Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Pharmaceutical and Pharmacokinetic Aspects of Nanoformulation Based Drug Delivery Systems for Anti-cancer Drugs

Author(s): Arun Kumar Singh, Shiv Bahadur*, Deepika Yadav and Hunny Dabas

Volume 29, Issue 24, 2023

Published on: 29 August, 2023

Page: [1896 - 1906] Pages: 11

DOI: 10.2174/1381612829666230824144727

Price: $65

Abstract

Many nanodrug delivery systems used with various routes of administration have been developed recently. These may be dendrimers, nanocrystals, emulsions, liposomes, solid lipid nanoparticles, micelles, or polymeric nanoparticles. The nanodrug delivery systems may improve effectiveness, safety, physicochemical qualities, and pharmacokinetic/pharmacodynamic profile. Functionalized nanodrug delivery systems can increase the half-life, improve the bioavailability of orally administered pharmaceuticals, and target tissue distribution. By decreasing the number of dosage intervals required, increasing the magnitude of the intended pharmacological effects, and decreasing the severity of undesirable systemic side effects, nanodrug systems show promise for improving treatment adherence and clinical results. Nanodrugs have been demonstrated to exhibit cytotoxicity, oxidative stress, inflammation, and genotoxicity in vitro and in vivo; however, this attention has recently been refocused on their potentially harmful potential owing to their beneficial pharmacokinetic features for the treatment of cancer. Researchers require a more profound knowledge of the pharmacokinetic and safety aspects of nanodrugs and the limits of each administration route to continue creating safe and efficacious nanodrugs with high therapeutic potential. The benefits and risks associated with pharmacokinetics have been highlighted in this article, which describes the current state of nanodrug system development.

Keywords: Nanodrug, pharmacokinetic, cytotoxicity, dendrimers, nanoparticles, cancer.

[1]
Arnold J, Kilmartin D, Olson J, Neville S, Robinson K, Laird A. Verteporfin therapy of subfoveal choroidal neovascularization in age-related macular degeneration: Two-year results of a randomized clinical trial including lesions with occult with no classic choroidal neovascularization-verteporfin in photodynamic therapy report 2. Am J Ophthalmol 2001; 131(5): 541-60.
[http://dx.doi.org/10.1016/S0002-9394(01)00967-9] [PMID: 11336929]
[2]
Asthana A, Chauhan AS, Diwan PV, Jain NK. Poly(amidoamine) (PAMAM) dendritic nanostructures for controlled sitespecific delivery of acidic anti-inflammatory active ingredient. AAPS PharmSciTech 2005; 6(3): E536-42.
[http://dx.doi.org/10.1208/pt060367] [PMID: 16354015]
[3]
Barenholz YC. Doxil® - The first FDA-approved nano-drug: Lessons learned. J Control Release 2012; 160(2): 117-34.
[http://dx.doi.org/10.1016/j.jconrel.2012.03.020] [PMID: 22484195]
[4]
Benbrook DM. Biotechnology and biopharmaceuticals: Transforming proteins and genes into drugs (2nd Edition.), 2015.
[5]
Berges R, Eligard R. Pharmacokinetics, effect on testosterone and PSA levels and tolerability. Eur Urol Suppl 2005; 4(5): 20-5.
[http://dx.doi.org/10.1016/j.eursup.2005.04.001]
[6]
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(10): 2373-87.
[http://dx.doi.org/10.1007/s11095-016-1958-5] [PMID: 27299311]
[7]
Bur M, Henning A, Hein S, Schneider M, Lehr CM. Inhalative nanomedicine-opportunities and challenges. Inhal Toxicol 2009; 21(S1): 137-43.
[http://dx.doi.org/10.1080/08958370902962283] [PMID: 19558246]
[8]
Dawidczyk CM, Russell LM, Searson PC. Nanomedicines for cancer therapy: State-of-the-art and limitations to pre-clinical studies that hinder future developments. Front Chem 2014; 2: 69.
[http://dx.doi.org/10.3389/fchem.2014.00069] [PMID: 25202689]
[9]
Chaturvedi K, Ganguly K, Nadagouda MN, Aminabhavi TM. Polymeric hydrogels for oral insulin delivery. J Control Release 2013; 165(2): 129-38.
[http://dx.doi.org/10.1016/j.jconrel.2012.11.005] [PMID: 23159827]
[10]
Dawidczyk CM, Kim C, Park JH, et al. State-of-the-art in design rules for drug delivery platforms: Lessons learned from FDA-approved nanomedicines. J Control Release 2014; 187: 133-44.
[http://dx.doi.org/10.1016/j.jconrel.2014.05.036] [PMID: 24874289]
[11]
de Jong WH, Borm PJ. Drug delivery and nanoparticles: Applications and hazards. Int J Nanomedicine 2008; 3(2): 133-49.
[http://dx.doi.org/10.2147/IJN.S596] [PMID: 18686775]
[12]
des Rieux A, Fievez V, Garinot M, Schneider YJ, Préat V. Nanoparticles as potential oral delivery systems of proteins and vaccines: A mechanistic approach. J Control Release 2006; 116(1): 1-27.
[http://dx.doi.org/10.1016/j.jconrel.2006.08.013] [PMID: 17050027]
[13]
Desai N, Trieu V, Yao Z, et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res 2006; 12(4): 1317-24.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-1634] [PMID: 16489089]
[14]
Devalapally H, Chakilam A, Amiji MM. Role of nanotechnology in pharmaceutical product development. J Pharm Sci 2007; 96(10): 2547-65.
[http://dx.doi.org/10.1002/jps.20875] [PMID: 17688284]
[15]
Draca N, Lazic R, Simic P, Dumic-Cule I, Luetic AT, Gabric N. Potential beneficial role of sevelamer hydrochloride in diabetic retinopathy. Med Hypotheses 2013; 80(4): 431-5.
[http://dx.doi.org/10.1016/j.mehy.2012.12.035] [PMID: 23357670]
[16]
Duncan R. Polymer therapeutics: Top 10 selling pharmaceuticals - What next? J Control Release 2014; 190: 371-80.
[http://dx.doi.org/10.1016/j.jconrel.2014.05.001] [PMID: 24818766]
[17]
Ehmann F, Sakai-Kato K, Duncan R, et al. Next-generation nanomedicines and nanosimilars: EU regulators’ initiatives relating to the development and evaluation of nanomedicines. Nanomedicine 2013; 8(5): 849-56.
[http://dx.doi.org/10.2217/nnm.13.68] [PMID: 23656268]
[18]
Eifler AC, Thaxton CS. Nanoparticle therapeutics: FDA approval, clinical trials, regulatory pathways, and case study. Methods Mol Biol 2011; 726: 325-38.
[http://dx.doi.org/10.1007/978-1-61779-052-2_21] [PMID: 21424459]
[19]
European Food Safety Authority. Guidance on the risk assessment of the application of nanoscience and nanotechnologies in the food and feed chain. EFSA J 2011; 9: 2140.
[http://dx.doi.org/10.2903/j.efsa.2018.5327]
[20]
European Medicines Agency. 2013. Reflection paper on surface coatings: General issues for consideration regarding parenteral administration of coated nanomedicine products. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2013/08/WC500147874.pdf
[21]
European Medicines Agency. Reflection paper on the development of 5 block copolymer micelle medicinal products. 2013. Available at: http://www.ema.europa.eu/docs/en_GB/document_ library/Scientific_guideline/2013/02/WC500138390.pdf
[23]
European Medicines Agency. Reflection paper on the data requirements for intravenous iron-based nano-colloidal products developed with reference to an innovator medicinalproduct. 2015. Available at: http://www.ema.europa.eu/docs/en_GB/document_lib- rary/Scientific_guideline/2015/03/WC500184922.pdf
[24]
Fanciullino R, Ciccolini J, Milano G. Challenges, expectations and limits for nanoparticles-based therapeutics in cancer: A focus on nano-albumin-bound drugs. Crit Rev Oncol Hematol 2013; 88(3): 504-13.
[http://dx.doi.org/10.1016/j.critrevonc.2013.06.010] [PMID: 23871532]
[25]
Choi HS, Frangioni JV. Nanoparticles for biomedical imaging: Fundamentals of clinical translation. Mol Imaging 2010; 9(6): 7290.2010.00031.
[http://dx.doi.org/10.2310/7290.2010.00031] [PMID: 21084027]
[26]
Wick P, Manser P, Limbach L, et al. The degree and kind of agglomeration affect carbon nanotube cytotoxicity. Toxicol Lett 2007; 168(2): 121-31.
[http://dx.doi.org/10.1016/j.toxlet.2006.08.019] [PMID: 17169512]
[27]
Stern ST, McNeil SE. Nanotechnology safety concerns revisited. Toxicol Sci 2008; 101(1): 4-21.
[http://dx.doi.org/10.1093/toxsci/kfm169] [PMID: 17602205]
[28]
Liu M, Fréchet JMJ. Designing dendrimers for drug delivery. Pharm Sci Technol Today 1999; 2(10): 393-401.
[http://dx.doi.org/10.1016/S1461-5347(99)00203-5] [PMID: 10498919]
[29]
Honda M, Asai T, Oku N, Araki Y, Tanaka M, Ebihara N. Liposomes and nanotechnology in drug development: Focus on ocular targets. Int J Nanomedicine 2013; 8: 495-503.
[http://dx.doi.org/10.2147/IJN.S30725] [PMID: 23439842]
[30]
Kaminskas LM, McLeod VM, Kelly BD, et al. A comparison of changes to doxorubicin pharmacokinetics, antitumor activity, and toxicity mediated by PEGylated dendrimer and PEGylated liposome drug delivery systems. Nanomedicine 2012; 8(1): 103-11.
[http://dx.doi.org/10.1016/j.nano.2011.05.013] [PMID: 21704192]
[31]
Maeda H, Sawa T, Konno T. Mechanism of tumor-targeted delivery of macromolecular drugs, including the EPR effect in solid tumor and clinical overview of the prototype polymeric drug SMANCS. J Control Release 2001; 74(1-3): 47-61.
[http://dx.doi.org/10.1016/S0168-3659(01)00309-1] [PMID: 11489482]
[32]
Kaminskas LM, Kelly BD, McLeod VM, et al. Capping methotrexate α-carboxyl groups enhances systemic exposure and retains the cytotoxicity of drug conjugated PEGylated polylysine dendrimers. Mol Pharm 2011; 8(2): 338-49.
[http://dx.doi.org/10.1021/mp1001872] [PMID: 21171585]
[33]
Kurmi BD, Gajbhiye V, Kayat J, Jain NK. Lactoferrin-conjugated dendritic nanoconstructs for lung targeting of methotrexate. J Pharm Sci 2011; 100(6): 2311-20.
[http://dx.doi.org/10.1002/jps.22469] [PMID: 21491447]
[34]
Prajapati RN, Tekade RK, Gupta U, Gajbhiye V, Jain NK. Dendimer-mediated solubilization, formulation development and in vitro-in vivo assessment of piroxicam. Mol Pharm 2009; 6(3): 940-50.
[http://dx.doi.org/10.1021/mp8002489] [PMID: 19231841]
[35]
Kaminskas LM, Porter CJH. Targeting the lymphatics using dendritic polymers (dendrimers). Adv Drug Deliv Rev 2011; 63(10-11): 890-900.
[http://dx.doi.org/10.1016/j.addr.2011.05.016] [PMID: 21683746]
[36]
Ziemba B, Matuszko G, Bryszewska M, Klajnert B. Influence of dendrimers on red blood cells. Cell Mol Biol Lett 2012; 17(1): 21-35.
[http://dx.doi.org/10.2478/s11658-011-0033-9] [PMID: 22086186]
[37]
Kawabata Y, Wada K, Nakatani M, Yamada S, Onoue S. Formulation design for poorly water-soluble drugs based on biopharmaceutics classification system: Basic approaches and practical applications. Int J Pharm 2011; 420(1): 1-10.
[http://dx.doi.org/10.1016/j.ijpharm.2011.08.032] [PMID: 21884771]
[38]
Gao L, Liu G, Ma J, Wang X, Zhou L, Li X. Drug nanocrystals: In vivo performances. J Control Release 2012; 160(3): 418-30.
[http://dx.doi.org/10.1016/j.jconrel.2012.03.013] [PMID: 22465393]
[39]
Chan HK, Kwok PCL. Production methods for nanodrug particles using the bottom-up approach. Adv Drug Deliv Rev 2011; 63(6): 406-16.
[http://dx.doi.org/10.1016/j.addr.2011.03.011] [PMID: 21457742]
[40]
Jia L, Wong H, Wang Y, Garza M, Weitman SD. Carbendazim: disposition, cellular permeability, metabolite identification, and pharmacokinetic comparison with its nanoparticle. J Pharm Sci 2003; 92(1): 161-72.
[http://dx.doi.org/10.1002/jps.10272] [PMID: 12486692]
[41]
Jinno J, Kamada N, Miyake M, et al. Effect of particle size reduction on dissolution and oral absorption of a poorly water-soluble drug, cilostazol, in beagle dogs. J Control Release 2006; 111(1-2): 56-64.
[http://dx.doi.org/10.1016/j.jconrel.2005.11.013] [PMID: 16410029]
[42]
Onoue S, Takahashi H, Kawabata Y, et al. Formulation design and photochemical studies on nanocrystal solid dispersion of curcumin with improved oral bioavailability. J Pharm Sci 2010; 99(4): 1871-81.
[http://dx.doi.org/10.1002/jps.21964] [PMID: 19827133]
[43]
Wu CY, Benet LZ. Predicting drug disposition via application of BCS: Transport/absorption/ elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res 2005; 22(1): 11-23.
[http://dx.doi.org/10.1007/s11095-004-9004-4] [PMID: 15771225]
[44]
Hanafy A, Spahnlangguth H, Vergnault G, et al. Pharmacokinetic evaluation of oral fenofibrate nanosuspensions and SLN in comparison to conventional suspensions of micronized drug. Adv Drug Deliv Rev 2007; 59(6): 419-26.
[http://dx.doi.org/10.1016/j.addr.2007.04.005] [PMID: 17566595]
[45]
Sylvestre JP, Tang MC, Furtos A, Leclair G, Meunier M, Leroux JC. Nanonization of megestrol acetate by laser fragmentation in aqueous milieu. J Control Release 2011; 149(3): 273-80.
[http://dx.doi.org/10.1016/j.jconrel.2010.10.034] [PMID: 21047539]
[46]
Xia D, Cui F, Piao H, et al. Effect of crystal size on the in vitro dissolution and oral absorption of nitrendipine in rats. Pharm Res 2010; 27(9): 1965-76.
[http://dx.doi.org/10.1007/s11095-010-0200-0] [PMID: 20585842]
[47]
Onoue S, Nakamura T, Uchida A, et al. Physicochemical and biopharmaceutical characterization of amorphous solid dispersion of nobiletin, a citrus polymethoxylated flavone, with improved hepatoprotective effects. Eur J Pharm Sci 2013; 49(4): 453-60.
[http://dx.doi.org/10.1016/j.ejps.2013.05.014] [PMID: 23707470]
[48]
Kawabata Y, Yamamoto K, Debari K, Onoue S, Yamada S. Novel crystalline solid dispersion of tranilast with high photostability and improved oral bioavailability. Eur J Pharm Sci 2010; 39(4): 256-62.
[http://dx.doi.org/10.1016/j.ejps.2009.12.009] [PMID: 20038453]
[49]
Onoue S, Aoki Y, Kawabata Y, et al. Development of inhalable nanocrystalline solid dispersion of tranilast for airway inflammatory diseases. J Pharm Sci 2011; 100(2): 622-33.
[http://dx.doi.org/10.1002/jps.22299] [PMID: 20653048]
[50]
Manvelian G, Daniels S, Gibofsky A. The pharmacokinetic parameters of a single dose of a novel nano-formulated, lower-dose oral diclofenac. Postgrad Med 2012; 124(1): 117-23.
[http://dx.doi.org/10.3810/pgm.2012.01.2524] [PMID: 22314121]
[51]
Manvelian G, Daniels S, Altman R. A phase I study evaluating the pharmacokinetic profile of a novel, proprietary, nano-formulated, lower-dose oral indomethacin. Postgrad Med 2012; 124(4): 1-9.
[http://dx.doi.org/10.3810/pgm.2012.07.2580] [PMID: 22913908]
[52]
Feldman EJ, Kolitz JE, Trang JM, et al. Pharmacokinetics of CPX-351: A nano-scale liposomal fixed molar ratio formulation of cytarabine: daunorubicin, in patients with advanced leukemia. Leuk Res 2012; 36(10): 1283-9.
[http://dx.doi.org/10.1016/j.leukres.2012.07.006] [PMID: 22840315]
[53]
Fetterly GJ, Straubinger RM. Pharmacokinetics of paclitaxel-containing liposomes in rats. AAPS PharmSci 2003; 5(4): 90-100.
[http://dx.doi.org/10.1208/ps050432] [PMID: 15198520]
[54]
Foss F. Clinical experience with denileukin diftitox (ONTAK). Semin Oncol 2006; 33(1): 11-6.
[http://dx.doi.org/10.1053/j.seminoncol.2005.12.017] [PMID: 16516670]
[55]
Foss FM, Sjak-Shie N, Goy A, et al. A multicenter phase II trial to determine the safety and efficacy of combination therapy with denileukin diftitox and cyclophosphamide, doxorubicin, vincristine and prednisone in untreated peripheral T-cell lymphoma: The CONCEPT study. Leuk Lymphoma 2013; 54(7): 1373-9.
[http://dx.doi.org/10.3109/10428194.2012.742521] [PMID: 23278639]
[56]
Francis MF, Cristea M, Winnik FM. Exploiting the vitamin B12 pathway to enhance oral drug delivery via polymeric micelles. Biomacromolecules 2005; 6(5): 2462-7.
[http://dx.doi.org/10.1021/bm0503165] [PMID: 16153081]
[57]
Fuentes AC, Szwed E, Spears CD, Thaper S, Dang LH, Dang NH. Denileukin diftitox (Ontak) as maintenance therapy for peripheral T-Cell lymphomas: Three cases with sustained remission. Case Rep Oncol Med 2015; 2015: 1-5.
[http://dx.doi.org/10.1155/2015/123756] [PMID: 26240767]
[58]
Green MR, Manikhas GM, Orlov S, et al. Abraxane®, a novel Cremophor®-free, albumin-bound particle form of paclitaxel for the treatment of advanced non-small-cell lung cancer. Ann Oncol 2006; 17(8): 1263-8.
[http://dx.doi.org/10.1093/annonc/mdl104] [PMID: 16740598]
[59]
Hafner A, Lovrić J, Lakoš GP, Pepić I. Nanotherapeutics in the EU: An overview on current state and future directions. Int J Nanomedicine 2014; 9: 1005-23.
[PMID: 24600222]
[60]
Hann IM, Prentice HG. Lipid-based amphotericin B: A review of the last 10 years of use. Int J Antimicrob Agents 2001; 17(3): 161-9.
[http://dx.doi.org/10.1016/S0924-8579(00)00341-1] [PMID: 11282260]
[61]
Hrkach J, Von Hoff D, Ali MM, et al. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile. Sci Transl Med 2012; 4(128): 128ra39.
[http://dx.doi.org/10.1126/scitranslmed.3003651] [PMID: 22491949]
[62]
Hu X, Miller L, Richman S, et al. A novel PEGylated interferon beta-1a for multiple sclerosis: Safety, pharmacology, and biology. J Clin Pharmacol 2012; 52(6): 798-808.
[http://dx.doi.org/10.1177/0091270011407068] [PMID: 21680782]
[63]
Ing M, Gupta N, Teyssandier M, et al. Immunogenicity of long-lasting recombinant factor VIII products. Cell Immunol 2016; 301: 40-8.
[http://dx.doi.org/10.1016/j.cellimm.2015.12.006] [PMID: 26723503]
[64]
Jain A, Jain SK. In vitro and cell uptake studies for targeting of ligand anchored nanoparticles for colon tumors. Eur J Pharm Sci 2008; 35(5): 404-16.
[http://dx.doi.org/10.1016/j.ejps.2008.08.008] [PMID: 18824095]
[65]
James ND, Coker RJ, Tomlinson D, et al. Liposomal doxorubicin (Doxil): An effective new treatment for Kaposi’s sarcoma in AIDS. Clin Oncol 1994; 6(5): 294-6.
[http://dx.doi.org/10.1016/S0936-6555(05)80269-9] [PMID: 7530036]
[66]
Johnson KP, Brooks BR, Cohen JA, et al. Extended use of glatiramer acetate (Copaxone) is well tolerated and maintains its clinical effect on multiple sclerosis relapse rate and degree of disability. Neurology 1998; 50(3): 701-8.
[http://dx.doi.org/10.1212/WNL.50.3.701] [PMID: 9521260]
[67]
Kato K, Chin K, Yoshikawa T, et al. Phase II study of NK105, a paclitaxel-incorporating micellar nanoparticle, for previously treated advanced or recurrent gastric cancer. Invest New Drugs 2012; 30(4): 1621-7.
[http://dx.doi.org/10.1007/s10637-011-9709-2] [PMID: 21728023]
[68]
Larsen AT, Ohlsson AG, Polentarutti B, et al. Oral bioavailability of cinnarizine in dogs: Relation to SNEDDS droplet size, drug solubility and in vitro precipitation. Eur J Pharm Sci 2013; 48(1-2): 339-50.
[http://dx.doi.org/10.1016/j.ejps.2012.11.004] [PMID: 23178440]
[69]
Lawrence MJ, Rees GD. Microemulsion-based media as novel drug delivery systems. Adv Drug Deliv Rev 2000; 45(1): 89-121.
[http://dx.doi.org/10.1016/S0169-409X(00)00103-4] [PMID: 11104900]
[70]
Ling H, Luoma JT, Hilleman D. A review of currently available fenofibrate and fenofibric acid formulations. Cardiol Res 2013; 4(2): 47-55.
[http://dx.doi.org/10.4021/cr270w] [PMID: 28352420]
[71]
Liu W, Yang XL, Winston Ho WS. Preparation of uniform-sized multiple emulsions and micro/nano particulates for drug delivery by membrane emulsification. J Pharm Sci 2011; 100(1): 75-93.
[http://dx.doi.org/10.1002/jps.22272] [PMID: 20589949]
[72]
Manjunath K, Venkateswarlu V. Pharmacokinetics, tissue distribution and bioavailability of clozapine solid lipid nanoparticles after intravenous and intraduodenal administration. J Control Release 2005; 107(2): 215-28.
[http://dx.doi.org/10.1016/j.jconrel.2005.06.006] [PMID: 16014318]
[73]
Matsumura Y, Hamaguchi T, Ura T, et al. Phase I clinical trial and pharmacokinetic evaluation of NK911, a micelle-encapsulated doxorubicin. Br J Cancer 2004; 91(10): 1775-81.
[http://dx.doi.org/10.1038/sj.bjc.6602204] [PMID: 15477860]
[74]
May JP, Li SD. Hyperthermia-induced drug targeting. Expert Opin Drug Deliv 2013; 10(4): 511-27.
[http://dx.doi.org/10.1517/17425247.2013.758631] [PMID: 23289519]
[75]
Mora-Huertas CE, Fessi H, Elaissari A. Polymer-based nanocapsules for drug delivery. Int J Pharm 2010; 385(1-2): 113-42.
[http://dx.doi.org/10.1016/j.ijpharm.2009.10.018] [PMID: 19825408]
[76]
Morgen M, Bloom C, Beyerinck R, et al. Polymeric nanoparticles for increased oral bioavailability and rapid absorption using celecoxib as a model of a low-solubility, high-permeability drug. Pharm Res 2012; 29(2): 427-40.
[http://dx.doi.org/10.1007/s11095-011-0558-7] [PMID: 21863477]
[77]
Mori S, Matsuura A, Rama Prasad YV, Takada K. Studies on the intestinal absorption of low molecular weight heparin using saturated fatty acids and their derivatives as an absorption enhancer in rats. Biol Pharm Bull 2004; 27(3): 418-21.
[http://dx.doi.org/10.1248/bpb.27.418] [PMID: 14993814]
[78]
Möschwitzer J, Müller RH. New method for the effective production of ultrafine drug nanocrystals. J Nanosci Nanotechnol 2006; 6(9): 3145-53.
[http://dx.doi.org/10.1166/jnn.2006.480] [PMID: 17048530]
[79]
Onoue S, Uchida A, Kuriyama K, et al. Novel solid self-emulsifying drug delivery system of coenzyme Q10 with improved photochemical and pharmacokinetic behaviors. Eur J Pharm Sci 2012; 46(5): 492-9.
[http://dx.doi.org/10.1016/j.ejps.2012.03.015] [PMID: 22498005]
[80]
Onoue S, Kuriyama K, Uchida A, Mizumoto T, Yamada S. Inhalable sustained-release formulation of glucagon: In vitro amyloidogenic and inhalation properties, and in vivo absorption and bioactivity. Pharm Res 2011; 28(5): 1157-66.
[http://dx.doi.org/10.1007/s11095-011-0379-8] [PMID: 21287249]
[81]
Onoue S, Sato H, Ogawa K, et al. Inhalable dry-emulsion formulation of cyclosporine A with improved anti-inflammatory effects in experimental asthma/COPD-model rats. Eur J Pharm Biopharm 2012; 80(1): 54-60.
[http://dx.doi.org/10.1016/j.ejpb.2011.10.003] [PMID: 22008148]
[82]
Reddy LH, Sharma RK, Chuttani K, Mishra AK, Murthy RR. Etoposide-incorporated tripalmitin nanoparticles with different surface charge: Formulation, characterization, radiolabeling, and biodistribution studies. AAPS J 2004; 6(3): 55-64.
[http://dx.doi.org/10.1208/aapsj060323] [PMID: 15760108]
[83]
Piao H, Kamiya N, Hirata A, Fujii T, Goto M. A novel solid-in-oil nanosuspension for transdermal delivery of diclofenac sodium. Pharm Res 2008; 25(4): 896-901.
[http://dx.doi.org/10.1007/s11095-007-9445-7] [PMID: 17896098]
[84]
Zhang N, Ping Q, Huang G, Xu W, Cheng Y, Han X. Lectin-modified solid lipid nanoparticles as carriers for oral administration of insulin. Int J Pharm 2006; 327(1-2): 153-9.
[http://dx.doi.org/10.1016/j.ijpharm.2006.07.026] [PMID: 16935443]
[85]
Pathak P, Nagarsenker M. Formulation and evaluation of lidocaine lipid nanosystems for dermal delivery. AAPS PharmSciTech 2009; 10(3): 985-92.
[http://dx.doi.org/10.1208/s12249-009-9287-1] [PMID: 19641997]
[86]
Watanabe M, Kawano K, Yokoyama M, Opanasopit P, Okano T, Maitani Y. Preparation of camptothecin-loaded polymeric micelles and evaluation of their incorporation and circulation stability. Int J Pharm 2006; 308(1-2): 183-9.
[http://dx.doi.org/10.1016/j.ijpharm.2005.10.030] [PMID: 16324807]
[87]
Pepić I, Jalšenjak N, Jalšenjak I. Micellar solutions of triblock copolymer surfactants with pilocarpine. Int J Pharm 2004; 272(1-2): 57-64.
[http://dx.doi.org/10.1016/j.ijpharm.2003.11.032] [PMID: 15019069]
[88]
Onoue S, Kojo Y, Suzuki H, et al. Development of novel solid dispersion of tranilast using amphiphilic block copolymer for improved oral bioavailability. Int J Pharm 2013; 452(1-2): 220-6.
[http://dx.doi.org/10.1016/j.ijpharm.2013.05.022] [PMID: 23694807]
[89]
Kabanov AV, Alakhov VY. Pluronic block copolymers in drug delivery: From micellar nanocontainers to biological response modifiers. Crit Rev Ther Drug Carrier Syst 2002; 19(1): 1-72.
[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v19.i1.10] [PMID: 12046891]
[90]
Zhang J, He C, Tang C, Yin C. Ternary polymeric nanoparticles for oral siRNA delivery. Pharm Res 2013; 30(5): 1228-39.
[http://dx.doi.org/10.1007/s11095-012-0961-8] [PMID: 23307349]
[91]
Bahadur S, Prakash A. A comprehensive review on nanomedicine: Promising approach for treatment of brain tumor through intranasal administration. Curr Drug Targets 2023; 24(1): 71-88.
[http://dx.doi.org/10.2174/1389450124666221019141044] [PMID: 36278468]
[92]
Bahadur S, Sachan N, Harwansh RK, Deshmukh R. Nanoparticlized system: Promising approach for the management of alzheimer’s disease through intranasal delivery. Curr Pharm Des 2020; 26(12): 1331-44.
[http://dx.doi.org/10.2174/1381612826666200311131658] [PMID: 32160843]
[93]
Harwansh RK, Bahadur S, Deshmukh R, Rahman MA. Exciting potential of nanoparticlized lipidic system for effective treatment of breast cancer and clinical updates: A translational prospective. Curr Pharm Des 2020; 26(11): 1191-205.
[http://dx.doi.org/10.2174/1381612826666200131101156] [PMID: 32003686]
[94]
Sachan N, Bahadur S, Sharma PK. Recent advances and novel approaches for nose to brain drug delivery for treatment of migraine. Drug Deliv Lett 2019; 9(3): 182-98.
[http://dx.doi.org/10.2174/2210303109666190508083142]
[95]
Pandey R, Ahmad Z, Sharma S, Khuller GK. Nano-encapsulation of azole antifungals: Potential applications to improve oral drug delivery. Int J Pharm 2005; 301(1-2): 268-76.
[http://dx.doi.org/10.1016/j.ijpharm.2005.05.027] [PMID: 16023808]
[96]
Rahman MA, Shakeel K, Ahmad FJ, Harwansh RK. β-Artemether and lumefantrine dual drug loaded lipid nanoparticles: Physicochemical characterization, pharmacokinetic evaluation and biodistribution study. Pharm Nanotechnol 2022; 10(3): 210-9.
[http://dx.doi.org/10.2174/2211738510666220428133532] [PMID: 36029070]
[97]
Chaturvedi S, Garg A, Verma A. Nano lipid based carriers for lymphatic voyage of anti-cancer drugs: An insight into the in-vitro, ex-vivo, in-situ and in-vivo study models. J Drug Deliv Sci Technol 2020; 59(101899): 101899.
[http://dx.doi.org/10.1016/j.jddst.2020.101899]
[98]
Chaturvedi S, Garg A. An insight of techniques for the assessment of permeation flux across the skin for optimization of topical and transdermal drug delivery systems. J Drug Deliv Sci Technol 2021; 62: 102355.
[http://dx.doi.org/10.1016/j.jddst.2021.102355]
[99]
Garg A, Chaturvedi S. A comprehensive review on chrysin: Emphasis on molecular targets, pharmacological actions and bio-pharmaceutical aspects. Curr Drug Targets 2022; 23(4): 420-36.
[http://dx.doi.org/10.2174/1389450122666210824141044] [PMID: 34431464]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy