Nano based drug delivery systems: recent developments and future prospects

Journal of Nanobiotechnology - Tập 16 Số 1 - 2018
Jayanta Kumar Patra1, Gitishree Das1, Leonardo Fernandes Fraceto2, Estefânia Vangelie Ramos Campos2, María del Pilar Rodriguez-Torres3, Laura Susana Acosta-Torres3, L.A. Dı́az-Torres4, Renato Grillo5, Mallappa Kumara Swamy6, Shivesh Sharma7, Solomon Habtemariam8, Han-Seung Shin9
1Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyang-si 10326, Republic of Korea
2Sao Paulo State University (UNESP), Institute of Science and Technology, Sorocaba, São Paulo, Zip Code 18087-180, Brazil
3Laboratorio de Investigación Interdisciplinaria, Área de Nanoestructuras y Biomateriales, Escuela Nacional de Estudios Superiores, Unidad Leon, Universidad Nacional Autonóma de México (UNAM), Boulevard UNAM No 2011. Predio El Saucillo y El Potrero, 37684, León, Guanajuato, Mexico
4Centro de Investigaciones en Óptica, A.P. 1-948, C.P. 37000, León, Guanajuato, Mexico
5Department of Physics and Chemistry, School of Engineering, São Paulo State University (UNESP), Ilha Solteira, SP, 15385-000, Brazil
6Department of Crop Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
7Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad, Uttar Pradesh, 211004, India
8Pharmacognosy Research Laboratories & Herbal Analysis Services UK, University of Greenwich, Medway Campus-Science, Grenville Building (G102/G107), Central Avenue, Chatham-Maritime, Kent, ME4 4TB, UK
9Department of Food Science and Biotechnology, Dongguk University, Ilsandong-gu, Goyang, Gyeonggi-do 10326, Republic of Korea

Tóm tắt

Từ khóa


Tài liệu tham khảo

Swamy MK, Sinniah UR. Patchouli (Pogostemon cablin Benth.): botany, agrotechnology and biotechnological aspects. Ind Crops Prod. 2016;87:161–76.

Mohanty SK, Swamy MK, Sinniah UR, Anuradha M. Leptadenia reticulata (Retz.) Wight & Arn. (Jivanti): botanical, agronomical, phytochemical, pharmacological, and biotechnological aspects. Molecules. 1019;2017:22.

Rodrigues T, Reker D, Schneider P, Schneider G. Counting on natural products for drug design. Nat Chem. 2016;8:531.

Siddiqui AA, Iram F, Siddiqui S, Sahu K. Role of natural products in drug discovery process. Int J Drug Dev Res. 2014;6(2):172–204.

Beutler JA. Natural products as a foundation for drug discovery. Curr Prot Pharmacol. 2009;46(1):9–11.

Thilakarathna SH, Rupasinghe H. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients. 2013;5:3367–87.

Bonifácio BV, da Silva PB, dos Santos Ramos MA, Negri KMS, Bauab TM, Chorilli M. Nanotechnology-based drug delivery systems and herbal medicines: a review. Int J Nanomed. 2014;9:1.

Watkins R, Wu L, Zhang C, Davis RM, Xu B. Natural product-based nanomedicine: recent advances and issues. Int J Nanomed. 2015;10:6055.

Martinho N, Damgé C, Reis CP. Recent advances in drug delivery systems. J Biomater Nanobiotechnol. 2011;2:510.

Jahangirian H, Lemraski EG, Webster TJ, Rafiee-Moghaddam R, Abdollahi Y. A review of drug delivery systems based on nanotechnology and green chemistry: green nanomedicine. Int J Nanomed. 2017;12:2957.

Liu Z, Tabakman S, Welsher K, Dai H. Carbon nanotubes in biology and medicine: in vitro and in vivo detection, imaging and drug delivery. Nano Res. 2009;2:85–120.

Orive G, Gascon AR, Hernández RM, Domı́nguez-Gil A, Pedraz JL. Techniques: new approaches to the delivery of biopharmaceuticals. Trends Pharmacol Sci. 2004;25:382–7.

Razzacki SZ, Thwar PK, Yang M, Ugaz VM, Burns MA. Integrated microsystems for controlled drug delivery. Adv Drug Deliv Rev. 2004;56:185–98.

Arayne MS, Sultana N, Qureshi F. nanoparticles in delivery of cardiovascular drugs. Pak J Pharm Sci. 2007;20:340–8.

Patra JK, Baek K-H. Green nanobiotechnology: factors affecting synthesis and characterization techniques. J Nanomater. 2014;2014:219.

Joseph RR, Venkatraman SS. Drug delivery to the eye: what benefits do nanocarriers offer? Nanomedicine. 2017;12:683–702.

Mirza AZ, Siddiqui FA. Nanomedicine and drug delivery: a mini review. Int Nano Lett. 2014;4:94.

Rudramurthy GR, Swamy MK, Sinniah UR, Ghasemzadeh A. Nanoparticles: alternatives against drug-resistant pathogenic microbes. Molecules. 2016;21:836.

Lam P-L, Wong W-Y, Bian Z, Chui C-H, Gambari R. Recent advances in green nanoparticulate systems for drug delivery: efficient delivery and safety concern. Nanomedicine. 2017;12:357–85.

Haba Y, Kojima C, Harada A, Ura T, Horinaka H, Kono K. Preparation of poly (ethylene glycol)-modified poly (amido amine) dendrimers encapsulating gold nanoparticles and their heat-generating ability. Langmuir. 2007;23:5243–6.

Shi X, Sun K, Baker JR Jr. Spontaneous formation of functionalized dendrimer-stabilized gold nanoparticles. J Phys Chem C. 2008;112:8251–8.

Park S-H, Oh S-G, Mun J-Y, Han S-S. Loading of gold nanoparticles inside the DPPC bilayers of liposome and their effects on membrane fluidities. Coll Surf B. 2006;48:112–8.

de Villiers MM, Aramwit P, Kwon GS. Nanotechnology in drug delivery. New York: Springer; 2008.

Kabanov AV, Lemieux P, Vinogradov S, Alakhov V. Pluronic® block copolymers: novel functional molecules for gene therapy. Adv Drug Deliv Rev. 2002;54:223–33.

Wang N, Feng Y. Elaborating the role of natural products-induced autophagy in cancer treatment: achievements and artifacts in the state of the art. BioMed Res Int. 2015;2015:934207.

Ouattara B, Simard RE, Holley RA. Piette GJ-P, Bégin A: Antibacterial activity of selected fatty acids and essential oils against six meat spoilage organisms. Int J Food Microbiol. 1997;37:155–62.

Sharma G, Raturi K, Dang S, Gupta S, Gabrani R. Combinatorial antimicrobial effect of curcumin with selected phytochemicals on Staphylococcus epidermidis. J Asian Nat Prod Res. 2014;16:535–41.

Abdelwahab SI, Sheikh BY, Taha MME, How CW, Abdullah R, Yagoub U, El-Sunousi R, Eid EE. Thymoquinone-loaded nanostructured lipid carriers: preparation, gastroprotection, in vitro toxicity, and pharmacokinetic properties after extravascular administration. Int J Nanomed. 2013;8:2163.

Krauel K, Pitaksuteepong T, Davies NM, Rades T. Entrapment of bioactive molecules in poly (alkylcyanoacrylate) nanoparticles. Am J Drug Deliv. 2004;2:251–9.

Tan Q, Liu W, Guo C, Zhai G. Preparation and evaluation of quercetin-loaded lecithin-chitosan nanoparticles for topical delivery. Int J Nanomed. 2011;6:1621.

Sanna V, Roggio AM, Siliani S, Piccinini M, Marceddu S, Mariani A, Sechi M. Development of novel cationic chitosan-and anionic alginate–coated poly (d, l-lactide-co-glycolide) nanoparticles for controlled release and light protection of resveratrol. Int J Nanomed. 2012;7:5501.

Casettari L, Illum L. Chitosan in nasal delivery systems for therapeutic drugs. J Control Release. 2014;190:189–200.

Obeid MA, Al Qaraghuli MM, Alsaadi M, Alzahrani AR, Niwasabutra K, Ferro VA. Delivering natural products and biotherapeutics to improve drug efficacy. Ther Deliv. 2017;8:947–56.

Miele E, Spinelli GP, Miele E, Di Fabrizio E, Ferretti E, Tomao S, Gulino A. Nanoparticle-based delivery of small interfering RNA: challenges for cancer therapy. Int J Nanomed. 2012;7:3637.

McNamara K, Tofail SA. Nanosystems: the use of nanoalloys, metallic, bimetallic, and magnetic nanoparticles in biomedical applications. Phys Chem Chem Phys. 2015;17:27981–95.

Saadeh Y, Vyas D. Nanorobotic applications in medicine: current proposals and designs. Am J Robot Surg. 2014;1:4–11.

Oliveira ON Jr, Iost RM, Siqueira JR Jr, Crespilho FN, Caseli L. Nanomaterials for diagnosis: challenges and applications in smart devices based on molecular recognition. ACS Appl Mater Interfaces. 2014;6:14745–66.

De Jong WH, Borm PJ. Drug delivery and nanoparticles: applications and hazards. Int J Nanomed. 2008;3:133.

Holzinger M, Le Goff A, Cosnier S. Nanomaterials for biosensing applications: a review. Front Chem. 2014;2:63.

Golovin YI, Gribanovsky SL, Golovin DY, Klyachko NL, Majouga AG, Master AM, Sokolsky M, Kabanov AV. Towards nanomedicines of the future: remote magneto-mechanical actuation of nanomedicines by alternating magnetic fields. J Control Release. 2015;219:43–60.

Lu H, Wang J, Wang T, Zhong J, Bao Y, Hao H. Recent progress on nanostructures for drug delivery applications. J Nanomater. 2016;2016:20.

Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33:941.

Kumari A, Kumar V, Yadav S. Nanotechnology: a tool to enhance therapeutic values of natural plant products. Trends Med Res. 2012;7:34–42.

Chen F, Ehlerding EB, Cai W. Theranostic nanoparticles. J Nucl Med. 2014;55:1919–22.

Swierczewska M, Han H, Kim K, Park J, Lee S. Polysaccharide-based nanoparticles for theranostic nanomedicine. Adv Drug Deliv Rev. 2016;99:70–84.

Chen K, Chen X. Design and development of molecular imaging probes. Curr Top Med Chem. 2010;10:1227–36.

Yhee JY, Son S, Kim SH, Park K, Choi K, Kwon IC. Self-assembled glycol chitosan nanoparticles for disease-specific theranostics. J Control Release. 2014;193:202–13.

Lee C-M, Jang D, Kim J, Cheong S-J, Kim E-M, Jeong M-H, Kim S-H, Kim DW, Lim ST, Sohn M-H, et al. Oleyl-Chitosan nanoparticles based on a dual probe for Optical/MR imaging in vivo. Bioconjug Chem. 2011;22:186–92.

Yang S-J, Lin F-H, Tsai H-M, Lin C-F, Chin H-C, Wong J-M, Shieh M-J. Alginate-folic acid-modified chitosan nanoparticles for photodynamic detection of intestinal neoplasms. Biomaterials. 2011;32:2174–82.

Ryu JH, Na JH, Ko HK, You DG, Park S, Jun E, Yeom HJ, Seo DH, Park JH, Jeong SY. Non-invasive optical imaging of cathepsin B with activatable fluorogenic nanoprobes in various metastatic models. Biomaterials. 2014;35:2302–11.

Lapčík L, Lapcik L, De Smedt S, Demeester J, Chabrecek P. Hyaluronan: preparation, structure, properties, and applications. Chem Rev. 1998;98:2663–84.

Kim H, Kim Y, Kim I-H, Kim K, Choi Y. ROS-responsive activatable photosensitizing agent for imaging and photodynamic therapy of activated macrophages. Theranostics. 2014;4:1.

Choi KY, Chung H, Min KH, Yoon HY, Kim K, Park JH, Kwon IC, Jeong SY. Self-assembled hyaluronic acid nanoparticles for active tumor targeting. Biomaterials. 2010;31:106–14.

Kamat M, El-Boubbou K, Zhu DC, Lansdell T, Lu X, Li W, Huang X. Hyaluronic acid immobilized magnetic nanoparticles for active targeting and imaging of macrophages. Bioconjug Chem. 2010;21:2128–35.

Arpicco S, Lerda C, Dalla Pozza E, Costanzo C, Tsapis N, Stella B, Donadelli M, Dando I, Fattal E, Cattel L. Hyaluronic acid-coated liposomes for active targeting of gemcitabine. Eur J Pharm Biopharm. 2013;85:373–80.

Wang G, Gao S, Tian R, Miller-Kleinhenz J, Qin Z, Liu T, Li L, Zhang F, Ma Q, Zhu L. Theranostic hyaluronic acid-iron micellar nanoparticles for magnetic-field-enhanced in vivo cancer chemotherapy. ChemMedChem. 2018;13:78–86.

Choi KY, Jeon EJ, Yoon HY, Lee BS, Na JH, Min KH, Kim SY, Myung S-J, Lee S, Chen X. Theranostic nanoparticles based on PEGylated hyaluronic acid for the diagnosis, therapy and monitoring of colon cancer. Biomaterials. 2012;33:6186–93.

Gombotz WR, Wee S. Protein release from alginate matrices. Adv Drug Deliv Rev. 1998;31:267–85.

Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog Polym Sci. 2012;37:106–26.

Baghbani F, Moztarzadeh F, Mohandesi JA, Yazdian F, Mokhtari-Dizaji M. Novel alginate-stabilized doxorubicin-loaded nanodroplets for ultrasounic theranosis of breast cancer. Int J Biol Macromol. 2016;93:512–9.

Podgórna K, Szczepanowicz K, Piotrowski M, Gajdošová M, Štěpánek F, Warszyński P. Gadolinium alginate nanogels for theranostic applications. Coll Surf B. 2017;153:183–9.

Moscovici M. Present and future medical applications of microbial exopolysaccharides. Front Microbiol. 1012;2015:6.

Ding Z, Liu P, Hu D, Sheng Z, Yi H, Gao G, Wu Y, Zhang P, Ling S, Cai L. Redox-responsive dextran based theranostic nanoparticles for near-infrared/magnetic resonance imaging and magnetically targeted photodynamic therapy. Biomater Sci. 2017;5:762–71.

Hong S-P, Kang SH, Kim DK, Kang BS. Paramagnetic nanoparticle-based targeting theranostic agent for c6 rat glioma cell. J Nanomater. 2016; 2016:7617894. https://doi.org/10.1155/2016/7617894 .

Mignani S, El Kazzouli S, Bousmina M, Majoral JP. Expand classical drug administration ways by emerging routes using dendrimer drug delivery systems: a concise overview. Adv Drug Deliv Rev. 2013;65:1316–30.

Lounnas V, Ritschel T, Kelder J, McGuire R, Bywater RP, Foloppe N. Current progress in structure-based rational drug design marks a new mindset in drug discovery. Comput Struc Biotechnol J. 2013;5:e201302011.

Mavromoustakos T, Durdagi S, Koukoulitsa C, Simcic M, Papadopoulos M, Hodoscek M, Golic Grdadolnik S. Strategies in the rational drug design. Curr Med Chem. 2011;18:2517–30.

Wong PT, Choi SK. Mechanisms of drug release in nanotherapeutic delivery systems. Chem Rev. 2015;115:3388–432.

Prachayasittikul V, Worachartcheewan A, Shoombuatong W, Songtawee N, Simeon S, Prachayasittikul V, Nantasenamat C. Computer-aided drug design of bioactive natural products. Curr Top Med Chem. 2015;15:1780–800.

Chen G, Roy I, Yang C, Prasad PN. Nanochemistry and nanomedicine for nanoparticle-based diagnostics and therapy. Chem Rev. 2016;116:2826–85.

Pelaz B, Alexiou C, Alvarez-Puebla RA, Alves F, Andrews AM, Ashraf S, Balogh LP, Ballerini L, Bestetti A, Brendel C, Bosi S. Diverse applications of nanomedicine. Acs Nano. 2017;11:2313–81.

Mattos BD, Rojas OJ, Magalhaes WLE. Biogenic silica nanoparticles loaded with neem bark extract as green, slow-release biocide. J Clean Prod. 2017;142:4206–13.

Kinnear C, Moore TL, Rodriguez-Lorenzo L, Rothen-Rutishauser B, Petri-Fink A. Form follows function: nanoparticle shape and its implications for nanomedicine. Chem Rev. 2017;117:11476–521.

Sethi M, Sukumar R, Karve S, Werner ME, Wang EC, Moore DT, Kowalczyk SR, Zhang L, Wang AZ. Effect of drug release kinetics on nanoparticle therapeutic efficacy and toxicity. Nanoscale. 2014;6:2321–7.

Mattos BD, Tardy BL, Magalhaes WLE, Rojas OJ. Controlled release for crop and wood protection: recent progress toward sustainable and safe nanostructured biocidal systems. J Control Release. 2017;262:139–50.

Siepmann F, Herrmann S, Winter G, Siepmann J. A novel mathematical model quantifying drug release from lipid implants. J Control Release. 2008;128:233–40.

Ding CZ, Li ZB. A review of drug release mechanisms from nanocarrier systems. Mater Sci Eng. 2017;76:1440–53.

Lee JH, Yeo Y. Controlled drug release from pharmaceutical nanocarriers. Chem Eng Sci. 2015;125:75–84.

Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem Rev. 2016;116:2602–63.

Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev. 2012;64:302–15.

Pelaz B, del Pino P, Maffre P, Hartmann R, Gallego M, Rivera-Fernandez S, de la Fuente JM, Nienhaus GU, Parak WJ. Surface functionalization of nanoparticles with polyethylene glycol: effects on protein adsorption and cellular uptake. Acs Nano. 2015;9:6996–7008.

Almalik A, Benabdelkamel H, Masood A, Alanazi IO, Alradwan I, Majrashi MA, Alfadda AA, Alghamdi WM, Alrabiah H, Tirelli N, Alhasan AH. Hyaluronic acid coated chitosan nanoparticles reduced the immunogenicity of the formed protein corona. Sci Rep. 2017;7:10542.

Martens TF, Remaut K, Deschout H, Engbersen JFJ, Hennink WE, van Steenbergen MJ, Demeester J, De Smedt SC, Braeckmans K. Coating nanocarriers with hyaluronic acid facilitates intravitreal drug delivery for retinal gene therapy. J Control Release. 2015;202:83–92.

Kolhar P, Anselmo AC, Gupta V, Pant K, Prabhakarpandian B, Ruoslahti E, Mitragotri S. Using shape effects to target antibody-coated nanoparticles to lung and brain endothelium. Proc Natl Acad Sci USA. 2013;110:10753–8.

Gao WW, Zhang LF. Coating nanoparticles with cell membranes for targeted drug delivery. J Drug Target. 2015;23:619–26.

Muller J, Bauer KN, Prozeller D, Simon J, Mailander V, Wurm FR, Winzen S, Landfester K. Coating nanoparticles with tunable surfactants facilitates control over the protein corona. Biomaterials. 2017;115:1–8.

Gao H, Yang Z, Zhang S, Cao S, Shen S, Pang Z, Jiang X. Ligand modified nanoparticles increases cell uptake, alters endocytosis and elevates glioma distribution and internalization. Sci Rep. 2013;3:2534.

Jain A, Jain SK. Ligand-appended BBB-targeted nanocarriers (LABTNs). Crit Rev Ther Drug Carrier Syst. 2015;32:149–80.

Shen HX, Shi SJ, Zhang ZR, Gong T, Sun X. Coating solid lipid nanoparticles with hyaluronic acid enhances antitumor activity against melanoma stem-like cells. Theranostics. 2015;5:755–71.

Gao X, Zhang J, Xu Q, Huang Z, Wang YY, Shen Q. Hyaluronic acid-coated cationic nanostructured lipid carriers for oral vincristine sulfate delivery. Drug Dev Ind Pharm. 2017;43:661–7.

Wang T, Hou JH, Su C, Zhao L, Shi YJ. Hyaluronic acid-coated chitosan nanoparticles induce ROS-mediated tumor cell apoptosis and enhance antitumor efficiency by targeted drug delivery via CD44. J Nanobiotechnol. 2017;15:7.

Muro S. Challenges in design and characterization of ligand-targeted drug delivery systems. J Control Release. 2012;164:125–37.

Kou L, Sun J, Zhai Y, He Z. The endocytosis and intracellular fate of nanomedicines: implication for rational design. Asian J Pharm Sci. 2013;8:1–10.

Li Z, Zhang Y, Zhu D, Li S, Yu X, Zhao Y, Ouyang X, Xie Z, Li L. Transporting carriers for intracellular targeting delivery via non-endocytic uptake pathways. Drug delivery. 2017;24:45–55.

Salatin S, Yari Khosroushahi A. Overviews on the cellular uptake mechanism of polysaccharide colloidal nanoparticles. J Cell Mol Med. 2017;21:1668–86.

Anirudhan TS, Nair AS. Temperature and ultrasound sensitive gatekeepers for the controlled release of chemotherapeutic drugs from mesoporous silica nanoparticles. J Mater Chem B. 2018;6:428–39.

Al-Ahmady Z, Kostarelos K. Chemical components for the design of temperature-responsive vesicles as cancer therapeutics. Chem Rev. 2016;116:3883–918.

Bai Y, Xie FY, Tian W. Controlled self-assembly of thermo-responsive amphiphilic h-shaped polymer for adjustable drug release. Chin J Polym Sci. 2018;36:406–16.

Zhang Z, Zhang D, Wei L, Wang X, Xu YL, Li HW, Ma M, Chen B, Xiao LH. Temperature responsive fluorescent polymer nanoparticles (TRFNPs) for cellular imaging and controlled releasing of drug to living cells. Coll Surf B. 2017;159:905–12.

Guo Y, Zhang Y, Ma J, Li Q, Li Y, Zhou X, Zhao D, Song H, Chen Q, Zhu X. Light/magnetic hyperthermia triggered drug released from multi-functional thermo-sensitive magnetoliposomes for precise cancer synergetic theranostics. J Control Release. 2017;272:145–58.

Hervault A, Thanh NT. Magnetic nanoparticle-based therapeutic agents for thermo-chemotherapy treatment of cancer. Nanoscale. 2014;6:11553–73.

Mathiyazhakan M, Wiraja C, Xu CJ: A Concise Review of Gold Nanoparticles-Based Photo-Responsive Liposomes for Controlled Drug Delivery. Nano-Micro Letters 2018, 10.

Xu L, Qiu LZ, Sheng Y, Sun YX, Deng LH, Li XQ, Bradley M, Zhang R. Biodegradable pH-responsive hydrogels for controlled dual-drug release. J Mater Chem B. 2018;6:510–7.

Ma GL, Lin WF, Yuan ZF, Wu J, Qian HF, Xua LB, Chen SF. Development of ionic strength/pH/enzyme triple-responsive zwitterionic hydrogel of the mixed l-glutamic acid and l-lysine polypeptide for site-specific drug delivery. J Mater Chem B. 2017;5:935–43.

Grillo R, Gallo J, Stroppa DG, Carbo-Argibay E, Lima R, Fraceto LF, Banobre-Lopez M. Sub-micrometer magnetic nanocomposites: insights into the effect of magnetic nanoparticles interactions on the optimization of SAR and MRI performance. Acs Appl Mater Interfaces. 2016;8:25777–87.

Alonso J, Khurshid H, Devkota J, Nemati Z, Khadka NK, Srikanth H, Pan JJ, Phan MH. Superparamagnetic nanoparticles encapsulated in lipid vesicles for advanced magnetic hyperthermia and biodetection. J Appl Phys. 2016;119:083904.

Ulbrich K, Hola K, Subr V, Bakandritsos A, Tucek J, Zboril R. Targeted drug delivery with polymers and magnetic nanoparticles: covalent and noncovalent approaches, release control, and clinical studies. Chem Rev. 2016;116:5338–431.

Chen CW, Syu WJ, Huang TC, Lee YC, Hsiao JK, Huang KY, Yu HP, Liao MY, Lai PS. Encapsulation of Au/Fe3O4 nanoparticles into a polymer nanoarchitecture with combined near infrared-triggered chemo-photothermal therapy based on intracellular secondary protein understanding. J Mater Chem B. 2017;5:5774–82.

Portero A, Remunan-Lopez C, Criado M, Alonso M. Reacetylated chitosan microspheres for controlled delivery of anti-microbial agents to the gastric mucosa. J Microencapsul. 2002;19:797–809.

Artursson P, Lindmark T, Davis SS, Illum L. Effect of chitosan on the permeability of monolayers of intestinal epithelial cells (Caco-2). Pharm Res. 1994;11:1358–61.

Fernández-Urrusuno R, Calvo P, Remuñán-López C, Vila-Jato JL, Alonso MJ. Enhancement of nasal absorption of insulin using chitosan nanoparticles. Pharm Res. 1999;16:1576–81.

De Campos AM, Sánchez A, Alonso MJ. Chitosan nanoparticles: a new vehicle for the improvement of the delivery of drugs to the ocular surface. Application to cyclosporin A. Int J Pharm. 2001;224:159–68.

Al-Qadi S, Grenha A, Carrión-Recio D, Seijo B, Remuñán-López C. Microencapsulated chitosan nanoparticles for pulmonary protein delivery: in vivo evaluation of insulin-loaded formulations. J Control Release. 2012;157:383–90.

Silva MM, Calado R, Marto J, Bettencourt A, Almeida AJ, Gonçalves L. Chitosan Nanoparticles as a mucoadhesive drug delivery system for ocular administration. Mar Drugs. 2017;15:370.

Pistone S, Goycoolea FM, Young A, Smistad G, Hiorth M. Formulation of polysaccharide-based nanoparticles for local administration into the oral cavity. Eur J Pharm Sci. 2017;96:381–9.

Liu S, Yang S, Ho PC. Intranasal administration of carbamazepine-loaded carboxymethyl chitosan nanoparticles for drug delivery to the brain. Asian J Pharm Sci. 2018;13:72–81.

Jain A, Jain SK. Optimization of chitosan nanoparticles for colon tumors using experimental design methodology. Artif Cells Nanomed Biotechnol. 2016;44:1917–26.

Sosnik A. Alginate particles as platform for drug delivery by the oral route: state-of-the-art. ISRN Pharm. 2014;2014:926157.

Patil NH, Devarajan PV. Insulin-loaded alginic acid nanoparticles for sublingual delivery. Drug Deliv. 2016;23:429–36.

Haque S, Md S, Sahni JK, Ali J, Baboota S. Development and evaluation of brain targeted intranasal alginate nanoparticles for treatment of depression. J Psychiatr Res. 2014;48:1–12.

Román JV, Galán MA, del Valle EMM. Preparation and preliminary evaluation of alginate crosslinked microcapsules as potential drug delivery system (DDS) for human lung cancer therapy. Biomed Phys Eng Expr. 2016;2:035015.

Garrait G, Beyssac E, Subirade M. Development of a novel drug delivery system: chitosan nanoparticles entrapped in alginate microparticles. J Microencapsul. 2014;31:363–72.

Costa J, Silva N, Sarmento B, Pintado M. Potential chitosan-coated alginate nanoparticles for ocular delivery of daptomycin. Eur J Clin Microbiol Infect Dis. 2015;34:1255–62.

Goswami S, Naik S. Natural gums and its pharmaceutical application. J Sci Innovative Res. 2014;3:112–21.

Laffleur F, Michalek M. Modified xanthan gum for buccal delivery—a promising approach in treating sialorrhea. Int J Biol Macromol. 2017;102:1250–6.

Huang J, Deng Y, Ren J, Chen G, Wang G, Wang F, Wu X. Novel in situ forming hydrogel based on xanthan and chitosan re-gelifying in liquids for local drug delivery. Carbohydr Polym. 2018;186:54–63.

Menzel C, Jelkmann M, Laffleur F, Bernkop-Schnürch A. Nasal drug delivery: design of a novel mucoadhesive and in situ gelling polymer. Int J Pharm. 2017;517:196–202.

Sun B, Zhang M, Shen J, He Z, Fatehi P, Ni Y. Applications of cellulose-based materials in sustained drug delivery systems. Curr Med Chem. 2017. https://doi.org/10.2174/0929867324666170705143308 .

Elseoud WSA, Hassan ML, Sabaa MW, Basha M, Hassan EA, Fadel SM. Chitosan nanoparticles/cellulose nanocrystals nanocomposites as a carrier system for the controlled release of repaglinide. Int J Biol Macromol. 2018;111:604–13.

Agarwal T, Narayana SGH, Pal K, Pramanik K, Giri S, Banerjee I. Calcium alginate-carboxymethyl cellulose beads for colon-targeted drug delivery. Int J Biol Macromol. 2015;75:409–17.

Hansen K, Kim G, Desai KG, Patel H, Olsen KF, Curtis-Fisk J, Tocce E, Jordan S, Schwendeman SP. Feasibility investigation of cellulose polymers for mucoadhesive nasal drug delivery applications. Mol Pharm. 2015;12:2732–41.

Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomed. 2015;10:975.

Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharm. 2015;6:286.

Kotla NG, Chandrasekar B, Rooney P, Sivaraman G, Larrañaga A, Krishna KV, Pandit A, Rochev Y. Biomimetic lipid-based nanosystems for enhanced dermal delivery of drugs and bioactive agents. ACS Biomater Sci Eng. 2017;3:1262–72.

Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, Samiei M, Kouhi M, Nejati-Koshki K. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8:102.

Mohan A, Narayanan S, Sethuraman S, Krishnan UM. Novel resveratrol and 5-fluorouracil coencapsulated in PEGylated nanoliposomes improve chemotherapeutic efficacy of combination against head and neck squamous cell carcinoma. BioMed res int. 2014;2014:424239.

Dimov N, Kastner E, Hussain M, Perrie Y, Szita N. Formation and purification of tailored liposomes for drug delivery using a module-based micro continuous-flow system. Sci Rep. 2017;7:12045.

Zylberberg C, Matosevic S. Pharmaceutical liposomal drug delivery: a review of new delivery systems and a look at the regulatory landscape. Drug Deliv. 2016;23:3319–29.

Sapsford KE, Algar WR, Berti L, Gemmill KB, Casey BJ, Oh E, Stewart MH, Medintz IL. Functionalizing nanoparticles with biological molecules: developing chemistries that facilitate nanotechnology. Chem Rev. 2013;113:1904–2074.

Zhang L, Gu F, Chan J, Wang A, Langer R, Farokhzad O. Nanoparticles in medicine: therapeutic applications and developments. Clin Pharmacol Ther. 2008;83:761–9.

Miyata K, Christie RJ, Kataoka K. Polymeric micelles for nano-scale drug delivery. React Funct Polym. 2011;71:227–34.

Xu W, Ling P, Zhang T. Polymeric micelles, a promising drug delivery system to enhance bioavailability of poorly water-soluble drugs. J Drug Deliv. 2013;2013:340315.

Kulthe SS, Choudhari YM, Inamdar NN, Mourya V. Polymeric micelles: authoritative aspects for drug delivery. Design Monomers Polym. 2012;15:465–521.

Devarajan PV, Jain S. Targeted drug delivery: concepts and design. Berlin: Springer; 2016.

Mourya V, Inamdar N, Nawale R, Kulthe S. Polymeric micelles: general considerations and their applications. Ind J Pharm Educ Res. 2011;45:128–38.

Wakaskar RR. Polymeric micelles for drug delivery. Int J Drug Dev Res. 2017;9:1–2.

Mandal A, Bisht R, Rupenthal ID, Mitra AK. Polymeric micelles for ocular drug delivery: from structural frameworks to recent preclinical studies. J Control Release. 2017;248:96–116.

Li Q, Lai KL, Chan PS, Leung SC, Li HY, Fang Y, To KK, Choi CHJ, Gao QY, Lee TW. Micellar delivery of dasatinib for the inhibition of pathologic cellular processes of the retinal pigment epithelium. Coll Surf B. 2016;140:278–86.

Kesharwani P, Xie L, Banerjee S, Mao G, Padhye S, Sarkar FH, Iyer AK. Hyaluronic acid-conjugated polyamidoamine dendrimers for targeted delivery of 3, 4-difluorobenzylidene curcumin to CD44 overexpressing pancreatic cancer cells. Coll Surf B. 2015;136:413–23.

Zhu J, Shi X. Dendrimer-based nanodevices for targeted drug delivery applications. J Mater Chem B. 2013;1:4199–211.

Madaan K, Kumar S, Poonia N, Lather V, Pandita D. Dendrimers in drug delivery and targeting: drug-dendrimer interactions and toxicity issues. J Pharm Bioallied Sci. 2014;6:139.

Cheng Y, Xu Z, Ma M, Xu T. Dendrimers as drug carriers: applications in different routes of drug administration. J Pharm Sci. 2008;97:123–43.

Noriega-Luna B, Godínez LA, Rodríguez FJ, Rodríguez A, Larrea G, Sosa-Ferreyra C, Mercado-Curiel R, Manríquez J, Bustos E. Applications of dendrimers in drug delivery agents, diagnosis, therapy, and detection. J Nanomater. 2014;2014:39.

Tripathy S, Das M. Dendrimers and their applications as novel drug delivery carriers. J Appl Pharm Sci. 2013;3:142–9.

Kesharwani P, Jain K, Jain NK. Dendrimer as nanocarrier for drug delivery. Progr Polym Sci. 2014;39:268–307.

Jain K, Gupta U, Jain NK. Dendronized nanoconjugates of lysine and folate for treatment of cancer. Eur J Pharm Biopharm. 2014;87:500–9.

Kaur A, Jain K, Mehra NK, Jain N. Development and characterization of surface engineered PPI dendrimers for targeted drug delivery. Artif Cells Nanomed Biotechnol. 2017;45:414–25.

Choi S-J, Lee JK, Jeong J, Choy J-H. Toxicity evaluation of inorganic nanoparticles: considerations and challenges. Mol Cell Toxicol. 2013;9:205–10.

Kong F-Y, Zhang J-W, Li R-F, Wang Z-X, Wang W-J, Wang W. Unique roles of gold nanoparticles in drug delivery, targeting and imaging applications. Molecules. 2017;22:1445.

Prusty K, Swain SK. Nano silver decorated polyacrylamide/dextran nanohydrogels hybrid composites for drug delivery applications. Mater Sci Eng. 2018;85:130–41.

Marcu A, Pop S, Dumitrache F, Mocanu M, Niculite C, Gherghiceanu M, Lungu C, Fleaca C, Ianchis R, Barbut A. Magnetic iron oxide nanoparticles as drug delivery system in breast cancer. Appl Surf Sci. 2013;281:60–5.

Junyaprasert VB, Morakul B. Nanocrystals for enhancement of oral bioavailability of poorly water-soluble drugs. Asian J Pharm Sci. 2015;10:13–23.

Du J, Li X, Zhao H, Zhou Y, Wang L, Tian S, Wang Y. Nanosuspensions of poorly water-soluble drugs prepared by bottom-up technologies. Int J Pharm. 2015;495:738–49.

Ni R, Zhao J, Liu Q, Liang Z, Muenster U, Mao S. Nanocrystals embedded in chitosan-based respirable swellable microparticles as dry powder for sustained pulmonary drug delivery. Eur J Pharm Sci. 2017;99:137–46.

McNamara K, Tofail SA. Nanoparticles in biomedical applications. Adv Phys. 2017;2:54–88.

Kudr J, Haddad Y, Richtera L, Heger Z, Cernak M, Adam V, Zitka O. Magnetic nanoparticles: from design and synthesis to real world applications. Nanomaterials. 2017;7:243.

Prasad PN. Nanophotonics. New York: Wiley; 2004.

Volkov Y. Quantum dots in nanomedicine: recent trends, advances and unresolved issues. Biochem Biophys Res Commun. 2015;468:419–27.

Liu J, Lau SK, Varma VA, Moffitt RA, Caldwell M, Liu T, Young AN, Petros JA, Osunkoya AO, Krogstad T. Molecular mapping of tumor heterogeneity on clinical tissue specimens with multiplexed quantum dots. ACS Nano. 2010;4:2755–65.

Xu G, Zeng S, Zhang B, Swihart MT, Yong K-T, Prasad PN. New generation cadmium-free quantum dots for biophotonics and nanomedicine. Chem Rev. 2016;116:12234–327.

Shi Y, Pramanik A, Tchounwou C, Pedraza F, Crouch RA, Chavva SR, Vangara A, Sinha SS, Jones S, Sardar D. Multifunctional biocompatible graphene oxide quantum dots decorated magnetic nanoplatform for efficient capture and two-photon imaging of rare tumor cells. ACS Appl Mater Interfaces. 2015;7:10935–43.

Han H-S, Niemeyer E, Huang Y, Kamoun WS, Martin JD, Bhaumik J, Chen Y, Roberge S, Cui J, Martin MR. Quantum dot/antibody conjugates for in vivo cytometric imaging in mice. Proc Natl Acad Sci. 2015;112:1350–5.

So M-K, Xu C, Loening AM, Gambhir SS, Rao J. Self-illuminating quantum dot conjugates for in vivo imaging. Nat Biotechnol. 2006;24:339.

Zheng F-F, Zhang P-H, Xi Y, Chen J-J, Li L-L, Zhu J-J. Aptamer/graphene quantum dots nanocomposite capped fluorescent mesoporous silica nanoparticles for intracellular drug delivery and real-time monitoring of drug release. Anal Chem. 2015;87:11739–45.

Huang C-L, Huang C-C, Mai F-D, Yen C-L, Tzing S-H, Hsieh H-T, Ling Y-C, Chang J-Y. Application of paramagnetic graphene quantum dots as a platform for simultaneous dual-modality bioimaging and tumor-targeted drug delivery. J Mater Chem B. 2015;3:651–64.

Olerile LD, Liu Y, Zhang B, Wang T, Mu S, Zhang J, Selotlegeng L, Zhang N. Near-infrared mediated quantum dots and paclitaxel co-loaded nanostructured lipid carriers for cancer theragnostic. Coll Surf B. 2017;150:121–30.

Cai X, Luo Y, Zhang W, Du D, Lin Y. pH-Sensitive ZnO quantum dots–doxorubicin nanoparticles for lung cancer targeted drug delivery. ACS Appl Mater Interfaces. 2016;8:22442–50.

Balaji AB, Pakalapati H, Khalid M, Walvekar R, Siddiqui H. Natural and synthetic biocompatible and biodegradable polymers. In: Shimpi NG (ed) Biodegradable and biocompatible polymer composites: processing, properties and applications. Woodhead Publishing series in composites science and engineering. Duxford: Woodhead Publishing; 2017. p. 3–32.

Bassas-Galia M, Follonier S, Pusnik M, Zinn M. Natural polymers: a source of inspiration. In: Bioresorbable polymers for biomedical applications. New York: Elsevier; 2017. p. 31–64.

Lohcharoenkal W, Wang L, Chen YC, Rojanasakul Y. Protein nanoparticles as drug delivery carriers for cancer therapy. BioMed Res Int. 2014;2014:180549.

Liu Z, Jiao Y, Wang Y, Zhou C, Zhang Z. Polysaccharides-based nanoparticles as drug delivery systems. Adv Drug Deliv Rev. 2008;60:1650–62.

Poole-Warren L, Patton A. Introduction to biomedical polymers and biocompatibility. In: Biosynthetic polymers for medical applications. New York: Elsevier; 2016. p. 3–31.

Pertici G. Introduction to bioresorbable polymers for biomedical applications. In: Biosynthetic polymers for medical applications. New York: Elsevier; 2016. p. 3–29.

Cardoso MJ, Costa RR, Mano JF. Marine origin polysaccharides in drug delivery systems. Mar Drugs. 2016;14:34.

Yu Z, Yu M, Zhang Z, Hong G, Xiong Q. Bovine serum albumin nanoparticles as controlled release carrier for local drug delivery to the inner ear. Nanoscale Res Lett. 2014;9:343.

Robinson M, Zhang X. The world medicines situation. Traditional medicines: global situation, issues and challenges. Geneva: World Health Organization; 2011. p. 1–12.

Atanasov AG, Waltenberger B, Pferschy-Wenzig E-M, Linder T, Wawrosch C, Uhrin P, Temml V, Wang L, Schwaiger S, Heiss EH. Discovery and resupply of pharmacologically active plant-derived natural products: a review. Biotechnol Adv. 2015;33:1582–614.

David B, Wolfender J-L, Dias DA. The pharmaceutical industry and natural products: historical status and new trends. Phytochem Rev. 2015;14:299–315.

Namdari M, Eatemadi A, Soleimaninejad M, Hammed AT. A brief review on the application of nanoparticle enclosed herbal medicine for the treatment of infective endocarditis. Biomed Pharm. 2017;87:321–31.

Heinrich M. Ethnopharmacology in the 21st century-grand challenges. Front Pharm. 2010;1:8.

Kinghorn AD, Pan L, Fletcher JN, Chai H. The relevance of higher plants in lead compound discovery programs. J Nat Prod. 2011;74:1539–55.

Yuan H, Ma Q, Ye L, Piao G. The traditional medicine and modern medicine from natural products. Molecules. 2016;21:559.

Patra JK, Das G, Baek K-H. Towards a greener environment: synthesis and applications of green nanoparticles. Pak J Agric Sci. 2016;53:59–79.

Duncan R, Gaspar R. Nanomedicine (s) under the microscope. Mol Pharm. 2011;8:2101–41.

Ramana KV, Singhal SS, Reddy AB. Therapeutic potential of natural pharmacological agents in the treatment of human diseases. BioMed Res Int. 2014;2014:573452.

Guo W. Green technology for nanoparticles in biomedical applications. In: Rai M, Posten C, editors. Green biosynthesis of nanoparticles: mechanisms and applications. Wallington: CABI; 2013.

Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release. 2015;200:138–57.

Brigger I, Dubernet C, Couvreur P. Nanoparticles in cancer therapy and diagnosis. Adv Drug Deliv Rev. 2002;54:631–51.

Yohan D, Chithrani BD. Applications of nanoparticles in nanomedicine. J Biomed Nanotechnol. 2014;10:2371–92.

Ambesh P, Campia U, Obiagwu C, Bansal R, Shetty V, Hollander G, Shani J. Nanomedicine in coronary artery disease. Indian Heart J. 2017;69:244–51.

Grazu V, Moros M, Sánchez-Espinel C. Nanocarriers as nanomedicines: design concepts and recent advances. In: Frontiers of nanoscience. Vol. 4, New York: Elsevier; 2012. p. 337–440.

Rizzo LY, Theek B, Storm G, Kiessling F, Lammers T. Recent progress in nanomedicine: therapeutic, diagnostic and theranostic applications. Curr Opin Biotechnol. 2013;24:1159–66.

Devasena T. Diagnostic and therapeutic nanomaterials. In: Therapeutic and diagnostic nanomaterials. New York: Springer; 2017. p. 1–13.

Ventola CL. Progress in nanomedicine: approved and investigational nanodrugs. Pharm Ther. 2017;42:742.

Havel H, Finch G, Strode P, Wolfgang M, Zale S, Bobe I, Youssoufian H, Peterson M, Liu M. Nanomedicines: from bench to bedside and beyond. AAPS J. 2016;18:1373–8.

Kumar A, Chen F, Mozhi A, Zhang X, Zhao Y, Xue X, Hao Y, Zhang X, Wang PC, Liang X-J. Innovative pharmaceutical development based on unique properties of nanoscale delivery formulation. Nanoscale. 2013;5:8307–25.

Boroumand Moghaddam A, Namvar F, Moniri M, Md Tahir P, Azizi S, Mohamad R. Nanoparticles biosynthesized by fungi and yeast: a review of their preparation, properties, and medical applications. Molecules. 2015;20:16540–65.

Metz KM, Sanders SE, Pender JP, Dix MR, Hinds DT, Quinn SJ, Ward AD, Duffy P, Cullen RJ, Colavita PE. Green synthesis of metal nanoparticles via natural extracts: the biogenic nanoparticle corona and its effects on reactivity. ACS Sustain Chem Eng. 2015;3:1610–7.

Paul D, Sinha SN. Extracellular synthesis of silver nanoparticles using Pseudomonas aeruginosa KUPSB12 and its antibacterial activity. JJBS. 2014;7:245–50.

Kushwaha A, Singh VK, Bhartariya J, Singh P, Yasmeen K. Isolation and identification of E. coli bacteria for the synthesis of silver nanoparticles: characterization of the particles and study of antibacterial activity. Eur J Exp Biol. 2015;5:65–70.

Iravani S. Bacteria in nanoparticle synthesis: current status and future prospects. Int Sch Res Notices. 2014;2014:359316.

Mittal AK, Chisti Y, Banerjee UC. Synthesis of metallic nanoparticles using plant extracts. Biotechnol Adv. 2013;31:346–56.

Khan HA, Sakharkar MK, Nayak A, Kishore U, Khan A. 14-nanoparticles for biomedical applications: an overview. In: Narayan R, editor. Nanobiomaterials. Cambridge: Woodhead Publishing; 2018. p. 357–84.

Aravamudhan A, Ramos DM, Nada AA, Kumbar SG. Natural polymers: polysaccharides and their derivatives for biomedical applications. In: Natural and synthetic biomedical polymers. New York: Elsevier; 2014. p. 67–89.

Franci G, Falanga A, Galdiero S, Palomba L, Rai M, Morelli G, Galdiero M. Silver nanoparticles as potential antibacterial agents. Molecules. 2015;20:8856–74.

Pajardi G, Rapisarda V, Somalvico F, Scotti A, Russo GL, Ciancio F, Sgrò A, Nebuloni M, Allevi R, Torre ML. Skin substitutes based on allogenic fibroblasts or keratinocytes for chronic wounds not responding to conventional therapy: a retrospective observational study. Int Wound J. 2016;13:44–52.

Rahimi G, Alizadeh F, Khodavandi A. Mycosynthesis of silver nanoparticles from Candida albicans and its antibacterial activity against Escherichia coli and Staphylococcus aureus. Trop J Pharm Res. 2016;15:371–5.

Ali M, Kim B, Belfield KD, Norman D, Brennan M, Ali GS. Inhibition of Phytophthora parasitica and P. capsici by silver nanoparticles synthesized using aqueous extract of Artemisia absinthium. Phytopathology. 2015;105:1183–90.

Malapermal V, Botha I, Krishna SBN, Mbatha JN. Enhancing antidiabetic and antimicrobial performance of Ocimum basilicum, and Ocimum sanctum (L.) using silver nanoparticles. Saudi J Biol Sci. 2017;24:1294–305.

Sankar R, Karthik A, Prabu A, Karthik S, Shivashangari KS, Ravikumar V. Origanum vulgare mediated biosynthesis of silver nanoparticles for its antibacterial and anticancer activity. Coll Surf B. 2013;108:80–4.

Patra JK, Ali MS, Oh I-G, Baek K-H. Proteasome inhibitory, antioxidant, and synergistic antibacterial and anticandidal activity of green biosynthesized magnetic Fe3O4 nanoparticles using the aqueous extract of corn (Zea mays L.) ear leaves. Artif Cells Nanomed Biotechnol. 2017;45:349–56.

Patra JK, Baek K-H. Antibacterial activity and synergistic antibacterial potential of biosynthesized silver nanoparticles against foodborne pathogenic bacteria along with its anticandidal and antioxidant effects. Front Microbiol. 2017;8:167.

Patra JK, Kwon Y, Baek K-H. Green biosynthesis of gold nanoparticles by onion peel extract: synthesis, characterization and biological activities. Adv Powder Technol. 2016;27:2204–13.

Patra JK, Baek K-H. Biosynthesis of silver nanoparticles using aqueous extract of silky hairs of corn and investigation of its antibacterial and anticandidal synergistic activity and antioxidant potential. IET Nanobiotechnol. 2016;10:326–33.

Patra JK, Baek K-H. Comparative study of proteasome inhibitory, synergistic antibacterial, synergistic anticandidal, and antioxidant activities of gold nanoparticles biosynthesized using fruit waste materials. Int J Nanomed. 2016;11:4691.

Patra JK, Baek K-H. Green synthesis of silver chloride nanoparticles using Prunus persica L. outer peel extract and investigation of antibacterial, anticandidal, antioxidant potential. Green Chem Lett Rev. 2016;9:132–42.

Patra JK, Das G, Baek K-H. Phyto-mediated biosynthesis of silver nanoparticles using the rind extract of watermelon (Citrullus lanatus) under photo-catalyzed condition and investigation of its antibacterial, anticandidal and antioxidant efficacy. J Photochem Photobiol B. 2016;161:200–10.

Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery systems. Pharmacol Rep. 2012;64:1020–37.

Zhu Z, Li Y, Yang X, Pan W, Pan H. The reversion of anti-cancer drug antagonism of tamoxifen and docetaxel by the hyaluronic acid-decorated polymeric nanoparticles. Pharmacol Res. 2017;126:84–96.

Dias DA, Urban S, Roessner U. A historical overview of natural products in drug discovery. Metabolites. 2012;2:303–36.

Gupta U, Sharma S, Khan I, Gothwal A, Sharma AK, Singh Y, Chourasia MK, Kumar V. Enhanced apoptotic and anticancer potential of paclitaxel loaded biodegradable nanoparticles based on chitosan. Int J Biol Macromol. 2017;98:810–9.

Chang C-H, Huang W-Y, Lai C-H, Hsu Y-M, Yao Y-H, Chen T-Y, Wu J-Y, Peng S-F, Lin Y-H. Development of novel nanoparticles shelled with heparin for berberine delivery to treat Helicobacter pylori. Acta Biomaterialia. 2011;7:593–603.

Aldawsari HM, Hosny KM. Solid lipid nanoparticles of Vancomycin loaded with Ellagic acid as a tool for overcoming nephrotoxic side effects: preparation, characterization, and nephrotoxicity evaluation. J Drug Deliv Sci Technol. 2018;45:76–80.

Dian L, Yu E, Chen X, Wen X, Zhang Z, Qin L, Wang Q, Li G, Wu C. Enhancing oral bioavailability of quercetin using novel soluplus polymeric micelles. Nanoscale Res Lett. 2014;9:684.

Spillmann CM, Naciri J, Algar WR, Medintz IL, Delehanty JB. Multifunctional liquid crystal nanoparticles for intracellular fluorescent imaging and drug delivery. ACS Nano. 2014;8:6986–97.

Purama RK, Goswami P, Khan AT, Goyal A. Structural analysis and properties of dextran produced by Leuconostoc mesenteroides NRRL B-640. Carbohydr Polym. 2009;76:30–5.

Agarwal A, Gupta U, Asthana A, Jain NK. Dextran conjugated dendritic nanoconstructs as potential vectors for anti-cancer agent. Biomaterials. 2009;30:3588–96.

Barenholz YC. Doxil®—the first FDA-approved nano-drug: lessons learned. J Control Release. 2012;160:117–34.

Maeng JH, Lee D-H, Jung KH, Bae Y-H, Park I-S, Jeong S, Jeon Y-S, Shim C-K, Kim W, Kim J. Multifunctional doxorubicin loaded superparamagnetic iron oxide nanoparticles for chemotherapy and magnetic resonance imaging in liver cancer. Biomaterials. 2010;31:4995–5006.

Bonechi C, Martini S, Ciani L, Lamponi S, Rebmann H, Rossi C, Ristori S. Using liposomes as carriers for polyphenolic compounds: the case of trans-resveratrol. PLoS ONE. 2012;7:e41438.

Noorafshan A, Ashkani-Esfahani S. A review of therapeutic effects of curcumin. Curr Pharm Des. 2013;19:2032–46.

Wei X, Senanayake TH, Bohling A, Vinogradov SV. Targeted nanogel conjugate for improved stability and cellular permeability of curcumin: synthesis, pharmacokinetics, and tumor growth inhibition. Mol Pharm. 2014;11:3112–22.

Feng T, Wei Y, Lee RJ, Zhao L. Liposomal curcumin and its application in cancer. Int J Nanomed. 2017;12:6027.

Cheng C, Peng S, Li Z, Zou L, Liu W, Liu C. Improved bioavailability of curcumin in liposomes prepared using a pH-driven, organic solvent-free, easily scalable process. RSC Adv. 2017;7:25978–86.

Bilia AR, Guccione C, Isacchi B, Righeschi C, Firenzuoli F, Bergonzi MC. Essential oils loaded in nanosystems: a developing strategy for a successful therapeutic approach. Evid Based Complement Alternat Med. 2014;2014:651593.

Sainz V, Conniot J, Matos AI, Peres C, Zupanǒiǒ E, Moura L, Silva LC, Florindo HF, Gaspar RS. Regulatory aspects on nanomedicines. Biochem Biophys Res Commun. 2015;468:504–10.

Hassan S, Prakash G, Ozturk AB, Saghazadeh S, Sohail MF, Seo J, Dokmeci MR, Zhang YS, Khademhosseini A. Evolution and clinical translation of drug delivery nanomaterials. Nano Today. 2017;15:91–106.

Agrahari V, Agrahari V. Facilitating the translation of nanomedicines to a clinical product: challenges and opportunities. Drug Discov Today. 2018;23(5):974–91.

Caster JM, Patel AN, Zhang T, Wang A. Investigational nanomedicines in 2016: a review of nanotherapeutics currently undergoing clinical trials. Wiley Interdiscip Rev. 2016;2017:9.

Wacker MG, Proykova A, Santos GML. Dealing with nanosafety around the globe—regulation vs. innovation. Int J Pharm. 2016;509:95–106.

Lin P-C, Lin S, Wang PC, Sridhar R. Techniques for physicochemical characterization of nanomaterials. Biotechnol Adv. 2014;32:711–26.

Grossman JH, Crist RM, Clogston JD. Early development challenges for drug products containing nanomaterials. AAPS J. 2017;19:92–102.

Tinkle S, McNeil SE, Mühlebach S, Bawa R, Borchard G, Barenholz YC, Tamarkin L, Desai N. Nanomedicines: addressing the scientific and regulatory gap. Ann NY Acad Sci. 2014;1313:35–56.

Pandit A, Zeugolis DI. Twenty-five years of nano-bio-materials: have we revolutionized healthcare? Fut Med. 2016;11(9):985–7.

Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date. Pharm Res. 2016;33:2373–87.

Tran S, DeGiovanni P-J, Piel B, Rai P. Cancer nanomedicine: a review of recent success in drug delivery. Clin Transl Med. 2017;6:44.

Anselmo AC, Mitragotri S. Nanoparticles in the clinic. Bioeng Transl Med. 2016;1:10–29.

Grumezescu AM. Nanoscale fabrication, optimization, scale-up and biological aspects of pharmaceutical nanotechnology. New York: William Andrew; 2017.

Caster JM, Patel AN, Zhang T, Wang A. Investigational nanomedicines in 2016: a review of nanotherapeutics currently undergoing clinical trials. Wiley Interdiscip Rev. 2017;9:e1416.

Drug approvals and databases. https://www.fda.gov/Drugs/InformationOnDrugs/default.htm . Accessed 16 Aug 2018.

D’Mello SR, Cruz CN, Chen M-L, Kapoor M, Lee SL, Tyner KM. The evolving landscape of drug products containing nanomaterials in the United States. Nat Nanotechnol. 2017;12:523.