Applications of Nanoparticle Tracking Analysis in Drug and Gene Delivery

Nanoparticle tracking analysis (NTA) is a unique method that provides fast and multi-parameter information about nanoparticles. With the help of this technique, users can acquire number frequency distributions of particle sizes in polydisperse nanoparticulate systems.

NTA represents a novel technique, and as such has been implemented in many different sectors within the pharmaceutical sciences. This article describes some of the recent work published in the literature, where NTA has been recommended, employed, and evaluated for studying nanoparticle-based drug delivery systems.

Nanomedicine

The application of nanotechnology in medicine, particularly in drug and gene delivery, has become increasingly popular. This trend can be attributed to the fact that fewer novel drugs are entering into the market, and the discovery of novel biologically active compounds, which can be therapeutically utilized to treat many diseases, has come down drastically. As a result, the use of versatile and multifunctional structures of nanoparticles has grown quickly.

Nanoparticles provide improved pharmacokinetic properties, ensure sustained and controlled release, and target only certain tissues, cells, or organs. These aspects enhance the pharmacological efficacy of current drugs. Some of the commonly defined nanoparticle vectors are as follows:

  • liposomes
  • dendrimers
  • micelles
  • solid lipid nanoparticles
  • semiconductor nanoparticles
  • metallic nanoparticles
  • polymeric nanoparticles

Therefore, nanoparticles are widely used to deliver drugs, vaccines, genes, and diagnostics into particular cells and tissues. Conversely, while these particles are increasingly being utilized to bring down the side effects and toxicity levels of the drugs, it has been found that the so-called carrier systems themselves can pose hazards to patients.

In fact, the type of risks posed by utilizing nanoparticles for drug delivery are greater than the risks posed by chemicals used in traditional delivery matrices. As such, many different substances are being analyzed to formulate nanoparticles for drug delivery. These substances range from biological substances such as phospholipids, gelatin and albumin for liposomes through to substances of chemical nature such as metal containing nanoparticles and polymers.

Nanoparticles in Drug Delivery

Considering the various applications of nanoparticles, nanoparticle-based drug delivery and targeting has been the topic of a recent review, which discusses the advantages of nanotechnology and also provides warnings regarding the physical nature of the nanoparticles, and how they can impede with standardized and conventional immunotoxicity and biocompatibility testing procedures.

Yet another comprehensive review describes many assays that are needed to find out the chemical and physical characteristics of nanoparticles such as MALDI-TOF, batch-mode dynamic light scattering, TEM, AFM, zeta potential measurement, and SEM X-ray microanalysis of nanoparticles existing in cultured cell thin sections or tissues.

Since NTA is a newly developed method, it was not accounted for in this review. However, NTA is being used in the characterization of nanoparticulate suspensions, which are being formulated for drug delivery application. Before introducing nanoparticles to cellular systems for cytotoxicity testing, a better understanding of nanoparticle size distribution is very important. In this context, the NTA technique has been shown to be useful when compared to DLS and other nanoparticle characterization techniques.

Earlier Applications of NTA

Maher et al., 2009 used NTA for studying sodium caproate mediated promotion of oral drug absorption; Bult et al., 2010 utilized NTA for studying holonium; and Bhuiyan (2010) demonstrated that drug release from thermosensitive liposomes can be caused by hypothermia by utilizing the NTA technique to define the liposome preparations.

Recently, Sunshine et al. (2012) utilized the NTA technique to measure the particle size before subretinal injection. The effective transfection of the RPE in vivo indicated that nanoparticles can possibly be used for studying genetic diseases to treat various eye diseases.

Nanoparticles in Targeting

Molecular structures with an affinity for particular cell surface biomarkers are generally added during the targeting of drug delivery nanoparticles to certain sites. However, adding these capture molecules to the surface of the drug delivery nanoparticles can be quite challenging.

To ensure optimum performance, adequate loading, retention of activity, and reduced aggregation are very important. Likewise, when other biochemical species, developed to stabilize the functional structures, are added to the nanoparticles, it may cause similar toxic effects.

In contrast, when macromolecules are added to nanoparticles, NTA can detect even trace-level changes in hydrodynamic diameter and can even detect and specify aggregates that may occur during such changes. As such, NTA has been employed in many of these studies, including the influence of conjugating polymer-alendronate-taxane complexes used for targeting bone metastases (Miller et al., 2009).

NTA was used by the same group to demonstrate that conjugation for the targeting of angiogenesis-dependent calcified neoplasms utilizing varied polymers led to polydispersities of narrower and smaller sizes. This along with a cathepsin-K-cleavable system, a more specific drug release was eventually obtained. As a result, the group directed the toxicity of the free drugs to the bone cancer.

In another experimental study, both NTA and DLS techniques were utilized to demonstrate the particle size between 1 and 50nm in aqueous solution (Dellinger et al., 2013). This was done by using a number of advanced methods such as NMR, matrix assisted laser desorption ionization mass spectrometry (MALDI-TOF), and high performance liquid chromatography to define stabilized FcεRI-mediated mast cells, fullerene derivatives, and peripheral blood basophils.

NTA was also applied to size the calcium phosphate nanoparticles used for transporting supramolecular drugs through the cell membrane (Rotan et al., 2013). NTA was also used to size bioresorbable polymersomes for delivery of cisplatin (Petersen et al., 2013).

Gene, RNA and DNA Delivery

Novel biologically active compounds can be therapeutically exploited to treat many types of diseases. However, with fewer number of discoveries made in this regard, there is a need to bridge this critical gap. Here, the use of nanotechnology, predominantly in gene and drug delivery, represents a key milestone.

In this context, using nanoparticles to transport and deliver cargoes of various genetic materials presents an interesting approach. The use of using polymeric nanoparticles to deliver non-viral genes is one attractive method for treating genetic diseases and developing sophisticated technologies for regenerative medicine.

Viruses come with major safety issues, but polymeric nanoparticles can be formulated to be non-toxic, non-mutagenic, and non-immunogenic. Such particles are chemically versatile, environmentally responsive, biodegradable, easier to produce, and can carry larger nucleic acid cargo.

siRNA delivery to cell systems has been recently researched to improve human mesenchymal stem cell differentiation through RNA interference (RNAi). This approach may provide a suitable means for regulating cellular conditions for tissue engineering. However, this would require the development of a safe and effective delivery system.

The delivery of SiRNA has also been researched by using cell-penetrating peptides (CPPs), which have been widely studied as drug delivery systems for nanoparticles, nucleic acids, and proteins. Ezzat et al., 2012 also used NTA to show the stability of nanoparticles on drying and re-suspension. In a similar way, Troiber et al., 2012 compared four types of particle sizing techniques for the characterization of siRNA polyplex, as a standard method for measuring the particle size was not available.

As such, four types of analytical techniques, such as AFM, DLS, fluorescence correlation spectroscopy (FCS), and “nanoparticle trafficking analysis” (NTA), were assessed for their viability for studying the characteristics of both heterogeneous and homogeneous siRNA polyplexes. While larger particles measuring 120nm can be sized by all techniques, particles measuring 40nm had very low refractive index to be detected by NTA.

Recently, NTA was used to demonstrate that dendrimer structures, which were being used as siRNA delivery vehicles, experienced changes in terms of polydispersity and size at higher dendrimer concentrations. This suggested that electrostatic complexation leads to an equilibrium between complex aggregates of various sizes (Jensen, 2011). This analysis made it possible to detect the optimum dendrimer structure for subsequent delivery of nucleic acids.

The NTA technique has even been applied to develop non-viral gene delivery systems, which are based on poly(β-amino ester)s (PBAEs) (Tzeng et al., 2011), a lipophilic plasmid DNA condensate (Do et al., 2011), and in the screening of these structures in vitro (van Gael et al., 2011).

Recently, Shmueli et al., 2013 described a novel procedure to define PBEA nanoparticles through the NTA technique. These hydrolytically degradable PBAEs have been found to be effective at gene delivery to hard-to-transfect cell types, namely human brain cancer cells, human retinal endothelial cells, macrovascular endothelial cells, and mouse mammary epithelial cells. This NTA-based procedure can be used for assessing polymeric nanoparticles and any target cell type in a multiwell or 96-well plate format for transfection assay for quick screening of the transfection efficacy.

According to Witwer et al., 2013, droplet digital PCR and real-time quantitative PCR or RT-qPCR for plant miRNAs in mammalian blood does not provide significant proof for the uptake of dietary miRNAs. This is contrary to the earlier evidence that exogenous dietary miRNAs penetrate the tissues and bloodstream of ingesting animals. This evidence indicates that at least a single plant miRNA, miR168, takes part in cross-kingdom regulation of a mammalian transcript.

However, when RT-qPCR was used to determine plant and endogenous miRNAs from pigtailed macaques, which were injected with a miRNA-rich, plant-based substance, the outcomes did not support the uptake of dietary plant miRNAs. Here, NTA was applied to demonstrate a change in population and particle size after food intake.

Further, NTA technique was also used for studying chitosan-based nanoparticles for delivery of gene-and siRNA (Malmo, 2012) and for “The in vitro assessment of alkylglyceryl-functionalized chitosan nanoparticles as permeating vectors for the blood–brain barrier.”

Conclusion

NTA is a unique tool used for the characterization of nanoparticles. A better understanding of the distribution of nanoparticles before their introduction to cellular systems for cytotoxicity testing purposes is very important. NTA has proved handy in this regard when compared to other types of nanoparticle characterization methods, and therefore is used in a wide range of applications within the pharmaceutical sciences.

References

  1. Abdel-Hafez SM, Hathout RM, Sammour OA (2013) Towards better modelling of chitosan nanoparticles production: Screening different factors and comparing two experimental designs - International Journal of Biological Macromolecules, http://www.sciencedirect.com/science/article/pii/S0141813013006545
  2. Ballarín-González B, Dagnaes-Hansen F, Fenton RA, Gao S, Hein S, Dong M, Kjems J and Howard KA (2013) Protection and Systemic Translocation of siRNA Following Oral Administration of Chitosan/siRNA Nanoparticles, Molecular Therapy Nucleic Acids () 2, e76; doi:10.1038/mtna.2013.2 Published online 5 March 2013
  3. Battice LD (2011), Characterization of the interaction between protein loaded polymeric nanoparticles and supported lipid bilayers towards improved drug delivery systems, Master's Thesis, Department of Applied Physics and Biological Physics, Chalmers University of Technology, Göteborg, Sweden. http://publications.lib.chalmers.se/records/fulltext/141679.pdf
  4. Bender EA, Adorne MD, Colomé LM, Abdalla DP, Guterres SS, Pohlmann AR (2012) Hemocompatibility of poly(ɛ-caprolactone) lipid-core nanocapsules stabilized with polysorbate 80-lecithin and uncoated or coated with chitosan, International Journal of Pharmaceutics Volume 426, Issues 1–2, 15 April 2012, Pages 271–279
  5. Bhise NS, Gray RS, Sunshine JC, Htet S, Ewald AJ. and Green JJ. (2010), The relationship between terminal functionalization and molecular weight of a gene delivery polymer and transfection efficacy in mammary epithelial 2-D cultures and 3-D organotypic cultures, Biomaterials, DOI:10.1016/j.biomaterials.2010.07.023
  6. Bhise NS, Wahlin KJ, Zack DJ, Green J (2013) Evaluating the potential of poly (beta-amino ester) nanoparticles for reprogramming human fibroblasts to become induced pluripotent stem cells, International Journal of Nanomedicine, http://dx.doi.org/10.2147/IJN.S53830
  7. Blagbrough IS., Metwally AA., and Ghonaim HM (2012), Asymmetrical N4,N9-Diacyl Spermines: SAR Studies of Non-Viral Lipopolyamine Vectors for Efficient siRNA Delivery with Silencing of EGFP Reporter Gene, Mol. Pharmaceutics, Just Accepted Manuscript, DOI: 10.1021/mp200428d
  8. Bloembergen S, Mclennan IJ, Jones N, Wagner R, Shermon AKG, Elsayed AR and Liu J (2013) Aptamer bioconjugate drug delivery device, United States Patent Application 20130090467
  9. Bhuiyan DB (2010) Application of hyperthermia for localized drug release from thermosensitive liposomes, Master's Thesis in Biomedical Engineering, Chalmers University of Technology, Goteborg, Sweden 2010.
  10. Bult W, Varkevisser R, Soulimani F, Seevinck PR, de Leeuw H, Bakker CJG, Luijten PR, van het Schip AD, Hennink WE and Nijsen JFW (2010) Holmium Nanoparticles: Preparation and In vitro Characterization of a New Device for Radioablation of Solid Malignancies, Pharmaceutical Research, DOI: 10.1007/s11095-010-0226-3 Online First Open Access, Research Paper
  11. Case RI (2013) Methods Of Separating Nucleic Acid Polymer Conjugates United States Patent Application 20130236986
  12. Chen H and Walsh S (2013) Polymer Protein Microparticles, - US Patent 20,130,129,830, 2013
  13. Cheng K, Zhou Y-M, Sun Z-Y, Hu H-B, Zhong H, Kong X-K and Chen Q-W (2012) Synthesis of carbon-coated, porous and water-dispersive Fe3O4 nanocapsules and their excellent performance for heavy metal removal applications, Dalton Trans., 2012, Advance Article, DOI: 10.1039/C2DT12312F
  14. Ciolkowski M, Rozanek M, Szewczyk M, Klajnert B and Bryszewska M (2011), The influence of PAMAM-OH dendrimers on the activity of human erythrocytes ATPases, Biochimica et Biophysica Acta (BBA) - Biomembranes, Article in Press, doi:10.1016/j.bbamem.2011.07.021
  15. Corradetti B, Freile P, Pells S, Bagnaninchi P, Park J, Fahmy TM, de Sousa PA (2012) Paracrine signalling events in embryonic stem cell renewal mediated by affinity targeted nanoparticles, Biomaterials, http://dx.doi.org/10.1016/j.biomaterials.2012.06.011
  16. Cunha-Azevedo EP (2011) Biodegradable nanoparticles of PLGA, covered with DMSA, containing itraconazole for treatment of Paracoccidioidomycosis.PhD Thesis (PhD in health sciences)-University of Brasília, Brasília, 2011.
  17. Debotton N, Harush-Frenkel O, Gofrit O and Benita S (2010) Antibody-nanocarrier conjugates for drug targeting and improved cancer therapy, Unither Nanomedical & Telemedical Technology Conference – The Future’s Approach to Medicine, Hotel Manoir Des Sables, Orford (Quebec), Canada 23-26th February, 2010.
  18. Dellinger A, Brooks DB, Plunkett B, Vonakis BM, Sandros M (2012) Effects of Novel Nanomaterials on Allergic Mediator Release from Human Mast Cells and Basophils through Non-IgE Mediated Pathways. J Nanomed Nanotechol 3:153.doi:10.4172/2157-7439.1000153
  19. De Jong WH and Borm PJ (2008) Drug delivery and nanoparticles: Applications and hazards, International Journal of Nanomedicine, Volume 3(2), Pages 133 – 149
  20. Demirci H, Sierra RG, Laksmono H, Shoeman RL, Botha S, Barends TRM, Nass K, Schlichting I, . Doak RB, Gati C, Williams GJ, Boutet S, Messerschmidt M, Jog lG, Dahlberg AE, Gregory ST and Bogan MJ (2013) Serial femtosecond X-ray diffraction of 30S ribosomal subunit microcrystals in liquid suspension at ambient temperature using an X-ray free-electron laser, Acta Crystallographica Section F, Structural Biology and Crystallization Communications, Volume 69, Part 9 (September 2013)
  21. Demento SL, Bonafé N, Cui W, Kaech SM, Caplan MJ, Fikrig E Ledizet M and Fahmy T M (2010) TLR9-Targeted Biodegradable Nanoparticles as Immunization Vectors Protect against West Nile Encephalitis, The Journal of Immunology September 1, 2010 vol. 185 no. 5 2989-2997
  22. Dimitrova M (2011) Applications of sub-visible particle analysis in the development of protein therapeutics, Proc FIP Pharmaceutical Sciences 2010 World Congress, November 14-18, 2010, Morial Convention Center, New Orleans, Louisiana, USA.
  23. Do TT, Tang VJ, Aguilera JA, Perry CC and Milligan JR (2011) Characterization of a Lipophilic Plasmid DNA Condensate Formed with a Cationic Peptide Fatty Acid Conjugate, Biomacromolecules, Articles ASAP (As Soon As Publishable), Publication Date (Web): March 16, 2011 (Article)
  24. Ezzat K, Zaghloul EM., EL Andaloussi S, Lehto T, El-Sayed R, Magdy T, Smith ECI and Langel Ü (2012) Solid formulation of cell-penetrating peptide nanocomplexes with siRNA and their stability in simulated gastric conditions, Journal of Controlled Release, Volume 162, Issue 1, 20 August 2012, Pages 1–8
  25. Figueiredo M (2013) Sizing Nanoparticles in Liquids: An Overview of Methods, Drug Delivery Systems: Advanced Technologies Potentially Applicable in Personalised Treatment, Advances in Predictive, Preventive and Personalised Medicine, Volume 4, 2013, pp 87-107
  26. Garrier J,Reshetov V,Gräfe S, Guillemin F, Zorin V, Bezdetnaya L (2013) Factors affecting the selectivity of nanoparticle-based photoinduced damage in free and xenografted chorioallantoïc membrane model, Journal of Drug Targeting 0 0:0, 1-12
  27. Geng X, Ye H, Feng Z, Lao X, Zhang L, Huang J, Wu Z-R (2012) Synthesis and characterization of cisplatinloaded, EGFR-targeted biopolymer and in vitro evaluation for targeted delivery J Biomed Mater Res Part A 2012:00A:000.
  28. Ghonaim HM, Li S, Soltan MK, Pourzand C and Blagbrough IS (2007a), Chain Length Modulation in Symmetrical Lipopolyamines and the effect on Nanoparticle Formulations for Gene Delivery, in British Pharmaceutical Conference BPC2007, Manchester, 10th Sept.
  29. Ghonaim HM, Li S, Pourzand C and Blagbrough IS (2007b), Efficient Novel Unsymmetrical Lipopolyamine Formulations for Gene Delivery, in British Pharmaceutical Conference BPC2007, Manchester, 10th Sept.
  30. Ghonaim HM, Li S, Pourzand C and Blagbrough IS (2007c), Formulation and Delivery of Fluorescent siRNA by Lipospermine Nanoparticle Complex Formation, in British Pharmaceutical Conference BPC2007, Manchester, 10th Sept.
  31. Ghonaim HM, (2008) Design and Development of Pharmaceutical Dosage Forms for Gene and siRNA Delivery, PhD Thesis University of Bath, Department of Pharmacy and Pharmacology, September 2008
  32. Ghonaim H, Li S and Blagbrough IS (2009) Very Long Chain N4 , N9 –Diacyl Spermines: Non-Viral Lipopolyamine Vectors for Efficient Plasmid DNA and siRNA Delivery Pharmaceutical Research, Volume 26, Number 1, p19-31
  33. Ghonaim HM, Li S and Blagbrough IS (2010) N1,N12-Diacyl Spermines: SAR Studies on Non-viral Lipopolyamine Vectors for Plasmid DNA and siRNA Formulation Pharmaceutical Research, Vol 27, (1) p17-29
  34. Haag R, Fischer W, Quadir MAl and Ofek P (2011), Compounds suited as nanocarriers for active agents and their use, United States Patent Application 20110082090
  35. Howard KA, Peer D (2013) Providing the full picture: a mandate for standardizing nanoparticle-based drug delivery, Nanomedicine (2013) 8(7), 1031–1033, ISSN 1743-5889 10.2217/NNM.13.95 © 2013 Future Medicine Ltd
  36. Hsu J, Serrano D, Bhowmick T, Kumar K, Shen Y, Kuo YC, Garnacho C and Muro S (2010), Enhanced Endothelial Delivery and Biochemical Effects of α-Galactosidase by ICAM-1-Targeted Nanocarriers for Fabry Disease, Journal of Controlled Release, Article in Press, DOI:10.1016/j.jconrel.2010.10.031
  37. Jensen LB, Pavan GM, Kasimova MR, Rutherford S, Danani A, Nielsen HM and Foged C (2011) Elucidating the molecular mechanism of PAMAM-siRNA dendriplex self-assembly: Effect of dendrimer charge density, International Journal of Pharmaceutics, Article in Press, Accepted Manuscript, doi:10.1016/j.ijpharm.2011.03.015
  38. Katzer T, Chaves P, Bernardi A, Pohlmann AR, Guterres SS and Beck RC (2014) Castor oil and mineral oil nanoemulsion: development and compatibility with a soft contact lens.Pharm Dev Technol 19 (2) p232-7. DOi:10.3109/10837450.2013.769569. Epub 2013 Feb 25.
  39. Kendall M, Ding P, Kendall K and Clark H (2010), Particles Interact with Key Components of Lung Lining Fluid, American Association for Aerosol Research (AAAR) Specialty Conference – Air Pollution and Health: Bridging the Gap from Sources to Health Outcomes March 22 – 26 ,San Diego, CA
  40. Kim J, Sunshine JC, Green JJ (2013), Bioconjugate Chem., Article ASAP, DOI:10.1021/bc4002322
  41. Klyachko NL, Haney MJ, Zhao Y, Manickam DS, Mahajan V, Suresh P, Hingtgen SD, Mosley LR, Gendelman HE, Kabanov AV & Batrakova EV (2013) Macrophages offer a paradigm switch for CNS delivery of therapeutic proteins , Nanomedicine, Posted online on November 18, 2013, (doi:10.2217/nnm.13.115)
  42. Kolluru LP, Rizvi SAA., D’Souza M, and D’Souza MJ (2012) Formulation development of albumin based theragnostic nanoparticles as a potential delivery system for tumor targeting, Journal of Drug Targeting, Ahead of Print : Pages 1-10, (doi: 10.3109/1061186X.2012.729214)
  43. Kooijmans SAA, Stremersch S, Braeckmans K, de Smedt S, Hendrix A, Wood MJA, Schiffelers RM, Raemdonck K, Vader P (2013) Electroporationinduced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles, Journal of Controlled Release, http://dx.doi.org/10.1016/j.jconrel.2013.08.014 , Available online 29 August 2013
  44. Kozielski KL, Tzeng SY and Green JJ (2013) A bioreducible linear poly(β-amino ester) for siRNA delivery, Chem. Commun., 2013, Accepted Manuscript, DOI:10.1039/C3CC40718G
  45. Kumar A, Saienni AE, Dixit N (2013) Synthesis and Characterization of Nanoencapsulated Drugs, in Nanomedicine in Drug Delivery (Eds Arun Kumar, Heidi M. Mansour, Adam Friedman, Eric R. Blough), CRC Press, 2013, Ch 2, p Page 41.
  46. Kumru O S, Liu J, Ji JA, Cheng W, Wang YJ, Wang T, Joshi SB, Midduagh CR and Volkin DB (2012), Compatibility, physical stability, and characterization of an IgG4 monoclonal antibody after dilution into different intravenous administration bags. J. Pharm. Sci.. doi: 10.1002/jps.23224
  47. Lien C-F, Molnár É, Toman P, Tsibouklis J, Pilkington GJ, Górecki DC and Barbu E (2012) In vitro Assessment of Alkylglyceryl-Functionalized Chitosan Nanoparticles as Permeating Vectors for the Blood–Brain Barrier, Biomacromolecules, 2012, 13 (4), pp 1067–1073, Publication Date (Web): March 11, 2012 (Article), DOI: 10.1021/bm201790s
  48. Look M, Saltzman WM, Craft J, Fahmy TM (2013) The nanomaterial-dependent modulation of dendritic cells and its potential influence on therapeutic immunosuppression in lupus, Biomaterials, Available online 1 November 2013, http://dx.doi.org/10.1016/j.biomaterials.2013.10.046
  49. Lundin P (2011), Delivery of gene-regulating agents: internalization mechanisms and novel vectors, PhD Thesis, Karolinska Institute, Stockholm, ISBN 978-91-7457-432-6
  50. Maher S, Leonard TW, Jacobsen J and Brayden DJ (2009) Safety and efficacy of sodium caprate in promoting oral drug absorption: from in vitro to the clinic, Advanced Drug Delivery Reviews, Volume 61, Issue 15, 17 December 2009, Pages 1427-1449
  51. Malam Y, Lim E and Seifalian, A.(2011) Current Trends in the Application of Nanoparticles in Drug Delivery, Current Medicinal Chemistry, Volume 18, Number 7, March 2011 , pp. 1067-1078(12)
  52. Malmo J (2012) Chitosan-based nanocarriers for gene-and siRNA-delivery, PhD Thesis, Norwegian University of Science and Technology, Faculty of Natural Sciences and Technology, Department of Biotechnology, ISBN 978-82-471-3748-2 (electronic ver.)
  53. Mbah CC, Builders PF, Attama AA (2013) Nanovesicular carriers as alternative drug delivery systems: ethosomes in focus, International Journal of Nanomedicine, January 2014, Vol. 11, No. 1 , Pages 45-59 (doi:10.1517/17425247.2013.860130)
  54. McNeil SE (2011a) Unique Benefits of Nanotechnology to Drug Delivery and Diagnostics: in Characterization of Nanoparticles Intended for Drug Delivery (Ed Scott E. McNeil) , Methods in Molecular Biology, 2011, Volume 697, Part 1, 3-8, DOI: 10.1007/978-1-60327-198-1_1
  55. McNeil SE (2011b) Challenges for Nanoparticle Characterization: in Characterization of Nanoparticles Intended for Drug Delivery (Ed Scott E. McNeil) , Methods in Molecular Biology, 2011, Volume 697, Part 1, 3-8, DOI:10.1007/978-1-60327-198-1_1
  56. Mendes LP, Gaeti MPN, de Ávila PHM, Vieira MdS, Rodrigues BdS, Marcelino RIdÁ, dos Santos LCR, Valadares MC, Lima EM (2013) Multicompartimental Nanoparticles for Co-Encapsulation and Multimodal Drug Delivery to Tumor Cells and Neovasculature, Pharmaceutical Research, October 2013, DOI 10.1007/s11095-013-1234-x
  57. Miller K, Erez R, Segal E, Shabat D, Satchi-Fainaro R (2009), Targeting Bone Metastases with a Bispecific Anticancer and Antiangiogenic Polymer-Alendronate-Taxane Conjugate, Angewandte Chemie International Edition, 48 (16) 2949-2954
  58. Moddaresi M, Brown MB, Zhao Y, Tamburic S and Jones SA (2010) The role of vehicle–nanoparticle interactions in topical drug delivery International Journal of Pharmaceutics, Volume 400, Issues 1-2, Pages 176-182
  59. Moquin A and Winnik FM (2012) The Use of FieldFlow Fractionation for the Analysis of Drug and Gene Delivery Systems, in “Field-Flow Fractionation in Biopolymer Analysis” (S. Kim R. Williams, Karin D. Caldwell (Eds)) ISBN3709101530, Chapter 13, p 187-207
  60. Morch Y, Stenstad P, Schmid R, Hansen R, Berg S, Eggen S, Aslund A, von Hartmann E, Afadzi M and Boe A (2013) Ultrasound mediated delivery of a novel Nanoparticle-Microbubble platform, European CLINAM and ETPN Conference, Basel, June 23-26.2013.
  61. Mostaghaci B, Loretz B, Haberkorn R, Kickelbick G, and Lehr C-M (2013) Onestep synthesis of nano-sized and stable amino-functionalized calcium phosphate particles for DNA transfection, Chem. Mater., Just Accepted Manuscript, DOI:10.1021/cm401886u, Publication Date (Web): August 21, 2013
  62. Mund R (2013) Titania Nanoparticles for the Intracellular Delivery of Paclitaxel in Breast Cancers, B Tech Thesis, http://ethesis.nitrkl.ac.in/5055/1/109BM0658.pdf
  63. Nassar T, Rom A, Nyska A and Benita S (2009) Novel double coated nanocapsules for intestinal delivery and enhanced oral bioavailability of tacrolimus, a P-gp substrate drug, Journal of Controlled Release, Volume 133, Issue 1, Pages 77-84
  64. Nyska A and Benita S (2009) Novel double coated nanocapsules for intestinal delivery and enhanced oral bioavailability of tacrolimus, a P-gp substrate drug, Journal of Controlled Release, Vol 133, Issue 1, Pages 77-84
  65. Ofek P, Fischer W, Calderón M, Haag R, and Satchi-Fainaro R (2010), In vivo delivery of small interfering RNA to tumors and their vasculature by novel dendritic nanocarriers, FASEB J. DOI: 10.1096/fj.09-149641
  66. Papanikolaou E, Kontostathi G, Drakopoulou E, Georgomanoli M, Stamateris E, Vougas K, Vlahou A, Malloy A, Ware M, Anagnou NP. (2013) Characterization and comparative performance of lentiviral vector preparations, concentrated by either one-step ultrafiltration or ultracentrifugation, Virus Research 175 (2013) 1– 11, Available online 11 April 2013, http://dx.doi.org/10.1016/j.virusres.2013.03.015
  67. Park J, Gao W, Whiston R, Strom T, Metcalfe S and Fahmy TM (2010) Modulation of CD4+ T Lymphocyte Lineage Outcomes with Targeted, Nanoparticle-Mediated Cytokine Delivery, Mol. Pharmaceutics, 2011, 8 (1), pp 143–152
  68. Petersen MA, Hillmyer MA, and Kokkoli E (2013) Bioresorbable Polymersomes for Targeted Delivery of Cisplatin, Bioconjugate Chem., Just Accepted Manuscript, DOI: 10.1021/bc3003259, Publication Date (Web): March 22, 2013
  69. Parker AL., Bradshaw AC., Alba R, Nicklin SA., Baker AH (2013) Capsid Modification Strategies for Detargeting Adenoviral Vectors, Adenovirus; Methods in Molecular Biology Volume 1089, 2014, pp 45-59
  70. Ram M, Yaduvanshi K.S, Yadav H; Singh N, Mangla G, Shivakumar H (2011) Nanoparticles, Promising Carriers in Drug Targeting: A Review Current Drug Therapy, Volume 6, Number 2, May 2011 , pp. 87-96(10)
  71. Reis CP, Silva C, Martinho N and Rosado C (2013) Review; Drug carriers for oral delivery of peptides and proteins: accomplishments and future perspectives, Therapeutic Delivery February 2013, Vol. 4, No. 2, Pages 251-265 , DOI 10.4155/tde.12.143 , (doi:10.4155/tde.12.143)
  72. Piotrowski M, Szczepanowicz K, Jantas D, Leśkiewicz M, Lasoń W, Warszyński P (2013) Emulsion-core and polyelectrolyte-shell nanocapsules: biocompatibility and neuroprotection against SH-SY5Y cells, Journal of Nanoparticle Research, October 2013, 15:2035
  73. Rotan O, Sokolova V, Gilles P, Hu W, Dutt S, Schrader T and Epple M (2013), Transport of supramolecular drugs across the cell membrane by calcium phosphate nanoparticles. Mat.-wiss. u. Werkstofftech., 44: 176–182. doi: 10.1002/ mawe.201300085
  74. Sander A.A. Kooijmansa SAA, Stremerschb S, Braeckmans K, de Smedt SC, Hendrix A, Wood MJA, Schiffelers RM, Raemdonck K, and Vader P. (2013) Electroporation-induced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles. J Controlled Release 172 (1) p 229-238. DOI:10.1016/j.jconrel.2013.08.014
  75. Sangwai M, Sardar S, Vavia P (2012) Nanoemulsified orlistat-embedded multi-unit pellet system (MUPS) with improved dissolution and pancreatic lipase inhibition, Pharmaceutical Development and Technology, Posted online on December 24, 2012. (doi:10.3109/10837450.2012.751404).
  76. Satchi-fainaro R and Docon V, Jesus M (2011), Novel conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases, United States Patent Application 20110286923
  77. Segal E, Pan H, Ofek P, Udagawa T, Kopečková P, Kopeček J and Satchi-Fainaro R (2009) Targeting Angiogenesis-Dependent Calcified Neoplasms Using Combined Polymer Therapeutics, PLoS ONE. 2009; 4(4): e5233.
  78. Shimizu C, Kim J, Stepanowsky P, Trinh C, Lau HD (2013) Differential Expression of miR-145 in Children with Kawasaki Disease. PLoS ONE 8(3):e58159.doi:10.1371/journal.pone.0058159
  79. Shirali AC, Look M, Du W, Kassis E, Stout-Delgado HW, Fahmy TM and Goldstein DR (2011), Nanoparticle Delivery of Mycophenolic Acid Upregulates PD-L1 on Dendritic Cells to Prolong Murine Allograft Survival. American Journal of Transplantation. doi: 10.1111/j.1600-6143.2011.03725.x, Article first published online: 30 AUG 2011
  80. Shmueli RB, Bhise NS, Green JJ (2013) Evaluation of Polymeric Gene Delivery Nanoparticles by Nanoparticle Tracking Analysis and High-throughput Flow Cytometry, J Vis Exp. 2013 Mar 1;(73). doi: 10.3791/50176.
  81. Sokolova V, Knuschke T, Kovtun A, Buer J, Epple M and Westendorf AM (2010) The use of calcium phosphate nanoparticles encapsulating Toll-like receptor ligands and the antigen hemagglutinin to induce dendritic cell maturation and T cell activation, Biomaterials, Vol 31, Issue 21, July 2010, Pages 5627-33
  82. Sokolova V, Knuschke T, Buer J, Westendorf AM and Epple M (2012a), Quantitative determination of the composition of multi-shell calcium phosphate–oligonucleotide nanoparticles and their application for the activation of dendritic cells, Acta Biomaterialia, Article in Press, 7, 4029–4036, doi:10.1016/j.actbio.2011.07.010
  83. Sokolova V, Rotan O, Klesing J, Nalbant P, Buer J, Knuschke T, Westendorf AM and Epple M (2012) Calcium phosphate nanoparticles as versatile carrier for small and large molecules across cell membranes Journal of Nanoparticle Research, Volume 14, Number 6 (2012b), 910, DOI: 10.1007/s11051-012-0910-9
  84. Soltan MK, Ghonaim HM, El Sadek M, Kull MA, El-aziz LA and Blagbrough IS (2009), Design and Synthesis of N4, N9-Disubstituted Spermines for Non-viral siRNA Delivery – Structure-Activity Relationship Studies of siFection Efficiency Versus Toxicity, Pharmaceutical Research, Volume 26, Number 2, p 286-295
  85. Stremersch S (2013) Encapsulation of siRNA into Extracellular Vesicles by Electroporation is Biased by siRNA Precipitation, Molecular Medicine Congress, 3-4 September 2013, Frankfurt, Germany
  86. Sunshine JC, Sunshine SB, Bhutto I, Handa JT, Green JJ (2012) Poly(β-Amino Ester)-Nanoparticle Mediated Transfection of Retinal Pigment Epithelial Cells In vitro and In vivo. PLoS ONE 7(5): e37543. doi:10.1371/journal.pone.0037543
  87. Tagalakis AD, Grosse SM, Meng Q-H, Mustapa MFM, Kwok A, Salehi SE, Tabor AB, Hailes HC and Hart SL (2010) Integrin-targeted nanocomplexes for tumor specific delivery and therapy by systemic administration Biomaterials, Vol 32, Issue 5, February 2011, p1370-6
  88. Troiber CM (2013) Sequence-defined polycationic oligomers for nucleic acid delivery, PhD Thesis, University of Munich, http://edoc.ub.unimuenchen.de/15343/1/Troiber_Christina.pdf
  89. Tzeng SY., Guerrero-Cázares H, Martinez EE., Sunshine J C., Quiñones-Hinojosa A and Green JJ (2011), Non-viral gene delivery nanoparticles based on Poly(β-amino esters) for treatment of glioblastoma, Biomaterials, Article in Press, DOI:10.1016, j.biomaterials.2011.04.016
  90. Tzeng SY, Hung BP., Grayson WL, Green JJ (2012) Cystamine-terminated poly (beta-amino ester)s for siRNA delivery to human mesenchymal stem cells and enhancement of osteogenic differentiation Biomaterials, Volume 33, Issue 32, November 2012, Pages 8142–8151
  91. Tzeng SY and Green JJ (2012), Subtle Changes to Polymer Structure and Degradation Mechanism Enable Highly Effective Nanoparticles for siRNA and DNA Delivery to Human Brain Cancer. Advanced Healthcare Materials. doi: 10.1002/adhm.201200257
  92. van Gael EVB, van Eijk R, Oosting RS, Kok RJ, Hennink WE., Crommelin DJA and Mastrobattista E (2011), How to screen non-viral gene delivery systems in vitro? Journal of Controlled Release, Article in Press, DOI:10.1016/j.jconrel.2011.05.001
  93. Verderio P, Bonetti P, Colombo M, Pandolfi L, and Prosperi D (2013) Intracellular drug release from curcumin-loaded PLGA nanoparticles induces G2/M block in breast cancer cells, Biomacromolecules, Just Accepted Manuscript, DOI: 10.1021/ bm3017324, Publication Date (Web): January 27, 2013
  94. Widjaja LK, Bora M, Chan PNPH, Lipik V, Wong TTL, Venkatraman SS. (2013) Hyaluronic acid-based nanocomposite hydrogels for ocular drug delivery applications. J Biomed Mater Res Part A 2013:00A:000–000
  95. Wiley DT, Webster P, Gale A, Davis ME (2013) Transcytosis and brain uptake of transferrin-containing nanoparticles by tuning avidity to transferrin receptor, PNAS, 10.1073/pnas.1307152110
  96. Witwer KW, McAlexander MA, Queen SE, Adams RJ (2013) Real-time quantitative PCR and droplet digital PCR for plant miRNAs in mammalian blood provide little evidence for general uptake of dietary miRNAs: Limited evidence for general uptake of dietary plant xenomiRs. RNA Biology, 2013; 10(7), http://dx.doi.org/10.4161/rna.25246, www.landesbioscience.com/journals/rnabiology/article/25246/
  97. Yandrapu SK, Kanujia P, Chalasani K, Mangamoori L, Kolapalli RV and Chauhan A (2012) Development and optimization of thiolated dendrimer as a viable mucoadhesive excipient for the controlled drug delivery: An acyclovir model formulation, Nanomedicine: Nanotechnology, Biology and Medicine, doi:10.1016/j.nano.2012.10.005
  98. Zanotto-Filho A, Coradini K, Braganhol E, Schröder R, de Oliveira C M, Simões-Pires A, Battastini OAM, Pohlmann AR, Guterres SS, Forcelini CM, Beck RCR and Moreira JCF (2012) Curcumin-loaded lipid-core nanocapsules as a strategy to improve pharmacological efficacy of curcumin in glioma treatment, European Journal of Pharmaceutics and Biopharmaceutics, http://dx.doi.org/10.1016/j.ejpb.2012.10.019, Available online 28 November 2012
  99. Zhao J, Mi Y, Liu Y, Feng S-S (2011), Quantitative control of targeting effect of anticancer drugs formulated by ligand-conjugated nanoparticles of biodegradable copolymer blend, Biomaterials, Volume 33, Issue 6, February 2012, Pages 1948-1958
  100. Zhu T, Jiang Z, Ma Y (2012) Lipid Exchange between Membranes: Effects of Membrane Surface Charge, Composition, and Curvature, Colloids and Surfaces B: Biointerfaces http://dx.doi.org/10.1016/j.colsurfb.2012.04.024 Available online 25 April 2012.
  101. Zuckerman JE, Choi CHJ., Han H, and Davis ME (2012), Polycation-siRNA nanoparticles can disassemble at the kidney glomerular basement membrane, PNAS, February 6, 2012, doi: 10.1073/pnas.1200718109

About Malvern Panalytical

Malvern Panalytical provides the materials and biophysical characterization technology and expertise that enable scientists and engineers to understand and control the properties of dispersed systems.

These systems range from proteins and polymers in solution, particle and nanoparticle suspensions and emulsions, through to sprays and aerosols, industrial bulk powders and high concentration slurries.

Used at all stages of research, development and manufacturing, Malvern Panalytical’s materials characterization instruments provide critical information that helps accelerate research and product development, enhance and maintain product quality and optimize process efficiency.


Sponsored Content Policy: News-Medical.net publishes articles and related content that may be derived from sources where we have existing commercial relationships, provided such content adds value to the core editorial ethos of News-Medical.Net which is to educate and inform site visitors interested in medical research, science, medical devices and treatments.

Last updated: May 31, 2023 at 9:58 AM

Citations

Please use one of the following formats to cite this article in your essay, paper or report:

  • APA

    Malvern Panalytical. (2023, May 31). Applications of Nanoparticle Tracking Analysis in Drug and Gene Delivery. News-Medical. Retrieved on April 18, 2024 from https://www.news-medical.net/whitepaper/20160606/Applications-of-Nanoparticle-Tracking-Analysis-in-Drug-and-Gene-Delivery.aspx.

  • MLA

    Malvern Panalytical. "Applications of Nanoparticle Tracking Analysis in Drug and Gene Delivery". News-Medical. 18 April 2024. <https://www.news-medical.net/whitepaper/20160606/Applications-of-Nanoparticle-Tracking-Analysis-in-Drug-and-Gene-Delivery.aspx>.

  • Chicago

    Malvern Panalytical. "Applications of Nanoparticle Tracking Analysis in Drug and Gene Delivery". News-Medical. https://www.news-medical.net/whitepaper/20160606/Applications-of-Nanoparticle-Tracking-Analysis-in-Drug-and-Gene-Delivery.aspx. (accessed April 18, 2024).

  • Harvard

    Malvern Panalytical. 2023. Applications of Nanoparticle Tracking Analysis in Drug and Gene Delivery. News-Medical, viewed 18 April 2024, https://www.news-medical.net/whitepaper/20160606/Applications-of-Nanoparticle-Tracking-Analysis-in-Drug-and-Gene-Delivery.aspx.

Other White Papers by this Supplier

While we only use edited and approved content for Azthena answers, it may on occasions provide incorrect responses. Please confirm any data provided with the related suppliers or authors. We do not provide medical advice, if you search for medical information you must always consult a medical professional before acting on any information provided.

Your questions, but not your email details will be shared with OpenAI and retained for 30 days in accordance with their privacy principles.

Please do not ask questions that use sensitive or confidential information.

Read the full Terms & Conditions.