Skip to main content
Log in

Sizing up the Next Generation of Nanomedicines

  • Perspective
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

During the past two decades the nanomedicine field has experienced significant progress. To date, over sixty nanoparticle (NP) formulations have been approved in the US and EU while many others are in clinical or preclinical development, indicating a concerted effort to translate promising bench research to commercially viable pharmaceutical products. The use of NPs as novel drug delivery systems, for example, can improve drug safety and efficacy profiles and enable access to intracellular domains of diseased cells, thus paving the way to previously intractable biological targets. However, the measurement of their physicochemical properties presents substantial challenges relative to conventional injectable formulations. In this perspective, we focus exclusively on particle size, a core property and critical quality attribute of nanomedicines. We present an overview of relevant state-of-the-art technologies for particle sizing, highlighting the main parameters that can influence the selection of techniques suitable for a specific size range or material. We consider the increasing need, and associated challenge, to measure size in physiologically relevant media. We detail the importance of standards, key to validate any measurement, and the need for suitable reference materials for processes used to characterize novel and complex NPs. This perspective highlights issues critical to achieve compliance with regulatory guidelines and to support research and manufacturing quality control.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Abbreviations

CDER:

FDA Center for Drug Evaluation and Research

EMA:

European Medicine Agency

FDA:

US Food and Drug Administration

ISO:

International Organization for Standardization

JRC:

European Commission Joint Research Centre

NIST:

National Institute of Standards and Technology

US NCL:

US National Cancer Institute, Nanotechnology Characterization Laboratory

EU NCL:

European Union NanoMedicine Characterization Laboratory

SRM:

Standard Reference Material (NIST trademarked product)

RM:

Reference Material

References

  1. Freitas RA Jr. What is nanomedicine? Nanomedicine. 2005;1(1):2–9.

    Article  CAS  Google Scholar 

  2. Koo OM, Rubinstein I, Onyuksel H. Role of nanotechnology in targeted drug delivery and imaging: a concise review. Nanomedicine. 2005;1(3):193–212.

    Article  CAS  Google Scholar 

  3. Halamoda-Kenzaoui B, Calzolai L, Urban P, Zuang V, Baconnier S, Boisseau P, et al. Bridging communities in the field of nanomedicine. Regul Toxicol Pharmacol. 2019;106:187–96.

    Article  Google Scholar 

  4. Faria M, Bjornmalm M, Thurecht KJ, Kent SJ, Parton RG, Kavallaris M, et al. Minimum information reporting in bio-nano experimental literature. Nat Nanotechnol. 2018;13(9):777–85.

    Article  CAS  Google Scholar 

  5. D'Mello SR, Cruz CN, Chen M-L, Kapoor M, Lee SL, Tyner KM. The evolving landscape of drug products containing nanomaterials in the United States. Nat Nanotechnol. 2017;12(6):523–9.

    Article  CAS  Google Scholar 

  6. Evans ER, Bugga P, Asthana V, Drezek R. Metallic nanoparticles for cancer immunotherapy. Mater Today (Oxford, U K). 2018;21(6):673–685.

    Article  CAS  Google Scholar 

  7. Svenson S. The dendrimer paradox - high medical expectations but poor clinical translation. Chem Soc Rev. 2015;44(12):4131–44.

    Article  CAS  Google Scholar 

  8. Tsai D-H, Lu Y-F, Del Rio FW, Cho TJ, Guha S, Zachariah MR, et al. Orthogonal analysis of functional gold nanoparticles for biomedical applications. Anal Bioanal Chem. 2015;407(28):8411–22.

    Article  CAS  Google Scholar 

  9. Clogston JD, Crist RM, SE MN. Physicochemical Characterization of Polymer Nanoparticles: Challenges and Present Limitations. In: Vauthier C, Ponchel G, editors. Polymer Nanoparticles for Nanomedicines A Guide for their Design, Preparation and Development. Cham: Springer; 2016. p. 641.

    Google Scholar 

  10. Renaud J-P, Chari A, Ciferri C. Liu W-t, Remigy H-W, stark H, Wiesmann C. Cryo-EM in drug discovery: achievements, limitations and prospects. Nat Rev Drug Discov. 2018;17(7):471–92.

    Article  CAS  Google Scholar 

  11. Gigault J, Pettibone JM, Schmitt C, Hackley VA. Rational strategy for characterization of nanoscale particles by asymmetric-flow field flow fractionation: a tutorial. Anal Chim Acta. 2014;809:9–24.

    Article  CAS  Google Scholar 

  12. Pitkanen L, Striegel AM. Size-exclusion chromatography of metal nanoparticles and quantum dots. TrAC Trends Anal Chem. 2016;80:311–20.

    Article  CAS  Google Scholar 

  13. Zattoni A, Roda B, Borghi F, Marassi V, Reschiglian P. Flow field-flow fractionation for the analysis of nanoparticles used in drug delivery. J Pharm Biomed Anal. 2014;87:53–61.

    Article  CAS  Google Scholar 

  14. ISO. Nanotechnologies -- Analysis of nano-objects using asymmetrical-flow and centrifugal field-flow fractionation. ISO Technical Specification 21362:2018. In.; 2018. p. 38.

  15. Weatherall E, Willmott GR. Applications of tunable resistive pulse sensing. Analyst (Cambridge, U K). 2015;140(10):3318–3334.

    Article  CAS  Google Scholar 

  16. Maguire CM, Prina-Mello A, Maguire CM, Prina-Mello A, Rosslein M, Wick P. Characterisation of particles in solution - a perspective on light scattering and comparative technologies. Sci Technol Adv Mater. 2018;19(1):732–45.

    Article  CAS  Google Scholar 

  17. Maas SLN, de Vrij J, van der Vlist EJ, Geragousian B, van Bloois L, Mastrobattista E, et al. Possibilities and limitations of current technologies for quantification of biological extracellular vesicles and synthetic mimics. J Control Release. 2015;200:87–96.

    Article  CAS  Google Scholar 

  18. Caputo F, Clogston J, Calzolai L, Rosslein M, Prina-Mello A. Measuring particle size distribution of nanoparticle enabled medicinal products, the joint view of EUNCL and NCI-NCL. A step by step approach combining orthogonal measurements with increasing complexity. J Control Release. 2019;299:31–43.

    Article  CAS  Google Scholar 

  19. Taylor G. Dispersion of soluble matter in solvent flowing slowly through a tube. Proc R Soc London, Ser A. 1953;219:186–203.

    Article  CAS  Google Scholar 

  20. Cottet H, Martin M, Papillaud A, Souaied E, Collet H, Commeyras A. Determination of Dendrigraft poly-L-lysine diffusion coefficients by Taylor dispersion analysis. Biomacromolecules. 2007;8(10):3235–43.

    Article  CAS  Google Scholar 

  21. Clogston JD, Patri AK. Importance of Physicochemical Characterization Prior to Immunological Studies. In: Dobrovolskaia MA, McNeil SE, editors. Handbook of Immunological Properties of Engineered Nanomaterials: World Scientific; 2013. p. 720.

  22. Grossman JH, Crist RM, Clogston JD. Early development challenges for drug Products containing Nanomaterials. AAPS J. 2017;19(1):92–102.

    Article  CAS  Google Scholar 

  23. Wolfram J, Suri K, Yang Y, Shen J, Celia C, Fresta M, et al. Shrinkage of pegylated and non-pegylated liposomes in serum. Colloids Surf, B. 2014;114:294–300.

    Article  CAS  Google Scholar 

  24. Gioria S, Caputo F, Urban P, Maguire CM, Bremer-Hoffmann S, Prina-Mello A, et al. Are existing standard methods suitable for the evaluation of nanomedicines: some case studies. Nanomedicine (London, U K). 2018;13(5):539–54.

    Article  CAS  Google Scholar 

  25. Capomaccio R, Ojea Jimenez I, Colpo P, Gilliland D, Ceccone G, Rossi F, et al. Determination of the structure and morphology of gold nanoparticle-HSA protein complexes. Nanoscale. 2015;7(42):17653–7.

    Article  CAS  Google Scholar 

  26. Ashby J, Schachermeyer S, Pan S, Zhong W. Dissociation-Based Screening of Nanoparticle-Protein Interaction via Flow Field-Flow Fractionation. Anal Chem (Washington, DC, U S). 2013;85(15):7494–501.

    Article  CAS  Google Scholar 

  27. Spadavecchia J, Movia D, Moore C, Maguire CM, Moustaoui H, Casale S, et al. Targeted polyethylene glycol gold nanoparticles for the treatment of pancreatic cancer: from synthesis to proof-of-concept in vitro studies. Int J Nanomedicine. 2016;11:791–822.

    Article  CAS  Google Scholar 

  28. Moustaoui H, Saber J, Djeddi I, Liu Q, Movia D, Prina-Mello A, et al. Lamy de la Chapelle M, Djaker N. a protein corona study by scattering correlation spectroscopy: a comparative study between spherical and urchin-shaped gold nanoparticles. Nanoscale. 2019;11(8):3665–73.

    Article  CAS  Google Scholar 

  29. Gollwitzer C, Bartczak D, Goenaga-Infante H, Kestens V, Krumrey M, Minelli C, et al. A comparison of techniques for size measurement of nanoparticles in cell culture medium. Anal Methods. 2016;8(26):5272–82.

    Article  CAS  Google Scholar 

  30. Sikora A, Shard AG, Minelli C. Size and ζ-potential measurement of silica nanoparticles in serum using tunable resistive pulse sensing. Langmuir. 2016;32(9):2216–24.

    Article  CAS  Google Scholar 

  31. Pal AK, Aalaei I, Gadde S, Gaines P, Schmidt D, Demokritou P, et al. High resolution characterization of engineered nanomaterial dispersions in complex media using tunable resistive pulse sensing technology. ACS Nano. 2014;8(9):9003–15.

    Article  CAS  Google Scholar 

  32. FDA US. Drug Products, Including Biological Products, that Contain Nanomaterials - Guidance for Industry- Draft Guidance Document. In: CDER, editor.; 2017. p. 29.

  33. ISO. Reference materials -- Selected terms and definitions. ISO Guide 30: 2015. In.; 2015. p. 8.

  34. German Federal Institute for Material Research and Testing (BAM), Nanoscaled Reference Materials. Available from: http://www.nano-refmat.bam.de/en/category_10_nanoobjects_nanoparticles_nanomaterials.htm.

  35. Tyner K. Regulatory considerations for drug products containing nanomaterials: US FDA perspective. In. Seminar presented at the US National Institute of Standards and Technology. Gaithersburg, MD; Jan 17, 2017.

  36. FDA US. Analytical Procedures and Methods Validation for Drugs and Biologics- Guidance for Industry. In: CDER, editor.; July 2015. p. 18.

  37. Report of the 2016 Global Summit on Regulatory Science (GSRS16) Nanotechnology Standards and Applications. In. Bathesda, Maryland: U.S. National Institutes of Health; 2016. p. 35.

  38. National Institute of Standards and Technology (NIST). Available from: https://www.nist.gov/.

  39. Nanotechnology Characterization Laboratory (NCL) webpage. Available from: https://ncl.cancer.gov/.

  40. EU Nanomedicine Characterisation Laboratory (EU NCL). Available from: http://www.euncl.eu/.

  41. FDA US. CDER’s Program for the Recognition of Voluntary Consensus Standards Related to Pharmaceutical Quality- Draft Guidance Document. In: CDER, editor.; 2019. p. 10.

  42. European Medicines Agency. Reflection paper on the data requirements for intravenous liposomal products developed with reference to an innovator liposomal product. EMA/CHMP/806058/2009/Rev. 02. . In. United Kingdom: Committee for Human Medicinal Product (CHMP); 2013.

  43. European Medicines Agency. Reflection paper on the data requirements for intravenous iron-based nano-colloidal products developed with reference to an innovator medicinal product. EMA/CHMP/SWP/620008/2012. In. Amsterdam: Committee for Medicinal Products for Human Use; 2015.

  44. Kaga S, Truong NP, Esser L, Senyschyn D, Sanyal A, Sanyal R, et al. Influence of size and shape on the biodistribution of nanoparticles prepared by polymerization-induced self-assembly. Biomacromolecules. 2017;18(12):3963–70.

    Article  CAS  Google Scholar 

  45. Mulholland GW, Bryner NP, Croarkin C. Measurement of the 100 nm NIST SRM 1963 by differential mobility analysis. Aerosol Sci Technol. 1999;31(1):39–55.

    Article  CAS  Google Scholar 

  46. Rauscher H, Roebben G, Mech A, Gibson N, Kestens V, Linsinger TPJ, Riego Sintes J. JRC Science for Policy Report. An overview of concepts and terms used in the European Commission's definition of nanomaterial In. Luxembourgh: Publications Office of the European Union; 2019. p. 44.

Download references

ACKNOWLEDGMENTS AND DISCLOSURES

J.D.C., V.A.H., A.P-M, S.P. and P.L.S. would like to thank all of the speakers who participated in the ACS symposium on “Particle Sizing of Nanoparticles: From Regulatory & Metrology Aspects to Application & Analysis” in Boston, MA and the speakers who participated in the two symposia organized at CLINAM 2018 in Basel, Switzerland. The authors thank Joseph Meyer, Leidos Biomedical Research, Inc., for graphic illustrations. The authors confirm that this article has no conflict of interest to declare in the contents of the manuscript. The identification of any commercial product or trade name does not imply endorsement or recommendation by the National Institute of Standards and Technology, the Nanotechnology Characterization Lab, AstraZeneca or Pfizer.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Jeffrey D. Clogston or Patrick Lim Soo.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

ESM 1

(DOCX 15 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Clogston, J.D., Hackley, V.A., Prina-Mello, A. et al. Sizing up the Next Generation of Nanomedicines. Pharm Res 37, 6 (2020). https://doi.org/10.1007/s11095-019-2736-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11095-019-2736-y

Keywords

Navigation