1
|
Li H, Murugesan A, Shoaib M, Chen Q. Emerging Trends and Future Prospects of Peptide-Based Hydrogels: Revolutionizing Food Technology Applications. Compr Rev Food Sci Food Saf 2025; 24:e70187. [PMID: 40371450 DOI: 10.1111/1541-4337.70187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/28/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
Peptide-based hydrogels (PHs) are versatile materials with considerable potential in food technology. Advances in synthesis techniques, such as self-assembly, click chemistry, enzymatic cross-linking, and co-assembly with polymers, have improved their production efficiency and scalability. Derived from natural amino acids, PHs are biocompatible, biodegradable, and responsive to environmental factors like pH and temperature. In food technology, encapsulation and controlled release of bioactive compounds enhance nutrient stability, flavor preservation, and bioavailability. PHs serve as texture modifiers, improve product consistency, and possess antimicrobial properties for food preservation by inhibiting spoilage and pathogens. Their biodegradability supports eco-friendly practices and sustainable packaging, including edible films and coatings that extend shelf life. Adjustable properties such as ionic strength make PHs adaptable to specific needs. PHs also show potential in developing advanced food equipment, including 3D printers and encapsulation systems, promoting efficiency and sustainability. This review emphasizes that PHs offer innovative, sustainable solutions to enhance food functionality, quality, and safety, with broad applications in food processing and preservation.
Collapse
Affiliation(s)
- Huanhuan Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, P. R. China
| | - Arul Murugesan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, P. R. China
| | - Muhammad Shoaib
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, P. R. China
| | - Quansheng Chen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, P. R. China
| |
Collapse
|
2
|
Mahmoudi Gharehbaba A, Soltanmohammadi F, Vandghanooni S, Eskandani M, Adibkia K. A comprehensive review on overcoming the multifaceted challenge of cancer multidrug resistance: The emerging role of mesoporous silica nanoparticles. Biomed Pharmacother 2025; 186:118045. [PMID: 40215648 DOI: 10.1016/j.biopha.2025.118045] [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: 12/11/2024] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
Multidrug resistance (MDR) is a significant challenge in tumor treatment, severely reducing the effectiveness of anticancer drugs and contributing to high mortality rates. This article overviews the various factors involved in the development of MDR, such as changes in drug targets, increased DNA repair mechanisms, and the impact of the tumor microenvironment. It also emphasizes the potential of mesoporous silica nanoparticles (MSNs) as a drug delivery system to combat MDR. With their unique characteristics-such as a high surface area, adjustable pore sizes, and the ability to be functionalized for targeted delivery-MSNs serve as excellent carriers for the simultaneous delivery of chemotherapeutics and siRNAs aimed at reversing resistance pathways. The paper focuses on innovative methods using MSNs for direct intranuclear delivery of their cargos to overcome efflux barrier and improve the effectiveness of combination therapies. This review highlights a promising approach for enhancing cancer treatment outcomes by integrating advanced nanotechnology with traditional therapies, addressing the ongoing challenge of MDR in oncology.
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
| | - Fatemeh Soltanmohammadi
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - 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
|
3
|
Hu J, Arvejeh PM, Bone S, Hett E, Marincola FM, Roh KH. Nanocarriers for cutting-edge cancer immunotherapies. J Transl Med 2025; 23:447. [PMID: 40234928 PMCID: PMC12001629 DOI: 10.1186/s12967-025-06435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer immunotherapy aims to harness the body's own immune system for effective and long-lasting elimination of malignant neoplastic tissues. Owing to the advance in understanding of cancer pathology and immunology, many novel strategies for enhancing immunological responses against various cancers have been successfully developed, and some have translated into excellent clinical outcomes. As one promising strategy for the next generation of immunotherapies, activating the multi-cellular network (MCN) within the tumor microenvironment (TME) to deploy multiple mechanisms of action (MOAs) has attracted significant attention. To achieve this effectively and safely, delivering multiple or pleiotropic therapeutic cargoes to the targeted sites of cancerous tissues, cells, and intracellular organelles is critical, for which numerous nanocarriers have been developed and leveraged. In this review, we first introduce therapeutic payloads categorized according to their predicted functions in cancer immunotherapy and their physicochemical structures and forms. Then, various nanocarriers, along with their unique characteristics, properties, advantages, and limitations, are introduced with notable recent applications in cancer immunotherapy. Following discussions on targeting strategies, a summary of each nanocarrier matching with suitable therapeutic cargoes is provided with comprehensive background information for designing cancer immunotherapy regimens.
Collapse
Affiliation(s)
- Joyce Hu
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | - Pooria M Arvejeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sydney Bone
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Erik Hett
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | | | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
- Biotechnology Science and Engineering Program, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
| |
Collapse
|
4
|
Desai S, Thorat P, Majumdar A. A promise of nose to brain delivery of bevacizumab intranasal sol-gel formulation substantiated in rat C6 glioma model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4123-4148. [PMID: 39417842 DOI: 10.1007/s00210-024-03536-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Glioblastoma is one of the rapidly spreading cancers, with its potent malignancy often linked to pronounced angiogenesis within tumors. To mitigate this vascularization profile, bevacizumab (Avastin®), a monoclonal antibody, has been utilized for its antiangiogenic activity. However, its effectiveness is hindered by challenges in crossing the blood-brain barrier and the risk of off-target organ toxicity. Delivering drugs directly from the nose to the brain through the olfactory or trigeminal nerves bypassing the blood-brain barrier offers enhanced bioavailability and a more precise targeting strategy. To overcome these challenges, we aimed to develop bevacizumab in situ gel loaded mesoporous silica nanoparticles for intranasal delivery and further examine their pharmacokinetic and pharmacodynamic characteristics. The intranasal gel of mesoporous silica nanoparticles loaded with bevacizumab was optimized and formulated using a factorial and quality by design approach. In the case of bevacizumab mesoporous silica nanoparticles, lower particle size and most negative zeta potential were selected as quality target product profiles which is important for drug loading on the mesoporous silica nanoparticles and also transport of these nanoparticles across the nasal mucosa to the brain. A design space with a multidimensional combination of input variables and process parameters has been demonstrated to assure quality. To optimize the design space and achieve the desired quality standards, the base catalyst and surfactant concentration were chosen as the critical process parameters, while particle size and zeta potential were identified as the critical quality attributes. The novel formulation was assessed for physicochemical parameters such as particle size, zeta potential, entrapment efficiency, appearance, color, consistency, and pH. Additionally, studies on in vitro release, ex vivo permeation, stability, nasal toxicity, organ safety, and bioavailability were conducted. The efficacy study was conducted in an orthotopic murine glioblastoma rat model in which C6 Luc cells were instilled in the striatum of the rat's brain. In vivo, bioluminescence imaging of brain tumors was carried out to observe the tumor regression after treatment with the intranasal and intravenous bevacizumab formulation. Biochemical parameters and histopathology were performed for organ safety studies. The optimized intranasal formulation exhibited an average particle size of 318.8 nm and a zeta potential of - 14.7 mV for the mesoporous silica nanoparticles. The formulation also demonstrated an entrapment efficiency of 91.34% and a loading capacity of 45.67%. Further pharmacokinetic studies revealed that the optimized intranasal bevacizumab formulation achieved a significantly higher brain concentration Cmax = 147.9 ng/ml, indicating improved bioavailability compared to rats administered with intravenous bevacizumab formulation (BEVATAS®), which had a Cmax of 127.2 ng/ml. Moreover, this nanoparticle formulation entirely mitigated systemic exposure to bevacizumab. Organ safety evaluation of different biochemical parameters and histopathological analyses revealed that the intranasal bevacizumab-treated group was showing less off-target organ toxicity compared to the group treated with intravenous bevacizumab formulation. Subsequently, the efficacy of this nanosystem was evaluated in an orthotopic glioblastoma rat model, monitoring tumor growth over time through in vivo bioluminescence imaging and assessing anti-angiogenic effects. Twenty-one days post-induction, mesoporous silica nanoparticles loaded with bevacizumab in situ gel exhibited a marked reduction in average bioluminescence radiance (4.39 × 103) from day 7 (1.35 × 107) emphasizing an enhanced anti-angiogenic effect compared to the group treated with intravenous bevacizumab formulation which showed a gradual decrease in average bioluminescence radiance (4.82 × 104) from day 7 (1.42 × 107). These results suggest that the proposed novel formulation of mesoporous silica nanoparticles loaded bevacizumab in situ gel could serve as a promising avenue to enhance glioblastoma treatment efficacy, thereby potentially improving patient quality of life and survival rates significantly. Furthermore, the success of this delivery method could open new avenues for treating other neurological disorders, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and stroke. By providing effective brain-targeted therapies, this approach has the potential to revolutionize treatment options and improve outcomes for a broad spectrum of neurological conditions.
Collapse
Affiliation(s)
- Siddhesh Desai
- Department of Pharmacology, Bombay College of Pharmacy, Santacruz East, Mumbai, 400098, India
| | - Prajakta Thorat
- Department of Pharmacology, Bombay College of Pharmacy, Santacruz East, Mumbai, 400098, India
| | - Anuradha Majumdar
- Department of Pharmacology, Bombay College of Pharmacy, Santacruz East, Mumbai, 400098, India.
| |
Collapse
|
5
|
Shamshirgaran MA, Golchin M. Necrotic enteritis in chickens: a comprehensive review of vaccine advancements over the last two decades. Avian Pathol 2025; 54:1-26. [PMID: 39190009 DOI: 10.1080/03079457.2024.2398028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/29/2024] [Accepted: 08/26/2024] [Indexed: 08/28/2024]
Abstract
ABSTRACTNecrotic enteritis (NE) is a severe gastrointestinal disease that poses a significant threat to poultry, leading to progressive deterioration of the small intestine, reduced performance, and increased mortality rates, causing economic losses in the poultry industry. The elimination of antimicrobial agents from chicken feed has imposed a need to explore alternative approaches for NE control, with vaccination emerging as a promising strategy to counteract the detrimental consequences associated with NE. This comprehensive review presents an overview of the extensive efforts made in NE vaccination from 2004 to 2023. The review focuses on the development and evaluation of vaccine candidates designed to combat NE. Rigorous evaluations were conducted in both experimental chickens and broiler chickens, the target population, to assess the vaccines' capacity to elicit an immune response and provide substantial protection against toxin challenges and experimental NE infections. The review encompasses the design of vaccine candidates, the antigens employed, in vivo immune responses, and the efficacy of these vaccines in protecting birds from experimental NE infection. This review contributes to the existing knowledge of NE vaccination strategies, offering valuable insights for future research and development in this field.
Collapse
Affiliation(s)
- Mohammad Ali Shamshirgaran
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mehdi Golchin
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
6
|
Primavera R, Wang J, Buchwald P, Ganguly A, Patel S, Bettencourt L, Chetty S, Yarani R, Regmi S, Levitte S, Kevadiya B, Guindani M, Decuzzi P, Thakor AS. Controlled Nutrient Delivery to Pancreatic Islets Using Polydopamine-Coated Mesoporous Silica Nanoparticles. NANO LETTERS 2025; 25:939-950. [PMID: 39791700 DOI: 10.1021/acs.nanolett.4c03613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In this study, we designed a nanoscale platform for sustained amino acid delivery to support transplanted pancreatic islets. The platform features mesoporous silica nanoparticles (MSNPs) loaded with glutamine (G), an essential amino acid required for islet survival and function, and coated with polydopamine (PD). We investigated various PD concentrations (0.5-2 mg/mL) and incubation times (0.5-2 h) to optimize G release, identifying that a PD concentration of 0.5 mg/mL incubated for 0.5 h yielded the best results to support islet viability and functionality ex vivo, particularly under inflammatory conditions. In syngeneic islet transplantation in STZ-diabetic mice, G alone provided only temporary benefits; however, PD-G-MSNPs significantly improved islet engraftment and function, with animals maintaining glycemic control for 30 days due to controlled G release. Our findings support the use of this nanoscale platform to provide essential nutrients like G to transplanted islets until they can establish their own blood and nutrient supply.
Collapse
Affiliation(s)
- Rosita Primavera
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Jing Wang
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Abantika Ganguly
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shaini Patel
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Lili Bettencourt
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shashank Chetty
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Shobha Regmi
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Steven Levitte
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Bhavesh Kevadiya
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Michele Guindani
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Avnesh S Thakor
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| |
Collapse
|
7
|
ORMAN B, KOÇ A, KARAMAN DŞEN, NALBANTSOY A. Intrinsic apoptotic effect of Anatolian honeybee ( Apis mellifera anatoliaca) venom promoted with mesoporous silica nanocarriers. Turk J Biol 2024; 49:185-197. [PMID: 40365105 PMCID: PMC12068665 DOI: 10.55730/1300-0152.2736] [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: 08/09/2024] [Revised: 04/25/2025] [Accepted: 12/30/2024] [Indexed: 05/15/2025] Open
Abstract
Background/aim The use of bee products or treatment with bees, as a complement to conventional medicine is attracting considerable attention in cancer research. Although discoveries related to the potential anticancer effects of bee venom are increasing, the unstable nature of venom biomolecules remains a limiting factor for their usage. In this study, we employed mesoporous silica nanocarriers (MSNs) to provide precise dosing and prevent carriers from biomolecule degradation thanks to the outstanding loading capacity provided by the pores, excellent chemical and biological robustness, and ability to improve bioavailability. Materials and methods MSNs were synthesized and physicochemical characterizations were carried out. The cytotoxicity of Apis mellifera anatoliaca bee venom and venom-complexed MSNs (MSNs@Venom) were determined for the MDA-MB 231, PC3, and HeLa cancer cell lines and the cytotoxicity of pristine MSNs was investigated for the HEK-293 and CCD34-Lu cell lines. The cellular uptake of MSNs@Venom by PC3 and MDA-MB 231 cells was investigated by fluorescence microscopy and flow cytometry. The apoptotic effect on cancer cells was examined by flow cytometry. Results The MSNs exhibited significant cellular uptake of MSN by the PC3 and MDA-MB 231 cell lines, resulting in a 1.5-fold enhancement in the apoptotic effect of venom on the PC3 cell line when combined with MSNs, compared to cells exposed alone to venom. Conclusion MSNs could effectively be taken up by MDA-MB 231 and PC3 cancer cells, enhancing the action of bee venom by the particle-mediated delivery. MSNs@Venom have the potential to offer cost-effective complementary and innovative cancer treatment options.
Collapse
Affiliation(s)
- Batuhan ORMAN
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, İzmir,
Turkiye
| | - Aylin KOÇ
- Department of Biomedical Engineering, Graduate School of Natural and Applied Sciences, İzmir Kâtip Çelebi University, İzmir,
Turkiye
| | - Didem ŞEN KARAMAN
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, İzmir Kâtip Çelebi University, İzmir,
Turkiye
| | - Ayşe NALBANTSOY
- Department of Bioengineering, Faculty of Engineering, Ege University, İzmir,
Turkiye
| |
Collapse
|
8
|
Godakhindi V, Yazdimamaghani M, Dam SK, Ferdous F, Wang AZ, Tarannum M, Serody J, Vivero‐Escoto JL. Optimized Fabrication of Dendritic Mesoporous Silica Nanoparticles as Efficient Delivery System for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402802. [PMID: 39375971 PMCID: PMC11636195 DOI: 10.1002/smll.202402802] [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: 04/08/2024] [Revised: 09/13/2024] [Indexed: 10/09/2024]
Abstract
In the past decade, cancer immunotherapy has revolutionized the field of oncology. Major immunotherapy approaches such as immune checkpoint inhibitors, cancer vaccines, adoptive cell therapy, cytokines, and immunomodulators have shown great promise in preclinical and clinical settings. Among them, immunomodulatory agents including cancer vaccines are particularly appealing; however, they face limitations, notably the absence of efficient and precise targeted delivery of immune-modulatory agents to specific immune cells and the potential for off-target toxicity. Nanomaterials can play a pivotal role in addressing targeting and other challenges in cancer immunotherapy. Dendritic mesoporous silica nanoparticles (DMSNs) can enhance the efficacy of cancer vaccines by enhancing the effective loading of immune modulatory agents owing to their tunable pore sizes. In this work, an emulsion-based method is optimized to customize the pore size of DMSNs and loaded DMSNs with ovalbumin (OVA) and cytosine-phosphate-guanine (CpG) oligodeoxynucleotides (CpG-OVA-DMSNs). The immunotherapeutic effect of DMSNs is achieved through controlled chemical release of OVA and CpG in antigen-presenting cells (APCs). The results demonstrated that CpG-OVA-DMSNs efficiently activated the immune response in APCs and reduced tumor growth in the murine B16-OVA tumor model.
Collapse
Affiliation(s)
- Varsha Godakhindi
- Department of ChemistryUniversity of North Carolina at CharlotteCharlotteNC28223USA
- Nanoscale Science ProgramThe University of North Carolina at CharlotteCharlotteNC28223USA
| | - Mostafa Yazdimamaghani
- Center for Nanotechnology in Drug Delivery and Division of pharmacoengineering and Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Sudip Kumar Dam
- Department of ChemistryUniversity of North Carolina at CharlotteCharlotteNC28223USA
- Nanoscale Science ProgramThe University of North Carolina at CharlotteCharlotteNC28223USA
| | - Farzana Ferdous
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNC28223USA
| | - Andrew Z. Wang
- Center for Nanotechnology in Drug Delivery and Division of pharmacoengineering and Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNC27599USA
- Department of Radiation OncologyUT Southwestern Medical CenterDallasTX75390USA
| | - Mubin Tarannum
- Division of Medical OncologyDana Farber Cancer InstituteHarvard Medical SchoolBostonMA02215USA
| | - Jonathan Serody
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNC27599USA
- Department of Microbiology and ImmunologyUNC School of MedicineMarsico HallChapel HillNC27599USA
- Department of MedicineUNC School of MedicineHaupt BuildingChapel HillNC27599USA
| | - Juan L. Vivero‐Escoto
- Department of ChemistryUniversity of North Carolina at CharlotteCharlotteNC28223USA
- Nanoscale Science ProgramThe University of North Carolina at CharlotteCharlotteNC28223USA
| |
Collapse
|
9
|
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
|
10
|
Torpey A, Bellow E, Samojedny V, Ahluwalia S, Desai A, Caldwell W, Bergese S. Nanotechnology in Pain Management. Pharmaceutics 2024; 16:1479. [PMID: 39598601 PMCID: PMC11597168 DOI: 10.3390/pharmaceutics16111479] [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: 09/19/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024] Open
Abstract
Chronic pain is a debilitating condition that affects millions of patients worldwide, contributing to a high disease burden and millions of dollars in lost wages, missed workdays, and healthcare costs. Opioids, NSAIDs, acetaminophen, gabapentinoids, muscle relaxants, anticonvulsants, and antidepressants are the most used medications for chronic pain and carry significant side effects, including gastric bleeding, hepatotoxicity, stroke, kidney damage, constipation, dizziness, and arrhythmias. Opioids in particular carry the risk of long-term dependence, drug tolerance, and overdose. In 2022, 81,806 people died from opioid overdose in the United States alone. Alternative treatments for chronic pain are critically needed, and nanotechnology has emerged as a promising means of achieving effective long-term analgesia while avoiding the adverse side effects associated with conventional pharmacological agents. Nanotechnology-based treatments include liposomes, Poly Lactic-co-Glycolic Acid (PLGA) and other polymeric nanoparticles, and carbon-based polymers, which can help mitigate those adverse side effects. These nanomaterials can serve as drug delivery systems that facilitate controlled release and drug stability via the encapsulation of free molecules and protein-based drugs, leading to longer-lasting analgesia and minimizing side effects. In this review, we examine the role of nanotechnology in addressing concerns associated with conventional chronic pain treatments and discuss the ongoing efforts to develop novel, nanotechnology-based treatments for chronic pain such as nanocapacitor patches, gene therapy, the use of both viral and non-viral vectors, CRISPR, and scavengers.
Collapse
Affiliation(s)
- Andrew Torpey
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.T.); (A.D.); (W.C.)
| | - Emily Bellow
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (E.B.); (V.S.)
| | - Veronica Samojedny
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (E.B.); (V.S.)
| | - Sukhpreet Ahluwalia
- Department of Surgery, Stony Brook University Hospital, Stony Brook, NY 11794, USA;
| | - Amruta Desai
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.T.); (A.D.); (W.C.)
| | - William Caldwell
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.T.); (A.D.); (W.C.)
| | - Sergio Bergese
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.T.); (A.D.); (W.C.)
- Department of Neurosurgery, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| |
Collapse
|
11
|
Fan X, Xu X, Xia S, Cheng Y, Guo X. Inhibition of Polyphenol Oxidase Activity by Mesoporous Silica Nanoparticles and Multiwalled Carbon Nanotubes Modified with Surfactants. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:24185-24192. [PMID: 39485261 DOI: 10.1021/acs.langmuir.4c03850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Polyphenol oxidase (PPO) is the culprit behind the browning of fruits and vegetables. Therefore, how to reduce the thermal deactivation temperature of PPO or use as few safe reagents as possible to inhibit enzymatic browning has practical significance. Mesoporous silica nanoparticles (MSNs) and multiwalled carbon nanotubes (MWCNTs) are stable and have high biosafety. In the present study, efficient PPO inhibitors were developed based on MSNs and MWCNTs. It is found that after modification with a very small amount of dodecyl trimethylammonium bromide (DTAB, ≥60 μg/mL), MSNs can significantly inhibit the activity of PPO although single MSNs and single DTAB show very limited effect on PPO activity. After modification with a very small amount of sodium dodecyl sulfate (SDS, 5.7-9.5 μg/mL), MWCNTs almost completely inactivate PPO. However, SDS@MSN and DTAB@MWCNT cannot decrease PPO activity significantly.
Collapse
Affiliation(s)
- Xiaonan Fan
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, People's Republic of China
- School of Materials Science and Engineering, China University of Petroleum (East China), Qingdao, Shandong 266580, People's Republic of China
| | - Xin Xu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, People's Republic of China
| | - Shuhuai Xia
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, People's Republic of China
| | - Yanrong Cheng
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, People's Republic of China
| | - Xia Guo
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, People's Republic of China
| |
Collapse
|
12
|
Elbedwehy AM, Wu J, Na HK, Baek A, Jung H, Kwon IH, Lee SW, Kim JH, Lee TG. ROS-responsive charge reversal mesoporous silica nanoparticles as promising drug delivery system for neovascular retinal diseases. J Control Release 2024; 373:224-239. [PMID: 39002796 DOI: 10.1016/j.jconrel.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Intravitreal injection of biodegradable implant drug carriers shows promise in reducing the injection frequency for neovascular retinal diseases. However, current intravitreal ocular devices have limitations in adjusting drug release rates for individual patients, thereby affecting treatment effectiveness. Accordingly, we developed mesoporous silica nanoparticles (MSNs) featuring a surface that reverse its charge in response to reactive oxygen species (ROS) for efficient delivery of humanin peptide (HN) to retinal epithelial cells (ARPE-19). The MSN core, designed with a pore size of 2.8 nm, ensures a high HN loading capacity 64.4% (w/w). We fine-tuned the external surface of the MSNs by incorporating 20% Acetyl-L-arginine (Ar) to create a partial positive charge, while 80% conjugated thioketal (TK) methoxy polyethylene glycol (mPEG) act as ROS gatekeeper. Ex vivo experiments using bovine eyes revealed the immobilization of Ar-MSNs-TK-PEG (mean zeta potential: 2 mV) in the negatively charged vitreous. However, oxidative stress reversed the surface charge to -25 mV by mPEG loss, facilitating the diffusion of the nanoparticles impeded with HN. In vitro studies showed that ARPE-19 cells effectively internalize HN-loaded Ar-MSNs-TK, subsequently releasing the peptide, which offered protection against oxidative stress-induced apoptosis, as evidenced by reduced TUNEL and caspase3 activation. The inhibition of retinal neovascularization was further validated in an in vivo oxygen-induced retinopathy (OIR) mouse model.
Collapse
Affiliation(s)
- Ahmed M Elbedwehy
- Department of Nano Science, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea; Nanotechnology Center, Mansoura University, Mansoura 35516, Egypt
| | - Jun Wu
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hee-Kyung Na
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Ahruem Baek
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Haejin Jung
- Flow Cytometry Core Facility of Research Solution Center, Institute of Basic Science, Daejeon 34126, Republic of Korea
| | - Ik Hwan Kwon
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Sang Won Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Ophthalmology, Seoul National University Hospital, Seoul 03080, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea; Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Tae Geol Lee
- Department of Nano Science, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea.
| |
Collapse
|
13
|
Mohanan S, Sathish CI, Ramadass K, Liang M, Vinu A. Design and Synthesis of Cabazitaxel Loaded Core-Shell Mesoporous Silica Nanoparticles with Different Morphologies for Prostate Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2303269. [PMID: 37386787 DOI: 10.1002/smll.202303269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/06/2023] [Indexed: 07/01/2023]
Abstract
In this work, the synthesis of core-shell ordered mesoporous silica nanoparticles (CSMS) with tunable particle size and shape through a dual surfactant-assisted approach is demonstrated. By varying the synthesis conditions, including the type of the solvent and the concentration of the surfactant, monodispersed and ordered mesoporous silica nanoparticles with tunable particle size (140-600 nm) and morphologies (hexagonal prism (HP), oblong, spherical, and hollow-core) can be realized. Comparative studies of the Cabazitaxel (CBZ)-loaded HP and spherical-shaped CSMS are conducted to evaluate their drug delivery efficiency to PC3 (prostate cancer) cell lines. These nanoparticles showed good biocompatibility and displayed a faster drug release at acidic pH than at basic pH. The cellular uptake of CSMS measured using confocal microscopy, flow cytometry, microplate reader, and ICP-MS (inductively coupled plasma mass spectrometry) techniques in PC3 cell lines revealed a better uptake of CSMS with HP morphology than its spherical counterparts. Cytotoxicity study showed that the anticancer activity of CBZ is improved with a higher free radical production when loaded onto CSMS. These unique materials with tunable morphology can serve as an excellent drug delivery system and will have potential applications for treating various cancers.
Collapse
Affiliation(s)
- Shan Mohanan
- Global Innovative Centre for Advanced Nanomaterials, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - C I Sathish
- Global Innovative Centre for Advanced Nanomaterials, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Kavitha Ramadass
- Global Innovative Centre for Advanced Nanomaterials, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| |
Collapse
|
14
|
Dubey N, Rahimnejad M, Swanson WB, Xu J, de Ruijter M, Malda J, Squarize CH, Castilho RM, Bottino MC. Integration of Melt Electrowritten Polymeric Scaffolds and Bioprinting for Epithelial Healing via Localized Periostin Delivery. ACS Macro Lett 2024; 13:959-965. [PMID: 39024469 DOI: 10.1021/acsmacrolett.4c00240] [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] [Indexed: 07/20/2024]
Abstract
Management of skin injuries imposes a substantial financial burden on patients and hospitals, leading to diminished quality of life. Periostin (rhOSF), an extracellular matrix component, regulates cell function, including a proliferative healing phase, representing a key protein to promote wound healing. Despite its proven efficacy in vitro, there is a lack of scaffolds that facilitate the in situ delivery of rhOSF. In addition, there is a need for a scaffold to not only support cell growth, but also to resist the mechanical forces involved in wound healing. In this work, we synthesized rhOSF-loaded mesoporous nanoparticles (MSNs) and incorporated them into a cell-laden gelatin methacryloyl (GelMA) ink that was bioprinted into melt electrowritten poly(ε-caprolactone) (PCL) microfibrous (MF-PCL) meshes to develop mechanically competent constructs. Diffraction light scattering (DLS) analysis showed a narrow nanoparticle size distribution with an average size of 82.7 ± 13.2 nm. The rhOSF-loaded hydrogels showed a steady and controlled release of rhOSF over 16 days at a daily dose of ∼40 ng/mL. Compared with blank MSNs, the incorporation of rhOSF markedly augmented cell proliferation, underscoring its contribution to cellular performance. Our findings suggest a promising approach to address challenges such as prolonged healing, offering a potential solution for developing robust, biocompatible, and cell-laden grafts for burn wound healing applications.
Collapse
Affiliation(s)
- Nileshkumar Dubey
- Faculty of Dentistry, National University of Singapore, 119077 Singapore
| | - Maedeh Rahimnejad
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jinping Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mylène de Ruijter
- Regenerative Medicine Center Utrecht, 3584 Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, 3584 Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| | - Cristiane H Squarize
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan Ann Arbor, Michigan 48109, United States
| | - Rogerio M Castilho
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan Ann Arbor, Michigan 48109, United States
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
15
|
Godakhindi V, Tarannum M, Dam SK, Vivero-Escoto JL. Mesoporous Silica Nanoparticles as an Ideal Platform for Cancer Immunotherapy: Recent Advances and Future Directions. Adv Healthc Mater 2024; 13:e2400323. [PMID: 38653190 PMCID: PMC11305940 DOI: 10.1002/adhm.202400323] [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: 01/27/2024] [Revised: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Cancer immunotherapy recently transforms the traditional approaches against various cancer malignancies. Immunotherapy includes systemic and local treatments to enhance immune responses against cancer and involves strategies such as immune checkpoints, cancer vaccines, immune modulatory agents, mimetic antigen-presenting cells, and adoptive cell therapy. Despite promising results, these approaches still suffer from several limitations including lack of precise delivery of immune-modulatory agents to the target cells and off-target toxicity, among others, that can be overcome using nanotechnology. Mesoporous silica nanoparticles (MSNs) are investigated to improve various aspects of cancer immunotherapy attributed to the advantageous structural features of this nanomaterial. MSNs can be engineered to alter their properties such as size, shape, porosity, surface functionality, and adjuvanticity. This review explores the immunological properties of MSNs and the use of MSNs as delivery vehicles for immune-adjuvants, vaccines, and mimetic antigen-presenting cells (APCs). The review also details the current strategies to remodel the tumor microenvironment to positively reciprocate toward the anti-tumor immune cells and the use of MSNs for immunotherapy in combination with other anti-tumor therapies including photodynamic/thermal therapies to enhance the therapeutic effect against cancer. Last, the present demands and future scenarios for the use of MSNs for cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Varsha Godakhindi
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Mubin Tarannum
- Division of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Sudip Kumar Dam
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Juan L Vivero-Escoto
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| |
Collapse
|
16
|
Li L, Xie Y, Wang J, Sun Q, Gao M, Li C. Biofilm microenvironment-activated multimodal therapy nanoplatform for effective anti-bacterial treatment and wound healing. Acta Biomater 2024; 183:221-234. [PMID: 38849021 DOI: 10.1016/j.actbio.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
Antimicrobial drug development faces challenges from bacterial resistance, biofilms, and excessive inflammation. Here, we design an intelligent nanoplatform utilizing mesoporous silica nanoparticles doped with copper ions for loading copper sulfide (DM/Cu2+-CuS). The mesoporous silica doped with tetrasulfide bonds responds to the biofilm microenvironment (BME), releasing Cu2+ions, CuS along with hydrogen sulfide (H2S) gas. The release of hydrogen sulfide within 72 h reached 793.5 µM, significantly higher than that observed with conventional small molecule donors. H2S induces macrophages polarization towards the M2 phenotype, reducing inflammation and synergistically accelerating endothelial cell proliferation and migration with Cu2+ions. In addition, H2S disrupts extracellular DNA within biofilms, synergistically photothermal enhanced peroxidase-like activity of CuS to effectively eradicate biofilms. Remarkably, DM-mediated consumption of endogenous glutathione enhances the anti-biofilm activity of H2S and improves oxygen species (ROS) destruction efficiency. The combination of photothermal therapy (PTT), chemodynamic therapy (CDT), and gas treatment achieves sterilization rates of 99.3 % and 99.6 % against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli), respectively, in vitro under 808 nm laser irradiation. Additionally, in vivo experiments demonstrate a significant biosafety and antibacterial potential. In summary, the H2S donor developed in this study exhibits enhanced biocompatibility and controlled release properties. By integrating BME-responsive gas therapy with antibacterial ions, PTT and CDT, a synergistic multimodal strategy is proposed to offer new therapeutic approaches for wound healing. STATEMENT OF SIGNIFICANCE: The advanced DMOS/Cu2+-CuS (DMCC) multimodal therapeutic nanoplatform has been developed for the treatment of drug-resistant bacterial wound infections and has exhibited enhanced therapeutic efficacy through the synergistic effects of photothermal therapy, chemodynamic therapy, Cu2+ions, and H2S. The DMCC exhibited exceptional biocompatibility and could release CuS, Cu2+, and H2S in response to elevated concentrations of glutathione within the biofilm microenvironment. H2S effectively disrupted the biofilm structure. Meanwhile, peroxidase activity of CuS combined with GSH-mediated reduction of Cu2+ to Cu+ generated abundant hydroxyl radicals under acidic conditions, leading to efficient eradication of pathogenic bacteria. Furthermore, both H2S and Cu2+ could modulate M2 macrophages polarization and regulate immune microenvironment dynamics. These strategies collectively provided a novel approach for developing antibacterial nanomedical platforms.
Collapse
Affiliation(s)
- Lei Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Yulin Xie
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Junrong Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Qianqian Sun
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Minghong Gao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China.
| | - Chunxia Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China.
| |
Collapse
|
17
|
Wang T, Nakata H, Shen B, Jiao Z, Yokota K, Kuroda S, Kasugai S, Marukawa E. Involvement of miR-199a-5p-loaded mesoporous silica nanoparticle-polyethyleneimine-KALA in osteogenic differentiation. J Dent Sci 2024; 19:1506-1514. [PMID: 39035341 PMCID: PMC11259637 DOI: 10.1016/j.jds.2024.01.007] [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: 12/24/2023] [Revised: 01/08/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose While there are numerous reports on surgical techniques and materials for bone grafting, limited methods are available to enhance the body's inherent capacity to heal bones. Here we investigated microRNA-199a (miR-199a), a molecular that promotes osteoblast differentiation and bone healing. Materials and methods To construct a miR-199a delivery complex, miR-199a-5p mimics were coated with mesoporous silica nanoparticles (MSNs) following modified with polyethyleneimine (PEI) and peptide WEAKLAKALAKALAKHLAKALAKALKACEA (KALA) to obtain 199a-5p-loaded MSN-PEI-KALA. Nanoparticle complexes are assessed for particle size and zeta potential using transmission electron microscopy and dynamic light scattering. Then MC3T3-E1 cells are exposed to MSN_miR-199a-5p @PEI-KALA. The impact of MSN_miR-199a-5p@PEI-KALA at varying concentrations on cell viability is assessed using Cell Counting Kit-8. Cell uptake and distribution were analyzed by double fluorescent staining with fluorescein amidite-labeled MSN_miR-199a@PEI-KALA and lysosome labeling. On day 7 after osteogenic induction, alkaline phosphatase (ALP) staining was conducted. Results The findings indicated that the nanoparticle complexes encapsulating PEI and peptide exhibited an augmentation in both particle size and zeta potential. At a dosage of 10 μg/mL, MSN_miR-199a@PEI-KALA displayed the lowest cytotoxicity compared to the control group. MC3T3-E1 cells treated with MSN_miR-199a-5p@PEI-KALA exhibited intensified ALP staining and elevated mRNA expression levels of ALP, runt-related transcription factor 2, and osteopontin, suggesting the involvement of miR-199a-5p-loaded MSN-PEI-KALA in osteogenic differentiation. Conclusion The successful construction of the delivering complex MSN_miR-199a@PEI-KALA in present research highlights the promise of this biomaterial carrier for the application of miRNAs in treating bone defects.
Collapse
Affiliation(s)
- Tianyue Wang
- Department of Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Regenerative and Reconstructive Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidemi Nakata
- Department of Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Regenerative and Reconstructive Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Bing Shen
- Department of Physiology, School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Ziying Jiao
- Department of Physiology, School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Kaori Yokota
- Department of Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Regenerative and Reconstructive Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Kuroda
- Department of Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Regenerative and Reconstructive Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Improvement of Gnatho-oral Function, Department of Stomatognathic, Faculty of Dental Medicine, Hokkaido University, Hokkaido, Japan
| | - Shohei Kasugai
- Department of Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Regenerative and Reconstructive Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Dental Clinic, Southern TOHOKU General Hospital, Fukushima, Japan
| | - Eriko Marukawa
- Department of Regenerative and Reconstructive Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
18
|
Abdelwahab WM, Le-Vinh B, Riffey A, Hicks L, Buhl C, Ettenger G, Jackson KJ, Weiss AM, Miller S, Ryter K, Evans JT, Burkhart DJ. Promotion of Th17 Polarized Immunity via Co-Delivery of Mincle Agonist and Tuberculosis Antigen Using Silica Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:3877-3889. [PMID: 38832760 DOI: 10.1021/acsabm.4c00245] [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] [Indexed: 06/05/2024]
Abstract
Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.
Collapse
Affiliation(s)
- Walid M Abdelwahab
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Bao Le-Vinh
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Alexander Riffey
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Linda Hicks
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Cassandra Buhl
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - George Ettenger
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Konner J Jackson
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Adam M Weiss
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Shannon Miller
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Kendal Ryter
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - David J Burkhart
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| |
Collapse
|
19
|
Zhang Y, Lin X, Chen X, Fang W, Yu K, Gu W, Wei Y, Zheng H, Piao J, Li F. Strategies to Regulate the Degradation and Clearance of Mesoporous Silica Nanoparticles: A Review. Int J Nanomedicine 2024; 19:5859-5878. [PMID: 38887691 PMCID: PMC11182361 DOI: 10.2147/ijn.s451919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/26/2024] [Indexed: 06/20/2024] Open
Abstract
Mesoporous silica nanoparticles (MSNs) have attracted extensive attention as drug delivery systems because of their unique meso-structural features (high specific surface area, large pore volume, and tunable pore structure), easily modified surface, high drug-loading capacity, and sustained-release profiles. However, the enduring and non-specific enrichment of MSNs in healthy tissues may lead to toxicity due to their slow degradability and hinder their clinical application. The emergence of degradable MSNs provided a solution to this problem. The understanding of strategies to regulate degradation and clearance of these MSNs for promoting clinical trials and expanding their biological applications is essential. Here, a diverse variety of degradable MSNs regarding considerations of physiochemical properties and doping strategies of degradation, the biodistribution of MSNs in vivo, internal clearance mechanism, and adjusting physical parameters of clearance are highlighted. Finally, an overview of these degradable and clearable MSNs strategies for biosafety is provided along with an outlook of the encountered challenges.
Collapse
Affiliation(s)
- Yuelin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Xue Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Xinxin Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Weixiang Fang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Kailing Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Wenting Gu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Hangsheng Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Jigang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Fanzhu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
20
|
Shen Y, Yu Y, Zhang X, Hu B, Wang N. Progress of nanomaterials in the treatment of thrombus. Drug Deliv Transl Res 2024; 14:1154-1172. [PMID: 38006448 DOI: 10.1007/s13346-023-01478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Thrombus has long been the major contributor of death and disability because it can cause adverse effects to varying degrees on the body, resulting in vascular blockage, embolism, heart valve deformation, widespread bleeding, etc. However, clinically, conventional thrombolytic drug treatments have hemorrhagic complication risks and easy to miss the best time of treatment window. Thus, it is an urgent need to investigate newly alternative treatment strategies that can reduce adverse effects and improve treatment effectiveness. Drugs based on nanomaterials act as a new biomedical strategy and promising tools, and have already been investigated for both diagnostic and therapeutic purposes in thrombus therapy. Recent studies have some encouraging progress. In the present review, we primarily concern with the latest developments in the areas of nanomedicines targeting thrombosis therapy. We present the thrombus' formation, characteristics, and biomarkers for diagnosis, overview recent emerging nanomedicine strategies for thrombus therapy, and focus on the future design directions, challenges, and prospects in the nanomedicine application in thrombus therapy.
Collapse
Affiliation(s)
- Yetong Shen
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China
- College of Life and Health Sciences, Northeastern University, Shenyang, 110167, China
| | - Yang Yu
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xin Zhang
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China
| | - Bo Hu
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China.
| | - Ning Wang
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China.
- Department of Forensic Medicine, China Medical University, No.77 Puhe Road, Shenyang, 110122, China.
| |
Collapse
|
21
|
Arshad T, Shoaib Khan HM, Akhtar N, Hanan H, Hussain MD, Kazi M. Structural elucidation and development of azelaic acid loaded mesoporous silica nanoparticles infused gel: Revolutionizing nanodrug delivery for cosmetics and pharmaceuticals. Heliyon 2024; 10:e29460. [PMID: 38665554 PMCID: PMC11043944 DOI: 10.1016/j.heliyon.2024.e29460] [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: 02/02/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
This research aimed to enhance dermal delivery and optimize depigmentation therapy by designing mesoporous silica nanoparticles (MSNs) encapsulating azelaic acid (AZA) within a gel matrix. The MSNs were prepared using the sol-gel method. After subsequent processes, including acid extraction and drug loading, were then elucidated through PDI, size, zeta-potential, entrapment efficiency, nitrogen adsorption assay, FE-SEM, thermogravimetric analysis, differential scanning calorimetry, Fourier transform infrared spectroscopy, X-ray diffraction, and tyrosinase inhibition assay, were employed to assess the formulation. In-vitro stability tests for both AZA-MSN gel (AZCG) and AZA-loaded mesoporous silica gel (AZMG) were conducted at 8 °C, 25 °C, 40 °C, and 40 °C + 75 % RH, encompassing assessments of color, liquefaction, pH, and conductivity. Our findings showed a notable entrapment efficiency of 93.46 % for AZA-MSNs, with FE-SEM illustrating porous spherical MSNs. The particle size of AZA-MSNs was determined to be 211.9 nm, with a pore size of 2.47 nm and XRD analysis confirmed the amorphous state of AZA within the MSN carriers. Rheology examination indicated a non-Newtonian flow, while ex-vivo rat skin permeation studies conducted in a phosphate buffer (pH = 5.5) demonstrated a biphasic release pattern with 85.53 % cumulative drug permeation for AZA-MSNs. Overall, the study endorse the potential of AZA-MSNs as an efficacious and stable formulation for AZA delivery, highlighting their promise in addressing pigmentation concerns compared to conventional approaches.
Collapse
Affiliation(s)
- Tahreem Arshad
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Haji Muhammad Shoaib Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Naveed Akhtar
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Hanasul Hanan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Muhammad Delwar Hussain
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, USA
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box-2457, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
22
|
Broering MF, Oseliero Filho PL, Borges PP, da Silva LCC, Knirsch MC, Xavier LF, Scharf P, Sandri S, Stephano MA, de Oliveira FA, Sayed IM, Gamarra LF, Das S, Fantini MCA, Farsky SHP. Development of Ac2-26 Mesoporous Microparticle System as a Potential Therapeutic Agent for Inflammatory Bowel Diseases. Int J Nanomedicine 2024; 19:3537-3554. [PMID: 38638365 PMCID: PMC11024051 DOI: 10.2147/ijn.s451589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction Inflammatory bowel diseases (IBDs) disrupt the intestinal epithelium, leading to severe chronic inflammation. Current therapies cause adverse effects and are expensive, invasive, and ineffective for most patients. Annexin A1 (AnxA1) is a pivotal endogenous anti-inflammatory and tissue repair protein in IBD. Nanostructured compounds loading AnxA1 or its active N-terminal mimetic peptides improve IBD symptomatology. Methods To further explore their potential as a therapeutic candidate, the AnxA1 N-terminal mimetic peptide Ac2-26 was incorporated into SBA-15 ordered mesoporous silica and covered with EL30D-55 to deliver it by oral treatment into the inflamed gut. Results The systems SBA-Ac2-26 developed measurements revealed self-assembled rod-shaped particles, likely on the external surface of SBA-15, and 88% of peptide incorporation. SBA-15 carried the peptide Ac2-26 into cultured Raw 264.7 macrophages and Caco-2 epithelial cells. Moreover, oral administration of Eudragit-SBA-15-Ac2-26 (200 μg; once a day; for 4 days) reduced colitis clinical symptoms, inflammation, and improved epithelium recovery in mice under dextran-sodium sulfate-induced colitis. Discussion The absorption of SBA-15 in gut epithelial cells is typically low; however, the permeable inflamed barrier can enable microparticles to cross, being phagocyted by macrophages. These findings suggest that Ac2-26 is successfully delivered and binds to its receptors in both epithelial and immune cells, aligning with the clinical results. Conclusion Our findings demonstrate a simple and cost-effective approach to delivering Ac2-26 orally into the inflamed gut, highlighting its potential as non-invasive IBD therapy.
Collapse
Affiliation(s)
- Milena Fronza Broering
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Pedro Leonidas Oseliero Filho
- Department of Applied Physics, Physics Institute, University of Sao Paulo, São Paulo, Brazil
- Materials Innovation Factory, University of Liverpool, Liverpool, MSY, UK
| | - Pâmela Pacassa Borges
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Marcos Camargo Knirsch
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luana Filippi Xavier
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marco Antonio Stephano
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Fernando Anselmo de Oliveira
- Instituto do Cérebro, Instituto Israelita de Ensino e Pesquisa, Sociedade Beneficente Israelita Brasileira Hospital Albert Einstein, São Paulo, SP, Brazil
| | - Ibrahim M Sayed
- Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Lionel Fernel Gamarra
- Instituto do Cérebro, Instituto Israelita de Ensino e Pesquisa, Sociedade Beneficente Israelita Brasileira Hospital Albert Einstein, São Paulo, SP, Brazil
| | - Soumita Das
- Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Márcia C A Fantini
- Department of Applied Physics, Physics Institute, University of Sao Paulo, São Paulo, Brazil
| | - Sandra H P Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
23
|
Turky NO, Abdelmonem NA, Tammam SN, Gad MZ, Breitinger HG, Breitinger U. Antibacterial and in vitro anticancer activities of the antimicrobial peptide NRC-07 encapsulated in chitosan nanoparticles. J Pept Sci 2024; 30:e3550. [PMID: 37853814 DOI: 10.1002/psc.3550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/09/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
Antimicrobial peptides (AMPs) are promising alternatives to conventional antibiotics and chemotherapy in the treatment of multidrug-resistant pathogens and drug-resistant cancers. Clinical application of AMPs is limited due to low stability and inefficient transport. Encapsulation in nanocarriers may improve their therapeutic potential. Chitosan nanoparticles (CS-NPs) are efficient carriers for proteins and peptides, improving the treatment of microbial infections and targeted drug delivery. We examined toxicity against cancer cell lines and antibacterial activities of the pleurocidin-like AMP NRC-07 upon encapsulation in CS-NPs by ionotropic gelation. The biological activities of various formulations of free and encapsulated NRC-07 and free nanoparticles were evaluated against Pseudomonas aeruginosa and breast cancer cells, using assays for cell viability and lactate dehydrogenase cytolysis with non-cancer cell lines as controls. NRC-07-containing nanoparticles decreased the bacterial and cancer cell viability in a concentration-dependent manner. Activities of encapsulated peptide were >2-fold higher than those of free NRC-07 peptide. Unloaded CS-NPs and free peptide were not cytotoxic against control cells. Encapsulation of NRC-07 into CS-NPs enhanced the antibacterial and selective cytotoxicity of the peptide, possibly enhancing anticancer activities. Encapsulation presents a promising tool for the development of efficient drug delivery systems.
Collapse
Affiliation(s)
- Nancy O Turky
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, Egypt
| | - Noura A Abdelmonem
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, Egypt
| | - Salma N Tammam
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, Egypt
| | | | - Hans-Georg Breitinger
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, Egypt
| | - Ulrike Breitinger
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, Egypt
| |
Collapse
|
24
|
Sarkar A, Sarkhel S, Bisht D, Jaiswal A. Cationic dextrin nanoparticles for effective intracellular delivery of cytochrome C in cancer therapy. RSC Chem Biol 2024; 5:249-261. [PMID: 38456040 PMCID: PMC10915965 DOI: 10.1039/d3cb00090g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 11/19/2023] [Indexed: 03/09/2024] Open
Abstract
Intracellular protein delivery shows promise as a selective and specific approach to cancer therapy. However, a major challenge is posed by delivering proteins into the target cells. Despite the development of nanoparticle (NP)-based approaches, a versatile and biocompatible delivery system that can deliver active therapeutic cargo into the cytosol while escaping endosome degradation remains elusive. In order to overcome these challenges, a polymeric nanocarrier was prepared using cationic dextrin (CD), a biocompatible and biodegradable polymer, to encapsulate and deliver cytochrome C (Cyt C), a therapeutic protein. The challenge of endosomal escape of the nanoparticles was addressed by co-delivering the synthesized NP construct with chloroquine, which enhances the endosomal escape of the therapeutic protein. No toxicity was observed for both CD NPs and chloroquine at the concentration tested in this study. Spectroscopic investigations confirmed that the delivered protein, Cyt C, was structurally and functionally active. Additionally, the delivered Cyt C was able to induce apoptosis by causing depolarization of the mitochondrial membrane in HeLa cells, as evidenced by flow cytometry and microscopic observations. Our findings demonstrate that an engineered delivery system using CD NPs is a promising platform in nanomedicine for protein delivery applications.
Collapse
Affiliation(s)
- Ankita Sarkar
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| | - Sanchita Sarkhel
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| | - Deepali Bisht
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| | - Amit Jaiswal
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| |
Collapse
|
25
|
Li K, Chen W, Ma L, Yan L, Wang B. Approaches for reducing chemo/radiation-induced cardiotoxicity by nanoparticles. ENVIRONMENTAL RESEARCH 2024; 244:117264. [PMID: 37776941 DOI: 10.1016/j.envres.2023.117264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Nanoparticles are fascinating and encouraging carriers for cancer treatment due to their extraordinary properties and potential applications in targeted drug delivery, treatment, and diagnosis. Experimental studies including in vitro and in vivo examinations show that nanoparticles can cause a revolution in different aspects of cancer therapy. Normal tissue toxicity and early and late consequences are the major limitations of cancer therapy by radiotherapy and chemotherapy. However, the delivery of drugs into tumors or reducing the accumulation of drugs in normal tissues can permit a more satisfactory response of malignancies to therapy with more inferior side effects. Cardiac toxicity is one of the major problems for chemotherapy and radiotherapy. Therefore, several experimental studies have been performed to minimize the degenerative impacts of cancer treatment on the heart and also enhance the influences of radiotherapy and chemotherapy agents in cancers. This review article emphasizes the benefits of nanoparticle-based drug delivery techniques, including minimizing the exposure of the heart to anticancer drugs, enhancing the accumulation of drugs in cancers, and expanding the effectiveness of radiotherapy. The article also discusses the challenges and problems accompanied with nanoparticle-based drug delivery techniques such as toxicity, which need to be addressed through further research. Moreover, the article emphasizes the importance of developing safe and effective nanoparticle-based therapies that can be translated into clinical practice.
Collapse
Affiliation(s)
- Ketao Li
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Wan Chen
- Department of Cardiology, Jiulongpo First People's Hospital, Chongqing, 400051, China
| | - Liping Ma
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Laixing Yan
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Bing Wang
- Department of Cardiology, Zouping People's Hospital, Zouping, shandong, 256299, China.
| |
Collapse
|
26
|
Beitzinger B, Schmid R, Jung C, Tiwary K, Hermann P, Jacob T, Lindén M. Confinement and Polarity Effects on the Peptide Packing Density on Mesoporous Silica Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:4294-4305. [PMID: 38346113 PMCID: PMC10905996 DOI: 10.1021/acs.langmuir.3c03513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/28/2024]
Abstract
The adsorption of cationic peptide JM21 onto different mesoporous silica nanoparticles (MSNs) from an aqueous solution was studied as a function of pH. In agreement with the literature, the highest loading degrees could be achieved at pH close to the isoelectric point of the peptide where the peptide-peptide repulsion is minimum. However, mesopore size, mesopore geometry, and surface polarity all had an influence on the peptide adsorption in terms of both affinity and maximum loading at a given pH. This adsorption behavior could largely be explained by a combination of pH-dependent electrostatic interactions and confinement effects. It is demonstrated that hydrophobic interactions enhance the degree of peptide adsorption under pH conditions where the electrostatic attraction was absent in the case of mesoporous organosilica nanoparticles (MONs). The lower surface concentration of silanol groups for MON led to a lower level of peptide adsorption under optimum pH conditions compared to all-silica particles. Finally, the study confirmed the protective role of MSNs in preserving the biological activity of JM#21 against enzymatic degradation, even for large-pore MSNs, emphasizing their potential as nanocarriers for therapeutic peptides. By integrating experimental findings with theoretical modeling, this research elucidates the complex interplay of factors that influence peptide-silica interactions, providing vital insights for optimizing peptide loading and stabilization in biomedical applications.
Collapse
Affiliation(s)
- Bastian Beitzinger
- Institute
of Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, Ulm 89081, Germany
| | - Roman Schmid
- Institute
of Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, Ulm 89081, Germany
| | - Christoph Jung
- Institute
of Electrochemistry, Ulm University, Albert-Einstein-Allee 47, Ulm 89081, Germany
| | - Kanishka Tiwary
- Department
of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89070, Germany
| | - Patrick Hermann
- Department
of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89070, Germany
| | - Timo Jacob
- Institute
of Electrochemistry, Ulm University, Albert-Einstein-Allee 47, Ulm 89081, Germany
| | - Mika Lindén
- Institute
of Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, Ulm 89081, Germany
| |
Collapse
|
27
|
Howaili F, Saadabadi A, Mäkilä E, Korotkova E, Eklund PC, Salo-Ahen OMH, Rosenholm JM. Investigating the Effectiveness of Different Porous Nanoparticles as Drug Carriers for Retaining the Photostability of Pinosylvin Derivative. Pharmaceutics 2024; 16:276. [PMID: 38399330 PMCID: PMC10892027 DOI: 10.3390/pharmaceutics16020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Pinosylvin monomethyl ether (PsMME) is a natural compound known for its valuable bioactive properties, including antioxidant and anti-inflammatory effects. However, PsMME's susceptibility to photodegradation upon exposure to ultraviolet (UV) radiation poses a significant limitation to its applications in the pharmaceutical field. This study, for the first time, introduces a strategy to enhance the photostability of PsMME by employing various nanoformulations. We utilized mesoporous silica nanoparticles (MSNs) coated with polydopamine via a poly(ethylene imine) layer (PDA-PEI-MSNs), thermally carbonized porous silicon nanoparticles (TCPSi), and pure mesoporous polydopamine nanoparticles (MPDA). All these nanocarriers exhibit unique characteristics, including the potential for shielding the drug from UV light, which makes them promising for enhancing the photostability of loaded drugs. Here, these three nanoparticles were synthesized and their morphological and physicochemical properties, including size and ζ-potential, were characterized. They were subsequently loaded with PsMME, and the release profiles and kinetics of all three nanoformulations were determined. To assess their photoprotection ability, we employed gas chromatography with a flame ionization detector (GC-FID) and gas chromatography-mass spectrometry (GC-MS) to assess the recovery percentage of loaded PsMME before and after UV exposure for each nanoformulation. Our findings reveal that MPDA exhibits the highest protection ability, with a remarkable 90% protection against UV light on average. This positions MPDA as an ideal carrier for PsMME, and by extension, potentially for other photolabile drugs as well. As a final confirmation of its suitability as a drug nanocarrier, we conducted cytotoxicity evaluations of PsMME-loaded MPDA, demonstrating dose-dependent drug toxicity for this formulation.
Collapse
Affiliation(s)
- Fadak Howaili
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland; (F.H.); (A.S.); (O.M.H.S.-A.)
| | - Atefeh Saadabadi
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland; (F.H.); (A.S.); (O.M.H.S.-A.)
- Laboratory of Molecular Science and Engineering, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland;
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, 20014 Turku, Finland;
| | - Ekaterina Korotkova
- Laboratory of Natural Materials Technology, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland;
| | - Patrik C. Eklund
- Laboratory of Molecular Science and Engineering, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland;
| | - Outi M. H. Salo-Ahen
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland; (F.H.); (A.S.); (O.M.H.S.-A.)
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland; (F.H.); (A.S.); (O.M.H.S.-A.)
| |
Collapse
|
28
|
Dourado D, Miranda JA, de Oliveira MC, Freire DT, Xavier-Júnior FH, Paredes-Gamero EJ, Alencar ÉDN. Recent Trends in Curcumin-Containing Inorganic-Based Nanoparticles Intended for In Vivo Cancer Therapy. Pharmaceutics 2024; 16:177. [PMID: 38399238 PMCID: PMC10891663 DOI: 10.3390/pharmaceutics16020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Curcumin is a natural compound that has been widely investigated thanks to its various biological properties, including antiproliferative. This molecule acts on different cancers such as lung, breast, pancreatic, colorectal, etc. However, the bioactive actions of curcumin have limitations when its physicochemical properties compromise its pharmacological potential. As a therapeutic strategy against cancer, curcumin has been associated with inorganic nanoparticles. These nanocarriers are capable of delivering curcumin and offering physicochemical properties that synergistically enhance anticancer properties. This review highlights the different types of curcumin-based inorganic nanoparticles and discusses their physicochemical properties and in vivo anticancer activity in different models of cancer.
Collapse
Affiliation(s)
- Douglas Dourado
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife 50670-420, PE, Brazil;
| | - Júlio Abreu Miranda
- Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal 59010-180, RN, Brazil; (J.A.M.); (M.C.d.O.)
| | - Matheus Cardoso de Oliveira
- Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal 59010-180, RN, Brazil; (J.A.M.); (M.C.d.O.)
| | - Danielle Teixeira Freire
- College of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil; (D.T.F.); (E.J.P.-G.)
| | - Francisco Humberto Xavier-Júnior
- Laboratory of Pharmaceutical Biotechnology (BioTecFarm), Department of Pharmacy, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil;
| | - Edgar Julian Paredes-Gamero
- College of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil; (D.T.F.); (E.J.P.-G.)
| | - Éverton do Nascimento Alencar
- College of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil; (D.T.F.); (E.J.P.-G.)
| |
Collapse
|
29
|
Li F, Wang Y, Chen D, Du Y. Nanoparticle-Based Immunotherapy for Reversing T-Cell Exhaustion. Int J Mol Sci 2024; 25:1396. [PMID: 38338674 PMCID: PMC10855737 DOI: 10.3390/ijms25031396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
T-cell exhaustion refers to a state of T-cell dysfunction commonly observed in chronic infections and cancer. Immune checkpoint molecules blockading using PD-1 and TIM-3 antibodies have shown promising results in reversing exhaustion, but this approach has several limitations. The treatment of T-cell exhaustion is still facing great challenges, making it imperative to explore new therapeutic strategies. With the development of nanotechnology, nanoparticles have successfully been applied as drug carriers and delivery systems in the treatment of cancer and infectious diseases. Furthermore, nanoparticle-based immunotherapy has emerged as a crucial approach to reverse exhaustion. Here, we have compiled the latest advances in T-cell exhaustion, with a particular focus on the characteristics of exhaustion that can be targeted. Additionally, the emerging nanoparticle-based delivery systems were also reviewed. Moreover, we have discussed, in detail, nanoparticle-based immunotherapies that aim to reverse exhaustion, including targeting immune checkpoint blockades, remodeling the tumor microenvironment, and targeting the metabolism of exhausted T cells, etc. These data could aid in comprehending the immunopathogenesis of exhaustion and accomplishing the objective of preventing and treating chronic diseases or cancer.
Collapse
Affiliation(s)
- Fei Li
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Yahong Wang
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (Y.W.); (D.C.)
| | - Dandan Chen
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (Y.W.); (D.C.)
| | - Yunjie Du
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
30
|
Sun P, Cheng B, Ru J, Li X, Fang G, Xie Y, Shi G, Hou J, Zhao L, Gan L, Ma L, Liang C, Chen Y, Li Z. Nano-carrier DMSN for effective multi-antigen vaccination against SARS-CoV-2. J Nanobiotechnology 2024; 22:11. [PMID: 38167103 PMCID: PMC10763455 DOI: 10.1186/s12951-023-02271-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
The pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has had a profound impact on the global health and economy. While mass vaccination for herd immunity is effective, emerging SARS-CoV-2 variants can evade spike protein-based COVID-19 vaccines. In this study, we develop a new immunization strategy by utilizing a nanocarrier, dendritic mesoporous silica nanoparticle (DMSN), to deliver the receptor-binding domain (RBD) and conserved T-cell epitope peptides (DMSN-P-R), aiming to activate both humoral and cellular immune responses in the host. The synthesized DMSN had good uniformity and dispersion and showed a strong ability to load the RBD and peptide antigens, enhancing their uptake by antigen-presenting cells (APCs) and promoting antigen delivery to lymph nodes. The DMSN-P-R vaccine elicited potent humoral immunity, characterized by highly specific RBD antibodies. Neutralization tests demonstrated significant antibody-mediated neutralizing activity against live SARS-CoV-2. Crucially, the DMSN-P-R vaccine also induced robust T-cell responses that were specifically stimulated by the RBD and conserved T-cell epitope peptides of SARS-CoV-2. The DMSN demonstrated excellent biocompatibility and biosafety in vitro and in vivo, along with degradability. Our study introduces a promising vaccine strategy that utilizes nanocarriers to deliver a range of antigens, effectively enhancing both humoral and cellular immune responses to prevent virus transmission.
Collapse
Affiliation(s)
- Peng Sun
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Bingsheng Cheng
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jiaxi Ru
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Xiaoyan Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310051, China
| | - Guicun Fang
- Microscopy Core Facility, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Yinli Xie
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Guangjiang Shi
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Jichao Hou
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Longwei Zhao
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Lipeng Gan
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Lina Ma
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Chao Liang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| | - Yin Chen
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310051, China.
| | - Zhiyong Li
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China.
| |
Collapse
|
31
|
Kirla H, Henry DJ, Jansen S, Thompson PL, Hamzah J. Use of Silica Nanoparticles for Drug Delivery in Cardiovascular Disease. Clin Ther 2023; 45:1060-1068. [PMID: 37783646 DOI: 10.1016/j.clinthera.2023.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
PURPOSE Cardiovascular disease (CVD) is the leading cause of death worldwide. The current CVD therapeutic drugs require long-term treatment with high doses, which increases the risk of adverse effects while offering only marginal treatment efficacy. Silica nanoparticles (SNPs) have been proven to be an efficient drug delivery vehicle for numerous diseases, including CVD. This article reviews recent progress and advancement in targeted delivery for drugs and diagnostic and theranostic agents using silica nanoparticles to achieve therapeutic efficacy and improved detection of CVD in clinical and preclinical settings. METHODS A search of PubMed, Scopus, and Google Scholar databases from 1990 to 2023 was conducted. Current clinical trials on silica nanoparticles were identified through ClinicalTrials.gov. Search terms include silica nanoparticles, cardiovascular diseases, drug delivery, and therapy. FINDINGS Silica nanoparticles exhibit biocompatibility in biological systems, and their shape, size, surface area, and surface functionalization can be customized for the safe transport and protection of drugs in blood circulation. These properties also enable effective drug uptake in specific tissues and controlled drug release after systemic, localized, or oral delivery. A range of silica nanoparticles have been used as nanocarrier for drug delivery to treat conditions such as atherosclerosis, hypertension, ischemia, thrombosis, and myocardial infarction. IMPLICATIONS The use of silica nanoparticles for drug delivery and their ongoing development has emerged as a promising strategy to improve the effectiveness of drugs, imaging agents, and theranostics with the potential to revolutionize the treatment of CVD.
Collapse
Affiliation(s)
- Haritha Kirla
- Targeted Drug Delivery, Imaging & Therapy Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Chemistry and Physics, College of Science, Health, Engineering and Education, Murdoch University, Western Australia, Australia.
| | - David J Henry
- Chemistry and Physics, College of Science, Health, Engineering and Education, Murdoch University, Western Australia, Australia
| | - Shirley Jansen
- Targeted Drug Delivery, Imaging & Therapy Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Curtin Health Innovation Research Institute and Curtin Medical School, Curtin University, Perth, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia; Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Peter L Thompson
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
| | - Juliana Hamzah
- Targeted Drug Delivery, Imaging & Therapy Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Curtin Health Innovation Research Institute and Curtin Medical School, Curtin University, Perth, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.
| |
Collapse
|
32
|
Salarkia E, Mehdipoor M, Molaakbari E, Khosravi A, Sazegar MR, Salari Z, Rad I, Dabiri S, Joukar S, Sharifi I, Ren G. Exploring mesoporous silica nanoparticles as oral insulin carriers: In-silico and in vivo evaluation. Heliyon 2023; 9:e20430. [PMID: 37810809 PMCID: PMC10556789 DOI: 10.1016/j.heliyon.2023.e20430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023] Open
Abstract
The advancements in nanoscience have brought attention to the potential of utilizing nanoparticles as carriers for oral insulin administration. This study aims to investigate the effectiveness of synthesized polymeric mesoporous silica nanoparticles (MSN) as carriers for oral insulin and their interactions with insulin and IR through in-silico docking. Diabetic rats were treated with various MSN samples, including pure MSN, Amin-grafted MSN/PEG/Insulin (AMPI), Al-grafted MSN/PEG/Insulin (AlMPI), Zinc-grafted MSN/PEG/Insulin (ZNPI), and Co-grafted MSN/PEG/Insulin (CMPI). The nanocomposites were synthesized using a hybrid organic-inorganic method involving MSNs, graphene oxide, and insulin. Characterization of the nanocomposites was conducted using X-ray diffraction (XRD), Fourier-transform infrared (FTIR) spectroscopy, and scanning electron microscopy (SEM). In vivo tests included the examination of blood glucose levels and histopathological parameters of the liver and pancreas in type 1 diabetic rats. The MSN family demonstrated a significant reduction in blood glucose levels compared to the diabetic control group (p < 0.001). The synthesized nanocomposites exhibited safety, non-toxicity, fast operation, self-repairing pancreas, cost-effectiveness, and high efficiency in the oral insulin delivery system. In the in-silico study, Zn-grafted MSN, Co-grafted MSN, and Al-grafted MSN were selected. Docking results revealed strong interactions between MSN compounds and insulin and IR, characterized by the formation of hydrogen bonds and high binding energy. Notably, Co-grafted MSN showed the highest docking scores of -308.171 kcal/mol and -337.608 kcal/mol to insulin and IR, respectively. These findings demonstrate the potential of polymeric MSN as effective carriers for oral insulin, offering promising prospects for diabetes treatment.
Collapse
Affiliation(s)
- Ehsan Salarkia
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdis Mehdipoor
- Faculty of Chemistry, North Tehran Branch, Islamic Azad University, Hakimiyeh, Tehran, Iran
| | - Elahe Molaakbari
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Sazegar
- Faculty of Chemistry, North Tehran Branch, Islamic Azad University, Hakimiyeh, Tehran, Iran
| | - Zohreh Salari
- Department of Obstetrics and Gynecology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Iman Rad
- Afzalipour School of Medicine & Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Afzalipour School of Medicine & Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Siyavash Joukar
- Neuroscience Research Center, Institute of Basic and Clinical Physiology Sciences, Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Guogang Ren
- School of Engineering and Computer Science, University of Hertfordshire, Hatfield, AL10 9AB, UK
| |
Collapse
|
33
|
Eom YS, Park JH, Kim TH. Recent Advances in Stem Cell Differentiation Control Using Drug Delivery Systems Based on Porous Functional Materials. J Funct Biomater 2023; 14:483. [PMID: 37754897 PMCID: PMC10532449 DOI: 10.3390/jfb14090483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
The unique characteristics of stem cells, which include self-renewal and differentiation into specific cell types, have paved the way for the development of various biomedical applications such as stem cell therapy, disease modelling, and drug screening. The establishment of effective stem cell differentiation techniques is essential for the effective application of stem cells for various purposes. Ongoing research has sought to induce stem cell differentiation using diverse differentiation factors, including chemicals, proteins, and integrin expression. These differentiation factors play a pivotal role in a variety of applications. However, it is equally essential to acknowledge the potential hazards of uncontrolled differentiation. For example, uncontrolled differentiation can give rise to undesirable consequences, including cancerous mutations and stem cell death. Therefore, the development of innovative methods to control stem cell differentiation is crucial. In this review, we discuss recent research cases that have effectively utilised porous functional material-based drug delivery systems to regulate stem cell differentiation. Due to their unique substrate properties, drug delivery systems based on porous functional materials effectively induce stem cell differentiation through the steady release of differentiation factors. These ground-breaking techniques hold considerable promise for guiding and controlling the fate of stem cells for a wide range of biomedical applications, including stem cell therapy, disease modelling, and drug screening.
Collapse
Affiliation(s)
| | | | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea; (Y.-S.E.); (J.-H.P.)
| |
Collapse
|
34
|
Potnis CS, Grapperhaus CA, Gupta G. Investigating BioCaRGOS, a Sol-Gel Matrix for the Stability of Heme Proteins under Enzymatic Degradation and Low pH. ACS OMEGA 2023; 8:32053-32059. [PMID: 37692240 PMCID: PMC10483679 DOI: 10.1021/acsomega.3c04012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/02/2023] [Indexed: 09/12/2023]
Abstract
There have been significant advances in the development of vaccines for the prevention of various infectious diseases in the last few decades. These vaccines are mainly composed of proteins and nucleic acids. Poor handling and storage, exposure to high temperatures that lead to enzymatic degradation, pH variation, and various other stresses can denature the proteins or nucleic acids present in any vaccine formulation. Therefore, it is necessary to maintain a proper environment to preserve the integrity of biospecimens. To overcome these challenges, we report a practical and user-friendly approach for sol-gels called "BioCaRGOS" that can stabilize heme proteins not only in the presence of degrading enzymes and acidic pH but simultaneously maintain stability at room temperature. Heme proteins, such as myoglobin and cytochrome c, have been used for this study.
Collapse
Affiliation(s)
- Chinmay S Potnis
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| | - Craig A Grapperhaus
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| | - Gautam Gupta
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky 40292, United States
| |
Collapse
|
35
|
P. Singh R, S. Bischoff D, S Singh S, H. Hahn B. Peptide-based immunotherapy in lupus: Where are we now? RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:139-149. [PMID: 37781681 PMCID: PMC10538607 DOI: 10.2478/rir-2023-0020] [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: 06/01/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023]
Abstract
In autoimmune rheumatic diseases, immune hyperactivity and chronic inflammation associate with immune dysregulation and the breakdown of immune self-tolerance. A continued, unresolved imbalance between effector and regulatory immune responses further exacerbates inflammation that ultimately causes tissue and organ damage. Many treatment modalities have been developed to restore the immune tolerance and immmunoregulatory balance in autoimmune rheumatic diseases, including the use of peptide-based therapeutics or the use of nanoparticles-based nanotechnology. This review summarizes the state-of-the-art therapeutic use of peptide-based therapies in autoimmune rheumatic diseases, with a specific focus on lupus.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, 90073 CA, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, 90073 CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095 CA, USA
| | | | - Bevra H. Hahn
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095 CA, USA
| |
Collapse
|
36
|
Eckenberger E, Raczka T, Neuhuber W, Distel LVR, Klein S. Acriflavine-Functionalized Silica@Manganese Ferrite Nanostructures for Synergistic Radiation and Hypoxia Therapies. ACS APPLIED BIO MATERIALS 2023; 6:3089-3102. [PMID: 37433114 DOI: 10.1021/acsabm.2c01021] [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: 07/13/2023]
Abstract
Mesoporous and nonmesoporous SiO2@MnFe2O4 nanostructures were loaded with the hypoxia-inducible factor-1 inhibitor acriflavine for combined radiation and hypoxia therapies. The X-ray irradiation of the drug-loaded nanostructures not only triggered the release of the acriflavine inside the cells but also initiated an energy transfer from the nanostructures to surface-adsorbed oxygen to generate singlet oxygen. While the drug-loaded mesoporous nanostructures showed an initial drug release before the irradiation, the drug was primarily released upon X-ray radiation in the case of the nonmesoporous nanostructures. However, the drug loading capacity was less efficient for the nonmesoporous nanostructures. Both drug-loaded nanostructures proved to be very efficient in irradiated MCF-7 multicellular tumor spheroids. The damage of these nanostructures toward the nontumorigenic MCF-10A multicellular spheroids was very limited because of the small number of nanostructures that entered the MCF-10A spheroids, while similar concentrations of acriflavine without nanostructures were toxic for the MCF-10A spheroids.
Collapse
Affiliation(s)
- Elisabeth Eckenberger
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| | - Theodor Raczka
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| | - Winfried Neuhuber
- Institute of Anatomy and Cell Biology, Chair of Anatomy I, Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 9, D-91054 Erlangen, Germany
| | - Luitpold V R Distel
- Department of Radiation Oncology, Friedrich-Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, D-91054 Erlangen, Germany
| | - Stefanie Klein
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| |
Collapse
|
37
|
Dayanandan AP, Cho WJ, Kang H, Bello AB, Kim BJ, Arai Y, Lee SH. Emerging nano-scale delivery systems for the treatment of osteoporosis. Biomater Res 2023; 27:68. [PMID: 37443121 DOI: 10.1186/s40824-023-00413-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/11/2023] [Indexed: 07/15/2023] Open
Abstract
Osteoporosis is a pathological condition characterized by an accelerated bone resorption rate, resulting in decreased bone density and increased susceptibility to fractures, particularly among the elderly population. While conventional treatments for osteoporosis have shown efficacy, they are associated with certain limitations, including limited drug bioavailability, non-specific administration, and the occurrence of adverse effects. In recent years, nanoparticle-based drug delivery systems have emerged as a promising approach for managing osteoporosis. Nanoparticles possess unique physicochemical properties, such as a small size, large surface area-to-volume ratio, and tunable surface characteristics, which enable them to overcome the limitations of conventional therapies. These nanoparticles offer several advantages, including enhanced drug stability, controlled release kinetics, targeted bone tissue delivery, and improved drug bioavailability. This comprehensive review aims to provide insights into the recent advancements in nanoparticle-based therapy for osteoporosis. It elucidates the various types of nanoparticles employed in this context, including silica, polymeric, solid lipid, and metallic nanoparticles, along with their specific processing techniques and inherent properties that render them suitable as potential drug carriers for osteoporosis treatment. Furthermore, this review discusses the challenges and future suggestions associated with the development and translation of nanoparticle drug delivery systems for clinical use. These challenges encompass issues such as scalability, safety assessment, and regulatory considerations. However, despite these challenges, the utilization of nanoparticle-based drug delivery systems holds immense promise in revolutionizing the field of osteoporosis management by enabling more effective and targeted therapies, ultimately leading to improved patient outcomes.
Collapse
Affiliation(s)
| | - Woong Jin Cho
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Hyemin Kang
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | | | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
38
|
Filipić B, Pantelić I, Nikolić I, Majhen D, Stojić-Vukanić Z, Savić S, Krajišnik D. Nanoparticle-Based Adjuvants and Delivery Systems for Modern Vaccines. Vaccines (Basel) 2023; 11:1172. [PMID: 37514991 PMCID: PMC10385383 DOI: 10.3390/vaccines11071172] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Ever since the development of the first vaccine, vaccination has had the great impact on global health, leading to the decrease in the burden of numerous infectious diseases. However, there is a constant need to improve existing vaccines and develop new vaccination strategies and vaccine platforms that induce a broader immune response compared to traditional vaccines. Modern vaccines tend to rely on certain nanotechnology platforms but are still expected to be readily available and easy for large-scale manufacturing and to induce a durable immune response. In this review, we present an overview of the most promising nanoadjuvants and nanoparticulate delivery systems and discuss their benefits from tehchnological and immunological standpoints as well as their objective drawbacks and possible side effects. The presented nano alums, silica and clay nanoparticles, nanoemulsions, adenoviral-vectored systems, adeno-associated viral vectors, vesicular stomatitis viral vectors, lentiviral vectors, virus-like particles (including bacteriophage-based ones) and virosomes indicate that vaccine developers can now choose different adjuvants and/or delivery systems as per the requirement, specific to combatting different infectious diseases.
Collapse
Affiliation(s)
- Brankica Filipić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
- Section of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| |
Collapse
|
39
|
Theivendran S, Lazarev S, Yu C. Mesoporous silica/organosilica nanoparticles for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2023; 3:20220086. [PMID: 37933387 PMCID: PMC10624378 DOI: 10.1002/exp.20220086] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/09/2023] [Indexed: 11/08/2023]
Abstract
Cancer is one of the fatal diseases in the history of humankind. In this regard, cancer immunotherapeutic strategies have revolutionized the traditional mode of cancer treatment. Silica based nano-platforms have been extensively applied in nanomedicine including cancer immunotherapy. Mesoporous silica nanoparticles (MSN) and mesoporous organosilica nanoparticles (MON) are attractive candidates due to the ease in controlling the structural parameters as needed for the targeted immunotherapeutic applications. Especially, the MON provide an additional advantage of controlling the composition and modulating the biological functions to actively synergize with other immunotherapeutic strategies. In this review, the applications of MSN, MON, and metal-doped MSN/MON in the field of cancer immunotherapy and tumor microenvironment regulation are comprehensively summarized by highlighting the structural and compositional attributes of the silica-based nanoplatforms.
Collapse
Affiliation(s)
- Shevanuja Theivendran
- Australian Institute for Bioengineering and NanotechnologyThe University of Queensland, BrisbaneSt LuciaAustralia
| | - Sergei Lazarev
- Australian Institute for Bioengineering and NanotechnologyThe University of Queensland, BrisbaneSt LuciaAustralia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and NanotechnologyThe University of Queensland, BrisbaneSt LuciaAustralia
| |
Collapse
|
40
|
Lisina S, Inam W, Huhtala M, Howaili F, Zhang H, Rosenholm JM. Nano Differential Scanning Fluorimetry as a Rapid Stability Assessment Tool in the Nanoformulation of Proteins. Pharmaceutics 2023; 15:pharmaceutics15051473. [PMID: 37242715 DOI: 10.3390/pharmaceutics15051473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/20/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The development and production of innovative protein-based therapeutics is a complex and challenging avenue. External conditions such as buffers, solvents, pH, salts, polymers, surfactants, and nanoparticles may affect the stability and integrity of proteins during formulation. In this study, poly (ethylene imine) (PEI) functionalized mesoporous silica nanoparticles (MSNs) were used as a carrier for the model protein bovine serum albumin (BSA). To protect the protein inside MSNs after loading, polymeric encapsulation with poly (sodium 4-styrenesulfonate) (NaPSS) was used to seal the pores. Nano differential scanning fluorimetry (NanoDSF) was used to assess protein thermal stability during the formulation process. The MSN-PEI carrier matrix or conditions used did not destabilize the protein during loading, but the coating polymer NaPSS was incompatible with the NanoDSF technique due to autofluorescence. Thus, another pH-responsive polymer, spermine-modified acetylated dextran (SpAcDEX), was applied as a second coating after NaPSS. It possessed low autofluorescence and was successfully evaluated with the NanoDSF method. Circular dichroism (CD) spectroscopy was used to determine protein integrity in the case of interfering polymers such as NaPSS. Despite this limitation, NanoDSF was found to be a feasible and rapid tool to monitor protein stability during all steps needed to create a viable nanocarrier system for protein delivery.
Collapse
Affiliation(s)
- Sofia Lisina
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Wali Inam
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Mikko Huhtala
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, 20500 Turku, Finland
| | - Fadak Howaili
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| |
Collapse
|
41
|
Khaliq NU, Lee J, Kim J, Kim Y, Yu S, Kim J, Kim S, Sung D, Kim H. Mesoporous Silica Nanoparticles as a Gene Delivery Platform for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051432. [PMID: 37242674 DOI: 10.3390/pharmaceutics15051432] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer remains a major global health challenge. Traditional chemotherapy often results in side effects and drug resistance, necessitating the development of alternative treatment strategies such as gene therapy. Mesoporous silica nanoparticles (MSNs) offer many advantages as a gene delivery carrier, including high loading capacity, controlled drug release, and easy surface functionalization. MSNs are biodegradable and biocompatible, making them promising candidates for drug delivery applications. Recent studies demonstrating the use of MSNs for the delivery of therapeutic nucleic acids to cancer cells have been reviewed, along with their potential as a tool for cancer therapy. The major challenges and future interventions of MSNs as gene delivery carriers for cancer therapy are discussed.
Collapse
Affiliation(s)
- Nisar Ul Khaliq
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Juyeon Lee
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Joohyeon Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Yejin Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Sohyeon Yu
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jisu Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sangwoo Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Daekyung Sung
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea
| | - Hyungjun Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| |
Collapse
|
42
|
LaBauve AE, Saada EA, Jones IKA, Mosesso R, Noureddine A, Techel J, Gomez A, Collette N, Sherman MB, Serda RE, Butler KS, Brinker CJ, Schoeniger JS, Sasaki D, Negrete OA. Lipid-coated mesoporous silica nanoparticles for anti-viral applications via delivery of CRISPR-Cas9 ribonucleoproteins. Sci Rep 2023; 13:6873. [PMID: 37105997 PMCID: PMC10133914 DOI: 10.1038/s41598-023-33092-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Emerging and re-emerging viral pathogens present a unique challenge for anti-viral therapeutic development. Anti-viral approaches with high flexibility and rapid production times are essential for combating these high-pandemic risk viruses. CRISPR-Cas technologies have been extensively repurposed to treat a variety of diseases, with recent work expanding into potential applications against viral infections. However, delivery still presents a major challenge for these technologies. Lipid-coated mesoporous silica nanoparticles (LCMSNs) offer an attractive delivery vehicle for a variety of cargos due to their high biocompatibility, tractable synthesis, and amenability to chemical functionalization. Here, we report the use of LCMSNs to deliver CRISPR-Cas9 ribonucleoproteins (RNPs) that target the Niemann-Pick disease type C1 gene, an essential host factor required for entry of the high-pandemic risk pathogen Ebola virus, demonstrating an efficient reduction in viral infection. We further highlight successful in vivo delivery of the RNP-LCMSN platform to the mouse liver via systemic administration.
Collapse
Affiliation(s)
- Annette E LaBauve
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, USA
| | - Edwin A Saada
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA
- Biotechnology and Biosciences Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, USA
| | - Iris K A Jones
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA
| | - Richard Mosesso
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA
| | - Achraf Noureddine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, USA
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, USA
| | - Jessica Techel
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, USA
| | - Andrew Gomez
- Department of Active Ceramics Value Stream, Sandia National Laboratories, Albuquerque, USA
| | - Nicole Collette
- Biotechnology and Biosciences Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, USA
| | - Michael B Sherman
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, USA
| | - Rita E Serda
- Department of Internal Medicine, Health Sciences Center, University of New Mexico, Albuquerque, USA
| | - Kimberly S Butler
- Department of Molecular and Microbiology, Sandia National Laboratories, Albuquerque, USA
| | - C Jeffery Brinker
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, USA
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, USA
- Advanced Materials Laboratory, Sandia National Laboratories, Albuquerque, USA
| | | | - Darryl Sasaki
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, USA
| | - Oscar A Negrete
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA.
| |
Collapse
|
43
|
Wen J, Li H, Dai H, Hua S, Long X, Li H, Ivanovski S, Xu C. Intra-articular nanoparticles based therapies for osteoarthritis and rheumatoid arthritis management. Mater Today Bio 2023; 19:100597. [PMID: 36910270 PMCID: PMC9999238 DOI: 10.1016/j.mtbio.2023.100597] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 02/27/2023] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are chronic and progressive inflammatory joint diseases that affect a large population worldwide. Intra-articular administration of various therapeutics is applied to alleviate pain, prevent further progression, and promote cartilage regeneration and bone remodeling in both OA and RA. However, the effectiveness of intra-articular injection with traditional drugs is uncertain and controversial due to issues such as rapid drug clearance and the barrier afforded by the dense structure of cartilage. Nanoparticles can improve the efficacy of intra-articular injection by facilitating controlled drug release, prolonged retention time, and enhanced penetration into joint tissue. This review systematically summarizes nanoparticle-based therapies for OA and RA management. Firstly, we explore the interaction between nanoparticles and joints, including articular fluids and cells. This is followed by a comprehensive analysis of current nanoparticles designed for OA/RA, divided into two categories based on therapeutic mechanisms: direct therapeutic nanoparticles and nanoparticles-based drug delivery systems. We highlight nanoparticle design for tissue/cell targeting and controlled drug release before discussing challenges of nanoparticle-based therapies for efficient OA and RA treatment and their future clinical translation. We anticipate that rationally designed local injection of nanoparticles will be more effective, convenient, and safer than the current therapeutic approach.
Collapse
Affiliation(s)
- Juan Wen
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Huimin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huan Dai
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Shu Hua
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210009, China
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Corresponding author. School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia.
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Corresponding author. School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia.
| |
Collapse
|
44
|
Zangi AR, Amiri A, Borzouee F, Bagherifar R, Pazooki P, Hamishehkar H, Javadzadeh Y. Immobilized nanoparticles-mediated enzyme therapy; promising way into clinical development. DISCOVER NANO 2023; 18:55. [PMID: 37382752 PMCID: PMC10409955 DOI: 10.1186/s11671-023-03823-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/06/2023] [Indexed: 06/30/2023]
Abstract
Enzyme (Enz)-mediated therapy indicated a remarkable effect in the treatment of many human cancers and diseases with an insight into clinical phases. Because of insufficient immobilization (Imb) approach and ineffective carrier, Enz therapeutic exhibits low biological efficacy and bio-physicochemical stability. Although efforts have been made to remove the limitations mentioned in clinical trials, efficient Imb-destabilization and modification of nanoparticles (NPs) remain challenging. NP internalization through insufficient membrane permeability, precise endosomal escape, and endonuclease protection following release are the primary development approaches. In recent years, innovative manipulation of the material for Enz immobilization (EI) fabrication and NP preparation has enabled nanomaterial platforms to improve Enz therapeutic outcomes and provide low-diverse clinical applications. In this review article, we examine recent advances in EI approaches and emerging views and explore the impact of Enz-mediated NPs on clinical therapeutic outcomes with at least diverse effects.
Collapse
Affiliation(s)
- Ali Rajabi Zangi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ala Amiri
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Fatemeh Borzouee
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rafieh Bagherifar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouya Pazooki
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Science, Tabriz, 5166-15731, Iran
| | - Yousef Javadzadeh
- Biotechnology Research Center, and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, 5166-15731, Iran.
| |
Collapse
|
45
|
Villarruel LA, Brie B, Municoy S, Becú-Villalobos D, Desimone MF, Catalano PN. Silica-collagen nanoformulations with extended human growth hormone release. Int J Pharm 2023; 634:122662. [PMID: 36736675 DOI: 10.1016/j.ijpharm.2023.122662] [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: 09/02/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Growth hormone deficiency has been treated by the daily administration of recombinant human growth hormone (hGH) for decades. Patient compliance to this treatment is generally incomplete due to challenges including dose frequency and lack of perceived benefits. This stimulates the research on new formulations to reduce the number of periodic administrations. In this study silica nanoparticles and silica-collagen nanocomposites were evaluated for hGH loading and release. Bare nanoparticles showed higher hGH adsorption capacity than thiol- and isobutyl-bearing particles of similar diameters. Monitoring of bound protein conformation changes indicated hGH structure retention when adsorbed on bare silica nanoparticles and suggested no alterations on protein activity. Protein-loaded particles incorporated into collagen matrices (silica-collagen nanocomposites) showed a progressive protein release profile different from the observed for hGH-loaded silica nanoparticles and hGH-loaded collagen matrices. While both the collagen and the silica nanoparticle systems reached a 100 % release after 4 and 7 days respectively, silica-collagen nanocomposites showed a bi-phasic prolonged hGH release reaching approximately an 80 % after 15 days. These findings suggest that biocompatible silica-collagen nanocomposites could be used as vehicles for the prolonged delivery of hGH which could lead to a potential reduction in the number of periodic administrations.
Collapse
Affiliation(s)
- Luis A Villarruel
- Instituto de Nanociencia y Nanotecnología (CNEA - CONICET), Nodo Constituyentes, Av. Gral.Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina; Departamento de Micro y Nanotecnología, Gerencia de Desarrollo Tecnológico y Proyectos Especiales, Gerencia de Área de Investigación y Aplicaciones No Nucleares, Centro Atómico Constituyentes, Comisión Nacional de Energía Atómica, Av. Gral. Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Belén Brie
- Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490 (C1428ADN), Buenos Aires, Argentina
| | - Sofía Municoy
- Universidad de Buenos Aires (UBA), CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junin 954 (1113), Buenos Aires, Argentina
| | - Damasia Becú-Villalobos
- Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490 (C1428ADN), Buenos Aires, Argentina
| | - Martín F Desimone
- Universidad de Buenos Aires (UBA), CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junin 954 (1113), Buenos Aires, Argentina.
| | - Paolo N Catalano
- Instituto de Nanociencia y Nanotecnología (CNEA - CONICET), Nodo Constituyentes, Av. Gral.Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina; Departamento de Micro y Nanotecnología, Gerencia de Desarrollo Tecnológico y Proyectos Especiales, Gerencia de Área de Investigación y Aplicaciones No Nucleares, Centro Atómico Constituyentes, Comisión Nacional de Energía Atómica, Av. Gral. Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junin 954 (1113), Buenos Aires, Argentina.
| |
Collapse
|
46
|
Zhang J, Kothalawala S, Yu C. Engineered silica nanomaterials in pesticide delivery: Challenges and perspectives. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 320:121045. [PMID: 36639042 DOI: 10.1016/j.envpol.2023.121045] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/04/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Over the past decade, nanopesticide has been developed rapidly for exploring effective and safe alternatives to conventional pesticides with significant drawbacks and risks. Many nanotechnologies, including pesticide nanoemulsions, polymer-based nanopesticides, and metal/metal oxide nanoparticle-based pesticides have emerged and are extensively reviewed. Engineered silica nanomaterials (ESNs) have also shown promising potential as carriers in nanopesticides for modern agriculture. However, there are limited reviews specifically on ESN-based nanopesticides. Herein, we provide a comprehensive review on the recent progress of ESN-based nanopesticide technologies. An introduction of synthetic technology, formation mechanism, and surface engineering technology is firstly presented. Then, the advantages of ESN-based pesticide formulation and their structure-function-relationship are illustrated in detail. Finally, our perspectives on challenges and future research in ESN-based nanopesticide development are discussed.
Collapse
Affiliation(s)
- Jun Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Sukitha Kothalawala
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
47
|
Zhuang J, Yu Y, Lu R. Mesoporous silica nanoparticles as carrier to overcome bacterial drug resistant barriers. Int J Pharm 2023; 631:122529. [PMID: 36563796 DOI: 10.1016/j.ijpharm.2022.122529] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/27/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Antibiotic resistance has become a global threat to health due to abuse of antibiotics. Lots of existing antibiotics have lost their effect on drug resistant bacteria. Moreover, the discovery of novel antibiotics becomes more and more difficult. It is necessary to develop new strategies to fight against antibiotic resistance. Nano-drug delivery systems endow old antibiotics with new vitality to defeat the antibiotic resistant barrier by protecting antibiotics against hydrolysis, increasing uptake and circumventing efflux pump. Among them, mesoporous silica nanoparticles (MSNs) are one of the most extensively investigated as carrier of antibiotics due to large drug loading capability, tunable physicochemical characteristics, and biocompatibility. MSNs can improve the delivery of antibiotics to bacteria greatly by reducing size, modifying surface, and regulating shapes. Furthermore, MSNs hybridized metal ions or metal nanoparticles exert stronger antibacterial effect by controlling the release of metal ions or increasing active oxygen species. In addition, metal capped MSNs are also able to load antibiotics to exert synergistic antibacterial effect. This paper firstly reviewed the current application of various nanomaterials as antibacterial agents, and then focused on the MSNs including the introduction of MSNs and various approaches for improving antibacterial effect of MSNs.
Collapse
Affiliation(s)
- Jie Zhuang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Yiming Yu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Rui Lu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
48
|
Gándara Z, Rubio N, Castillo RR. Delivery of Therapeutic Biopolymers Employing Silica-Based Nanosystems. Pharmaceutics 2023; 15:pharmaceutics15020351. [PMID: 36839672 PMCID: PMC9963032 DOI: 10.3390/pharmaceutics15020351] [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: 12/01/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
The use of nanoparticles is crucial for the development of a new generation of nanodevices for clinical applications. Silica-based nanoparticles can be tailored with a wide range of functional biopolymers with unique physicochemical properties thus providing several advantages: (1) limitation of interparticle interaction, (2) preservation of cargo and particle integrity, (3) reduction of immune response, (4) additional therapeutic effects and (5) cell targeting. Therefore, the engineering of advanced functional coatings is of utmost importance to enhance the biocompatibility of existing biomaterials. Herein we will focus on the most recent advances reported on the delivery and therapeutic use of silica-based nanoparticles containing biopolymers (proteins, nucleotides, and polysaccharides) with proven biological effects.
Collapse
Affiliation(s)
- Zoila Gándara
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| | - Noelia Rubio
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| | - Rafael R. Castillo
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| |
Collapse
|
49
|
Rani R, Malik P, Dhania S, Mukherjee TK. Recent Advances in Mesoporous Silica Nanoparticle-Mediated Drug Delivery for Breast Cancer Treatment. Pharmaceutics 2023; 15:227. [PMID: 36678856 PMCID: PMC9860911 DOI: 10.3390/pharmaceutics15010227] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Breast cancer (BC) currently occupies the second rank in cancer-related global female deaths. Although consistent awareness and improved diagnosis have reduced mortality in recent years, late diagnosis and resistant response still limit the therapeutic efficacy of chemotherapeutic drugs (CDs), leading to relapse with consequent invasion and metastasis. Treatment with CDs is indeed well-versed but it is badly curtailed with accompanying side effects and inadequacies of site-specific drug delivery. As a result, drug carriers ensuring stealth delivery and sustained drug release with improved pharmacokinetics and biodistribution are urgently needed. Core-shell mesoporous silica nanoparticles (MSNPs) have recently been a cornerstone in this context, attributed to their high surface area, low density, robust functionalization, high drug loading capacity, size-shape-controlled functioning, and homogeneous shell architecture, enabling stealth drug delivery. Recent interest in using MSNPs as drug delivery vehicles has been due to their functionalization and size-shape-driven versatilities. With such insights, this article focuses on the preparation methods and drug delivery mechanisms of MSNPs, before discussing their emerging utility in BC treatment. The information compiled herein could consolidate the database for using inorganic nanoparticles (NPs) as BC drug delivery vehicles in terms of design, application and resolving post-therapy complications.
Collapse
Affiliation(s)
- Ruma Rani
- ICAR-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Sunena Dhania
- Department of Bio & Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Tapan Kumar Mukherjee
- Institute of Biotechnology (AIB), Amity University, Noida 201313, Uttar Pradesh, India
| |
Collapse
|
50
|
Rathod K, Ahmed H, Gomte SS, Chougule S, A P, Dethe MR, Patel RJ, PVP DB, Alexander A. Exploring the potential of anti-inflammatory activity of berberine chloride-loaded mesoporous silica nanoparticles in carrageenan-induced rat paw edema model. J SOLID STATE CHEM 2023. [DOI: 10.1016/j.jssc.2022.123639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|