Superior penetration and retention behavior of 50 nm gold nanoparticles in tumors.

Nanoparticles offer potential as drug delivery systems for chemotherapeutics based on certain advantages of molecular drugs. In this study, we report that particle size exerts great influence on the penetration and retention behavior of nanoparticles entering tumors. On comparing gold-coated Au@tiopronin nanoparticles that were prepared with identical coating and surface properties, we found that 50 nanoparticles were more effective in all in vitro, ex vivo, and in vivo assays conducted using MCF-7 breast cells as a model system. Beyond superior penetration in cultured cell monolayers, 50 nm Au@tiopronin nanoparticles also penetrated more deeply into tumor spheroids ex vivo and accumulated more effectively in tumor xenografts in vivo after a single intravenous dose. In contrast, larger gold-coated nanoparticles were primarily localized in the periphery of the tumor spheroid and around blood vessels, hindering deep penetration into tumors. We found multicellular spheroids to offer a simple ex vivo tumor model to simulate tumor tissue for screening the nanoparticle penetration behavior. Taken together, our findings define an optimal smaller size for nanoparticles that maximizes their effective accumulation in tumor tissue.

[1]  C. M. Roth,et al.  Binding and transport of PAMAM‐RGD in a tumor spheroid model: The effect of RGD targeting ligand density , 2011, Biotechnology and bioengineering.

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

[3]  D. Fernig,et al.  Determination of size and concentration of gold nanoparticles from UV-vis spectra. , 2007, Analytical chemistry.

[4]  J. Brown,et al.  Exploiting tumour hypoxia in cancer treatment , 2004, Nature Reviews Cancer.

[5]  James E Hutchison,et al.  Toward greener nanosynthesis. , 2007, Chemical reviews.

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

[7]  Kenneth A. Dawson,et al.  Role of cell cycle on the cellular uptake and dilution of nanoparticles in a cell population. , 2011, Nature nanotechnology.

[8]  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.

[9]  Arezou A Ghazani,et al.  Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. , 2006, Nano letters.

[10]  S. Pun,et al.  Increased nanoparticle penetration in collagenase-treated multicellular spheroids , 2007, International journal of nanomedicine.

[11]  C. Allen,et al.  Block copolymer micelles for delivery of cancer therapy: transport at the whole body, tissue and cellular levels. , 2009, Journal of controlled release : official journal of the Controlled Release Society.

[12]  Chung-Yuan Mou,et al.  Size effect on cell uptake in well-suspended, uniform mesoporous silica nanoparticles. , 2009, Small.

[13]  Catherine J. Murphy,et al.  Evidence for Seed-Mediated Nucleation in the Chemical Reduction of Gold Salts to Gold Nanoparticles , 2001 .

[14]  G. Fracasso,et al.  Effect of therapeutic macromolecules in spheroids. , 2000, Critical reviews in oncology/hematology.

[15]  Shinsuke Sando,et al.  A quantum dot conjugated sugar ball and its cellular uptake. On the size effects of endocytosis in the subviral region. , 2004, Journal of the American Chemical Society.

[16]  Ou Chen,et al.  Fluorescent nanorods and nanospheres for real-time in vivo probing of nanoparticle shape-dependent tumor penetration. , 2011, Angewandte Chemie.

[17]  Shufang Zhang,et al.  Engineering a scaffold-free 3D tumor model for in vitro drug penetration studies. , 2010, Biomaterials.

[18]  Weipeng Cao,et al.  Gold nanoparticles functionalized with therapeutic and targeted peptides for cancer treatment. , 2012, Biomaterials.

[19]  A. Giaccia,et al.  The unique physiology of solid tumors: opportunities (and problems) for cancer therapy. , 1998, Cancer research.

[20]  C. Ries,et al.  Comparison of 3D and 2D tumor models reveals enhanced HER2 activation in 3D associated with an increased response to trastuzumab , 2009, Oncogene.

[21]  I. Tannock,et al.  Drug penetration in solid tumours , 2006, Nature Reviews Cancer.

[22]  Malcolm E. Kenney,et al.  Deep penetration of a PDT drug into tumors by noncovalent drug-gold nanoparticle conjugates. , 2011, Journal of the American Chemical Society.

[23]  Vladimir P Torchilin,et al.  Cationic charge determines the distribution of liposomes between the vascular and extravascular compartments of tumors. , 2002, Cancer research.

[24]  M. Gottesman,et al.  Collateral sensitivity of multidrug-resistant cells to the orphan drug tiopronin. , 2011, Journal of medicinal chemistry.

[25]  Juergen Friedrich,et al.  Spheroid-based drug screen: considerations and practical approach , 2009, Nature Protocols.

[26]  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.

[27]  Jessie L.-S. Au,et al.  Drug Delivery and Transport to Solid Tumors , 2003, Pharmaceutical Research.

[28]  H. Maeda,et al.  Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. , 2000, Journal of controlled release : official journal of the Controlled Release Society.

[29]  Rakesh K Jain,et al.  Molecular regulation of vessel maturation , 2003, Nature Medicine.

[30]  Charles R. Martin Nanomedicine: a great first year and, with your help, a bright future ahead , 2007 .

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

[32]  H. Maeda,et al.  A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. , 1986, Cancer research.

[33]  C. M. Roth,et al.  PAMAM-RGD conjugates enhance siRNA delivery through a multicellular spheroid model of malignant glioma. , 2009, Bioconjugate chemistry.

[34]  Hwan-You Chang,et al.  Recent advances in three‐dimensional multicellular spheroid culture for biomedical research , 2008, Biotechnology journal.

[35]  Rakesh K. Jain,et al.  Pathology: Cancer cells compress intratumour vessels , 2004, Nature.

[36]  Vincent M. Rotello,et al.  Tuning Payload Delivery in Tumour Cylindroids using Gold Nanoparticles , 2010, Nature nanotechnology.

[37]  G. Frens Controlled Nucleation for the Regulation of the Particle Size in Monodisperse Gold Suspensions , 1973 .

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

[39]  R. Langer,et al.  Photoswitchable Nanoparticles for Triggered Tissue Penetration and Drug Delivery , 2012, Journal of the American Chemical Society.

[40]  Yang Ren,et al.  Structure of gold nanoparticles suspended in water studied by x-ray diffraction and computer simulations , 2005 .