Investigating the impact of nanoparticle size on active and passive tumor targeting efficiency.

Understanding the principles governing the design of nanoparticles for tumor targeting is essential for the effective diagnosis and treatment of solid tumors. There is currently a poor understanding of how to rationally engineer nanoparticles for tumor targeting. Here, we engineered different-sized spherical gold nanoparticles to discern the effect of particle diameter on passive (poly(ethylene glycol)-coated) and active (transferrin-coated) targeting of MDA-MB-435 orthotopic tumor xenografts. Tumor accumulation of actively targeted nanoparticles was found to be 5 times faster and approximately 2-fold higher relative to their passive counterparts within the 60 nm diameter range. For 15, 30, and 100 nm, we observed no significant differences. We hypothesize that such enhancements are the result of an increased capacity to penetrate into tumors and preferentially associate with cancer cells. We also use computational modeling to explore the mechanistic parameters that can impact tumor accumulation efficacy. We demonstrate that tumor accumulation can be mediated by high nanoparticle avidity and are weakly dependent on their plasma clearance rate. Such findings suggest that empirical models can be used to rapidly screen novel nanomaterials for relative differences in tumor targeting without the need for animal work. Although our findings are specific to MDA-MB-435 tumor xenografts, our experimental and computational findings help to enrich knowledge of design considerations that will aid in the optimal engineering of spherical gold nanoparticles for cancer applications in the future.

[1]  Jiwon Bang,et al.  Surface engineering of inorganic nanoparticles for imaging and therapy. , 2013, Advanced drug delivery reviews.

[2]  Iseult Lynch,et al.  Physical-chemical aspects of protein corona: relevance to in vitro and in vivo biological impacts of nanoparticles. , 2011, Journal of the American Chemical Society.

[3]  R. Jain,et al.  Delivering nanomedicine to solid tumors , 2010, Nature Reviews Clinical Oncology.

[4]  S M Moghimi,et al.  Long-circulating and target-specific nanoparticles: theory to practice. , 2001, Pharmacological reviews.

[5]  Brahim Lounis,et al.  Cathepsin L digestion of nanobioconjugates upon endocytosis. , 2009, ACS nano.

[6]  M. Amiji,et al.  Biodistribution and pharmacokinetic analysis of long-circulating thiolated gelatin nanoparticles following systemic administration in breast cancer-bearing mice. , 2007, Journal of pharmaceutical sciences.

[7]  Tanya S. Hauck,et al.  Exploring Primary Liver Macrophages for Studying Quantum Dot Interactions with Biological Systems , 2010, Advanced materials.

[8]  Warren C W Chan,et al.  Mediating tumor targeting efficiency of nanoparticles through design. , 2009, Nano letters.

[9]  C. Unger,et al.  In vitro and in vivo Efficacy of Acid-Sensitive Transferrin and Albumin Doxorubicin Conjugates in a Human Xenograft Panel and in the MDA-MB-435 Mamma Carcinoma Model , 2000, Journal of drug targeting.

[10]  Michael M. Schmidt,et al.  A modeling analysis of the effects of molecular size and binding affinity on tumor targeting , 2009, Molecular Cancer Therapeutics.

[11]  Wolfgang A. Weber,et al.  Impact of tumor-specific targeting on the biodistribution and efficacy of siRNA nanoparticles measured by multimodality in vivo imaging , 2007, Proceedings of the National Academy of Sciences.

[12]  S. Nie,et al.  A reexamination of active and passive tumor targeting by using rod-shaped gold nanocrystals and covalently conjugated peptide ligands. , 2010, ACS nano.

[13]  May D. Wang,et al.  In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tags , 2008, Nature Biotechnology.

[14]  Warren C W Chan,et al.  Elucidating the mechanism of cellular uptake and removal of protein-coated gold nanoparticles of different sizes and shapes. , 2007, Nano letters.

[15]  Mark E. Davis,et al.  Targeting kidney mesangium by nanoparticles of defined size , 2011, Proceedings of the National Academy of Sciences.

[16]  M. Uesaka,et al.  Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. , 2011, Nature nanotechnology.

[17]  M. Dobrovolskaia,et al.  Immunological properties of engineered nanomaterials , 2007, Nature Nanotechnology.

[18]  M. Kurfürst Detection and molecular weight determination of polyethylene glycol-modified hirudin by staining after sodium dodecyl sulfate-polyacrylamide gel electrophoresis. , 1992, Analytical biochemistry.

[19]  F. Kiessling,et al.  Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress. , 2012, Journal of controlled release : official journal of the Controlled Release Society.

[20]  Warren C W Chan,et al.  Fluorescence‐Tagged Gold Nanoparticles for Rapidly Characterizing the Size‐Dependent Biodistribution in Tumor Models , 2012, Advanced healthcare materials.

[21]  D. Wei,et al.  Discarded free PEG-based assay for obtaining the modification extent of pegylated proteins. , 2007, Talanta.

[22]  Mark E. Davis,et al.  Impact of tumor‐specific targeting and dosing schedule on tumor growth inhibition after intravenous administration of siRNA‐containing nanoparticles , 2008, Biotechnology and bioengineering.

[23]  Michael S. Strano,et al.  Size-dependent cellular uptake and expulsion of single-walled carbon nanotubes: single particle tracking and a generic uptake model for nanoparticles. , 2009, ACS nano.

[24]  Stephen J. Lomnes,et al.  Tissue-like phantoms for near-infrared fluorescence imaging system assessment and the training of surgeons. , 2006, Journal of biomedical optics.

[25]  Mark E. Davis,et al.  Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles , 2009, Proceedings of the National Academy of Sciences.

[26]  Huajian Gao,et al.  Mechanics of receptor-mediated endocytosis. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[27]  G. Adams,et al.  High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. , 2001, Cancer research.

[28]  R K Jain,et al.  Transport of fluid and macromolecules in tumors. III. Role of binding and metabolism. , 1991 .

[29]  Warren C W Chan,et al.  The effect of nanoparticle size, shape, and surface chemistry on biological systems. , 2012, Annual review of biomedical engineering.

[30]  R K Jain,et al.  Transport of fluid and macromolecules in tumors. I. Role of interstitial pressure and convection. , 1989, Microvascular research.

[31]  W. Chan,et al.  Synthesis and surface modification of highly monodispersed, spherical gold nanoparticles of 50-200 nm. , 2009, Journal of the American Chemical Society.

[32]  U. Nielsen,et al.  Antibody targeting of long-circulating lipidic nanoparticles does not increase tumor localization but does increase internalization in animal models. , 2006, Cancer research.

[33]  Manuela Semmler-Behnke,et al.  Particle size-dependent and surface charge-dependent biodistribution of gold nanoparticles after intravenous administration. , 2011, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V.

[34]  Warren C W Chan,et al.  Nanoparticle-mediated cellular response is size-dependent. , 2008, Nature nanotechnology.

[35]  G. Ulrich Nienhaus,et al.  Impact of protein modification on the protein corona on nanoparticles and nanoparticle-cell interactions. , 2014, ACS nano.

[36]  Kenneth A. Dawson,et al.  Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts , 2008, Proceedings of the National Academy of Sciences.

[37]  Andrew Emili,et al.  Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake. , 2012, Journal of the American Chemical Society.

[38]  Keishiro Tomoda,et al.  Biodistribution of colloidal gold nanoparticles after intravenous administration: effect of particle size. , 2008, Colloids and surfaces. B, Biointerfaces.

[39]  B. Murray,et al.  The effect of geometry on capillary wall dose for boron neutron capture therapy. , 1976, Physics in medicine and biology.

[40]  Susan Newbigging,et al.  In vivo quantum-dot toxicity assessment. , 2010, Small.

[41]  Philip M. Kelly,et al.  Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. , 2013, Nature nanotechnology.

[42]  Subra Suresh,et al.  Size‐Dependent Endocytosis of Nanoparticles , 2009, Advanced materials.

[43]  Dai Fukumura,et al.  Multistage nanoparticle delivery system for deep penetration into tumor tissue , 2011, Proceedings of the National Academy of Sciences.

[44]  Warren C. W. Chan,et al.  Polyethylene glycol backfilling mitigates the negative impact of the protein corona on nanoparticle cell targeting. , 2014, Angewandte Chemie.

[45]  S. Krantz,et al.  Transferrin receptor number, synthesis, and endocytosis during erythropoietin-induced maturation of Friend virus-infected erythroid cells. , 1986, The Journal of biological chemistry.