Review on nanoparticles used in drug delivery for cancer

Authors

  • Sonam B Bisen Department of Pharmaceutics, Sinhgad College of Pharmacy, Vadgaon (Bk), Pune 411041, Maharashtra, India.

DOI:

https://doi.org/10.30574/gscbps.2021.16.1.0194

Keywords:

Nanoparticles, Nanocarriers, Drug delivery, Cancer

Abstract

Current cancer treatments include surgical intervention, radiation, and chemotherapy medications. Nanoparticles have a variety of advantages as medication delivery systems. Nanoparticles (NPs) are newly discovered methods for delivering medicines to tumour cells with little drug leakage into healthy cells. To enhance biodistribution and increase circulation duration in the bloodstream, nanoparticles have been developed with optimum size and surface properties. Here, I look at the many types and features of nanoparticles. Examples of commercially available nanocarrier-based medicines include: Therapeutic nanoparticles, the function of metal nanoparticles in cancer diagnosis and therapy, are important ideas in nanoparticle medication delivery for cancer.

Metrics

Metrics Loading ...

References

Stewart BW, Kleihues P. World Cancer Report. Lyon: IARC press. 2003.

Society AC. Breast cancer facts & figures. Atlanta (GA): American Cancer Society. 2007.

Ross JS, Schenkein DP, Pietrusko R, et al. Targeted therapies for cancer 2004. Am J Clin Pathol. 2004; 122: 598–609.

Cho K, Wang X, Nie S, et al. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res. 2008; 14: 1310–1316.

Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Adv Enzyme Regul. 2001; 41: 189–207.

Allen TM. Ligand-targeted therapeutics in anticancer therapy. Nat Rev Cancer. 2002; 2: 750–763.

Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer. 2006; 6: 688–701.

Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer. 2005; 5: 161–171.

Dadwal A, Baldi A, Kumar Narang R. Nanoparticles as carriers for drug delivery in cancer. Artificial cells, nanomedicine, and biotechnology. 2018; 46(2): 295-305.

Moghimi SM, Hunter AC, Murray JC. Long-circulating and targetspecific nanoparticles: theory to practice. Pharmacol Rev. 2001; 53: 283–318.

Moghimi SM, Szebeni J. Stealth liposomes and long circulating nanoparticles: critical issues in pharmacokinetics, opsonization and protein-binding properties. Prog Lipid Res. 2003; 42: 463–478.

Harris JM, Martin NE, Modi M. Pegylation. Clin Pharmacokinet. 2001; 40: 539–551.

Wisse E, Braet F, Luo D, et al. Structure and function of sinusoidal lining cells in the liver. Toxicol Pathol. 1996; 24: 100–111.

Yuan F, Dellian M, Fukumura D, et al. Vascular permeability in a human tumor xenograft: molecular size dependence and cutoff size. Cancer Res. 1995; 55: 3752–3756.

Gradishar WJ, Tjulandin S, Davidson N, et al. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer. J Clin Oncol. 2005; 23: 7794–7803.

Sabbatini P, Aghajanian C, Dizon D, et al. Phase II study of CT2103 in patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. J Clin Oncol. 2004; 22: 4523–4531.

Bhatt R, DE Vries P, Tulinsky J, et al. Synthesis and in vivo antitumor activity of poly (L-glutamic acid) conjugates of 20 (S)-camptothecin. J Med Chem. 2003; 46: 190–193.

Vasey PA, Kaye SB, Morrison R, et al. Phase I clinical and pharmacokinetic study of PK1 [N-(2-hydroxypropyl) methacrylamide copolymer doxorubicin]: first member of a new class of chemotherapeutic agents – drug-polymer conjugates. Clin Cancer Res. 1999; 5: 83–94.

Markman M. Pegylated liposomal doxorubicin in the treatment of cancers of the breast and ovary. Expert Opin Pharmacother. 2006; 7: 1469–1474.

Rivera E. Current status of liposomal anthracycline therapy in metastatic breast cancer. Clin Breast Cancer. 2003; 4: S76–S83.

Rosenthal E, Poizot-Martin I, Saint-Marc T, et al. Phase IV study of liposomal daunorubicin (DaunoXome) in AIDS-related Kaposi sarcoma. Am J Clin Oncol. 2002; 25: 57–59.

Batrakova E, Dorodnych TY, Klinskii EY, et al. Anthracycline antibiotics non-covalently incorporated into the block copolymer micelles: in vivo evaluation of anti-cancer activity. Br J Cancer. 1996; 74: 1545–1552.

Nakanishi T, Fukushima S, Okamoto K, et al. Development of the polymer micelle carrier system for doxorubicin. J Control Release. 2001; 74: 295–302.

Kim T-Y, Kim D-W, Chung J-Y, 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.

Malik N, Evagorou EG, Duncan R. Dendrimer-platinate: a novel approach to cancer chemotherapy. Anti-Cancer Drugs. 1999; 10: 767–776.

CJ Hawker, KL Wooley. The convergence of synthetic organic and polymer chemistries, Science. 2005; 309(5738): 1200–1205.

S Johnson, A Bangham. Potassium permeabilityof single compartment liposomes with and without valinomycin, Biochim. et Biophys. Acta (BBA): Biomembr. 1969; 193(1): 82–91.

Y Malam, M Loizidou, AM Seifalian. Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer, Trends Pharmacol. Sci. 2009; 30(11): 592–599.

VP Torchilin. Recent advances with liposomes as pharmaceutical carriers, Nat. Rev. Drug Discov. 2005; 4(2): 145–160.

CMJ Hu, S Kaushal, HST Cao, S Aryal, M Sartor, S Esener, et al., Halfantibody functionalized lipid- polymer hybrid nanoparticles for targeted drug delivery to carcinoembryonic antigen presenting pancreatic cancer cells, Mol. Pharm. 2010; 7(3): 914–920.

R Misra, S Acharya, SK Sahoo. Cancer nanotechnology: application of nanotechnology in cancer therapy, Drug Discov. Today. 2010; 15(19): 842–850.

S Doktorovova, EB Souto, AM Silva. Nanotoxicology applied to solid lipid nanoparticles and nanostructured lipid carriers–a systematic review of in vitro data, Eur. J. Pharm. Biopharm. 2014; 87(1): 1–18.

S Selvamuthukumar, R Velmurugan. Nanostructured lipid carriers: a potential drug carrier for cancer chemotherapy, Lipids Health Dis. 2012; 11: 159.

Z Shao, J Shao, B Tan, S Guan, Z Liu, Z Zhao, et al., Targeted lung cancer therapy: preparation and optimization of transferrin-decorated nanostructured lipid carriers as novel nanomedicine for co-delivery of anticancer drugs and DNA, Int. J. Nanomed. 2015; 10: 1223.

R Müller, W Mehnert, JS Lucks, C Schwarz, A Zur Mühlen, H Meyhers, et al., Solid lipid nanoparticles (SLN): an alternative colloidal carrier system for controlled drug delivery, Eur. J. Pharm. Biopharm. 1995; 41(1): 62–69.

HL Wong, R Bendayan, AM Rauth, Y Li, XY Wu, Chemotherapy with anticancer drugs encapsulated in solid lipid nanoparticles, Adv. Drug Deliv. Rev. 2007; 59(6): 491–504.

S Acharya, SK Sahoo, PLGA nanoparticles containing various anticancer agents and tumour delivery by EPR effect, Adv. Drug Deliv. Rev. 2011; 63(3): 170–183.

D Astruc, E Boisselier, Ct Ornelas. Dendrimers designed for functions: from physical, photophysical, and supramolecular properties to applications in sensing, catalysis, molecular electronics, photonics, and nanomedicine, Chem. Rev. 2010; 110(4): 1857–1959.

D Tomalia, S Uppuluri, D Swanson, H Brothers, L Piehler, J Li, et al., Dendritic Macromolecules: A Fourth Major Class of Polymer Architecture–New Properties Driven by Architecture. MRS Proceedings, Cambridge Univ Press. 1998.

CJ Hawker, JM Frechet. Preparation of polymers with controlled molecular architecture: a new convergent approach to dendritic macromolecules, J. Am. Chem. Soc. 1990; 112(21): 7638–7647.

S Svenson, DA Tomalia. Dendrimers in biomedical applications—reflections on the field, Adv. Drug Deliv. Rev. 2005; 57(15): 2106–2129.

CO Turrin, AM Caminade. Dendrimers for imaging. dendrimers: towards catalytic, Mater. Biomed. Uses. 2011; 393–412.

AR Menjoge, RM Kannan, DA Tomalia. Dendrimer-based drug and imaging conjugates: design considerations for nanomedical applications, Drug Discov. Today. 2010; 15(5): 171–185.

XH Peng, X Qian, H Mao, AY Wang. Targeted magnetic iron oxide nanoparticles for tumor imaging and therapy, Int. J. Nanomed. 2008; 3(3): 311.

MK Yu, YY Jeong, J Park, S Park, JW Kim, JJ Min, et al., Drug-loaded superparamagnetic iron oxide nanoparticles for combined cancer imaging and therapy in vivo, Angew. Chem. Int. Ed. 2008; 47(29): 5362–5365.

C Sun, K Du, C Fang, N Bhattarai, O Veiseh, F Kievit, et al., PEG-mediated synthesis of highly dispersive multifunctional superparamagnetic nanoparticles: their physicochemical properties and function in vivo, ACS Nano. 2010; 4(4): 2402–2410.

FM Kievit, M Zhang. Surface engineering of iron oxide nanoparticles for targeted cancer therapy, Acc. Chem. Res. 2011; 44(10): 853–862.

DM Eigler, EK Schweizer. Positioning single atoms with a scanning tunnelling microscope, Nature. 1990; 344(6266): 524–526.

JH Park, G von Maltzahn, E Ruoslahti, SN Bhatia, MJ Sailor. Micellar hybrid nanoparticles for simultaneous magnetofluorescent imaging and drug delivery, Angew. Chem. 2008; 120(38): 7394–7398.

S Libutti, G Paciotti, L Myer, R Haynes, W Gannon, M Walker, et al., Results of a Completed Phase I Clinical Trial of CYT-6091: A Pegylated Colloidal Gold-TNF Nanomedicine. ASCO Annual Meeting Proceedings. 2009.

B Blasiak, FC van Veggel, B Tomanek. Applications of nanoparticles for MRI cancer diagnosis and therapy, J. Nanomater. 2013; 12.

Bahrami B, Hojjat-Farsangi M, Mohammadi H, Anvari E, Ghalamfarsa G, Yousefi M, Jadidi-Niaragh F. Nanoparticles and targeted drug delivery in cancer therapy. Immunology letters. 1 Oct 2017; 190: 64-83.

Sinha R, Kim GJ, Nie S, Shin DM. Nanotechnology in cancer therapeutics: bioconjugated nanoparticles for drug delivery. Molecular cancer therapeutics. 1 Aug 2006; 5(8): 1909-17.

Jain RK. Barriers to drug delivery in solid tumors. Sci Am. 1994; 271: 58–65.

De Menezes DEL, Pilarski LM, Allen TM. In vitro and in vivo targeting of immunoliposomal doxorubicin to human B-cell lymphoma. Cancer Res. 1998; 58: 3320–3330.

Park JW, Hong K, Kirpotin DB, et al. Anti-HER2 immunoliposomes. Clin Cancer Res. 2002; 8: 1172–1181.

Allen TM. Ligand-targeted therapeutics in anticancer therapy. Nat Rev Cancer. 2002; 2: 750–763.

Pastan I, Hassan R, Fitzgerald DJ, et al. Immunotoxin therapy of cancer. Nat Rev Cancer. 2006; 6: 559–565.

Peer D, Zhu P, Carman CV, et al. Selective gene silencing in activated leukocytes by targeting siRNAs to the integrin lymphocyte function-associated antigen-1. Proc Natl Acad Sci USA. 2007; 104: 4095–4100.

Khlebtsov NG, Dykman LA. Optical properties and biomedical applications of plasmonic nanoparticles. J Quant Spect Rad Trans. 2010; 111: 1-35.

Jain PK, El-Sayed IH, El-Sayed, MA. Au nanoparticles target cancer. Nano Today. 2007; 2: 18-29.

Guo SR, Gong JY, Jiang P, Wu M, Lu Y, Yu SH. Biocompatible, luminescent silver@phenol formaldehyde resin core/shell nanospheres: large-scale synthesis and application for in vivo bioimaging. Adv Func Mat. 2008; 18: 872-879.

Portney, N.G., Ozkan, M. Nano-oncology: drug delivery, imaging, and sensing. Anal Bioanal Chem. 2006; 384: 620-630.

O’Neal D, Hirsch L, Halas N, et al. Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. Can Lett. 2004; 209: 171–176.

Xu R, Ma J, Sun XC, Chen ZP, Jiang XL, Guo ZR, Huang L, Li Y, Wang M, Wang CL, Liu JW, Fan X, Gu JY, Chen X, Zhang Y, Gu N. Ag nanoparti‐ cles sensitize IR-induced killing of cancer cells. Cell Res. 2009; 19: 1031-1034.

Ficai D, Ficai A, Andronescu E. Advances in cancer treatment: Role of nanoparticles. Larramendy LM, Soloneski S, Nanomaterials-Toxicit y and Risk Assessment. InTech. 15 Jul 2015; 1-22.

Downloads

Published

2021-07-30

How to Cite

Bisen, S. B. . (2021). Review on nanoparticles used in drug delivery for cancer. GSC Biological and Pharmaceutical Sciences, 16(1), 062–069. https://doi.org/10.30574/gscbps.2021.16.1.0194

Issue

Section

Review Article