Integrated nanotechnology platform for tumor-targeted multimodal imaging and therapeutic cargo release

Significance The main goal in the emerging field of cancer nanomedicine is to generate, standardize, and produce multifunctional carriers designed to improve the response of drugs against tumors. Here we report the design, development, and preclinical validation of a ligand-directed bioinorganic platform that integrates tumor targeting, receptor-mediated cell internalization, photon-to-heat conversion, and drug delivery. This enabling hydrogel-based technology can accommodate a broad variety of ligands, nanoparticles, and payloads. We show experimental proof-of-concept in mouse models of breast and prostate cancer with molecular imaging and marked reduction of tumor growth. However, with future proof that this technology is translatable, medical applications beyond cancer may also be leveraged. A major challenge of targeted molecular imaging and drug delivery in cancer is establishing a functional combination of ligand-directed cargo with a triggered release system. Here we develop a hydrogel-based nanotechnology platform that integrates tumor targeting, photon-to-heat conversion, and triggered drug delivery within a single nanostructure to enable multimodal imaging and controlled release of therapeutic cargo. In proof-of-concept experiments, we show a broad range of ligand peptide-based applications with phage particles, heat-sensitive liposomes, or mesoporous silica nanoparticles that self-assemble into a hydrogel for tumor-targeted drug delivery. Because nanoparticles pack densely within the nanocarrier, their surface plasmon resonance shifts to near-infrared, thereby enabling a laser-mediated photothermal mechanism of cargo release. We demonstrate both noninvasive imaging and targeted drug delivery in preclinical mouse models of breast and prostate cancer. Finally, we applied mathematical modeling to predict and confirm tumor targeting and drug delivery. These results are meaningful steps toward the design and initial translation of an enabling nanotechnology platform with potential for broad clinical applications.

[1]  B. Ghosh,et al.  Nanocarriers for cancer-targeted drug delivery , 2016, Journal of drug targeting.

[2]  K. Landfester,et al.  Natural liposomes and synthetic polymeric structures for biomedical applications. , 2015, Biochemical and biophysical research communications.

[3]  Hiroshi Maeda,et al.  Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. , 2015, Advanced drug delivery reviews.

[4]  E. Rampazzo,et al.  Targeted tumor imaging of anti-CD20-polymeric nanoparticles developed for the diagnosis of B-cell malignancies , 2015, International journal of nanomedicine.

[5]  K. Landfester,et al.  Carbohydrate-Based Nanocarriers Exhibiting Specific Cell Targeting with Minimum Influence from the Protein Corona. , 2015, Angewandte Chemie.

[6]  D. Dixon,et al.  Gold nanoparticle surface functionalization: a necessary requirement in the development of novel nanotherapeutics. , 2015, Nanomedicine.

[7]  R. Sidman,et al.  Selection and identification of ligand peptides targeting a model of castrate-resistant osteogenic prostate cancer and their receptors , 2015, Proceedings of the National Academy of Sciences.

[8]  V. Patravale,et al.  Polymeric nanoparticles for targeted treatment in oncology: current insights , 2015, International journal of nanomedicine.

[9]  Sang-Jun Park,et al.  Temperature-triggered tumor-specific delivery of anticancer agents by cRGD-conjugated thermosensitive liposomes. , 2014, Colloids and surfaces. B, Biointerfaces.

[10]  Mauro Ferrari,et al.  Transport properties of pancreatic cancer describe gemcitabine delivery and response. , 2014, The Journal of clinical investigation.

[11]  Vittorio Cristini,et al.  Mechanistic modeling identifies drug-uptake history as predictor of tumor drug resistance and nano-carrier-mediated response. , 2013, ACS nano.

[12]  S. L. Kumar,et al.  NANOTECHNOLOGY IN CANCER THERAPY , 2013 .

[13]  Vittorio Cristini,et al.  Mechanistic patient-specific predictive correlation of tumor drug response with microenvironment and perfusion measurements , 2013, Proceedings of the National Academy of Sciences.

[14]  Mark Thomas Smith,et al.  Reengineering viruses and virus-like particles through chemical functionalization strategies. , 2013, Current opinion in biotechnology.

[15]  Eric C. Carnes,et al.  Mesoporous silica nanoparticle nanocarriers: biofunctionality and biocompatibility. , 2013, Accounts of chemical research.

[16]  Andrew Tsourkas,et al.  Effect of ligand density, receptor density, and nanoparticle size on cell targeting. , 2013, Nanomedicine : nanotechnology, biology, and medicine.

[17]  P. Stockley,et al.  MS2 viruslike particles: a robust, semisynthetic targeted drug delivery platform. , 2013, Molecular pharmaceutics.

[18]  D. Koppers-Lalic,et al.  Virus-modified exosomes for targeted RNA delivery; A new approach in nanomedicine☆ , 2012, Advanced Drug Delivery Reviews.

[19]  Joseph W. Nichols,et al.  Odyssey of a cancer nanoparticle: from injection site to site of action. , 2012, Nano today.

[20]  V. Muzykantov,et al.  Multifunctional Nanoparticles: Cost Versus Benefit of Adding Targeting and Imaging Capabilities , 2012, Science.

[21]  Jason L. Townson,et al.  Delivery of Ricin Toxin A‐Chain by Peptide‐Targeted Mesoporous Silica Nanoparticle‐Supported Lipid Bilayers , 2012, Advanced healthcare materials.

[22]  Jason L. Townson,et al.  Delivery of small interfering RNA by peptide-targeted mesoporous silica nanoparticle-supported lipid bilayers. , 2012, ACS nano.

[23]  Mark B. Carter,et al.  The Targeted Delivery of Multicomponent Cargos to Cancer Cells via Nanoporous Particle-Supported Lipid Bilayers , 2011, Nature materials.

[24]  Eric D. Pressly,et al.  Evaluation of multivalent, functional polymeric nanoparticles for imaging applications. , 2011, ACS nano.

[25]  P. Decuzzi,et al.  On the synergistic effects of ligand-mediated and phage-intrinsic properties during in vivo selection. , 2010, Advances in genetics.

[26]  M. Ozawa,et al.  An unrecognized extracellular function for an intracellular adapter protein released from the cytoplasm into the tumor microenvironment , 2009, Proceedings of the National Academy of Sciences.

[27]  C. Jeffrey Brinker,et al.  Porous nanoparticle supported lipid bilayers (protocells) as delivery vehicles. , 2009, Journal of the American Chemical Society.

[28]  M. Ferrari,et al.  Predicting drug pharmacokinetics and effect in vascularized tumors using computer simulation , 2006, Journal of mathematical biology.

[29]  W. Arap,et al.  Bottom-Up Assembly of Hydrogels from Bacteriophage and Au Nanoparticles: The Effect of Cis- and Trans-Acting Factors , 2008, PloS one.

[30]  M. Ozawa,et al.  Techniques to decipher molecular diversity by phage display. , 2007, Methods in molecular biology.

[31]  W. Arap,et al.  In vivo detection of gold-imidazole self-assembly complexes: NIR-SERS signal reporters. , 2006, Analytical chemistry.

[32]  Wadih Arap,et al.  Networks of gold nanoparticles and bacteriophage as biological sensors and cell-targeting agents , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[33]  R. Stafford,et al.  Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[34]  T. Allen Ligand-targeted therapeutics in anticancer therapy , 2002, Nature Reviews Cancer.

[35]  George M. Whitesides,et al.  Molecular Self-Assembly of Aliphatic Thiols on Gold Colloids , 1996 .

[36]  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.