420
Views
46
CrossRef citations to date
0
Altmetric
Review Article

Promising effects of nanomedicine in cancer drug delivery

ORCID Icon
Pages 319-324 | Received 27 Jun 2017, Accepted 05 Sep 2017, Published online: 18 Sep 2017

References

  • Alexis F, Pridgen EM, Langer R, et al. Nanoparticle technologies for cancer therapy. Drug Deliv. 2010;197:55–86.
  • Farokhzad OC, Langer R. Impact of nanotechnology on drug delivery. ACS Nano. 2009;3:16–20.
  • Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer. 2005;5:161–171.
  • Ferrari M. Nanogeometry: beyond drug delivery. Nat Nanotechnol. 2008;3:131–132.
  • Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer. 2006;6:688–701.
  • Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9:615–627.
  • Kopeček J, Kopečková P. HPMA copolymers: origins, early developments, present, and future. Adv Drug Deliv Rev. 2010;62:122–149.
  • Owens DE, Peppas NA. Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int J Pharm. 2006;307:93–102.
  • Champion JA, Katare YK, Mitragotri S. Particle shape: a new design parameter for micro-and nanoscale drug delivery carriers. J Control Release. 2007;121:3–9.
  • Vinogradov SV, Bronich TK, Kabanov AV. Nanosized cationic hydrogels for drug delivery: preparation, properties and interactions with cells. Adv Drug Deliv Rev. 2002;54:135–147.
  • Wong J, Brugger A, Khare A, et al. Suspensions for intravenous (IV) injection: a review of development, preclinical and clinical aspects. Adv Drug Deliv Rev. 2008;60:939–954.
  • Choi HS, Liu W, Misra P, et al. Renal clearance of quantum dots. Nat Biotechnol. 2007;25:1165–1170.
  • Sa LTM, Albernaz Mde S, Patricio BF, et al. Biodistribution of nanoparticles: initial considerations. J Pharm Biomed Anal. 2012;70:602–604.
  • Kazunori K, Kwon GS, Yokoyama M, et al. Block copolymer micelles as vehicles for drug delivery. J Control Release. 1993;24:119–132.
  • Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharm Res. 2007;24:1.
  • Chithrani BD, Chan WC. Elucidating the mechanism of cellular uptake and removal of protein-coated gold nanoparticles of different sizes and shapes. Nano Lett. 2007;7:1542–1550.
  • Dobrovolskaia MA, McNeil SE. Immunological properties of engineered nanomaterials. Nature Nanotech. 2007;2:469–478.
  • Serda RE, Ferrati S, Godin B, et al. Mitotic trafficking of silicon microparticles. Nanoscale. 2009;1:250–259.
  • Moghimi SM, Hamad I, Andresen TL, et al. Methylation of the phosphate oxygen moiety of phospholipid-methoxy (polyethylene glycol) conjugate prevents PEGylated liposome-mediated complement activation and anaphylatoxin production. FASEB J. 2006;20:2591–2593.
  • Hamad I, Christy Hunter A, Rutt KJ, et al. Complement activation by PEGylated single-walled carbon nanotubes is independent of C1q and alternative pathway turnover. Mol Immunol. 2008;45:3797–3803.
  • Chow EK-H, Ho D. Cancer nanomedicine: from drug delivery to imaging. Sc Transl Med. 2013;5:216rv4–216rv4.
  • Xu X, Ho W, Zhang X, et al. Cancer nanomedicine: from targeted delivery to combination therapy. Trends Mol Med. 2015;21:223–232.
  • Wakaskar RR, Bathena SP, Tallapaka SB, et al. Peripherally cross-linking the shell of core-shell polymer micelles decreases premature release of physically loaded combretastatin A4 in whole blood and increases its mean residence time and subsequent potency against primary murine breast tumors after IV administration. Pharm Res. 2015;32:1028–1044.
  • Ambardekar VV, Wakaskar RR, Sharma B, et al. The efficacy of nuclease-resistant Chol-siRNA in primary breast tumors following complexation with PLL-PEG (5K). Biomaterials. 2013;34:4839–4848.
  • Wakaskar RR. Effect of peripheral shell cross-linking on the efficacy of a hydrophobic vascular disrupting agent physically loaded in core-shell polymeric micelles. University of Nebraska Medical Center; 2015.
  • Almeida JPM, Chen AL, Foster A, et al. In vivo biodistribution of nanoparticles. Nanomedicine (Lond). 2011;6:815–835.
  • Duan X, Li Y. Physicochemical characteristics of nanoparticles affect circulation, biodistribution, cellular internalization, and trafficking. Small. 2013;9:1521–1532.
  • Ernsting MJ, Murakami M, Roy A, et al. Factors controlling the pharmacokinetics, biodistribution and intratumoral penetration of nanoparticles. J Control Release. 2013;172:782–794.
  • Kwon GS, Okano T. Polymeric micelles as new drug carriers. Adv Drug Deliv Rev. 1996;21:107–116.
  • van Nostrum CF. Covalently cross-linked amphiphilic block copolymer micelles. Soft Matter. 2011;7:3246–3259.
  • Kamaly N, Xiao Z, Valencia PM, et al. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem Soc Rev. 2012;41:2971–3010.
  • Acharya S, Sahoo SK. PLGA nanoparticles containing various anticancer agents and tumour delivery by EPR effect. Adv Drug Deliv Rev. 2011;63:170–183.
  • Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Deliv Rev. 2011;63:131–135.
  • Barenholz YC. Doxil®-the first FDA-approved nano-drug: lessons learned. J Control Release. 2012;160:117–134.
  • Kim SC, Kim DW, Shim YH, et al. In vivo evaluation of polymeric micellar paclitaxel formulation: toxicity and efficacy. J Control Release. 2001;72:191–202.
  • Kim T-Y, Kim DW, Chung JY, et al. Phase I and pharmacokinetic study of Genexol-PM, a cremophor-free, polymeric micelle-formulated paclitaxel, in patients with advanced malignancies. Clin Cancer Res. 2004;10:3708–3716.
  • Semple SC, Akinc A, Chen J, et al. Rational design of cationic lipids for siRNA delivery. Nat Biotechnol. 2010;28:172–176.
  • Alexis F, Pridgen E, Molnar LK, et al. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm. 2008;5:505–515.
  • Arvizo RR, Miranda OR, Moyano DF, et al. Modulating pharmacokinetics, tumor uptake and biodistribution by engineered nanoparticles. PLoS One. 2011;6:e24374.
  • Gaumet M, Vargas A, Gurny R, et al. Nanoparticles for drug delivery: the need for precision in reporting particle size parameters. Eur J Pharm Biopharm. 2008;69:1–9.
  • Li S-D, Huang L. Pharmacokinetics and biodistribution of nanoparticles. Mol Pharm. 2008;5:496–504.
  • Wakaskar R. Types of nanocarriers–formulation method and applications. J Bioequiv Avail. 2017;9:e77.
  • Bobo D, Robinson KJ, Islam J, et al. Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date. Pharm Res. 2016;33:2373–2387.
  • Wakaskar RR. General overview of lipid–polymer hybrid nanoparticles, dendrimers, micelles, liposomes, spongosomes and cubosomes. J Drug Target. 2017 [Aug 18]; [8 p.]. DOI:10.1080/1061186X.2017.1367006
  • Bertrand N, Wu J, Xu X, et al. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:2–25.
  • Danquah MK, Zhang XA, Mahato RI. Extravasation of polymeric nanomedicines across tumor vasculature. Adv Drug Deliv Rev. 2011;63:623–639.
  • Wakaskar RR. Passive and active targeting in tumor microenvironment. Int J Drug Develop Res. 2017;9:37–41.
  • Peer D, Karp JM, Hong S, et al. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2:751–760.
  • Ge Z, Liu S. Functional block copolymer assemblies responsive to tumor and intracellular microenvironments for site-specific drug delivery and enhanced imaging performance. Chem Soc Rev. 2013;42:7289–7325.
  • Hrkach J, Von Hoff D, Mukkaram Ali M, et al. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile. Sci Transl Med. 2012;4:128ra39–128ra39.
  • Davis ME. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol Pharma. 2009;6:659–668.
  • Davis ME, Zuckerman JE, Hang C, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464:1067–1070.
  • Lee RJ, Low PS. Delivery of liposomes into cultured KB cells via folate receptor-mediated endocytosis. J Biol Chem 1994;269:3198–3204.
  • Rege BD, Kao JP, Polli JE. Effects of nonionic surfactants on membrane transporters in Caco-2 cell monolayers. Eur J Pharm Sci. 2002;16:237–246.
  • Pan J, Feng S-S. Targeted delivery of paclitaxel using folate-decorated poly (lactide)–vitamin E TPGS nanoparticles. Biomaterials. 2008;292663–2672.
  • Mishra B, Patel BB, Tiwari S. Colloidal nanocarriers: a review on formulation technology, types and applications toward targeted drug delivery. Nanomed Nanotechnol Biol Med. 2010;6:9–24.
  • Chen WC, Zhang AX, Li S-D. Limitations and niches of the active targeting approach for nanoparticle drug delivery. Eur J Nanomed. 2012;4:89–93.
  • Wakaskar RR. Challenges pertaining to adverse effects of drugs. Int J Drug Develop Res. 2017;9:1–2.
  • Wakaskar RR. Brief overview of nanoparticulate therapy in cancer. Journal of Drug Targeting 2017 [Jul 4]; [4 p.]. DOI:10.1080/1061186X.2017.1347175
  • Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer drugs. Annu Rev Med. 2012;63:185–198.
  • Arrowsmith J. A decade of change. Nat Rev Drug Discov. 2012;11:17–18.
  • Schütz CA, Juillerat-Jeanneret L, Mueller H, et al. Therapeutic nanoparticles in clinics and under clinical evaluation. Nanomedicine. 2013;8:449–467.
  • Kou L, Luo Y, Liu X, et al. Effects of mild heat treatment on microbial growth and product quality of packaged fresh‐cut table grapes. J Food Sci. 2007;72:S567–S573.
  • Mousavi SR, Rezaei M. Nanotechnology in agriculture and food production. J Appl Environ Biol Sci. 2011;1:414–419.
  • Rashidi L, Khosravi-Darani K. The applications of nanotechnology in food industry. Critic Rev Food Sci Nutrit. 2011;51:723–730.
  • Taglietti A, Arciola CR, D'Agostino A, et al. Antibiofilm activity of a monolayer of silver nanoparticles anchored to an amino-silanized glass surface. Biomaterials. 2014;35:1779–1788.
  • Eleftheriadou M, Pyrgiotakis G, Demokritou P. Nanotechnology to the rescue: using nano-enabled approaches in microbiological food safety and quality. Curr Opin Biotechnol. 2017;44:87–93.
  • Leoni L, Desai TA. Nanoporous biocapsules for the encapsulation of insulinoma cells: biotransport and biocompatibility considerations. IEEE Trans Biomed Eng. 2001;48:1335–1341.
  • Freitas RA. Current status of nanomedicine and medical nanorobotics. J Comput Theoret Nanosci. 2005;2:1–25.
  • Lowery AR, Gobin AM, Day ES, et al. Immunonanoshells for targeted photothermal ablation of tumor cells. Int J Nanomed. 2006;1(2):149–154.
  • Wakaskar RR. Cancer therapy with drug delivery systems. J Pharmacogeno Pharmacoproteom. 2017;8:100e1578.
  • Wakaskar RR. Polymeric Micelles and their properties. J Nanomed Nanotechnol. 2017;8:1000433.

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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