1
|
Tarvirdipour S, Abdollahi SN, Köser J, Bina M, Schoenenberger CA, Palivan CG. Enhanced antimicrobial protection through surface immobilization of antibiotic-loaded peptide multicompartment micelles. J Mater Chem B 2025. [PMID: 40227831 PMCID: PMC11996027 DOI: 10.1039/d5tb00246j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
The escalating global threat of antibiotic-resistant bacterial infections, driven by biofilm formation on medical device surfaces, prompts the need for innovative therapeutic strategies. To address this growing challenge, we develop rifampicin-loaded multicompartment micelles (RIF-MCMs) immobilized on surfaces, offering a dual-functional approach to enhance antimicrobial efficacy for localized therapeutic applications. We first optimize the physicochemical properties of RIF-MCMs, and subsequently coat the optimal formulation onto a glass substrate, as confirmed by quartz crystal microbalance and atomic force microscopy. Surface-immobilized RIF-MCMs facilitate sustained antibiotic release in response to biologically relevant temperatures (37 °C and 42 °C). In addition, their heterogeneous distribution enhances the surface's roughness, contributing to the antibacterial activity through passive mechanisms such as hindering bacterial adhesion and biofilm formation. In vitro antimicrobial testing demonstrates that RIF-MCM-modified surfaces achieve a 98% reduction in Staphylococcus aureus viability and a three-order-of-magnitude decrease in colony formation compared to unmodified surfaces. In contrast, RIF-MCMs exhibit minimal cytotoxicity to mammalian cells, making them suitable candidates for medical device coatings. Our dual-function antimicrobial strategy, combining sustained antibiotic release and enhanced surface roughness, presents a promising approach to locally prevent implant-associated infections and biofilm formation.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel-4058, Switzerland.
- NCCR-Molecular Systems Engineering, Mattenstrasse 24a, Basel-4058, Switzerland
| | - S Narjes Abdollahi
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel-4058, Switzerland.
| | - Joachim Köser
- School of Life Sciences, Institute for Chemistry and Bioanalytics, University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasse 30, Muttenz-4132, Switzerland
| | - Maryame Bina
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel-4058, Switzerland.
| | | | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel-4058, Switzerland.
- NCCR-Molecular Systems Engineering, Mattenstrasse 24a, Basel-4058, Switzerland
| |
Collapse
|
2
|
Xia Q, Zhou S, Zhou J, Zhao X, Saif MS, Wang J, Hasan M, Zhao M, Liu Q. Recent Advances and Challenges for Biological Materials in Micro/Nanocarrier Synthesis for Bone Infection and Tissue Engineering. ACS Biomater Sci Eng 2025; 11:1945-1969. [PMID: 40067283 DOI: 10.1021/acsbiomaterials.4c02118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Roughly 1.71 billion people worldwide suffer from large bone abnormalities, which are the primary cause of disability. Traditional bone grafting procedures have several drawbacks that impair their therapeutic efficacy and restrict their use in clinical settings. A great deal of work has been done to create fresh, more potent strategies. Under these circumstances, a crucial technique for the regeneration of major lesions has emerged: bone tissue engineering (BTE). BTE involves the use of biomaterials that can imitate the natural design of bone. To yet, no biological material has been able to fully meet the parameters of the perfect implantable material, even though several varieties have been created and investigated for bone regeneration. Against this backdrop, researchers have focused a great deal of interest over the past few years on the subject of nanotechnology and the use of nanostructures in regenerative medicine. The ability to create nanoengineered particles that can overcome the current constraints in regenerative strategies─such as decreased cell proliferation and differentiation, insufficient mechanical strength in biological materials, and insufficient production of extrinsic factors required for effective osteogenesis has revolutionized the field of bone and tissue engineering. The effects of nanoparticles on cell characteristics and the application of biological materials for bone regeneration are the main topics of our review, which summarizes the most recent in vitro and in vivo research on the application of nanotechnology in the context of BTE.
Collapse
Affiliation(s)
- Qipeng Xia
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Shuyan Zhou
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, PR China
| | - Jingya Zhou
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- College of Acupuncture and Massage, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, PR China
| | - Xia Zhao
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| | - Muhammad Saqib Saif
- Department of Biochemistry, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Jianping Wang
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Murtaza Hasan
- Department of Biotechnology, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Min Zhao
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Qiang Liu
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| |
Collapse
|
3
|
Ashoub MH, Golestani A, Amiri M, Razavi R, Farsinejad A. pH-Sensitive Sulfasalazine Release from Green-Synthesized Mesoporous Fe 3O 4@SiO 2 Nanocomposites using Opuntia ficus-indica Extract. J Pharm Sci 2025:103792. [PMID: 40222721 DOI: 10.1016/j.xphs.2025.103792] [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: 12/02/2024] [Revised: 03/27/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025]
Abstract
This study presents the green synthesis of mesoporous Fe3O4@SiO2 nanocomposites using Opuntia ficus-indica extract for pH-sensitive delivery of sulfasalazine. The synthesized nanocomposites exhibited well-defined spherical morphology with particle sizes ranging from 80-120 nm and superior superparamagnetic properties with a saturation magnetization of 75.8 emu/g. X-ray diffraction analysis confirmed the formation of a pure magnetite phase with an average crystallite size of 17.37 nm, while nitrogen physisorption revealed a high specific surface area of 165.8 m²/g with uniform mesopores centered at 4.2 nm. The nanocomposites demonstrated exceptional drug loading characteristics with 81.1% loading efficiency and 23.9% loading capacity. pH-dependent release studies showed enhanced release under acidic conditions (82% at pH 3.5) compared to physiological pH (50% at pH 7.4), indicating potential for targeted drug delivery applications. Cytotoxicity studies using L929 cells and peripheral blood mononuclear cells (PBMCs) revealed remarkable biocompatibility at concentrations up to 200 µg/mL over 72 hours. These findings establish the potential of green-synthesized mesoporous Fe3O4@SiO2 nanocomposites as efficient carriers for pH-sensitive drug delivery systems, offering an environmentally friendly approach to developing advanced therapeutic platforms.
Collapse
Affiliation(s)
- Muhammad Hossein Ashoub
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amin Golestani
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mahnaz Amiri
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran.
| | - Alireza Farsinejad
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
4
|
Wang L, Wang Y, Deng C, Eggleston I, Gao S, Li A, Alvarez Reyes WR, Cai K, Qiu R, Haynes CL, White JC, Xing B. Optimizing SiO 2 Nanoparticle Structures to Enhance Drought Resistance in Tomato ( Solanum lycopersicum L.): Insights into Nanoparticle Dissolution and Plant Stress Response. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40200726 DOI: 10.1021/acs.jafc.5c03048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Drought stress significantly limits crop productivity and poses a critical threat to global food security. Silica nanoparticles (SiO2NPs) have shown a potential to mitigate drought stress, but the role of the nanostructure on overall efficacy remains unclear. This study evaluated solid (SSiO2NPs), porous (PSiO2NPs), and hollow (HSiO2NPs) SiO2NPs for their effects on drought-stressed tomatoes (Solanum lycopersicum L.). Silicic acid release rates followed the order: HSiO2NPs > PSiO2NPs > SSiO2NPs > Bulk-SiO2. Compared to untreated controls, foliar application of PSiO2NPs and HSiO2NPs under drought stress significantly improved shoot Si concentrations and plants' dry weight. These treatments also enhanced antioxidant enzyme activities (catalase, peroxidase, and superoxide dismutase) and phytohormone-targeted metabolome levels (jasmonic acid, salicylic acid, and auxin), contributing to greater drought tolerance. Conversely, SSiO2NPs, silicic acid, and Bulk-SiO2 had minimal impact on plant dry weight or physiological responses. These results highlight the importance of nanoparticles architecture in alleviating drought stress and promoting sustainable agriculture.
Collapse
Affiliation(s)
- Lei Wang
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
- Stockbridge School of Agriculture, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Analytical Chemistry, The Connecticut Agricultural Experiment Station, 123 Huntington St., New Haven, Connecticut 06511, United States
| | - Yi Wang
- Department of Analytical Chemistry, The Connecticut Agricultural Experiment Station, 123 Huntington St., New Haven, Connecticut 06511, United States
| | - Chaoyi Deng
- Department of Analytical Chemistry, The Connecticut Agricultural Experiment Station, 123 Huntington St., New Haven, Connecticut 06511, United States
| | - Ian Eggleston
- Stockbridge School of Agriculture, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Shang Gao
- Stockbridge School of Agriculture, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Aoze Li
- Stockbridge School of Agriculture, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Wilanyi R Alvarez Reyes
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kunzheng Cai
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
| | - Rongliang Qiu
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Christy L Haynes
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jason C White
- Department of Analytical Chemistry, The Connecticut Agricultural Experiment Station, 123 Huntington St., New Haven, Connecticut 06511, United States
| | - Baoshan Xing
- Stockbridge School of Agriculture, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
5
|
Xu Y, An D, Zhang T, Wu X, Wang S, Shao J, Qu LL, Guo Y, Dong X. Mitochondrion-Targeted Type I Photodynamic Therapy for Agonist Independent cGAS-STING Activation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418894. [PMID: 39988853 DOI: 10.1002/adma.202418894] [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: 12/03/2024] [Revised: 01/11/2025] [Indexed: 02/25/2025]
Abstract
CGAS-STING agonists generally lead to hyperimmunity and systemic toxicity, hindering their immunotherapeutic outcomes. Herein, a mitochondrion-targeted nanoagonist (termed HABH) containing boron dipyrromethene (BODIPY)-derived type I photosensitizer (BDP) and Au nanoparticle-engineered hollow mesoporous silica (HMSN/AuNPs) has been fabricated for light-controlled mitochondrial stress-inducing and agonist-independent cGAS-STING pathway activation. The HABH nanoagonist can actively target tumor tissues and release the mitochondrion-targeted BDP. Under light illumination, BDP achieves type I photodynamic therapy (PDT) in mitochondria, generating massive hydroxyl radicals (•OH) and inducing mitochondrial stress in an oxygen-independent manner, promoting the release of mitochondrial DNA (mtDNA). Simultaneously, the HMSN/AuNPs act as dual nanozymes to derive cascade reactions for •OH production, elevating the intracellular oxidative state, and together with the BDP-induced mitochondrial stress, finally evoking the cGAS-STING pathway and facilitating the release of type I interferon. In the orthotopic breast tumor models, the HABH nanoagonist achieved intratumoral and systemic immunoactivation for eradicating primary tumors and preventing metastasis tumors. Therefore, the constructed mitochondrion-targeted nanoagonist enabled light-controlled and agonist-independent cGAS-STING activation, providing a paradigm for photoimmunotherapy.
Collapse
Affiliation(s)
- Yin Xu
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
| | - Daokuan An
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
| | - Tian Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xiaochen Wu
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
| | - Shuang Wang
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
| | - Jinjun Shao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Lu-Lu Qu
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
| | - Yuxin Guo
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
| | - Xiaochen Dong
- School of Chemistry & Materials Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou, 221116, China
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| |
Collapse
|
6
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
7
|
Tan H, Wang S, He X, Yang G, Zhu Y, Yang S, Yan S, Gong C, Bai W, Hu Y, Song J, Zheng L. Microneedles Loaded with Nitric-Oxide Driven Nanomotors Improve Force-Induced Efferocytosis Impairment and Sterile Inflammation by Revitalizing Macrophage Energy Metabolism. ACS NANO 2025; 19:9390-9411. [PMID: 40025734 DOI: 10.1021/acsnano.5c01877] [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: 03/04/2025]
Abstract
Mechanical force initiates sterile inflammation, a process implicated in diverse physiological and pathological processes. The timely clearance of apoptotic cells by macrophages via efferocytosis is crucial for the proper resolution of sterile inflammation and for averting excessive tissue damage. Despite this, the specific role and underlying mechanisms of mechanical force on macrophage efferocytosis remain obscure. By integrating bioinformatics and metabolomics analyses, we uncovered how mechanical force disrupts the "arginine metabolism─TCA cycle─mitochondrial function" metabolic cascade, thereby impairing macrophage efferocytosis and intensifying sterile inflammation. Notably, we discovered that elevating l-arginine levels can ameliorate these crises by restoring energy metabolism. Leveraging this insight, we engineered a microneedle drug delivery system loaded with nitric-oxide driven nanomotors (MSN-LA@MNs) for targeted delivery of l-arginine. The active component, MSN-LA, exploits the heightened expression of inducible nitric oxide synthase (iNOS) in force-loaded tissues as a chemoattractant, harnessing NO generated from iNOS-catalyzed l-arginine for autonomous propulsion. In a force-induced rat orthodontic tooth movement (OTM) model, we confirmed that MSN-LA@MNs enhance macrophage efferocytosis and, under iNOS guidance, dynamically modulate sterile inflammation levels in OTM, thus facilitating the OTM process. Collectively, our findings elucidate previously unclear mechanistic links between force, macrophage efferocytosis, and sterile inflammation from a metabolic vantage point, offering a promising targeted strategy for modulating force-related biological processes such as OTM.
Collapse
Affiliation(s)
- Hao Tan
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Shan Wang
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Xinyi He
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Guoyin Yang
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Ye Zhu
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Sihan Yang
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Shengnan Yan
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Chu Gong
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Wenya Bai
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Yun Hu
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Jinlin Song
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| | - Leilei Zheng
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
8
|
Zhang Y, Huang Q, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Wang F. Exploring New Bioorthogonal Catalysts: Scaffold Diversity in Catalysis for Chemical Biology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404431. [PMID: 39921286 PMCID: PMC11884534 DOI: 10.1002/advs.202404431] [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: 04/25/2024] [Revised: 01/11/2025] [Indexed: 02/10/2025]
Abstract
Bioorthogonal catalysis has revolutionized the field of chemical biology by enabling selective and controlled chemical transformations within living systems. Research has converged on the development of innovative catalyst scaffolds, seeking to broaden the scope of bioorthogonal reactions, boost their efficiency, and surpass the limitations of conventional catalysts. This review provides a comprehensive overview of the latest advancements in bioorthogonal catalyst research based on different scaffold materials. Through an in-depth analysis of fabrication strategies and applications of bioorthogonal catalysts, this review discusses the design principles, mechanisms of action, and applications of these novel catalysts in chemical biology. Current challenges and future directions in exploring the scaffold diversity are also highlighted. The integration of diverse catalyst scaffolds offers exciting prospects for precise manipulation of biomolecules and the development of innovative therapeutic strategies in chemical biology. In addition, the review fills in the gaps in previous reviews, such as in fully summarizing the presented scaffold materials applied in bioorthogonal catalysts, emphasizing the potential impact on advancing bioorthogonal chemistry, and offering prospects for future development in this field.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qizhen Huang
- School of Public HealthNantong UniversityNantong226019China
| | - Fang Lei
- School of Public HealthNantong UniversityNantong226019China
| | - Wanlong Qian
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Chengfeng Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qi Wang
- School of Public HealthNantong UniversityNantong226019China
| | - Chaoqun Liu
- School of PharmacyHenan UniversityKaifeng475004China
| | - Haiwei Ji
- School of Public HealthNantong UniversityNantong226019China
| | - Faming Wang
- School of Public HealthNantong UniversityNantong226019China
| |
Collapse
|
9
|
Yu S, Rejinold NS, Choi G, Choy JH. Revolutionizing healthcare: inorganic medicinal nanoarchitectonics for advanced theranostics. NANOSCALE HORIZONS 2025; 10:460-483. [PMID: 39648727 DOI: 10.1039/d4nh00497c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Over the last two decades, advancements in nanomaterials and nanoscience have paved the path for the emergence of nano-medical convergence science, significantly impacting healthcare. In our review, we highlight how these advancements are applied in various biomedical technologies such as drug delivery systems, bio-imaging for diagnostic and therapeutic purposes. Recently, novel inorganic nanohybrid drugs have been developed, combining multifunctional inorganic nanomaterials with therapeutic agents (known as inorganic medicinal nanoarchitectonics). These innovative drugs are actively utilized in cutting-edge medical treatments, including targeted anti-cancer therapy, photo and radiation therapy, and immunotherapy. This review provides a detailed overview of the current development status of inorganic medicinal nanoarchitectonics and explores potential future directions in their advancements.
Collapse
Affiliation(s)
- Seungjin Yu
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - N Sanoj Rejinold
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
| | - Goeun Choi
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Ho Choy
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Division of Natural Sciences, The National Academy of Sciences, Seoul 06579, Republic of Korea
- Tokyo Tech Tokyo Tech World Research Hub Initiative (WRHI), Institute of Innovative Research, Institute of Science Tokyo, Yokohama 226853, Japan
| |
Collapse
|
10
|
Du H, Wang Z, Long S, Li Y, Yang D. The advancement of nanosystems for drug delivery in the prevention and treatment of dental caries. Front Cell Infect Microbiol 2025; 15:1546816. [PMID: 40007606 PMCID: PMC11850577 DOI: 10.3389/fcimb.2025.1546816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
The dental caries remains a globally prevalent disease. Although its incidence has decrease due to enhancements in sanitation policies and public health measures, the treatment and prevention of dental caries still pose significant challenges. Within the oral cavity, traditional drug delivery systems suffer from limitation such as inadequate tissue penetration, short duration of action at target site, and low specificity, which minimally affect the prevention and treatment of dental caries. Consequently, nanosystem for drug delivery, offering enhanced drug stability, solubility, and bio-availability while reducing side effects, garnering attention increasing attention in the fight against dental caries. Therefore, this review examines the role of nanosystems for drug delivery in combating dental caries by inhibiting bacteria survival, biofilm formation, demineralization, and promoting remineralization, and exploring their potential to become the mainstream means of prevention and treatment of dental caries in future.
Collapse
Affiliation(s)
- Han Du
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Zheng Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Shenglan Long
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Yiding Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Deqin Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Department of Conservative Dentistry and Endodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Yuan Y, Lin Q, Feng HY, Zhang Y, Lai X, Zhu MH, Wang J, Shi J, Huang Y, Zhang L, Lu Q, Yuan Z, Lovell JF, Chen HZ, Sun P, Fang C. A multistage drug delivery approach for colorectal primary tumors and lymph node metastases. Nat Commun 2025; 16:1439. [PMID: 39920155 PMCID: PMC11806101 DOI: 10.1038/s41467-025-56768-z] [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: 01/26/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
The presence of lymph node (LN) metastases guides cancer staging and worsens prognoses. Incomplete lymphadenectomy of metastatic LNs may end up with disease recurrence, while excessive resection can result in increased postoperative complications with even no survival benefit. Thus, effective non-invasive methods to treat metastatic LNs would be highly desirable. Here, we develop an enzyme-responsive formulation of small-sized doxorubicin-loaded mesoporous silica nanoparticles (DMSN, 40 nm) encapsulated in nanoliposomes (DMSN@Pla-Lipo, 160 nm). The liposomal membrane contains 1,2-dipalmitoyl-sn-glycero-3-phospho-rac-(1-glycerol) (DPPG) and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), two phospholipids sensitive to secreted phospholipase A2 in human colorectal tumors. In an orthotopic colorectal murine tumor model, phospholipase-induced membrane permeabilization triggers the liberation of DMSN from liposomes for enhanced tumor penetration, conferring an enhanced suppression for the primary tumor. Furthermore, through translocation into metastatic LNs via tumor lymphatics, metastatic tumor cells in LNs are eradicated. Metastases to other major organs are also suppressed, which can be ascribed to the inhibition of colorectal cancer metastasis-associated TGF-β, Wnt, and Hippo signaling pathways in metastatic LNs. The treatment confers an 80% 90-day survival rate in this aggressive tumor model. Taken together, this study demonstrates a deliberate treatment approach for management of both primary tumors and metastatic LNs through multistage drug delivery.
Collapse
Affiliation(s)
- Yihang Yuan
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Quanjun Lin
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Hai-Yi Feng
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yunpeng Zhang
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Jue Wang
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Jiangpei Shi
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yanhu Huang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Lele Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Zeli Yuan
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China.
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China.
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
12
|
Ge H, Wang X, Bai S, Bi Y, Liu F, Sun J, Fu W, Xu D. Dual pH- and Temperature-Responsive Performance and Cytotoxicity of N-Isopropylacrylamide and Acrylic Acid Functionalized Bimodal Mesoporous Silicas with Core-Shell Structure and Fluorescent Feature for Hela Cell. Pharmaceutics 2025; 17:206. [PMID: 40006572 PMCID: PMC11859581 DOI: 10.3390/pharmaceutics17020206] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Polymer-coated mesoporous silica nanoparticles have attracted immense research interest in stimuli-responsive drug delivery systems due to their drug-releasing ability on demand at specific sites in response to external or internal signals. However, the relationships between the coated-copolymer encapsulation and drug delivery performance in the hybrid nanocomposites was rarely reported. Therefore, the main objectives of the present work are to explore the cell uptake, cellular internalization, cytotoxicity, and hemolysis performance of the fluorescent hybrid materials with different polymer-encapsulated amounts. Methods: Using (2-(2-aminoethyl)-6-(dimethylamino)-1H-benzo[de]isoquinoline-1,3(2H)-dione)-doped poly[(N-isopropylacrylamide)-co-(acrylic acid)] (PAN) as a shell and bimodal mesoporous silicas (BMMs) as a core, the dual pH- and temperature-responsive mesoporous PAN@M-BMMs with the fluorescent performances were synthesized via a radical polymerization approach. The effects of the PAN-coated thicknesses on their physicochemical properties and structural features were demonstrated via XRD and SAXS patterns, SEM and TEM images, FT-IR spectra, and TG analysis. Their mass fractal (Dm) evolutions were elucidated on the basis of the SAXS patterns and fluorescence spectra. Results: The Dm values increased from 2.74 to 2.87 with an increase of the PAN-coated amount from 17 to 26.5% along with the particle size from 76.1 to 85.6 nm and blue-shifting of their fluorescent emission wavelength from 470 to 444 nm. Meanwhile, the PAN@M-BMMs exhibited a high ibuprofen (IBU) loading capacity (13.8%) and strong dual pH-/temperature-responsive drug-releasing performances (83.1%) at pH 7.4 and 25 °C, as comparison with that (17.9%) at pH 2.0 and 37 °C. The simulated results confirmed that the adsorption energy decreased from -67.18 kJ/mol for pure BMMs to -116.76 kJ/mol for PAN@M-BMMs, indicating the PAN-grafting on the surfaces of the BMMs core was beneficial to improve its IBU-adsorption capacity. Its uptake in the HeLa cell line was performed via microplate readers, confocal microscopy, flow cytometry, and ICP measurement, showing a low cytotoxicity at a concentration up to 100 µg/mL. Specially, P0.2AN@M-BMMs had a superior cellular uptake and fluorescence properties via the time-dependent uptake experiments, and exhibited the highest silicon content via the cellular internalization analysis, as compared to other carriers. Hemolysis tests confirmed the hemolysis rates below 5%. Conclusions: These demonstrations verified that PAN@M-BMMs should be a promising biomedical application prospect.
Collapse
Affiliation(s)
- Huijie Ge
- Beijing Key Laboratory for Green Catalysis and Separation, Institute of Matter Science, Beijing University of Technology, Beijing 100124, China; (H.G.); (X.W.); (S.B.); (Y.B.); (F.L.)
| | - Xiaoli Wang
- Beijing Key Laboratory for Green Catalysis and Separation, Institute of Matter Science, Beijing University of Technology, Beijing 100124, China; (H.G.); (X.W.); (S.B.); (Y.B.); (F.L.)
| | - Shiyang Bai
- Beijing Key Laboratory for Green Catalysis and Separation, Institute of Matter Science, Beijing University of Technology, Beijing 100124, China; (H.G.); (X.W.); (S.B.); (Y.B.); (F.L.)
| | - Yuhua Bi
- Beijing Key Laboratory for Green Catalysis and Separation, Institute of Matter Science, Beijing University of Technology, Beijing 100124, China; (H.G.); (X.W.); (S.B.); (Y.B.); (F.L.)
| | - Fei Liu
- Beijing Key Laboratory for Green Catalysis and Separation, Institute of Matter Science, Beijing University of Technology, Beijing 100124, China; (H.G.); (X.W.); (S.B.); (Y.B.); (F.L.)
| | - Jihong Sun
- Beijing Key Laboratory for Green Catalysis and Separation, Institute of Matter Science, Beijing University of Technology, Beijing 100124, China; (H.G.); (X.W.); (S.B.); (Y.B.); (F.L.)
| | - Wenliang Fu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China;
| | - Donggang Xu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China;
| |
Collapse
|
13
|
Xu L, Xiong W, Zhang S, Pan J, Liu Y, Liu Q, Wu H, Li L, Zhu JJ, Zheng F. Near-Infrared Photothermally Assisted Nanoprobes Boost Signal Amplification for Fluorescence Imaging and Urinalysis of Tumor. Anal Chem 2025; 97:2463-2471. [PMID: 39865715 DOI: 10.1021/acs.analchem.4c06166] [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: 01/28/2025]
Abstract
Early diagnosis of tumors allows effective treatment of primary cancers through localized therapeutic interventions. However, developing diagnostic tools for sensitive, simple, and early tumor (especially less than 2 mm in diameter) detection remains a challenge. Herein, we presented a biomarker-activatable nanoprobe that enabled a near-infrared (NIR) photothermally amplified signal for fluorescence imaging and urinalysis of tumor. This activatable nanoprobe was constructed by encapsulating renal-clearable NIR ZW800 dyes into a CuS@mSiO2 core-shell nanoparticle with hyaluronic acid. The nanoprobes could accumulate at the tumor site and respond to tumor-associated hyaluronidase, undergoing in situ enzyme-catalyzed decapsulation to release renal-clearable ZW800 dyes for fluorescence imaging and uranalysis of tumor in living mice. Notedly, with the aid of NIR laser irradiation, the photothermal effect of CuS core boosted signal amplification for tumor diagnosis via photothermally enhanced ZW800 release, providing high specificity and sensitivity. On account of the biomarker specificity and the spatiotemporally controlled NIR irradiation, the nanoprobes could selectively activate their NIR fluorescence (NIRF) signal to visualize tumors in living mice and allow for the easy translation of the nanoprobe as artificial urinary biomarker probes for in vitro diagnosis of tumor progression. In 4T1 tumor-bearing mice models, the activatable nanoprobes enabled ultrasensitive detection of tumors (1.9 mm in diameter). This study offers a noninvasive and photothermally signal-amplified approach for early tumor diagnosis via NIRF imaging or simple urine tests.
Collapse
Affiliation(s)
- Limei Xu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Weiwei Xiong
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Shiling Zhang
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Jiajia Pan
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Yingqi Liu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Qiulin Liu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Haojie Wu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Lingling Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Xianlin Ave 163, Nanjing, Jiangsu 210023, China
| | - Fenfen Zheng
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| |
Collapse
|
14
|
Wang X, Guo X, Ren H, Song X, Chen L, Yu L, Ren J, Chen Y. An "Outer Piezoelectric and Inner Epigenetic" Logic-Gated PANoptosis for Osteosarcoma Sono-Immunotherapy and Bone Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415814. [PMID: 39726343 DOI: 10.1002/adma.202415814] [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: 10/16/2024] [Indexed: 12/28/2024]
Abstract
The precise manipulation of PANoptosis, a newly defined cell death pathway encompassing pyroptosis, apoptosis, and necroptosis, is highly desired to achieve safer cancer immunotherapy with tumor-specific inflammatory responses and minimal side effects. Nonetheless, this objective remains a formidable challenge. Herein, an "AND" logic-gated strategy for accurately localized PANoptosis activation, utilizing composite 3D-printed bioactive glasses scaffolds integrated with epigenetic regulator-loaded porous piezoelectric SrTiO3 nanoparticles is proposed. The "logic-gated" strategy is co-programmed by an "outer" input signal of exogenous ultrasound irradiation to produce reactive oxygen species and an "inner" input signal of acid tumor microenvironment to ensure the epigenetic demethylation regulation, guaranteeing the tumor-specific PANoptosis. Specifically, immunogenic PANoptosis triggers dendritic cell maturation and cytotoxic T cell activation, amplifying antitumor immune responses and significantly suppressing osteosarcoma growth, with a suppression rate of ≈73.47 ± 5.2%. In addition, the well-known bioactivities of Sr-doped scaffolds expedite osteogenic differentiation and reinforce bone regeneration. Therefore, this work provides a paradigm of logic-gated sono-piezoelectric biomaterial platform with concurrently exogenous/endogenous activated PANoptosis for controlled sono-immunotherapy of osteosarcoma, and related bone defects repair.
Collapse
Affiliation(s)
- Xiaoting Wang
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, 400010, P. R. China
| | - Xun Guo
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, 400010, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Luodan Yu
- Department of Radiology, Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jianli Ren
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, 400010, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
15
|
Preedanorawut P, Chatchawankanphanich O, Yiamsawas D, Crespy D. Controlled Release of Hydrophilic Drug from Hollow Nanodots. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409112. [PMID: 39888222 DOI: 10.1002/smll.202409112] [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: 10/04/2024] [Revised: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Here the challenge of limited encapsulation efficiency of ionizable hydrophilic molecules in silica materials is addressed. Two effective strategies are showcased that allow high encapsulation efficiency of salicylic acid, while simultaneously maintaining the morphology and particle size of silica nanocapsules. These promising approaches involve the formation and encapsulation of a prodrug or the complexation of the hydrophilic payload with a hydrophobic moiety to form a complex that is dissociated in acidic conditions. Well-defined core-shell silica nanocapsules with a diameter of 6 nm are obtained and exhibited an encapsulation efficiency of over 90%. High amounts of salicylic acid are released in acidic conditions from silica nanocapsules entrapping the prodrug or the complex, leading to pH-responsive characteristics. This work demonstrates promising strategies for the encapsulation and the controlled release of hydrophilic fertilizers, pesticides or drugs.
Collapse
Affiliation(s)
- Patitta Preedanorawut
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| | - Orawan Chatchawankanphanich
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Doungporn Yiamsawas
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Daniel Crespy
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| |
Collapse
|
16
|
Spada A, Gerber-Lemaire S. Surface Functionalization of Nanocarriers with Anti-EGFR Ligands for Cancer Active Targeting. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:158. [PMID: 39940134 PMCID: PMC11820047 DOI: 10.3390/nano15030158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/14/2025]
Abstract
Active cancer targeting consists of the selective recognition of overexpressed biomarkers on cancer cell surfaces or within the tumor microenvironment, enabled by ligands conjugated to drug carriers. Nanoparticle (NP)-based systems are highly relevant for such an approach due to their large surface area which is amenable to a variety of chemical modifications. Over the past decades, several studies have debated the efficiency of passive targeting, highlighting active targeting as a more specific and selective approach. The choice of conjugation chemistry for attaching ligands to nanocarriers is critical to ensure a stable and robust system. Among the panel of cancer biomarkers, the epidermal growth factor receptor (EGFR) stands as one of the most frequently overexpressed receptors in different cancer types. The design and development of nanocarriers with surface-bound anti-EGFR ligands are vital for targeted therapy, relying on their facilitated capture by EGFR-overexpressing tumor cells and enabling receptor-mediated endocytosis to improve drug accumulation within the tumor microenvironment. In this review, we examine several examples of the most recent and significant anti-EGFR nanocarriers and explore the various conjugation strategies for NP functionalization with anti-EGFR biomolecules and small molecular ligands. In addition, we also describe some of the most common characterization techniques to confirm and analyze the conjugation patterns.
Collapse
Affiliation(s)
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
17
|
Qian S, Wang Y, Fan J, Kong T, Wang Y, Wang K, Liao Y, Wang L, Zheng J. pH-Sensitive oligopeptide magnetic mesoporous silica beads for deoxyribonucleic acid extraction. NANOSCALE ADVANCES 2025:d4na00987h. [PMID: 39898280 PMCID: PMC11780327 DOI: 10.1039/d4na00987h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Exploring novel synthesis strategies for magnetic beads to extract nucleic acids is of great significance in the field of in vitro diagnostics. In the present research, monodisperse magnetic mesoporous silica beads were synthesized via the thermolysis reaction of Fe(acac)3 by using large-pore dendritic silica colloids as templates, and were further functionalized with a highly pH-sensitive histidine-glutamate co-oligopeptide for deoxyribonucleic acid extraction. The large-pore dendritic silica colloid scaffolds were utilized for high-density incorporation of superparamagnetic iron oxide nanoparticles within the vertical channels. The morphology and properties of the as-prepared pH-sensitive oligopeptide magnetic mesoporous silica beads were evaluated by transmission electron microscopy, scanning electron microscopy, vibrating sample magnetometry, X-ray photoelectron spectroscopy, X-ray diffraction testing and so on. The average size of the obtained magnetic beads was 370 nm in diameter with a narrow size distribution. The saturation magnetization and magnetic content of the resultant magnetic beads were 25 emu g-1 and 59%, respectively. Moreover, the magnetic mesoporous silica beads exhibited an obvious pH-responsive behavior. Due to these remarkable features, successful deoxyribonucleic acid capture using the as-prepared pH-sensitive oligopeptide magnetic mesoporous silica beads was achieved.
Collapse
Affiliation(s)
- Sihua Qian
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| | - Yiting Wang
- College of Chemistry, Jilin Normal University Siping 136000 P. R. China
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| | - Junjie Fan
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| | - Tong Kong
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| | - Yuhui Wang
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| | - Kaizhe Wang
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| | - Yufeng Liao
- Department of Clinical Laboratory Ningbo No. 2 Hospital Ningbo 315010 P.R. China
| | - Li Wang
- College of Chemistry, Jilin Normal University Siping 136000 P. R. China
| | - Jianping Zheng
- Ningbo Cixi Institute of Biomedical Engineering, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences Ningbo 315300 P. R. China
| |
Collapse
|
18
|
Mundada AB, Pradhan P, Raju R, Sujitha YS, Kulkarni PA, Mundada PA, Tiwari R, Sharma P. Molecular dynamics in pharmaceutical nanotechnology: simulating interactions and advancing applications. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-27. [PMID: 39786352 DOI: 10.1080/09205063.2025.2450150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Molecular Dynamics (MD) simulations are now widely utilized in pharmaceutical nanotechnology to gain deeper understanding of nanoscale processes imperative to drug design. This review has also detailed how MD simulation can be employed in the study of drug-nanocarrier interactions, controlling release of chemical compounds from drug delivery systems and increasing solubility and bioavailability of nanocarriers. Furthermore, MD contributes to examining the drug delivery systems, measuring the toxic effects, and determining biocompatibility of nanomedical systems. With the incorporation of artificial intelligence and the use of hybrid simulation systems, MD has gone a step ahead to model other niches of biology that make a tremendous opening to develop highly selective nanomedications. Nevertheless, with well-known issues such as computational constraints and the discrepancy between in silico and experiment results, MD remains a work in progress, with considerable promise for replacing or supplementing existing approaches to the development of precision medicine and nanomedicine, the continued progression of healthcare hopeful.
Collapse
Affiliation(s)
- Anand Badrivishal Mundada
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District Dhule, Maharashtra, India
| | - Pankaj Pradhan
- Department of Pharmacy, Swami Keshvanand Institute of Pharmacy, Ramnagaria, Jagatpura, Jaipur, Rajasthan, India
| | - Rajapandi Raju
- Department of Pharmacy, St. John's College of Pharmaceutical Sciences & Research, Kattappana, Kerala, India - Idukki
| | - Y Sarah Sujitha
- Department of Pharmacy, Krishna Teja Pharmacy College, Tirupati, India
| | - Parag Arun Kulkarni
- Department of Pharmaceutics, Shastry Institute of Pharmacy, Erandol, Maharashtra, India
| | - Pooja Anand Mundada
- Department of Pharmacy, R. C. Patel Institute of Pharmacy, Shirpur, District Dhule, Maharashtra, India
| | - Ruchi Tiwari
- Department of Pharmaceutics, PSIT-Pranveer Singh Institute of Technology (Pharmacy), Kanpur, Uttar Pradesh, India
| | - Pankaj Sharma
- Department of Pharmaceutics, ShriRam College of Pharmacy, Banmore, Morena, Madhya Pradesh, India
| |
Collapse
|
19
|
Cai B, Rong X, Sun Y, Liu L, Li Z. Engineered 3D DNA Crystals: A Molecular Design Perspective. SMALL METHODS 2025:e2401455. [PMID: 39777863 DOI: 10.1002/smtd.202401455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Recent advances in biomolecular self-assembly have transformed material science, enabling the creation of novel materials with unparalleled precision and functionality. Among these innovations, 3D DNA crystals have emerged as a distinctive class of macroscopic materials, engineered through the bottom-up approach by DNA self-assembly. These structures uniquely combine precise molecular ordering with high programmability, establishing their importance in advanced material design. This review delves into the molecular design of engineered 3D DNA crystals, classifying current crystal structures based on "crystal bond orientations" and examining key aspects of in-silico molecular design, self-assembly, and crystal modifications. The functionalization of 3D DNA crystals for applications in crystallization scaffolding, biocatalysis, biosensing, electrical and optical devices, as well as in the emerging fields of DNA computing and data storage are explored. Finally, the ongoing challenges are addressed and future directions to advance the field of engineered 3D DNA crystals are proposed.
Collapse
Affiliation(s)
- Baoshuo Cai
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xiao Rong
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yifan Sun
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Longfei Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06484, USA
- Nanobiology Institute, Yale University, West Haven, CT, 06484, USA
| | - Zhe Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
20
|
Gharehbaba AM, Omidi Y, Barar J, Eskandani M, Adibkia K. Synergistic pH-responsive MUC-1 aptamer-conjugated Ag/MSN Janus nanoparticles for targeted chemotherapy, photothermal therapy, and gene therapy in breast cancer. BIOMATERIALS ADVANCES 2025; 166:214081. [PMID: 39454415 DOI: 10.1016/j.bioadv.2024.214081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Drug resistance in cancer treatment, primarily attributed to the overexpression of the multidrug resistance (MDR) gene, significantly hampers the effectiveness of chemotherapy. This mechanism, driven by the increased production of P-glycoprotein (P-gp) efflux pumps, highlights the urgent need for innovative strategies to combat drug resistance in cancer patients. This study explores the application of antisense technology to suppress MDR gene expression, while addressing the challenges of instability and limited cellular uptake associated with antisense oligonucleotides. We synthesized Janus silver-mesoporous silica nanoparticles (Ag/MSN JNPs) using a sol-gel method, characterized by transmission electron microscopy (TEM) and dynamic light scattering (DLS), revealing uniformly sized, dumbbell-shaped nanoparticles with an average size of 285 ± 5.12 nm. Doxorubicin (DOX) was loaded into the porous structure of the mesoporous silica, and JNPs were functionalized with chitosan (CS) to incorporate P-gp antisense and a MUC-1 aptamer, serving as a pH-responsive gatekeeper. Our findings indicate that the Ap-As-DOX-JNPs achieved a remarkable 89 ± 0.59 % cell death in drug-resistant MCF-7/ADR cells after 48 h, alongside an 80 % reduction in P-gp expression. The combination of DOX, antisense technology, and photothermal therapy utilizing these JNPs demonstrates a promising strategy to effectively overcome drug resistance. Notably, normal MCF-7 cells exhibited reduced viability from 39.11 ± 1.12 % to 30.05 ± 1.07 % when treated with DOX-JNPs under near-infrared (NIR) irradiation. These results underscore the potential of utilizing MUC-1 aptamer-conjugated Janus nanoparticles in conjunction with chitosan as a gatekeeper to enhance the efficacy of chemotherapy, photothermal therapy, and gene therapy in overcoming multidrug resistance in cancer treatment.
Collapse
Affiliation(s)
- Adel Mahmoudi Gharehbaba
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Khosro Adibkia
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Mura M, Carucci C, Caddeo E, Sovová Š, Piludu M, Pekař M, Jachimska B, Parsons DF, Salis A. Specific buffer effects on the formation of BSA protein corona around amino-functionalized mesoporous silica nanoparticles. J Colloid Interface Sci 2025; 677:540-547. [PMID: 39106779 DOI: 10.1016/j.jcis.2024.07.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/13/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
The effect of buffer species on biomolecules and biomolecule-nanoparticle interactions is a phenomenon that has been either neglected, or not understood. Here, we study the formation of a BSA protein corona (PC) around amino-functionalized mesoporous silica nanoparticles (MSN-NH2) in the presence of different buffers (Tris, BES, cacodylate, phosphate, and citrate) at the same pH (7.15) and different concentrations (10, 50, and 100 mM). We find that BSA adsorption is buffer specific, with the adsorbed amount of BSA being 4.4 times higher in the presence of 100 mM Tris (184 ± 3 mg/g) than for 100 mM citrate (42 ± 2 mg/g). That is a considerable difference that cannot be explained by conventional theories. The results become clearer if the interaction energies between BSA and MSN-NH2, considering the electric double layer (EEDL) and the van der Waals (EvdW) terms, are evaluated. The buffer specific PC derives from buffer specific zeta potentials that, for MSN-NH2, are positive with Tris and negative with citrate buffers. A reversed sign of zeta potentials can be obtained by considering polarizability-dependent dispersion forces acting together with electrostatics to give the buffer specific outcome. These results are relevant not only to our understanding of the formation of the PC but may also apply to other bio- and nanosystems in biological media.
Collapse
Affiliation(s)
- Monica Mura
- Department of Chemical and Geological Sciences, University of Cagliari &CSGI, Cittadella Universitaria, S.S. 554 bivio Sestu, 09042 Monserrato, CA, Italy
| | - Cristina Carucci
- Department of Chemical and Geological Sciences, University of Cagliari &CSGI, Cittadella Universitaria, S.S. 554 bivio Sestu, 09042 Monserrato, CA, Italy
| | - Elena Caddeo
- Department of Chemical and Geological Sciences, University of Cagliari &CSGI, Cittadella Universitaria, S.S. 554 bivio Sestu, 09042 Monserrato, CA, Italy
| | - Šárka Sovová
- Faculty of Chemistry, Brno University of Technology, Purkynova 464/118, 612 00 Brno, Czechia
| | - Marco Piludu
- Department of Chemical and Geological Sciences, University of Cagliari &CSGI, Cittadella Universitaria, S.S. 554 bivio Sestu, 09042 Monserrato, CA, Italy
| | - Miloslav Pekař
- Faculty of Chemistry, Brno University of Technology, Purkynova 464/118, 612 00 Brno, Czechia
| | - Barbara Jachimska
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Krakow, Poland
| | - Drew F Parsons
- Department of Chemical and Geological Sciences, University of Cagliari &CSGI, Cittadella Universitaria, S.S. 554 bivio Sestu, 09042 Monserrato, CA, Italy
| | - Andrea Salis
- Department of Chemical and Geological Sciences, University of Cagliari &CSGI, Cittadella Universitaria, S.S. 554 bivio Sestu, 09042 Monserrato, CA, Italy.
| |
Collapse
|
22
|
Liang J, Yao L, Liu Z, Chen Y, Lin Y, Tian T. Nanoparticles in Subunit Vaccines: Immunological Foundations, Categories, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407649. [PMID: 39501996 DOI: 10.1002/smll.202407649] [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: 08/28/2024] [Revised: 10/12/2024] [Indexed: 01/11/2025]
Abstract
Subunit vaccines, significant in next-generation vaccine development, offer precise targeting of immune responses by focusing on specific antigens. However, this precision often comes at the cost of eliciting strong and durable immunity, posing a great challenge to vaccine design. To address this limitation, recent advancements in nanoparticles (NPs) are utilized to enhance antigen delivery efficiency and boost vaccine efficacy. This review examines how the physicochemical properties of NPs influence various stages of the immune response during vaccine delivery and analyzes how different NP types contribute to immune activation and enhance vaccine performance. It then explores the unique characteristics and immune activation mechanisms of these NPs, along with their recent advancements, and highlights their application in subunit vaccines targeting infectious diseases and cancer. Finally, it discusses the challenges in NP-based vaccine development and proposes future directions for innovation in this promising field.
Collapse
Affiliation(s)
- Jiale Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Taoran Tian
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
23
|
Yazdan M, Naghib SM. Smart Ultrasound-responsive Polymers for Drug Delivery: An Overview on Advanced Stimuli-sensitive Materials and Techniques. Curr Drug Deliv 2025; 22:283-309. [PMID: 38288800 DOI: 10.2174/0115672018283792240115053302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 04/11/2025]
Abstract
In recent years, a notable advancement has occurred in the domain of drug delivery systems via the integration of intelligent polymers that respond to ultrasound. The implementation of this groundbreaking methodology has significantly revolutionised the controlled and precise delivery of therapeutic interventions. An in-depth investigation is conducted into the most recent developments in ultrasonic stimulus-responsive materials and techniques for the purpose of accomplishing precise medication administration. The investigation begins with an exhaustive synopsis of the foundational principles underlying drug delivery systems that react to ultrasonic stimuli, focusing specifically on the complex interplay between polymers and ultrasound waves. Significant attention is devoted to the development of polymers that demonstrate tailored responsiveness to ultrasound, thereby exemplifying their versatility in generating controlled drug release patterns. Numerous classifications of intelligent polymers are examined in the discussion, including those that react to variations in temperature, pH, and enzymes. When coupled with ultrasonic stimuli, these polymers offer a sophisticated framework for the precise manipulation of drug release in terms of both temporal and spatial dimensions. The present study aims to examine the synergistic effects of responsive polymers and ultrasound in overcoming biological barriers such as the blood-brain barrier and the gastrointestinal tract. By doing so, it seeks to shed light on the potential applications of these materials in intricate clinical scenarios. The issues and future prospects of intelligent ultrasound-responsive polymers in the context of drug delivery are critically analysed in this article. The objective of this study is to offer valuable perspectives on the challenges that must be overcome to enable the effective implementation of these technologies. The primary objective of this comprehensive review is to furnish researchers, clinicians, and pharmaceutical scientists with a wealth of information that will serve as a guide for forthcoming developments in the development and enhancement of intelligent drug delivery systems that employ ultrasound-responsive polymers to attain superior therapeutic outcomes.
Collapse
Affiliation(s)
- Mostafa Yazdan
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| |
Collapse
|
24
|
Pallathadka H, Jabir M, Rasool KH, Hanumanthaiah M, Sharma N, Pramanik A, Rab SO, Jawad SF, Oghenemaro EF, Mustafa YF. siRNA-based therapy for overcoming drug resistance in human solid tumours; molecular and immunological approaches. Hum Immunol 2025; 86:111221. [PMID: 39700968 DOI: 10.1016/j.humimm.2024.111221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
RNA interference (RNAi) is a primordial biological process that protects against external intrusion. SiRNA has the potential to selectively silence disease-related genes in a sequence-specific way, thus offering a promising therapeutic approach. The efficacy of siRNA-based therapies in cancer treatment has gained significant recognition due to multiple studies demonstrating its ability to effectively suppress cancer cells' growth and multiplication. Moreover, siRNA-based medicines have shown considerable promise in enhancing the sensitivity of cancer cells to chemotherapy and other treatment methods by suppressing genes that play a role in the development of drug resistance. Exploring and identifying functional genes linked to cancer cell characteristics and drug resistance is crucial for developing effective siRNAs for cancer treatment and advancing targeted and personalized therapeutics. Targeting and silencing genes in charge of resistance mechanisms, such as those involved in drug efflux, cell survival, or DNA repair, is possible with siRNA therapy in the context of drug resistance, especially cancer. Through inhibiting these genes, siRNA therapy can prevent resistance and restore the efficacy of traditional medications. This review addresses the potential of siRNAs in addressing drug resistance in human tumours, opening up new possibilities in cancer therapy. This review article offers a non-systematic summary of how different siRNA types contribute to cancer cells' treatment resistance. Using pertinent keywords, sources were chosen from reliable databases, including PubMed, Scopus, and Google Scholar. The review covered essential papers in this area and those that mainly addressed the function of siRNA in drug resistance. The articles examined in connection with the title of this review were primarily published from 2020 onward and are based on in vitro studies. Furthermore, this article examines the potential barriers and prospective perspectives of siRNA therapies.
Collapse
Affiliation(s)
| | - Majid Jabir
- Department of Applied Sciences, University of Technology, Iraq
| | | | - Malathi Hanumanthaiah
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri - 140307, Mohali, Punjab, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001 Babil, Iraq.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Delta State University, Faculty of Pharmacy, PMB 1 Abraka, Delta State, Nigeria
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
25
|
Farjadian F, Parsi F, Heidari R, Zarkesh K, Mohammadi HR, Mohammadi-Samani S, Tayebi L. Mesoporous Silica Administration as a New Strategy in the Management of Warfarin Toxicity: An In-Vitro and In-Vivo Study. Adv Pharm Bull 2024; 14:883-891. [PMID: 40190678 PMCID: PMC11970487 DOI: 10.34172/apb.42665] [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: 01/14/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose Warfarin is one of the most widely used anticoagulants that functions by inhibiting vitamin K epoxide reductase. Warfarin overdose, whether intentional or unintentional, can cause life-threatening bleeding. Here, we present a novel warfarin adsorbent based on mesoporous silica that could serve as an antidote to warfarin toxicity. Methods Amino-functionalized mesoporous silica (MS-NH2) was synthesized based on the co-condensation method through a soft template technique followed by template removal. The prepared structure and functional group were studied by Fourier transform infrared spectroscopy (FT-IR), and X-ray diffraction (XRD). Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) checked the morphology. The capacity of MS-NH2 in the adsorption of warfarin was evaluated in vitro, at pH=7.4 and pH=1.2. In vivo evaluations were performed in control and warfarin-overdosed animal models. Overdosed animals were treated with MS-NH2 by oral gavage. Biomarkers of organ injury were assessed in animal serum. Results The MS-NH2 was relatively uniform, spherical with defined diameters (400 nm) and porous structure. Synthesized particles had a large surface area (1015 m2 g-1) and mean pore diameter of 2.4 nm which led to considerable adsorption capacity for warfarin 1666 mg/g. In vivo studies revealed that oral administration of MS-NH2 in mice poisoned with warfarin caused a significant difference (P<0.05) in the International Normalized Ratio (INR) and prothrombin time (PT). Moreover, the warfarin with MS-NH2 group demonstrated a notable decrease in biomarkers associated with tissue damage, such as bilirubin, lactate dehydrogenase (LDH), alanine aminotransferase (ALT), and aspartate aminotransferase (AST). Conclusion The results confirm that MS-NH2 administration can be an effective treatment for warfarin toxicity and could potentially mitigate the adverse effects of warfarin poisoning.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Parsi
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khatereh Zarkesh
- Department of Pharmaceutics, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hamid Reza Mohammadi
- Department of Toxicology, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, USA
- Institute for Engineering in Medicine, Health & Human Performance (EnMed), Batten College of Engineering and Technology, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
26
|
Zhang D, Wei W, Xie T, Zhou X, He X, Qiao J, Guo R, Jin G, Li N. Magnetic Nanocarriers for pH/GSH/NIR Triple-Responsive Drug Release and Synergistic Therapy in Tumor Cells. ACS OMEGA 2024; 9:49749-49758. [PMID: 39713612 PMCID: PMC11656227 DOI: 10.1021/acsomega.4c08267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
In this study, the mesoporous Fe3O4 nanodrug carriers containing disulfide bonds (CHO-SMNPs) were successfully synthesized and characterized. Doxorubicin (DOX) was loaded onto the CHO-SMNPs as a model drug and gatekeeper through the formation of imine bonds with the aldehyde groups on the surface of the mesoporous materials. This drug carrier demonstrates effective drug release triggered by pH, glutathione (GSH), and near-infrared (NIR) light, along with satisfactory photothermal conversion efficiency under NIR irradiation at 808 nm. Furthermore, CHO-SMNPs exhibit excellent blood compatibility and biodegradability. They also show good biocompatibility and efficient cellular uptake in HeLa and MCF-7 cancer cells. Most importantly, the CHO-SMNPs/DOX has shown significant effectiveness in killing both HeLa and MCF-7 cancer cells. Consequently, CHO-SMNPs/DOX presents substantial potential as a magnetic-targeted, pH/GSH/NIR triple-triggered drug delivery system for synergistic chemo-photothermal therapy in tumor treatment.
Collapse
Affiliation(s)
- Di Zhang
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| | - Wanyu Wei
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| | - Tianxiang Xie
- School
of Stomatology, Shanxi Medical University, Taiyuan 030001, China
| | - Xue Zhou
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| | - Xu He
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| | - Jie Qiao
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| | - Rui Guo
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| | - Gang Jin
- Department
of Medical Oncology, Second Hospital of
Shanxi Medical University, Taiyuan 030001, China
| | - Ningbo Li
- School
of Basic Medical Sciences, Shanxi Medical
University, Taiyuan 030001, China
| |
Collapse
|
27
|
Biswas P, Livni N, Paul D, Aram L, Safadi R, Varsano N, Elad N, Kamyshinsky R, Leskes M, Gal A. A pH-Dependent Phase Separation Drives Polyamine-Mediated Silicification from Undersaturated Solutions. ACS NANO 2024; 18:33998-34006. [PMID: 39641753 DOI: 10.1021/acsnano.4c08707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Silica polymerization from its soluble monomers is fundamental to many chemical processes. Although industrial methods require harsh conditions and concentrated precursors, biological silica precipitation occurs under ambient conditions from dilute solutions. The hallmark of biosilica is the presence of amine-rich organic macromolecules, but their functional role remains elusive. Here, we show a pH-dependent stimulatory effect of such polyamines on silica polymerization. Notably, this process is decoupled from the saturation degree, allowing the synthesis of polymer-silica hybrid products with controlled network morphologies from undersaturated solutions. The data suggest a two-step phase separation process. First, an associative liquid-liquid phase separation forms a micrometer-size dense phase. Second, silica undergoes a liquid-to-solid transition in the supersaturated condensates to form a bicontinuous silica structure. This study can inspire "soft chemistry" routes to design organic-inorganic nanomaterials with regulatory principles optimized by evolution.
Collapse
Affiliation(s)
- Protap Biswas
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nitzan Livni
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Debojit Paul
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lior Aram
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Razi Safadi
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Neta Varsano
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nadav Elad
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Roman Kamyshinsky
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Leskes
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Assaf Gal
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
28
|
Rocío Hernández A, Bogdanova E, Campos Pacheco JE, Kocherbitov V, Ekström M, Pilkington G, Valetti S. Disordered mesoporous silica particles: an emerging platform to deliver proteins to the lungs. Drug Deliv 2024; 31:2381340. [PMID: 39041383 PMCID: PMC11268259 DOI: 10.1080/10717544.2024.2381340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
Pulmonary delivery and formulation of biologics are among the more complex and growing scientific topics in drug delivery. We herein developed a dry powder formulation using disordered mesoporous silica particles (MSP) as the sole excipient and lysozyme, the most abundant antimicrobial proteins in the airways, as model protein. The MSP had the optimal size for lung deposition (2.43 ± 0.13 µm). A maximum lysozyme loading capacity (0.35 mg/mg) was achieved in 150 mM PBS, which was seven times greater than that in water. After washing and freeze-drying, we obtained a dry powder consisting of spherical, non-aggregated particles, free from residual buffer, or unabsorbed lysozyme. The presence of lysozyme was confirmed by TGA and FT-IR, while N2 adsorption/desorption and SAXS analysis indicate that the protein is confined within the internal mesoporous structure. The dry powder exhibited excellent aerodynamic performance (fine particle fraction <5 µm of 70.32%). Lysozyme was released in simulated lung fluid in a sustained kinetics and maintaining high enzymatic activity (71-91%), whereas LYS-MSP were shown to degrade into aggregated nanoparticulate microstructures, reaching almost complete dissolution (93%) within 24 h. MSPs were nontoxic to in vitro lung epithelium. The study demonstrates disordered MSP as viable carriers to successfully deliver protein to the lungs, with high deposition and retained activity.
Collapse
Affiliation(s)
- Aura Rocío Hernández
- Biofilms – Research Center for Biointerfaces (BRCB), Malmö, Sweden
- Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Ekaterina Bogdanova
- Biofilms – Research Center for Biointerfaces (BRCB), Malmö, Sweden
- Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Jesus E. Campos Pacheco
- Biofilms – Research Center for Biointerfaces (BRCB), Malmö, Sweden
- Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Vitaly Kocherbitov
- Biofilms – Research Center for Biointerfaces (BRCB), Malmö, Sweden
- Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | | | | | - Sabrina Valetti
- Biofilms – Research Center for Biointerfaces (BRCB), Malmö, Sweden
- Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| |
Collapse
|
29
|
Pablos JL, Lozano D, Manzano M, Vallet-Regí M. Regenerative medicine: Hydrogels and mesoporous silica nanoparticles. Mater Today Bio 2024; 29:101342. [PMID: 39649249 PMCID: PMC11625165 DOI: 10.1016/j.mtbio.2024.101342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 12/10/2024] Open
Abstract
Hydrogels, that are crosslinked polymer networks, can absorb huge quantities of water and/or biological fluids. Their physical properties, such as elasticity and soft tissue, together with their biocompatibility and biodegradability, closely resemble living tissues. The versatility of hydrogels has fuelled their application in various fields, such as agriculture, biomaterials, the food industry, drug delivery, tissue engineering, and regenerative medicine. Their combination with nanoparticles, specifically with Mesoporous Silica Nanoparticles (MSNs), have elevated these composites to the next level, since MSNs could improve the hydrogel mechanical properties, their ability to encapsulate and controlled release great amounts of different therapeutic agents, and their responsiveness to a variety of external and internal stimuli. In this review, the main features of both MSNs and hydrogels are introduced, followed by the discussion of different hydrogels-MSNs structures and an overview of their use in different applications, such as drug delivery technologies and tissue engineering.
Collapse
Affiliation(s)
- Jesús L. Pablos
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
| | - Daniel Lozano
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
- Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| | - Miguel Manzano
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
- Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
- Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| |
Collapse
|
30
|
Yan J, Siwakoti P, Shaw S, Bose S, Kokil G, Kumeria T. Porous silicon and silica carriers for delivery of peptide therapeutics. Drug Deliv Transl Res 2024; 14:3549-3567. [PMID: 38819767 PMCID: PMC11499345 DOI: 10.1007/s13346-024-01609-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
Peptides have gained tremendous popularity as biological therapeutic agents in recent years due to their favourable specificity, diversity of targets, well-established screening methods, ease of production, and lower cost. However, their poor physiological and storage stability, pharmacokinetics, and fast clearance have limited their clinical translation. Novel nanocarrier-based strategies have shown promise in overcoming these issues. In this direction, porous silicon (pSi) and mesoporous silica nanoparticles (MSNs) have been widely explored as potential carriers for the delivery of peptide therapeutics. These materials possess several advantages, including large surface areas, tunable pore sizes, and adjustable pore architectures, which make them attractive carriers for peptide delivery systems. In this review, we cover pSi and MSNs as drug carriers focusing on their use in peptide delivery. The review provides a brief overview of their fabrication, surface modification, and interesting properties that make them ideal peptide drug carriers. The review provides a systematic account of various studies that have utilised these unique porous carriers for peptide delivery describing significant in vitro and in vivo results. We have also provided a critical comparison of the two carriers in terms of their physicochemical properties and short-term and long-term biocompatibility. Lastly, we have concluded the review with our opinion of this field and identified key areas for future research for clinical translation of pSi and MSN-based peptide therapeutic formulations.
Collapse
Affiliation(s)
- Jiachen Yan
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Prakriti Siwakoti
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Siuli Shaw
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Sudeep Bose
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Ganesh Kokil
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
31
|
Chen S, Lai J, Chen J, Zheng L, Wang M. 3D printed gelatin/PTMC core/shell scaffolds with NIR laser-tuned drug/biomolecule release for cancer therapy and uterine regeneration. Int J Biol Macromol 2024; 283:137193. [PMID: 39500434 DOI: 10.1016/j.ijbiomac.2024.137193] [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: 09/01/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/10/2024]
Abstract
Surgical resection is an efficient treatment for cancerous tissues and uterine fibroids in the women uterus. However, the insufficiency of clinical interventions could result in tumor recurrence, and the defective tissues remained would cause intrauterine adhesions (IUAs) and further affect reproduction capacity. In this study, 3D printed hydrogel/poly(l-lactide-co-trimethylene carbonate) (PLLA-co-TMC, "PTMC" in short) core/shell scaffolds with NIR-tuned doxorubicin hydrochloride (DOX) and estradiol (E2) dual release were designed and fabricated for cancer therapy and uterine regeneration. Gelatin (Gel) and DOX were homogeneously mixed and then 3D printed to form Gel-DOX scaffolds. Gel-DOX scaffolds were then immersed in PTMC-PDA@E2 solution to fabricate Gel-DOX/PTMC-PDA@E2 core/shell scaffolds. Consequently, Gel-DOX/PTMC-PDA@E2 scaffolds could release DOX and E2 in a chronological manner, firstly delivering DOX assisted by phototherapy (PTT) to effectively kill Hela cells and then sustainably releasing E2 to promote uterine tissue regeneration. In vitro experiments showed that core/shell scaffolds exhibited excellent anticancer efficiency through the synergy of DOX release and hyperthermia ablation. Moreover, E2 could be sustainably released for over 28 days in vitro to promote the proliferation of bone marrow-derived mesenchymal stem cells (BMSCs). The novel Gel-DOX/PTMC-PDA@E2 core/shell scaffolds have therefore exhibited potential promise for the treatment of cancer therapy and uterine regeneration.
Collapse
Affiliation(s)
- Shangsi Chen
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China
| | - Jiahui Lai
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China
| | - Jizhuo Chen
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China
| | - Liwu Zheng
- Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong Special Administrative Region of China
| | - Min Wang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China.
| |
Collapse
|
32
|
He L, Javid Anbardan Z, Habibovic P, van Rijt S. Doxorubicin- and Selenium-Incorporated Mesoporous Silica Nanoparticles as a Combination Therapy for Osteosarcoma. ACS APPLIED NANO MATERIALS 2024; 7:25400-25411. [PMID: 39606122 PMCID: PMC11590048 DOI: 10.1021/acsanm.4c04294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024]
Abstract
Doxorubicin (Dox) is a promising anticancer chemotherapeutic, which has been widely investigated in osteosarcoma (OS) treatment. However, there are several disadvantages regarding its clinical use. Specifically, Dox has low specificity toward cancer cells, which can lead to serious side effects. In addition, cancer cells can develop resistance toward Dox, reducing its therapeutic efficiency. Combination therapy (CT) facilitated by nanoparticle delivery systems is a promising strategy to overcome these drawbacks. In this study, we investigated the effectiveness of Dox and selenium (Se) CT using mesoporous silica nanoparticles (MSN) coated with hyaluronic acid (HA) as drug carriers. We hypothesized that combining Se as a second agent can increase Dox anti-OS effectiveness and that MSN can be used to facilitate dual drug delivery. In our system, HA was used as a gatekeeper to control the intracellular release of Se/Dox by means of its pH-responsive degradation. CT therapy using MSNs coated with HA led to a higher OS inhibitory efficiency in vitro compared to MSNs carrying either Se or Dox alone. This study demonstrates that using MSNs for the dual delivery of Se and Dox is a promising method for OS therapy.
Collapse
Affiliation(s)
- Lei He
- Department of Instructive
Biomaterials Engineering, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Zahra Javid Anbardan
- Department of Instructive
Biomaterials Engineering, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Pamela Habibovic
- Department of Instructive
Biomaterials Engineering, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sabine van Rijt
- Department of Instructive
Biomaterials Engineering, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
33
|
Lathakumari RH, Vajravelu LK, Satheesan A, Thulukanam J. Advancing cryptococcal treatment: The role of nanoparticles in mitigating antifungal resistance. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100323. [PMID: 39678065 PMCID: PMC11638651 DOI: 10.1016/j.crmicr.2024.100323] [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] [Indexed: 12/17/2024] Open
Abstract
Cryptococcus, a ubiquitous and formidable fungal pathogen, contributes to a substantial global disease burden, with nearly 250,000 cases and 181,000 fatalities attributed to cryptococcal meningitis annually worldwide. The invasive nature of Cryptococcus presents significant challenges in treatment and management, as it mostly affects vulnerable populations, including HIV patients, organ transplant recipients, pregnant women, and elderly individuals. Moreover, these difficulties are exacerbated by the development of antifungal resistance, which emphasizes the need for efficient control measures. In this context, research efforts focusing on infection control and novel therapeutic strategies become paramount. Nanoparticle-based therapies emerge as a solution, offering advanced antifungal properties and improved efficacy. Developing effective treatment options requires understanding the complex landscape of cryptococcal infections and the innovative potential of nanoparticle-based therapies. This review highlights the urgent need for novel strategies to combat the growing threat posed by antifungal resistance while offering insights into the intricate realm of cryptococcal infections, particularly focusing on the promising role of nanoparticle-based therapies.
Collapse
Affiliation(s)
- Rahul Harikumar Lathakumari
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| | - Leela Kakithakara Vajravelu
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| | - Abhishek Satheesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| | - Jayaprakash Thulukanam
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| |
Collapse
|
34
|
Bhuyan T, Choudhury K, Das P, Sharma S, Mazumder JA, Mohanta YK. Biosynthesis of pH-Responsive Mesoporous Silica Nanoparticles from Cucumber Peels for Targeting 3D Lung Tumor Spheroids. ACS APPLIED BIO MATERIALS 2024; 7:7494-7508. [PMID: 39467160 DOI: 10.1021/acsabm.4c01092] [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: 10/30/2024]
Abstract
Lung adenocarcinoma is considered to be one of the primary causes of cancer-related deaths globally. Conventional treatments, such as chemotherapy and radiation therapy taken together, have not significantly lowered mortality rates. Repositioning of authorized anticancer medications supported by nanotechnology has therefore emerged as an effective strategy to close such gaps. In this context, mesoporous silica nanoparticles (MSNs) were biosynthesized from cucumber peels and were loaded with doxorubicin, a common anticancer drug to form doxorubicin-bound mesoporous silica nanoparticles (DMSNs). The study addresses a sustainable method for turning waste materials into MSNs, which can be used to create multifunctional nanosystems. The therapeutic module (DMSNs) was designed specifically to target 2D monolayer cells and 3D tumor spheroids of lung adenocarcinoma cancer. The DMSNs demonstrated notable antiproliferative activity and effective intracellular localization in addition to being biocompatible and innately fluorescent. Subsequent investigations revealed significant antibacterial activity against Staphylococcal infection, which is primarily prevalent in lung cancer patients. Thus, the developed MSNs held promising potential for anticancer drug delivery systems and have antibacterial potential to treat bacterial infections in patients with lung cancer. Furthermore, the cucumber peel-mediated synthesis of MSNs could also aid in the management of food waste and promote the adoption of the waste-to-health paradigm for sustainable solutions.
Collapse
Affiliation(s)
- Tamanna Bhuyan
- Nanobiotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Ninth Mile, Technocity, Baridua, Ri-Bhoi, 793101 Meghalaya, India
| | - Konika Choudhury
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Pranjoli Das
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Sanathoibi Sharma
- Nanobiotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Ninth Mile, Technocity, Baridua, Ri-Bhoi, 793101 Meghalaya, India
| | - Jahirul Ahmed Mazumder
- Department of Chemical Engineering, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Yugal Kishore Mohanta
- Nanobiotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Ninth Mile, Technocity, Baridua, Ri-Bhoi, 793101 Meghalaya, India
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam603103, Tamil Nadu, India
| |
Collapse
|
35
|
Guo M, Lin R, Xu W, Xu L, Liu M, Huang X, Zhang J, Li X, Ma Y, Yuan M, Li Q, Dong Q, Li X, Zhao T, Zhao D. Replenishing Cation-π Interactions for the Fabrication of Mesoporous Levodopa Nanoformulations for Parkinson Remission. ACS NANO 2024; 18:30605-30615. [PMID: 39436831 DOI: 10.1021/acsnano.4c09326] [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: 10/25/2024]
Abstract
Directly assembling drugs into mesoporous nanoformulations will be greatly favored due to the combination of enhanced drug delivery efficiency and mesostructure-enabled nanobio interactions. However, such an approach is hindered due to the lack of understanding of polymer nanoparticles' formation mechanism, especially the relationship between polymerization, self-assembly, and the nucleation process. Here, by investigating the levodopa and dopamine polymerization process, we identify π-cation interaction as pivotal in the self-assembly and nucleation control of dopa molecules. Thus, through manipulation of the π-cation interaction, we present the direct assembly of a commercial drug, levodopa, into mesoporous nanoformulations. The synthesized nanospheres, approximately 200 nm in diameter, exhibit uniform mesopores of around 8 nm. These nanoformulations, abundant in mesopores, enhance chiral phenylalanine interaction with α-synuclein (Syn), curbing aggregation, safeguarding neurons, and alleviating Parkinson's pathology. When combating α-synuclein, the nanoformulation achieved ∼100% inhibition of protein aggregation and sustained neuron viability up to 300%. We believe that this study may advance mesoscale self-assembly knowledge, guiding future nanopharmaceutical developments.
Collapse
Affiliation(s)
- Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200433, P. R. China
| | - Runfeng Lin
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Wenqing Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200433, P. R. China
| | - Li Xu
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Minchao Liu
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Xirui Huang
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Jie Zhang
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Xingjin Li
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Yanming Ma
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Minjia Yuan
- Shanghai Qiran Biotechnology Co., Ltd., Shanghai 201702, P. R. China
| | - Qi Li
- Shanghai Qiran Biotechnology Co., Ltd., Shanghai 201702, P. R. China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200433, P. R. China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200433, P. R. China
| | - Xiaomin Li
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Tiancong Zhao
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Dongyuan Zhao
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, 2011-iChEM, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
36
|
Rasool A, Sri S, Zulfajri M, Sri Herwahyu Krismastuti F. Nature inspired nanomaterials, advancements in green synthesis for biological sustainability. INORG CHEM COMMUN 2024; 169:112954. [DOI: 10.1016/j.inoche.2024.112954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Chamani M, Asgari S, Najmeddin A, Pourjavadi A, Amin M, Gholami M, Dorkoosh FA. Antibacterial activity of a silver-incorporated vancomycin-modified mesoporous silica against methicillin-resistant Staphylococcus aureus. J Biomater Appl 2024; 39:439-454. [PMID: 39193668 DOI: 10.1177/08853282241274517] [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: 08/29/2024]
Abstract
Since conventional antibiotics are almost ineffective on methicillin-resistant Staphylococcus aureus (MRSA) strains, designing their antibacterial alternatives is necessary. Besides, the use of vancomycin is applied for specific detection of the bacteria. Silver-incorporated vancomycin-modified mesoporous silica nanoparticles (MSNs@Van@Ag NPs) were designed for detection and treatment of MRSA bacteria. Mesoporous silica nanoparticles (MSNs) were synthesized through the template method, modified with vancomycin, and finally incorporated with silver nanoparticles (Ag NPs). The MSNs@Van@Ag NPs with a homogenously spherical shape, average size of 50-100 nm, surface area of 955.8 m2/g, and thermal stability up to 200°C were successfully characterized. The amount of Ag incorporated into the MSNs@Van@Ag was calculated at 3.9 ppm and the release amount of Ag was received at 2.92 ppm (75%) after 100 h. The in vitro antibacterial susceptibility test showed the MIC of 100 μg mL-1 for MSNs@Van and 50 μg mL-1 for MSNs@Van@Ag, showing in vitro enhanced effect of Ag and vancomycin in the bactericidal process. An in vivo acute pneumonia model was performed and biochemical assays and pathological studies confirmed the nanomedicine's short-term safety for in vivo application. Cytokine assay using ELISA showed that MSN@Van@Ag causes a reduction of pro-inflammatory cytokines and bacterial proliferation leading to alleviation of inflammatory response.
Collapse
Affiliation(s)
- Mehdi Chamani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadi Asgari
- Polymer Research Laboratory, Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Ali Najmeddin
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Pourjavadi
- Polymer Research Laboratory, Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Khoramian R, Issakhov M, Pourafshary P, Gabdullin M, Sharipova A. Surface modification of nanoparticles for enhanced applicability of nanofluids in harsh reservoir conditions: A comprehensive review for improved oil recovery. Adv Colloid Interface Sci 2024; 333:103296. [PMID: 39241391 DOI: 10.1016/j.cis.2024.103296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/22/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Nanoparticles improve traditional Enhanced Oil Recovery (EOR) methods but face instability issues. Surface modification resolves these, making it vital to understand its impact on EOR effectiveness. This paper examines how surface-modified nanoparticles can increase oil recovery rates. We discuss post-synthesis modifications like chemical functionalization, surfactant and polymer coatings, surface etching, and oxidation, and during-synthesis modifications like core-shell formation, in-situ ligand exchange, and surface passivation. Oil displacement studies show surface-engineered nanoparticles outperform conventional EOR methods. Coatings or functionalizations alter nanoparticle size by 1-5 nm, ensuring colloidal stability for 7 to 30 days at 25 to 65 °C and 30,000 to 150,000 ppm NaCl. This stability ensures uniform distribution and enhanced penetration through low-permeability (1-10 md) rocks, improving oil recovery by 5 to 50 %. Enhanced recovery is achieved through 1-25 μm oil-in-water emulsions, increased viscosity by ≥30 %, wettability changes from 170° to <10°, and interfacial tension reductions of up to 95 %. Surface oxidation is suitable for carbon-based nanoparticles in high-permeability (≥500 md) reservoirs, leading to 80 % oil recovery in micromodel studies. Surface etching is efficient for all nanoparticle types, and combining it with chemical functionalization enhances resistance to harsh conditions (≥40,000 ppm salinity and ≥ 50 °C). Modifying nanoparticle surfaces with a silane coupling agent before using polymers and surfactants improves EOR parameters and reduces polymer thermal degradation (e.g., only 10 % viscosity decrease after 90 days). Economically, 500 ppm of nanoparticles requires 56.25 kg in a 112,500 m3 reservoir, averaging $200/kg, and 2000 ppm of surface modifiers require 4 kg at $3.39/kg. This results in 188,694.30 barrels, or $16,039,015.50 at $85 per barrel for a 20 % increase in oil recovery. The economic benefits justify the initial costs, highlighting the importance of cost-effective nanoparticles for EOR applications.
Collapse
Affiliation(s)
- Reza Khoramian
- School of Mining and Geosciences, Nazarbayev University, Astana, Kazakhstan
| | | | - Peyman Pourafshary
- School of Mining and Geosciences, Nazarbayev University, Astana, Kazakhstan.
| | | | | |
Collapse
|
39
|
Umair Amin M, Ali S, Engelhardt KH, Nasrullah U, Preis E, Schaefer J, Pfeilschifter J, Bakowsky U. Enhanced photodynamic therapy of curcumin using biodegradable PLGA coated mesoporous silica nanoparticles. Eur J Pharm Biopharm 2024; 204:114503. [PMID: 39303950 DOI: 10.1016/j.ejpb.2024.114503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Since the available treatments are not highly effective to combat cancer, therefore, the alternative strategies are unavoidable. Photodynamic therapy (PDT) is one of the emerging approaches which is target specific and minimally invasive. This study explores the successful development of Poly (D,L-lactide-co-glycolide) (PLGA) coated mesoporous silica nanoparticles (MSNs) and their augmented effects achieved by integrating curcumin (Cur) and cetyltrimethylammonium bromide (CTAB) in the polymeric layer and silica's pores, respectively. The synthesized nanocarriers (Cur-PLGA-cMSNs) have shown preferential targeting to the cellular organelles facilitated by CTAB's and Cur's affinity to mitochondria. CTAB and Cur-based PDT induced oxidative stress and generation of reactive oxygen species (ROS), resulting in dysfunctional mitochondria and triggered apoptotic pathways. PLGA coating has produced multifunctional effects, including; gatekeeping effects at pore openings, providing an extra loading site, enhancing the hemocompatibility of MSNs, and masking the free cur-related prolonged coagulation time. Cur-PLGA-cMSNs, as a multifaceted and combative approach with synergistic effects demonstrate promising potential to enhance outcomes in cancer treatment.
Collapse
Affiliation(s)
- Muhammad Umair Amin
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg, Germany.
| | - Sajid Ali
- Department of Chemistry, Ångstr¨ om Laboratory, Uppsala University, 75237 Uppsala, Sweden
| | - Konrad H Engelhardt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg, Germany
| | - Usman Nasrullah
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt am Main, Germany
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg, Germany
| | - Jens Schaefer
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg, Germany
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt am Main, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg, Germany
| |
Collapse
|
40
|
Vashishtha A, Phimphachanh A, Gaillard T, Schmitt J, Gerardin C, Rydzek G, Aubert T. Hybrid Silica Cage-Type Nanostructures Made from Triply Hydrophilic Block Copolymers Single Micelles. ACS NANO 2024; 18:29008-29020. [PMID: 39396244 DOI: 10.1021/acsnano.4c09887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Controlling the structure and functionality of porous silica nanoparticles has been a continuous source of innovation with important potential for advanced biomedical applications. Their synthesis, however, usually involves passive surfactants or amphiphilic copolymers that do not add value to the material after synthesis. In contrast, polyion complex (PIC) micelles based on hydrophilic block copolymers allow for the direct synthesis of intrinsically functional hybrid materials. While most previous studies have focused on bulk materials made from double-hydrophilic block copolymers (DHBC), in this work we have synthesized a triple-hydrophilic block copolymer (THBC) and demonstrated both its PIC micellization and its potential for hybrid mesoporous silica nanomaterials. Introducing this THBC has allowed to direct the transition from bulk three-dimensional (3D) materials to zero-dimensional (0D) nanomaterials with cage-type structures. The stabilization and isolation of these nanostructures formed around discrete individual micelles has been made possible by the careful design of the three different blocks that each play a key role. These nanostructures could also be synthesized from hybrid PIC micelles based on THBC-multivalent metal ions complexes, offering a direct route to metal/silica composite nanoparticles. This class of THBC polymers therefore creates significant opportunities for the synthesis of nanostructures with complex and functional architectures.
Collapse
Affiliation(s)
- Anu Vashishtha
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France
| | | | - Thomas Gaillard
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France
| | - Julien Schmitt
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France
- Saint-Gobain Research Provence, LSFC, CNRS, 84300 Cavaillon, France
| | - Corine Gerardin
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France
| | | | - Tangi Aubert
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France
| |
Collapse
|
41
|
Trayford C, van Rijt S. In situ modified mesoporous silica nanoparticles: synthesis, properties and theranostic applications. Biomater Sci 2024; 12:5450-5467. [PMID: 39371000 PMCID: PMC11457002 DOI: 10.1039/d4bm00094c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 08/29/2024] [Indexed: 10/08/2024]
Abstract
Over the last 20 years, mesoporous silica nanoparticles (MSNs) have drawn considerable attention in the biomedical field due to their large surface area, porous network, biocompatibility, and abundant modification possibilities. In situ MSN modification refers to the incorporation of materials such as alkoxysilanes, ions and nanoparticles (NPs) in the silica matrix during synthesis. Matrix modification is a popular approach for endowing MSNs with additional functionalities such as imaging properties, bioactivity, and degradability, while leaving the mesopores free for drug loading. As such, in situ modified MSNs are considered promising theranostic agents. This review provides an extensive overview of different materials and modification strategies that have been used and their effect on MSN properties. We also highlight how in situ modified MSNs have been applied in theranostic applications, oncology and regenerative medicine. We conclude with perspectives on the future outlooks and current challenges for the widespread clinical use of in situ modified MSNs.
Collapse
Affiliation(s)
- Chloe Trayford
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Sabine van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
42
|
Kusz J, Boissiere C, Bretonnière Y, Sanchez C, Parola S. Pyrene monomer-excimer dynamics to reveal molecular organization in mesoporous hybrid silica films. NANOSCALE 2024; 16:18918-18932. [PMID: 39267607 DOI: 10.1039/d4nr02987a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Self-assembly and characteristics of hybrid mesoporous silica film templates remain a subject of inquiry. The short time scale of the inorganic condensation and formation of micelles makes our understanding of this process insufficient. To provide an insight into the evaporation-induced self-assembly of such films, we synthesized an efficient molecular probe of the triethoxysilane precursor bearing a pyrene derivative. The probe was introduced into the porous film at the synthesis stage through the sol-gel co-condensation method. At different synthesis stages, the emission of pyrene moieties was measured by fluorescence spectroscopy, revealing the placement of probes within the film. We also report dynamic excimer formation upon template removal. Moreover, we evaluate the influence of several parameters on the pyrene excimer formation phenomenon. The pore geometry, probe concentration, and the presence of another organosilane precursor are investigated in this work.
Collapse
Affiliation(s)
- Jakub Kusz
- École Normale Supérieure de Lyon, CNRS, Université Claude Bernard Lyon 1, Laboratoire de Chimie, UMR 5182, 46 Allée d'Italie, 69364 Lyon, France.
| | - Cédric Boissiere
- Sorbonne Université, CNRS, Collège de France, Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), UMR 7574, 4 place Jussieu, 75005 Paris, France.
| | - Yann Bretonnière
- École Normale Supérieure de Lyon, CNRS, Université Claude Bernard Lyon 1, Laboratoire de Chimie, UMR 5182, 46 Allée d'Italie, 69364 Lyon, France.
| | - Clément Sanchez
- Sorbonne Université, CNRS, Collège de France, Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), UMR 7574, 4 place Jussieu, 75005 Paris, France.
| | - Stephane Parola
- École Normale Supérieure de Lyon, CNRS, Université Claude Bernard Lyon 1, Laboratoire de Chimie, UMR 5182, 46 Allée d'Italie, 69364 Lyon, France.
| |
Collapse
|
43
|
Martín‐Morales C, Caspani S, Desco M, Tavares de Sousa C, Gómez‐Gaviro MV. Controlled Drug Release Systems for Cerebrovascular Diseases. ADVANCED THERAPEUTICS 2024. [DOI: 10.1002/adtp.202400239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Indexed: 01/06/2025]
Abstract
AbstractThis review offers a comprehensive exploration of optimized drug delivery systems tailored for controlled release and their crucial role in addressing cerebrovascular diseases. Through an in‐depth analysis, various controlled release methods, including nanoparticles, liposomes, hydrogels, and other emerging technologies are examined. Highlighting the importance of precise drug targeting, it is delved into the underlying mechanisms of these delivery systems and their potential to improve therapeutic outcomes while minimizing adverse effects. Additionally, the specific applications of these optimized drug delivery systems in treating cerebrovascular disorders such as ischemic stroke, cerebral aneurysms, and intracranial hemorrhage are discussed. By shedding light on the advancements in drug delivery techniques and their implications in cerebrovascular medicine, this review offers valuable insights into the future of therapeutic interventions in neurology.
Collapse
Affiliation(s)
- Celia Martín‐Morales
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
| | - Sofia Caspani
- IFIMUP – Institute of Physics for Advanced Materials Departamento de Física e Astronomia, Nanotechnology and Photonics of University of Porto Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre s/n Porto 4169‐007 Portugal
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
- Departamento de Bioingeniería Universidad Carlos III de Madrid Leganés 28911 Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) Madrid 28029 Spain
- Centro de Investigaciones Cardiovasculares (CNIC) Melchor Fernández Almagro Madrid 28029 Spain
| | - Célia Tavares de Sousa
- Departamento de Física Aplicada and IAdChem Facultad de Ciencias Universidad Autonoma de Madrid (UAM) Campus de Cantoblanco, C/ Francisco Tomas y Valiente, 7 Madrid 28049 Spain
| | - María Victoria Gómez‐Gaviro
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
- Departamento de Bioingeniería Universidad Carlos III de Madrid Leganés 28911 Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) Madrid 28029 Spain
| |
Collapse
|
44
|
Li M, Tang Q, Wan H, Zhu G, Yin D, Lei L, Li S. Functional inorganic nanoparticles in cancer: Biomarker detection, imaging, and therapy. APL MATERIALS 2024; 12. [DOI: 10.1063/5.0231279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Cancer poses a major global public health challenge. Developing more effective early diagnosis methods and efficient treatment techniques is crucial to enhance early detection sensitivity and treatment outcomes. Nanomaterials offer sensitive, accurate, rapid, and straightforward approaches for cancer detection, diagnosis, and treatment. Inorganic nanoparticles are widely used in medicine because of their high stability, large specific surface area, unique surface properties, and unique quantum size effects. Functional inorganic nanoparticles involve modifying inorganic nanoparticles to enhance their physical properties, enrichment capabilities, and drug-loading efficiency and to minimize toxicity. This Review provides an overview of various types of inorganic nanoparticles and their functionalization characteristics. We then discuss the progress of functional inorganic nanoparticles in cancer biomarker detection and imaging. Furthermore, we discuss the application of functional inorganic nanoparticles in radiotherapy, chemotherapy, gene therapy, immunotherapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and combination therapy, highlighting their characteristics and advantages. Finally, the toxicity and potential challenges of functional inorganic nanoparticles are analyzed. The purpose of this Review is to explore the application of functional inorganic nanoparticles in diagnosing and treating cancers, while also presenting a new avenue for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University 1 , Changsha 410011, Hunan,
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University 1 , Changsha 410011, Hunan,
| | - Hua Wan
- Department of Otorhinolaryngology Head and Neck Surgery 2 , 331 Hospital of Zhuzhou, Zhuzhou 412002, Hunan,
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University 1 , Changsha 410011, Hunan,
| | - Danhui Yin
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University 1 , Changsha 410011, Hunan,
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University 3 , Hangzhou 310015, Zhejiang,
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University 1 , Changsha 410011, Hunan,
| |
Collapse
|
45
|
Tu B, Jonnalagadda S. Amorphous stabilization of BCS II drugs using mesoporous silica. Int J Pharm 2024; 663:124555. [PMID: 39111354 DOI: 10.1016/j.ijpharm.2024.124555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
This study aimed to investigate the amorphous stabilization of BCS Class II drugs using mesoporous silica as a carrier to produce amorphous solid dispersions. Ibuprofen, fenofibrate, and budesonide were selected as model drugs to evaluate the impact of molecular weight and partition coefficient on the solid state of drug-loaded mesoporous silica (MS) particles. The model drugs were loaded into three grades of MS, SYLYSIA SY730, SYLYSIA SY430, and SYLYSIA SY350, with pore diameters of 2.5 nm, 17 nm, and 21 nm, respectively, at 1:1, 2:1, and 3:1, carrier to drug ratios, and three different loading concentrations using solvent immersion and spray drying techniques. Differential scanning calorimetry (DSC) thermograms of SY430 and SY350 samples exhibited melting point depressions indicating constricted crystallization inside the pores, whereas SY730 samples with melting points matching the pure API may be a result of surface crystallization. Powder x-ray diffraction (PXRD) diffractograms showed all crystalline samples matched the diffraction patterns of the pure API indicating no polymorphic transitions and all 3:1 ratio samples exhibited amorphous halo profiles. Response surface regression analysis and Classification and Regression Tree (CART) analysis suggest carrier to drug ratios, followed by molecular weight, have the most significant impact on the crystallinity of a drug loaded into MS particles.
Collapse
Affiliation(s)
- Buu Tu
- Saint Joseph's University, 600 S 43rd Street, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
46
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [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: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
- Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
47
|
Huang J, Hong X, Chen S, He Y, Xie L, Gao F, Zhu C, Jin X, Yan H, Ye Y, Shao M, Du X, Feng G. Biomimetic Metal-Organic Framework Gated Nanoplatform for Sonodynamic Therapy against Extensively Drug Resistant Bacterial Lung Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402473. [PMID: 38962911 PMCID: PMC11434100 DOI: 10.1002/advs.202402473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/03/2024] [Indexed: 07/05/2024]
Abstract
Novel antimicrobial strategies are urgently needed to treat extensively drug-resistant (XDR) bacterial infections due to the high mortality rate and lack of effective therapeutic agents. Herein, nanoengineered human umbilical cord mesenchymal stem cells (hUC-MSCs), named PMZMU, are designed as a sonosensitizer for synergistic sonodynamic-nano-antimicrobial therapy against gram-negative XDR bacteria. PMZMU is composed of a bacterial targeting peptide (UBI29-41) modified hUC-MSCs membrane (MSCm), a sonosensitizer meso-tetra(4-car-boxyphenyl) porphine doped mesoporous organo-silica nanoparticle and an acidity-responsive metal-organic framework ZIF-8. This innovative formulation enables efficient loading of polymyxin B, reduces off-target drug release, increases circulation and targeting efficacy, and generates reactive oxygen species upon ultrasound irradiation. PMZMU exhibits remarkable in vitro inhibitory activity against four XDR bacteria: Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa (PA), and Escherichia coli. Taking advantage of the bacterial targeting ability of UBI29-41 and the inflammatory chemotaxis of hUC-MSC, PMZMU can be precisely delivered to lung infection sites thereby augmenting polymyxin B concentration. PMZMU-mediated sonodynamic therapy significantly reduces bacterial burden, relieves inflammatory damage by promoting the polarization of macrophages toward M2 phenotype, and improves survival rates without introducing adverse events. Overall, this study offers promising strategies for treating deep-tissue XDR bacterial infections, and guides the design and optimization of biomimetic nanomedicine.
Collapse
Affiliation(s)
- Jianling Huang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiuwen Hong
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Sixi Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Lixu Xie
- Department of Pulmonary and Critical Care Medicine, Qi Lu Hospital of Shandong University, Wen hua xi Road 107#, Jinan, 250012, China
| | - Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Chenghua Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiao Jin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Haihao Yan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yongxia Ye
- Department of Radiology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, 210009, China
| | - Mingyue Shao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xingran Du
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| |
Collapse
|
48
|
Shao A, Jin L, Ge Y, Ye Z, Xu M, Zhou Y, Li Y, Wang L, Xu P, Jin K, Mao Z, Ye J. C176-loaded and phosphatidylserine-modified nanoparticles treat retinal neovascularization by promoting M2 macrophage polarization. Bioact Mater 2024; 39:392-405. [PMID: 38855060 PMCID: PMC11157223 DOI: 10.1016/j.bioactmat.2024.05.038] [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: 03/12/2024] [Revised: 05/08/2024] [Accepted: 05/25/2024] [Indexed: 06/11/2024] Open
Abstract
Retinal neovascularization (RNV), a typical pathological manifestation involved in most neovascular diseases, causes retinal detachment, vision loss, and ultimately irreversible blindness. Repeated intravitreal injections of anti-VEGF drugs were developed against RNV, with limitations of incomplete responses and adverse effects. Therefore, a new treatment with a better curative effect and more prolonged dosage is demanding. Here, we induced macrophage polarization to anti-inflammatory M2 phenotype by inhibiting cGAS-STING signaling with an antagonist C176, appreciating the role of cGAS-STING signaling in the retina in pro-inflammatory M1 polarization. C176-loaded and phosphatidylserine-modified dendritic mesoporous silica nanoparticles were constructed and examined by a single intravitreal injection. The biosafe nanoparticles were phagocytosed by retinal macrophages through a phosphatidylserine-mediated "eat me" signal, which persistently release C176 to suppress STING signaling and thereby promote macrophage M2 polarization specifically. A single dosage can effectively alleviate pathological angiogenesis phenotypes in murine oxygen-induced retinopathy models. In conclusion, these C176-loaded nanoparticles with enhanced cell uptake and long-lasting STING inhibition effects might serve as a promising way for treating RNV.
Collapse
Affiliation(s)
- An Shao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yanni Ge
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Ziqiang Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Mingyu Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yifan Zhou
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yingyu Li
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Linyan Wang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Pinglong Xu
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310030, China
| | - Kai Jin
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
49
|
Carnero Canales CS, Marquez Cazorla JI, Marquez Cazorla RM, Roque-Borda CA, Polinário G, Figueroa Banda RA, Sábio RM, Chorilli M, Santos HA, Pavan FR. Breaking barriers: The potential of nanosystems in antituberculosis therapy. Bioact Mater 2024; 39:106-134. [PMID: 38783925 PMCID: PMC11112550 DOI: 10.1016/j.bioactmat.2024.05.013] [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: 01/31/2024] [Revised: 04/17/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, continues to pose a significant threat to global health. The resilience of TB is amplified by a myriad of physical, biological, and biopharmaceutical barriers that challenge conventional therapeutic approaches. This review navigates the intricate landscape of TB treatment, from the stealth of latent infections and the strength of granuloma formations to the daunting specters of drug resistance and altered gene expression. Amidst these challenges, traditional therapies often fail, contending with inconsistent bioavailability, prolonged treatment regimens, and socioeconomic burdens. Nanoscale Drug Delivery Systems (NDDSs) emerge as a promising beacon, ready to overcome these barriers, offering better drug targeting and improved patient adherence. Through a critical approach, we evaluate a spectrum of nanosystems and their efficacy against MTB both in vitro and in vivo. This review advocates for the intensification of research in NDDSs, heralding their potential to reshape the contours of global TB treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Cesar Augusto Roque-Borda
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Giulia Polinário
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | | | - Rafael Miguel Sábio
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
| | - Marlus Chorilli
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Fernando Rogério Pavan
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
50
|
Prabhakar N, Långbacka E, Özliseli E, Mattsson J, Mahran A, Suleymanova I, Sahlgren C, Rosenholm JM, Åkerfelt M, Nees M. Surface Modification of Mesoporous Silica Nanoparticles as a Means to Introduce Inherent Cancer-Targeting Ability in a 3D Tumor Microenvironment. SMALL SCIENCE 2024; 4:2400084. [PMID: 40212075 PMCID: PMC11935100 DOI: 10.1002/smsc.202400084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Indexed: 04/13/2025] Open
Abstract
Mesoporous silica nanoparticles (MSNs) have emerged as promising drug carriers that can facilitate targeted anticancer drug delivery, but efficiency studies relying on active targeting mechanisms remain elusive. This study implements in vitro 3D cocultures, so-called microtissues, to model a physiologically relevant tumor microenvironment (TME) to examine the impact of surface-modified MSNs without targeting ligands on the internalization, cargo delivery, and cargo release in tumor cells and cancer-associated fibroblasts. Among these, acetylated MSNs most effectively localized in tumor cells in a 3D setting containing collagen, while other MSNs did so to a lesser degree, most likely due to remaining trapped in the extracellular matrix of the TME. Confocal imaging of hydrophobic model drug-loaded MSNs demonstrated effective cargo release predominantly in tumor cells, both in 2D and 3D cocultures. MSN-mediated delivery of an anticancer drug in the microtissues exhibited a significant reduction in tumor organoid size and enhanced the tumor-specific cytotoxic effects of a γ-secretase inhibitor, compared to the highly hydrophobic drug in free form. This inherent targeting potential suggests reduced off-target effects and increased drug efficacy, showcasing the promise of surface modification of MSNs as a means of direct cell-specific targeting and delivery for precise and successful targeted drug delivery.
Collapse
Affiliation(s)
- Neeraj Prabhakar
- Pharmaceutical Sciences Laboratory, Faculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
- Centre for Structural Systems Biology (CSSB)c/o DESYNotkestrasse 8522607HamburgGermany
- Department of PhysicsUniversity of Hamburg20355HamburgGermany
| | - Erica Långbacka
- BiosciencesFaculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
- Institute of Biomedicine and FICAN West Cancer CentreUniversity of TurkuTurku20520Finland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurku20520Finland
- Turku Bioscience CentreÅbo Akademi University and University of TurkuTurku20520Finland
| | - Ezgi Özliseli
- Pharmaceutical Sciences Laboratory, Faculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
| | - Jesse Mattsson
- Institute of Biomedicine and FICAN West Cancer CentreUniversity of TurkuTurku20520Finland
| | - Alaa Mahran
- Pharmaceutical Sciences Laboratory, Faculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
- Department of PharmaceuticsFaculty of PharmacyAssiut UniversityAssiut71526Egypt
| | - Ilida Suleymanova
- Faculty of Biological and Environmental SciencesHelsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinki00014Finland
| | - Cecilia Sahlgren
- BiosciencesFaculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurku20520Finland
- Turku Bioscience CentreÅbo Akademi University and University of TurkuTurku20520Finland
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
| | - Malin Åkerfelt
- BiosciencesFaculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
- Institute of Biomedicine and FICAN West Cancer CentreUniversity of TurkuTurku20520Finland
| | - Matthias Nees
- Institute of Biomedicine and FICAN West Cancer CentreUniversity of TurkuTurku20520Finland
- Department of Biochemistry and Molecular BiologyMedical University of LublinLublin20‐093Poland
| |
Collapse
|