483
Views
81
CrossRef citations to date
0
Altmetric
Review

Chitosan nanoparticles in drug therapy of infectious and inflammatory diseases

, , , &
Pages 1177-1194 | Received 15 Jan 2016, Accepted 04 Apr 2016, Published online: 02 May 2016

References

  • Pradeep KD, Joy DD, Tripathi VS. Chitin and chitosan: chemistry, properties and applications. JSIR. 2004;63:23–30.
  • Rinaudo M. Chitin and chitosan: properties and applications. Prog Polym Sci. 2006;31:603–632.
  • Jolanta K, Weinhold MX, Thöming J, et al. Biomedical activity of chitin/chitosan based material influence of physicochemical properties apart from molecular weight and degree of N-Acetylation. Polymers. 2011;3:1875–01.
  • Bo M, Bavestrello G, Kurek D, et al. Isolation and identification of chitin in black coral paranthipates larix (Anthozoa: Cnidaria). Int J Biol Macromol. 2012;51:129–137.
  • Brunner E, Richthammer P, Ehrlich H, et al. Chitin-based organic networks: an integral part of cell wall biosilica in the diatom Thalassiosira pseudonana. Angew Chem Int Ed Engl. 2009;48:9724–9727.
  • Ehrlich H, Maldonado M, Spindler KD, et al. First evidence of chitin as a component of the skeletal fibers of marine sponges. Part I. Verongidae (Demospongia: Porifera). J Exp Zool B Mol Dev Evol. 2007;308:347–356.
  • Ehrlich H, Krautter M, Hanke T, et al. First evidence of the presence of chitin in skeletons of marine sponges. Part II. Glass sponges (Hexactinellida: Porifera). J Exp Zool B Mol Dev Evol. 2007;308:473–483.
  • Ehrlich H, Ilan M, Maldonado M, et al. Three dimensional chitin-based scaffolds from Verongida sponges (Demospongiae: Porifera). Part I. Isolation and identification of chitin. Int J Biol Macromol. 2010;7:132–140.
  • Ehrlich H. Chitin and collagen as universal and alternative templates in bio-mineralization. Int Geol Rev. 2010;52:661–699.
  • Anitha A, Sowmya S, Sudheesh PT, et al. Chitin and chitosan in selected biomedical applications. Prog Polym Sci. 2014;39:1644–1667.
  • Riccardo A, Muzzarelli A. Biomedical exploitation of chitin and chitosan via mechano-chemical disassembly, electro spinning, dissolution in imidazolium ionic liquids, and supercritical drying. Mar Drugs. 2011;9:1510–1533.
  • Dasha M, Chiellini F, Ottenbriteb RM, et al. Chitosan-A versatile semi-synthetic polymer in biomedical applications. Prog Polym Sci. 2011;36:981–1014.
  • Gaurav L, Yon SK, Jin WH, et al. Chito oligosaccharide and its derivatives: preparation and biological applications. Bio Med Res Int. 2014;10:1–13.
  • CAutho F, Christine J. Chitosan-based biomaterials for tissue engineering. Eur Polym J. 2013;49:780–792.
  • Susana R, Marita D, Carmen RL, et al. Biocompatibility of chitosan carriers with application in drug delivery. J Funct Biomater. 2012;3:615–641.
  • Agnihotri SA, Mallikarjuna NN, Aminabhavi TM, et al. Recent advances on chitosan-based micro- and nanoparticles in drug delivery. J. Control Release. 2004;100:5–28.
  • Sneha P, Aiswarya J, Changam S, et al. Extraction and purification of chitosan from chitin isolated from sea prawn Fenneropenaeus indicus. Asian J Pharm Clin Res. 2014;7:201–204.
  • Rejinold NS, Nair A, Sabitha M, et al. Synthesis, characterization and in vitro cyto compatibility studies of chitin nanogels for biomedical applications. Carbohyd Polym. 2012;87:943–949.
  • Senel S, McClure SJ. Potential applications of chitosan in veterinary medicine. Adv Drug Deliv Rev. 2004;56:1467–1480.
  • Manish PP, Ravi RP, Jayvadan KP. Chitosan mediated targeted drug delivery system: a review. J Pharm Pharmaceut Sci. 2010;13:536–557.
  • Kenneth CO, Franklin CK. Formulation development and evaluation of drug release kinetics from colon-targeted ibuprofen tablets based on eudragit RL 100-chitosan inter polyelectrolyte complexes. ISRN Pharm. 2013;13:1–8.
  • Aruna U, Rajalakshmi R, Indira MY, et al. Role of chitosan nanoparticles in cancer therapy. IJIPSR. 2013;4:318–324.
  • Ehrlich H, Krajewska B, Hanke T, et al. Chitosan membrane as a template for hydroxyl apatite crystal growth in a model dual membrane diffusion system. J Membrane Sci. 2006;273:124–128.
  • Lee J, Lee C, Kim TH, et al. Self-assembled glycol chitosan nanogels containing palmityl-acylated exendin-4peptide as a long-acting anti-diabetic inhalation system. J Control Release. 2012;161:728–734.
  • Yang T-L. Chitin-based materials in tissue engineering: applications in soft tissue and epithelial organ. Int J Mol Sci. 2011;12:1936–1963.
  • Dev A, Binulal NS, Anitha A, et al. Preparation of poly (lactic acid)/chitosan nanoparticles for anti-HIV drug delivery applications. Carbohydr Polym. 2010;80:833–838.
  • Manjusha R, Anuja A, Yuvraj SN. Review: chitosan based hydrogel polymeric beads- as drug delivery system. BioResources. 2010;5:2765–2807.
  • Yi H, Haiqing S, Yihong G, et al. Covalently cross linked chitosan hydrogel: properties of in vitro degradation and chondrocyte encapsulation. Acta Biomaterialia. 2007;3:23–31.
  • Nwe N, Furuike T, Tamura H. The mechanical and biological properties of chitosan scaffolds for tissue regeneration templates are significantly enhanced by chitosan from Gongronella butleri. Materials. 2009;2:374–398.
  • Tarun G, Arsh C, Joshi R. Preparation of chitosan scaffolds for tissue engineering using freeze drying technology. J Pharm Res. 2012;2:72–73.
  • Koa JA, Park HJ, Hwang SJ, et al. Preparation and characterization of chitosan micro particles intended for controlled drug delivery. Int J Pharm. 2002;24:165–174.
  • Sezer AD, Cevher E. Topical drug delivery using chitosan nano and microparticles. Expert Opin Drug Deliv. 2012;9:1129–1146.
  • Yassin AEB, Alsarra IA, Al-Mohizea AM. Chitosan beads as a new gastro retentive system of verapamil. Sci Pharm. 2006;74:175–188.
  • Ikeda T, Ikeda K, Kouhei Y, et al. Fabrication and characteristics of chitosan sponge as a tissue engineering scaffold. Bio Med Res Int. 2014;10:1–8.
  • Cunxian D, Dianrui Z, Feihu W, et al. Chitosan-g-poly(N-isopropylacrylamide) based nanogels for tumor extracellular targeting. Int J Pharm. 2011;409:252–259.
  • Rapha R, Heloıse R, Annedes R. Chitosan and chitosan derivatives in drug delivery and tissue engineering. Adv Polym Sci. 2011;244:19–44.
  • Anitha A, Divya Rani V, Krishna R, et al. Synthesis, characterization, cytotoxicity and antibacterial studies of chitosan, O-carboxymethyl and N, O-carboxymethyl chitosan nanoparticles. Carbohydr Polym. 2009;9:672–677.
  • Nagpal K, Singh SK, Mishra DN. Chitosan nanoparticles: a promising system in novel drug delivery. Chem Pharm Bull. 2010;58:1423–1430.
  • Rajalakshmi R, Indira MY, Aruna U, et al. Chitosan nanoparticles- an emerging trend in nanotechnology. IJDDT. 2014;6:204–229.
  • Yosefine A, Yun-OO O, Hyun WK, et al. Chitosan-coated Fe3O4 magnetic nanoparticles as carrier of cisplatin for drug delivery. Fish Aquat Sci. 2015;18:89–98.
  • Yateendra SP, Saikishore V, Satyanarayana J. Drug delivery systems using chitosan nanoparticles. Am J Pharm Tech Res. 2012;2:1–19.
  • Grenha A. Biocompatibility of chitosan carriers with application in drug delivery. J Funct Biomater. 2012;3:615–641.
  • Venkatesan C, Vimal S, Sahul Hameed AS. Synthesis and characterization of chitosan tri polyphosphate nanoparticles and its encapsulation efficiency containing Russell’s viper snake venom. J Bio Chem Mol Toxic. 2013;8:406–411.
  • Grenha A. Chitosan nanoparticles: a survey of preparation methods. J Drug Target. 2012;20:291–300.
  • Sailaja AK, Amareshwar P, Chakravarty P. Different techniques used for the preparation of nanoparticles using natural polymers and their application. Int J Pharm Pharm Sci. 2011;3:45–50.
  • Barbi MS, Carvalho FC, Kiill CP, et al. Preparation and characterization of chitosan nanoparticles for zidovudine nasal delivery. J Nanosci Nanotechnol. 2015;15:865–874.
  • Rajalakshmi R, Indira Muzib Y, Aruna U, et al. Chitosan nanoparticles - an emerging trend in nanotechnology. Int J Drug Dev Res. 2014;6:204–229.
  • Balcerzak J, Kucharska M, Gruchała B. Preparation of micro and nanostructures of chitosan by ultrasonic coalescence of w/o emulsions. PCACD. 2013;18:13–20.
  • Shering AM, Kannan C, Sabari KK, et al. Formulation of 5-fluorouracil loaded chitosan nanoparticles by emulsion droplet coalescence method for cancer therapy. Int J Pharm Biol Sci Arch. 2011;2:926–931.
  • Ahmed TA, El-Say KM. Development of alginate-reinforced chitosan nanoparticles utilizing W/O nanoemulsification/internal cross linking technique for transdermal delivery of rabeprazole. Life Sci. 2014;110:35–43.
  • Wang JJ, Zeng ZW, Xiao RZ, et al. Recent advances of chitosan nanoparticles as drug carriers. Int J Nanomedicine. 2011;6:765–774.
  • Pourshahab PS, Gilani K, Moazeni E, et al. Preparation and characterization of spray dried inhalable powders containing chitosan nanoparticles for pulmonary delivery of isoniazid. J Microencapsul. 2011;287:605–613.
  • Kafshgari MH, Khorram M, Mansouri M, et al. Preparation of alginate and chitosan nanoparticles using a new reverse micellar system. Iran Polymer J. 2012;21:99–107.
  • Lv -P-P, Wei W, Gong F-L, et al. Preparation of uniformly sized chitosan nanospheres by a premix membrane emulsification technique. Ind Eng Chem Res. 2009;48:8819–8828.
  • Poovi G, Dhana Lekshmi UM, Narayanan N, et al. Preparation and characterization of repaglinide loaded chitosan polymeric nanoparticles. Res J Nanosci Nanotechnol. 2001;1:12–24.
  • Shahverdi S, Hajimiri M, Atyabi F, et al. Preparation and characterization of aFGF- loaded chitosan nanoparticles with two different methods. Res Pharm Sci. 2012;7:233–239.
  • Agnihotri SA, Aminabhavi TM. Chitosan nanoparticles for prolonged of timolol maleate. Drug Dev Ind Pharm. 2007;33:1254–1262.
  • Kenneth VIR. Challenges in the Treatment of infections caused by gram-positive and gram-negative bacteria in patients with cancer and neutropenia. Clin Infect Dis. 2005;40:S246–52.
  • Stephen SM. Factors in the emergence of infectious diseases. Emerg Infect Dis. 1995;1:7–15.
  • Zhang L, Pornpattananangkul D, Huang M. Development of nanoparticles for antimicrobial drug delivery. Curr Med Chem. 2010;17:585–594.
  • Chia HK, Chien YH, Tin YH. Assessment of chitosan-affected metabolic response by peroxisome proliferator-activated receptor bioluminescent imaging-guided transcriptomic analysis. PLoS ONE. 2012;4:4–15.
  • Monarul I, Shah M, Khandaker RM, et al. Antibacterial activity of crab-chitosan against Staphylococcus aureus and Escherichia coli. J AdvScient Res. 2011;2:63–66.
  • Ying CC, Jan YY, Cheng FT. Antibacterial characteristics and activity of water-soluble chitosan derivatives prepared by the maillard reaction. Molecules. 2011;16:8504–8514.
  • Rejane CG, De Douglas B, Odilio BG. A review of the antimicrobial activity of chitosan. Polimeros Ciencia Tecnologia. 2009;19:241–247.
  • Fernanda A, Francisco G, Diego A, et al. Chitosan-grafted copolymers and chitosan-ligand conjugates as matrices for pulmonary drug delivery. Int J Carbohydr Chem. 2011;14:1–14.
  • Rancan F, Blume-Peytavi U, Vogt A. Utilization of biodegradable polymeric materials as delivery agents in dermatology. Clin Cosmet Investig Dermatol. 2014;7:23–34.
  • Saharan V, Sharma G, Yadav M, et al. Synthesis and in vitro antifungal efficacy of Cu- chitosan nanoparticles against pathogenic fungi of tomato. Int J BiolMacromol. 2015;75:346–353.
  • Narendhar C, Murugu MK, Rejitha M, et al. Potency of multi component drug delivery systems: a critical reveiw. J Chem Pharm Si. 2013;6:55–61.
  • Devika RB, Hrushikesh MJ, Murali S, et al. Chitosan reduced gold nanoparticles as novel carriers for transmucosal delivery of insulin. Pharmaceutical Res. 2007;24:1415–1426.
  • Gao P, Nie X, Zou M, et al. Recent advances in materials for extended-release antibiotic delivery system. J Antibiot. 2011;9:625–634.
  • Heidi MM, Min JS, Abeer AG, et al. for pharmaceutical dosage forms: molecular pharmaceutics and controlled release drug delivery aspects. Int J MolSci. 2010;11:3298–3322.
  • Zhang L, Pornpattanan D, Hu MJ, et al. Development of nanoparticles for anti microbial drug delivery. Curr Med Chem. 2010;17:585–594.
  • Shweta R, Parul S, Pritam V, et al. Nanoparticle-based drug delivery systems: promising approaches against infections. Braz Arch Biol Technol. 2014;57:209–222.
  • Jian MS, Luan X, Yan L, et al. Chitosan-based luminescent/magnetic hybrid nanogels for insulin delivery, cell imaging, and anti-diabetic research of dietary supplements. Int J Pharm. 2012;427:400–409.
  • Bushra J, Huma H, Shahid A, et al. Cefazolin loaded chitosan nanoparticles to cure multi drug resistant gram-negative pathogens. Carbohydrpolym. 2015;78:1–33.
  • Bhavin KP, Rajesh HP, Pooja SA, et al. Development of oral sustained release rifampicin loaded chitosan nanoparticles by design of experiment. JDDST. 2013;1:1–10.
  • Costa JR, Silva NC, Sarmento B, et al. Potential chitosan-coated alginate nanoparticles for ocular delivery of daptomycin. J ClinMicrobiol Infect Dis. 2015;34:1255–1262.
  • Admire D, Jessica LR, Law CW, et al. Multimodal nanoparticles that provide immune modulation and intracellular drug delivery for infectious diseases. Nanomedicine. 2014;4:831–838.
  • Ustundag-Okur N, Gokçe EH, Bozbıyık D, et al. Preparation and in vitro-in vivo evaluation of ofloxacin loaded ophthalmic nano structured lipid carriers modified with chitosan oligosaccharide lactate for the treatment of bacterial keratitis. Eur J Pharm Sci. 2014;63:204–215.
  • Jain V, Gupta A, Pawar VK, et al. Chitosan-assisted immunotherapy for intervention of experimental leishmaniasis via amphotericin B-loaded solid lipid nanoparticles. Appl Bio Chem Biotechnol. 2014;174:1309–1330.
  • Zhou W, Wang Y, Jian J, et al. Self-aggregated nanoparticles based on amphiphilicpoly (lactic acid)-grafted-chitosan copolymer for ocular delivery of amphotericin B. Int J Nanomedicine. 2013;8:3715–3728.
  • Tarun G, Goutam R, Amit KG. Inhalable chitosan nanoparticles as anti tubercular drug carriers for an effective treatment of tuberculosis. Artif Cell Nanomed Biotechnol. 2015;14:1–5.
  • Kong Z, Yu M, Cheng K, et al. Incorporation of chitosan nanospheres into thin mineralized collagen coatings for improving the antibacterial effect. Colloids Surf B Biointerfaces. 2013;111:536–541.
  • Gnanadhas DP, Ben Thomas M, Elango M, et al. Chitosan-dextran sulphate nanocapsule drug delivery system as an effective therapeutic against intra phagosomal pathogen Salmonella. J Antimicrob Chemother. 2013;68:2576–2586.
  • Lin YH, Tsai SC, Lai CH, et al. Genipin-cross-linked fucose-chitosan/heparin nanoparticles for the eradication of Helicobacter pylori. Biomaterials. 2013;34:4466–4479.
  • Moretton MA, Chiappetta DA, Andrade F, et al. Hydrolyzed galactomannan-modified nanoparticles and flower-like polymeric micelles for the active targeting of rifampicin to macrophages. J Biomed Nanotechnol. 2013;9:1076–1087.
  • Chhonker YS, Prasad YD, Chandasana H, et al. Amphotericin-B entrapped lecithin/chitosan nanoparticles for prolonged ocular application. Int J Biol Macromol. 2015;72:1451–1458.
  • Pornpattananangkul D, Zhang L, Olson S, et al. Bacterial toxin-triggered drug release from gold nanoparticle-stabilized liposomes for the treatment of bacterial infection. J Am Chem Soc. 2011;133:4132–4139.
  • Junise V, Saraswathi R. Development and characterization of inhaled chitosan nanoparticles loaded with isoniazid. JPTRM. 2014;2:159–170.
  • Maya S, Indulekha S, Sukhithasri V, et al. Efficacy of tetracycline encapsulated O-carboxymethyl chitosan nanoparticles against intracellular infections of Staphylococcus aureus. Int J Biol Macromol. 2012;51:392–399.
  • El Zowalaty ME, Hussein Al Ali SH, Husseiny MI, et al. The ability of streptomycin-loaded chitosan-coated magnetic nano composites to possess antimicrobial and anti-tuberculosis activities. Int J Nanomedicine. 2015;10:3269–3274.
  • Kiruthika V, Maya S, Maneesha KS, et al. Comparative efficacy of chloramphenicol loaded chondroitin sulfate and dextran sulfate nanoparticles to treat intracellular Salmonella infections. Colloids Surf B Biointerfaces. 2015;15:1–10.
  • Ling Y, Noraziah MZ, Atif S, et al. Antifungal activity of chitosan nanoparticles and correlation with their physical properties. Int J Bio Met. 2012;1:1–9.
  • Anna MP, Giuseppantonio M, Stefania S, et al. Chitosan nanoparticles loaded with the anti-microbial peptide temporin B exert a long term anti-bacterial activity in vitro against clinical isolates of Staphylococcus epidermidis. Front MicroBiol. 2015;6:1–10.
  • Tripathy S, Mahapatra SK, Chattopadhyay S, et al. A novel chitosan based anti-malarial drug delivery against Plasmodium berghei infection. Acta Trop. 2013;3:494–503.
  • Pandey R, Khuller GK. Nanotechnology based drug delivery system(s) for the management of tuberculosis. Indian J Exp Biol. 2006;44:357–366.
  • Paranjpe M, Müller-Goymann CC. Nanoparticle mediated pulmonary drug delivery: a review. Int J Mol Sci. 2014;15:5852–5873.
  • Gareth G, Nystrom B, Suraj B, et al. Nano bead-based interventions for the treatment and prevention of tuberculosis. Nature Rev. 2010;8:827–834.
  • Amit KG, Tarun G, Goutam R, et al. Development and characterization of nano embedded microparticles for pulmonary delivery of antitubercular drugs against experimental tuberculosis. Mol Pharm. 2015;11:3839–3850.
  • Elsabahy M, Nazarali A, Foldvari M. Non-viral nucleic acid delivery: key challenges and future directions. Curr Drug Deliv. 2011;8:235–244.
  • Layek B, Lipp L, Singh J. Cell penetrating peptide conjugated chitosan for enhanced delivery of nucleic acid. Int J Mol Sci. 2015;16:28912–28930.
  • Vimal S, Majeed A, Taju G, et al. Chitosan tripolyphosphate (CS/TPP) nanoparticles: preparation, characterization and application for gene delivery in shrimpS. Acta Trop. 2013;128:486–493.
  • Lu H-D, Zhao H-Q, Wang K, et al. Novel hyaluronic acid–chitosan nanoparticles as non-viral gene delivery vectorstargeting osteoarthritis. Int J Pharm. 2011;420:358–65.
  • Lu H, Dai Y, Lulu L, et al. Chitosan-graft-polyethylenimine/DNA nanoparticles as novel non-viral gene delivery vectors targeting osteoarthritis. PLoS One. 2014;9:1–12.
  • Layek B, Singh J. Amino acid grafted chitosan for high performance gene delivery: comparison of amino acid hydrophobicity on vector and polyplex characteristics. Biomacromolecules. 2013;14:485–494.
  • Boyapalle S, Xu W, Raulji P, et al. A multiple siRNA-based anti-HIV/SHIV microbicide shows protection in both in vitroand in vivo models. PLoS One. 2015;10:1–16.
  • Bird GH, Boyapalle S, Wong T, et al. Mucosal delivery of a double-stapled RSV peptide prevents nasopulmonary infection. J Clin Invest. 2014;124:2113–2124.
  • Oliveira CR, Rezende CF, Silva MR, et al. Oral vaccination based on DNA-chitosan nanoparticles againstSchistosomamansoni Infection. Scientific World J. 2012;2012:1–11.
  • Xu J, Dai W, Wang Z, et al. Intranasal vaccination with chitosan-DNA nanoparticles expressing pneumococcal surface antigen a protects mice against nasopharyngeal colonization by streptococcus pneumonia. Clin Vaccine Immunol. 2011;18:75–81.
  • Xin Zhang, Mysore K, Flannery E. et al. Chitosan/interfering RNA nanoparticle mediated gene silencing in disease vector mosquito larvae. J Vis Exp. 2015;97:1–20.
  • Foger F, Noonpakdee W, Loretz B, et al. Inhibition of malarial topoisomerase II in Plasmodium falciparum by antisense nanoparticles. Int J Pharm. 2006;319:139–146.
  • Park BK, Kim -M-M. Applications of chitin and its derivatives in biological medicine. Int J Mol Sci. 2010;11:5152–5164.
  • Kerch G. The potential of chitosan and its derivatives in prevention and treatment of age-related diseases. Mar Drugs. 2015;13:2158–2182.
  • Sarwar A, Katas H, Samsudin SN, et al. Regioselective sequential modification of Chitosan via Azide-Alkyne click reaction: synthesis, characterization, and antimicrobial activity of chitosan derivatives and nanoparticles. PLoS ONE. 2015;10:1–22.
  • Smitha KT, Sreelakshmi M, Biswas R, et al. Amidase encapsulated O-carboxymethyl chitosan nanoparticles for vaccine delivery. Int J Biol Macromol. 2014;63:154–157.
  • Ahmed TA, Aljaeid BM. Preparation, characterization, and potential application of chitosan, chitosan derivatives, and chitosan metal nanoparticles in pharmaceutical drug delivery. Drug Des Devel Ther. 2016;10:483–507.
  • Alamdarnejad G, Sharif A, Taranejoo S. Synthesis and characterization of thiolatedcarboxymethylchitosan-graft-cyclodextrin nanoparticles as a drug deliveryvehicle for albendazole. J Mater Sci Mater Med. 2013;24:1939–1949.
  • Billich A, Aschauer H, Aszódi A, et al. Percutaneous absorption of drugs used in atopic eczema: pimecrolimus permeates less through skin than corticosteroids and tacrolimus. Int J Pharm. 2004;269:29–35.
  • Jessy S, Maria L. Nanocarriers for targeting in inflammation. Asian J Pharm Clin Res. 2013;6:3–12.
  • Barksby HE, Lea SR, Preshaw PM. The expanding family of interleukin-1 cytokines and their role in destructive inflammatory disorders. Clin ExpImmunol. 2007;149:217–225.
  • Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev. 2013;65:36–48.
  • Tayal P. Implication of solid lipid nanoparticles for topical drug delivery. IJPRS. 2015;4:301–316.
  • Sivaram AJ, Rajitha P, Maya S, et al. Nanogels for delivery, imaging and therapy. WIRES Nanomed Nanobiotechnol. 2015;7:509–533.
  • Nitha V, Ramya DD. Progress in psoriasis therapy via novel drug delivery systems. Curr Dermatol Rep. 2014;6:14–18.
  • Park BK, Kim MM. Applications of chitin and its derivatives in biological medicine. Int J Mol Sci. 2010;11:5152–5164.
  • Oliveira MI, Santos SG, Oliveira MJ, et al. Chitosan drives anti-inflammatory macrophage polarisation and pro-inflammatory dendritic cell stimulation. Eur Cell Mater. 2012;24:136–153.
  • Wenshui X, Ping L, Jiali Z, et al. Biological activities of chitosan and chitooligo saccharides. Food Hydrocoll. 2011;2:170–179.
  • Hirva AS, Rakesh PP. Statistical modeling of zaltoprofen loaded biopolymeric nanoparticles: characterization and anti-inflammatory activity of nanoparticles loaded gel. Int J Pharma Investig. 2015;5:120–127.
  • Haliza K, Zahid H, Tay CL. Chitosan nanoparticles as a percutaneous drug delivery system for hydrocortisone. J Nanomater. 2012;1:1–11.
  • Fardet L, Flahault A, Kettaneh A, et al. Corticosteroid-induced clinical adverse events: frequency, risk factors and patient’s opinion. Br J Dermatol. 2007;157:142–148.
  • Hernández-Díaz S, Rodríguez LA. Steroids and risk of upper gastrointestinal complications. Am J Epidemiol. 2001;153:1089–1093.
  • Mehmet D, Nagihan U, Fatma Y, et al. Ketorolac tromethamine loaded chitosan nanoparticles as a nanotherapeutic system for ocular diseases. J Biol Chem. 2013;41:81–86.
  • Ridolfi DM, Marcato PD, Justo GZ, et al. Chitosan-solid lipid nanoparticles as carriers for topical delivery of tretinoin. Colloids Surf B Biointerfaces. 2012;93:36–40.
  • Zhang Z, Tsai P-C, Ramezanli T, et al. Polymeric nanoparticles-based topical delivery systems for the treatment of dermatological diseases. Wiley Interdiscip Rev NanomedNanobiotechnol. 2013;5:205–218.
  • Shah PP, Desai PR, Patel AR, et al. Skin permeating nanogel for the cutaneous co-delivery of two anti-inflammatory drugs. Biomaterials. 2012;33:1607–1617.
  • Lowenstein PR, Castro MG. Inflammation and adaptive immune responses to adenoviral vectors injected into the brain: peculiarities, mechanisms, and consequences. Gene Ther. 2003;10:946–954.
  • Kim -S-S, Ye C, Kumar P, et al. Targeted delivery of siRNA to macrophagesfor anti-inflammatory treatment. Mol Ther. 2010;18:993–1001.
  • Lee J, Yun K-S, Choi CS, et al. T cell-specific siRNA delivery using antibody-conjugated chitosan nanoparticles. Bio Conjugate Chem. 2012;23:1174–1180.
  • Jeong EJ, Choi M, Lee J, et al. The spacer arm length in cell-penetrating peptides influences chitosan/siRNA nanoparticle delivery for pulmonary inflammation treatment. Nanoscale. 2015;7:20095–20104.
  • Kumar M, Kong X, Behera AK, et al. IFN-γ-pDNA nanoparticle (CIN) therapy for allergic Asthma. Genet Vaccines Ther. 2003;1:1–10.
  • Venugopal V, Kumar J, Muralidharan S. Targeted delivery of silymarin to liver cells by galactosylated nanoparticles: in-vitro and in-vivo evaluation studies. AJPhSci. 2014;2:1–8.
  • Lee D-W, Shirley SA, Lockey RF, et al. Thiolated chitosan nanoparticles enhance anti-inflammatory effectsofintranasally delivered theophylline. Respir Res. 2006;7:1–10.
  • Trapania A, Sitterbergb J, Bakowskyb U, et al. The potential of glycol chitosan nanoparticles as carrier for lowwater soluble drugs. Int J Pharm. 2009;375:97–106.
  • Coco R, Plapied L, Pourcelle L, et al. Drug delivery to inflamed colon by nanoparticles: comparison of different strategies. Int J Pharm. 2013;440:3–12.
  • Lee SJ, Lee A, Hwang SR, et al. TNF-α Gene silencing using polymerized siRNA/ thiolated glycol chitosan nanoparticles for rheumatoid arthritis. Mol Ther. 2014;22:397–408.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.