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.

  • Review Article
  • Published:

Targeted drug delivery strategies for precision medicines

Abstract

Progress in the field of precision medicine has changed the landscape of cancer therapy. Precision medicine is propelled by technologies that enable molecular profiling, genomic analysis and optimized drug design to tailor treatments for individual patients. Although precision medicines have resulted in some clinical successes, the use of many potential therapeutics has been hindered by pharmacological issues, including toxicities and drug resistance. Drug delivery materials and approaches have now advanced to a point where they can enable the modulation of a drug’s pharmacological parameters, without compromising the desired effect on molecular targets. Specifically, they can modulate a drug’s pharmacokinetics, stability, absorption and exposure to tumours and healthy tissues, and facilitate the administration of synergistic drug combinations. This Review highlights recent progress in precision therapeutics and drug delivery, and identifies opportunities for strategies to improve the therapeutic index of cancer drugs and, consequently, clinical outcomes.

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

Access options

Buy this article

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

Fig. 1: Major side effects of kinase inhibitors.
Fig. 2: Pharmacological properties of kinase inhibitors.
Fig. 3: Nanoscale delivery approaches for small-molecule cargoes.
Fig. 4: Delivery routes and targets in the tumour microenvironment.
Fig. 5: Organ targeting with drug delivery systems.
Fig. 6: Proposed patient selection and clinical correlate measurements for a precision drug nanomedicine trial.

Similar content being viewed by others

References

  1. Tran, S., DeGiovanni, P. J., Piel, B. & Rai, P. Cancer nanomedicine: a review of recent success in drug delivery. Clin. Transl. Med. 6, 44 (2017).

    Article  Google Scholar 

  2. Dugger, S. A., Platt, A. & Goldstein, D. B. Drug development in the era of precision medicine. Nat. Rev. Drug Discov. 17, 183–196 (2017).

    Article  Google Scholar 

  3. Blanco, E., Shen, H. & Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 33, 941–951 (2015).

    Article  CAS  Google Scholar 

  4. Bhullar, K. S. et al. Kinase-targeted cancer therapies: progress, challenges and future directions. Mol. Cancer 17, 48 (2018).

    Article  Google Scholar 

  5. Ferguson, F. M. & Gray, N. S. Kinase inhibitors: the road ahead. Nat. Rev. Drug Discov. 17, 353–377 (2018). Review of kinase inhibitors as a leading modality in personalized medicine.

    Article  CAS  Google Scholar 

  6. Roskoski, R. Properties of FDA-approved small molecule protein kinase inhibitors. Pharmacol. Res. 144, 19–50 (2019).

    Article  CAS  Google Scholar 

  7. Smalley, I. & Smalley, K. S. M. ERK inhibition: a new front in the war against MAPK pathway–driven cancers? Cancer Discov. 8, 140–142 (2018).

    Article  CAS  Google Scholar 

  8. Yaeger, R. & Corcoran, R. B. Targeting alterations in the RAF–MEK pathway. Cancer Discov. 9, 329–341 (2019).

    Article  CAS  Google Scholar 

  9. Gabizon, A., Shmeeda, H. & Barenholz, Y. Pharmacokinetics of pegylated liposomal doxorubicin. Clin. Pharmacokinet. 42, 419–436 (2003).

    Article  CAS  Google Scholar 

  10. Wilhelm, S. et al. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 1, 16014 (2016). Major challenges of nanomedicines in oncology, with an emphasis on drug loading.

    Article  CAS  Google Scholar 

  11. Song, Q. et al. Reduction responsive self-assembled nanoparticles based on disulfide-linked drug–drug conjugate with high drug loading and antitumor efficacy. Mol. Pharm. 13, 190–201 (2016).

    Article  CAS  Google Scholar 

  12. Shamay, Y. et al. Quantitative self-assembly prediction yields targeted nanomedicines. Nat. Mater. 17, 361–368 (2018).

    Article  CAS  Google Scholar 

  13. Bamrungsap, S. et al. Nanotechnology in therapeutics: a focus on nanoparticles as a drug delivery system. Nanomedicine 7, 1253–1271 (2012).

    Article  CAS  Google Scholar 

  14. Hong, D. et al. Phase I study of LY2606368, a checkpoint kinase 1 inhibitor, in patients with advanced cancer. J. Clin. Oncol. 34, 1764–1771 (2016).

    Article  CAS  Google Scholar 

  15. Mak, G. et al. A phase Ib dose-finding, pharmacokinetic study of the focal adhesion kinase inhibitor GSK2256098 and trametinib in patients with advanced solid tumours. Br. J. Cancer 120, 975–981 (2019).

    Article  CAS  Google Scholar 

  16. Postel-Vinay, S. et al. Clinical benefit in Phase-I trials of novel molecularly targeted agents: does dose matter? Br. J. Cancer 100, 1373–1378 (2009).

    Article  CAS  Google Scholar 

  17. Wong, H. H., Barton, C., Acton, G., McLeod, R. & Halford, S. Trends in the characteristics, dose-limiting toxicities and efficacy of phase I oncology trials: The Cancer Research UK experience. Eur. J. Cancer 66, 9–16 (2016).

    Article  CAS  Google Scholar 

  18. Hanker, A. B., Kaklamani, V. & Arteaga, C. L. Challenges for the clinical development of PI3K inhibitors: strategies to improve their impact in solid tumors. Cancer Discov. 9, 482–491 (2019).

    Article  CAS  Google Scholar 

  19. Bullock, J. M., Rahman, A. & Liu, Q. Lessons learned: dose selection of small molecule-targeted oncology drugs. Clin. Cancer Res. 22, 2630–2638 (2016).

    Article  CAS  Google Scholar 

  20. Waldner, M., Fantus, D., Solari, M. & Thomson, A. W. New perspectives on mTOR inhibitors (rapamycin, rapalogs and TORKinibs) in transplantation. Br. J. Clin. Pharmacol. 82, 1158–1170 (2016).

    Article  CAS  Google Scholar 

  21. Infante, J. R. et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. Lancet Oncol. 13, 773–781 (2012).

    Article  CAS  Google Scholar 

  22. Tyagi, P. & Santiago, C. New features in MEK retinopathy. BMC Ophthalmol. 18, 221–221 (2018).

    Article  Google Scholar 

  23. Khan, K. H. et al. Hyperglycemia and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) inhibitors in phase I trials: incidence, predictive factors, and management. Oncologist 21, 855–860 (2016).

    Article  CAS  Google Scholar 

  24. Touyz, R. M., Herrmann, S. M. S. & Herrmann, J. Vascular toxicities with VEGF inhibitor therapies–focus on hypertension and arterial thrombotic events. J. Am. Soc. Hypertens. 12, 409–425 (2018).

    Article  CAS  Google Scholar 

  25. Touyz, R. M. & Herrmann, J. Cardiotoxicity with vascular endothelial growth factor inhibitor therapy. NPJ Precis. Oncol. 2, 13 (2018).

    Article  Google Scholar 

  26. Massey, P. R., Okman, J. S., Wilkerson, J. & Cowen, E. W. Tyrosine kinase inhibitors directed against the vascular endothelial growth factor receptor (VEGFR) have distinct cutaneous toxicity profiles: a meta-analysis and review of the literature. Support. Care Cancer 23, 1827–1835 (2015).

    Article  Google Scholar 

  27. Thill, M. & Schmidt, M. Management of adverse events during cyclin-dependent kinase 4/6 (CDK4/6) inhibitor-based treatment in breast cancer. Ther. Adv. Med. Oncol. https://doi.org/10.1177/175883591879332 (2018).

  28. Hantschel, O. Unexpected off-targets and paradoxical pathway activation by kinase Inhibitors. ACS Chem. Biol. 10, 234–245 (2015). A description of on-target toxicity of PI3K/AKT/mTOR pathway inhibitors.

    Article  CAS  Google Scholar 

  29. Force, T., Krause, D. S. & Van Etten, R. A. Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat. Rev. Cancer 7, 332–344 (2007).

    Article  CAS  Google Scholar 

  30. Miyazawa, K. Encountering unpredicted off-target effects of pharmacological inhibitors. J. Biochem. 150, 1–3 (2011).

    Article  CAS  Google Scholar 

  31. de Groot, C. O. et al. A cell biologist’s field guide to aurora kinase inhibitors. Front. Oncol. 5, 285 (2015).

    Article  Google Scholar 

  32. Roberts, A. W. et al. Phase 1 study of the safety, pharmacokinetics, and antitumour activity of the BCL2 inhibitor navitoclax in combination with rituximab in patients with relapsed or refractory CD20+ lymphoid malignancies. Br. J. Haematol. 170, 669–678 (2015).

    Article  CAS  Google Scholar 

  33. Cang, S., Iragavarapu, C., Savooji, J., Song, Y. & Liu, D. ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J. Hematol. Oncol. 8, 129 (2015).

    Article  Google Scholar 

  34. Bazak, R., Houri, M., El Achy, S., Kamel, S. & Refaat, T. Cancer active targeting by nanoparticles: a comprehensive review of literature. J. Cancer Res. Clin. Oncol. 141, 769–784 (2015).

    Article  CAS  Google Scholar 

  35. Kantarjian, H. M. et al. Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer 119, 2611–2619 (2013).

    Article  CAS  Google Scholar 

  36. Ashton, S. et al. Aurora kinase inhibitor nanoparticles target tumors with favorable therapeutic index in vivo. Sci. Transl. Med. 8, 325ra17 (2016).

    Article  Google Scholar 

  37. Keen, N. & Taylor, S. Mitotic drivers — inhibitors of the Aurora B Kinase. Cancer Metastasis Rev. 28, 185–195 (2009).

    Article  CAS  Google Scholar 

  38. Burris, H. A. et al. A phase I, open-label, first-time-in-patient dose escalation and expansion study to assess the safety, tolerability, and pharmacokinetics of nanoparticle encapsulated Aurora B kinase inhibitor AZD2811 in patients with advanced solid tumours. J. Clin. Oncol. 35, 15 (2017).

    Article  Google Scholar 

  39. Shamay, Y. et al. P-selectin is a nanotherapeutic delivery target in the tumor microenvironment. Sci. Transl. Med. 8, 345ra87 (2016).

    Article  Google Scholar 

  40. Bachelet, L. et al. Affinity of low molecular weight fucoidan for P-selectin triggers its binding to activated human platelets. Biochim. Biophys. Acta 1790, 141–146 (2009).

    Article  CAS  Google Scholar 

  41. Mizrachi, A. et al. Tumour-specific PI3K inhibition via nanoparticle-targeted delivery in head and neck squamous cell carcinoma. Nat. Commun. 8, 14292 (2017).

    Article  CAS  Google Scholar 

  42. Zumsteg, Z. S. et al. Taselisib (GDC-0032), a potent β-sparing small molecule inhibitor of PI3K, radiosensitizes head and neck squamous carcinomas containing activating PIK3CA alterations. Clin. Cancer Res. 22, 2009–2019 (2016).

    Article  CAS  Google Scholar 

  43. Horn, D., Hess, J., Freier, K., Hoffmann, J. & Freudlsperger, C. Targeting EGFR-PI3K-AKT-mTOR signaling enhances radiosensitivity in head and neck squamous cell carcinoma. Expert Opin. Ther. Targets 19, 795–805 (2015).

    Article  CAS  Google Scholar 

  44. Josephs, D. H., Fisher, D. S., Spicer, J. & Flanagan, R. J. Clinical pharmacokinetics of tyrosine kinase inhibitors: implications for therapeutic drug monitoring. Ther. Drug Monit. 35, 562–587 (2013).

    Article  CAS  Google Scholar 

  45. van Erp, N. P., Gelderblom, H. & Guchelaar, H. J. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat. Rev. 35, 692–706 (2009).

    Article  Google Scholar 

  46. Wells, S. A. et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. J. Clin. Oncol. 30, 134–141 (2012).

    Article  CAS  Google Scholar 

  47. Poggi, L. & Kolesar, J. M. Vismodegib for the treatment of basal cell skin cancer. Am. J. Health Syst. Pharm. 70, 1033–1038 (2013).

    Article  CAS  Google Scholar 

  48. Goel, V. et al. Population pharmacokinetics of sonidegib (LDE225), an oral inhibitor of hedgehog pathway signaling, in healthy subjects and in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 77, 745–755 (2016).

    Article  CAS  Google Scholar 

  49. Burris, H. A. 3rd et al. Phase I safety, pharmacokinetics, and clinical activity study of lapatinib (GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases, in heavily pretreated patients with metastatic carcinomas. J. Clin. Oncol. 23, 5305–5313 (2005).

    Article  CAS  Google Scholar 

  50. Kramkimel, N. et al. Vemurafenib pharmacokinetics and its correlation with efficacy and safety in outpatients with advanced BRAF-mutated melanoma. Target. Oncol. 11, 59–69 (2016).

    Article  CAS  Google Scholar 

  51. Wagner, M. C. et al. Nilotinib shows prolonged intracellular accumulation upon pulse-exposure: a novel mechanism for induction of apoptosis in CML cells. Leukemia 27, 1567–1570 (2013).

    Article  CAS  Google Scholar 

  52. Lipka, D. B., Wagner, M. C., Dziadosz, M. & Fischer, T. Prolonged cellular midostaurin retention suggests potential alternative dosing strategies for FLT3-ITD-positive leukemias. Leukemia 30, 2090–2093 (2016).

    Article  CAS  Google Scholar 

  53. Schafranek, L. et al. Sustained inhibition of STAT5, but not JAK2, is essential for TKI-induced cell death in chronic myeloid leukemia. Leukemia 29, 76–85 (2015).

    Article  CAS  Google Scholar 

  54. Vasconcelos, T., Sarmento, B. & Costa, P. Solid dispersions as strategy to improve oral bioavailability of poor water soluble drugs. Drug Discov. Today 12, 1068–1075 (2007).

    Article  CAS  Google Scholar 

  55. Farouk, F. & Shamma, R. Chemical structure modifications and nano-technology applications for improving ADME-Tox properties, a review. Arch. Pharm. 352, 1800213 (2019).

    Article  Google Scholar 

  56. Chow, E. K. & Ho, D. Cancer nanomedicine: from drug delivery to imaging. Sci. Transl. Med. 5, 216rv14 (2013).

    Article  Google Scholar 

  57. Muhamad, N., Plengsuriyakarn, T. & Na-Bangchang, K. Application of active targeting nanoparticle delivery system for chemotherapeutic drugs and traditional/herbal medicines in cancer therapy: a systematic review. Int. J. Nanomed. 13, 3921–3935 (2018).

    Article  CAS  Google Scholar 

  58. Sykes, E. A. et al. Tailoring nanoparticle designs to target cancer based on tumor pathophysiology. Proc. Natl Acad. Sci. USA 113, E1142–E1151 (2016).

    Article  CAS  Google Scholar 

  59. Carmeliet, P. & Jain, R. K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 10, 417–427 (2011).

    Article  CAS  Google Scholar 

  60. Peng, F. et al. Nanoparticles promote in vivo breast cancer cell intravasation and extravasation by inducing endothelial leakiness. Nat. Nanotechnol. 14, 279–286 (2019).

    Article  CAS  Google Scholar 

  61. Pérez-Medina, C. et al. Nanoreporter PET predicts the efficacy of anti-cancer nanotherapy. Nat. Commun. 7, 11838 (2016).

    Article  Google Scholar 

  62. Boulikas, T. Clinical overview on Lipoplatin: a successful liposomal formulation of cisplatin. Expert Opin. Investig. Drugs 18, 1197–1218 (2009).

    Article  CAS  Google Scholar 

  63. Bartelink, I. H. et al. Tumor drug penetration measurements could be the neglected piece of the personalized cancer treatment puzzle. Clin. Pharmacol. Ther. 106, 148–163 (2019). A road map for the clinical implementation of precision dosing.

    Article  Google Scholar 

  64. Reddy, L. H. & Couvreur, P. Nanotechnology for therapy and imaging of liver diseases. J. Hepatol. 55, 1461–1466 (2011).

    Article  CAS  Google Scholar 

  65. In, G. K. & Nieva, J. Emerging chemotherapy agents in lung cancer: nanoparticles therapeutics for non-small cell lung cancer. Transl. Cancer Res. 4, 340–355 (2015).

    CAS  Google Scholar 

  66. Harisinghani, M. G. et al. Noninvasive detection of clinically occult lymph-node metastases in prostate cancer. N. Engl. J. Med. 348, 2491–2499 (2003).

    Article  Google Scholar 

  67. Heller, D. A. et al. Modular ‘click-in-emulsion’ bone-targeted nanogels. Adv. Mater. 25, 1449–1454 (2013).

    Article  CAS  Google Scholar 

  68. Sago, C. D. et al. Nanoparticles that deliver RNA to bone marrow identified by in vivo directed evolution. J. Am. Chem. Soc. 140, 17095–17105 (2018).

    Article  CAS  Google Scholar 

  69. Alidori, S. et al. Targeted fibrillar nanocarbon RNAi treatment of acute kidney injury. Sci. Transl. Med. 8, 331ra39 (2016).

    Article  Google Scholar 

  70. Williams, R. M. et al. Selective nanoparticle targeting of the renal tubules. Hypertension 71, 87–94 (2018).

    Article  CAS  Google Scholar 

  71. Williams, R. M. et al. Mesoscale nanoparticles selectively target the renal proximal tubule epithelium. Nano Lett. 15, 2358–2364 (2015).

    Article  CAS  Google Scholar 

  72. Choi, C. H. J., Zuckerman, J. E., Webster, P. & Davis, M. E. Targeting kidney mesangium by nanoparticles of defined size. Proc. Natl Acad. Sci. USA 108, 6656–6661 (2011).

    Article  CAS  Google Scholar 

  73. Zuckerman, J. E., Choi, C. H. J., Han, H. & Davis, M. E. Polycation-siRNA nanoparticles can disassemble at the kidney glomerular basement membrane. Proc. Natl Acad. Sci. USA 109, 3137–3142 (2012).

    Article  CAS  Google Scholar 

  74. Lee, H. et al. An endoscope with integrated transparent bioelectronics and theranostic nanoparticles for colon cancer treatment. Nat. Commun. 6, 10059 (2015).

    Article  CAS  Google Scholar 

  75. Elci, S. G. et al. Surface charge controls the suborgan biodistributions of gold nanoparticles. ACS Nano 10, 5536–5542 (2016).

    Article  CAS  Google Scholar 

  76. Hirn, S. et al. Particle size-dependent and surface charge-dependent biodistribution of gold nanoparticles after intravenous administration. Eur. J. Pharm. Biopharm. 77, 407–416 (2011).

    Article  CAS  Google Scholar 

  77. Thapa, B., Kumar, P., Zeng, H. B. & Narain, R. Asialoglycoprotein receptor-mediated gene delivery to hepatocytes using galactosylated polymers. Biomacromolecules 16, 3008–3020 (2015).

    Article  CAS  Google Scholar 

  78. Han, J. et al. Acute and chronic shear stress differently regulate endothelial internalization of nanocarriers targeted to platelet-endothelial cell adhesion molecule-1. ACS Nano 6, 8824–8836 (2012).

    Article  CAS  Google Scholar 

  79. Parhiz, H. et al. PECAM-1 directed re-targeting of exogenous mRNA providing two orders of magnitude enhancement of vascular delivery and expression in lungs independent of apolipoprotein E-mediated uptake. J. Control. Rel. 291, 106–115 (2018).

    Article  CAS  Google Scholar 

  80. Dan, M., Cochran, D. B., Yokel, R. A. & Dziubla, T. D. Binding, transcytosis and biodistribution of anti-PECAM-1 iron oxide nanoparticles for brain-targeted delivery. PLoS ONE 8, e81051 (2013).

    Article  Google Scholar 

  81. Reychler, G. & Michotte, J. B. Development challenges and opportunities in aerosol drug delivery systems in non-invasive ventilation in adults. Expert Opin. Drug Deliv. 16, 153–162 (2019).

    Article  CAS  Google Scholar 

  82. Mainprize, T. et al. Blood-brain barrier opening in primary brain tumors with non-invasive MR-guided focused ultrasound: a clinical safety and feasibility study. Sci. Rep. 9, 321 (2019).

    Article  Google Scholar 

  83. Wang, C., Ye, Y., Hochu, G. M., Sadeghifar, H. & Gu, Z. Enhanced cancer immunotherapy by microneedle patch-assisted delivery of anti-PD1 antibody. Nano Lett. 16, 2334–2340 (2016).

    Article  CAS  Google Scholar 

  84. Shi, J., Kantoff, P. W., Wooster, R. & Farokhzad, O. C. Cancer nanomedicine: progress, challenges and opportunities. Nat. Rev. Cancer 17, 20–37 (2017).

    Article  CAS  Google Scholar 

  85. Hrkach, J. et al. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile. Sci. Transl. Med. 4, 128ra39 (2012).

    Article  Google Scholar 

  86. Wang, Y., Huang, H. Y., Yang, L., Zhang, Z. & Ji, H. Cetuximab-modified mesoporous silica nano-medicine specifically targets EGFR-mutant lung cancer and overcomes drug resistance. Sci. Rep. 6, 25468 (2016).

    Article  CAS  Google Scholar 

  87. Shamay, Y., Golan, M., Tyomkin, D. & David, A. Assessing the therapeutic efficacy of VEGFR-1-targeted polymer drug conjugates in mouse tumor models. J. Control. Rel. 229, 192–199 (2016).

    Article  CAS  Google Scholar 

  88. Oh, P. et al. In vivo proteomic imaging analysis of caveolae reveals pumping system to penetrate solid tumors. Nat. Med. 20, 1062–1068 (2014).

    Article  CAS  Google Scholar 

  89. Shamay, Y., Paulin, D., Ashkenasy, G. & David, A. E-selectin binding peptide–polymer–drug conjugates and their selective cytotoxicity against vascular endothelial cells. Biomaterials 30, 6460–6468 (2009).

    Article  CAS  Google Scholar 

  90. Shamay, Y. et al. Inhibition of primary and metastatic tumors in mice by E-selectin-targeted polymer-drug conjugates. J. Control. Rel. 217, 102–112 (2015).

    Article  CAS  Google Scholar 

  91. Seguin, L., Desgrosellier, J. S., Weis, S. M. & Cheresh, D. A. Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol. 25, 234–240 (2015).

    Article  CAS  Google Scholar 

  92. Hallahan, D. E. et al. Targeting drug delivery to radiation-induced neoantigens in tumor microvasculature. J. Control. Rel. 74, 183–191 (2001).

    Article  CAS  Google Scholar 

  93. Temming, K. et al. Delivery of the p38 MAPkinase inhibitor SB202190 to angiogenic endothelial cells: development of novel RGD-equipped and PEGylated drug–albumin conjugates using platinum(II)-based drug linker technology. Bioconjug. Chem. 17, 1246–1255 (2006).

    Article  CAS  Google Scholar 

  94. Li, X. et al. Synthesis and biological evaluation of RGD-conjugated MEK1/2 kinase inhibitors for integrin-targeted cancer therapy. Molecules 18, 13957–13978 (2013).

    Article  CAS  Google Scholar 

  95. Chandarlapaty, S. Negative feedback and adaptive resistance to the targeted therapy of cancer. Cancer Discov. 2, 311–319 (2012).

    Article  CAS  Google Scholar 

  96. Carracedo, A. et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J. Clin. Invest. 118, 3065–3074 (2008).

    CAS  Google Scholar 

  97. O’Reilly, K. E. et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–1508 (2006).

    Article  Google Scholar 

  98. Sergina, N. V. et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 445, 437–441 (2007).

    Article  CAS  Google Scholar 

  99. Coffee, E. M. et al. Concomitant BRAF and PI3K/mTOR blockade is required for effective treatment of BRAF(V600E) colorectal cancer. Clin. Cancer Res. 19, 2688–2698 (2013).

    Article  CAS  Google Scholar 

  100. Wee, S. et al. PI3K pathway activation mediates resistance to MEK inhibitors in KRAS mutant cancers. Cancer Res. 69, 4286–4293 (2009).

    Article  CAS  Google Scholar 

  101. Molina-Arcas, M., Hancock, D. C., Sheridan, C., Kumar, M. S. & Downward, J. Coordinate direct input of both KRAS and IGF1 receptor to activation of PI3 kinase in KRAS-mutant lung cancer. Cancer Discov. 3, 548–563 (2013).

    Article  CAS  Google Scholar 

  102. Murillo, M. M. et al. RAS interaction with PI3K p110alpha is required for tumor-induced angiogenesis. J. Clin. Invest. 124, 3601–3611 (2014).

    Article  CAS  Google Scholar 

  103. Manchado, E. et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature 534, 647–651 (2016).

    Article  CAS  Google Scholar 

  104. Chandarlapaty, S. et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell 19, 58–71 (2011).

    Article  CAS  Google Scholar 

  105. Serra, V. et al. PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer. Oncogene 30, 2547–2557 (2011).

    Article  CAS  Google Scholar 

  106. Chakrabarty, A., Sanchez, V., Kuba, M. G., Rinehart, C. & Arteaga, C. L. Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. Proc. Natl Acad. Sci. USA 109, 2718–2723 (2012).

    Article  CAS  Google Scholar 

  107. Garrett, J. T. et al. Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase. Proc. Natl Acad. Sci. USA 108, 5021–5026 (2011).

    Article  CAS  Google Scholar 

  108. Tao, J. J. et al. Antagonism of EGFR and HER3 enhances the response to inhibitors of the PI3K-Akt pathway in triple-negative breast cancer. Sci. Signal. 7, ra29 (2014).

    Article  Google Scholar 

  109. Carver, B. S. et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell 19, 575–586 (2011).

    Article  CAS  Google Scholar 

  110. Bosch, A. et al. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor–positive breast cancer. Sci. Transl. Med. 7, 283ra51 (2015).

    Article  Google Scholar 

  111. Toska, E. et al. PI3K pathway regulates ER-dependent transcription in breast cancer through the epigenetic regulator KMT2D. Science 355, 1324–1330 (2017).

    Article  CAS  Google Scholar 

  112. Dickler, M. N. et al. A phase II study of the PI3K inhibitor taselisib (GDC-0032) combined with fulvestrant (F) in patients (pts) with HER2-negative (HER2-), hormone receptor-positive (HR+) advanced breast cancer (BC). J. Clin. Oncol. 34, 520 (2016).

    Article  Google Scholar 

  113. Saura, C. et al. Primary results of LORELEI: a phase II randomized, double-blind study of neoadjuvant letrozole (LET) plus taselisib versus LET plus placebo (PLA) in postmenopausal patients (pts) with ER+/HER2-negative early breast cancer (EBC). Ann. Oncol. https://doi.org/10.1093/annonc/mdx440.001 (2017).

  114. Juvekar, A. et al. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov. 2, 1048–1063 (2012).

    Article  CAS  Google Scholar 

  115. Sun, C. et al. Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers. Sci. Transl. Med. 9, eaal5148 (2017).

    Article  Google Scholar 

  116. Lee, H.-Y. et al. Response of non-small cell lung cancer cells to the inhibitors of phosphatidylinositol 3-kinase/Akt- and MAPK kinase 4/c-Jun NH2-terminal kinase pathways: an effective therapeutic strategy for lung cancer. Clin. Cancer Res. 11, 6065–6074 (2005).

    Article  CAS  Google Scholar 

  117. Duong, H.-Q., Kim, H. J., Kang, H. J., Seong, Y.-S. & Bae, I. ZSTK474, a PI3K inhibitor, suppresses proliferation and sensitizes to gemcitabine in human pancreatic adenocarcinoma cells. Oncol. Rep. 27, 182–188 (2012).

    CAS  Google Scholar 

  118. Dreaden, E. C. et al. Tumor-targeted synergistic blockade of MAPK and PI3K from a layer-by-layer nanoparticle. Clin. Cancer Res. 21, 4410–4419 (2015). A notable example of the use of nanomedicine to enable a therapeutic combination to combat drug resistance and avoid toxicity of kinase inhibitors.

    Article  CAS  Google Scholar 

  119. Pisarevsky, E. et al. Rational design of polyglutamic acid delivering an optimized combination of drugs targeting mutated BRAF and MEK in melanoma. Adv. Ther. https://doi.org/10.1002/adtp.202000028 (2020).

  120. Morton, S. W. et al. A nanoparticle-based combination chemotherapy delivery system for enhanced tumor killing by dynamic rewiring of signaling pathways. Sci. Signal. 7, ra44 (2014).

    Article  Google Scholar 

  121. Ibrahim, Y. H. et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov. 2, 1036–1047 (2012).

    Article  CAS  Google Scholar 

  122. Patel, S. et al. Boosting intracellular delivery of lipid nanoparticle-encapsulated mRNA. Nano Lett. 17, 5711–5718 (2017).

    Article  CAS  Google Scholar 

  123. Fire, A. et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391, 806–811 (1998).

    Article  CAS  Google Scholar 

  124. Ting, A. H., Schuebel, K. E., Herman, J. G. & Baylin, S. B. Short double-stranded RNA induces transcriptional gene silencing in human cancer cells in the absence of DNA methylation. Nat. Genet. 37, 906–910 (2005).

    Article  CAS  Google Scholar 

  125. Xu, C.-f & Wang, J. Delivery systems for siRNA drug development in cancer therapy. Asian J. Pharm. Sci. 10, 1–12 (2015).

    Article  Google Scholar 

  126. Jackson, A. L. & Linsley, P. S. Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application. Nat. Rev. Drug Discov. 9, 57–67 (2010).

    Article  CAS  Google Scholar 

  127. Wang, J., Lu, Z., Wientjes, M. G. & Au, J. L. S. Delivery of siRNA therapeutics: barriers and carriers. AAPS J. 12, 492–503 (2010).

    Article  CAS  Google Scholar 

  128. Wong, A. W., Baginski, T. K. & Reilly, D. E. Enhancement of DNA uptake in FUT8-deleted CHO cells for transient production of afucosylated antibodies. Biotechnol. Bioeng. 106, 751–763 (2010).

    Article  CAS  Google Scholar 

  129. Kim, Y. G., Cha, J. & Chandrasegaran, S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc. Natl Acad. Sci. USA 93, 1156–1160 (1996).

    Article  CAS  Google Scholar 

  130. Chou, S. T., Leng, Q. & Mixson, A. J. Zinc finger nucleases: tailor-made for gene therapy. Drugs Future 37, 183–196 (2012).

    Article  CAS  Google Scholar 

  131. LaFountaine, J. S., Fathe, K. & Smyth, H. D. C. Delivery and therapeutic applications of gene editing technologies ZFNs, TALENs, and CRISPR/Cas9. Int. J. Pharm. 494, 180–194 (2015).

    Article  CAS  Google Scholar 

  132. Perez, E. E. et al. Establishment of HIV-1 resistance in CD4(+) T cells by genome editing using zinc-finger nucleases. Nat. Biotechnol. 26, 808–816 (2008).

    Article  CAS  Google Scholar 

  133. Mussolino, C. et al. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 39, 9283–9293 (2011).

    Article  CAS  Google Scholar 

  134. Zhu, H. et al. Baculoviral transduction facilitates TALEN-mediated targeted transgene integration and Cre/LoxP cassette exchange in human-induced pluripotent stem cells. Nucleic Acids Res. 41, e180 (2013).

    Article  CAS  Google Scholar 

  135. Hu, Z. et al. TALEN-mediated targeting of HPV oncogenes ameliorates HPV-related cervical malignancy. J. Clin. Invest. 125, 425–436 (2015).

    Article  Google Scholar 

  136. Ru, R. et al. Targeted genome engineering in human induced pluripotent stem cells by penetrating TALENs. Cell Regen. 2, 5 (2013).

    Article  CAS  Google Scholar 

  137. Liu, J., Gaj, T., Patterson, J. T., Sirk, S. J. & Barbas III, C. F. Cell-penetrating peptide-mediated delivery of TALEN proteins via bioconjugation for genome engineering. PLoS ONE 9, e85755 (2014).

    Article  Google Scholar 

  138. Wang, J. & Quake, S. R. RNA-guided endonuclease provides a therapeutic strategy to cure latent herpesviridae infection. Proc. Natl Acad. Sci. USA 111, 13157–13162 (2014).

    Article  CAS  Google Scholar 

  139. Cheng, R. et al. Efficient gene editing in adult mouse livers via adenoviral delivery of CRISPR/Cas9. FEBS Lett. 588, 3954–3958 (2014).

    Article  CAS  Google Scholar 

  140. Mintzer, M. A. & Simanek, E. E. Nonviral vectors for gene delivery. Chem. Rev. 109, 259–302 (2009).

    Article  CAS  Google Scholar 

  141. Putnam, D. Polymers for gene delivery across length scales. Nat. Mater. 5, 439–451 (2006).

    Article  CAS  Google Scholar 

  142. Davis, M. E. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol. Pharm. 6, 659–668 (2009).

    Article  CAS  Google Scholar 

  143. Semple, S. C. et al. Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 28, 172–176 (2010).

    Article  CAS  Google Scholar 

  144. Love, K. T. et al. Lipid-like materials for low-dose, in vivo gene silencing. Proc. Natl Acad. Sci. USA 107, 1864–1869 (2010).

    Article  CAS  Google Scholar 

  145. Monopoli, M. P., Aberg, C., Salvati, A. & Dawson, K. A. Biomolecular coronas provide the biological identity of nanosized materials. Nat. Nano 7, 779–786 (2012).

    Article  CAS  Google Scholar 

  146. Ando, H. et al. Polycation liposomes as a vector for potential intracellular delivery of microRNA. J. Gene Med. 15, 375–383 (2013).

    Article  CAS  Google Scholar 

  147. Kim, J. S., Oh, M. H., Park, J. Y., Park, T. G. & Nam, Y. S. Protein-resistant, reductively dissociable polyplexes for in vivo systemic delivery and tumor-targeting of siRNA. Biomaterials 34, 2370–2379 (2013).

    Article  CAS  Google Scholar 

  148. Guo, J. et al. Systemic delivery of therapeutic small interfering RNA using a pH-triggered amphiphilic poly-l-lysine nanocarrier to suppress prostate cancer growth in mice. Eur. J. Pharm. Sci. 45, 521–532 (2012).

    Article  CAS  Google Scholar 

  149. Weinstein, S. et al. Harnessing RNAi-based nanomedicines for therapeutic gene silencing in B-cell malignancies. Proc. Natl Acad. Sci. USA 113, E16–E22 (2016).

    Article  CAS  Google Scholar 

  150. Tabernero, J. et al. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer Discov. 3, 406–417 (2013).

    Article  CAS  Google Scholar 

  151. Liang, X. et al. Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. J. Biotechnol. 208, 44–53 (2015).

    Article  CAS  Google Scholar 

  152. Zuris, J. A. et al. Efficient delivery of genome-editing proteins in vitro and in vivo. Nat. Biotechnol. 33, 73–80 (2015).

    Article  CAS  Google Scholar 

  153. Wang, M. et al. Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc. Natl Acad. Sci. USA 113, 2868–2873 (2016).

    Article  CAS  Google Scholar 

  154. Yin, H. et al. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat. Biotechnol. 34, 328–333 (2016).

    Article  CAS  Google Scholar 

  155. Miller, J. B. et al. Non-viral CRISPR/Cas gene editing in vitro and in vivo enabled by synthetic nanoparticle co-delivery of Cas9 mRNA and sgRNA. Angew. Chem. Int. Ed. Engl. 56, 1059–1063 (2017).

    Article  CAS  Google Scholar 

  156. Ramakrishna, S. et al. Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA. Genome Res. 24, 1020–1027 (2014).

    Article  CAS  Google Scholar 

  157. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  Google Scholar 

  158. Wei, T., Cheng, Q., Min, Y.-L., Olson, E. N. & Siegwart, D. J. Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat. Commun. 11, 3232 (2020).

    Article  CAS  Google Scholar 

  159. Hu, Q. et al. Conjugation of haematopoietic stem cells and platelets decorated with anti-PD-1 antibodies augments anti-leukaemia efficacy. Nat. Biomed. Eng. 2, 831–840 (2018).

    Article  CAS  Google Scholar 

  160. Robert, C. et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 372, 320–330 (2014).

    Article  Google Scholar 

  161. Chen, Q. et al. Photothermal therapy promotes tumor infiltration and antitumor activity of CAR T cells. Adv. Mater. 31, 1900192 (2019).

    Article  Google Scholar 

  162. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    Article  Google Scholar 

  163. Menon, S., Shin, S. & Dy, G. Advances in cancer immunotherapy in solid tumors. Cancers 8, 106 (2016).

    Article  Google Scholar 

  164. de Charette, M., Marabelle, A. & Houot, R. Turning tumour cells into antigen presenting cells: The next step to improve cancer immunotherapy? Eur. J. Cancer 68, 134–147 (2016).

    Article  Google Scholar 

  165. Santomasso, B., Bachier, C., Westin, J., Rezvani, K. & Shpall, E. J. The other side of CAR T-cell therapy: cytokine release syndrome, neurologic toxicity, and financial burden. Am. Soc. Clin. Oncol. Educ. Book 39, 433–444 (2019).

    Article  Google Scholar 

  166. Jo, S. D., Nam, G.-H., Kwak, G., Yang, Y. & Kwon, I. Harnessing designed nanoparticles: Current strategies and future perspectives in cancer immunotherapy. Nano Today 17, 23–37 (2017).

    Article  CAS  Google Scholar 

  167. Nam, J. et al. Cancer nanomedicine for combination cancer immunotherapy. Nat. Rev. Mater. 4, 398–414 (2019). Opportunities for combination immunotherapy based on nanoparticle platforms designed for chemotherapy, photothermal therapy, photodynamic therapy, radiotherapy and gene therapy.

    Article  Google Scholar 

  168. Liu, Z., Jiang, W., Nam, J., Moon, J. J. & Kim, B. Y. S. Immunomodulating nanomedicine for cancer therapy. Nano Lett. 18, 6655–6659 (2018).

    Article  CAS  Google Scholar 

  169. Zhang, L.-x, Xie, X.-x, Liu, D.-q, Xu, Z. P. & Liu, R.-t Efficient co-delivery of neo-epitopes using dispersion-stable layered double hydroxide nanoparticles for enhanced melanoma immunotherapy. Biomaterials 174, 54–66 (2018).

    Article  CAS  Google Scholar 

  170. Alskar, L. C., Porter, C. J. & Bergstrom, C. A. Tools for early prediction of drug loading in lipid-based formulations. Mol. Pharm. 13, 251–261 (2016).

    Article  Google Scholar 

  171. Kuai, R., Ochyl, L. J., Bahjat, K. S., Schwendeman, A. & Moon, J. J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 16, 489–496 (2016).

    Article  Google Scholar 

  172. Jeanbart, L. et al. Enhancing efficacy of anticancer vaccines by targeted delivery to tumor-draining lymph nodes. Cancer Immunol. Res. 2, 436–447 (2014).

    Article  CAS  Google Scholar 

  173. Kim, J. et al. Injectable, spontaneously assembling, inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy. Nat. Biotechnol. 33, 64–72 (2015).

    Article  CAS  Google Scholar 

  174. Li, A. W. et al. A facile approach to enhance antigen response for personalized cancer vaccination. Nat. Mater. 17, 528–534 (2018).

    Article  CAS  Google Scholar 

  175. Lu, K. et al. Low-dose X-ray radiotherapy–radiodynamic therapy via nanoscale metal–organic frameworks enhances checkpoint blockade immunotherapy. Nat. Biomed. Eng. 2, 600–610 (2018).

    Article  CAS  Google Scholar 

  176. Sau, S. et al. Multifunctional nanoparticles for cancer immunotherapy: A groundbreaking approach for reprogramming malfunctioned tumor environment. J. Control. Rel. 274, 24–34 (2018).

    Article  CAS  Google Scholar 

  177. Lu, C. et al. Phase I clinical trial of systemically administered TUSC2(FUS1)-nanoparticles mediating functional gene transfer in humans. PLoS ONE 7, e34833 (2012).

    Article  CAS  Google Scholar 

  178. Ji, L. et al. Expression of several genes in the human chromosome 3p21.3 homozygous deletion region by an adenovirus vector results in tumor suppressor activities in vitro and in vivo. Cancer Res. 62, 2715–2720 (2002).

    CAS  Google Scholar 

  179. Demeure, M. J. et al. A phase I/II study of TKM-080301, a PLK1-targeted RNAi in patients with adrenocortical cancer (ACC). J. Clin. Oncol. 34, 2547–2547 (2016).

    Article  Google Scholar 

  180. M.D. Anderson Cancer Center https://ClinicalTrials.gov/show/NCT01591356 (2015).

  181. Sarker, D. et al. First-in-human, first-in-class phase I study of MTL-CEBPA, a small activating RNA (saRNA) targeting the transcription factor C/EBP-α in patients with advanced liver cancer. J. Clin. Oncol. 35, TPS2612 (2017).

    Article  Google Scholar 

  182. Gonzalez-Angulo, A. M. et al. Weekly nab-rapamycin in patients with advanced nonhematologic malignancies: final results of a phase I trial. Clin. Cancer Res. 19, 5474–5484 (2013).

    Article  CAS  Google Scholar 

  183. Plummer, R. et al. A Phase I clinical study of cisplatin-incorporated polymeric micelles (NC-6004) in patients with solid tumours. Br. J. Cancer 104, 593–598 (2011).

    Article  CAS  Google Scholar 

  184. Shamay, Y. Quantitative prediction of self assembly yields tumor targeted nanoparticles. Nat. Mater. 17, 361–368 (2018). Predicting drug encapsulation into nanocarriers based on drug molecular structure.

    Article  CAS  Google Scholar 

  185. Tuomela, A., Hirvonen, J. & Peltonen, L. Stabilizing agents for drug nanocrystals: effect on bioavailability. Pharmaceutics 8, 16 (2016).

    Article  Google Scholar 

  186. Guo, S. & Huang, L. Nanoparticles containing insoluble drug for cancer therapy. Biotechnol. Adv. 32, 778–788 (2014).

    Article  CAS  Google Scholar 

  187. Woodhead, J. L. & Hall, C. K. Encapsulation efficiency and micellar structure of solute-carrying block copolymer nanoparticles. Macromolecules 44, 5443–5451 (2011).

    Article  CAS  Google Scholar 

  188. Dinarvand, R., Sepehri, N., Manoochehri, S., Rouhani, H. & Atyabi, F. Polylactide-co-glycolide nanoparticles for controlled delivery of anticancer agents. Int. J. Nanomed. 6, 877–895 (2011).

    Article  CAS  Google Scholar 

  189. Maojo, V. et al. Nanoinformatics: a new area of research in nanomedicine. Int. J. Nanomed. 7, 3867–3890 (2012).

    Article  Google Scholar 

  190. Seidler, J., McGovern, S. L., Doman, T. N. & Shoichet, B. K. Identification and prediction of promiscuous aggregating inhibitors among known drugs. J. Med. Chem. 46, 4477–4486 (2003).

    Article  CAS  Google Scholar 

  191. Irwin, J. J. et al. An aggregation advisor for ligand discovery. J. Med. Chem. 58, 7076–7087 (2015).

    Article  CAS  Google Scholar 

  192. Feng, B. Y., Shelat, A., Doman, T. N., Guy, R. K. & Shoichet, B. K. High-throughput assays for promiscuous inhibitors. Nat. Chem. Biol. 1, 146–148 (2005).

    Article  CAS  Google Scholar 

  193. Puzyn, T. et al. Using nano-QSAR to predict the cytotoxicity of metal oxide nanoparticles. Nat. Nanotechnol. 6, 175–178 (2011).

    Article  CAS  Google Scholar 

  194. Zhang, Y. et al. Lipid-modified aminoglycoside derivatives for in vivo siRNA delivery. Adv. Mater. 25, 4641–4645 (2013).

    Article  CAS  Google Scholar 

  195. Guo, C., Luo, Y., Zhou, R. & Wei, G. Probing the self-assembly mechanism of diphenylalanine-based peptide nanovesicles and nanotubes. ACS Nano 6, 3907–3918 (2012).

    Article  CAS  Google Scholar 

  196. Guo, C., Luo, Y., Zhou, R. & Wei, G. Triphenylalanine peptides self-assemble into nanospheres and nanorods that are different from the nanovesicles and nanotubes formed by diphenylalanine peptides. Nanoscale 6, 2800–2811 (2014).

    Article  CAS  Google Scholar 

  197. Lee, O. S., Stupp, S. I. & Schatz, G. C. Atomistic molecular dynamics simulations of peptide amphiphile self-assembly into cylindrical nanofibers. J. Am. Chem. Soc. 133, 3677–3683 (2011).

    Article  CAS  Google Scholar 

  198. Frederix, P. W. et al. Exploring the sequence space for (tri-)peptide self-assembly to design and discover new hydrogels. Nat. Chem. 7, 30–37 (2015).

    Article  CAS  Google Scholar 

  199. Shi, C. et al. A drug-specific nanocarrier design for efficient anticancer therapy. Nat. Commun. 6, 7449 (2015). De novo nanocarrier design based on drug structure to enhance activity.

    Article  Google Scholar 

  200. Yaari, Z. et al. Theranostic barcoded nanoparticles for personalized cancer medicine. Nat. Commun. 7, 13325 (2016).

    Article  CAS  Google Scholar 

  201. Gebhart, G. et al. Molecular imaging as a tool to investigate heterogeneity of advanced HER2-positive breast cancer and to predict patient outcome under trastuzumab emtansine (T-DM1): the ZEPHIR trial. Ann. Oncol. 27, 619–624 (2016).

    Article  CAS  Google Scholar 

  202. Perez-Gracia, J. L. et al. Strategies to design clinical studies to identify predictive biomarkers in cancer research. Cancer Treat. Rev. 53, 79–97 (2017).

    Article  Google Scholar 

  203. Arai, N., Sasaki, H., Tamura, R., Ohara, K. & Yoshida, K. Unusual magnetic resonance imaging findings of a glioblastoma arising during treatment with lenvatinib for thyroid cancer. World Neurosurg. 107, 1047.e9–1047.e15 (2017).

    Article  Google Scholar 

  204. Man, F., Lammers, T. & de Rosales, R. T. M. Imaging nanomedicine-based drug delivery: a review of clinical studies. Mol. Imaging Biol. 20, 683–695 (2018). A review of clinical data on the imaging of nanoparticle uptake into tumours to predict efficacy of drug delivery in tumours.

    Article  Google Scholar 

  205. Weickhardt, A. J. et al. Rapid-onset hypogonadism secondary to crizotinib use in men with metastatic nonsmall cell lung cancer. Cancer 118, 5302–5309 (2012).

    Article  CAS  Google Scholar 

  206. Chapman, P. B. et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 364, 2507–2516 (2011).

    Article  CAS  Google Scholar 

  207. Lee, H. et al. (64)Cu-MM-302 positron emission tomography quantifies variability of enhanced permeability and retention of nanoparticles in relation to treatment response in patients with metastatic breast cancer. Clin. Cancer Res. 23, 4190–4202 (2017).

    Article  CAS  Google Scholar 

  208. Golombek, S. K. et al. Tumor targeting via EPR: Strategies to enhance patient responses. Adv. Drug. Deliv. Rev. 130, 17–38 (2018).

    Article  CAS  Google Scholar 

  209. Lammers, T. et al. Cancer nanomedicine: is targeting our target? Nat. Rev. Mater. 1, 16069 (2016).

    Article  CAS  Google Scholar 

  210. Schmid, D. et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 8, 1747 (2017).

    Article  Google Scholar 

  211. Lucky, S. S., Soo, K. C. & Zhang, Y. Nanoparticles in photodynamic therapy. Chem. Rev. 115, 1990–2042 (2015).

    Article  CAS  Google Scholar 

  212. Tinkle, S. et al. Nanomedicines: addressing the scientific and regulatory gap. Ann. N. Y. Acad. Sci. 1313, 35–56 (2014).

    Article  CAS  Google Scholar 

  213. Crist, R. M. et al. Common pitfalls in nanotechnology: lessons learned from NCI’s Nanotechnology Characterization Laboratory. Integr. Biol. 5, 66–73 (2013). Trends in nanoparticle safety and biocompatibility, and common mistakes regarding nanoparticle development for translation.

    Article  CAS  Google Scholar 

  214. Piret, J.-P. et al. Pan-European inter-laboratory studies on a panel of in vitro cytotoxicity and pro-inflammation assays for nanoparticles. Arch. Toxicol. 91, 2315–2330 (2017).

    Article  CAS  Google Scholar 

  215. Kantarjian, H. M. et al. Bosutinib safety and management of toxicity in leukemia patients with resistance or intolerance to imatinib and other tyrosine kinase inhibitors. Blood 9, 1309–1318 (2014).

    Article  Google Scholar 

  216. Shaw, A. T. et al. Ceritinib in ALK-rearranged non–small-cell lung cancer. N. Engl. J. Med. 370, 1189–1197 (2014).

    Article  CAS  Google Scholar 

  217. Hauschild, A. et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 380, 358–365 (2012).

    Article  CAS  Google Scholar 

  218. Robert, C. et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 372, 30–39 (2015).

    Article  Google Scholar 

  219. Welsh, S. J. & Corrie, P. G. Management of BRAF and MEK inhibitor toxicities in patients with metastatic melanoma. Ther. Adv. Med. Oncol. 7, 122–136 (2015).

    Article  CAS  Google Scholar 

  220. Sequist, L. V. et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J. Clin. Oncol. 31, 3327–3334 (2013).

    Article  CAS  Google Scholar 

  221. Flaherty, K. T. et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 367, 107–114 (2012).

    Article  CAS  Google Scholar 

  222. Wells, E. M., Rao, A. A. N., Scafidi, J. & Packer, R. J. Neurotoxicity of biologically targeted agents in pediatric cancer trials. Pediatr. Neurol. 46, 212–221 (2012).

    Article  Google Scholar 

  223. Dreyling, M. et al. Phase 2A study of copanlisib, aNnovel PI3K Inhibitor, in patients with indolent lymphoma. Blood 124, 1701 (2014).

    Article  Google Scholar 

  224. Younes, A. et al. An open-label phase II study of buparlisib (BKM120) in patients with relapsed and refractory diffuse large B-cell lymphoma, mantle cell lymphoma or follicular lymphoma. Blood 124, 1718 (2014).

    Article  Google Scholar 

  225. Younes, A. et al. An open-label phase II study of buparlisib (BKM120) in patients with relapsed and refractory diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL) and follicular lymphoma (FL). Blood 126, 1493 (2015).

    Article  Google Scholar 

  226. Coutré, S. E. et al. Management of adverse events associated with idelalisib treatment: expert panel opinion. Leuk. Lymphoma 56, 2779–2786 (2015).

    Article  Google Scholar 

  227. Cheah, C. Y. & Fowler, N. H. Idelalisib in the management of lymphoma. Blood 128, 331–336 (2016).

    Article  CAS  Google Scholar 

  228. Robinson, G. W. et al. Irreversible growth plate fusions in children with medulloblastoma treated with a targeted hedgehog pathway inhibitor. Oncotarget 8, 69295–69302 (2017).

    Article  Google Scholar 

  229. Seet, R. C. S., Rabinstein, A. A., Lindell, P. E., Uhm, J. H. & Wijdicks, E. F. Cerebrovascular events after bevacizumab treatment: an early and severe complication. Neurocrit. Care 15, 421–427 (2011).

    Article  CAS  Google Scholar 

  230. Seet, R. C. S. & Rabinstein, A. A. Clinical features and outcomes of posterior reversible encephalopathy syndrome following bevacizumab treatment. QJM 105, 69–75 (2012).

    Article  CAS  Google Scholar 

  231. Grothey, A. et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 381, 303–312 (2013).

    Article  CAS  Google Scholar 

  232. Jhaveri, K. D., Wanchoo, R., Sakhiya, V., Ross, D. W. & Fishbane, S. Adverse renal effects of novel molecular oncologic targeted therapies: a narrative review. Kidney Int. Rep. 2, 108–123 (2017).

    Article  Google Scholar 

  233. Elisei, R. et al. Cabozantinib in progressive medullary thyroid cancer. J. Clin. Oncol. 31, 3639–3646 (2013).

    Article  CAS  Google Scholar 

  234. Schlumberger, M. et al. Lenvatinib versus placebo in radioiodine-refractory thyroid caner. N. Engl. J. Med. 372, 621–630 (2015).

    Article  Google Scholar 

  235. Qi, W.-X., Shen, Z., Tang, L.-N. & Yao, Y. Congestive heart failure risk in cancer patients treated with vascular endothelial growth factor tyrosine kinase inhibitors: a systematic review and meta-analysis of 36 clinical trials. Br. J. Clin. Pharmacol. 78, 748–762 (2014).

    Article  CAS  Google Scholar 

  236. Alibolandi, M. et al. The chemotherapeutic potential of doxorubicin-loaded PEG-b-PLGA nanopolymersomes in mouse breast cancer model. Eur. J. Pharm. Biopharm. 94, 521–531 (2015).

    Article  CAS  Google Scholar 

  237. Sirsi, S. R., Williams, J. H. & Lutz, G. J. Poly(ethylene imine)–poly(ethylene glycol) copolymers facilitate efficient delivery of antisense oligonucleotides to nuclei of mature muscle cells of mdx mice. Hum. Gene Ther. 16, 1307–1317 (2005).

    Article  CAS  Google Scholar 

  238. Tian, H. Y. et al. Biodegradable cationic PEG-PEI-PBLG hyperbranched block copolymer: synthesis and micelle characterization. Biomaterials 26, 4209–4217 (2005).

    Article  CAS  Google Scholar 

  239. Kulkarni, S. B., Betageri, G. V. & Singh, M. Factors affecting microencapsulation of drugs in liposomes. J. Microencapsul. 12, 229–246 (1995).

    Article  CAS  Google Scholar 

  240. Allen, T. M., Sapra, P. & Moase, E. Use of the post-insertion method for the formation of ligand-coupled liposomes. Cell. Mol. Biol. Lett. 7, 889–894 (2002).

    CAS  Google Scholar 

  241. Nii, T. & Ishii, F. Encapsulation efficiency of water-soluble and insoluble drugs in liposomes prepared by the microencapsulation vesicle method. Int. J. Pharm. 298, 198–205 (2005).

    Article  CAS  Google Scholar 

  242. Muller, R. H. & Keck, C. M. Challenges and solutions for the delivery of biotech drugs–a review of drug nanocrystal technology and lipid nanoparticles. J. Biotechnol. 113, 151–170 (2004).

    Article  CAS  Google Scholar 

  243. Lv, P. P. et al. Porous quaternized chitosan nanoparticles containing paclitaxel nanocrystals improved therapeutic efficacy in non-small-cell lung cancer after oral administration. Biomacromolecules 12, 4230–4239 (2011).

    Article  CAS  Google Scholar 

  244. Junghanns, J. U. & Muller, R. H. Nanocrystal technology, drug delivery and clinical applications. Int. J. Nanomed. 3, 295–309 (2008).

    CAS  Google Scholar 

  245. Peterson, C. M. et al. HPMA copolymer delivery of chemotherapy and photodynamic therapy in ovarian cancer. Adv. Exp. Med. Biol. 519, 101–123 (2003).

    Article  CAS  Google Scholar 

  246. Veronese, F. M. et al. PEG-doxorubicin conjugates: influence of polymer structure on drug release, in vitro cytotoxicity, biodistribution, and antitumor activity. Bioconjug. Chem. 16, 775–784 (2005).

    Article  CAS  Google Scholar 

  247. Fox, M. E., Szoka, F. C. & Frechet, J. M. Soluble polymer carriers for the treatment of cancer: the importance of molecular architecture. Acc. Chem. Res. 42, 1141–1151 (2009).

    Article  CAS  Google Scholar 

  248. Desai, N. et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer Res. 12, 1317–1324 (2006).

    Article  CAS  Google Scholar 

  249. Altintas, I. et al. Nanobody-albumin nanoparticles (NANAPs) for the delivery of a multikinase inhibitor 17864 to EGFR overexpressing tumor cells. J. Control. Rel. 165, 110–118 (2013).

    Article  CAS  Google Scholar 

  250. Dong, C., Li, B., Li, Z., Shetty, S. & Fu, J. Dasatinib-loaded albumin nanoparticles possess diminished endothelial cell barrier disruption and retain potent anti-leukemia cell activity. Oncotarget 7, 49699–49709 (2016).

    Article  Google Scholar 

  251. Gu, J., Fan, W., Shimojima, A. & Okubo, T. Organic–inorganic mesoporous nanocarriers integrated with biogenic ligands. Small 3, 1740–1744 (2007).

    Article  CAS  Google Scholar 

  252. Loh, X. J. & Lee, T. C. Gene delivery by functional inorganic nanocarriers. Recent Pat. DNA Gene Seq. 6, 108–114 (2012).

    Article  CAS  Google Scholar 

  253. Lin, G., Mi, P., Chu, C., Zhang, J. & Liu, G. Inorganic nanocarriers overcoming multidrug resistance for cancer theranostics. Adv. Sci. 3, 1600134 (2016).

    Article  Google Scholar 

  254. Nissinen, T. et al. Tailored dual PEGylation of inorganic porous nanocarriers for extremely long blood circulation in vivo. ACS Appl. Mater. Interfaces 8, 32723–32731 (2016).

    Article  CAS  Google Scholar 

  255. Pan, G. et al. Mesoporous silica nanoparticles (MSNs)-based organic/inorganic hybrid nanocarriers loading 5-Fluorouracil for the treatment of colon cancer with improved anticancer efficacy. Colloids Surf. B Biointerfaces 159, 375–385 (2017).

    Article  CAS  Google Scholar 

  256. Levina, M. & Rajabi-Siahboomi, A. R. The influence of excipients on drug release from hydroxypropyl methylcellulose matrices. J. Pharm. Sci. 93, 2746–2754 (2004).

    Article  CAS  Google Scholar 

  257. Jeong, B., Bae, Y. H. & Kim, S. W. Drug release from biodegradable injectable thermosensitive hydrogel of PEG–PLGA–PEG triblock copolymers. J. Control. Rel. 63, 155–163 (2000).

    Article  CAS  Google Scholar 

  258. Kamaly, N., Yameen, B., Wu, J. & Farokhzad, O. C. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem. Rev. 116, 2602–2663 (2016).

    Article  CAS  Google Scholar 

  259. Manzari, M. T. et al. Genomically informed small-molecule drugs overcome resistance to a sustained-release formulation of an engineered death receptor agonist in patient-derived tumor models. Sci. Adv. 5, eaaw9162 (2019).

    Article  CAS  Google Scholar 

  260. Tanner, T. & Marks, R. Delivering drugs by the transdermal route: review and comment. Skin Res. Technol. 14, 249–260 (2008).

    Article  CAS  Google Scholar 

  261. Ma, G. & Wu, C. Microneedle, bio-microneedle and bio-inspired microneedle: a review. J. Control. Rel. 251, 11–23 (2017).

    Article  CAS  Google Scholar 

  262. Prausnitz, M. R. Microneedles for transdermal drug delivery. Adv. Drug Deliv. Rev. 56, 581–587 (2004).

    Article  CAS  Google Scholar 

  263. Ruel-Gariépy, E. & Leroux, J.-C. In situ-forming hydrogels—review of temperature-sensitive systems. Eur. J. Pharm. Biopharm. 58, 409–426 (2004).

    Article  Google Scholar 

  264. Murdan, S. Electro-responsive drug delivery from hydrogels. J. Control. Rel. 92, 1–17 (2003).

    Article  CAS  Google Scholar 

  265. Lin, C.-C. & Metters, A. T. Hydrogels in controlled release formulations: Network design and mathematical modeling. Adv. Drug Deliv. Rev. 58, 1379–1408 (2006).

    Article  CAS  Google Scholar 

  266. Sham, J. O. H., Zhang, Y., Finlay, W. H., Roa, W. H. & Löbenberg, R. Formulation and characterization of spray-dried powders containing nanoparticles for aerosol delivery to the lung. Int. J. Pharm. 269, 457–467 (2004).

    Article  CAS  Google Scholar 

  267. Sung, J. C., Pulliam, B. L. & Edwards, D. A. Nanoparticles for drug delivery to the lungs. Trends Biotechnol. 25, 563–570 (2007).

    Article  CAS  Google Scholar 

  268. Mansour, H. M., Rhee, Y.-S. & Wu, X. Nanomedicine in pulmonary delivery. Int. J. Nanomed. 4, 299–319 (2009).

    Article  CAS  Google Scholar 

  269. Liu, Z. et al. Drug delivery with carbon nanotubes for in vivo cancer treatment. Cancer Res. 68, 6652–6660 (2008).

    Article  CAS  Google Scholar 

  270. Gillies, E. R. & Fréchet, J. M. J. Dendrimers and dendritic polymers in drug delivery. Drug Discov. Today 10, 35–43 (2005).

    Article  CAS  Google Scholar 

  271. Liu, M. & Fréchet, J. M. Designing dendrimers for drug delivery. Pharm. Sci. Technol. Today 2, 393–401 (1999).

    Article  CAS  Google Scholar 

  272. Patri, A. K., Majoros, I. J. & Baker, J. R. Dendritic polymer macromolecular carriers for drug delivery. Curr. Opin. Chem. Biol. 6, 466–471 (2002).

    Article  CAS  Google Scholar 

  273. Leto, S. M. & Trusolino, L. Primary and acquired resistance to EGFR-targeted therapies in colorectal cancer: impact on future treatment strategies. J. Mol. Med. 92, 709–722 (2014).

    Article  CAS  Google Scholar 

  274. Sato, H. et al. Combined inhibition of MEK and PI3K pathways overcomes acquired resistance to EGFR-TKIs in non-small cell lung cancer. Cancer Sci. 109, 3183–3196 (2018).

    Article  CAS  Google Scholar 

  275. Wilson, F. H. et al. A functional landscape of resistance to ALK inhibition in lung cancer. Cancer Cell 27, 397–408 (2015).

    Article  CAS  Google Scholar 

  276. Masoud, V. & Pagès, G. Targeted therapies in breast cancer: New challenges to fight against resistance. World J. Clin. Oncol. 8, 120–134 (2017).

    Article  Google Scholar 

  277. Higgins, M. J. & Baselga, J. Targeted therapies for breast cancer. J. Clin. Investig. 121, 3797–3803 (2011).

    Article  CAS  Google Scholar 

  278. Villanueva, J. et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 18, 683–695 (2010).

    Article  CAS  Google Scholar 

  279. McCubrey, J. A. et al. Targeting survival cascades induced by activation of Ras/Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways for effective leukemia therapy. Leukemia 22, 708–722 (2008).

    Article  CAS  Google Scholar 

  280. McLornan, D., Percy, M. & McMullin, M. F. JAK2 V617F: a single mutation in the myeloproliferative group of disorders. Ulster Med. J. 75, 112–119 (2006).

    Google Scholar 

  281. Piloto, O. et al. Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood 109, 1643–1652 (2007).

    Article  CAS  Google Scholar 

  282. Chu, S. et al. Detection of BCR-ABL kinase mutations in CD34+ cells from chronic myelogenous leukemia patients in complete cytogenetic remission on imatinib mesylate treatment. Blood 105, 2093–2098 (2005).

    Article  CAS  Google Scholar 

  283. Jiang, Z.-M. et al. Crizotinib-loaded polymeric nanoparticles in lung cancer chemotherapy. Med. Oncol. 32, 193 (2015).

    Article  Google Scholar 

  284. Hazkani, I. et al. Can molecular profiling enhance radiotherapy? Impact of personalized targeted gold nanoparticles on radiosensitivity and imaging of adenoid cystic carcinoma. Theranostics 7, 3962–3971 (2017).

    Article  CAS  Google Scholar 

  285. Cortese, B., D’Amone, S. & Palamà, I. E. Wool-like hollow polymeric nanoparticles for CML chemo-combinatorial therapy. Pharmaceutics 10, 52 (2018).

    Article  Google Scholar 

  286. James, A. R. et al. Computational and mechanistic studies on the effect of galactoxyloglucan: Imatinib nanoconjugate in imatinib resistant K562 cells. Tumor Biol. 39, 1010428317695946 (2017).

    Article  Google Scholar 

  287. Theochari, I. et al. Drug nanocarriers for cancer chemotherapy based on microemulsions: The case of Vemurafenib analog PLX4720. Colloids Surf. B. Biointerfaces 154, 350–356 (2017).

    Article  CAS  Google Scholar 

  288. Savla, R., Garbuzenko, O. B., Chen, S., Rodriguez-Rodriguez, L. & Minko, T. Tumor-targeted responsive nanoparticle-based systems for magnetic resonance imaging and therapy. Pharm. Res. 31, 3487–3502 (2014).

    Article  CAS  Google Scholar 

  289. Liu, C. et al. Tumor-targeted nanoparticles deliver a vitamin D-based drug payload for the treatment of EGFR tyrosine kinase inhibitor-resistant lung cancer. Mol. Pharm. 15, 3216–3226 (2018).

    Article  CAS  Google Scholar 

  290. He, Y., Su, Z., Xue, L., Xu, H. & Zhang, C. Co-delivery of erlotinib and doxorubicin by pH-sensitive charge conversion nanocarrier for synergistic therapy. J. Control. Rel. 229, 80–92 (2016).

    Article  CAS  Google Scholar 

  291. Ali, A. A. A. et al. Erlotinib-conjugated iron oxide nanoparticles as a smart cancer-targeted theranostic probe for MRI. Sci. Rep. 6, 36650 (2016).

    Article  CAS  Google Scholar 

  292. Lam, A. T. N. et al. Colloidal gold nanoparticle conjugates of gefitinib. Colloids Surf. B. Biointerfaces 123, 61–67 (2014).

    Article  CAS  Google Scholar 

  293. Shende, P., Patil, S. & Gaud, R. S. A combinatorial approach of inclusion complexation and dendrimer synthesization for effective targeting EGFR-TK. Mater. Sci. Eng. C 76, 959–965 (2017).

    Article  CAS  Google Scholar 

  294. Liu, Y., Zhao, Y., Luo, H., Liu, F. & Wu, Y. Construction of EGFR peptide gefitinib/quantum dots long circulating polymeric liposomes for treatment and detection of nasopharyngeal carcinoma. Biochem. Biophys. Res. Commun. 490, 141–146 (2017).

    Article  CAS  Google Scholar 

  295. Zhou, Z. et al. Delayed sequential co-delivery of gefitinib and doxorubicin for targeted combination chemotherapy. Mol. Pharm. 14, 4551–4559 (2017).

    Article  CAS  Google Scholar 

  296. Wang, J. et al. iRGD-decorated polymeric nanoparticles for the efficient delivery of vandetanib to hepatocellular carcinoma: preparation and in vitro and in vivo evaluation. ACS Appl. Mater. Interfaces 8, 19228–19237 (2016).

    Article  CAS  Google Scholar 

  297. Kallus, S. et al. Nanoformulations of anticancer FGFR inhibitors with improved therapeutic index. Nanomedicine 14, 2632–2643 (2018).

    Article  CAS  Google Scholar 

  298. Simon, T. et al. Design of FLT3 inhibitor - gold nanoparticle conjugates as potential therapeutic agents for the treatment of acute myeloid leukemia. Nanoscale Res. Lett. 10, 466 (2015).

    Article  Google Scholar 

  299. Suarasan, S., Simon, T., Boca, S., Tomuleasa, C. & Astilean, S. Gelatin-coated gold nanoparticles as carriers of FLT3 Inhibitors for acute myeloid leukemia treatment. Chem. Biol. Drug Des. 87, 927–935 (2016).

    Article  CAS  Google Scholar 

  300. Segura-Ibarra, V. et al. Rapamycin nanoparticles localize in diseased lung vasculature and prevent pulmonary arterial hypertension. Int. J. Pharm. 524, 257–267 (2017).

    Article  CAS  Google Scholar 

  301. Rizwan, H., Mohanta, J., Si, S. & Pal, A. Gold nanoparticles reduce high glucose-induced oxidative-nitrosative stress regulated inflammation and apoptosis via tuberin-mTOR/NF-κB pathways in macrophages. Int. J. Nanomed. 12, 5841–5862 (2017).

    Article  CAS  Google Scholar 

  302. Li, H., Teng, Y., Sun, J. & Liu, J. Inhibition of hemangioma growth using polymer–lipid hybrid nanoparticles for delivery of rapamycin. Biomed. Pharmacother. 95, 875–884 (2017).

    Article  CAS  Google Scholar 

  303. Haeri, A., Osouli, M., Bayat, F., Alavi, S. & Dadashzadeh, S. Nanomedicine approaches for sirolimus delivery: a review of pharmaceutical properties and preclinical studies. Artif. Cell Nanomed. Biotechnol. 46, 1–14 (2018).

    Article  CAS  Google Scholar 

  304. Kasper, M. et al. Novel everolimus-loaded nanocarriers for topical treatment of murine experimental autoimmune uveoretinitis (EAU). Exp. Eye Res. 168, 49–56 (2018).

    Article  CAS  Google Scholar 

  305. Kabary, D. M. et al. Inhalable multi-compartmental phospholipid enveloped lipid core nanocomposites for localized mTOR inhibitor/herbal combined therapy of lung carcinoma. Eur. J. Pharm. Biopharm. 130, 152–164 (2018).

    Article  CAS  Google Scholar 

  306. Lunova, M. et al. Targeting the mTOR signaling pathway utilizing nanoparticles: a critical overview. Cancers 11, 82 (2019).

    Article  CAS  Google Scholar 

  307. Dasa, S. S. K. et al. Plectin-targeted liposomes enhance the therapeutic efficacy of a PARP inhibitor in the treatment of ovarian cancer. Theranostics 8, 2782–2798 (2018).

    Article  CAS  Google Scholar 

  308. Novohradsky, V., Zajac, J., Vrana, O., Kasparkova, J. & Brabec, V. Simultaneous delivery of olaparib and carboplatin in PEGylated liposomes imparts this drug combination hypersensitivity and selectivity for breast tumor cells. Oncotarget 9, 28456–28473 (2018).

    Article  Google Scholar 

  309. Du, C. et al. Epidermal growth factor receptor-targeting peptide nanoparticles simultaneously deliver gemcitabine and olaparib to treat pancreatic cancer with breast cancer 2 (BRCA2) mutation. ACS Nano 12, 10785–10796 (2018).

    Article  CAS  Google Scholar 

  310. Duan, W. & Liu, Y. Targeted and synergistic therapy for hepatocellular carcinoma: monosaccharide modified lipid nanoparticles for the co-delivery of doxorubicin and sorafenib. Drug Des. Devel. Ther. 12, 2149–2161 (2018).

    Article  CAS  Google Scholar 

  311. Ke, Y. & Xiang, C. Transferrin receptor-targeted HMSN for sorafenib delivery in refractory differentiated thyroid cancer therapy. Int. J. Nanomed. 13, 8339–8354 (2018).

    Article  CAS  Google Scholar 

  312. Tang, S. & Li, Y. Sorafenib-loaded ligand-functionalized polymer-lipid hybrid nanoparticles for enhanced therapeutic effect against liver cancer. J. Nanosci. Nanotechnol. 19, 6866–6871 (2019).

    Article  CAS  Google Scholar 

  313. Hamarat Şanlıer, Ş. et al. Development of ultrasound-triggered and magnetic-targeted nanobubble system for dual-drug delivery. J. Pharm. Sci. 108, 1272–1283 (2019).

    Article  Google Scholar 

  314. Chakravarty, R. et al. Hollow mesoporous silica nanoparticles for tumor vasculature targeting and PET image-guided drug delivery. Nanomedicine 10, 1233–1246 (2015).

    Article  CAS  Google Scholar 

  315. Shi, J.-F. et al. A combination of targeted sunitinib liposomes and targeted vinorelbine liposomes for treating invasive breast cancer. J. Biomed. Nanotechnol. 11, 1568–1582 (2015).

    Article  CAS  Google Scholar 

  316. Huo, M. et al. Tumor-targeted delivery of sunitinib base enhances vaccine therapy for advanced melanoma by remodeling the tumor microenvironment. J. Control. Rel. 245, 81–94 (2017).

    Article  CAS  Google Scholar 

  317. Saber, M. M., Bahrainian, S., Dinarvand, R. & Atyabi, F. Targeted drug delivery of Sunitinib Malate to tumor blood vessels by cRGD-chiotosan-gold nanoparticles. Int. J. Pharm. 517, 269–278 (2017).

    Article  CAS  Google Scholar 

  318. Nazari-Vanani, R., Azarpira, N., Heli, H., Karimian, K. & Sattarahmady, N. A novel self-nanoemulsifying formulation for sunitinib: Evaluation of anticancer efficacy. Colloids Surf. B. Biointerfaces 160, 65–72 (2017).

    Article  CAS  Google Scholar 

  319. Kim, H. et al. Combination of sunitinib and PD-L1 blockade enhances anticancer efficacy of TLR7/8 agonist-based nanovaccine. Mol. Pharm. 16, 1200–1210 (2019).

    Article  CAS  Google Scholar 

  320. Fang, S. et al. Dimeric camptothecin derived phospholipid assembled liposomes with high drug loading for cancer therapy. Colloids Surf. B. Biointerfaces 166, 235–244 (2018).

    Article  CAS  Google Scholar 

  321. Wu, Y. et al. Co-delivery of dual chemo-drugs with precisely controlled, high drug loading polymeric micelles for synergistic anti-cancer therapy. Biomater. Sci. 8, 949–959 (2020).

    Article  CAS  Google Scholar 

  322. Ménard, M. et al. Mesoporous silica templated-albumin nanoparticles with high doxorubicin payload for drug delivery assessed with a 3-D tumor cell model. Biochim. Biophys. Acta Biomembr. 1863, 332–341 (2019).

    Article  Google Scholar 

  323. Kang, S. J., Durairaj, C., Kompella, U. B., O’Brien, J. M. & Grossniklaus, H. E. Subconjunctival nanoparticle carboplatin in the treatment of murine retinoblastoma. Arch. Ophthalmol. 127, 1043–1047 (2009).

    Article  CAS  Google Scholar 

  324. You, Y., He, L., Ma, B. & Chen, T. High-drug-loading mesoporous silica nanorods with reduced toxicity for precise cancer therapy against nasopharyngeal carcinoma. Adv. Funct. Mater. 27, 1703313 (2017).

    Article  Google Scholar 

  325. Kather, J. N. et al. Predicting survival from colorectal cancer histology slides using deep learning: A retrospective multicenter study. PLoS Med. 16, e1002730 (2019).

    Article  Google Scholar 

  326. Bradbury, M. et al. Clinical translation of an ultrasmall inorganic optical-PET imaging nanoparticle probe. Sci. Transl. Med. 6, 260ra149 (2014).

    Google Scholar 

Download references

Acknowledgements

This work is supported, in part, by the NIH Director’s New Innovator Award (DP2-HD075698) and NIH grants R01-CA215719, R01-DK114321, R01-NS116353, R01-CA190642, UL1-TR002384, T32-CA062948 and P30-CA008748, the National Science Foundation (CAREER 1752506 and PHY 1545832), the American Cancer Society Research Scholar Grant (GC230452), the Pershing Square Sohn Cancer Research Alliance, the Breast Cancer Foundation, the Stand Up to Cancer Foundation, the Geoffrey Beene Cancer Research Center, the Expect Miracles Foundation - Financial Services Against Cancer, the Anna Fuller Fund, the Louis V. Gerstner Jr. Young Investigator’s Fund, the Frank A. Howard Scholars Program, the Honorable Tina Brozman Foundation for Ovarian Cancer Research, Cycle for Survival, the Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center, Mr. William H. Goodwin and Mrs. Alice Goodwin and the Commonwealth Foundation for Cancer Research, the Center for Experimental Therapeutics, the Imaging & Radiation Sciences Program and the Center for Molecular Imaging and Nanotechnology of Memorial Sloan Kettering Cancer. Y.S. is a Jack Klein fellow. The opinions, results, findings and/or interpretations of data contained therein are the responsibility of the contractor and do not necessarily represent the opinions, interpretations or policy of the state or, if funded with federal funds, the applicable federal funding agency. Special thanks to S. D’Angelo for helpful discussions.

Author information

Authors and Affiliations

Authors

Contributions

M.T.M., Y.S. and H.K. contributed equally to the literature research for the article, creating figures, writing the manuscript and contributing to the discussion of the content. D.A.H., N.R. and M.S. discussed the content of the article and wrote perspective content relevant to their respective fields of expertise. D.A.H. conceived the article. All authors contributed to reviewing and editing the manuscript.

Corresponding author

Correspondence to Daniel A. Heller.

Ethics declarations

Competing interests

D.A.H. is a co-founder and officer with equity interest in Goldilocks Therapeutics, Inc., LipidSense, Inc. and Nirova BioSense, Inc., as well as a member of the scientific advisory boards of Concarlo Holdings, LLC and Nanorobotics, Inc. M.S. is a full-time employee at AstraZeneca, has received research funds from Puma Biotechnology, AstraZeneca, Daiichi Sankyo, Immunomedics, Targimmune and Menarini Ricerche, is a co-founder of Medendi.org and is on the advisory board of Menarini Ricerche. N.R. is on the scientific advisory board (SAB) of Chugai, BeiGene, Fortress Biotech, Daiichi Sankyo, AstraZeneca, F-Prime, Zai Lab, Arvinas and Array BioPharma, and is a past SAB member of Millennium Takeda, Kadmon, Kura Oncology and Araxes. N.R. is also a consultant to Novartis Biomedical, Boehringer Ingelheim, Tarveda, Foresite Capital, Array BioPharma and Revolution Medicines, and, in recent years, has also consulted with Eli Lilly, Merrimack, Kura Oncology, Araxes and Kadmon. N.R. owns equity in BeiGene, Zai Lab, Fortress Biotech, Kura Oncology, Araxes, Kadmon and Effector. N.R. is a full-time employee at National Resilience Inc., collaborates with Plexxikon and receives research support from Chugai.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Manzari, M.T., Shamay, Y., Kiguchi, H. et al. Targeted drug delivery strategies for precision medicines. Nat Rev Mater 6, 351–370 (2021). https://doi.org/10.1038/s41578-020-00269-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41578-020-00269-6

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research