Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Analysis of nanoparticle delivery to tumours

Abstract

Targeting nanoparticles to malignant tissues for improved diagnosis and therapy is a popular concept. However, after surveying the literature from the past 10 years, only 0.7% (median) of the administered nanoparticle dose is found to be delivered to a solid tumour. This has negative consequences on the translation of nanotechnology for human use with respect to manufacturing, cost, toxicity, and imaging and therapeutic efficacy. In this article, we conduct a multivariate analysis on the compiled data to reveal the contributions of nanoparticle physicochemical parameters, tumour models and cancer types on the low delivery efficiency. We explore the potential causes of the poor delivery efficiency from the perspectives of tumour biology (intercellular versus transcellular transport, enhanced permeability and retention effect, and physicochemical-dependent nanoparticle transport through the tumour stroma) as well as competing organs (mononuclear phagocytic and renal systems) and present a 30-year research strategy to overcome this fundamental limitation. Solving the nanoparticle delivery problem will accelerate the clinical translation of nanomedicine.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Analysis of nanoparticle delivery efficiency to solid tumours from studies published in 2005–2015.
Figure 2: Estimation of nanoparticle dose for human tumour treatment.
Figure 3: Tumour blood vessels and mode for nanoparticle extravasation in mother vessels versus capillaries.
Figure 4: Mechanisms for nanoparticle elimination from the bloodstream.
Figure 5: Proposed 30-year strategy for nanoparticle delivery research.

Similar content being viewed by others

References

  1. Peer, D. et al. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2, 751–760 (2007).

    CAS  Google Scholar 

  2. Rao, W. et al. Chitosan-decorated doxorubicin-encapsulated nanoparticle targets and eliminates tumor reinitiating cancer stem-like cells. ACS Nano 9, 5725–5740 (2015).

    CAS  Google Scholar 

  3. Min, Y., Caster, J. M., Eblan, M. J. & Wang, A. Z. Clinical translation of nanomedicine. Chem. Rev. 115, 11147–11190 (2015).

    CAS  Google Scholar 

  4. Nel, A. E. et al. Understanding biophysicochemical interactions at the nano–bio interface. Nat. Mater. 8, 543–557 (2009).

    CAS  Google Scholar 

  5. Albanese, A., Tang, P. S. & Chan, W. C. W. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu. Rev. Biomed. Eng. 14, 1–16 (2012).

    CAS  Google Scholar 

  6. Endres, T. et al. Optimising the self-assembly of siRNA loaded PEG–PCL–lPEI nano-carriers employing different preparation techniques. J. Control. Release 160, 583–591 (2012).

    CAS  Google Scholar 

  7. Huang, X., El-Sayed, I. H., Qian, W. & El-Sayed, M. A. Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J. Am. Chem. Soc. 128, 2115–2120 (2006).

    CAS  Google Scholar 

  8. Wolfbeis, O. S. An overview of nanoparticles commonly used in fluorescent bioimaging. Chem. Soc. Rev. 44, 4743–4768 (2015).

    CAS  Google Scholar 

  9. Attia, M. F. et al. Biodistribution of X-ray iodinated contrast agent in nano-emulsions is controlled by the chemical nature of the oily core. ACS Nano 8, 10537–10550 (2014).

    CAS  Google Scholar 

  10. Kircher, M. F. et al. A brain tumor molecular imaging strategy using a new triple-modality MRI-photoacoustic-Raman nanoparticle. Nat. Med. 18, 829–834 (2012).

    CAS  Google Scholar 

  11. Guo, X. Shi, C., Wang, J., Di, S. & Zhou, S. pH-triggered intracellular release from actively targeting polymer micelles. Biomaterials 34, 4544–4554 (2013).

    CAS  Google Scholar 

  12. Gao, W., Chan, J. M. & Farokhzad, O. C. pH-responsive nanoparticles for drug delivery. Mol. Pharmaceut. 7, 1913–1920 (2010).

    CAS  Google Scholar 

  13. Cheng, R., Meng, F., Deng, C., Klok, H.-A. & Zhong, Z. Dual and multi-stimuli responsive polymeric nanoparticles for programmed site-specific drug delivery. Biomaterials 34, 3647–3657 (2013).

    CAS  Google Scholar 

  14. Hu, Q., Katti, P. S. & Gu, Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale 6, 12273–12286 (2014).

    CAS  Google Scholar 

  15. de la Rica, R., Aili, D. & Stevens, M. M. Enzyme-responsive nanoparticles for drug release and diagnostics. Adv. Drug Deliv. Rev. 64, 967–978 (2012).

    CAS  Google Scholar 

  16. Chen, F., Ehlerding, E. B. & Cai, W. Theranostic nanoparticles. J. Nucl. Med. 55, 1919–1922 (2014).

    CAS  Google Scholar 

  17. Chen, Q. et al. Drug-induced self-assembly of modified albumins as nano-theranostics for tumor-targeted combination therapy. ACS Nano 9, 5223–5233 (2015).

    CAS  Google Scholar 

  18. Chou, L. Y. T., Zagorovsky, K. & Chan, W. C. W. DNA assembly of nanoparticle superstructures for controlled biological delivery and elimination. Nat. Nanotechnol. 9, 148–155 (2014).

    CAS  Google Scholar 

  19. Bae, Y. H. & Park, K. Targeted drug delivery to tumors: myths, reality and possibility. J. Control. Release 153, 198–205 (2011).

    CAS  Google Scholar 

  20. Wang, A. Z., Langer, R. & Farokhzad, O. C. Nanoparticle delivery of cancer drugs. Annu. Rev. Med. 63, 185–198 (2012).

    CAS  Google Scholar 

  21. Park, K. Facing the truth about nanotechnology in drug delivery. ACS Nano 7, 7442–7447(2013).

    CAS  Google Scholar 

  22. Lazarovits, J., Chen, Y. Y., Sykes, E. A. & Chan, W. C. W. Nanoparticle–blood interactions: the implications on solid tumour targeting. Chem. Commun. 51, 2756–2767 (2015).

    CAS  Google Scholar 

  23. Nichols, J. W. & Bae, Y. H. Odyssey of a cancer nanoparticle: from injection site to site of action. Nano Today 7, 606–618 (2012).

    CAS  Google Scholar 

  24. Jain, R. K. & Stylianopoulos, T. Delivering nanomedicine to solid tumors. Nat. Rev. Clin. Oncol. 7, 653–664 (2010).

    CAS  Google Scholar 

  25. Florence, A. T. “Targeting” nanoparticles: the constraints of physical laws and physical barriers. J. Control. Release 164, 115–124 (2012).

    CAS  Google Scholar 

  26. Choi, H. S. et al. Renal clearance of quantum dots. Nat. Biotechnol. 25, 1165–1170 (2007).

    CAS  Google Scholar 

  27. Liu, J. et al. PEGylation and zwitterionization: pros and cons in the renal clearance and tumor targeting of near-IR-emitting gold nanoparticles. Angew. Chem. Int. Ed. Engl. 52, 12572–12576 (2013).

    CAS  Google Scholar 

  28. Liu, J. et al. Passive tumor targeting of renal-clearable luminescent gold nanoparticles: long tumor retention and fast normal tissue clearance. J. Am. Chem. Soc. 135, 4978–4981 (2013).

    CAS  Google Scholar 

  29. Yu, M. & Zheng, J. Clearance pathways and tumor targeting of imaging nanoparticles. ACS Nano 9, 6655–6674 (2015).

    CAS  Google Scholar 

  30. Dawidczyk, C. M. et al. State-of-the-art in design rules for drug delivery platforms: lessons learned from FDA-approved nanomedicines. J. Control. Release 187, 133–144 (2014).

    CAS  Google Scholar 

  31. Chiou, W. L. Critical evaluation of the potential error in pharmacokinetic studies of using the linear trapezoidal rule method for the calculation of the area under the plasma level-time curve. J. Pharmacokinet. Biopharm. 6, 539–546 (1978).

    CAS  Google Scholar 

  32. Sykes, E. A., Chen, J., Zheng, G. & Chan, W. C. W. Investigating the impact of nanoparticle size on active and passive tumor targeting efficiency. ACS Nano 8, 5696–5706 (2014).

    CAS  Google Scholar 

  33. Tsai, C.-C. et al. Biodistribution and pharmacokinetics of 188Re-liposomes and their comparative therapeutic efficacy with 5-fluorouracil in C26 colonic peritoneal carcinomatosis mice. Int. J. Nanomed. 6, 2607–2619 (2011).

    CAS  Google Scholar 

  34. Kukowska-Latallo, J. F. et al. Nanoparticle targeting of anticancer drug improves therapeutic response in animal model of human epithelial cancer. Cancer Res. 65, 5317–5324 (2005).

    CAS  Google Scholar 

  35. Sadekar, S., Ray, A., Janà t-Amsbury, M., Peterson, C. M. & Ghandehari, H. Comparative biodistribution of PAMAM dendrimers and HPMA copolymers in ovarian-tumor-bearing mice. Biomacromolecules 12, 88–96 (2011).

    CAS  Google Scholar 

  36. Reagan-Shaw, S., Nihal, M. & Ahmad, N. Dose translation from animal to human studies revisited. FASEB J. 22, 659–661 (2008).

    CAS  Google Scholar 

  37. Maeda, H., Nakamura, H. & Fang, J. The EPR effect for macromolecular drug delivery to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv. Drug Deliv. Rev. 65, 71–79 (2013).

    CAS  Google Scholar 

  38. Ruoslahti, E., Bhatia, S. N. & Sailor, M. J. Targeting of drugs and nanoparticles to tumors. J. Cell Biol. 188, 759–768 (2010).

    CAS  Google Scholar 

  39. Dvorak, H. F., Nagy, J. A., Dvorak, J. T. & Dvorak, A. M. Identification and characterization of the blood vessels of solid tumors that are leaky to circulating macromolecules. Am. J. Pathol. 133, 95–109 (1988).

    CAS  Google Scholar 

  40. Warren, B. A. in Tumor Blood Circulation: Angiogenesis, Vascular Morphology and Blood Flow of Experimental and Human Tumors (ed. Peterson, H.-I. ) 1–47 (CRC Press, 1979).

    Google Scholar 

  41. Nagy, J. A. et al. Permeability properties of tumor surrogate blood vessels induced by VEGF-A. Lab. Invest. 86, 767–780 (2006).

    CAS  Google Scholar 

  42. Dvorak, H. F. Rous-Whipple Award Lecture. How tumors make bad blood vessels and stroma. Am. J. Pathol. 162, 1747–1757 (2003).

    CAS  Google Scholar 

  43. Dvorak, H. F. in The Endothelium: A Comprehensive Reference (ed. Aird, W. ) 1457–1470 (Cambridge Univ. Press, 2007).

    Google Scholar 

  44. Zeng, H. et al. Orphan nuclear receptor TR3/Nur77 regulates VEGF-A-induced angiogenesis through its transcriptional activity. J. Exp. Med. 203, 719–729 (2006).

    CAS  Google Scholar 

  45. Paku, S. & Paweletz, N. First steps of tumor-related angiogenesis. Lab. Invest. 65, 334–346 (1991).

    CAS  Google Scholar 

  46. Nagy, J. A., Benjamin, L., Zeng, H., Dvorak, A. M. & Dvorak, H. F. Vascular permeability, vascular hyperpermeability and angiogenesis. Angiogenesis 11, 109–119 (2008).

    CAS  Google Scholar 

  47. Chang, S. H. et al. VEGF-A induces angiogenesis by perturbing the cathepsin–cysteine protease inhibitor balance in venules, causing basement membrane degradation and mother vessel formation. Cancer Res. 69, 4537–4544 (2009).

    CAS  Google Scholar 

  48. Nagy, J. A., Chang, S. H., Shih, S. C., Dvorak, A. M. & Dvorak, H. F. Heterogeneity of the tumor vasculature. Semin. Thromb. Hemostasis 36, 321–331 (2010).

    CAS  Google Scholar 

  49. Pettersson, A. et al. Heterogeneity of the angiogenic response induced in different normal adult tissues by vascular permeability factor/vascular endothelial growth factor. Lab. Invest. 80, 99–115 (2000).

    CAS  Google Scholar 

  50. Fidler, I. J., Yano, S., Zhang, R. D., Fujimaki, T. & Bucana, C. D. The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol. 3, 53–57 (2002).

    CAS  Google Scholar 

  51. Sundberg, C. et al. Glomeruloid microvascular proliferation follows adenoviral vascular permeability factor/vascular endothelial growth factor-164 gene delivery. Am. J. Pathol. 158, 1145–1160 (2001).

    CAS  Google Scholar 

  52. Nagy, J. A., Shih, S. C., Wong, W. H., Dvorak, A. M. & Dvorak, H. F. Chapter 3. The adenoviral vector angiogenesis/lymphangiogenesis assay. Methods Enzymol. 444, 43–64 (2008).

    CAS  Google Scholar 

  53. Nagy, J. A., Dvorak, A. M. & Dvorak, H. F. Vascular hyperpermeability, angiogenesis, and stroma generation. Cold Spring Harbor Perspect. Med. 2, a006544 (2012).

    Google Scholar 

  54. Kobayashi, H., Watanabe, R. & Choyke, P. L. Improving conventional enhanced permeability and retention (EPR) effects; what is the appropriate target? Theranostics 4, 81–89 (2013).

    Google Scholar 

  55. Prabhakar, U. et al. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 73, 2412–2417 (2013).

    CAS  Google Scholar 

  56. Matsumura, Y. & Maeda, H. A. New concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 46, 6387–6392 (1986).

    CAS  Google Scholar 

  57. Dvorak, H. F. Tumors: wounds that do not heal–redux. Cancer Immunol. Res. 3, 1–11 (2015).

    CAS  Google Scholar 

  58. Hobbs, S. et al. Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc. Natl Acad. Sci. USA 95, 4607–4612 (1998).

    CAS  Google Scholar 

  59. Dvorak, A. M. et al. The vesiculo-vacuolar organelle (VVO): a distinct endothelial cell structure that provides a transcellular pathway for macromolecular extravasation. J. Leukocyte Biol. 59, 100–115 (1996).

    CAS  Google Scholar 

  60. Feng, D., Nagy, J. A., Hipp, J., Dvorak, H. F. & Dvorak, A. M. Vesiculo-vacuolar organelles and the regulation of venule permeability to macromolecules by vascular permeability factor, histamine, and serotonin. J. Exp. Med. 183, 1981–1986 (1996).

    CAS  Google Scholar 

  61. Pickup, M. W., Mouw, J. K. & Weaver, V. M. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 15, 1243–1253 (2014).

    CAS  Google Scholar 

  62. Box, C., Rogers, S. J., Mendiola, M. & Eccles, S. A. Tumour-microenvironmental interactions: paths to progression and targets for treatment. Semin. Cancer Biol. 20, 128–138 (2010).

    CAS  Google Scholar 

  63. Eccles, S. A. & Alexander, P. Macrophage content of tumours in relation to metastatic spread and host immune reaction. Nature 250, 667–669 (1974).

    CAS  Google Scholar 

  64. Heldin, C.-H., Rubin, K., Pietras, K. & Ostman, A. High interstitial fluid pressure — an obstacle in cancer therapy. Nat. Rev. Cancer 4, 806–813 (2004).

    CAS  Google Scholar 

  65. Swartz, M. A. & Lund, A. W. Lymphatic and interstitial flow in the tumour microenvironment: linking mechanobiology with immunity. Nat. Rev. Cancer 12, 210–219 (2012).

    CAS  Google Scholar 

  66. Perrault, S. D., Walkey, C., Jennings, T., Fischer, H. C. & Chan, W. C. W. Mediating tumor targeting efficiency of nanoparticles through design. Nano Lett. 9, 1909–1915 (2009).

    CAS  Google Scholar 

  67. Chauhan, V. P. et al. Fluorescent nanorods and nanospheres for real-time in vivo probing of nanoparticle shape-dependent tumor penetration. Angew. Chem. Int. Ed. Engl. 50, 11417–11420 (2011).

    CAS  Google Scholar 

  68. Yuan, F. et al. Vascular permeability and microcirculation of gliomas and mammary carcinomas transplanted in rat and mouse cranial windows. Cancer Res. 54, 4564–4568 (1994).

    CAS  Google Scholar 

  69. Albanese, A., Lam, A. K., Sykes, E. A., Rocheleau, J. V. & Chan, W. C. W. Tumour-on-a-chip provides an optical window into nanoparticle tissue transport. Nat. Commun. 4, 2718 (2013).

    Google Scholar 

  70. Huang, X. et al. A reexamination of active and passive tumor targeting by using rod-shaped gold nanocrystals and covalently conjugated peptide ligands. ACS Nano 4, 5887–5896 (2010).

    CAS  Google Scholar 

  71. Kunjachan, S. et al. Passive versus active tumor targeting using RGD- and NGR-modified polymeric nanomedicines. Nano Lett. 14, 972–981 (2014).

    CAS  Google Scholar 

  72. Choi, C. H. J., Alabi, C. A., Webster, P. & Davis, M. E. Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles. Proc. Natl Acad. Sci. USA 107, 1235–1240 (2010).

    CAS  Google Scholar 

  73. Gordon, S. & Taylor, P. R. Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 5, 953–964 (2005).

    CAS  Google Scholar 

  74. Fischer, H. C., Hauck, T. S., Gómez-Aristizá bal, A. & Chan, W. C. W. Exploring primary liver macrophages for studying quantum dot interactions with biological systems. Adv. Mater. 22, 2520–2524 (2010).

    CAS  Google Scholar 

  75. Huang, S. et al. In vivo splenic clearance correlates with in vitro deformability of red blood cells from Plasmodium yoelii-infected mice. Infect. Immun. 82, 2532–2541 (2014).

    Google Scholar 

  76. Davies, L. C., Jenkins, S. J., Allen, J. E. & Taylor, P. R. Tissue-resident macrophages. Nat. Immunol. 14, 986–995 (2013).

    CAS  Google Scholar 

  77. Syed, A. & Chan, W. C. W. How nanoparticles interact with cancer cells. Cancer Treat. Res. 166, 227–244 (2015).

    CAS  Google Scholar 

  78. Patel, P. C. et al. Scavenger receptors mediate cellular uptake of polyvalent oligonucleotide-functionalized gold nanoparticles. Bioconjugate Chem. 21, 2250–2256 (2010).

    CAS  Google Scholar 

  79. Wang, H., Wu, L. & Reinhard, B. M. Scavenger receptor mediated endocytosis of silver nanoparticles into J774A.1 macrophages is heterogeneous. ACS Nano 6, 7122–7132 (2012).

    CAS  Google Scholar 

  80. Cedervall, T. et al. Understanding the nanoparticle–protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles. Proc. Natl Acad. Sci. USA 104, 2050–2055 (2007).

    CAS  Google Scholar 

  81. Walkey, C. D. & Chan, W. C. W. Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. Chem. Soc. Rev. 41, 2780–2799 (2012).

    CAS  Google Scholar 

  82. Albanese, A. et al. Secreted biomolecules alter the biological identity and cellular interactions of nanoparticles. ACS Nano 8, 5515–5526 (2014).

    CAS  Google Scholar 

  83. Walkey, C. D., Olsen, J. B., Guo, H., Emili, A. & Chan, W. C. W. Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake. J. Am. Chem. Soc. 134, 2139–2147 (2012).

    CAS  Google Scholar 

  84. Walkey, C. D. et al. Protein corona fingerprinting predicts the cellular interaction of gold and silver nanoparticles. ACS Nano 8, 2439–2455 (2014).

    CAS  Google Scholar 

  85. Jong, W. H. de et al. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials 29, 1912–1919 (2008).

    Google Scholar 

  86. Deen, W. M., Lazzara, M. J. & Myers, B. D. Structural determinants of glomerular permeability. Am. J. Physiol. Renal Physiol. 281, F579–F596 (2001).

    CAS  Google Scholar 

  87. Venkatachalam, M. A. & Rennke, H. G. The structural and molecular basis of glomerular filtration. Circ. Res. 43, 337–347 (1978).

    CAS  Google Scholar 

  88. Nair, A. V., Keliher, E. J., Core, A. B., Brown, D. & Weissleder, R. Characterizing the interactions of organic nanoparticles with renal epithelial cells in vivo. ACS Nano 9, 3641–3653 (2015).

    CAS  Google Scholar 

  89. Pillai, G. Nanomedicines for cancer therapy: an update of FDA approved and those under various stages of development. SOJ Pharm. Pharm. Sci. 1, 1–13 (2014).

    Google Scholar 

  90. Venditto, V. J. & Szoka, F. C. Cancer nanomedicines: so many papers and so few drugs! Adv. Drug Deliv. Rev. 65, 80–88 (2013).

    CAS  Google Scholar 

  91. Allen, T. M. & Cullis, P. R. Liposomal drug delivery systems: from concept to clinical applications. Adv. Drug Deliv. Rev. 65, 36–48 (2013).

    CAS  Google Scholar 

  92. Barenholz, Y. Doxil® — the first FDA-approved nano-drug: lessons learned. J. Control. Release 160, 117–134 (2012).

    CAS  Google Scholar 

  93. Leonard, R. C. F., Williams, S., Tulpule, A., Levine, A. M. & Oliveros, S. Improving the therapeutic index of anthracycline chemotherapy: focus on liposomal doxorubicin (Myocet). Breast 18, 218–224 (2009).

    CAS  Google Scholar 

  94. Chauhan, V. P. et al. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat. Nanotechnol. 7, 383–388 (2012).

    CAS  Google Scholar 

  95. Jiang, W., Huang, Y., An, Y. & Kim, B. Y. S. Remodeling tumor vasculature to enhance eelivery of intermediate-sized nanoparticles. ACS Nano 9, 8689–8696 (2015).

    CAS  Google Scholar 

  96. Tailor, T. D. et al. Effect of pazopanib on tumor microenvironment and liposome delivery. Mol. Cancer Ther. 9, 1798–1808 (2010).

    CAS  Google Scholar 

  97. Pastuskovas, C. V. et al. Effects of anti-VEGF on pharmacokinetics, biodistribution, and tumor penetration of trastuzumab in a preclinical breast cancer model. Mol. Cancer Ther. 11, 752–762 (2012).

    CAS  Google Scholar 

  98. Dobosz, M., Ntziachristos, V., Scheuer, W. & Strobel, S. Multispectral fluorescence ultramicroscopy: three-dimensional visualization and automatic quantification of tumor morphology, drug penetration, and antiangiogenic treatment response. Neoplasia 16, 1–13 (2014).

    Google Scholar 

  99. Roger, M. et al. Mesenchymal stem cells as cellular vehicles for delivery of nanoparticles to brain tumors. Biomaterials 31, 8393–8401 (2010).

    CAS  Google Scholar 

  100. Li, L. et al. Silica nanorattle–doxorubicin-anchored mesenchymal stem cells for tumor-tropic therapy. ACS Nano 5, 7462–7470 (2011).

    CAS  Google Scholar 

  101. Cheng, H. et al. Nanoparticulate cellular patches for cell-mediated tumoritropic delivery. ACS Nano 4, 625–631 (2010).

    CAS  Google Scholar 

  102. Hu, Q. et al. Engineering nanoparticle-coated bacteria as oral DNA vaccines for cancer immunotherapy. Nano Lett. 15, 2732–2739 (2015).

    CAS  Google Scholar 

  103. MacDiarmid, J. A. et al. Bacterially derived 400 nm particles for encapsulation and cancer cell targeting of chemotherapeutics. Cancer Cell 11, 431–445 (2007).

    CAS  Google Scholar 

  104. Park, S. J. et al. New paradigm for tumor theranostic methodology using bacteria-based microrobot. Sci. Rep. 3, 3394 (2013).

    Google Scholar 

  105. Doshi, N. et al. Cell-based drug delivery devices using phagocytosis-resistant backpacks. Adv. Mater. 23, H105–H109 (2011).

    CAS  Google Scholar 

  106. Akin, D. et al. Bacteria-mediated delivery of nanoparticles and cargo into cells. Nat. Nanotechnol 2, 441–449 (2007).

    CAS  Google Scholar 

  107. Kuhn, S. J., Finch, S. K., Hallahan, D. E. & Giorgio, T. D. Proteolytic surface functionalization enhances in vitro magnetic nanoparticle mobility through extracellular matrix. Nano Lett. 6, 306–312 (2006).

    CAS  Google Scholar 

  108. Cui, M. et al. Multifunctional albumin nanoparticles as combination drug carriers for intra-tumoral chemotherapy. Adv. Healthcare Mater. 2, 1236–1245 (2013).

    CAS  Google Scholar 

  109. Gormley, A. J. et al. Plasmonic photothermal therapy increases the tumor mass penetration of HPMA copolymers. J. Control Release 166, 130–138 (2013).

    CAS  Google Scholar 

  110. Diagaradjane, P. et al. Modulation of in vivo tumor radiation response via gold nanoshell-mediated vascular-focused hyperthermia: characterizing an integrated antihypoxic and localized vascular disrupting targeting strategy. Nano Lett. 8, 1492–1500 (2008).

    CAS  Google Scholar 

  111. Ohara, Y. et al. Effective delivery of chemotherapeutic nanoparticles by depleting host Kupffer cells. Int. J. Cancer 131, 2402–2410 (2012).

    CAS  Google Scholar 

  112. van Rooijen, N. & Sanders, A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J. Immunol. Methods 174, 83–93 (1994).

    CAS  Google Scholar 

  113. Diagaradjane, P., Deorukhkar, A., Gelovani, J. G., Maru, D. M. & Krishnan, S. Gadolinium chloride augments tumor-specific imaging of targeted quantum dots in vivo. ACS Nano 4, 4131–4141 (2010).

    CAS  Google Scholar 

  114. Parodi, A. et al. Synthetic nanoparticles functionalized with biomimetic leukocyte membranes possess cell-like functions. Nat. Nanotechnol. 8, 61–68 (2013).

    CAS  Google Scholar 

  115. Piao, J.-G. et al. Erythrocyte membrane is an alternative coating to polyethylene glycol for prolonging the circulation lifetime of gold nanocages for photothermal therapy. ACS Nano 8, 10414–10425 (2014).

    CAS  Google Scholar 

  116. Rodriguez, P. L. et al. Minimal “self” peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science 339, 971–975 (2013).

    CAS  Google Scholar 

  117. Barua, S. & Mitragotri, S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: a review of current status and future prospects. Nano Today 9, 223–243 (2014).

    CAS  Google Scholar 

  118. Pascal, J. et al. Mechanistic patient-specific predictive correlation of tumor drug response with microenvironment and perfusion measurements. Proc. Natl Acad. Sci. USA 110, 14266–14271 (2013).

    CAS  Google Scholar 

  119. Koay, E. J. et al. Transport properties of pancreatic cancer describe gemcitabine delivery and response. J. Clin. Invest. 124, 1525–1536 (2014).

    CAS  Google Scholar 

  120. Shao, K. et al. Nanoparticle-based immunotherapy for cancer. ACS Nano 9, 16–30 (2015).

    CAS  Google Scholar 

  121. Zuckerman, J. E. et al. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc. Natl Acad. Sci. USA 111, 11449–11454 (2014).

    CAS  Google Scholar 

  122. Bangham, A. D., Standish, M. M. & Watkins, J. C. Diffusion of univalent ions across the lamellae of swollen phospholipids. J. Mol. Biol. 13, 238–252 (1965).

    CAS  Google Scholar 

Download references

Acknowledgements

The authors thank all authors of the studies that were surveyed for providing additional information on request to calculate the nanoparticle delivery efficiencies (Supplementary information S1 (table)).The authors thank M. Samarakoon and A. Göpferich for fruitful discussions. The authors also thank S. Patel, M. Chalsev, A. Mahmood, J. D. Mora, and Y. Y. Chen for creating the database system. W.C.W.C. acknowledges the Canadian Institutes of Health Research (CIHR), Natural Sciences and Engineering Research Council (NSERC), and Prostate Cancer Canada for supporting his research program. H.F.D. acknowledges financial support from the US National Institutes of Health (NIH) grant P01 CA92644 and by a contract from the National Foundation for Cancer Research. S.O. acknowledges the Japan Society for the Promotion of Science (JSPS) for a Research Fellowship (PD, No. 5621) and the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan for a Grant-in-Aid for Young Scientists (B) (No. 26820356).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Warren C. W. Chan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Supplementary information

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wilhelm, S., Tavares, A., Dai, Q. et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater 1, 16014 (2016). https://doi.org/10.1038/natrevmats.2016.14

Download citation

  • Published:

  • DOI: https://doi.org/10.1038/natrevmats.2016.14

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer