophoretic light scattering. It was found that SER retained its proteolytic activity after immobilization in polymeric carriers. Moreover, the complexes have a concentration-dependent enhancing effect on the proliferation and migration of human keratinocyte HaCaT and gingival fibroblast HGF cells.
Collapse
Affiliation(s)
- Katya Kamenova
- Institute of Polymers, Bulgarian Academy of Sciences, bl.103 Akad. G. Bonchev Str., 1113 Sofia, Bulgaria; (K.K.); (A.P.)
| | - Anna Prancheva
- Institute of Polymers, Bulgarian Academy of Sciences, bl.103 Akad. G. Bonchev Str., 1113 Sofia, Bulgaria; (K.K.); (A.P.)
| | - Lyubomira Radeva
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (L.R.); (K.Y.)
| | - Krassimira Yoncheva
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (L.R.); (K.Y.)
| | - Maya M. Zaharieva
- The Stephan Angeloff Institute of Microbiology, 1113 Sofia, Bulgaria; (M.M.Z.); (H.M.N.)
| | - Hristo M. Najdenski
- The Stephan Angeloff Institute of Microbiology, 1113 Sofia, Bulgaria; (M.M.Z.); (H.M.N.)
| | - Petar D. Petrov
- Institute of Polymers, Bulgarian Academy of Sciences, bl.103 Akad. G. Bonchev Str., 1113 Sofia, Bulgaria; (K.K.); (A.P.)
| |
Collapse
|
40
|
Liu Y, Li C, Yang X, Yang B, Fu Q. Stimuli-responsive polymer-based nanosystems for cardiovascular disease theranostics. Biomater Sci 2024; 12:3805-3825. [PMID: 38967109 DOI: 10.1039/d4bm00415a] [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: 07/06/2024]
Abstract
Stimulus-responsive polymers have found widespread use in biomedicine due to their ability to alter their own structure in response to various stimuli, including internal factors such as pH, reactive oxygen species (ROS), and enzymes, as well as external factors like light. In the context of atherosclerotic cardiovascular diseases (CVDs), stimulus-response polymers have been extensively employed for the preparation of smart nanocarriers that can deliver therapeutic and diagnostic drugs specifically to inflammatory lesions. Compared with traditional drug delivery systems, stimulus-responsive nanosystems offer higher sensitivity, greater versatility, wider applicability, and enhanced biosafety. Recent research has made significant contributions towards designing stimulus-responsive polymer nanosystems for CVDs diagnosis and treatment. This review summarizes recent advances in this field by classifying stimulus-responsive polymer nanocarriers according to different responsiveness types and describing numerous stimuli relevant to these materials. Additionally, we discuss various applications of stimulus-responsive polymer nanomaterials in CVDs theranostics. We hope that this review will provide valuable insights into optimizing the design of stimulus-response polymers for accelerating their clinical application in diagnosing and treating CVDs.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Congcong Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
41
|
Hamzeheipour S, Rezaei A, Behniafar H. Novel amphiphilic (AB)3‐type star block polymer: Synthesis and micellization study. J Appl Polym Sci 2024; 141. [DOI: 10.1002/app.55565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 04/01/2024] [Indexed: 01/06/2025]
Abstract
AbstractAn amphiphilic three‐armed star polymer was synthesized through the polar cycloaddition reaction of a polyethylene glycol‐based macro‐triazide with an Yne‐terminated polycaprolactone in a molar ratio of 1:3 (click chemistry). Thus, each arm is itself a di‐block copolymer branched from the core, which is a 1,3,5‐triazine ring. The attachment of the pre‐synthesized hydrophobic segments to the hydrophilic segments in each arm leads to the creation of a 1,2,3‐triazole ring in the structure. Fourier transform infrared (FTIR) and proton nuclear magnetic resonance (H‐NMR) spectroscopies were used to verify the structures. The micellization process of the prepared amphiphilic polymer was investigated in aqueous medium. For this purpose, the critical micelle concentration (CMC) was specified by the fluorometric method. Using the transmission electron microscopy (TEM) technique, it was observed that spherical self‐assemblies with a mean diameter of <100 nm are formed from the aggregation of the amphiphilic macromolecules. In addition, the hydrodynamic diameter (H) of the polymeric micelles and their size distribution were examined by dynamic light scattering (DLS) technique.
Collapse
Affiliation(s)
| | - Abbas Rezaei
- School of Chemistry Damghan University Damghan Iran
| | | |
Collapse
|
42
|
Mukherjee D, Raikwar S. Recent Update on Nanocarrier(s) as the Targeted Therapy for Breast Cancer. AAPS PharmSciTech 2024; 25:153. [PMID: 38961013 DOI: 10.1208/s12249-024-02867-x] [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: 11/28/2023] [Accepted: 06/11/2024] [Indexed: 07/05/2024] Open
Abstract
Despite ongoing advances in cancer therapy, the results for the treatment of breast cancer are not satisfactory. The advent of nanotechnology promises to be an essential tool to improve drug delivery effectiveness in cancer therapy. Nanotechnology provides an opportunity to enhance the treatment modality by preventing degradation, improving tumour targeting, and controlling drug release. Recent advances have revealed several strategies to prevent cancer metastasis using nano-drug delivery systems (NDDS). These strategies include the design of appropriate nanocarriers loaded with anti-cancer drugs that target the optimization of physicochemical properties, modulate the tumour microenvironment, and target biomimetic techniques. Nanocarriers have emerged as a preferential approach in the chemotropic treatment for breast cancer due to their pivotal role in safeguarding the therapeutic agents against degradation. They facilitate efficient drug concentration in targeted cells, surmount the resistance of drugs, and possess a small size. Nevertheless, these nanocarrier(s) have some limitations, such as less permeability across the barrier and low bioavailability of loaded drugs. To overcome these challenges, integrating external stimuli has been employed, encompassing infrared light, thermal stimulation, microwaves, and X-rays. Among these stimuli, ultrasound-triggered nanocarriers have gained significant attention due to their cost-effectiveness, non-invasive nature, specificity, ability to penetrate tissues, and capacity to deliver elevated drug concentrations to intended targets. This article comprehensively reviews recent advancements in different nanocarriers for breast cancer chemotherapy. It also delves into the associated hurdles and offers valuable insights into the prospective directions for this innovative field.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sarjana Raikwar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
43
|
Zhou SY, Giang NN, Kim H, Chien PN, Le LTT, Trinh TT, Nga PT, Kwon HJ, Ham JR, Lee WK, Gu YJ, Zhang XR, Jin YX, Nam SY, Heo CY. Assessing the efficacy of mesotherapy products: Ultra Exo Booster, and Ultra S Line Plus in hair growth: An ex vivo study. Skin Res Technol 2024; 30:e13780. [PMID: 39031929 PMCID: PMC11259544 DOI: 10.1111/srt.13780] [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: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 07/22/2024]
Abstract
In this study, scalp tissues from Korean adults between 20 and 80 without skin disease were used. Scalp tissues were processed, and hair follicles were isolated and cultured with different treatments (including Bioscalp, Ultra Exo Booster, and Ultra S Line Plus) from Ultra V company. Over 12 days, observations and measurements of hair follicle characteristics were recorded at intervals (Days 0, 3, 6, 9, and 12). The study assessed the impact of these substances on hair follicle growth and morphology. Bioscalp, combined with Ultra Exo Booster and Ultra S Line Plus, showed significant hair elongation in ex vivo. Preservation of hair bulb diameter was observed, indicating potential for sustained hair growth by exosome-based products. The hair growth cycle analysis suggested a lower transition to the catagen stage in test products from Ultra V compared to non-treated groups. The research findings indicated that the tested formulations, especially the combination of Bioscalp, Ultra Exo Booster, and Ultra S Line Plus, demonstrated significant effectiveness in promoting hair growth, maintaining the integrity of the hair bulb, and reducing the transition to the catagen stage. The study suggests promising alternative treatments for hair loss, illustrating results that were as good as those of the conventional testing product groups.
Collapse
Affiliation(s)
- Shu Yi Zhou
- Department of MedicineCollege of MedicineSeoul National UniversitySeoulSouth Korea
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
| | - Nguyen Ngan Giang
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
- Department of Medical Device DevelopmentCollege of MedicineSeoul National UniversitySeoulSouth Korea
| | - Hyunjee Kim
- Korean Institute of Nonclinical StudyH&Bio. Co. Ltd.SeongnamSouth Korea
| | - Pham Ngoc Chien
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
- Korean Institute of Nonclinical StudyH&Bio. Co. Ltd.SeongnamSouth Korea
| | - Linh Thi Thuy Le
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
- Department of Biomedical ScienceCollege of MedicineSeoul National UniversitySeoulSouth Korea
- Faculty of Medical TechniqueHai Phong University of Medicine and PharmacyHaiphongVietnam
| | - Thuy‐Tien Thi Trinh
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
- Korean Institute of Nonclinical StudyH&Bio. Co. Ltd.SeongnamSouth Korea
| | - Pham Thi Nga
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
- Korean Institute of Nonclinical StudyH&Bio. Co. Ltd.SeongnamSouth Korea
| | | | | | - Won Ku Lee
- UltraV Co., Ltd. R&D CenterSeoulSouth Korea
| | - Yeon Ju Gu
- UltraV Co., Ltd. R&D CenterSeoulSouth Korea
| | - Xin Rui Zhang
- Department of MedicineCollege of MedicineSeoul National UniversitySeoulSouth Korea
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
| | - Yong Xun Jin
- Department of MedicineCollege of MedicineSeoul National UniversitySeoulSouth Korea
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
| | - Sun Young Nam
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
| | - Chan Yeong Heo
- Department of MedicineCollege of MedicineSeoul National UniversitySeoulSouth Korea
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamSouth Korea
- Department of Medical Device DevelopmentCollege of MedicineSeoul National UniversitySeoulSouth Korea
- Korean Institute of Nonclinical StudyH&Bio. Co. Ltd.SeongnamSouth Korea
| |
Collapse
|
44
|
Zhou Y, Xu M, Shen W, Xu Y, Shao A, Xu P, Yao K, Han H, Ye J. Recent Advances in Nanomedicine for Ocular Fundus Neovascularization Disease Management. Adv Healthc Mater 2024; 13:e2304626. [PMID: 38406994 PMCID: PMC11468720 DOI: 10.1002/adhm.202304626] [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: 12/26/2023] [Revised: 02/22/2024] [Indexed: 02/27/2024]
Abstract
As an indispensable part of the human sensory system, visual acuity may be impaired and even develop into irreversible blindness due to various ocular pathologies. Among ocular diseases, fundus neovascularization diseases (FNDs) are prominent etiologies of visual impairment worldwide. Intravitreal injection of anti-vascular endothelial growth factor drugs remains the primary therapy but is hurdled by common complications and incomplete potency. To renovate the current therapeutic modalities, nanomedicine emerged as the times required, which is endowed with advanced capabilities, able to fulfill the effective ocular fundus drug delivery and achieve precise drug release control, thus further improving the therapeutic effect. This review provides a comprehensive summary of advances in nanomedicine for FND management from state-of-the-art studies. First, the current therapeutic modalities for FNDs are thoroughly introduced, focusing on the key challenges of ocular fundus drug delivery. Second, nanocarriers are comprehensively reviewed for ocular posterior drug delivery based on the nanostructures: polymer-based nanocarriers, lipid-based nanocarriers, and inorganic nanoparticles. Thirdly, the characteristics of the fundus microenvironment, their pathological changes during FNDs, and corresponding strategies for constructing smart nanocarriers are elaborated. Furthermore, the challenges and prospects of nanomedicine for FND management are thoroughly discussed.
Collapse
Affiliation(s)
- Yifan Zhou
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Mingyu Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Wenyue Shen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Yufeng Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - An Shao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Peifang Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| |
Collapse
|
45
|
Zhang Y, Zhang Y, Ding R, Zhang K, Guo H, Lin Y. Self-Assembled Nanocarrier Delivery Systems for Bioactive Compounds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310838. [PMID: 38214694 DOI: 10.1002/smll.202310838] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/25/2023] [Indexed: 01/13/2024]
Abstract
Although bioactive compounds (BCs) have many important functions, their applications are greatly limited due to their own defects. The development of nanocarriers (NCs) technology has gradually overcome the defects of BCs. NCs are equally important as BCs to some extent. Self-assembly (SA) methods to build NCs have many advantages than chemical methods, and SA has significant impact on the structure and function of NCs. However, the relationship among SA mechanism, structure, and function has not been given enough attention. Therefore, from the perspective of bottom-up building mechanism, the concept of SA-structure-function of NCs is emphasized to promote the development of SA-based NCs. First, the conditions and forces for occurring SA are introduced, and then the SA basis and molecular mechanism of protein, polysaccharide, and lipid are summarized. Then, varieties of the structures formed based on SA are introduced in detail. Finally, facing the defects of BCs and how to be well solved by NCs are also elaborated. This review attempts to describe the great significance of constructing artificial NCs to deliver BCs from the aspects of SA-structure-function, so as to promote the development of SA-based NCs and the wide application of BCs.
Collapse
Affiliation(s)
- Yafei Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yuning Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Rui Ding
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
| | - Kai Zhang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
- Food Laboratory of Zhongyuan, Luohe, 462300, China
| |
Collapse
|
46
|
An X, Yang J, Cui X, Zhao J, Jiang C, Tang M, Dong Y, Lin L, Li H, Wang F. Advances in local drug delivery technologies for improved rheumatoid arthritis therapy. Adv Drug Deliv Rev 2024; 209:115325. [PMID: 38670229 DOI: 10.1016/j.addr.2024.115325] [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: 11/03/2023] [Revised: 03/25/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by an inflammatory microenvironment and cartilage erosion within the joint cavity. Currently, antirheumatic agents yield significant outcomes in RA treatment. However, their systemic administration is limited by inadequate drug retention in lesion areas and non-specific tissue distribution, reducing efficacy and increasing risks such as infection due to systemic immunosuppression. Development in local drug delivery technologies, such as nanostructure-based and scaffold-assisted delivery platforms, facilitate enhanced drug accumulation at the target site, controlled drug release, extended duration of the drug action, reduced both dosage and administration frequency, and ultimately improve therapeutic outcomes with minimized damage to healthy tissues. In this review, we introduced pathogenesis and clinically used therapeutic agents for RA, comprehensively summarized locally administered nanostructure-based and scaffold-assisted drug delivery systems, aiming at improving the therapeutic efficiency of RA by alleviating the inflammatory response, preventing bone erosion and promoting cartilage regeneration. In addition, the challenges and future prospects of local delivery for clinical translation in RA are discussed.
Collapse
Affiliation(s)
- Xiaoran An
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jiapei Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Xiaolin Cui
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jiaxuan Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Chenwei Jiang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Minglu Tang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yabing Dong
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Longfei Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Hui Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330000, PR China
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| |
Collapse
|
47
|
Karabulut H, Xu D, Ma Y, Tut TA, Ulag S, Pinar O, Kazan D, Guncu MM, Sahin A, Wei H, Chen J, Gunduz O. A new strategy for the treatment of middle ear infection using ciprofloxacin/amoxicillin-loaded ethyl cellulose/polyhydroxybutyrate nanofibers. Int J Biol Macromol 2024; 269:131794. [PMID: 38697434 DOI: 10.1016/j.ijbiomac.2024.131794] [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: 07/29/2023] [Revised: 03/23/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
A middle ear infection occurs due to the presence of several microorganisms behind the eardrum (tympanic membrane) and is very challenging to treat due to its unique location and requires a well-designed treatment. If not treated properly, the infection can result in severe symptoms and unavoidable side effects. In this study, excellent biocompatible ethyl cellulose (EC) and biodegradable polyhydroxybutyrate (PHB) biopolymer were used to fabricate drug-loaded nanofiber scaffolds using an electrospinning technique to overcome antibiotic overdose and insufficient efficacy of drug release during treatment. PHB polymer was produced from Halomonas sp., and the purity of PHB was found to around be 90 %. Additionally, ciprofloxacin (CIP) and amoxicillin (AMX) are highly preferable since both drugs are highly effective against gram-negative and gram-positive bacteria to treat several infections. Obtained smooth nanofibers were between 116.24 and 171.82 nm in diameter and the addition of PHB polymer and antibiotics improved the morphology of the nanofiber scaffolds. Thermal properties of the nanofiber scaffolds were tested and the highest Tg temperature resulted at 229 °C. The mechanical properties of the scaffolds were tested, and the highest tensile strength resulted in 4.65 ± 6.33 MPa. Also, drug-loaded scaffolds were treated against the most common microorganisms that cause the infection, such as S.aureus, E.coli, and P.aeruginosa, and resulted in inhibition zones between 10 and 21 mm. MTT assay was performed by culturing human adipose-derived mesenchymal stem cells (hAD MSCs) on the scaffolds. The morphology of the hAD MSCs' attachment was tested with SEM analysis and hAD MSCs were able to attach, spread, and live on each scaffold even on the day of 7. The cumulative drug release kinetics of CIP and AMX from drug-loaded scaffolds were analysed in phosphate-buffered saline (pH: 7.4) within different time intervals of up to 14 days using a UV spectrophotometer. Furthermore, the drug release showed that the First-Order and Korsmeyer-Peppas models were the most suitable kinetic models. Animal testing was performed on SD rats, matrix and collagen deposition occurred on days 5 and 10, which were observed using Hematoxylin-eosin and Masson's trichrome staining. At the highest drug concentration, a better repair effect was observed. Results were promising and showed potential for novel treatment.
Collapse
Affiliation(s)
- Hatice Karabulut
- Department of Systems Science and Industrial Engineering, State University of New York at Binghamton, New York, USA; Center for Nanotechnology & Biomaterials Research, Marmara University, Istanbul, Turkey
| | - Dingli Xu
- Health Science Center, Ningbo University, Zhejiang, China
| | - Yuxi Ma
- Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, China; University of Chinese Academy of Sciences, Beijing, China
| | - Tufan Arslan Tut
- Center for Nanotechnology & Biomaterials Research, Marmara University, Istanbul, Turkey; Department of Metallurgy and Materials Engineering, Faculty of Technology, Marmara University, Istanbul, Turkey
| | - Songul Ulag
- Center for Nanotechnology & Biomaterials Research, Marmara University, Istanbul, Turkey; Department of Metallurgy and Materials Engineering, Faculty of Technology, Marmara University, Istanbul, Turkey
| | - Orkun Pinar
- Department of Separation Science, LUT School of Engineering Science, LUT University, Sammonkatu 12, 50130, Mikkeli, Finland
| | - Dilek Kazan
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey; Bacpolyzyme Bioengineering LLC., Marmara University Technopark., Istanbul, Turkey
| | - Mehmet Mucahit Guncu
- Institute of Health Sciences, Department of Microbiology, Marmara University, Istanbul, Turkey
| | - Ali Sahin
- Department of Biochemistry, School of Medicine/ Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, Istanbul, Turkey
| | - Hua Wei
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China.
| | - Jing Chen
- Institute of Medical Sciences, The Second Hospital & Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan 250033, China..
| | - Oguzhan Gunduz
- Center for Nanotechnology & Biomaterials Research, Marmara University, Istanbul, Turkey; Department of Metallurgy and Materials Engineering, Faculty of Technology, Marmara University, Istanbul, Turkey.
| |
Collapse
|
48
|
Alavifar SM, Golshan M, Hosseini MS, Salami-Kalajahi M. Coumarin-Modified Starch Fluorescent Nanoparticles as Sensor of Fe 3+ and Zn 2+ ions Utilizing Dynamic Quenching and Chelation Mechanisms. J Fluoresc 2024:10.1007/s10895-024-03752-3. [PMID: 38739316 DOI: 10.1007/s10895-024-03752-3] [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: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 05/14/2024]
Abstract
Zinc and iron are two essential trace minerals that play a pivotal role in maintaining optimal health and well-being in the human body. Despite being required in relatively small quantities, their significance can be understated as they participate in a wide array of critical physiological processes such as oxygen transport, DNA synthesis, controlling nutrient availability, etc. Understanding the distribution and behavior of these ions in natural water bodies is essential for assessing water quality, studying ecological processes, and managing environmental impacts. In this study, we have developed a dual fluorescence probe using starch which was functionalized with coumarin derivatives, for efficient detection of Fe3+ and Zn2+ ions. This structure led a self-assembled starch/coumarin (SC) fluorescent nanoparticles with strong fluorescence intensity under ultraviolet light (365 nm). The quenching effect of Fe3+ on the SC fluorescent probe enabled efficient specific detection of Fe3+. Furthermore, Zn2+ ions increased fluorescence intensity of coumarin compounds (λemission = 459). This phenomenon occurs when the coumarin compound forms a complex or interacts with the zinc ion, resulting in enhanced fluorescence emission. In summary, the developed fluorescent probe offered a promising approach for sensitive and specific detection of iron and zinc ions in aqueous solutions.
Collapse
Affiliation(s)
- Seyyed-Mahdi Alavifar
- Faculty of Polymer Engineering, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
- Institute of Polymeric Materials, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
| | - Marzieh Golshan
- Faculty of Polymer Engineering, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
- Institute of Polymeric Materials, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
| | - Mahdi Salami Hosseini
- Faculty of Polymer Engineering, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
- Institute of Polymeric Materials, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
| | - Mehdi Salami-Kalajahi
- Faculty of Polymer Engineering, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran.
- Institute of Polymeric Materials, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran.
| |
Collapse
|
49
|
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
50
|
Yang C, Du Y, Li Q, Gao X, Zha P, Zhan W, Liu K, Bi F, Hua Z, Yang G. Morphological Transformation and Surface Engineering of Glycovesicles Driven by Bioinspired Hydrogen Bonds of Nucleobases. ACS Macro Lett 2024; 13:468-474. [PMID: 38574471 DOI: 10.1021/acsmacrolett.4c00037] [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: 04/06/2024]
Abstract
Glycopolymer-based supramolecular glycoassemblies with signal-driven cascade morphological deformation and accessible surface engineering toward bioinspired functional glycomaterials have attracted much attention due to their diverse applications in fundamental and practical scenarios. Herein, we achieved the cascade morphological transformation and surface engineering of a nucleobase-containing polymeric glycovesicle through exploiting the bioinspired complementary multiple hydrogen bonds of complementary nucleobases. First, the synthesized thymine-containing glycopolymers (PGal30-b-PTAm249) are capable of self-assembling into well-defined glycovesicles. Several kinds of amphiphilic adenine-containing block copolymers with neutral, positive, and negative charges were synthesized to engineer the glycovesicles through the multiple hydrogen bonds between adenine and thymine. A cascade of morphological transformations from vesicles to ruptured vesicles with tails, to worm-like micelles, and finally to spherical micelles were observed via continuously adding the adenine-containing polymer into the thymine-containing glycovesicles. Furthermore, the surface charge properties of these glyconano-objects can be facilely regulated through incorporating various adenine-containing polymers. This work demonstrates the potential application of a unique bioinspired approach to precisely engineer the morphology and surface properties of glycovesicles for boosting their biological applications.
Collapse
Affiliation(s)
- Caiyun Yang
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yixuan Du
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Qiaoran Li
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xinru Gao
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Peng Zha
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wanli Zhan
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ketao Liu
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Feihu Bi
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zan Hua
- The Key Laboratory of Functional Molecular Solids, Ministry of Education, and Department of Materials Chemistry, School of Chemistry and Materials Science, Anhui Normal University, Wuhu, Anhui 214002, China
| | - Guang Yang
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei, Anhui 230036, China
| |
Collapse
|