Skip to main content

Advertisement

Log in

Nanomaterial Drug Products: Manufacturing and Analytical Perspectives

  • Review Article
  • Theme: Nanotechnology in Complex Drug Products: Learning from the Past, Preparing for the Future
  • Published:
The AAPS Journal Aims and scope Submit manuscript

ABSTRACT

The increasing use of nanotechnology, including nanoparticles, in the preparation of drug products requires both manufacturing and analytical considerations in order to establish the quality metrics suitable for performance and risk assessment. A range of different nanoparticle systems exists including (but not limited to) nano-drugs, nano-additives, and nano-carriers. These systems generally require more complex production and characterization strategies than conventional pharmaceutical dosage forms. The advantage of using nanoparticle systems in pharmaceutical science is that the effective and desired function of the material can be designed through modern manufacturing processes. This paper offers a systematic nomenclature which allows for greater understanding of the drug product under evaluation based on available data from other nanoparticle reports. Analytical considerations of nano-drugs, nano-additives, and nano-carriers and the way in which they are measured are directly connected to quality control. Ultimately, the objective is to consider the entire nano-drug, nano-additive, and nano-carrier product life cycle with respect to its manufacture, use, and eventual fate. The tools and approaches to address the needs of these products exist; it should be the task of the pharmaceutical scientists and those in related disciplines to increase their understanding of nanomedicine and its novel products.

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
Fig. 3

Similar content being viewed by others

References

  1. Etheridge ML et al. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomed: Nanotechnol, Biol Med. 2013;9(1):1–14.

    CAS  Google Scholar 

  2. Foldvari M, Bagonluri M. Carbon nanotubes as functional excipients for nanomedicines: I. Pharmaceutical properties. Nanomed: Nanotechnol, Biol Med. 2008;4(3):173–82.

    CAS  Google Scholar 

  3. Duncan R, Gaspar R. Nanomedicine (s) under the microscope. Mol Pharmaceutics. 2011;8(6):2101–41.

    Article  CAS  Google Scholar 

  4. Weissig V, Pettinger TK, Murdock N. Nanopharmaceuticals (part 1): products on the market. Int J Nanomed. 2014;9:4357.

    Article  CAS  Google Scholar 

  5. Peer D et al. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751–60.

    Article  CAS  PubMed  Google Scholar 

  6. Barenholz YC. Doxil®—the first FDA-approved nano-drug: lessons learned. J Controlled Release. 2012;160(2):117–34.

    Article  CAS  Google Scholar 

  7. Haley, B. and E. Frenkel. Nanoparticles for drug delivery in cancer treatment. in Urologic Oncology: Seminars and original investigations. Elsevier;2008.

  8. Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Delivery Rev. 2004;56(11):1649–59.

    Article  CAS  Google Scholar 

  9. Bawarski WE et al. Emerging nanopharmaceuticals. Nanomed: Nanotechnol, Biol Med. 2008;4(4):273–82.

    CAS  Google Scholar 

  10. Tyner, K. and N. Sadrieh, Considerations when submitting nanotherapeutics to FDA/CDER for regulatory review. Characterization of nanoparticles intended for drug delivery, 2011: p. 17–31.

  11. De Jong WH, Borm PJ. Drug delivery and nanoparticles: applications and hazards. Int J Nanomed. 2008;3(2):133.

    Article  Google Scholar 

  12. Horcajada P et al. Porous metal-organic-framework nanoscale carriers as a potential platform for drug delivery and imaging. Nat Mater. 2010;9(2):172–8.

    Article  CAS  PubMed  Google Scholar 

  13. Hickey, A.J. and D. Ganderton, Statistical experimental design, in Pharmaceutical Process Engineering, Informa Healthcare: New York, NY;2010 p. 197–201.

  14. Mansour HM, Hickey AJ. Raman characterization and chemical imaging of biocolloidal self-assemblies, drug delivery systems, and pulmonary inhalation aerosols: a review. AAPS PharmSciTech. 2007;8, E99.

    Article  PubMed  Google Scholar 

  15. Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharm Res. 2007;24(1):1–16.

    Article  CAS  PubMed  Google Scholar 

  16. Torchilin VP. Multifunctional nanocarriers. Adv Drug Delivery Rev. 2012;64:302–15.

    Article  Google Scholar 

  17. Warheit DB et al. Health effects related to nanoparticle exposures: environmental, health and safety considerations for assessing hazards and risks. Pharmacol Ther. 2008;120(1):35–42.

    Article  CAS  PubMed  Google Scholar 

  18. Stebounova LV et al. Health and safety implications of occupational exposure to engineered nanomaterials. Wiley Interdisciplinary Reviews. Nanomed Nanobiotechnol. 2012;4(3):310–21.

    Article  CAS  Google Scholar 

  19. Wang X et al. A general strategy for nanocrystal synthesis. Nature. 2005;437(7055):121–4.

    Article  CAS  PubMed  Google Scholar 

  20. Grzelczak M et al. Shape control in gold nanoparticle synthesis. Chem Soc Rev. 2008;37(9):1783–91.

    Article  CAS  PubMed  Google Scholar 

  21. Wang Y, Xia Y. Bottom-up and top-down approaches to the synthesis of monodispersed spherical colloids of low melting-point metals. Nano Lett. 2004;4(10):2047–50.

    Article  CAS  Google Scholar 

  22. Bartlett JA et al. Summary report of PQRI workshop on nanomaterial in drug products: current experience and management of potential risks. AAPS J. 2014;17(1):44–64.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Merisko-Liversidge E, Liversidge GG. Nanosizing for oral and parenteral drug delivery: a perspective on formulating poorly-water soluble compounds using wet media milling technology. Adv Drug Deliv Rev. 2011;63(6):427–40.

    Article  CAS  PubMed  Google Scholar 

  24. Van Eerdenbrugh B et al. Characterization of physico-chemical properties and pharmaceutical performance of sucrose co-freeze–dried solid nanoparticulate powders of the anti-HIV agent loviride prepared by media milling. Int J Pharm. 2007;338(1):198–206.

    Article  PubMed  Google Scholar 

  25. He Q, Shi J. Mesoporous silica nanoparticle based nano drug delivery systems: synthesis, controlled drug release and delivery, pharmacokinetics and biocompatibility. J Mater Chem. 2011;21(16):5845–55.

    Article  CAS  Google Scholar 

  26. Champion JA, Katare YK, Mitragotri S. Particle shape: a new design parameter for micro-and nanoscale drug delivery carriers. J Controlled Release. 2007;121(1):3–9.

    Article  CAS  Google Scholar 

  27. Bemis GW, Murcko MA. The properties of known drugs. 1. Molecular frameworks. J Med Chem. 1996;39(15):2887–93.

    Article  CAS  PubMed  Google Scholar 

  28. Stöber W, Fink A, Bohn E. Controlled growth of monodisperse silica spheres in the micron size range. J Colloid Interface Sci. 1968;26(1):62–9.

    Article  Google Scholar 

  29. Gaumet M et al. Nanoparticles for drug delivery: the need for precision in reporting particle size parameters. Eur J Pharm Biopharm. 2008;69(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  30. Vasconcelos T, Sarmento B, Costa P. Solid dispersions as strategy to improve oral bioavailability of poor water soluble drugs. Drug Discovery Today. 2007;12(23):1068–75.

    Article  CAS  PubMed  Google Scholar 

  31. Whitesides, G.M., J.P. Mathias, and C.T. Seto, Molecular self-assembly and nanochemistry: a chemical strategy for the synthesis of nanostructures, DTIC Document;1991.

  32. Kim H et al. Controlled production of emulsion drops using an electric field in a flow-focusing microfluidic device. Appl Phys Lett. 2007;91(13):133106.

    Article  Google Scholar 

  33. Mozafari, M., Nanoliposomes: preparation and analysis. Liposomes: methods and protocols, Volume 1: Pharmaceutical Nanocarriers, 2010: p. 29–50.

  34. Lu Y, Park K. Polymeric micelles and alternative nanonized delivery vehicles for poorly soluble drugs. Int J Pharm. 2013;453(1):198–214.

    Article  CAS  PubMed  Google Scholar 

  35. Reis CP et al. Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles. Nanomed: Nanotechnol, Biol Med. 2006;2(1):8–21.

    CAS  Google Scholar 

  36. Jones M-C, Leroux J-C. Polymeric micelles—a new generation of colloidal drug carriers. Eur J Pharm Biopharm. 1999;48(2):101–11.

    Article  CAS  PubMed  Google Scholar 

  37. Zanen P et al. The effect of the inhalation flow on the performance of a dry powder inhalation system. Int J Pharm. 1992;81(2):199–203.

    Article  CAS  Google Scholar 

  38. Hickey AJ, Misra A, Fourie PB. Dry powder antibiotic aerosol product development: inhaled therapy for tuberculosis. J Pharm Sci. 2013;102(11):3900–7.

    Article  CAS  PubMed  Google Scholar 

  39. Hickey AJ. Back to the future: inhaled drug products. J Pharm Sci. 2013;102(4):1165–72.

    Article  CAS  PubMed  Google Scholar 

  40. Xu Z, Mansour HM, Hickey AJ. Particle interactions in dry powder inhaler unit processes: a review. J Adhes Sci Technol. 2011;25(4–5):451–82.

    Article  CAS  Google Scholar 

  41. Ostraat ML et al. The Nanomaterial Registry: facilitating the sharing and analysis of data in the diverse nanomaterial community. Int J Nanomed. 2013;8 Suppl 1:7.

    CAS  Google Scholar 

  42. Gaheen S et al. caNanoLab: data sharing to expedite the use of nanotechnology in biomedicine. Comput Sci Dis. 2013;6(1):p. 014010.

    Google Scholar 

  43. Sayes CM et al. Correlating nanoscale titania structure with toxicity: a cytotoxicity and inflammatory response study with human dermal fibroblasts and human lung epithelial cells. Toxicol Sci. 2006;92(1):174–85.

    Article  CAS  PubMed  Google Scholar 

  44. Sayes CM et al. Nano-C 60 cytotoxicity is due to lipid peroxidation. Biomaterials. 2005;26(36):7587–95.

    Article  CAS  PubMed  Google Scholar 

  45. Mills KC et al. Nanomaterial registry: database that captures the minimal information about nanomaterial physico-chemical characteristics. J Nanopart Res. 2014;16(2):1–9.

    Article  Google Scholar 

  46. Guzan K et al. Integration of data: the Nanomaterial Registry project and data curation. Comput Sci Dis. 2013;6(1):014007.

    Article  Google Scholar 

  47. Sayes CM, Reed KL, Warheit DB. Assessing toxicity of fine and nanoparticles: comparing in vitro measurements to in vivo pulmonary toxicity profiles. Toxicol Sci. 2007;97(1):163–80.

    Article  CAS  PubMed  Google Scholar 

  48. Alanazi FK, Radwan AA, Alsarra IA. Biopharmaceutical applications of nanogold. Saudi Pharm J. 2010;18(4):179–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen PC, Mwakwari SC, Oyelere AK. Gold nanoparticles: from nanomedicine to nanosensing. Nanotechnol Sci Appl. 2008;1:45–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hickey AJ et al. Size reduction and classification. Pharmaceutical process engineering. New York: Marcel Dekker; 2001. p. p. 17.

    Google Scholar 

  51. Dobashi, R. Risk of dust explosions of combustible nanomaterials. in Journal of Physics: Conference Series. IOP Publishing;2009.

  52. Berg JM et al. Internalization of carbon black and maghemite iron oxide nanoparticle mixtures leads to oxidant production. Chem Res Toxicol. 2010;23(12):1874–82.

    Article  CAS  PubMed  Google Scholar 

  53. Warheit DB et al. Pulmonary toxicity study in rats with three forms of ultrafine-TiO 2 particles: differential responses related to surface properties. Toxicology. 2007;230(1):90–104.

    Article  CAS  PubMed  Google Scholar 

  54. Wang TC et al. Ultrahigh surface area zirconium MOFs and insights into the applicability of the BET theory. J Am Chem Soc. 2015;137(10):3585–91.

    Article  CAS  PubMed  Google Scholar 

  55. Patil S et al. Protein adsorption and cellular uptake of cerium oxide nanoparticles as a function of zeta potential. Biomaterials. 2007;28(31):4600–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Clogston, J.D. and A.K. Patri, Zeta potential measurement. Characterization of nanoparticles intended for drug delivery, 2011: p. 63–70.

  57. Berg JM et al. The relationship between pH and zeta potential of 30 nm metal oxide nanoparticle suspensions relevant to in vitro toxicological evaluations. Nanotoxicology. 2009;3(4):276–83.

    Article  CAS  Google Scholar 

  58. Riddick, T.M., Control of colloid stability through zeta potential. Blood, 1968. 10(1).

  59. Sung JC, Pulliam BL, Edwards DA. Nanoparticles for drug delivery to the lungs. TRENDS Biotechnol. 2007;25(12):563–70.

    Article  CAS  PubMed  Google Scholar 

  60. Sham JO-H et al. Formulation and characterization of spray-dried powders containing nanoparticles for aerosol delivery to the lung. Int J Pharm. 2004;269(2):457–67.

    Article  CAS  PubMed  Google Scholar 

  61. Pilcer G, Amighi K. Formulation strategy and use of excipients in pulmonary drug delivery. Int J Pharm. 2010;392(1):1–19.

    Article  CAS  PubMed  Google Scholar 

  62. Ankley GT et al. Adverse outcome pathways: a conceptual framework to support ecotoxicology research and risk assessment. Environ Toxicol Chem. 2010;29(3):730–41.

    Article  CAS  PubMed  Google Scholar 

  63. Vinken M et al. Development of an adverse outcome pathway from drug-mediated bile salt export pump inhibition to cholestatic liver injury. Toxicol Sci. 2013;136(1):97–106.

    Article  CAS  PubMed  Google Scholar 

  64. Morita, S., Quantifying Exposure to Engineered Nanomaterials (QEEN) from manufactured products: addressing environmental, health, and safety implications, P. Johnson and G. Holdridge, Editors. Consumer Product Safety Commission (CPSC) and National Nanotechnology Initiative (NNI): Arlington, VA;2016.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Christie M. Sayes.

Additional information

Guest Editors: Katherine Tyner, Sau (Larry) Lee, and Marc Wolfgang

Glossary

Nano-additive (abbrev. NA)

An inactive ingredient or excipient; excipients on the nanoscale that are added to dosage forms; used as a transport module for a drug

Nano-carrier (abbrev. NC)

Non-drug components (additives) prepared as nanoparticles in which the drug is either dispersed (in a single particle) or to which drug is added (particles of drug and carrier); used as a transport module for a drug

Nanocrystal

An example of a nanoparticle and used as an active pharmaceutical ingredient (API) in nanomedicine; a nanoparticle of near-pure drug form; usually composed of atoms, molecules, or other ingredients in a crystalline arrangement

Nano-drug (abbrev. ND)

An example of a therapeutic agent that is the drug alone on the nanoscale; can be a hard solid (e.g., tablets, capsules), soft solid (ointments, suppositories), liquid (solutions, suspensions), or gas (aerosol); a.k.a. nano-pharmaceutical

Nano-enabled

An adjective that describes something as functionally enhanced as compared to its non-nano form; objects may only have one or two dimensions on a nanoscale

Nanomedicine

The medical application of nanoscience; inclusive of drugs (or pharmaceuticals), therapeutic agents, devices, sensors, and machines

Nanoparticle

A particle, either naturally occurring or anthropogenic, that exists with mass and density on the nanoscale; equivalent to nanoparticulate and includes examples such as nanocrystal and nanopowder

Nanoparticle system

Population references for individual nanoparticle groups

Nanopowder

An example of a nanoparticle that exists in the solid state and not suspended in a liquid or gas

Nanoscale

Refers to the size of an agent on or within the nanometer size domain; usually reserved for materials between 1 and 1000 but sometimes defined as 1–100 for purposes outside pharmaceutical applications

Nanoscience

The foundational discipline of nano-related discoveries; the study of materials on the scale of nanometers

Nanotechnology

The application of nanoscience through engineering principles utilizing a material with dimensions of less than 100 nm or with a material engineered to exhibit properties unique from its bulk counterpart up to 1000 nm

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sayes, C.M., Aquino, G.V. & Hickey, A.J. Nanomaterial Drug Products: Manufacturing and Analytical Perspectives. AAPS J 19, 18–25 (2017). https://doi.org/10.1208/s12248-016-0008-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12248-016-0008-x

KEY WORDS

Navigation