TAILORING THE NANOPARTICLES SURFACE FOR EFFICIENT CANCER THERAPEUTICS DELIVERY

Authors

  • ARUSHI CHAUHAN Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India 160012
  • RAVI RANJAN Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India 160012
  • PRAMOD AVTI Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India 160012
  • ARVIND GULBAKE Centre for Interdisciplinary Research, D. Y. Patil Education Society, Deemed to be University, Kolhapur, M. S., 416006, India

DOI:

https://doi.org/10.22159/ijap.2020.v12s4.40099

Keywords:

Cancer therapeutics, Dendrimers, Nanomaterials, Iron Oxide Nanoparticles, Lipid nanoparticles, Polyethyleneglycol, Polymeric nanoparticle, Polyvinyl alcohol, Transferrin

Abstract

Nanotechnology has tremendous advantages in many areas of scientific as well as clinical research. The development of nanoparticles (NPs) that can efficiently deliver drugs specifically to the cancer cells can help reduce normal cells toxicity and co-morbidities. Cancer can be treated by exploiting the unique physiochemical of the NPs, and modulating their surface modifications using ligands which further could be used as drug cargo vehicles. To enhance biocompatibility and drug delivery towards the target site, various modifications can be included to modify the surface of the NPs, such as carbohydrates, dendrimers, DNA, RNA, siRNA, drugs, and other ligands. These ligand-coated NPs have potential applications in the field of biomedical research, including diagnosis, contrast agents for molecular and clinical imaging (Magnetic Resonance Imaging (MRI), Computed tomography (CT), positron emission tomography (PET)), as cargo vehicles for drugs, increasing the blood circulation half-life, and blood detoxification. Further, the conjugation of anti-cancer drugs to the NPs can be efficiently used to target the cancer disease. This review highlights some of the features and surface modification strategies of the NPs, such as an iron oxide (IO), liposomes (LP)-based NPs, and polymer-based NPs, which show their effectiveness as cargo agents for cancer therapeutics.

Downloads

Download data is not yet available.

References

Fischer T, Wilharm N, Hayn A, Mierke CT. Matrix and cellular mechanical properties are the driving factors for facilitating human cancer cell motility into 3D engineered matrices. Converg Sci Phys Oncol 2017;3:044003.

Suhail Y, Cain M, Vanaja K, Kurywchak P, Levchenko A, Kalluri R Kshitiz. Systems biology of cancer metastasis. Cell Syst 2019;9:109-27.

Xi W, Schmidt CK, Sanchez S, Gracias DH, Carazo-Salas RE, Butler R, et al. Molecular insights into division of single human cancer cells in on-chip transparent microtubes. ACS Nano 2016;10:5835-46.

Ramakrishna N, Temin S, Chandarlapaty S, Crews JR, Davidson NE, Esteva FJ, et al. Recommendations on disease management for patients with advanced human epidermal growth factor receptor 2–positive breast cancer and brain metastases: american society of clinical oncology clinical practice guideline. J Clin Oncol 2014;32:2100.

Mottet N, Bellmunt J, Bolla M, Briers E, Cumberbatch MG, De Santis M, et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol 2017;71:618-29.

Oronsky BT, Carter CA, Oronsky AL, Salacz ME, Reid T. “No patient left behind”: an alternative to “the War on Cancer” metaphor. Springer; 2016.

Wang Y, Dou L, He H, Zhang Y, Shen Q. Multifunctional nanoparticles as nanocarrier for vincristine sulfate delivery to overcome tumor multidrug resistance. Mol Pharm 2014; 11:885-94.

Schofield PN, Kondratowicz M. Evolving paradigms for the biological response to low dose ionizing radiation; the role of epigenetics. Int J Radiat Biol 2018;94:769-81.

Cartmell KB, Bonilha HS, Matson T, Bryant DC, Zapka J, Bentz TA, et al. Patient participation in cancer clinical trials: a pilot test of lay navigation. Contemp Clin Trials Commun 2016;3:86-93.

Fogel DB. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contemp Clin Trials Commun 2018;11:156-64.

Ovais M, Raza A, Naz S, Islam NU, Khalil AT, Ali S, et al. Current state and prospects of the phytosynthesized colloidal gold nanoparticles and their applications in cancer theranostics. Appl Microbiol Biotechnol 2017;101:3551-65.

Piktel E, Niemirowicz K, Wątek M, Wollny T, Deptuła P, Bucki R. Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J. Nanobiotechnol 2016;14:39.

Ng VW, Avti PK, Bedard M, Lam T, Rouleau L, Tardif JC, et al. Miktoarm star conjugated multifunctional gold nanoshells: synthesis and evaluation of biocompatibility and cellular uptake. J Mater Chem B 2014;2:6334-44.

Bédard M, Avti PK, Lam T, Rouleau L, Tardif JC, Rheaume E, et al. Conjugation of multivalent ligands to gold nanoshells and designing a dual-modality imaging probe. J Mater Chem B 2015;3:1788-800.

Saleem J, Wang L, Chen C. Carbon‐based nanomaterials for cancer therapy via targeting tumor microenvironment. Adv Healthc Mater 2018;7:1800525.

Wang Y, Sun S, Zhang Z, Shi D. Nanomaterials for cancer precision medicine. Adv Mater 2018;30:1705660.

Haley B, Frenkel E. editors. Nanoparticles for drug delivery in cancer treatment. Urol Oncol Sem Ori: Elsevier; 2008.

Palazzolo S, Bayda S, Hadla M, Caligiuri I, Corona G, Toffoli G, et al. The clinical translation of organic nanomaterials for cancer therapy: a focus on polymeric nanoparticles, micelles, liposomes and exosomes. Curr Med Chem 2018;25:4224-68.

Bi Y, Hao F, Yan G, Teng L, J Lee R, Xie J. Actively targeted nanoparticles for drug delivery to tumor. Curr Drug Metab 2016;17:763-82.

Avti PK, Kakkar A. Dendrimers as anti-inflammatory agents. Braz J Pharm Sci 2013;49:57-65.

Avti PK, Maysinger D, Kakkar A. Alkyne-azide “click” chemistry in designing nanocarriers for applications in biology. Molecules 2013;18:9531-49.

Barenholz Y. Doxil-the first FDA-approved nano-drug: from an idea to a product. Handb Harnessing Biomater Nanomed 2012:335-98. DOI:10.4032/9789814364270

Farooq MA, Aquib M, Farooq A, Haleem Khan D, Joelle Maviah MB, Sied Filli M, et al. Recent progress in nanotechnology-based novel drug delivery systems in designing of cisplatin for cancer therapy: an overview. Artif Cell Nanomed B 2019;47:1674-92.

Lam T, Pouliot P, Avti PK, Lesage F, Kakkar AK. Superparamagnetic iron oxide-based nanoprobes for imaging and theranostics. Adv Colloid Interface Sci 2013;199:95-113.

Singh G, Kumar N, Avti PK. Bioheat physics for hyperthermia therapy. Application of Biomedical Engineering in Neuroscience: Springer; 2019. p. 381-97.

Singh G, Kumar N, Avti PK. Computational evaluation of effectiveness for intratumoral injection strategies in magnetic nanoparticle assisted thermotherapy. Int J Heat Mass Tran 2020;148:119129.

Lam T, Avti PK, Pouliot P, Maafi F, Tardif JC, Rheaume E, et al. Fabricating water-dispersible superparamagnetic iron oxide nanoparticles for biomedical applications through ligand exchange and direct conjugation. J Nanomater 2016;6:100.

Lam T, Avti PK, Pouliot P, Tardif JC, Rheaume E, Lesage F, et al. Surface engineering of SPIONs: role of phosphonate ligand multivalency in tailoring their efficacy. Nanotechnology 2016;27:415602.

Lam T, Avti PK, Pouliot P, Tardif JC, Rheaume E, Lesage F, et al. Magnetic resonance imaging/fluorescence dual modality protocol using designed phosphonate ligands coupled to superparamagnetic iron oxide nanoparticles. J Mater Chem 2016;4:3969-81.

Wu W, Jiang CZ, Roy VA. Designed synthesis and surface engineering strategies of magnetic iron oxide nanoparticles for biomedical applications. Nanoscale 2016;8:19421-74.

Thorat ND, Bohara RA, Malgras V, Tofail SA, Ahamad T, Alshehri SM, et al. Multimodal super paramagnetic nanoparticles with unusually enhanced specific absorption rate for synergetic cancer therapeutics and magnetic resonance imaging. ACS Appl Mater Inter 2016;8:14656-64.

Schwaminger S, Bauer D, Fraga Garcia P, Wagner F, Berensmeier S. Oxidation of magnetite nanoparticles: impact on surface and crystal properties. Cryst Eng Comm 2017;19:246-55.

Wang S, Jiao Q, Liu X, Xu Y, Shi Q, Yue S, et al. Controllable synthesis of γ-Fe2O3 nanotube/porous rGO composites and their enhanced microwave absorption properties. ACS Sustain Chem Eng 2019;7:7004-13.

Chen J, Sun X, Shao R, Xu Y, Gao J, Liang W. VEGF siRNA delivered by polycation liposome-encapsulated calcium phosphate nanoparticles for tumor angiogenesis inhibition in breast cancer. Int J Nanomed 2017;12:6075.

Danafar H, Sharafi A. Co-delivery of sulforaphane and curcumin with pegylated iron oxide-gold core-shell nanoparticles for delivery to breast cancer cell line. Iran J Pharm Res 2018;17:480.

Ding W, Jin X, Ma B, Wang X, Lou C, Zheng J, et al. Determination of prussian blue nanoparticles in rat tissue in the presence of endogenous iron interferences by inductively coupled plasma–optical emission spectrometry (ICP-OES). Anal Lett 2020:1-14. https://doi.org/10.1080/00032719.2020.1762630

Tian X, Liu S, Zhu J, Qian Z, Bai L, Pan Y. Biofunctional magnetic hybrid nanomaterials for theranostic applications. Nanotechnology 2018;30:032002.

Guo T, Wu Y, Lin Y, Xu X, Lian H, Huang G, et al. Black phosphorus quantum dots with renal clearance property for efficient photodynamic therapy. Small 2018;14:1702815.

Tsoi KM, MacParland SA, Ma XZ, Spetzler VN, Echeverri J, Ouyang B, et al. Mechanism of hard-nanomaterial clearance by the liver. Nat Mater 2016;15:1212-21.

Liang Q, Wang YX, Ding JS, He W, Deng Ll, Li N, et al. Intra-arterial delivery of superparamagnetic iron-oxide nanoshell based chemoembolization system for the treatment of liver tumor. Discovery Med 2017;23:27-39.

Carvalho A, Fernandes AR, Baptista PV. Nanoparticles as delivery systems in cancer therapy: focus on gold nanoparticles and drugs. Applications of Targeted Nano Drugs and Delivery Systems: Elsevier; 2019. p. 257-95.

Nan X, Zhang X, Liu Y, Zhou M, Chen X, Zhang X. Dual-targeted multifunctional nanoparticles for magnetic resonance imaging-guided cancer diagnosis and therapy. ACS Appl Mater Interfaces 2017;9:9986-95.

Jang H, Lee C, Nam GE, Quan B, Choi HJ, Yoo JS, et al. In vivo magnetic resonance and dual fluorescence imaging of tumor sites by using dye-doped silica-coated iron oxide nanoparticles. J Nanopart Res 2016;18:41.

Ahn SH, Lee N, Choi C, Shin SW, Han Y, Park HC. Feasibility study of Fe3O4/TaO x nanoparticles as a radiosensitizer for proton therapy. Phys Med Biol 2018;63:114001.

Ma D, Chen J, Luo Y, Wang H, Shi X. Zwitterion-coated ultrasmall iron oxide nanoparticles for enhanced T 1-weighted magnetic resonance imaging applications. J Mater Chem B 2017;5:7267-73.

Lachowicz D, Szpak A, Malek Zietek KE, Kepczynski M, Muller RN, Laurent S, et al. Biocompatible and fluorescent superparamagnetic iron oxide nanoparticles with superior magnetic properties coated with charged polysaccharide derivatives. Colloids Surf B 2017;150:402-10.

Qasim M, Asghar K, Dharmapuri G, Das D. Investigation of novel superparamagnetic Ni0. 5Zn0. 5Fe2O4@ albumen nanoparticles for controlled delivery of anticancer drug. Nanotechnology 2017;28:365101.

Patitsa M, Karathanou K, Kanaki Z, Tzioga L, Pippa N, Demetzos C, et al. Magnetic nanoparticles coated with polyarabic acid demonstrate enhanced drug delivery and imaging properties for cancer theranostic applications. Sci Rep 2017;7:1-8.

Zhao M, van Straten D, Broekman ML, Preat V, Schiffelers RM. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics 2020;10:1355.

El-Boubbou K. Magnetic iron oxide nanoparticles as drug carriers: preparation, conjugation and delivery. Nanomed J 2018;13:929-52.

Motomura M, Ichihara H, Matsumoto Y. Nano-chemotherapy using cationic liposome that strategically targets the cell membrane potential of pancreatic cancer cells with a negative charge. Bioorg Med Chem Lett 2018;28:1161-5.

Vassilev P, Tien HT. Planar lipid bilayers in relation to biomembranes. Structure and properties of cell membrane structure and properties of cell membranes: Volume III; 2018. p. 39.

Alavi M, Hamidi M. Passive and active targeting in cancer therapy by liposomes and lipid nanoparticles. Drug Metabol Person Therapy 2019;34:1-8.

Maeki M, Kimura N, Sato Y, Harashima H, Tokeshi M. Advances in microfluidics for lipid nanoparticles and extracellular vesicles and applications in drug delivery systems. Adv Drug Delivery Rev 2018;128:84-100.

Lopes NA, Pinilla CMB, Brandelli A. Pectin and polygalacturonic acid-coated liposomes as novel delivery system for nisin: preparation, characterization and release behavior. Food Hydrocoll 2017;70:1-7.

Pandey H, Rani R, Agarwal V. Liposome and their applications in cancer therapy. Braz Arch Biol 2016;59. DOI:10.1590/1678-4324-2016150477

Haeri A, Sadeghian S, Rabbani S, Shirani S, Anvari MS, Dadashzadeh S. Physicochemical characteristics of liposomes are decisive for their anti-restenosis efficacy following local delivery. Nanomedicine 2017;12:131-45.

Mastrotto F, Brazzale C, Bellato F, De Martin S, Grange G, Mahmoudzadeh M, et al. In vitro and in vivo behavior of liposomes decorated with PEGs with different chemical features. Mol Pharm 2019;17:472-87.

Wonder E, Simon Gracia L, Scodeller P, Majzoub RN, Kotamraju VR, Ewert KK, et al. Competition of charge-mediated and specific binding by peptide-tagged cationic liposome–DNA nanoparticles in vitro and in vivo. Biomaterials 2018;166:52-63.

Rajendran V, Rohra S, Raza M, Hasan GM, Dutt S, Ghosh PC. Stearylamine liposomal delivery of monensin in combination with free artemisinin eliminates blood stages of plasmodium falciparum in culture and P. berghei infection in murine malaria. Antimicrob Agents Chemother 2016;60:1304-18.

Mineart KP, Venkataraman S, Yang YY, Hedrick JL, Prabhu VM. Fabrication and characterization of hybrid stealth liposomes. Macromolecules 2018;51:3184-92.

Olusanya TO, Haj Ahmad RR, Ibegbu DM, Smith JR, Elkordy AA. Liposomal drug delivery systems and anticancer drugs. Molecules 2018;23:907.

Sutradhar KB, Amin M. Nanotechnology in cancer drug delivery and selective targeting. Int Sch Res Notices 2014;1-12. DOI:10.1155/2014/939378

Anselmo AC, Mitragotri S. Nanoparticles in the clinic: an update. Bioeng Transl Med 2019;4:e10143.

Berger JL, Smith A, Zorn KK, Sukumvanich P, Olawaiye AB, Kelley J, et al. Outcomes analysis of an alternative formulation of PEGylated liposomal doxorubicin in recurrent epithelial ovarian carcinoma during the drug shortage era. Onco Targets Ther 2014;7:1409.

Zhu Y, Zhang J, Meng F, Deng C, Cheng R, Feijen J, et al. cRGD/TAT dual-ligand reversibly cross-linked micelles loaded with docetaxel penetrate deeply into tumor tissue and show high antitumor efficacy in vivo. ACS Appl Mater Interfaces 2017;9:35651-63.

Kim CH, Lee SG, Kang MJ, Lee S, Choi YW. Surface modification of lipid-based nanocarriers for cancer cell-specific drug targeting. Int J Pharm Investig 2017;47:203-27.

Pereira NRC, Loiola RA, Rodrigues SF, de Oliveira CP, Buttenbender SL, Guterres SS, et al. Mechanisms of the effectiveness of poly (ε-caprolactone) lipid-core nanocapsules loaded with methotrexate on glioblastoma multiforme treatment. Int J Nanomed 2018;13:4563.

Perez Herrero E, Fernandez Medarde A. Advanced targeted therapies in cancer: drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm 2015;93:52-79.

Smith AG, Macleod KF. Autophagy, cancer stem cells and drug resistance. J Pathol 2019;247:708-18.

Olszowka M, Russo R, Mancini G, Cappelli C. A computational approach to the resonance Raman spectrum of doxorubicin in aqueous solution. Theor Chem Acc 2016;135:27.

Li B, Li Q, Mo J, Dai H. Drug-loaded polymeric nanoparticles for cancer stem cell targeting. Front Pharmacol 2017;8:51.

Silveira N, Longuinho MM, Leitao SG, Silva RS, Lourenço MC, Silva PE, et al. Synthesis and characterization of the antitubercular phenazine lapazine and development of PLGA and PCL nanoparticles for its entrapment. Mater Sci Eng 2016;58:458-66.

Ahsan SM, Thomas M, Reddy KK, Sooraparaju SG, Asthana A, Bhatnagar I. Chitosan as a biomaterial in drug delivery and tissue engineering. Int J Biol Macromol 2018;110:97-109.

Mahanta AK, Senapati S, Maiti P. A polyurethane–chitosan brush as an injectable hydrogel for controlled drug delivery and tissue engineering. Polym Chem 2017;8:6233-49.

Ni M, Xiong M, Zhang X, Cai G, Chen H, Zeng Q, et al. Poly (lactic-co-glycolic acid) nanoparticles conjugated with CD133 aptamers for targeted salinomycin delivery to CD133+osteosarcoma cancer stem cells. Int J Nanomed 2015;10:2537.

Hemshekhar M, Thushara RM, Chandranayaka S, Sherman LS, Kemparaju K, Girish KS. Emerging roles of hyaluronic acid bioscaffolds in tissue engineering and regenerative medicine. Int J Biol Macromol 2016;86:917-28.

Ke XY, Ng VWL, Gao SJ, Tong YW, Hedrick JL, Yang YY. Co-delivery of thioridazine and doxorubicin using polymeric micelles for targeting both cancer cells and cancer stem cells. Biomaterials 2014;35:1096-108.

Published

01-11-2020

How to Cite

CHAUHAN, A., RANJAN, R., AVTI, . P., & GULBAKE, A. (2020). TAILORING THE NANOPARTICLES SURFACE FOR EFFICIENT CANCER THERAPEUTICS DELIVERY. International Journal of Applied Pharmaceutics, 12(4), 11–17. https://doi.org/10.22159/ijap.2020.v12s4.40099

Issue

Section

Full Proceeding Paper