N. A. Alhaj, R. Abdullah, S. Ibrahim, and A. Bustamam, Tamoxifen drug loading solid lipid nanoparticles prepared by hot high pressure homogenization techniques, Am. J. Pharmacol. Toxicol, vol.3, pp.219-224, 2008.

K. Adibkia, M. R. Siahi-shadbad, A. Nokhodchi, and Y. Omidi, Piroxicam nanoparticles for ocular delivery: physicochemical characterization and implementation in endotoxin-induced uveitis, J. Drug Target, vol.15, issue.6, pp.407-416, 2007.

K. Adibkia, Y. Javadzadeh, S. Dastmalchi, G. Mohammadi, F. K. Niri et al., Naproxen-eudragit RS100 nanoparticles: preparation and physicochemical characterization, Colloids Surf. B Biointerfaces, vol.83, pp.155-159, 2011.

H. Ahmad, K. Kumar, M. A. Rahman, M. M. Rahman, M. A. Miah et al., Preparation and characterization of conducting polyaniline layered magnetic nano composite polymer particles, Polym. Adv. Technol, vol.24, pp.740-746, 2013.

J. Araújo, E. Gonzalez, M. A. Egea, M. L. Garcia, and E. B. Souto, Nanomedicines for ocular NSAIDs: safety on drug delivery, Nanomed. Nanotechnol. Biol. Med, vol.5, pp.394-401, 2009.

A. I. Arida and M. M. Al-tabakha, Encapsulation of ketoprofen for controlled drug release, Eur. J. Pharm. Biopharm, vol.66, issue.1, pp.48-54, 2007.

A. A. Attama, S. Reichl, and C. C. Müller-goymann, Diclofenac sodium delivery to the eye: in vitro evaluation of novel solid lipid nanoparticle formulation using human cornea construct, Int. J. Pharm, vol.355, pp.307-313, 2008.

A. Avachat, P. Bornare, and R. Dash, Sustained release microspheres of ropinirole hydrochloride: effect of process parameters, Acta Pharm, vol.61, pp.363-376, 2011.

A. A. Badawi, H. M. El-laithy, R. K. Qidra, H. E. Mofty, and M. E. Dally, Chitosan based nanocarriers for indomethacin ocular delivery, Arch. Pharm. Res, vol.31, pp.1040-1049, 2008.

W. Badri, K. Miladi, R. Eddabra, H. Fessi, and A. Elaissari, Elaboration of nanoparticles containing indomethacin: argan oil for transdermal local and cosmetic application, J. Nanomater, p.935439, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02092666

G. Barratt, Colloidal drug carriers: achievements and perspectives, CMLS, vol.60, pp.21-37, 2003.

S. Barua and S. Mitragotri, Challenges associated with penetration of nanoparticles across cell and tissue barriers: a review of current status and future prospects, Nano Today, vol.9, pp.223-243, 2014.

D. N. Bateman, Non-steroidal anti-inflammatory drugs, p.140, 2012.

S. Batzri and E. D. Korn, Single bilayer liposomes prepared without sonication, Biochim. Biophys. Acta, vol.298, pp.1015-1019, 1973.

B. Beetge, J. Du-plessis, and D. G. Müller, The influence of the physiochemical characteristics and pharmacokinetics properties of selected NSAID's on their transdermal absorption, Int. J. Pharm, vol.193, pp.261-264, 2000.

M. Beija, R. Salvayre, N. Lauth-de-viguerie, and J. Marty, Colloidal systems for drug delivery: from design to therapy, Trends Biotechnol, vol.30, pp.485-496, 2012.

K. Bhaskar, J. Anbu, V. Ravichandiran, V. Venkateswarlu, and Y. M. Rao, Lipid nanoparticles for transdermal delivery of flurbiprofen: formulation, in vitro, ex vivo and in vivo studies, Lipids Health Dis, vol.8, issue.6, 2009.

A. Budhian, S. J. Siegel, and K. I. Winey, Haloperidol-loaded PLGA nanoparticles: systematic study of particle size and drug content, Int. J. Pharm, vol.336, pp.367-375, 2007.

M. Burian and G. Geisslinger, COX-dependent mechanisms involved in the antinociceptive action of NSAIDs at central and peripheral sites, Pharmacol. Ther, vol.107, issue.2, pp.139-154, 2005.

Y. Ç?rpanl?, C. Robineau, Y. Çapan, and S. Çal??, Etodolac loaded poly (lactide-coglycolide) nanoparticles: formulation and in vitro characterization, Hacettepe Univ. J. Faculty Pharm, vol.29, issue.2, pp.105-114, 2009.

F. Castelli, C. Puglia, M. G. Sarpietro, L. Rizza, and F. Bonina, Characterization of indomethacin-loaded lipid nanoparticles by differential scanning calorimetry, Int. J. Pharm, vol.304, pp.231-238, 2005.
DOI : 10.1016/j.ijpharm.2005.08.011

E. Cenni, D. Granchi, S. Avnet, C. Fotia, M. Salerno et al., Biocompatibility of poly(D,L-lactideco-glycolide) nanoparticles conjugated with alendronate, Biomaterials, vol.29, pp.1400-1411, 2008.
DOI : 10.1016/j.biomaterials.2007.12.022

M. Cetin, A. Atila, and Y. Kadioglu, Formulation and In vitro characterization of Eudragit 1 L100 and Eudragit 1 L100-PLGA nanoparticles containing diclofenac sodium, AAPS PharmSciTech, vol.11, pp.1250-1256, 2010.
DOI : 10.1208/s12249-010-9489-6

URL : http://europepmc.org/articles/pmc2974120?pdf=render

C. Charcosset and H. Fessi, A new process for drug loaded nanocapsules preparation using a membrane contactor, Drug Dev. Ind. Pharm, vol.31, pp.987-992, 2005.
DOI : 10.1080/03639040500306237

L. Chen and D. M. Ashcroft, Risk of myocardial infarction associated with selective COX-2 inhibitors: meta-analysis of randomised controlled trials, Pharmacoepidemiol. Drug Saf, vol.16, pp.762-772, 2007.

P. C. Chen, J. Huang, J. Pang, P. C. Chen, J. Huang et al., An investigation of optimum NLC-sunscreen formulation using Taguchi analysis, J. Nanomater, vol.9, 2013.
DOI : 10.1155/2013/463732

URL : http://doi.org/10.1155/2013/463732

M. Chorny, I. Fishbein, H. D. Danenberg, and G. Golomb, Lipophilic drug loaded nanospheres prepared by nanoprecipitation: effect of formulation variables on size, drug recovery and release kinetics, J. Control. Release, vol.83, pp.389-400, 2002.
DOI : 10.1016/s0168-3659(02)00211-0

B. L. Chung, M. J. Toth, N. Kamaly, Y. J. Sei, J. Becraft et al., Nanomedicines for endothelial disorders, Nano Today, vol.10, pp.759-776, 2015.

D. 'aurizio, E. Van-nostrum, C. F. Van-steenbergen, M. J. Sozio, P. Siepmann et al., Preparation and characterization of poly(lactic-co-glycolic acid) microspheres loaded with a labile antiparkinson prodrug, Int. J. Pharm, vol.409, pp.289-296, 2011.

M. M. De-villiers, D. P. Otto, S. J. Strydom, and Y. M. Lvov, Introduction to nanocoatings produced by layer-by-layer (LbL) self-assembly, Adv. Drug Deliv. Rev, vol.63, pp.701-715, 2011.

A. Des-rieux, V. Fievez, M. Garinot, Y. Schneider, and V. Préat, Nanoparticles as potential oral delivery systems of proteins and vaccines: a mechanistic approach, J. Control. Release, vol.116, pp.1-27, 2006.

A. Deniz, A. Sade, F. Severcan, D. Keskin, A. Tezcaner et al., Celecoxibloaded liposomes: effect of cholesterol on encapsulation and in vitro release characteristics, Biosci. Rep, vol.30, pp.365-373, 2010.

I. Dey, M. Lejeune, and K. Chadee, Prostaglandin E2 receptor distribution and function in the gastrointestinal tract, Br. J. Pharmacol, vol.149, pp.611-623, 2006.
DOI : 10.1038/sj.bjp.0706923

URL : http://onlinelibrary.wiley.com/doi/10.1038/sj.bjp.0706923/pdf

F. U. Din, O. Mustapha, D. W. Kim, R. Rashid, J. H. Park et al., Novel dual-reverse thermosensitive solid lipid nanoparticle-loaded hydrogel for rectal administration of flurbiprofen with improved bioavailability and reduced initial burst effect, Eur. J. Pharm. Biopharm, vol.94, pp.64-72, 2015.
DOI : 10.1016/j.ejpb.2015.04.019

T. V. Doan, W. Couet, and J. C. Olivier, Formulation and in vitro characterization of inhalable rifampicin-loaded PLGA microspheres for sustained lung delivery, Int. J. Pharm, vol.414, pp.112-117, 2011.

. Silva, . Cintra, D. Oliveira, L. L. Estevanato, A. R. Simioni et al., Successful strategy for targeting the central nervous system using magnetic albumin nanospheres, J. Biomed. Nanotechnol, vol.8, pp.182-189, 2012.

D. C. Drummond, C. O. Noble, M. E. Hayes, J. W. Park, and D. B. Kirpotin, Pharmacokinetics and in vivo drug release rates in liposomal nanocarrier development, J. Pharm. Sci, vol.97, pp.4696-4740, 2008.
DOI : 10.1002/jps.21358

P. Ejaz, K. Bhojani, and V. R. Joshi, NSAIDs and kidney, J. Assoc. Phys. India, vol.52, pp.632-640, 2004.

M. L. Elsabahy and K. Wooleya, Design of polymeric nanoparticles for biomedical delivery applications, Chem. Soc. Rev, vol.41, issue.7, pp.2545-2561, 2012.

H. Ferreira, T. Matamá, R. Silva, C. Silva, A. C. Gomes et al., Functionalization of gauzes with liposomes entrapping an anti-inflammatory drug: a strategy to improve wound healing, React. Funct. Polym, vol.73, pp.1328-1334, 2013.

T. Fujisawa, H. Miyai, K. Hironaka, T. Tsukamoto, K. Tahara et al., Liposomal diclofenac eye drop formulations targeting the retina: formulation stability improvement using surface modification of liposomes, Int. J. Pharm, vol.436, pp.564-567, 2012.

M. Gagliardi, G. Bardi, and A. Bifone, Polymeric nanocarriers for controlled and enhanced delivery of therapeutic agents to the CNS, Ther. Deliv, vol.3, pp.875-887, 2012.

S. A. Galindo-rodriguez, E. Allemann, H. Fessi, and E. Doelker, Polymeric nanoparticles for oral delivery of drugs and vaccines: a critical evaluation of in vivo studies, Crit. Rev. Ther. Drug Carrier Syst, vol.22, pp.419-464, 2005.

N. Garti, Double emulsions-scope, limitations and new achievements, Colloids Surf. Physicochem. Eng. Aspects, vol.123, pp.233-246, 1997.

N. S. Gerstein, W. H. Gerstein, M. C. Carey, N. C. Kong-lam, H. Ram et al., The thrombotic and arrhythmogenic risks of perioperative NSAIDs, J. Cardiothorac. Vasc. Anesth, vol.28, pp.369-378, 2014.

T. K. Giri, C. Choudhary, A. Alexander, H. Badwaik, and D. K. Tripathi, Prospects of pharmaceuticals and biopharmaceuticals loaded microparticles prepared by double emulsion technique for controlled delivery, Saudi Pharm. J, vol.21, pp.125-141, 2013.

E. Gonzalez-mira, M. A. Egea, E. B. Souto, A. C. Calpena, and M. L. García, Optimizing flurbiprofen-loaded NLC by central composite factorial design for ocular delivery, Nanotechnology, vol.22, p.45101, 2011.

F. X. Gu, R. Karnik, A. Z. Wang, A. , F. Levy-nissenbaum et al., Targeted nanoparticles for cancer therapy, Nano Today, vol.2, pp.14-21, 2007.

S. Guinebretiere, Nanocapsules par émulsion-diffusion de solvant: obtention, caractérisation et mécanisme de formation, 2001.

R. Gupta, P. Yadav, and S. A. Saraf, Polyelectrolyte multilayer assembly bearing ketoprofen for transdermal delivery, Nat. Preced. Int. Con. Bioencap XVII, pp.24-26, 2009.

F. Han, R. Yin, X. Che, J. Yuan, Y. Cui et al., Nanostructured lipid carriers (NLC) based topical gel of flurbiprofen: design, characterization and in vivo evaluation, Int. J. Pharm, vol.439, pp.349-357, 2012.

C. Heneweer, S. E. Gendy, and O. Peñate-medina, Liposomes and inorganic nanoparticles for drug delivery and cancer imaging, Ther. Deliv, vol.3, pp.645-656, 2012.

E. P. Herrero, M. J. Alonso, and N. Csaba, Polymer-based oral peptide nanomedicines, Ther. Deliv, vol.3, pp.657-668, 2012.

K. Hippalgaonkar, G. R. Adelli, K. Hippalgaonkar, M. A. Repka, and S. Majumdar, Indomethacin-loaded solid lipid nanoparticles for ocular delivery: development, characterization, and in vitro evaluation, J. Ocul. Pharmacol. Ther, vol.29, pp.216-228, 2013.

S. Hornig, H. Bunjes, and T. Heinze, Preparation and characterization of nanoparticles based on dextran-drug conjugates, J. Colloid Interface Sci, vol.338, pp.56-62, 2009.

C. Huerta, M. Del-rosario-aberturas, and J. Molpeceres, Nimesulide-loaded nanoparticles for the potential coadjuvant treatment of prostate cancer, Int. J. Pharm, vol.493, pp.152-160, 2015.

M. M. Ibrahim, A. H. Abd-elgawad, O. A. Soliman, and M. M. Jablonski, Nanoparticle-based topical ophthalmic formulations for sustained celecoxib release, J. Pharm. Sci, vol.102, pp.1036-1053, 2013.

M. Iqbal, N. Zafar, H. Fessi, and A. Elaissari, Double emulsion solvent evaporation techniques used for drug encapsulation, Int. J. Pharm, vol.496, pp.173-190, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01958031

R. Jones, Nonsteroidal anti-inflammatory drug prescribing: past, present, and future, Am. J. Med, vol.110, pp.4-7, 2001.

M. Joshi and V. Patravale, Nanostructured lipid carrier (NLC) based gel of celecoxib, Int. J. Pharm, vol.346, pp.124-132, 2008.

R. Jukanti, G. Devaraj, R. Devaraj, and S. Apte, Drug targeting to inflammation: studies on antioxidant surface loaded diclofenac liposomes, Int. J. Pharm, vol.414, pp.179-185, 2011.

L. Kürti, R. Gáspár, Á. Márki, E. Kápolna, A. Bocsik et al., In vitro and in vivo characterization of meloxicam nanoparticles designed for nasal administration, Eur. J. Pharm. Sci, vol.50, pp.86-92, 2013.

R. Katara and D. K. Majumdar, Eudragit RL 100-based nanoparticulate system of aceclofenac for ocular delivery, Colloids Surf. B Biointerfaces, vol.103, pp.455-462, 2013.

P. Khachane, A. A. Date, and M. S. Nagarsenker, Positively charged polymeric nanoparticles: application in improving therapeutic efficacy of meloxicam after oral administration, J. Pharm, vol.66, pp.334-338, 2011.

K. A. Khaled, H. A. Sarhan, M. A. Ibrahim, A. H. Ali, and Y. W. Naguib, Prednisoloneloaded PLGA microspheres: in vitro characterization and in vivo application in adjuvant-induced arthritis in mice, AAPS PharmSciTech, vol.11, pp.859-869, 2010.

W. Badri, International Journal of Pharmaceutics, vol.515, p.771, 2016.

R. M. Khalil, A. Abd-elbary, M. A. Kassem, M. M. Ghorab, and M. Basha, Nanostructured lipid carriers (NLCs) versus solid lipid nanoparticles (SLNs) for topical delivery of meloxicam, Pharm. Dev. Technol, vol.19, pp.304-314, 2014.

S. Kheradmandnia, E. Vasheghani-farahani, M. Nosrati, and F. Atyabi, Preparation and characterization of ketoprofen-loaded solid lipid nanoparticles made from beeswax and carnauba wax, Nanomed. Nanotechnol. Biol. Med, vol.6, pp.753-759, 2010.

S. Khoee and M. Yaghoobian, An investigation into the role of surfactants in controlling particle size of polymeric nanocapsules containing penicillin-G in double emulsion, Eur. J. Med. Chem, vol.44, pp.2392-2399, 2009.

S. Khurana, N. K. Jain, and P. M. Bedi, Nanostructured lipid carriers based nanogel for meloxicam delivery: mechanistic, in-vivo and stability evaluation, Drug Dev. Ind. Pharm, vol.41, pp.1368-1375, 2015.

S. J. Kim, A. J. Flach, and L. M. Jampol, Nonsteroidal anti-inflammatory drugs in ophthalmology, Surv. Ophthalmol, vol.55, pp.108-133, 2010.

A. G. Kohli, P. H. Kierstead, V. J. Venditto, C. L. Walsh, and F. C. Szoka, Designer lipids for drug delivery: from heads to tails, J. Control. Release, vol.190, pp.274-287, 2014.

P. S. Kumar, J. V. Ratna, and N. Hematheerthani, Lornaxicam loaded solid lipid nanoparticles for transdermal drug delivery formulation and evaluation, J. Sci. Res. Phys, vol.3, issue.1, pp.54-57, 2014.

A. Kumari, S. K. Yadav, and S. C. Yadav, Biodegradable polymeric nanoparticles based drug delivery systems, Colloids Surf. B Biointerfaces, vol.75, pp.1-18, 2010.

A. Laouini, C. Jaafar-maalej, I. Limayem-blouza, S. Sfar, C. Charcosset et al., Preparation, characterization and applications of liposomes: state of the art, J. Colloid Sci. Biotechnol, vol.1, pp.147-168, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02002202

M. Lattuada and T. A. Hatton, Synthesis, properties and applications of Janus nanoparticles, Nano Today, vol.6, pp.286-308, 2011.

K. E. Lee, Y. J. Choi, B. R. Oh, I. K. Chun, and H. S. Gwak, Formulation and in vitro/in vivo evaluation of levodopa transdermal delivery systems, Int. J. Pharm, vol.456, pp.432-436, 2013.

P. Legrand, S. Lesieur, A. Bochot, R. Gref, W. Raatjes et al., Influence of polymer behaviour in organic solution on the production of polylactide nanoparticles by nanoprecipitation, Int. J. Pharm, vol.344, pp.33-43, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00180715

S. Lehrer, Nasal NSAIDs for alzheimer's disease, Am. J. Alzheimers Dis. Other Demen, vol.29, pp.401-403, 2014.

Q. F. Lenz, S. S. Guterres, A. Pohlmann, and M. P. Alves, Semi-solid topical formulations containing nimesulide-loaded nanocapsules showed in-vivo antiinflammatory activity in chronic arthritis and fibrovascular tissue models, Inflamm. Res, vol.61, pp.305-310, 2012.

T. S. Levchenko, W. C. Hartner, and V. P. Torchilin, Liposomes for cardiovascular targeting, Ther. Deliv, vol.3, pp.501-514, 2012.

L. Blouza, I. Charcosset, C. Sfar, S. Fessi, and H. , Preparation and characterization of spironolactone-loaded nanocapsules for paediatric use, Int. J. Pharm, vol.325, pp.124-131, 2006.

F. Lince, D. L. Marchisio, and A. A. Barresi, Strategies to control the particle size distribution of poly-epsilon-caprolactone nanoparticles for pharmaceutical applications, J. Colloid Interface Sci, vol.322, pp.505-515, 2008.

K. Manjunath, J. S. Reddy, and V. Venkateswarlu, Solid lipid nanoparticles as drug delivery systems, Methods Find. Exp. Clin. Pharmacol, vol.27, p.127, 2005.

S. Mazzaferro, K. Bouchemal, A. Maksimenko, R. Skanji, P. Opolon et al., Reduced intestinal toxicity of docetaxel loaded into mucoadhesive nanoparticles, in mouse xenograft model, J. Colloid Sci. Biotechnol, vol.1, pp.210-217, 2012.

P. L. Mcgeer and E. G. Mcgeer, NSAIDs and Alzheimer disease: epidemiological, animal model and clinical studies, Neurobiol. Aging, vol.28, pp.639-647, 2007.

W. Mehnert and K. Mäder, Solid lipid nanoparticles: production, characterization and applications, Adv. Drug Deliv. Rev, vol.47, pp.165-196, 2001.

K. Miladi, S. Sfar, H. Fessi, and A. Elaissari, Drug carriers in osteoporosis: preparation, drug encapsulation and applications, Int. J. Pharm, vol.445, pp.181-195, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01958042

K. Miladi, D. Ibraheem, M. Iqbal, S. Sfar, H. Fessi et al., Particles from preformed polymers as carriers for drug delivery, EXCLI J, vol.13, pp.28-57, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02093354

C. Mohanraj, Nanoparticles-a review, Trop. J. Pharm. Res, vol.5, issue.1, pp.561-573, 2006.

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Polymer-based nanocapsules for drug delivery, Int. J. Pharm, vol.385, pp.113-142, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02002194

C. E. Mora-huertas, O. Garrigues, H. Fessi, and A. Elaissari, Nanocapsules prepared via nanoprecipitation and emulsification-diffusion methods: comparative study, Eur. J. Pharm. Biopharm, vol.80, pp.235-239, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02093455

C. Nagda, N. Chotai, S. Patel, D. Nagda, U. Patel et al., Chitosan microspheres of aceclofenac: in vitro and in vivo evaluation, Pharm. Dev. Technol, vol.15, pp.442-451, 2010.

D. Narayanan, M. G. Geena, H. Lakshmi, M. Koyakutty, S. Nair et al., Poly-(ethylene glycol) modified gelatin nanoparticles for sustained delivery of the anti-inflammatory drug ibuprofen-sodium: an in vitro and in vivo analysis, Nanomedicine, vol.9, pp.818-828, 2013.

N. Naseri, H. Valizadeh, and P. Zakeri-milani, Solid lipid nanoparticles and nanostructured lipid carriers: structure, preparation and application, Adv. Pharm. Bull, vol.5, pp.305-313, 2015.

E. Nogueira, A. C. Gomes, A. Preto, and A. Cavaco-paulo, Design of liposomal formulations for cell targeting, Colloids Surf. B Biointerfaces, vol.136, pp.514-526, 2015.

A. Okyar, Y. Özsoy, and S. Güngör, Novel Formulation Approaches for Dermal and Transdermal Delivery of Non-Steroidal Anti-Inflammatory Drugs, InTechOpen, 2012.

T. Pal, S. Paul, and B. Sa, Polymethylmethacrylate coated alginate matrix microcapsules for controlled release of diclofenac sodium, Pharmacol. Amp Pharm, vol.2, pp.56-66, 2011.

J. Pardeike, A. Hommoss, and R. H. Müller, Lipid nanoparticles (SLN, NLC) in cosmetic and pharmaceutical dermal products, Int. J. Pharm, vol.366, pp.170-184, 2009.

J. Pardeike, K. Schwabe, and R. H. Müller, Influence of nanostructured lipid carriers (NLC) on the physical properties of the Cutanova Nanorepair Q10 cream and the in vivo skin hydration effect, Int. J. Pharm, vol.396, pp.166-173, 2010.

R. Parhi and P. Suresh, Preparation and characterization of solid lipid nanoparticles-a review, Curr. Drug Discov. Technol, vol.9, pp.2-16, 2012.
DOI : 10.2174/157016312799304552

A. Parra, M. Mallandrich, B. Clares, M. A. Egea, M. Espina et al., Design and elaboration of freeze-dried PLGA nanoparticles for the transcorneal permeation of carprofen: ocular anti-inflammatory applications, Colloids Surf. B Biointerfaces, vol.136, pp.935-943, 2015.

A. R. Patel, M. B. Chougule, I. Townlry, R. Patlolla, G. Wang et al., Efficacy of aerosolized celecoxib encapsulated nanostructured lipid carrier in non-small cell lung cancer in combination with docetaxel, Pharm. Res, vol.30, pp.1435-1446, 2013.

R. R. Patlolla, M. Chougule, A. R. Patel, T. Jackson, P. N. Tata et al., Formulation, characterization and pulmonary deposition of nebulized celecoxib encapsulated nanostructured lipid carriers, J. Control. Release Soc, vol.144, pp.233-241, 2010.
DOI : 10.1016/j.jconrel.2010.02.006

URL : http://europepmc.org/articles/pmc2868936?pdf=render

C. Patrono and B. Rocca, Nonsteroidal antiinflammatory drugs: past, present and future, Pharmacol. Res, vol.59, pp.285-289, 2009.
DOI : 10.1016/j.phrs.2009.01.011

F. Pavanetto, P. Perugini, B. Conti, T. Modena, and I. Genta, Evaluation of process parameters involved in chitosan microsphere preparation by the o/w/o multiple emulsion method, J. Microencapsul, vol.13, pp.679-688, 1996.

L. Peter, D. Klaus, E. Wolfhard, and W. Trummlitz-klaus, Structure and physicochemical properties of meloxicam a new NSAID, Eur. J. Pharm. Sci, vol.4, pp.175-187, 1996.

A. A. Phatak and P. D. Chaudhari, Development and evaluation of Nanostructured Lipid Carrier (NLC) based topical delivery of an anti-inflammatory drug, J. Pharm. Res, vol.7, pp.677-685, 2013.

M. A. Phillips, M. L. Gran, and N. A. Peppas, Targeted nanodelivery of drugs and diagnostics, Nano Today, vol.5, pp.143-159, 2010.
DOI : 10.1016/j.nantod.2010.03.003

URL : http://europepmc.org/articles/pmc2882307?pdf=render

E. Plasari, P. H. Grisoni, and J. Villermaux, Influence of process parameters on the precipitation of organic nanoparticles by drowning-out, Chem. Eng. Res. Des, vol.75, pp.237-244, 1997.

F. S. Poletto, L. A. Fiel, M. V. Lopes, G. Schaab, A. M. Gomes et al., Fluorescent-labeled poly(e-caprolactone) lipid-core nanocapsules: synthesis, physicochemical properties and macrophage uptake, J. Colloid Sci. Biotechnol, vol.1, pp.89-98, 2012.
DOI : 10.1166/jcsb.2012.1015

T. J. Powell, N. Palath, M. E. Derome, J. Tang, A. Jacobs et al., Synthetic nanoparticle vaccines produced by layer-by-layer assembly of artificial biofilms induce potent protective T-cell and antibody responses in vivo, Vaccine, vol.29, pp.558-569, 2011.

C. Puglia, P. Blasi, L. Rizza, A. Schoubben, F. Bonina et al., Lipid nanoparticles for prolonged topical delivery: an in vitro and in vivo investigation, Int. J. Pharm, vol.357, pp.295-304, 2008.

K. D. Rainsford, Profile and mechanisms of gastrointestinal and other side effects of nonsteroidal anti-inflammatory drugs (NSAIDs), Am. J. Med, vol.107, pp.27-35, 1999.

J. S. Refuerzo, J. F. Alexander, F. Leonard, M. Leon, M. Longo et al., Liposomes: a nanoscale drug carrying system to prevent indomethacin passage to the fetus in a pregnant mouse model, Am. J. Obstet. Gynecol, vol.212, issue.508, pp.1-7, 2015.

C. P. Reis, S. Candeias, C. Fernandes, N. Martinho, N. Aniceto et al., Ibuprofen nanoparticles for oral delivery: proof of concept, J. Nanomed. Biother. Discov, vol.4, p.119, 2013.

E. Ricciotti and G. A. Fitzgerald, Prostaglandins and inflammation, Arterioscler. Thromb. Vasc. Biol, vol.31, pp.986-1000, 2011.
DOI : 10.1161/atvbaha.110.207449

URL : http://atvb.ahajournals.org/content/atvbaha/31/5/986.full.pdf

V. Rosset, N. Ahmed, I. Zaanoun, B. Stella, H. Fessi et al., Elaboration of argan oil nanocapsules containing naproxen for cosmetic and transdermal local application, J. Colloid Sci. Biotechnol, vol.1, pp.218-224, 2012.

A. Y. Rwei, W. Wang, and D. S. Kohane, Photoresponsive nanoparticles for drug delivery, Nano Today, vol.10, pp.451-467, 2015.
DOI : 10.1016/j.nantod.2015.06.004

URL : http://europepmc.org/articles/pmc4669578?pdf=render

J. ?tecová, W. Mehnert, T. Blaschke, B. Kleuser, R. Sivaramakrishnan et al., Cyproterone acetate loading to lipid nanoparticles for topical acne treatment: particle characterisation and skin uptake, Pharm. Res, vol.24, pp.991-1000, 2007.

B. Sütå, S. Berkó, G. Kozma, Á. Kukovecz, M. Budai-szács et al., Development of ibuprofenloaded nanostructured lipid carrier-based gels: characterization and investigation of in vitro and in vivo penetration through the skin, Int. J. Nanomed, vol.11, pp.1201-1212, 2016.

S. K. Sahoo, S. Parveen, and J. J. Panda, The present and future of nanotechnology in human health care, Nanomed. Nanotechnol. Biol. Med, vol.3, pp.20-31, 2007.

J. C. Schwarz, N. Baisaeng, M. Hoppel, M. Löw, C. M. Keck et al., Ultrasmall NLC for improved dermal delivery of coenyzme Q10, Int. J. Pharm, vol.447, pp.213-217, 2013.
DOI : 10.1016/j.ijpharm.2013.02.037

A. Scott, K. Khan, J. Cook, and V. Duronio, What is inflammation? Are we ready to move beyond Celsus?, Br. J. Sports Med, vol.38, pp.248-249, 2004.
DOI : 10.1136/bjsm.2003.011221

URL : https://bjsm.bmj.com/content/38/3/248.full.pdf

A. S. Soares, A special issue on applications of microencapsulation, J. Colloid Sci. Biotechnol, vol.2, pp.77-77, 2013.
DOI : 10.1166/jcsb.2013.1038

C. Sostres, C. J. Gargallo, M. T. Arroyo, and A. Lanas, Adverse effects of nonsteroidal anti-inflammatory drugs (NSAIDs, aspirin and coxibs) on upper gastrointestinal tract, Best Pract. Res. Clin. Gastroenterol, vol.24, pp.121-132, 2010.
DOI : 10.1016/j.bpg.2009.11.005

P. Srinath, M. G. Chary, S. P. Vyas, and P. V. Diwan, Long-circulating liposomes of indomethacin in arthritic rats-a biodisposition study, Pharm. Acta Helv, vol.74, pp.399-404, 2000.
DOI : 10.1016/s0031-6865(00)00023-6

B. Stella, S. Arpicco, F. Rocco, V. Marsaud, J. Renoir et al., Encapsulation of gemcitabine lipophilic derivatives into polycyanoacrylate nanospheres and nanocapsules, Int. J. Pharm, vol.344, pp.71-77, 2007.
DOI : 10.1016/j.ijpharm.2007.06.006

Z. Su, F. Sun, Y. Shi, C. Jiang, Q. Meng et al., Effects of formulation parameters on encapsulation efficiency and release behavior of risperidone poly (D,L-lactide-co-glycolide) microsphere, Chem. Pharm. Bull. (Tokyo), vol.57, pp.1251-1256, 2009.

J. Suksiriworapong, K. Sripha, J. Kreuter, and V. B. Junyaprasert, Comparative study of Ibuprofen and Indomethacin loaded poly(e-caprolactone) nanoparticles: physicochemical properties, J. Magn. Magn. Mater, vol.37, pp.17-27, 2010.

S. C. Sweetman, Martindale, The Complete Drug Reference, 2009.

S. Tammam, S. Mathur, and N. Afifi, Preparation and biopharmaceutical evaluation of tacrolimus loaded biodegradable nanoparticles for liver targeting, J. Biomed. Nanotechnol, vol.8, pp.439-449, 2012.
DOI : 10.1166/jbn.2012.1403

T. Tsukamoto, K. Hironaka, T. Fujisawa, D. Yamaguchi, K. Tahara et al., Preparation of bromfenac-loaded liposomes modified with chitosan for ophthalmic drug delivery and evaluation of physicochemical properties and drug release profile, Asian J. Pharm. Sci, vol.8, pp.104-109, 2013.

P. Valot, M. Baba, J. Nedelec, and N. Sintes-zydowicz, Effects of process parameters on the properties of biocompatible ibuprofen-loaded microcapsules, Int. J. Pharm, vol.369, pp.53-63, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00374579

C. H. Vineela, K. Sailaja, and K. , Preparation of ibuprofen-loaded Eudragit S100 nanoparticles by solvent evaporation technique, Int. J. Pharm. Sci. Res, vol.5, pp.375-384, 2014.

A. Z. Wang, R. Langer, and O. C. Farokhzad, Nanoparticle delivery of cancer drugs, Ann. Rev. Med, vol.63, issue.1, p.185, 2012.
DOI : 10.1146/annurev-med-040210-162544

S. Weber, A. Zimmer, and J. Pardeike, Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC) for pulmonary application: a review of the state of the art, Eur. J. Pharm. Biopharm, vol.86, pp.7-22, 2014.

K. S. Yadav and K. K. Sawant, Formulation optimization of etoposide loaded PLGA nanoparticles by double factorial design and their evaluation, Curr. Drug Deliv, vol.7, pp.51-64, 2010.
DOI : 10.2174/156720110790396517

N. Yadav, S. Khatak, and U. V. Sara, Solid lipid nanoparticles-a review, Int. J. Appl. Pharm, vol.5, pp.8-18, 2013.

L. Yildirimer, N. T. Thanh, M. Loizidou, and A. M. Seifalian, Toxicology and clinical potential of nanoparticles, Nano Today, vol.6, pp.585-607, 2011.
DOI : 10.1016/j.nantod.2011.10.001

URL : https://doi.org/10.1016/j.nantod.2011.10.001

I. Youm, J. B. Murowchick, and B. C. Youan, Entrapment and release kinetics of furosemide from pegylated nanocarriers, Colloids Surf. B Biointerfaces, vol.94, pp.133-142, 2012.
DOI : 10.1016/j.colsurfb.2012.01.027

V. Závi?ová, M. Koneracká, O. ?trbák, N. Toma?ovi9-cová, P. Kop9-canský et al., Encapsulation of indomethacin in magnetic biodegradable polymer nanoparticles, J. Magn. Magn. Mater, vol.311, pp.379-382, 2007.

N. Zafar, H. Fessi, and A. Elaissari, Cyclodextrin containing biodegradable particles: from preparation to drug delivery applications, Int. J. Pharm, vol.461, pp.351-366, 2014.
DOI : 10.1016/j.ijpharm.2013.12.004

Y. Zhai and G. Zhai, Advances in lipid-based colloid systems as drug carrier for topic delivery, J. Control. Release, vol.193, pp.90-99, 2014.
DOI : 10.1016/j.jconrel.2014.05.054

X. Zhu, A. F. Radovic-moreno, J. Wu, R. Langer, and J. Shi, Nanomedicine in the management of microbial infection-overview and perspectives, Nano Today, vol.9, pp.478-498, 2014.

J. Ziltener, S. Leal, and P. Fournier, Non-steroidal anti-inflammatory drugs for athletes: an update, Ann. Phys. Rehabil. Med, vol.53, pp.282-288, 2010.
DOI : 10.1016/j.rehab.2010.03.001

URL : https://doi.org/10.1016/j.rehab.2010.03.001

W. Badri, International Journal of Pharmaceutics, vol.515, p.773, 2016.

, strains from clinical isolates. Asian Pac J Trop Med, vol.7, pp.435-441, 2014.

D. Njimoh, A. Lemuh, and S. E. Mokake, Antimicrobial activities of a plethora of medicinal plant extracts and hydrolates against human pathogens and their potential to reverse antibiotic resistance, Int J Microbiol, vol.2015, pp.1-15, 2015.

G. S. Patten, M. Y. Abeywardena, and L. E. Bennett, Inhibition of angiotensin converting enzyme, angiotensin II receptor blocking and blood pressure lowering bioactivity across plant families, Crit Rev Food Sci Nutr, vol.56, pp.181-214, 2013.

L. Rubió, M. J. Motilva, and M. P. Romero, Recent advances in biologically active compounds in herbs and spices: a review of the most effective antioxidant and anti-inflammatory active principles, Crit Rev Food Sci Nutr, vol.53, pp.943-953, 2013.

D. Brambilla, P. Luciani, and J. C. Leroux, Breakthrough discoveries in drug delivery technologies: the next 30 years, J Controlled Release, vol.190, pp.9-14, 2014.

, Torchilin VP Nanoparticulates as drug carriers, 2006.

A. Shakeri and A. Sahebkar, Nanotechnology: a successful approach to improve oral bioavailability of phytochemicals, Recent Pat Drug Deliv Formul, vol.10, issue.1, pp.4-6, 2016.

P. Khachane, A. A. Date, and M. S. Nagarsenker, Eudragit EPO nanoparticles: application in improving therapeutic efficacy and reducing ulcerogenicity of meloxicam on oral administration, J Biomed Nanotechnol, vol.7, pp.590-597, 2011.

S. Mazzaferro, K. Bouchemal, and A. Maksimenko, Reduced intestinal toxicity of docetaxel loaded into mucoadhesive nanoparticles, in mouse xenograft model, J Colloid Sci Biotechnol, vol.1, pp.210-217, 2012.

S. Tammam, S. Mathur, and N. Afifi, Preparation and biopharmaceutical evaluation of tacrolimus loaded biodegradable nanoparticles for liver targeting, J Biomed Nanotechnol, vol.8, pp.439-449, 2012.

G. Barratt, Colloidal drug carriers: achievements and perspectives, Cell Mol Life Sci, vol.60, pp.21-37, 2003.

M. Gagliardi, G. Bardi, and A. Bifone, Polymeric nanocarriers for controlled and enhanced delivery of therapeutic agents to the CNS, Ther Deliv, vol.3, pp.875-887, 2012.

C. Heneweer, S. Gendy, and O. Peñate-medina, Liposomes and inorganic nanoparticles for drug delivery and cancer imaging, Ther Deliv, vol.3, pp.645-656, 2012.

E. P. Herrero, M. J. Alonso, and N. Csaba, Polymer-based oral peptide nanomedicines, Ther Deliv, vol.3, pp.657-668, 2012.
DOI : 10.4155/tde.12.40

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Polymer-based nanocapsules for drug delivery, Int J Pharm, vol.385, pp.113-142, 2010.
DOI : 10.1016/j.ijpharm.2009.10.018

C. Silva, D. De-oliveira, and L. Estevanato, Successful strategy for targeting the central nervous system using magnetic albumin nanospheres, J Biomed Nanotechnol, vol.8, pp.182-189, 2012.

T. S. Levchenko, W. C. Hartner, and V. P. Torchilin, Liposomes for cardiovascular targeting, Ther Deliv, vol.3, pp.501-514, 2012.
DOI : 10.4155/tde.12.18

F. S. Poletto, L. A. Fiel, and M. V. Lopes, Fluorescent-labeled poly(?Caprolactone) lipid-core nanocapsules: synthesis, physicochemical properties and macrophage uptake, J Colloid Sci Biotechnol, vol.1, pp.89-98, 2012.
DOI : 10.1166/jcsb.2012.1015

F. Wang, J. Li, and C. Wang, Hydrophilic and fluorescent colloidal nanorods of MWNTs as effective targeted drug carrier, J Colloid Sci Biotechnol, vol.1, pp.192-200, 2012.
DOI : 10.1166/jcsb.2012.1024

E. Cenni, D. Granchi, and S. Avnet, Biocompatibility of poly(D,LLactide-Co-Glycolide) nanoparticles conjugated with alendronate, Biomaterials, vol.29, pp.1400-1411, 2008.
DOI : 10.1016/j.biomaterials.2007.12.022

S. K. Sahoo, S. Parveen, and J. J. Panda, The present and future of nanotechnology in human health care, Nanomedicine Nanotechnol Biol Med, vol.3, pp.20-31, 2007.

K. Miladi, S. Sfar, and H. Fessi, Drug carriers in osteoporosis: preparation, drug encapsulation and applications, Int J Pharm, vol.445, pp.181-195, 2013.
DOI : 10.1016/j.ijpharm.2013.01.031

H. Ahmad, A special issue on polymer and hybrid particles for biomedical applications, J Colloid Sci Biotechnol, vol.2, pp.153-154, 2013.
DOI : 10.1166/jcsb.2013.1049

A. Soares, A Special issue on applications of microencapsulation, J Colloid Sci Biotechnol, vol.2, p.77, 2013.
DOI : 10.1166/jcsb.2013.1038

K. Miladi, D. Ibraheem, and M. Iqbal, Particles from preformed polymers as carriers for drug delivery, EXCLI J, vol.13, pp.28-57, 2014.

A. Kumari, S. K. Yadav, and S. C. Yadav, Biodegradable polymeric nanoparticles based drug delivery systems, Colloids Surf B Biointerfaces, vol.75, pp.1-18, 2010.
DOI : 10.1016/j.colsurfb.2009.09.001

A. Des-rieux, V. Fievez, and M. Garinot, Nanoparticles as potential oral delivery systems of proteins and vaccines: a mechanistic approach, J Controlled Release, vol.116, pp.1-27, 2006.

S. A. Galindo-rodriguez, E. Allemann, and H. Fessi, Polymeric nanoparticles for oral delivery of drugs and vaccines: a critical evaluation of in vivo studies, Crit Rev Ther Drug Carrier Syst, vol.22, pp.419-464, 2005.

N. Zafar, H. Fessi, and A. Elaissari, Cyclodextrin containing biodegradable particles: from preparation to drug delivery applications, Int J Pharm, vol.461, pp.351-366, 2014.
DOI : 10.1016/j.ijpharm.2013.12.004

M. S. El-aasser, J. Ugelstad, and J. W. Vanderhoff, Polymer Emulsification Process. US4177177 A, 1979.

M. Li, O. Rouaud, and D. Poncelet, Microencapsulation by solvent evaporation: state of the art for process engineering approaches, Int J Pharm, vol.363, pp.26-39, 2008.

T. K. Giri, C. Houdhary, and A. Ajazuddin, Prospects of pharmaceuticals and biopharmaceuticals loaded microparticles prepared by double emulsion technique for controlled delivery, Saudi Pharm J, vol.21, pp.125-141, 2013.
DOI : 10.1016/j.jsps.2012.05.009

URL : https://doi.org/10.1016/j.jsps.2012.05.009

G. Crotts and T. G. Park, Protein delivery from poly(lactic-Co-Glycolic Acid) biodegradable microspheres: release kinetics and stability issues, J Microencapsul, vol.15, issue.6, pp.15699-15713, 1998.
DOI : 10.3109/02652049809008253

H. Okochi and M. Nakano, Preparation and evaluation of W/o/w type emulsions containing vancomycin, Adv Drug Deliv Rev, vol.45, pp.5-26, 2000.
DOI : 10.1016/s0169-409x(00)00097-1

V. R. Sinha and A. Trehan, Biodegradable microspheres for protein delivery, J Controlled Release, vol.90, pp.261-280, 2003.
DOI : 10.1016/s0168-3659(03)00194-9

S. Khoee and M. Yaghoobian, An investigation into the role of surfactants in controlling particle size of polymeric nanocapsules containing penicillin-G in double emulsion, Eur J Med Chem, vol.44, pp.2392-2399, 2009.

H. Fessi, F. Puisieux, and J. P. Devissaguet, Nanocapsule formation by interfacial polymer deposition following solvent displacement, Int J Pharm, vol.55, pp.1-4, 1989.
DOI : 10.1016/0378-5173(89)90281-0

U. Seju, A. Kumar, and K. K. Sawant, Development and evaluation of olanzapine-loaded PLGA nanoparticles for nose-to-brain delivery: in vitro and in vivo studies, Acta Biomater, vol.7, pp.4169-4176, 2011.

R. Katara, D. K. Majumdar, and R. L. Eudragit, 100-based nanoparticulate system of aceclofenac for ocular delivery, Colloids Surf B Biointerfaces, vol.103, pp.455-462, 2013.
DOI : 10.1016/j.colsurfb.2012.10.056

K. P. Seremeta, D. A. Chiappetta, and A. Sosnik, Poly(?-Caprolactone), Eudragit® RS 100 and poly(?-caprolactone)/Eudragit® RS 100 blend submicron particles for the sustained release of the antiretroviral efavirenz, Colloids Surf B Biointerfaces, vol.102, pp.441-449, 2013.

G. Wang, B. Yu, and Y. Wu, Controlled preparation and antitumor efficacy of vitamin E TPGS-functionalized PLGA nanoparticles for delivery of paclitaxel, Int J Pharm, vol.446, pp.24-33, 2013.

M. P. Siqueira-moura, F. L. Primo, and E. M. Espreafico, Development, characterization, and photocytotoxicity assessment on human melanoma of chloroaluminum phthalocyanine nanocapsules, Mater Sci Eng C, vol.33, pp.1744-1752, 2013.
DOI : 10.1016/j.msec.2012.12.088

H. Van-de-ven, C. Paulussen, and P. B. Feijens, PLGA nanoparticles and nanosuspensions with amphotericin B: potent in vitro and in vivo alternatives to fungizone and AmBisome, J Control Release, vol.161, pp.795-803, 2012.

L. Mazzarino, C. Travelet, and S. Ortega-murillo, Elaboration of chitosan-coated nanoparticles loaded with curcumin for mucoadhesive applications, J Colloid Interface Sci, vol.370, pp.58-66, 2012.
DOI : 10.1016/j.jcis.2011.12.063

URL : https://hal.archives-ouvertes.fr/hal-00720279

C. Sanson, C. Schatz, L. Meins, and J. F. , A simple method to achieve high doxorubicin loading in biodegradable polymersomes, J Control Release, vol.147, pp.428-435, 2010.
DOI : 10.1016/j.jconrel.2010.07.123

URL : https://hal.archives-ouvertes.fr/hal-00652812

I. Barwal, A. Sood, and M. Sharma, Development of stevioside pluronicF-68 copolymer based PLA-nanoparticles as an antidiabetic nanomedicine, Colloids Surf B Biointerfaces, vol.101, pp.510-516, 2013.
DOI : 10.1016/j.colsurfb.2012.07.005

H. Eidi, O. Joubert, and C. Némos, Drug delivery by polymeric nanoparticles induces autophagy in macrophages, Int J Pharm, vol.422, pp.495-503, 2012.
DOI : 10.1016/j.ijpharm.2011.11.020

URL : https://hal.archives-ouvertes.fr/hal-00746557

W. S. Cheow and K. Hadinoto, Enhancing encapsulation efficiency of highly water-soluble antibiotic in poly
DOI : 10.1016/j.colsurfa.2010.08.050

A. Alshamsan, Nanoprecipitation is more efficient than emulsion solvent evaporation method to encapsulate cucurbitacin I in PLGA nanoparticles, Saudi Pharm. J. SPJ, vol.22, pp.219-222, 2014.
DOI : 10.1016/j.jsps.2013.12.002

URL : https://doi.org/10.1016/j.jsps.2013.12.002

B. Armendáriz-barragán, N. Zafar, W. Badri, S. A. Galindo-rodríguez, D. Kabbaj et al., Plant extracts: from encapsulation to application, Expert Opin. Drug Deliv, vol.13, pp.1165-1175, 2016.

S. Arpicco, L. Battaglia, P. Brusa, R. Cavalli, D. Chirio et al., Recent studies on the delivery of hydrophilic drugs in nanoparticulate systems, J. Drug Deliv. Sci. Technol, vol.32, pp.298-312, 2016.

H. Asadi, K. Rostamizadeh, D. Salari, and M. Hamidi, Preparation of biodegradable nanoparticles of tri-block PLA-PEG-PLA copolymer and determination of factors controlling the particle size using artificial neural network, J. Microencapsul, vol.28, pp.406-416, 2011.

A. E. Asbahani, K. Miladi, W. Badri, M. Sala, E. H. Addi et al., Essential oils: from extraction to encapsulation, Int. J. Pharm, vol.483, pp.220-243, 2015.
DOI : 10.1016/j.ijpharm.2014.12.069

URL : https://hal.archives-ouvertes.fr/hal-01230084

E. Averina and E. Allémann, Encapsulation of alimentary bioactive oils of the Baikal Lake area into pH-sensitive micro-and nanoparticles, LWT-Food Sci. Technol, vol.53, pp.271-277, 2013.

W. Badri, K. Miladi, Q. A. Nazari, H. Fessi, and A. Elaissari, Effect of process and formulation parameters on polycaprolactone nanoparticles prepared by solvent displacement, Colloids Surf. Physicochem. Eng. Asp, vol.516, pp.238-244, 2017.
DOI : 10.1016/j.colsurfa.2016.12.029

R. Bansal, A. Guleria, and P. C. Acharya, FT-IR method development and validation for quantitative estimation of zidovudine in bulk and tablet dosage form, Drug Res, vol.63, pp.165-170, 2013.
DOI : 10.1055/s-0032-1333297

C. G. Bareras-urbina, B. Ramírez-wong, G. A. López-ahumada, S. E. Burruel-ibarra, O. Martínez-cruz et al., Polymeric nanocapsules and nanospheres for encapsulation and long sustained release of hydrophobic cyanine-type photosensitizer, Bazyli nska, vol.19, pp.42-49, 2014.

D. J. Bharali, I. A. Siddiqui, V. M. Adhami, J. C. Chamcheu, A. M. Aldahmash et al., Nanoparticle delivery of natural products in the prevention and treatment of cancers: current status and future prospects, Cancers, vol.3, pp.4024-4045, 2011.

S. Bhattacharjee, DLS and zeta potential ? What they are and what they are not?, J. Controlled Release, vol.235, pp.337-351, 2016.
DOI : 10.1016/j.jconrel.2016.06.017

X. Bian, S. Liang, J. John, C. Hsiao, X. Wei et al., Development of PLGA-based itraconazole injectable nanospheres for sustained release, Int. J. Nanomed, vol.8, pp.4521-4531, 2013.

U. Bilati, E. Allémann, and E. Doelker, Development of a nanoprecipitation method intended for the entrapment of hydrophilic drugs into nanoparticles, Eur. J. Pharm. Sci, vol.24, pp.67-75, 2005.

A. L. Boehm, I. Martinon, R. Zerrouk, E. Rump, and H. Fessi, Nanoprecipitation technique for the encapsulation of agrochemical active ingredients, J. Microencapsul, vol.20, pp.433-441, 2003.

L. Bomgaars, J. R. Geyer, J. Franklin, G. Dahl, J. Park et al., Phase I trial of intrathecal liposomal cytarabine in children with neoplastic meningitis, J. Clin. Oncol, vol.22, pp.3916-3921, 2004.
DOI : 10.1200/jco.2004.01.046

A. Budhian, S. J. Siegel, and K. I. Winey, Haloperidol-loaded PLGA nanoparticles: systematic study of particle size and drug content, Int. J. Pharm, vol.336, pp.367-375, 2007.
DOI : 10.1016/j.ijpharm.2006.11.061

D. D. Castro-enríquez, F. Rodríguez-félix, B. Ramírez-wong, P. I. Torres-chávez, M. M. Castillo-ortega et al., Preparation, characterization and release of urea from wheat gluten electrospun membranes, Materials, vol.5, pp.2903-2916, 2012.

H. Chang and M. Yeh, Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy, Int. J. Nanomedicine, vol.7, pp.49-60, 2012.

J. Chen, M. Zhou, L. Shao, Y. Wang, J. Yun et al., Feasibility of preparing nanodrugs by high-gravity reactive precipitation, Int. J. Pharm, vol.269, pp.267-274, 2004.
DOI : 10.1016/j.ijpharm.2003.09.044

L. S. Ching and S. Mohamed, Alpha-tocopherol content in 62 edible tropical plants, J. Agric. Food Chem, vol.49, pp.3101-3105, 2001.
DOI : 10.1021/jf000891u

M. Chorny, I. Fishbein, H. D. Danenberg, and G. Golomb, Lipophilic drug loaded nanospheres prepared by nanoprecipitation: effect of formulation variables on size, drug recovery and release kinetics, J. Control. Release, vol.83, pp.389-400, 2002.
DOI : 10.1016/s0168-3659(02)00211-0

V. Chourasiya, S. Bohrey, and A. Pandey, Formulation, optimization, characterization and in-vitro drug release kinetics of atenolol loaded PLGA nanoparticles using 33 factorial design for oral delivery, Mater. Discov, vol.5, pp.1-13, 2016.

M. Christofoli, E. C. Costa, K. U. Bicalho, V. De-cássia-domingues, M. F. Peixoto et al., Insecticidal effect of nanoencapsulated essential oils from Zanthoxylum rhoifolium, 2015.
DOI : 10.1016/j.indcrop.2015.03.025

, Bemisia tabaci populations. Ind. Crops Prod, vol.70, pp.301-308

C. Contado, E. Vighi, A. Dalpiaz, and E. Leo, Influence of secondary preparative parameters and aging effects on PLGA particle size distribution: a sedimentation field flow fractionation investigation, Anal. Bioanal. Chem, vol.405, pp.703-711, 2013.

S. M. Prud'homme and R. K. , Controlling drug nanoparticle formation by rapid precipitation, Adv. Drug Deliv. Rev, vol.63, pp.417-426, 2011.

A. Dalpiaz, F. Sacchetti, A. Baldisserotto, B. Pavan, E. Maretti et al., Application of the in-oil nanoprecipitation method in the encapsulation of hydrophilic drugs in PLGA nanoparticles, J. Drug Deliv. Sci. Technol. Drug Deliv. Res. Italy 32 Part B, vol.283, p.290, 2016.

F. Danhier, N. Lecouturier, B. Vroman, C. Jérôme, J. Marchand-brynaert et al., Paclitaxel-loaded PEGylated PLGA-based nanoparticles: in vitro and in vivo evaluation, J. Control. Release, vol.133, pp.11-17, 2009.
DOI : 10.1016/j.jconrel.2008.09.086

S. Das, P. K. Suresh, and R. Desmukh, Design of Eudragit RL 100 nanoparticles by nanoprecipitation method for ocular drug delivery, Nanomed. Nanotechnol. Biol. Med, vol.6, pp.318-323, 2010.
DOI : 10.1016/j.nano.2009.09.002

N. ;. Desai, P. F. Da-silva, L. F. Azevedo, L. G. Da-rocha, I. C. De-moraes-porto et al., Challenges in development of nanoparticle-based therapeutics, AAPS J, vol.14, issue.2, pp.282-295, 2012.

E. J. Fonseca, J. De, E. Oliveira, A. T. Zhang, and D. G. Watson, Polymeric nanoparticles of brazilian red propolis extract: preparation, characterization, antioxidant and leishmanicidal activity, Nanoscale Res. Lett, vol.11, p.301, 2016.

Y. Dong and S. Feng, Methoxy poly(ethylene glycol)-poly(lactide) (MPEG-PLA) nanoparticles for controlled delivery of anticancer drugs, Biomaterials, vol.25, pp.2843-2849, 2004.
DOI : 10.1016/j.biomaterials.2003.09.055

S. Dufort, L. Sancey, and J. Coll, Physico-chemical parameters that govern nanoparticles fate also dictate rules for their molecular evolution, Adv. Drug Deliv. Rev. Biol. Interact. Nanopart, vol.64, pp.179-189, 2012.
DOI : 10.1016/j.addr.2011.09.009

A. C. Eifler and C. S. Thaxton, Nanoparticle therapeutics: FDA approval, clinical trials, regulatory pathways, and case study, in: biomedical nanotechnology, Methods in Molecular Biology, pp.325-338, 2011.
DOI : 10.1007/978-1-61779-052-2_21

H. Fessi, F. Puisieux, J. P. Devissaguet, N. Ammoury, and S. Benita, Nanocapsule formation by interfacial polymer deposition following solvent displacement, Int. J. Pharm, vol.55, pp.1-4, 1989.
DOI : 10.1016/0378-5173(89)90281-0

C. Fonseca, S. Simões, and R. Gaspar, Paclitaxel-loaded PLGA nanoparticles: preparation, physicochemical characterization and in vitro anti-tumoral activity, J. Control. Release, vol.83, pp.273-286, 2002.
DOI : 10.1016/s0168-3659(02)00212-2

URL : http://estudogeral.sib.uc.pt/jspui/bitstream/10316/5788/1/filebe5e8c1d04e24a22835b02fd186cf7df.pdf

I. Gülseren, Y. Fang, and M. Corredig, Whey protein nanoparticles prepared with desolvation with ethanol: characterization, thermal stability and interfacial behavior, Food Hydrocolloids, vol.29, pp.258-264, 2012.

S. A. Galindo-rodríguez, F. Puel, S. Briançon, E. Allémann, E. Doelker et al., Comparative scale-up of three methods for producing ibuprofen-loaded nanoparticles, Eur. J. Pharm. Sci, vol.25, pp.357-367, 2005.

F. Galisteo-gonzález and J. A. Molina-bolívar, Systematic study on the preparation of BSA nanoparticles, Colloids Surf. B Biointerfaces, vol.123, pp.286-292, 2014.

J. Ge, E. Neofytou, J. Lei, R. E. Beygui, and R. N. Zare, Protein-polymer hybrid nanoparticles for drug delivery, Small, vol.8, pp.3573-3578, 2012.
DOI : 10.1002/smll.201200889

S. K. Ghosh, Functional Coatings: By Polymer Microencapsulation, 2006.

T. Govender, S. Stolnik, M. C. Garnett, L. Illum, and S. S. Davis, PLGA nanoparticles prepared by nanoprecipitation: drug loading and release studies of a water soluble drug, J. Control. Release, vol.57, pp.171-185, 1999.

A. Hafner, J. Lovri-c, G. P. Lako?, and I. Pepi-c, Nanotherapeutics in the EU: an overview on current state and future directions, Int. J. Nanomed, vol.9, pp.1005-1023, 2014.

S. Han, M. Li, X. Liu, H. Gao, and Y. Wu, Construction of amphiphilic copolymer nanoparticles based on gelatin as drug carriers for doxorubicin delivery, Colloids Surf. B Biointerfaces, vol.102, pp.833-841, 2013.

A. L. Harvey, Natural products in drug discovery, Drug Discov. Today, vol.13, pp.894-901, 2008.

M. Iqbal, N. Zafar, H. Fessi, and A. Elaissari, Double emulsion solvent evaporation techniques used for drug encapsulation, Int. J. Pharm, vol.496, pp.173-190, 2015.
DOI : 10.1016/j.ijpharm.2015.10.057

V. Jeannot, S. Mazzaferro, J. Lavaud, L. Vanwonterghem, M. Henry et al., Targeting CD44 receptor-positive lung tumors using polysaccharidebased nanocarriers: influence of nanoparticle size and administration route, Nanomed. Nanotechnol. Biol. Med, vol.12, pp.921-932, 2016.
DOI : 10.1016/j.nano.2015.11.018

URL : https://hal.archives-ouvertes.fr/hal-01360462

M. Jelvehgari and S. H. Montazam, Comparison of microencapsulation by emulsion-solvent extraction/evaporation technique using derivatives cellulose and acrylate-methacrylate copolymer as carriers, Jundishapur J. Nat. Pharm. Prod, vol.7, pp.144-152, 2012.
DOI : 10.17795/jjnpp-3986

URL : http://cdn.neoscriber.org/cdn/serve/313ea/738c11dc82916a1cd4dc818f30485d8a07083931/18288-pdf.pdf

B. K. Johnson and R. K. Prud'homme, Mechanism for rapid self-assembly of block copolymer nanoparticles, Phys. Rev. Lett, vol.91, p.118302, 2003.
DOI : 10.1103/physrevlett.91.118302

I. J. Joye and D. J. Mcclements, Production of nanoparticles by anti-solvent precipitation for use in food systems, Trends Food Sci. Technol, vol.34, pp.109-123, 2013.

M. Kalani and R. Yunus, Application of supercritical antisolvent method in drug encapsulation: a review, Int. J. Nanomed, vol.6, pp.9-42, 2011.

R. Katara and D. K. Majumdar, Eudragit RL 100-based nanoparticulate system of aceclofenac for ocular delivery, Colloids Surf. B Biointerfaces, vol.103, pp.455-462, 2013.
DOI : 10.1016/j.colsurfb.2012.10.056

O. Kayser, A. Lemke, and N. Hernández-trejo, The impact of nanobiotechnology on the development of new drug delivery systems, Curr. Pharm. Biotechnol, vol.6, pp.3-5, 2005.

P. Khachane, A. A. Date, and M. S. Nagarsenker, Eudragit EPO nanoparticles: application in improving therapeutic efficacy and reducing ulcerogenicity of meloxicam on oral administration, J. Biomed. Nanotechnol, vol.7, pp.590-597, 2011.

S. Kumar, S. S. Kesharwani, H. Mathur, M. Tyagi, G. J. Bhat et al., Molecular complexation of curcumin with pH sensitive cationic copolymer enhances the aqueous solubility, stability and bioavailability of curcumin, Eur. J. Pharm. Sci, vol.82, pp.86-96, 2016.

K. Langer, M. G. Anhorn, I. Steinhauser, S. Dreis, D. Celebi et al., Human serum albumin (HSA) nanoparticles: reproducibility of preparation process and kinetics of enzymatic degradation, Int. J. Pharm, vol.347, pp.109-117, 2008.

E. J. Lee, S. A. Khan, J. K. Park, and K. Lim, Studies on the characteristics of drugloaded gelatin nanoparticles prepared by nanoprecipitation, Bioprocess Biosyst. Eng, vol.35, pp.297-307, 2012.
DOI : 10.1007/s00449-011-0591-2

K. Letchford and H. Burt, A review of the formation and classification of amphiphilic block copolymer nanoparticulate structures: micelles, nanospheres, nanocapsules and polymersomes, Eur. J. Pharm. Biopharm, vol.65, pp.259-269, 2007.

L. Blouza, I. Charcosset, C. Sfar, S. Fessi, and H. , Preparation and characterization of spironolactone-loaded nanocapsules for paediatric use, Int. J. Pharm, vol.325, pp.124-131, 2006.

W. Li, M. Yalcin, Q. Lin, M. M. Ardawi, and S. A. Mousa, Self-assembly of green tea catechin derivatives in nanoparticles for oral lycopene delivery, J. Controlled Release, vol.248, pp.117-124, 2017.

F. Lince, D. L. Marchisio, and A. A. Barresi, Strategies to control the particle size distribution of poly-epsilon-caprolactone nanoparticles for pharmaceutical applications, J. Colloid Interface Sci, vol.322, pp.505-515, 2008.

T. Loftsson and M. E. Brewster, Pharmaceutical applications of cyclodextrins: basic science and product development, J. Pharm. Pharmacol, vol.62, pp.1607-1621, 2010.
DOI : 10.1111/j.2042-7158.2010.01030.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.2042-7158.2010.01030.x/pdf

W. Lohcharoenkal, L. Wang, Y. C. Chen, and Y. Rojanasakul, Protein nanoparticles as drug delivery carriers for cancer therapy, BioMed Res. Int, 2014.
DOI : 10.1155/2014/180549

URL : http://downloads.hindawi.com/journals/bmri/2014/180549.pdf

A. G. Luque-alcaraz, J. Lizardi-mendoza, F. M. Goycoolea, I. Higuera-ciapara, and W. Argüelles-monal, Preparation of chitosan nanoparticles by nanoprecipitation and their ability as a drug nanocarrier, RSC Adv, vol.6, pp.59250-59256, 2016.
DOI : 10.1039/c6ra06563e

D. L. Marchisio, L. Rivautella, and A. A. Barresi, Design and scale-up of chemical reactors for nanoparticle precipitation, AIChE J, vol.52, pp.1877-1887, 2006.
DOI : 10.1002/aic.10786

L. Martín-banderas, J. Álvarez-fuentes, M. Durán-lobato, J. Prados, C. Melguizo et al., Cannabinoid derivate-loaded PLGA nanocarriers for oral administration: formulation, characterization, and cytotoxicity studies, Int. J. Nanomed, vol.7, pp.5793-5806, 2012.

L. Mazzarino, C. Travelet, S. Ortega-murillo, I. Otsuka, I. Pignot-paintrand et al., Elaboration of chitosan-coated nanoparticles loaded with curcumin for mucoadhesive applications, J. Colloid Interface Sci, vol.370, pp.58-66, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00720279

K. Miladi, D. Ibraheem, M. Iqbal, S. Sfar, H. Fessi et al., Particles from preformed polymers as carriers for drug delivery, EXCLI J, vol.13, pp.28-57, 2014.

K. Miladi, S. Sfar, H. Fessi, and A. Elaissari, Encapsulation of alendronate sodium by nanoprecipitation and double emulsion: from preparation to in vitro studies, Special issue derived from International Conference on Bio-based Materials and Composites, vol.72, pp.24-33, 2015.
DOI : 10.1016/j.indcrop.2015.01.079

K. Miladi, S. Sfar, H. Fessi, and A. Elaissari, Nanoprecipitation process: from particle preparation to in vivo applications, pp.17-53, 2016.
DOI : 10.1007/978-3-319-41421-8_2

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Polymer-based nanocapsules for drug. Delivery, Int. J. Pharm, vol.385, pp.113-142, 2010.
DOI : 10.1016/j.ijpharm.2009.10.018

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Influence of process and formulation parameters on the formation of submicron particles by solvent displacement and emulsification-diffusion methods: critical comparison, Adv. Colloid Interface Sci, vol.163, pp.90-122, 2011.

J. Nicolas, S. Mura, D. Brambilla, N. Mackiewicz, and P. Couvreur, Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery, Chem. Soc. Rev, vol.42, pp.1147-1235, 2013.

C. M. Noronha, A. F. Granada, S. M. De-carvalho, R. C. Lino, O. B. De et al., Optimization of a-tocopherol loaded nanocapsules by the nanoprecipitation method, Ind. Crops Prod, vol.50, pp.896-903, 2013.

R. Paliwal, R. J. Babu, and S. Palakurthi, Nanomedicine scale-up technologies: feasibilities and challenges, AAPS PharmSciTech, vol.15, pp.1527-1534, 2014.
DOI : 10.1208/s12249-014-0177-9

URL : http://europepmc.org/articles/pmc4245446?pdf=render

E. A. Palombo, Traditional medicinal plant extracts and natural products with activity against oral bacteria: potential application in the prevention and treatment of oral diseases, Nat. Med, 2011.

A. R. Patel, Y. Hu, J. K. Tiwari, and K. P. Velikov, Synthesis and characterisation of zein-curcumin colloidal particles, Soft Matter, vol.6, pp.6192-6199, 2010.
DOI : 10.1039/c0sm00800a

Y. Pathak and D. Thassu, Drug Delivery Nanoparticles Formulation and Characterization, Drugs and the Pharmaceutical Sciences, 2009.

L. Peltonen, J. Aitta, S. Hyvönen, M. Karjalainen, and J. Hirvonen, Improved entrapment efficiency of hydrophilic drug substance during nanoprecipitation of poly(l)lactide nanoparticles, AAPS PharmSciTech, vol.5, 2004.
DOI : 10.1208/pt050116

URL : http://europepmc.org/articles/pmc2784850?pdf=render

R. Pereira, T. Julianto, K. Yuen, and A. Abdul-majeed, Anionic Eudragit nanoparticles as carriers for oral administration of peptidomimetic drugs, International Conference on Nanoscience and Nanotechnology, pp.298-301, 2006.
DOI : 10.1109/iconn.2006.340611

A. Popov, L. Schopf, J. Bourassa, and H. Chen, Enhanced pulmonary delivery of fluticasone propionate in rodents by mucus-penetrating nanoparticles, Int. J. Pharm, vol.502, pp.188-197, 2016.

A. M. Puga, A. Rey-rico, B. Magariños, C. Alvarez-lorenzo, and A. Concheiro, Hot melt poly-e-caprolactone/poloxamine implantable matrices for sustained delivery of ciprofloxacin, Acta Biomater, vol.8, pp.1507-1518, 2012.

K. M. Pustulka, A. R. Wohl, H. S. Lee, A. R. Michel, J. Han et al., Flash nanoprecipitation: particle structure and stability, Mol. Pharm, vol.10, pp.4367-4377, 2013.
DOI : 10.1021/mp400337f

URL : http://europepmc.org/articles/pmc3946569?pdf=render

Y. Qin, C. Liu, S. Jiang, L. Xiong, and Q. Sun, Characterization of starch nanoparticles prepared by nanoprecipitation: influence of amylose content and starch type, Ind. Crops Prod, vol.87, pp.182-190, 2016.

C. N. Quiroz-reyes, E. R. Jesús, N. E. Duran-caballero, and M. Á. Aguilar-méndez, Development and characterization of gelatin nanoparticles loaded with a cocoa-derived polyphenolic extract, Fruits, vol.69, pp.481-489, 2014.
DOI : 10.1051/fruits/2014034

URL : https://fruits.edpsciences.org/10.1051/fruits/2014034/pdf

C. S. Ritter, M. D. Baldissera, T. H. Grando, C. F. Souza, M. R. Sagrillo et al., Achyrocline satureioides essential oil-loaded in nanocapsules reduces cytotoxic damage in liver of rats infected by Trypanosoma evansi, Microb. Pathog, vol.103, pp.149-154, 2017.

F. F. Sahle, C. Gerecke, B. Kleuser, and R. Bodmeier, Formulation and comparative in vitro evaluation of various dexamethasone-loaded pH-sensitive polymeric nanoparticles intended for dermal applications, Int. J. Pharm, vol.516, pp.21-31, 2017.

S. Sahu, Biocompatible nanoparticles for sustained topical delivery of anticancer Phytoconstituent quercetin*Sneha Sahu, Swarnlata Saraf, Chanchal Deep Kaur and Shailendra Saraf Shri Rawatpura Sarkar Institute of Pharmacy, J. Biol. Sci, vol.16, pp.601-609, 2013.

D. Sharma, R. K. Sharma, N. Sharma, R. Gabrani, S. K. Sharma et al., Nose-to-brain delivery of PLGA-diazepam nanoparticles, AAPS PharmSciTech, vol.16, pp.1108-1121, 2015.
DOI : 10.1208/s12249-015-0294-0

URL : http://europepmc.org/articles/pmc4674633?pdf=render

S. ?im?ek, H. Ero-glu, B. Kurum, and K. Ulubayram, Brain targeting of Atorvastatin loaded amphiphilic PLGA-b-PEG nanoparticles, J. Microencapsul, vol.30, pp.10-20, 2013.

G. Singh and R. S. Pai, In-vitro/in-vivo characterization of trans-resveratrolloaded nanoparticulate drug delivery system for oral administration, J. Pharm. Pharmacol, vol.66, pp.1062-1076, 2014.

M. N. Singh, K. S. Hemant, M. Ram, and H. G. Shivakumar, Microencapsulation: a promising technique for controlled drug delivery, Res. Pharm. Sci, vol.5, pp.65-77, 2010.

V. R. Sinha, K. Bansal, R. Kaushik, R. Kumria, and A. Trehan, Poly-epsiloncaprolactone microspheres and nanospheres: an overview, Int. J. Pharm, vol.278, pp.1-23, 2004.

M. P. Siqueira-moura, F. L. Primo, E. M. Espreafico, and A. C. Tedesco, Development, characterization, and photocytotoxicity assessment on human melanoma of chloroaluminum phthalocyanine nanocapsules, Mater. Sci. Eng. C, vol.33, pp.1744-1752, 2013.
DOI : 10.1016/j.msec.2012.12.088

-. Vincristine-liposomal, lipid-encapsulated vincristine, onco TCS, transmembrane carrier system-vincristine, vincacine, vincristine sulfate liposomes for injection, VSLI, Drugs RD, vol.5, pp.119-123, 2004.

M. Wacker, Nanocarriers for intravenous injection-the long hard road to the market, Int. J. Pharm, vol.457, pp.50-62, 2013.
DOI : 10.1016/j.ijpharm.2013.08.079

T. Wang, D. Zhu, G. Liu, W. Tao, W. Cao et al., DTX-loaded star-shaped TAPP-PLA-b-TPGS nanoparticles for cancer chemical and photodynamic combination therapy, RSC Adv, vol.5, pp.50617-50627, 2015.
DOI : 10.1039/c5ra09042c

Y. Wang and Y. Tan, Enhanced drug loading capacity of 10-hydroxycamptothecinloaded nanoparticles prepared by two-step nanoprecipitation method, J. Drug Deliv. Sci. Technol, vol.36, pp.183-191, 2016.
DOI : 10.1016/j.jddst.2016.09.012

X. Yan, R. Ramos, E. Hoibian, C. Soulage, P. Alcouffe et al., Nanoprecipitation of PHPMA (Co)polymers into nanocapsules displaying tunable compositions, dimensions, and surface properties, ACS Macro Lett, vol.6, pp.447-451, 2017.
DOI : 10.1021/acsmacrolett.7b00094

URL : https://hal.archives-ouvertes.fr/hal-01637616

G. Yordanov, R. Skrobanska, and A. Evangelatov, Entrapment of epirubicin in poly (butyl cyanoacrylate) colloidal nanospheres by nanoprecipitation: formulation development and in vitro studies on cancer cell lines, Colloids Surf. B Biointerfaces, vol.92, pp.98-105, 2012.

G. Yuan, S. Yan, H. Xue, P. Zhang, J. Sun et al., Cucurbitacin I induces protective autophagy in glioblastoma in vitro and in vivo, J. Biol. Chem, vol.289, pp.10607-10619, 2014.
DOI : 10.1074/jbc.m113.528760

URL : http://www.jbc.org/content/289/15/10607.full.pdf

X. Zhang, X. Zhang, S. Wang, M. Liu, L. Tao et al., Surfactant modification of aggregation-induced emission material as biocompatible nanoparticles: facile preparation and cell imaging, Nanoscale, vol.5, pp.147-150, 2012.
DOI : 10.1039/c2nr32698a

P. Zhou, T. An, C. Zhao, Y. Li, R. Li et al., Lactosylated PLGA nanoparticles containing e-polylysine for the sustained release and livertargeted delivery of the negatively charged proteins, Int. J. Pharm, vol.478, pp.633-643, 2015.

C. J. Martínez-rivas, International Journal of Pharmaceutics, vol.532, p.81, 2017.

M. Iqbal, N. Zafar, H. Fessi, and A. Elaissari, Double emulsion solvent evaporation techniques used for drug encapsulation, Int. J. Pharm, vol.496, pp.173-190, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01958031

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Polymer-based nanocapsules for drug delivery, Int. J. Pharm, vol.385, pp.1-2, 2010.
DOI : 10.1016/j.ijpharm.2009.10.018

D. Quintanar-guerrero, E. Allémann, H. Fessi, and E. Doelker, Preparation techniques and mechanisms of formation of biodegradable nanoparticles from preformed polymers, Drug Dev. Ind. Pharm, vol.24, pp.1113-1128, 1998.

M. Gagliardi, G. Bardi, and A. Bifone, Polymeric nanocarriers for controlled and enhanced delivery of therapeutic agents to the CNS, Ther. Deliv, vol.3, pp.875-887, 2012.

E. P. Herrero, M. J. Alonso, and N. Csaba, Polymer-based oral peptide nanomedicines, Ther. Deliv, vol.3, pp.657-668, 2012.
DOI : 10.4155/tde.12.40

T. S. Levchenko, W. C. Hartner, and V. P. Torchilin, Liposomes for cardiovascular targeting, Ther. Deliv, vol.3, pp.501-514, 2012.
DOI : 10.4155/tde.12.18

F. S. Poletto, L. A. Fiel, M. V. Lopes, G. Schaab, A. M. Gomes et al., Fluorescent-labeled poly(-caprolactone) lipid-Core nanocapsules: synthesis, physicochemical properties and macrophage uptake, J. Colloid Sci. Biotechnol, vol.1, pp.89-98, 2012.
DOI : 10.1166/jcsb.2012.1015

F. Wang, J. Li, and C. Wang, Hydrophilic and fluorescent colloidal nanorods of MWNTs as effective targeted drug carrier, J. Colloid Sci. Biotechnol, vol.1, pp.192-200, 2012.
DOI : 10.1166/jcsb.2012.1024

E. Cenni, D. Granchi, S. Avnet, C. Fotia, M. Salerno et al., Biocompatibility of poly(d,l-lactide-co-glycolide) nanoparticles conjugated with alendronate, Biomaterials, vol.29, pp.1400-1411, 2008.
DOI : 10.1016/j.biomaterials.2007.12.022

S. K. Sahoo, S. Parveen, and J. J. Panda, The present and future of nanotechnology in human health care, Nanomed. Nanotechnol. Biol. Med, vol.3, pp.20-31, 2007.

K. Miladi, S. Sfar, H. Fessi, and A. Elaissari, Drug carriers in osteoporosis: preparation, drug encapsulation and applications, Int. J. Pharm, vol.445, pp.181-195, 2013.
DOI : 10.1016/j.ijpharm.2013.01.031

D. De, O. Cintra-e-silva, L. L. Estevanato, A. R. Simioni, M. M. De et al., Successful strategy for targeting the central nervous system using magnetic albumin nanospheres, J. Biomed. Nanotechnol, vol.8, pp.182-189, 2012.

M. S. Singh and D. Peer, RNA nanomedicines: the next generation drugs?, Curr. Opin. Biotechnol, vol.39, pp.28-34, 2016.
DOI : 10.1016/j.copbio.2015.12.011

J. Yang and J. Kope?ek, Polymeric biomaterials and nanomedicines, J. Drug Deliv. Sci. Technol, vol.30, pp.318-330, 2015.
DOI : 10.1016/j.jddst.2015.05.012

URL : https://manuscript.elsevier.com/S1773224715000842/pdf/S1773224715000842.pdf

L. Bregoli, D. Movia, J. D. Gavigan-imedio, J. Lysaght, J. Reynolds et al., Nanomedicine applied to translational oncology: a future perspective on cancer treatment, Nanomed. Nanotechnol. Biol. Med, vol.12, pp.81-103, 2016.
DOI : 10.1016/j.nano.2015.08.006

M. Gharagozloo, S. Majewski, and M. Foldvari, Therapeutic applications of nanomedicine in autoimmune diseases: from immunosuppression to tolerance induction, Nanomed. Nanotechnol. Biol. Med, vol.11, pp.1003-1018, 2015.

I. R. Bell, G. E. Schwartz, N. N. Boyer, M. Koithan, and A. J. Brooks, Advances in integrative nanomedicine for improving infectious disease treatment in public health, Eur. J. Integr. Med, vol.5, pp.126-140, 2013.
DOI : 10.1016/j.eujim.2012.11.002

URL : http://europepmc.org/articles/pmc3685499?pdf=render

R. G. Bai, K. Muthoosamy, and S. Manickam, Nanotechnology Applications for Tissue Engineering, p.12, 2015.

A. Kumari, S. K. Yadav, and S. C. Yadav, Biodegradable polymeric nanoparticles based drug delivery systems, Colloids Surf. B: Biointerfaces, vol.75, pp.1-18, 2010.
DOI : 10.1016/j.colsurfb.2009.09.001

A. Des-rieux, V. Fievez, M. Garinot, Y. Schneider, and V. Préat, Nanoparticles as potential oral delivery systems of proteins and vaccines: a mechanistic approach, J. Control. Release, vol.116, pp.1-27, 2006.

S. A. Galindo-rodriguez, E. Allemann, H. Fessi, and E. Doelker, Polymeric nanoparticles for oral delivery of drugs and vaccines: a critical evaluation of in vivo studies, Crit. Rev. Ther. Drug Carrier Syst, vol.22, pp.419-464, 2005.

N. Zafar, H. Fessi, and A. Elaissari, Cyclodextrin containing biodegradable particles: from preparation to drug delivery applications, Int. J. Pharm, vol.461, pp.351-366, 2014.
DOI : 10.1016/j.ijpharm.2013.12.004

W. Badri, K. Miladi, R. Eddabra, H. Fessi, and A. Elaissari, Elaboration of nanoparticles containing indomethacin: argan oil for transdermal local and cosmetic application, J. Nanomater, p.2015, 2015.
DOI : 10.1155/2015/935439

URL : http://doi.org/10.1155/2015/935439

B. D. Ulery, L. S. Nair, and C. T. Laurencin, Biomedical applications of biodegradable polymers, J. Polym. Sci. Part B: Polym. Phys, vol.49, pp.832-864, 2011.
DOI : 10.1002/polb.22259

URL : http://onlinelibrary.wiley.com/doi/10.1002/polb.22259/pdf

D. Bennet and S. Kim, Polymer nanoparticles for smart drug delivery, Application of Nanotechnology in Drug Delivery, 2014.

H. Fessi, F. Puisieux, J. P. Devissaguet, N. Ammoury, and S. Benita, Nanocapsule formation by interfacial polymer deposition following solvent displacement, Int. J. Pharm, vol.55, pp.1-4, 1989.
DOI : 10.1016/0378-5173(89)90281-0

M. Chorny, I. Fishbein, H. D. Danenberg, and G. Golomb, Lipophilic drug loaded nanospheres prepared by nanoprecipitation: effect of formulation variables on size, drug recovery and release kinetics, J. Control. Release, vol.83, pp.389-400, 2002.
DOI : 10.1016/s0168-3659(02)00211-0

C. Contado, E. Vighi, A. Dalpiaz, and E. Leo, Influence of secondary preparative parameters and aging effects on PLGA particle size distribution: a sedimentation field flow fractionation investigation, Anal. Bioanal. Chem, vol.405, pp.703-711, 2012.
DOI : 10.1007/s00216-012-6113-5

J. M. Lankveld and J. Lyklema, Adsorption of polyvinyl alcohol on the paraffin-water interface. II. Spread and adsorbed monolayers, J. Colloid Interface Sci, vol.41, pp.466-474, 1972.

R. Ali, G. K. Jain, Z. Iqbal, S. Talegaonkar, P. Pandit et al., Development and clinical trial of nano-atropine sulfate dry powder inhaler as a novel organophosphorous poisoning antidote, Nanomed. Nanotechnol. Biol. Med, vol.5, pp.55-63, 2009.
DOI : 10.1016/j.nano.2008.07.001

K. Miladi, S. Sfar, H. Fessi, and A. Elaissari, Encapsulation of alendronate sodium by nanoprecipitation and double emulsion: from preparation to in vitro studies, Ind. Crops Prod, vol.72, pp.24-33, 2015.

J. A. Cordero, M. Camacho, R. Obach, J. Domenech, and L. Vila, In vitro based index of topical anti-inflammatory activity to compare a series of NSAIDs, Eur J Pharm Biopharm, vol.51, pp.135-177, 2001.

C. Sostres, C. J. Gargallo, M. T. Arroyo, and A. Lanas, Adverse effects of non-steroidal anti-inflammatory drugs (NSAIDs, aspirin and coxibs) on upper gastrointestinal tract, Best Pract Res Clin Gastroenterol, vol.24, pp.121-153, 2010.
DOI : 10.1016/j.bpg.2009.11.005

D. N. Bateman, Non-steroidal anti-inflammatory drugs, Medicine (Baltimore), vol.40, p.140, 2012.

S. J. Kim, A. J. Flach, and L. M. Jampol, Nonsteroidal anti-inflammatory drugs in ophthalmology, Surv Ophthalmol, vol.55, pp.108-141, 2010.

J. Ziltener, S. Leal, and P. Fournier, Non-steroidal anti-inflammatory drugs for athletes: an update, Ann Phys Rehabil Med, vol.53, pp.278-82, 2010.
DOI : 10.1016/j.rehab.2010.03.001

URL : https://doi.org/10.1016/j.rehab.2010.03.001

V. Závi?ová, M. Koneracká, O. ?trbák, N. Toma?ovi?ová, P. Kop?anský et al., Encapsulation of indomethacin in magnetic biodegradable polymer nanoparticles, J Magn Magn Mater, vol.311, pp.379-82, 2007.

M. R. Prausnitz and R. Langer, Transdermal drug delivery, Nat Biotechnol, vol.26, pp.1261-1269, 2008.

K. Tomoda, H. Terashima, K. Suzuki, T. Inagi, H. Terada et al., Enhanced transdermal delivery of indomethacin using combination of PLGA nanoparticles and iontophoresis in vivo, Colloids Surf B: Biointerfaces, vol.92, pp.50-54, 2012.

J. Suksiriworapong, K. Sripha, J. Kreuter, and V. B. Junyaprasert, Comparative study of Ibuprofen and Indomethacin loaded poly (caprolactone) nanoparticles: physicochemical properties, J Magn Magn Mater, vol.37, pp.17-27, 2010.

M. Iqbal, N. Zafar, H. Fessi, and A. Elaissari, Double emulsion solvent evaporation techniques used for drug encapsulation, Int J Pharm, vol.496, pp.173-90, 2015.
DOI : 10.1016/j.ijpharm.2015.10.057

M. Jelvehgari and S. H. Montazam, Comparison of Microencapsulation by Emulsion-Solvent Extraction/Evaporation Technique Using Derivatives Cellulose and Acrylate-Methacrylate Copolymer as Carriers, Jundishapur J Nat Pharm Prod, vol.7, pp.144-52, 2012.
DOI : 10.17795/jjnpp-3986

URL : http://cdn.neoscriber.org/cdn/serve/313ea/738c11dc82916a1cd4dc818f30485d8a07083931/18288-pdf.pdf

H. Fessi, F. Puisieux, J. P. Devissaguet, N. Ammoury, and S. Benita, Nanocapsule formation by interfacial polymer deposition following solvent displacement, Int J Pharm, vol.55, pp.1-4, 1989.
DOI : 10.1016/0378-5173(89)90281-0

K. Miladi, S. Sfar, H. Fessi, and A. Elaissari, Nanoprecipitation process: from particle preparation to in vivo applications, pp.17-53, 2016.
DOI : 10.1007/978-3-319-41421-8_2

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Polymer-based nanocapsules for drug delivery, Int J Pharm, vol.385, pp.113-155, 2010.
DOI : 10.1016/j.ijpharm.2009.10.018

J. Park, M. G. Allen, and M. R. Prausnitz, Biodegradable polymer microneedles: fabrication, mechanics and transdermal drug delivery, J Control Release Off J Control Release Soc, vol.104, pp.51-66, 2005.

T. K. Dash and V. B. Konkimalla, Poly-?-caprolactone based formulations for drug delivery and tissue engineering: A review, J Control Release Off, vol.158, pp.15-33, 2012.
DOI : 10.1016/j.jconrel.2011.09.064

W. Badri, K. Miladi, Q. A. Nazari, H. Fessi, and A. Elaissari, Effect of process and formulation parameters on polycaprolactone nanoparticles prepared by solvent displacement, Colloids Surf A Physicochem Eng Asp, vol.516, pp.238-282, 2017.
DOI : 10.1016/j.colsurfa.2016.12.029

S. R. Pygall, J. Whetstone, P. Timmins, and C. D. Melia, Pharmaceutical applications of confocal laser scanning microscopy: The physical characterisation of pharmaceutical systems, Adv Drug Deliv Rev, vol.59, pp.1434-52, 2007.

R. Alvarez-román, A. Naik, Y. N. Kalia, R. H. Guy, and H. Fessi, Skin penetration and distribution of polymeric nanoparticles, J Control Release, vol.99, pp.53-62, 2004.

X. Zhang, X. Zhang, S. Wang, M. Liu, L. Tao et al., Surfactant modification of aggregation-induced emission material as Badri et al. 88 biocompatible nanoparticles: Facile preparation and cell imaging, Nanoscale, vol.5, pp.147-50, 2012.
DOI : 10.1039/c2nr32698a

K. Adibkia, Y. Javadzadeh, S. Dastmalchi, G. Mohammadi, F. K. Niri et al., Naproxen-eudragit RS100 nanoparticles: preparation and physicochemical characterization, Colloids Surf B: Biointerfaces, vol.83, pp.155-164, 2011.
DOI : 10.1016/j.colsurfb.2010.11.014

T. Gratieri, U. F. Schaefer, L. Jing, M. Gao, K. Kostka et al., Penetration of quantum dot particles through human skin, J Biomed Nanotechnol, vol.6, pp.586-95, 2010.
DOI : 10.1166/jbn.2010.1155

N. Vulovic, M. Primorac, M. Stupar, and J. L. Ford, Some studies into the properties of indomethacin suspensions intended for ophthalmic use, Int J Pharm, vol.55, pp.123-131, 1989.
DOI : 10.1016/0378-5173(89)90032-x

K. C. Gupta, A. Haider, Y. Choi, and I. Kang, Nanofibrous scaffolds in biomedical applications, Biomater Res, vol.18, p.5, 2014.
DOI : 10.1186/2055-7124-18-5

URL : https://biomaterialsres.biomedcentral.com/track/pdf/10.1186/2055-7124-18-5?site=biomaterialsres.biomedcentral.com

W. R. Braz, N. L. Rocha, E. H. De-faria, M. Silva, K. J. Ciuffi et al., Incorporation of anti-inflammatory agent into mesoporous silica, Nanotechnology, vol.27, issue.38, p.103, 2016.
DOI : 10.1088/0957-4484/27/38/385103

T. Elzein, M. Nasser-eddine, C. Delaite, S. Bistac, and P. Dumas, FTIR study of polycaprolactone chain organization at interfaces, J Colloid Interface Sci, vol.273, pp.381-388, 2004.
DOI : 10.1016/j.jcis.2004.02.001

H. Lin, G. Zhang, and S. Lin, Real-time co-crystal screening and formation between indomethacin and saccharin via DSC analytical technique or DSC-FTIR microspectroscopy, J Therm Anal Calorim, vol.120, pp.679-87, 2015.
DOI : 10.1007/s10973-014-3787-2

K. Tomoda, H. Terashima, K. Suzuki, T. Inagi, H. Terada et al., Enhanced transdermal delivery of indomethacin-loaded PLGA nanoparticles by iontophoresis, Colloids Surf B: Biointerfaces, vol.88, pp.706-716, 2011.
DOI : 10.1016/j.colsurfb.2011.08.004

, Drug Loaded Polymeric Nanoparticles 89

A. Ahmad, A. Husain, M. Mujeeb, S. A. Khan, A. K. Najmi et al., A review on therapeutic potential of Nigella sativa: a miracle herb, Asian Pac. J. Trop. Biomed, vol.3, pp.337-352, 2013.

F. Juhaimi, G. Figueredo, and P. Chalard, Comparison of chemical constituents of essential oils of black cumin (nigella sativa L.), Asian J. Chem, vol.25, p.10407, 2013.

S. H. Aljabre, O. M. Alakloby, and M. A. Randhawa, Dermatological effects of Nigella sativa, J. Dermatol. Dermatol. Surg, vol.19, pp.92-98, 2015.
DOI : 10.1016/j.jdds.2015.04.002

URL : https://doi.org/10.1016/j.jdds.2015.04.002

J. Ravindran, H. B. Nair, B. Sung, S. Prasad, R. R. Tekmal et al., Thymoquinone poly (lactide-co-glycolide) nanoparticles exhibit enhanced antiproliferative, anti-inflammatory, and chemosensitization potential, Biochem. Pharmacol, vol.79, pp.1640-1647, 2010.
DOI : 10.1016/j.bcp.2010.01.023

URL : http://europepmc.org/articles/pmc2846982?pdf=render

M. J. Hossen, W. S. Yang, D. Kim, A. Aravinthan, J. Kim et al., Thymoquinone: an IRAK1 inhibitor with in vivo and in vitro anti-inflammatory activities, Sci. Rep, vol.7, p.42995, 2017.

C. Sostres, C. J. Gargallo, M. T. Arroyo, and A. Lanas, Adverse effects of non-steroidal anti-inflammatory drugs (NSAIDs, aspirin and coxibs) on upper gastrointestinal tract, Best Pract. Res. Clin. Gastroenterol, vol.24, pp.121-132, 2010.
DOI : 10.1016/j.bpg.2009.11.005

J. A. Cordero, M. Camacho, R. Obach, J. Domenech, and L. Vila, In vitro based index of topical anti-inflammatory activity to compare a series of NSAIDs, Eur. J. Pharm. Biopharm. Off. J. Arbeitsgemeinschaft Pharm. Verfahrenstechnik EV, vol.51, pp.135-142, 2001.

J. A. Mitchell, P. Akarasereenont, C. Thiemermann, R. J. Flower, and J. R. Vane, Selectivity of nonsteroidal antiinflammatory drugs as inhibitors of constitutive and inducible cyclooxygenase, Proc. Natl. Acad. Sci. U. S. A, vol.90, pp.11693-11697, 1993.
DOI : 10.1073/pnas.90.24.11693

URL : http://www.pnas.org/content/90/24/11693.full.pdf

C. Reis, Ibuprofen nanoparticles for oral delivery: proof of concept, J. Nanomed. Biother. Discov, p.4, 2014.

C. Akermark and B. Forsskåhl, Topical indomethacin in overuse injuries in athletes. A randomized double-blind study comparing Elmetacin with oral indomethacin and placebo, Int. J. Sports Med, vol.11, pp.393-396, 1990.

V. Závi?ová, M. Koneracká, O. ?trbák, N. Toma?ovi?ová, P. Kop?anský et al., Encapsulation of indomethacin in magnetic biodegradable polymer nanoparticles, J. Magn. Magn Mater, vol.311, pp.379-382, 2007.

S. Lin, D. Wouessidjewe, M. Poelman, and D. Duchêne, In vivo evaluation of indomethacin/cyclodextrin complexes gastrointestinal tolerance and dermal antiinflammatory activity, Int. J. Pharm, vol.106, pp.63-67, 1994.

M. R. Prausnitz and R. Langer, Transdermal drug delivery, Nat. Biotechnol, vol.26, pp.1261-1268, 2008.

K. Tomoda, H. Terashima, K. Suzuki, T. Inagi, H. Terada et al., Enhanced transdermal delivery of indomethacin using combination of PLGA nanoparticles and iontophoresis in vivo, Colloids Surfaces B Biointerfaces, vol.92, pp.50-54, 2012.

M. Sala, A. Elaissari, and H. Fessi, Advances in psoriasis physiopathology and treatments: up to date of mechanistic insights and perspectives of novel therapies based on innovative skin drug delivery systems (ISDDS), J. Contr. Release, vol.239, pp.182-202, 2016.

M. A. Woodruff and D. W. Hutmacher, The return of a forgotten polymer-polycaprolactone in the 21st century, Prog. Polym. Sci, vol.35, pp.1217-1256, 2010.

W. Badri, K. Miladi, Q. A. Nazari, H. Fessi, and A. Elaissari, Effect of process and formulation parameters on polycaprolactone nanoparticles prepared by solvent displacement, Colloids Surf. Physicochem. Eng. Asp, vol.516, pp.238-244, 2017.
DOI : 10.1016/j.colsurfa.2016.12.029

W. Badri, K. Miladi, S. Robin, C. Viennet, Q. A. Nazari et al., Polycaprolactone based nanoparticles loaded with indomethacin for anti-inflammatory therapy: from preparation to Ex vivo study, Pharm. Res, vol.34, pp.1773-1783, 2017.
DOI : 10.1007/s11095-017-2166-7

A. Farhat, Vapo-diffusion assistée par micro-ondes ;: conception, optimisation et application, p.13, 2010.

Y. Li, A. Fabiano-tixier, and F. Chemat, Essential oils: from conventional to green extraction, Essent. Oils Reag. Green Chem, pp.9-20, 2014.
DOI : 10.1007/978-3-319-08449-7_2

N. Bousbia, M. A. Vian, M. A. Ferhat, B. Y. Meklati, and F. Chemat, A new process for extraction of essential oil from Citrus peels: microwave hydrodiffusion and gravity, J. Food Eng, vol.90, pp.409-413, 2009.

H. Fessi, F. Puisieux, J. P. Devissaguet, N. Ammoury, and S. Benita, Nanocapsule formation by interfacial polymer deposition following solvent displacement, Int. J. Pharm, vol.55, pp.1-4, 1989.
DOI : 10.1016/0378-5173(89)90281-0

A. E. Asbahani, A. Jilale, S. N. Voisin, E. Habib, A. Addi et al., Chemical composition and antimicrobial activity of nine essential oils obtained by steam distillation of plants from the Souss-Massa Region (Morocco), J. Essent. Oil Res, vol.27, pp.34-44, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01230130

E. A. Abdelhafed, K. Miladi, H. Addi, A. Bitar, H. Casabianca et al., Antimicrobial activity of nano-encapsulated essential oils: comparison to non-encapsulated essential oils, J. Colloid Sci. Biotechnol, vol.4, pp.39-48, 2015.

M. Bendahou, A. Muselli, M. Grignon-dubois, M. Benyoucef, J. Desjobert et al., Antimicrobial activity and chemical composition of Origanum glandulosum Desf. essential oil and extract obtained by microwave extraction: comparison with hydrodistillation, Food Chem, vol.106, pp.132-139, 2008.
DOI : 10.1016/j.foodchem.2007.05.050

URL : https://hal.archives-ouvertes.fr/hal-00449572

F. Benkaci-ali, A. Baaliouamer, B. Y. Meklati, and F. Chemat, Chemical composition of seed essential oils from Algerian Nigella sativa extracted by microwave and hydrodistillation, Flavour Fragrance J, vol.22, pp.148-153, 2007.

M. Hazzit, A. Baaliouamer, A. R. Veríssimo, M. L. Faleiro, and M. G. Miguel, Chemical composition and biological activities of Algerian Thymus oils, Food Chem, vol.116, pp.714-721, 2009.
DOI : 10.1016/j.foodchem.2009.03.018

A. J. Poulose and R. Croteau, Biosynthesis of aromatic monoterpenes: conversion of ?terpinene to p-cymene and thymol in Thymus vulgaris L, Arch. Biochem. Biophys, vol.187, pp.307-314, 1978.

Z. Gholamnezhad, S. Havakhah, and M. H. Boskabady, Preclinical and clinical effects of Nigella sativa and its constituent, thymoquinone: a review, J. Ethnopharmacol, vol.190, pp.372-386, 2016.
DOI : 10.1016/j.jep.2016.06.061

Z. T. Rushmi, D. M. Shariare, N. Akter, R. J. Mow, and M. Afroz, Preparation and Characterization of Black Seed Oil Loaded Liposomes, 2014.

K. Adibkia, Y. Javadzadeh, S. Dastmalchi, G. Mohammadi, F. K. Niri et al., Naproxen-eudragit RS100 nanoparticles: preparation and physicochemical characterization, vol.83, pp.155-159, 2011.
DOI : 10.1016/j.colsurfb.2010.11.014

A. Rohman and R. Ariani, Authentication of nigella sativa seed oil in binary and ternary mixtures with corn oil and soybean oil using FTIR spectroscopy coupled with partial least square, Sci. World J, vol.2013, p.740142, 2013.

A. A. Doolaanea, A. F. Ismail, and F. Mohamed, Quantification of nigella sativa oil (NSO) from biodegradable PLGA nanoparticles using FTIR spectroscopy, Int. J. Pharm. Pharmaceut. Sci, vol.6, pp.228-232, 2014.

K. C. Gupta, A. Haider, Y. Choi, and I. Kang, Nanofibrous scaffolds in biomedical applications, vol.18, p.5, 2014.
DOI : 10.1186/2055-7124-18-5

URL : https://biomaterialsres.biomedcentral.com/track/pdf/10.1186/2055-7124-18-5?site=biomaterialsres.biomedcentral.com

N. Vulovic, M. Primorac, M. Stupar, and J. L. Ford, Some studies into the properties of indomethacin suspensions intended for ophthalmic use, Int. J. Pharm, vol.55, pp.123-128, 1989.

W. R. Braz, N. L. Rocha, E. H. De-faria, M. L. Silva, K. J. Ciuffi et al., Incorporation of anti-inflammatory agent into mesoporous silica, Nanotechnology, vol.27, p.385103, 2016.
DOI : 10.1088/0957-4484/27/38/385103

T. Elzein, M. Nasser-eddine, C. Delaite, S. Bistac, and P. Dumas, FTIR study of polycaprolactone chain organization at interfaces, J. Colloid Interface Sci, vol.273, pp.381-387, 2004.
DOI : 10.1016/j.jcis.2004.02.001

K. S. Yadav and K. K. Sawant, Formulation optimization of etoposide loaded PLGA nanoparticles by double factorial design and their evaluation, Curr. Drug Deliv, vol.7, pp.51-64, 2010.
DOI : 10.2174/156720110790396517

C. E. Mora-huertas, H. Fessi, and A. Elaissari, Polymer-based nanocapsules for drug delivery, Int. J. Pharm, vol.385, pp.113-142, 2010.
DOI : 10.1016/j.ijpharm.2009.10.018

W. Badri, , pp.2320-5407

, All rights reserved, NSSEO. Copy Right, IJAR, vol.6, issue.4, pp.801-816, 2018.

?. .. , Introduction:ISSN, pp.2320-5407

, Int. J. Adv. Res, vol.6, issue.4, pp.801-816

. Figure7, Scale bars are 5?m. (A): control or untreated skin with colloidal dispersion, (B): mixture of 25 CSE and distilled water, nanoparticles loaded with indomethacin and CSE, and (D): nanoparticles loaded with indomethacin

H. Ahmad, K. Kumar, M. A. Rahman, M. M. Rahman, M. A. Miah et al., Preparation and characterization of conducting polyaniline layered magnetic nano composite polymer particles, Polym. Adv. Technol. 2013, vol.24, pp.740-746

A. Juhaimi, F. Figueredo, G. Chalard, and P. , Comparison of Chemical Constituents of Essential Oils of Black Cumin (Nigella sativa L.), Asian J. Chem, 2013.

R. Alvarez-román, A. Naik, Y. N. Kalia, R. H. Guy, and H. Fessi, Skin penetration and distribution of polymeric nanoparticles, J. Controlled Release2004, vol.99, pp.53-62

W. Badri, K. Miladi, Q. A. Nazari, H. Fessi, and A. Elaissari, Effect of process and formulation parameters on polycaprolactone nanoparticles prepared by solvent displacement, Colloids Surf. Physicochem. Eng. Asp, vol.516, pp.238-244, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02092748

H. A. Benson, Transdermal drug delivery: penetration enhancement techniques, Curr. Drug Deliv, vol.2, pp.23-33, 2005.

, Int. J. Adv. Res, vol.6, issue.4, pp.801-816

M. Bolzinger, S. Briançon, J. Pelletier, and Y. Chevalier, Penetration of drugs through skin, a complex rate-controlling membrane, Curr. Opin. Colloid Interface Sci. 2012, vol.17, pp.156-165
URL : https://hal.archives-ouvertes.fr/hal-02014114

J. Comer, S. Judge, D. Matthews, L. Towers, B. Falcone et al., The intrinsic aqueous solubility of indomethacin. ADMET DMPK2014, vol.2, pp.18-32

P. Desai, R. R. Patlolla, and M. Singh, Interaction of nanoparticles and cell-penetrating peptides with skin for transdermal drug delivery, Mol. Membr. Biol, vol.27, pp.247-259, 2010.

L. Ferrero-miliani, O. H. Nielsen, P. S. Andersen, and S. E. Girardin, Chronic inflammation: importance of NOD2 and NALP3 in interleukin-1beta generation, Clin. Exp. Immunol, vol.147, pp.227-235, 2007.

H. Fessi, F. Puisieux, J. P. Devissaguet, N. Ammoury, and S. Benita, Nanocapsule formation by interfacial polymer deposition following solvent displacement, Int. J. Pharm, vol.55, pp.1-4, 1989.

G. Garrido, D. González, Y. Lemus, D. García, L. Lodeiro et al., In vivo and in vitro anti-inflammatory activity of Mangifera indica L. extract (VIMANG), Pharmacol. Res, vol.50, pp.143-149, 2004.

P. G. Gunasekar, J. V. Rogers, M. B. Kabbur, C. M. Garrett, W. W. Brinkley et al., Molecular and histological responses in rat skin exposed to m-xylene, J. Biochem. Mol. Toxicol, vol.17, pp.92-94, 2003.

S. Y. Kim, Y. M. Lee, H. J. Shin, and J. S. Kang, Indomethacin-loaded methoxy poly(ethylene glycol)/ poly(epsilon-caprolactone) diblock copolymeric nanosphere: pharmacokinetic characteristics of indomethacin in the normal Sprague-Dawley rats, Biomaterials, vol.22, pp.2049-2056, 2001.

G. L. Li, T. J. Van-steeg, H. Putter, . Van-der, J. Spek et al., Cutaneous side-effects of transdermal iontophoresis with and without surfactant pretreatment: a single-blinded, randomized controlled trial, Br. J. Dermatol, vol.153, pp.404-412, 2005.

L. Maia-de-oliveira, M. Sousa-nunes-pinheiro, D. C. Oliveira-bezerra, B. M. Oliveira-leite, L. Rocha-tomé et al., Topical Anti-inflammatory Potential of Pumpkin (Cucurbita pepo L.) Seed Oil on Acute and Chronic Skin Inflammation in Mice, Acta Sci. Vet, p.41, 2013.

S. Lin, D. Wouessidjewe, M. Poelman, and D. Duchêne, In vivo evaluation of indomethacin/ cyclodextrin complexes gastrointestinal tolerance and dermal anti-inflammatory activity, Int. J. Pharm, vol.106, pp.63-67, 1994.

R. Medzhitov, N. Nagai, C. Yoshioka, and Y. Ito, Topical therapies for rheumatoid arthritis by gel ointments containing indomethacin nanoparticles in adjuvant-induced arthritis rat, J. Oleo Sci, vol.454, pp.337-346, 2008.

V. A. Pinheiro, D. Serikaku, A. R. Baby, M. V. Velasco, T. M. Kaneko et al., Development of ciclopirox olamine topical formulations: evaluation of drug release, penetration and cutaneous retention, Pharm. Dev. Technol, vol.20, pp.197-203, 2015.

S. R. Pygall, J. Whetstone, P. Timmins, and C. D. Melia, Pharmaceutical applications of confocal laser scanning microscopy: The physical characterisation of pharmaceutical systems, Adv. Drug Deliv. Rev, vol.59, pp.1434-1452, 2007.

J. Ravindran, H. B. Nair, B. Sung, S. Prasad, R. R. Tekmal et al., Thymoquinone Poly(lactide-co-glycolide) Nanoparticles Exhibit Enhanced Anti-proliferative, Anti-inflammatory, and Chemosensitization Potential, Biochem. Pharmacol, vol.79, pp.1640-1647, 2010.

H. Sadeghi, V. Zarezade, . Sadeghi, A. Hossein, M. Toori et al., Anti-inflammatory Activity of Stachys Pilifera Benth. Iran. Red Crescent Med, J, p.16, 2014.

M. Sala, R. Diab, A. Elaissari, and H. Fessi, Lipid nanocarriers as skin drug delivery systems: Properties, mechanisms of skin interactions and medical applications, Int. J. Pharm, vol.535, pp.1-17, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02093089

S. Sarkhel, Evaluation of the anti-inflammatory activities of Quillaja saponaria Mol. saponin extract in mice, Toxicol. Rep, vol.3, pp.1-3, 2016.

, Int. J. Adv. Res, vol.6, issue.4, pp.801-816

A. M. Shawesh, A. Kaukonen, S. Kallioinen, O. Antikainen, and J. Yliruusi, Development of indomethacin Carbopol ETD 2001 gels and the influence of storage time and temperature on their stability. Pharm, vol.58, pp.130-135, 2003.

N. Z. Yassin, S. M. El-shenawy, R. F. Abdel-rahman, M. Yakoot, M. Hassan et al., Effect of a topical copper indomethacin gel on inflammatory parameters in a rat model of osteoarthritis, Drug Des. Devel. Ther, vol.9, pp.1491-1498, 2015.

, Encapsulation of NSAIDs for inflammation management: Overview, progress, challenges and prospects

W. Badri, K. Miladi, Q. Agha-nazari, H. Greige-gerges, and H. Fessi, Abdelhamid Elaissari, 2016.

N. Zafar, W. Badri, S. A. Galindo-rodríguez, D. Kabbaj, and H. Fessi, Plant extracts: from encapsulation to application. Brenda Armendáriz Barragán, Abdelhamid Elaissari, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02002236

. , From encapsulation to drug delivery. Claudia Janeth Martínez Rivas, 2017.

, Effect of process and formulation parameters on polycaprolactone nanoparticles prepared by solvent displacement, 2016.

, Polycaprolactone Based Nanoparticles Loaded with Indomethacin for Anti-Inflammatory Therapy: From Preparation to Ex Vivo Study, 2017.

, Poly (?-caprolactone) nanoparticles loaded with indomethacin and Nigella Sativa L. essential oil for the topical treatment of inflammation, Inflammation topical enhanced treatment, 2018.

N. Indomethacin and L. Sativa, Essential oil inpoly-caprolactone nanoparticles: In Vivo study of anti-inflammatory activity, 2018.