Review

Prospect of nanomaterials as antimicrobial and antiviral regimen

  • Received: 05 January 2023 Revised: 09 March 2023 Accepted: 17 April 2023 Published: 10 May 2023
  • In recent years studies of nanomaterials have been explored in the field of microbiology due to the increasing evidence of antibiotic resistance. Nanomaterials could be inorganic or organic, and they may be synthesized from natural products from plant or animal origin. The therapeutic applications of nano-materials are wide, from diagnosis of disease to targeted delivery of drugs. Broad-spectrum antiviral and antimicrobial activities of nanoparticles are also well evident. The ratio of nanoparticles surface area to their volume is high and that allows them to be an advantageous vehicle of drugs in many respects. Effective uses of various materials for the synthesis of nanoparticles impart much specificity in them to meet the requirements of specific therapeutic strategies. The potential therapeutic use of nanoparticles and their mechanisms of action against infections from bacteria, fungi and viruses were the focus of this review. Further, their potential advantages, drawbacks, limitations and side effects are also included here. Researchers are characterizing the exposure pathways of nano-medicines that may cause serious toxicity to the subjects or the environment. Indeed, societal ethical issues in using nano-medicines pose a serious question to scientists beyond anything.

    Citation: Ashok Chakraborty, Anil Diwan, Jayant Tatake. Prospect of nanomaterials as antimicrobial and antiviral regimen[J]. AIMS Microbiology, 2023, 9(3): 444-466. doi: 10.3934/microbiol.2023024

    Related Papers:

  • In recent years studies of nanomaterials have been explored in the field of microbiology due to the increasing evidence of antibiotic resistance. Nanomaterials could be inorganic or organic, and they may be synthesized from natural products from plant or animal origin. The therapeutic applications of nano-materials are wide, from diagnosis of disease to targeted delivery of drugs. Broad-spectrum antiviral and antimicrobial activities of nanoparticles are also well evident. The ratio of nanoparticles surface area to their volume is high and that allows them to be an advantageous vehicle of drugs in many respects. Effective uses of various materials for the synthesis of nanoparticles impart much specificity in them to meet the requirements of specific therapeutic strategies. The potential therapeutic use of nanoparticles and their mechanisms of action against infections from bacteria, fungi and viruses were the focus of this review. Further, their potential advantages, drawbacks, limitations and side effects are also included here. Researchers are characterizing the exposure pathways of nano-medicines that may cause serious toxicity to the subjects or the environment. Indeed, societal ethical issues in using nano-medicines pose a serious question to scientists beyond anything.



    加载中

    Acknowledgments



    Thanks are due to Ms. Bethany Pond for her editorial assistance.

    Conflict of interest



    Authors Anil Diwan and Jayant Tatake are employed by the company NanoViricides, Inc, and they declare no conflicts of interest between the companies, AllExcel, Inc, TheraCour Pharma, Inc. and NanoViricides, Inc. The research was funded by NanoViricides, Inc.

    [1] Sethi S, Murphy TF (2001) Bacterial infection in chronic obstructive pulmonary disease in 2000: a state-of-the-art review. Clin Microbiol Rev 14: 336-363. https://doi.org/10.1128/CMR.14.2.336-363.2001
    [2] Durand ML, Calderwood SB, Weber DJ, et al. (1993) Acute bacterial meningitis in adults-A review of 493 episodes. N Engl J Med 328: 21-28. https://doi.org/10.1056/NEJM199301073280104
    [3] Lara HH, Ayala-Nu~nez NV, Ixtepan-Turrent L, et al. (2010) Mode of antiviral action of silver nanoparticles against HIV-1. J Nanobiotechno 8: 1-10. https://doi.org/10.1186/1477-3155-8-1
    [4] Singh SR, Krishnamurthy NB, Mathew BB (2014) A review on recent diseases caused by microbes. J Appl Environ Microbiol 2: 106-115.
    [5] Laxminarayan R, Duse A, Wattal C, et al. (2013) Antibiotic resistance—the need for global solutions. Lancet Infect Dis 13: 1057-1098. https://doi.org/10.1016/S1473-3099(13)70318-9
    [6] Friedman H, Newton C, Klein TW (2003) Microbial infections, immunomodulation, and drugs of abuse. Clin Microbiol Rev 16: 209-219. https://doi.org/10.1128/CMR.16.2.209-219.2003
    [7] Atolani O, Baker MT, Adeyemi OS, et al. (2020) Covid-19: critical discussion on the applications and implications of chemicals in sanitizers and disinfectants. EXCLI J 19: 785-799.
    [8] Gurunathan S, Han JW, Kwon DN, et al. (2014) Enhanced antibacterial and antibiofilm activities of silver nanoparticles against Gram-negative and Gram-positive bacteria. Nanoscale Res Lett 9: 1-17. https://doi.org/10.1186/1556-276X-9-373
    [9] Yien L, Zin NM, Sarwar A, et al. (2012) Antifungal activity of chitosan nanoparticles and correlation with their physical properties. Int J Biomater 632698. https://doi.org/10.1155/2012/632698
    [10] Sobhani Z, Samani SM, Montaseri H, et al. (2017) Nanoparticles of chitosan loaded ciprofloxacin: fabrication and antimicrobial activity. Adv Pharmaceut Bull 7: 427. https://doi.org/10.15171/apb.2017.051
    [11] Rai A, Prabhune A, Perry CC (2010) Antibiotic mediated synthesis of gold nanoparticles with potent antimicrobial activity and their application in antimicrobial coatings. J Mater Chem 20: 6789-6798. https://doi.org/10.1039/c0jm00817f
    [12] Alshammari F, Alshammari B, Moin A, et al. (2021) Ceftriaxone mediated synthesized gold nanoparticles: A nano-therapeutic tool to target bacterial resistance. Pharmaceutics 13: 1896. https://doi.org/10.3390/pharmaceutics13111896
    [13] Cepas V, López Y, Gabasa Y, et al. (2019) Inhibition of bacterial and fungal biofilm formation by 675 extracts from microalgae and cyanobacteria. Antibiotics (Basel) 8: 77. https://doi.org/10.3390/antibiotics8020077
    [14] Cremonini E, Zonaro E, Donini M, et al. (2016) Biogenic selenium nanoparticles: characterization, antimicrobial activity and effects on human dendritic cells and fibroblasts. Microb Biotechnol 9: 758-771. https://doi.org/10.1111/1751-7915.12374
    [15] Haghighi F, Roudba Mohammadir S, Mohammadi P, et al. (2013) Antifungal acitivity of TiO2 nanoparticles and EDTA on Candida abicans Biofilms. Infect Epidemiol Med 1: 133-138.
    [16] Singh P, Singh D, Sa P, et al. (2021) Insights from nanotechnology in COVID-19: prevention, detection, therapy and immunomodulation. Nanomedicine (Lond) 16: 1219-1235. https://doi.org/10.2217/nnm-2021-0004
    [17] Singh CK, Sodhi KK (2023) The emerging significance of nanomedicine-based approaches to fighting COVID-19 variants of concern: A perspective on the nanotechnology's role in COVID-19 diagnosis and treatment. Front Nanotechnol 4: 1084033. https://doi.org/10.3389/fnano.2022.1084033
    [18] Yang D (2021) Application of nanotechnology in the COVID-19 pandemic. Int J Nanomedicine 16: 623-649. https://doi.org/10.2147/IJN.S296383
    [19] Diwan A, Chakraborty A, Vijetha Chiniga V, et al. (2022) Dual effects of NV-CoV-2 biomimetic polymer: An antiviral regimen against COVID-19. PLOS One 17: e0278963. https://doi.org/10.1371/journal.pone.0278963
    [20] Chakraborty A, Diwan A, Arora V, et al. (2022) Mechanism of antiviral activities of nanoviricide's platform technology based biopolymer (NV-CoV-2). AIMS Public Health 9: 415-422. https://doi.org/10.3934/publichealth.2022028
    [21] Mubeen B, Ansar AN, Rasool R, et al. (2021) Nanotechnology as a novel approach in combating microbes providing an alternative to antibiotics. Antibiotics (Basel) 10: 1473. https://doi.org/10.3390/antibiotics10121473
    [22] Hung YP, Chen YF, Tsai PJ, et al. (2021) Advances in the application of nanomaterials as treatments for bacterial infectious diseases. Pharmaceutics 13: 1913. https://doi.org/10.3390/pharmaceutics13111913
    [23] Ozdal M, Gurkok S (2022) Recent advances in nanoparticles as antibacterial agent. ADMET DMPK 10: 115-129. https://doi.org/10.5599/admet.1172
    [24] Hussain FS, Abro NQ, Ahmed N, et al. (2022) Nano antivirals: A comprehensive review. Front Nanotechnol 4: 1064615. https://doi.org/10.3389/fnano.2022.1064615
    [25] Nami S, Aghebati-Maleki A, Aghebati-Maleki L (2021) Current applications and prospects of nanoparticles for antifungal drug delivery. EXCLI J 20: 562-584.
    [26] Khan I, Saeed K, Khan I (2017) Nanoparticles: properties, applications and toxicities. Arab J Chem 12: 908-931. https://doi.org/10.1016/j.arabjc.2017.05.011
    [27] Song X, Bayati P, Gupta M, et al. (2021) Fracture of magnesium matrix nanocomposites-a review. Int J Lightweight Mater Manufact 4: 67-98. https://doi.org/10.1016/j.ijlmm.2020.07.002
    [28] Vert M, Doi Y, Hellwich KH, et al. (2012) Terminology for biorelated polymers and applications (IUPAC Recommendations 2012). Pure Appl Chem 84: 377-410. https://doi.org/10.1515/amma-2016-0032
    [29] Von Nussbaum F, Brands M, Hinzen B, et al. (2006) Antibacterial natural products in medicinal chemistry—Exodus or revival?. Angew Chem Int Ed 45: 5072-5129. https://doi.org/10.1002/anie.200600350
    [30] Akhtar M, Swamy MK, Umar A, et al. (2015) Biosynthesis and characterization of silver nanoparticles from methanol leaf extract of Cassia didymobotyra and assessment of their antioxidant and antibacterial activities. J Nanosci Nanotechnol 15: 9818-9823. https://doi.org/10.1166/jnn.2015.10966
    [31] Fröhlich E, Salar-Behzadi S (2014) Toxicological assessment of inhaled nanoparticles: Role of in vivo, ex vivo, in vitro, and in silico studies. Int J Mol Sci 15: 4795-4822. https://doi.org/10.3390/ijms15034795
    [32] Qiu Y, Xu D, Sui G, et al. (2020) Gentamicin decorated phosphatidylcholine-chitosan nanoparticles against biofilms and intracellular bacteria. Int J Biol Macromol 156: 640-647. https://doi.org/10.1016/j.ijbiomac.2020.04.090
    [33] Evangelista TF, Andrade GR, Nascimento KN, et al. (2020) Supramolecular polyelectrolyte complexes based on cyclodextrin-grafted chitosan and carrageenan for controlled drug release. Carbohydr Polym 245: 116592. https://doi.org/10.1016/j.carbpol.2020.116592
    [34] Walvekar P, Gannimani R, Salih M, et al. (2019) Self-assembled oleylamine grafted hyaluronic acid polymersomes for delivery of vancomycin against methicillin resistant Staphylococcus aureus (MRSA). Colloids Surf B Biointerfaces 182: 110388. https://doi.org/10.1016/j.colsurfb.2019.110388
    [35] Ejaz S, Ihsan A, Noor T, et al. (2020) Mannose functionalized chitosan nanosystems for enhanced antimicrobial activity against multidrug resistant pathogens. Polym Test 91: 106814. https://doi.org/10.1016/j.polymertesting.2020.106814
    [36] Ucak S, Sudagidan M, Borsa BA, et al. (2020) Inhibitory effects of aptamer targeted teicoplanin encapsulated PLGA nanoparticles for Staphylococcus aureus strains. World J Microbiol Biotechnol 36: 69. https://doi.org/10.1007/s11274-020-02845-y
    [37] Vrouvaki I, Koutra E, Kornaros M, et al. (2020) Polymeric nanoparticles of pistacia lentiscus var. chia essential oil for cutaneous applications. Pharmaceutics 12: 353. https://doi.org/10.3390/pharmaceutics12040353
    [38] Gherasim O, Grumezescu AM, Grumezescu V, et al. (2020) Bioactive surfaces of polylactide and silver nanoparticles for the prevention of microbial contamination. Materials 13: 768. https://doi.org/10.3390/ma13030768
    [39] Grumezescu AM, Stoica AE, Dima-Balcescu MS, et al. (2019) Electrospun polyethylene terephthalate nanofibers loaded with silver nanoparticles: novel approach in anti-infective therapy. J Clin Med 8: 1039. https://doi.org/10.3390/jcm8071039
    [40] Khandelwal N, Kaur G, Kumara N, et al. (2014) Application of silver nanoparticles in viral inhibition: A new hope for antivirals. Dig J Nanomater Biostruct 9: 175-186. https://nanogo.co.uk/wp-content/uploads/2021/12/application-of-nanosilver.pdf
    [41] Alamdaran M, Movahedi B, Mohabatkar H, et al. (2018) In-vitro study of the novel nanocarrier of chitosan-based nanoparticles conjugated HIV-1 P24 protein-derived peptides. J Mol Liq 265: 243-250. https://doi.org/10.1016/j.molliq.2018.05.137
    [42] Belgamwar AV, Khan SA, Yeole PG (2019) Intranasal dolutegravir sodium loaded nanoparticles of hydroxypropyl-beta-cyclodextrin for brain delivery in Neuro-AIDS. J Drug Deliv Sci Technol 52: 1008-1020. https://doi.org/10.1016/j.jddst.2019.06.014
    [43] Costa AF, Araujo DE, Cabral MS, et al. (2018) Development, characterization, and in vitro-in vivo evaluation of polymeric nanoparticles containing miconazole and farnesol for treatment of vulvovaginal candidiasis. Med Mycol 7: 52-62. https://doi.org/10.1093/mmy/myx155
    [44] Reddy YC (2018) Formulation and evaluation of chitosan nanoparticles for improved efficacy of itraconazole antifungal drug. Asian J Pharm Clin Res 11: 147-152. https://doi.org/10.22159/ajpcr.2018.v11s4.31723
    [45] Sombra FM, Richter AR, De Araújo AR, et al. (2020) Development of amphotericin B-loaded propionate Sterculia striata polysaccharide nanocarrier. Int J Biol Macromol 146: 1133-1141. https://doi.org/10.1016/j.ijbiomac.2019.10.053
    [46] Real D, Hoffmann S, Leonardi D, et al. (2018) Chitosan-based nanodelivery systems applied to the development of novel triclabendazole formulations. PLOS One 13: e0207625. https://doi.org/10.1371/journal.pone.0207625
    [47] Durak S, Arasoglu T, Ates SC, et al. (2020) Enhanced antibacterial and antiparasitic activity of multifunctional polymeric nanoparticles. Nanotechnology 31: 175705. https://doi.org/10.1088/1361-6528/ab6ab9
    [48] Binder U, Aigner M, Risslegger B, et al. (2019) Minimal Inhibitory concentration (mic)-phenomena in candida albicans and their impact on the diagnosis of antifungal resistance. J Fungi (Basel) 5: 83. https://doi.org/10.3390/jof5030083
    [49] Kesharwani P, Fatima M, Singh V, et al. (2022) Itraconazole and difluorinated-curcumin containing chitosan nanoparticle loaded hydrogel for amelioration of onychomycosis. Biomimetics (Basel) 7: 206. https://doi.org/10.3390/biomimetics7040206
    [50] Butani D, Yewale C, Misra A (2016) Topical amphotericin B solid lipid nanoparticles: Design and development. Colloids Surf B 139: 17-24. https://doi.org/10.1016/j.colsurfb.2015.07.032
    [51] Souto E, Wissing S, Barbosa C, et al. (2004) Development of a controlled release formulation based on SLN and NLC for topical clotrimazole delivery. Int J Pharm 278: 71-7. https://doi.org/10.1016/j.ijpharm.2004.02.032
    [52] Cassano R, Ferrarelli T, Mauro MV, et al. (2016) Preparation, characterization and in vitro activities evaluation of solid lipid nanoparticles based on PEG-40 stearate for antifungal drugs vaginal delivery. Drug Deliv 23: 1037-46. https://doi.org/10.3109/10717544.2014.932862
    [53] Sanna V, Gavini E, Cossu M, et al. (2007) Solid lipid nanoparticles (SLN) as carriers for the topical delivery of econazole nitrate: In-vitro characterization, ex-vivo and in-vivo studies. J Pharm Pharmacol 59: 1057-64. https://doi.org/10.1211/jpp.59.8.0002
    [54] Bhalekar MR, Pokharkar V, Madgulkar A, et al. (2009) Preparation and evaluation of miconazole nitrate-loaded solid lipid nanoparticles for topical delivery. AAPS PharmSciTech 10: 289-96. https://doi.org/10.1208/s12249-009-9199-0
    [55] Jain S, Jain S, Khare P, et al. (2010) Design and development of solid lipid nanoparticles for topical delivery of an anti-fungal agent. Drug Deliv 17: 443-451. https://doi.org/10.3109/10717544.2010.483252
    [56] Kenechukwu FC, Attama AA, Ibezim EC (2017) Novel solidified reverse micellar solution-based mucoadhesive nano lipid gels encapsulating miconazole nitrate-loaded nanoparticles for improved treatment of oropharyngeal candidiasis. J Microencapsul 34: 592-609. https://doi.org/10.1080/02652048.2017.1370029
    [57] Mahato R, Tai W, Cheng K (2011) Prodrugs for improving tumor targetability and efficiency. Adv Drug Deliv Rev 63: 659-70. https://doi.org/10.1016/j.addr.2011.02.002
    [58] Kumar R, Sinha VR (2016) Solid lipid nanoparticle: An efficient carrier for improved ocular permeation of voriconazole. Drug Dev Ind Pharm 42: 1956-67. https://doi.org/10.1080/03639045.2016.1185437
    [59] Füredi P, Pápay ZE, Kovács K, et al. (2017) Development and characterization of the voriconazole loaded lipid-based nanoparticles. J Pharm Biomed Anal 132: 184-9. https://doi.org/10.1016/j.jpba.2016.09.047
    [60] El-Housiny S, Shams Eldeen MA, El-Attar YA, et al. (2018) Fluconazole-loaded solid lipid nanoparticles topical gel for treatment of pityriasis versicolor: formulation and clinical study. Drug Deliv 25: 78-90. https://doi.org/10.1080/10717544.2017.1413444
    [61] Anurak L, Chansiri G, Peankit D, et al. (2011) Griseofulvin solid lipid nanoparticles based on microemulsion technique. Adv Mater Res 197–198: 47-50. https://doi.org/10.4028/www.scientific.net/AMR.197-198.47
    [62] Ahmad A, Wei Y, Syed F, et al. (2016) Amphotericin B-conjugated biogenic silver nanoparticles as an innovative strategy for fungal infections. Microb Pathol 99: 271-81. https://doi.org/10.1016/j.micpath.2016.08.031
    [63] Tutaj K, Szlazak R, Szalapata K, et al. (2016) Amphotericin B-silver hybrid nanoparticles: Synthesis, properties and antifungal activity. Nanomedicine 12: 1095-103. https://doi.org/10.1016/j.nano.2015.12.378
    [64] Hussain MA, Ahmed D, Anwar A, et al. (2019) Combination therapy of clinically approved antifungal drugs is enhanced by conjugation with silver nanoparticles. Int Microbiol 22: 239-46. https://doi.org/10.1007/s10123-018-00043-3
    [65] Mussin JE, Roldán MV, Rojas F, et al. (2019) Antifungal activity of silver nanoparticles in combination with ketoconazole against Malassezia furfur. AMB Express 9: 131. https://doi.org/10.1186/s13568-019-0857-7
    [66] Souza AC, Nascimento AL, de Vasconcelos NM, et al. (2015) Activity and in vivo tracking of Amphotericin B loaded PLGA nanoparticles. Eur J Med Chem 95: 267-276. https://doi.org/10.1016/j.ejmech.2015.03.022
    [67] Vásquez Marcano RGDJ, Tominaga TT, Khalil NM, et al. (2018) Chitosan functionalized poly (ϵ-caprolactone) nanoparticles for amphotericin B delivery. Carbohydr Polym 202: 345-354. https://doi.org/10.1016/j.carbpol.2018.08.142
    [68] Chhonker YS, Prasad YD, Chandasana H, et al. (2015) Amphotericin-B entrapped lecithin/chitosan nanoparticles for prolonged ocular application. Int J Biol Macromol 72: 1451-8. https://doi.org/10.1016/j.ijbiomac.2014.10.014
    [69] Bhattacharjee S, Joshi R, Chughtai AA, et al. (2021) Graphene-and nanoparticle-embedded antimicrobial and biocompatible cotton/silk fabrics for protective clothing. ACS Appl Bio Mater 4: 6175-6185. https://doi.org/10.1021/acsabm.1c00508. 10.1021/acsabm.1c00508
    [70] Kharaghani D, Dutta D, Gitigard P, et al. (2019) Development of antibacterial contact lenses containing metallic nanoparticles. Polym Test 79: 106034. https://doi.org/10.1016/j.polymertesting.2019.106034
    [71] Kalita S, Kandimalla R, Bhowal AC, et al. (2018) Functionalization of β-lactam antibiotic on lysozyme capped gold nanoclusters retrogress MRSA and its persisters following awakening. Sci Rep 8: 1-13. https://doi.org/10.1038/s41598-018-22736-5
    [72] Konop M, Czuwara J, Kłodzińska E, et al. (2020) Evaluation of keratin biomaterial containing silver nanoparticles as a potential wound dressing in full-thickness skin wound model in diabetic mice. J Tissue Eng Regen Med 14: 334-346. https://doi.org/10.1002/term.2998
    [73] Gupta A, Briffa SM, Swingler S, et al. (2020) Synthesis of silver nanoparticles using curcumin-cyclodextrins loaded into bacterial cellulose-based hydrogels for wound dressing applications. Biomacromolecules 21: 1802-1811. https://doi.org/10.1021/acs.biomac.9b01724
    [74] Urbán P, Liptrott NJ, Bremer S (2019) Overview of the blood compatibility of nanomedicines: A trend analysis of in vitro and in vivo studies. Wiley Interdiscip Rev Nanomed Nanobiotechnol 11: e1546. https://doi.org/10.1002/wnan.1546
    [75] de la Harpe KM, Kondiah PPD, Choonara YE, et al. (2019) The hemocompatibility of nanoparticles: A review of cell-nanoparticle interactions and hemostasis. Cells 8: 1209. https://doi.org/10.3390/cells8101209
    [76] Guo S, Shi Y, Liang Y, et al. (2021) Relationship and improvement strategies between drug nanocarrier characteristics and hemocompatibility: What can we learn from the literature. Asian J Pharm Sci 16: 551-576. https://doi.org/10.1016/j.ajps.2020.12.002
    [77] Mikušová V, Mikuš P (2021) Advances in chitosan-based nanoparticles for drug delivery. Int J Mol Sci 22: 9652. https://doi.org/10.3390/ijms22179652
    [78] Pathak N, Singh P, Singh PK, et al. (2022) Biopolymeric nanoparticles based effective delivery of bioactive compounds toward the sustainable development of anticancerous therapeutics. Front Nutr 9: 963413. https://doi.org/10.3389/fnut.2022.963413
    [79] Chen HT, Neerman MF, Parrish AR, et al. (2004) Cytotoxicity, hemolysis, and acute in vivo toxicity of dendrimers based on melamine, candidate vehicles for drug delivery. J Am Chem Soc 126: 10044-10048. https://doi.org/10.1021/ja048548j
    [80] Jia YP, Ma BY, Wei XW, et al. (2017) The in vitro and in vivo toxicity of gold nanoparticles. Chin Chem Lett 28: 691-702. https://doi.org/10.1016/j.cclet.2017.01.021
    [81] Chetyrkina MR, Fedorov FS, Nasibulin AG (2022) In vitro toxicity of carbon nanotubes: a systematic review. RSC Adv 25: 16235-16256. https://doi.org/10.1039/D2RA02519A
    [82] Mahmoudi M, Hofmann H, Rothen-Rutishauser B, et al. (2012) Petri-Assessing the in vitro and in vivo Toxicity of Superparamagnetic Iron Oxide Nanoparticles. Chem Rev 112: 2323-2338. https://doi.org/10.1021/cr2002596
    [83] Chen L, Liu J, Zhang Y, et al. (2018) The toxicity of silica nanoparticles to the immune system. Nanomedicine 13: 1939-1962. https://doi.org/10.2217/nnm-2018-0076
    [84] Hadipour Moghaddam SP, Mohammadpour R, Ghandehari H (2019) In vitro and in vivo evaluation of degradation, toxicity, biodistribution, and clearance of silica nanoparticles as a function of size, porosity, density, and composition. J Control Release 311–312: 1-15. https://doi.org/10.1016/j.jconrel.2019.08.028
    [85] Zhang XF, Shen W, Gurunathan S (2016) Silver nanoparticle-mediated cellular responses in various cell lines: An in vitro model. Int J Mol Sci 17: 1603. https://doi.org/10.3390/ijms17101603
    [86] Yuan YG, Zhang S, Hwang JY, et al. (2018) Silver nanoparticles potentiates cytotoxicity and apoptotic potential of camptothecin in human cervical cancer cells. Oxid Med Cell Longev 18: 6121328. https://doi.org/10.1155/2018/6121328
    [87] Rim KT, Song SW, Kim HY (2013) Oxidative DNA damage from nanoparticle exposure and its application to workers' health: a literature review. Saf Health Work 4: 177-186. https://doi.org/10.1016/j.shaw.2013.07.006
    [88] Wen H, Dan M, Yang Y, et al. (2017) Acute toxicity and genotoxicity of silver nanoparticle in rats. PLOS One 2: e0185554. https://doi.org/10.1371/journal.pone.0185554
    [89] Hassanen EI, Khalaf AA, Tohamy AF, et al. (2019) Toxicopathological and immunological studies on different concentrations of chitosan-coated silver nanoparticles in rats. Int J Nanomedicine 14: 4723-4739. https://doi.org/10.2147/IJN.S207644
    [90] Panyam J, Labhasetwar V (2003) Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv Drug Deliv Rev 55: 329-347. https://doi.org/10.1016/S0169-409X(02)00228-4
    [91] Almofti MR, Ichikawa T, Yamashita K, et al. (2003) Silver ion induces a cyclosporine a-insensitive permeability transition in rat liver mitochondria and release of apoptogenic cytochrome C. J Biochem 134: 43-49. https://doi.org/10.1093/jb/mvg111
    [92] Iancu SD, Albu C, Chiriac L, et al. (2020) Assessment of gold-coated iron oxide nanoparticles as negative T2 contrast agent in small animal MRI studies. Int J Nanomedicine 4811–4824. https://doi.org/10.2147/IJN.S253184
    [93] Akhlaghi N, Najafpour-Darzi G (2021) Manganese ferrite (MnFe2O4) Nanoparticles: From synthesis to application-A review. J Ind Eng Chem 103: 292-304. https://doi.org/10.1016/j.jiec.2021.07.043
    [94] Ganapathe LS, Mohamed Md A, Yunus RM (2020) Magnetite (Fe3O4) Nanoparticles in biomedical application: from synthesis to surface functionalisation. Magnetochemistry 6: 68. https://doi.org/10.3390/magnetochemistry6040068
    [95] Liu Z, Liu Y, Liu S, et al. (2021) The effects of TiO2 nanotubes on the biocompatibility of 3D printed Cu-bearing TC4 alloy. Mater Des 207: 109831. https://doi.org/10.1016/j.matdes.2021.109831
    [96] Andrade RGD, Ferreira D, Veloso SRS, et al. (2022) Synthesis and cytotoxicity assessment of citrate-coated calcium and manganese ferrite nanoparticles for magnetic hyperthermia. Pharmaceutics 14: 2694. https://doi.org/10.3390/pharmaceutics14122694
    [97] Rudramurthy GR, Swamy MK, Sinniah UR, et al. (2016) Nanoparticles: alternatives against drug-resistant pathogenic microbes. Molecules 21: 836. https://doi.org/10.3390/molecules21070836
    [98] Shiri S, Abbasi N, Alizadeh K, et al. (2019) Novel and green synthesis of a nanopolymer and its use as a drug delivery system of silibinin and silymarin extracts in the olfactory ensheathing cells of rats in normal and high-glucose conditions. RSC Adv 9: 38912-38927. https://doi.org/10.1039/C9RA05608D
    [99] Sharma S, Sudhakara P, Singh J, et al. (2021) Critical review of biodegradable and bioactive polymer composites for bone tissue engineering and drug delivery applications. Polymers (Basel) 13: 2623. https://doi.org/10.3390/polym13162623
    [100] Makadia HK, Siegel SJ (2011) Poly Lactic-co-Glycolic Acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers (Basel) 3: 1377-1397. https://doi.org/10.3390/polym3031377
    [101] Félix Lanao RP, Jonker AM, Wolke JG, et al. (2013) Physicochemical properties and applications of poly(lactic-co-glycolic acid) for use in bone regeneration. Tissue Eng Part B Rev 19: 380-390. https://doi.org/10.1089/ten.teb.2012.0443
    [102] da Silva D, Kaduri M, Poley M, et al. (2018) Biocompatibility, biodegradation and excretion of polylactic acid (PLA) in medical implants and theranostic systems. Chem Eng J 340: 9-14. https://doi.org/10.1016/j.cej.2018.01.010
    [103] Ramos DP, Sarjinsky S, Alizadehgiashi M, et al. (2019) Polyelectrolyte vs polyampholyte behavior of composite chitosan/ gelatin films. ACS Omega 4: 8795-8803. http://pubs.acs.org/journal/acsodf
    [104] Olasehinde TA, Olaniran AO (2022) Neurotoxicity of polycyclic aromatic hydrocarbons: a systematic mapping and review of neuropathological mechanisms. Toxics 10: 417. https://doi.org/10.3390/toxics10080417
    [105] Liu Y, Hardie J, Zhang X, et al. (2017) Effects of engineered nanoparticles on the innate immune system. Semin Immunol 34: 25-32. https://doi.org/10.1016/j.smim.2017.09.011
    [106] Corbo C, Molinaro R, Parodi A, et al. (2016) The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery. Nanomedicine (Lond) 11: 81-100. https://doi.org/10.2217/nnm.15.188
    [107] Suk JS, Xu Q, Kim N, et al. (2016) PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 99: 28-51. https://doi.org/10.1016/j.addr.2015.09.012
    [108] Tan X, Fang Y, Ren Y, et al. (2019) D-α-tocopherol polyethylene glycol 1000 succinate-modified liposomes with an siRNA corona confer enhanced cellular uptake and targeted delivery of doxorubicin via tumor priming. Int J Nanomedicine 14: 1255-1268. https://doi.org/10.2147/IJN.S191858
    [109] Neophytou CM, Mesaritis A, Gregoriou G, et al. (2019) d-a-Tocopheryl Polyethylene Glycol 1000 Succinate and a small-molecule Survivin suppressant synergistically induce apoptosis in SKBR3 breast cancer cells. Sci Rep 9: 14375. https://doi.org/10.1038/s41598-019-50884-9
    [110] Dang Y, Guan J (2020) Nanoparticle-based drug delivery systems for cancer therapy. Smart Mater Med 1: 10-19. https://doi.org/10.1016/j.smaim.2020.04.001
    [111] Adepu S, Ramakrishna S (2021) Controlled drug delivery systems: current status and future directions. Molecules 26: 5905. https://doi.org/10.3390/molecules26195905
    [112] Yu B, Tai HC, Xue W, et al. (2010) Receptor-targeted nanocarriers for therapeutic delivery to cancer. Mol Membr Biol 27: 286-298. https://doi.org/10.3109/09687688.2010.521200
    [113] Yu H, Yang Z, Li F, et al. (2020) Cell-mediated targeting drugs delivery systems. Drug Deliv 27: 1425-1437. https://doi.org/10.1080/10717544.2020.1831103
    [114] Zhao Z, Ukidve A, Kim J, et al. (2020) Targeting strategies for tissue-specific drug delivery. Cell 181: 2020. https://doi.org/10.1016/j.cell.2020.02.001
    [115] Manzari MT, Shamay Y, Kiguchi H, et al. (2021) Targeted drug delivery strategies for precision medicines. Nat Rev Mater 6: 351-370. https://doi.org/10.1038/s41578-020-00269-6
    [116] Mitchell MJ, Billingsley MM, Haley RM, et al. (2021) Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov 20: 101-124. https://doi.org/10.1038/s41573-020-0090-8
    [117] Xie J, Lee S, Chen X (2010) Nanoparticle-based theranostic agents. Adv Drug Deliv Rev 62: 1064-1079. https://doi.org/10.1016/j.addr.2010.07.009
    [118] Chatterjee DK, Diagaradjane P, Krishnan S (2011) Nanoparticle-mediated Hyperthermia in Cancer Therapy. Ther Deliv 2: 1001-1014. https://doi.org/10.4155/tde.11.72
    [119] Jacque D, Martínez Maestro L, del Rosal B, et al. (2014) Nanoparticles for photothermal therapies. Nanoscale 6: 9494-9530. https://doi.org/10.1039/C4NR00708E
    [120] Zhou Y, Quan G, Wu Q, et al. (2018) Mesoporous silica nanoparticles for drug and gene delivery. Acta Pharmaceutica Sinica B 8: 165-177. https://doi.org/10.1016/j.apsb.2018.01.007
    [121] Li Z, Barnes JC, Bosoy A, et al. (2012) Mesoporous silica nanoparticles in biomedical applications. Chem Soc Rev 41: 2590-2605. https://doi.org/10.1039/c1cs15246g
    [122] De Jong WH, Borm PJ (2008) Drug delivery and nanoparticles:applications and hazards. Int J Nanomedicine 3: 133-149. https://doi.org/10.2147/IJN.S596
    [123] Patra JK, Das G, Fraceto LF, et al. (2018) Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol 16: 71. https://doi.org/10.1186/s12951-018-0392-8
    [124] Masarudin MJ, Cutts SM, Evison BJ, et al. (2015) Factors determining the stability, size distribution, and cellular accumulation of small, monodisperse chitosan nanoparticles as candidate vectors for anticancer drug delivery: application to the passive encapsulation of [(14)C]-doxorubicin. Nanotechnol Sci Appl 8: 67-80. https://doi.org/10.2147/NSA.S91785
    [125] Ajdary M, Moosavi MA, Rahmati M, et al. (2018) Health concerns of various nanoparticles: a review of their in vitro and in vivo toxicity. Nanomaterials (Basel) 8: 634. https://doi.org/10.3390/nano8090634
    [126] Ray P, Haideri N, Haque I, et al. (2021) The impact of nanoparticles on the immune system: a gray zone of nanomedicine. J Immunological Sci 5: 19-33. https://doi.org/10.29245/2578-3009/2021/1.1206
    [127] Abdal Dayem A, Hossain MK, Lee SB, et al. (2017) The role of reactive oxygen species (ros) in the biological activities of metallic nanoparticles. Int J Mol Sci 18: 120. https://doi.org/10.3390/ijms18010120
    [128] Ray PC, Yu H, Fu PP (2009) Toxicity and environmental risks of nanomaterials: challenges and future needs. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 27: 1-35. https://doi.org/10.1080/10590500802708267
    [129] Egbuna C, Parmar VK, Jeevanandam J, et al. (2021) Toxicity of nanoparticles in biomedical application: nanotoxicology. J Toxicol 2021:9954443: 1-21. https://doi.org/10.1155/2021/9954443
    [130] Huang YW, Cambre M, Lee HJ (2017) The toxicity of nanoparticles depends on multiple molecular and physicochemical mechanisms. Int J Mol Sci 18: 2702. https://doi.org/10.3390/ijms18122702
    [131] You DJ, Bonner JC (2020) Susceptibility factors in chronic lung inflammatory responses to engineered nanomaterials. Int J Mol Sci 21: 7310. https://doi.org/10.3390/ijms21197310
    [132] Singh A, Kukreti R, Saso L, et al. (2019) Oxidative stress: A key modulator in neurodegenerative diseases. Molecules 24: 1583. https://doi.org/10.3390/molecules24081583
    [133] Gänger S, Schindowski K (2018) Tailoring formulations for intranasal nose-to-brain delivery: a review on architecture, physico-chemical characteristics and mucociliary clearance of the nasal olfactory mucosa. Pharmaceutics 10: 116. https://doi.org/10.3390/pharmaceutics10030116
    [134] Guo C, Xia Y, Niu P, et al. (2015) Silica nanoparticles induce oxidative stress, inflammation, and endothelial dysfunction in vitro via activation of the MAPK/Nrf2 pathway and nuclear factor-κB signaling. Int J Nanomedicine 10: 1463-1477. https://doi.org/10.2147/IJN.S76114
    [135] Uskoković V (2021) Nanomedicine for the poor: a lost cause or an idea whose time has yet to come?. Nanomedicine (Lond) 16: 1203-1218. https://doi.org/10.2217/nnm-2021-0024
    [136] Shubhika Kwatra (2013) Nanotechnology and medicine–The upside and the downside. Int. J Drug Dev Res 5: 1-10. http://www.ijddr.in
    [137] Diwan A, Tatake J, Chakraborty A (2022) Therapeutic uses of TheraCour™ polymeric nanomicelles against cancer, infectious diseases and more. Nanomaterials for Cancer Detection Using Imaging Techniques and Their Clinical Applications.Springer Nature 473-506. https://doi.org/10.1007/978-3-031-09636-5_17
    [138] Chakraborty A, Diwan A, Barton R, et al. (2022) A new antiviral regimen against sars-cov-2 based on nanoviricide's biopolymer (NV-CoV-2). Front Nanotechnol 4: 891605. https://doi.org/10.3389/fnano.2022.891605
    [139] Barton RW, Tatake JG, Diwan AR (2011) Nanoviricides-A novel approach to antiviral therapeutics. Bionanotechnology II.CRC Press 141-154.
    [140] Chakraborty A, Diwan A (2020) NL-63: A better surrogate virus for studying SARS-CoV-2. Integr Mol Med 7: 1-9. https://doi.org/10.15761/IMM.1000408
    [141] NanoViricides, Inc., Pan-coronavirus COVID-19 Drug Candidates Are Highly Effective in Pre-clinical Animal Studies in Support of FDA Pre IND Application (2021). Available from: https://www.bloomberg.com/press-releases/2021-03-09/nanoviricides-inc-pan-coronavirus-covid-19-drug-candidates-are-highly-effective-in-pre-clinical-animal-studies-i.
    [142] Barton RW, Tatake JG, Diwan AR (2016) Nanoviricides: Targeted Anti-Viral Nanomaterials. Handbook of Clinical Nanomedicine, Nanoparticles, Imaging, Therapy, and Clinical Applications.Jenny Stanford Publishing 1039-1046.
    [143] NanoViricides is Developing Drugs Against SARS-CoV-2 with an Integrated Approach to Combat COVID-19, as Reported at The LD 500 Virtual Conference (2020). Available from: https://www.accesswire.com/604794/NanoViricides-is-Developing-Drugs-Against-SARS-CoV-2-with-an-Integrated-Approach-to-Combat-COVID-19-as-Reported-at-The-LD-500-Virtual-Conference.
    [144] Pal M, Berhanu G, Desalegn C, et al. (2020) Severe acute respiratory syndrome coronavirus-2 (sars-cov-2): an update. Cureus 12: e7423. https://doi.org/10.7759/cureus.7423
    [145] Chatterjee S, Bhattacharya M, Nag S, et al. (2023) A detailed overview of sars-cov-2 omicron: its sub-variants, mutations and pathophysiology, clinical characteristics, immunological landscape, immune escape, and therapies. Viruses 15: 167. https://doi.org/10.3390/v15010167
    [146] Beigel JM, Tomashek KM, Dodd LE, et al. (2020) Remdesivir for the treatment of Covid-19 final report. N Engl J Med 83: 1813-1826. https://doi.org/10.1056/NEJMoa2007764
    [147] ClinicalTrials.gov, multicenter, retrospective study of the effects of remdesivir in the treatment of severe Covid-19 infections, 2021. Available from: https://clinicaltrials.gov/ct2/show/NCT04365725
    [148] Chakraborty A, Diwan A, Arora V, et al. (2022) Polymer protects the drug and improves its pharmacokinetics. EC Pharmacol Toxicol 10.2: 108-118. https://ecronicon.org/assets/ecpt/pdf/ECPT-10-00707.pdf
    [149] Osorno LL, Brandley AN, Maldonado DE, et al. (2021) Review of contemporary self-assembled systems for the controlled delivery of therapeutics in medicine. Nanomaterials (Basel) 11: 278. https://doi.org/10.3390/nano11020278
    [150] Yadav S, Sharma AK, Kumar P (2020) Nanoscale self-assembly for therapeutic delivery. Front Bioeng Biotechnol 8: 127. https://doi.org/10.3389/fbioe.2020.00127
    [151] Li H, Labean TH, Leong KW (2011) Nucleic acid-based nanoengineering: novel structures for biomedical applications. Interface Focus 1: 702-24. https://doi.org/10.1098/rsfs.2011.0040
    [152] Marshall ML, Wagstaff KM (2020) Internalized functional DNA aptamers as alternative cancer therapies. Front Pharmacol 11: 1115. https://doi.org/10.3389/fphar.2020.01115
    [153] Ni X, Castanares M, Mukherjee A, et al. (2011) Nucleic acid aptamers: clinical applications and promising new horizons. Curr Med Chem 18: 4206-14. https://doi.org/10.2174/092986711797189600
    [154] Li J, Mo L, Lu CH, et al. (2016) Functional nucleic acid-based hydrogels for bioanalytical and biomedical applications. Chem Soc Rev 45: 1410-31. https://doi.org/10.1039/C5CS00586H
    [155] Abune L, Wang Y (2021) Affinity hydrogels for protein delivery. Trends Pharmacol Sci 42: 300-312. https://doi.org/10.1016/j.tips.2021.01.005
  • Reader Comments
  • © 2023 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1333) PDF downloads(166) Cited by(0)

Article outline

Figures and Tables

Figures(3)  /  Tables(10)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog