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.

  • Perspective
  • Published:

Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges

Abstract

Multiple nanotherapeutics have been approved for patients with cancer, but their effects on survival have been modest and, in some examples, less than those of other approved therapies. At the same time, the clinical successes achieved with immunotherapy have revolutionized the treatment of multiple advanced-stage malignancies. However, the majority of patients do not benefit from the currently available immunotherapies and many develop immune-related adverse events. By contrast, nanomedicines can reduce — but do not eliminate — the risk of certain life-threatening toxicities. Thus, the combination of these therapeutic classes is of intense research interest. The tumour microenvironment (TME) is a major cause of the failure of both nanomedicines and immunotherapies that not only limits delivery, but also can compromise efficacy, even when agents accumulate in the TME. Coincidentally, the same TME features that impair nanomedicine delivery can also cause immunosuppression. In this Perspective, we describe TME normalization strategies that have the potential to simultaneously promote the delivery of nanomedicines and reduce immunosuppression in the TME. Then, we discuss the potential of a combined nanomedicine-based TME normalization and immunotherapeutic strategy designed to overcome each step of the cancer-immunity cycle and propose a broadly applicable ‘minimal combination’ of therapies designed to increase the number of patients with cancer who are able to benefit from immunotherapy.

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: Cancer-immunity TME phenotypes affecting responsiveness to immunotherapy18.
Fig. 2: How nanomedicines can be used to perpetuate the cancer-immunity cycle.
Fig. 3: Normalizing the TME to increase the penetration of combination therapies.

Similar content being viewed by others

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Haslam, A. & Prasad, V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw. Open. 2, e192535–e192535 (2019).

    PubMed  PubMed Central  Google Scholar 

  3. Postow, M. A., Sidlow, R. & Hellmann, M. D. Immune-related adverse events associated with immune checkpoint blockade. N. Engl. J. Med. 378, 158–168 (2018).

    CAS  PubMed  Google Scholar 

  4. Riley, R. S., June, C. H., Langer, R. & Mitchell, M. J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug. Discovery 18, 175–196 (2019).

    CAS  PubMed  Google Scholar 

  5. Goldberg, M. S. et al. Improving cancer immunotherapy through nanotechnology. Nat. Rev. Cancer 19, 587–602 (2019).

    CAS  PubMed  Google Scholar 

  6. Schmid, P. et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379, 2108–2121 (2018).

    CAS  PubMed  Google Scholar 

  7. Jain, R. K. Physiological barriers to delivery of monoclonal antibodies and other macromolecules in tumors. Cancer Res. 50, 814s–819s (1990).

    CAS  PubMed  Google Scholar 

  8. Chauhan, V. P. & Jain, R. K. Strategies for advancing cancer nanomedicine. Nat. Mater. 12, 958–962 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Jain, R. K. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 26, 605–622 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Martin, J. D., Seano, G. & Jain, R. K. Normalizing function of tumor vessels: progress, opportunities and challenges. Annu. Rev. Physiol. 81, 505–534 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Fukumura, D., Kloepper, J., Amoozgar, Z., Duda, D. G. & Jain, R. K. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 15, 325–340 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee, H. et al. 64Cu-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).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ramanathan, R. K. et al. Correlation between ferumoxytol uptake in tumor lesions by MRI and response to nanoliposomal irinotecan in patients with advanced solid tumors: a pilot study. Clin. Cancer Res. 23, 3638–3648 (2017).

    CAS  PubMed  Google Scholar 

  14. Stylianopoulos, T., Munn, L. L. & Jain, R. K. Reengineering the physical microenvironment of tumors to improve drug delivery and efficacy: from mathematical modeling to bench to bedside. Trends Cancer 4, 292–319 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Chauhan, V. P., Stylianopoulos, T., Boucher, Y. & Jain, R. K. Delivery of molecular and nanoscale medicine to tumors: transport barriers and strategies. Annu. Rev. Chem. Biomol. Eng. 2, 281–298 (2011).

    CAS  PubMed  Google Scholar 

  16. Baine, M. K. et al. Characterization of tumor infiltrating lymphocytes in paired primary and metastatic renal cell carcinoma specimens. Oncotarget 6, 24990–25002 (2015).

    PubMed  PubMed Central  Google Scholar 

  17. Müller, P. et al. Metastatic spread in patients with non-small cell lung cancer is associated with a reduced density of tumor-infiltrating T cells. Cancer Immunol. Immunother. 65, 1–11 (2016).

    PubMed  Google Scholar 

  18. Chen, D. S. & Mellman, I. Elements of cancer immunity and the cancer–immune set point. Nature 541, 321–330 (2017).

    CAS  PubMed  Google Scholar 

  19. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Veglia, F. & Gabrilovich, D. I. Dendritic cells in cancer: the role revisited. Curr. Opin. immunol. 45, 43–51 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Facciabene, A. et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 475, 226–230 (2011).

    CAS  PubMed  Google Scholar 

  22. Togashi, Y., Shitara, K. & Nishikawa, H. Regulatory T cells in cancer immunosuppression — implications for anticancer therapy. Nat. Rev. Clin. Oncol. 16, 356–371 (2019).

    CAS  PubMed  Google Scholar 

  23. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen, I. X. et al. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc. Natl Acad. Sci. USA 116, 4558–4566 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chauhan, V. P. et al. Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy. Proc. Natl Acad. Sci. USA 116, 10674–10680 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Costa, A. et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 33, 463–479. e410 (2018).

    CAS  PubMed  Google Scholar 

  27. Rytelewski, M. et al. Merger of dynamic two-photon and phosphorescence lifetime microscopy reveals dependence of lymphocyte motility on oxygen in solid and hematological tumors. J. Immunother. Cancer 7, 78 (2019).

    PubMed  PubMed Central  Google Scholar 

  28. Hatfield, S. M. et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Transl. Med. 7, 277ra230 (2015).

    Google Scholar 

  29. Maenhout, S. K., Thielemans, K. & Aerts, J. L. Location, location, location: functional and phenotypic heterogeneity between tumor-infiltrating and non-infiltrating myeloid-derived suppressor cells. Oncoimmunology 3, e956579 (2014).

    PubMed  PubMed Central  Google Scholar 

  30. Voron, T. et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 212, 139–148 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Palazon, A. et al. An HIF-1α/VEGF-A axis in cytotoxic T cells regulates tumor progression. Cancer Cell 32, 669–683 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Wallin, J. J. et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 7, 12624 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Noman, M. Z. et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 211, 781–790 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Noman, M. Z. et al. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am. J. Physiol. Cell Physiol. 309, C569–C579 (2015).

    PubMed  PubMed Central  Google Scholar 

  35. Calcinotto, A. et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 72, 2746–2756 (2012).

    CAS  PubMed  Google Scholar 

  36. Kuczek, D. E. et al. Collagen density regulates the activity of tumor-infiltrating T cells. J. Immunother. Cancer 7, 68 (2019).

    PubMed  PubMed Central  Google Scholar 

  37. Mazzone, M. & Bergers, G. Regulation of blood and lymphatic vessels by immune cells in tumors and metastasis. Annu. Rev. Physiol. 81, 535–560 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Munn, L. L. & Jain, R. K. Vascular regulation of anti-tumor immunity. Science 365, 544–555 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Huang, Y. et al. Improving immune–vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 18, 195–203 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Jain, R. K. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat. Med. 7, 987–989 (2001).

    CAS  PubMed  Google Scholar 

  41. Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58–62 (2005).

    CAS  PubMed  Google Scholar 

  42. Goel, S. et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol. Rev. 91, 1071–1121 (2011).

    CAS  PubMed  Google Scholar 

  43. 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).

    CAS  PubMed  Google Scholar 

  44. Jain, R. K. Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J. Clin. Oncol. 31, 2205–2218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Viallard, C. & Larrivée, B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 20, 409–426 (2017).

    CAS  PubMed  Google Scholar 

  46. Jain, R. K., Martin, J. D. & Stylianopoulos, T. The role of mechanical forces in tumor growth and therapy. Annu. Rev. Biomed. Eng. 16, 321–346 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Whatcott, C. J., Han, H. & Von Hoff, D. D. Orchestrating the tumor microenvironment to improve survival for patients with pancreatic cancer: normalization, not destruction. Cancer J. 21, 299–306 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Stapleton, S., Allen, C., Pintilie, M. & Jaffray, D. A. Tumor perfusion imaging predicts the intra-tumoral accumulation of liposomes. J. Control. Rel. 172, 351–357 (2013).

    CAS  Google Scholar 

  49. Toy, R. et al. Multimodal in vivo imaging exposes the voyage of nanoparticles in tumor microcirculation. ACS Nano 7, 3118–3129 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. 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  PubMed  PubMed Central  Google Scholar 

  51. Martin, J. D. et al. Dexamethasone increases cisplatin-loaded nanocarrier delivery and efficacy in metastatic breast cancer by normalizing the tumor microenvironment. ACS Nano 13, 6396–6408 (2019).

    CAS  PubMed  Google Scholar 

  52. Stylianopoulos, T. & Jain, R. K. Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc. Natl Acad. Sci. USA 110, 18632–18637 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Jayson, G. C., Kerbel, R., Ellis, L. M. & Harris, A. L. Antiangiogenic therapy in oncology: current status and future directions. Lancet 388, 518–529 (2016).

    CAS  PubMed  Google Scholar 

  54. Chauhan, V. P. et al. Compression of pancreatic tumor blood vessels by hyaluronan is caused by solid stress and not interstitial fluid pressure. Cancer Cell 26, 14–15 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Stylianopoulos, T. et al. Coevolution of solid stress and interstitial fluid pressure in tumors during progression: implications for vascular collapse. Cancer Res. 73, 3833–3841 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Stylianopoulos, T. et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc. Natl Acad. Sci. USA 109, 15101–15108 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Papageorgis, P. et al. Tranilast-induced stress alleviation in solid tumors improves the efficacy of chemo-and nanotherapeutics in a size-independent manner. Sci. Rep. 7, 46140 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Netti, P. A., Berk, D. A., Swartz, M. A., Grodzinsky, A. J. & Jain, R. K. Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res. 60, 2497–2503 (2000).

    CAS  PubMed  Google Scholar 

  59. McKee, T. D. et al. Degradation of fibrillar collagen in a human melanoma xenograft improves the efficacy of an oncolytic herpes simplex virus vector. Cancer Res. 66, 2509–2513 (2006).

    CAS  PubMed  Google Scholar 

  60. Brown, E. et al. Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation. Nat. Med. 9, 796–800 (2003).

    CAS  PubMed  Google Scholar 

  61. Mok, W., Boucher, Y. & Jain, R. K. Matrix metalloproteinases-1 and -8 improve the distribution and efficacy of an oncolytic virus. Cancer Res. 67, 10664–10668 (2007).

    CAS  PubMed  Google Scholar 

  62. Sherman, M. H. et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 159, 80–93 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Sheridan, C. Pancreatic cancer provides test bed for first mechanotherapeutics. Nat. Biotechnol. 37, 829 (2019).

    CAS  PubMed  Google Scholar 

  64. Panagi, M. et al. TGF-β inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity. Theranostics 10, 1910–1922 (2020).

    PubMed  PubMed Central  Google Scholar 

  65. Incio, J. et al. Metformin reduces desmoplasia in pancreatic cancer by reprogramming stellate cells and tumor-associated macrophages. PLoS One 10, e0141392 (2015).

    PubMed  PubMed Central  Google Scholar 

  66. Polydorou, C., Mpekris, F., Papageorgis, P., Voutouri, C. & Stylianopoulos, T. Pirfenidone normalizes the tumor microenvironment to improve chemotherapy. Oncotarget 8, 24506–24517 (2017).

    PubMed  PubMed Central  Google Scholar 

  67. Zhao, Y. et al. Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma. Proc. Natl Acad. Sci. USA 116, 2210–2219 (2019).

    PubMed  PubMed Central  Google Scholar 

  68. Liu, H. et al. Use of angiotensin system inhibitors is associated with immune activation and longer survival in nonmetastatic pancreatic ductal adenocarcinoma. Clin. Cancer Res. 23, 5959–5969 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Geller, A. et al. Angiotensin system inhibitors during induction chemotherapy for esophageal adenocarcinoma: analysis of survival. J. Clin. Oncol. 36, e16066 (2018).

    Google Scholar 

  70. Pinter, M. et al. Use of inhibitors of the renin–angiotensin system is associated with longer survival in patients with hepatocellular carcinoma. United Eur. Gastroenterol. J. 5, 987–996 (2017).

    CAS  Google Scholar 

  71. Pinter, M. & Jain, R. K. Targeting the renin-angiotensin system to improve cancer treatment: implications for immunotherapy. Sci. Transl. Med. 9, eaan5616 (2017).

    PubMed  PubMed Central  Google Scholar 

  72. Levin, V. A., Chan, J., Datta, M., Yee, J. L. & Jain, R. K. Effect of angiotensin system inhibitors on survival in newly diagnosed glioma patients and recurrent glioblastoma patients receiving chemotherapy and/or bevacizumab. J. Neurooncol. 134, 325–330 (2017).

    CAS  PubMed  Google Scholar 

  73. Cleary, J. M. et al. FOLFOX plus ziv-aflibercept or placebo in first-line metastatic esophagogastric adenocarcinoma: a double-blind, randomized, multicenter phase 2 trial. Cancer 125, 2213–2221 (2019).

    CAS  PubMed  Google Scholar 

  74. Chauhan, V. P. et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat. Commun. 4, 2516 (2013).

    PubMed  Google Scholar 

  75. Diop-Frimpong, B., Chauhan, V. P., Krane, S., Boucher, Y. & Jain, R. K. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc. Natl Acad. Sci. USA 108, 2909–2914 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Murphy, J. E. et al. A phase II study of neoadjuvant FOLFIRINOX in combination with losartan followed by chemoradiotherapy in locally advanced pancreatic cancer: R0 resection rate and clinical outcomes. JAMA Oncol. 5, 1020–1027 (2019).

    PubMed  PubMed Central  Google Scholar 

  77. Zheng, X. et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J. Clin. Invest. 128, 2104–2115 (2018).

    PubMed  PubMed Central  Google Scholar 

  78. Allen, E. et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci. Transl. Med. 9, eaak9679 (2017).

    PubMed  PubMed Central  Google Scholar 

  79. Schmittnaegel, M. et al. Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade. Sci. Transl. Med. 9, eaak9670 (2017).

    PubMed  Google Scholar 

  80. Shigeta, K. et al. Dual programmed death receptor-1 and vascular endothelial growth factor receptor-2 blockade promotes vascular normalization and enhances antitumor immune responses in hepatocellular carcinoma. Hepatology https://doi.org/10.1002/hep.30889 (2019).

    Article  PubMed  Google Scholar 

  81. Huang, Y. et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl Acad. Sci. USA 109, 17561–17566 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Shrimali, R. K. et al. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res. 70, 6171–6180 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Socinski, M. A. et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 378, 2288–2301 (2018).

    CAS  PubMed  Google Scholar 

  84. Motzer, R. J. et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 380, 1103–1115 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Rini, B. I. et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 380, 1116–1127 (2019).

    CAS  PubMed  Google Scholar 

  86. Heist, R. S. et al. Improved tumor vascularization after anti-VEGF therapy with carboplatin and nab-paclitaxel associates with survival in lung cancer. Proc. Natl Acad. Sci. USA 112, 1547–1552 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Tian, L. et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 544, 250–254 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Chakravarthy, A., Khan, L., Bensler, N. P., Bose, P. & De Carvalho, D. D. TGF-β-associated extracellular matrix genes link cancer-associated fibroblasts to immune evasion and immunotherapy failure. Nat. Commun. 9, 4692 (2018).

    PubMed  PubMed Central  Google Scholar 

  89. Tauriello, D. V. et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 554, 538–543 (2018).

    CAS  PubMed  Google Scholar 

  90. Vanpouille-Box, C. et al. TGFβ is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 75, 2232–2242 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Medjebar, S. et al. Angiotensin-converting enzyme inhibitor prescription is associated with decreased progression-free survival (PFS) and overall survival (OS) in patients with lung cancers treated with PD-1/PD-L1 immune checkpoint blockers. J. Clin. Oncol. 37, e20512 (2019).

    Google Scholar 

  92. Regan, D. P. et al. The angiotensin receptor blocker losartan suppresses growth of pulmonary metastases via AT1R-independent inhibition of CCR2 signaling and monocyte recruitment. J. Immunol. 202, 3087–3102 (2019).

    CAS  PubMed  Google Scholar 

  93. Newick, K., O'Brien, S., Moon, E. & Albelda, S. M. CAR T cell therapy for solid tumors. Annu. Rev. Med. 68, 139–152 (2017).

    CAS  PubMed  Google Scholar 

  94. Segal, N. H. et al. Epitope landscape in breast and colorectal cancer. Cancer Res. 68, 889–892 (2008).

    CAS  PubMed  Google Scholar 

  95. Stevanovic´, S. et al. Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer. Science 356, 200–205 (2017).

    PubMed  PubMed Central  Google Scholar 

  96. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Hu, Z., Ott, P. A. & Wu, C. J. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat. Rev. Immunol. 18, 168–182 (2018).

    CAS  PubMed  Google Scholar 

  98. Soliman, H. H. nab-Paclitaxel as a potential partner with checkpoint inhibitors in solid tumors. Onco Targets Ther. 10, 101 (2017).

    CAS  PubMed  Google Scholar 

  99. Schuler, G. & Steinman, R. Dendritic cells as adjuvants for immune-mediated resistance to tumors. J. Exp. Med. 186, 1183–1187 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Albert, M. L., Sauter, B. & Bhardwaj, N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature 392, 86 (1998).

    CAS  PubMed  Google Scholar 

  101. Mpekris, F., Baish, J. W., Stylianopoulos, T. & Jain, R. K. Role of vascular normalization in benefit from metronomic chemotherapy. Proc. Natl Acad. Sci. USA 114, 1994–1999 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Voorwerk, L. et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat. Med. 25, 920–928 (2019).

    CAS  PubMed  Google Scholar 

  103. Zsiros, E. et al. A phase II trial of pembrolizumab in combination with bevacizumab and oral metronomic cyclophosphamide for recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer. Gynecol. Oncol. 154, 23 (2019).

    Google Scholar 

  104. Matulonis, U. et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Ann. Oncol. 30, 1080–1087 (2019).

    CAS  PubMed  Google Scholar 

  105. Min, Y. et al. Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat. Nanotechnol. 12, 877–882 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Peng, J. et al. Photosensitizer micelles together with IDO inhibitor enhance cancer photothermal therapy and immunotherapy. Adv. Sci. 5, 1700891 (2018).

    Google Scholar 

  107. Chen, Q. et al. Nanoparticle-enhanced radiotherapy to trigger robust cancer immunotherapy. Adv. Mater. 31, 1802228 (2019).

    Google Scholar 

  108. Chen, Z. et al. Bioinspired hybrid protein oxygen nanocarrier amplified photodynamic therapy for eliciting anti-tumor immunity and abscopal effect. ACS Nano 12, 8633–8645 (2018).

    CAS  PubMed  Google Scholar 

  109. He, C. et al. Core-shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. Nat. Commun. 7, 12499 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Dewan, M. Z. et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 15, 5379–5388 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Demaria, S. et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 11, 728–734 (2005).

    CAS  PubMed  Google Scholar 

  112. Vanpouille-Box, C. et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 8, 15618 (2017).

    PubMed  PubMed Central  Google Scholar 

  113. Twyman-Saint Victor, C. et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520, 373–377 (2015).

    CAS  PubMed  Google Scholar 

  114. Bonvalot, S. et al. NBTXR3, a first-in-class radioenhancer hafnium oxide nanoparticle, plus radiotherapy versus radiotherapy alone in patients with locally advanced soft-tissue sarcoma (Act. In. Sarc): a multicentre, phase 2–3, randomised, controlled trial. Lancet Oncol. 20, 1148–1159 (2019).

    CAS  PubMed  Google Scholar 

  115. Liu, J. et al. TGF-β blockade improves the distribution and efficacy of therapeutics in breast carcinoma by normalizing the tumor stroma. Proc. Natl Acad. Sci. USA 109, 16618–16623 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Batchelor, T. T. et al. Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation. Proc. Natl Acad. Sci. USA 110, 19059–19064 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Stapleton, S. et al. Radiation and heat improve the delivery and efficacy of nanotherapeutics by modulating intra-tumoral fluid dynamics. ACS Nano 12, 7583–7600 (2018).

    CAS  PubMed  Google Scholar 

  118. Miller, M. A. et al. Radiation therapy primes tumors for nanotherapeutic delivery via macrophage-mediated vascular bursts. Sci. Transl. Med. 9, eaal0225 (2017).

    PubMed  PubMed Central  Google Scholar 

  119. Shamay, Y. et al. P-selectin is a nanotherapeutic delivery target in the tumor microenvironment. Sci. Transl. Med. 8, 345ra387 (2016).

    Google Scholar 

  120. Irvine, D. J., Swartz, M. A. & Szeto, G. L. Engineering synthetic vaccines using cues from natural immunity. Nat. Mater. 12, 978–990 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Ramanjulu, J. M. et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature 564, 439–443 (2018).

    CAS  PubMed  Google Scholar 

  122. Milhem, M. M. et al. Phase 1b/2, open label, multicenter, study of the combination of SD-101 and pembrolizumab in patients with advanced melanoma who are naïve to anti-PD-1 therapy. J. Clin. Oncol. 37, 9534 (2019).

    Google Scholar 

  123. Meric-Bernstam, F. et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas. J. Clin. Oncol. 37, 2507 (2019).

    Google Scholar 

  124. Larkin, B. et al. Cutting edge: activation of STING in T cells induces type I IFN responses and cell death. J. Immunol. 199, 397–402 (2017).

    CAS  PubMed  Google Scholar 

  125. Nuhn, L. et al. Nanoparticle-conjugate TLR7/8 agonist localized immunotherapy provokes safe antitumoral responses. Adv. Mater. 30, 1803397 (2018).

    Google Scholar 

  126. Wilson, D. R. et al. Biodegradable STING agonist nanoparticles for enhanced cancer immunotherapy. Nanomedicine 14, 237–246 (2018).

    CAS  PubMed  Google Scholar 

  127. Koshy, S. T., Cheung, A. S., Gu, L., Graveline, A. R. & Mooney, D. J. Liposomal delivery enhances immune activation by STING agonists for cancer immunotherapy. Adv. Biosyst. 1, 1600013 (2017).

    PubMed  PubMed Central  Google Scholar 

  128. Momin, N. et al. Anchoring of intratumorally administered cytokines to collagen safely potentiates systemic cancer immunotherapy. Sci. Transl. Med. 11, eaaw2614 (2019).

    PubMed  PubMed Central  Google Scholar 

  129. Ishihara, J. et al. Targeted antibody and cytokine cancer immunotherapies through collagen affinity. Sci. Transl. Med. 11, eaau3259 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Irvine, D. J., Hanson, M. C., Rakhra, K. & Tokatlian, T. Synthetic nanoparticles for vaccines and immunotherapy. Chem. Rev. 115, 11109–11146 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547, 222–226 (2017).

    CAS  PubMed  Google Scholar 

  132. Ott, P. A. et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547, 217–221 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Hailemichael, Y. et al. Persistent antigen at vaccination sites induces tumor-specific CD8+ T cell sequestration, dysfunction and deletion. Nat. Med. 19, 465–472 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Rosalia, R. A. et al. CD40-targeted dendritic cell delivery of PLGA-nanoparticle vaccines induce potent anti-tumor responses. Biomaterials 40, 88–97 (2015).

    CAS  PubMed  Google Scholar 

  135. Yuba, E. et al. Dextran derivative-based pH-sensitive liposomes for cancer immunotherapy. Biomaterials 35, 3091–3101 (2014).

    CAS  PubMed  Google Scholar 

  136. 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 (2017).

    CAS  PubMed  Google Scholar 

  137. Ali, O. A., Huebsch, N., Cao, L., Dranoff, G. & Mooney, D. J. Infection-mimicking materials to program dendritic cells in situ. Nat. Mater. 8, 151–158 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Fan, Y. et al. Immunogenic cell death amplified by co-localized adjuvant delivery for cancer immunotherapy. Nano Lett. 17, 7387–7393 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Carreno, B. M. et al. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 348, 803–808 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Heil, F. et al. Species-specific recognition of single-stranded RNA via Toll-like receptor 7 and 8. Science 303, 1526–1529 (2004).

    CAS  PubMed  Google Scholar 

  141. Kranz, L. M. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–400 (2016).

    PubMed  Google Scholar 

  142. Kanchan, V. & Panda, A. K. Interactions of antigen-loaded polylactide particles with macrophages and their correlation with the immune response. Biomaterials 28, 5344–5357 (2007).

    CAS  PubMed  Google Scholar 

  143. Li, X., Sloat, B. R., Yanasarn, N. & Cui, Z. Relationship between the size of nanoparticles and their adjuvant activity: data from a study with an improved experimental design. Eur. J. Pharm. Biopharm. 78, 107–116 (2011).

    CAS  PubMed  Google Scholar 

  144. Tseng, Y.-C., Xu, Z., Guley, K., Yuan, H. & Huang, L. Lipid–calcium phosphate nanoparticles for delivery to the lymphatic system and SPECT/CT imaging of lymph node metastases. Biomaterials 35, 4688–4698 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Jewell, C. M., López, S. C. B. & Irvine, D. J. In situ engineering of the lymph node microenvironment via intranodal injection of adjuvant-releasing polymer particles. Proc. Natl Acad. Sci. USA 108, 15745–15750 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Oussoren, C. & Storm, G. Liposomes to target the lymphatics by subcutaneous administration. Adv. Drug. Deliv. Rev. 50, 143–156 (2001).

    CAS  PubMed  Google Scholar 

  147. Tokatlian, T. et al. Innate immune recognition of glycans targets HIV nanoparticle immunogens to germinal centers. Science 363, 649–654 (2019).

    CAS  PubMed  Google Scholar 

  148. Luo, M. et al. A STING-activating nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 12, 648–654 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Miao et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat. Biotechnol. 37, 1174–1185 (2019).

    CAS  PubMed  Google Scholar 

  150. Wolchok, J. D. et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369, 122–133 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Zhang, Y., Li, N., Suh, H. & Irvine, D. J. Nanoparticle anchoring targets immune agonists to tumors enabling anti-cancer immunity without systemic toxicity. Nat. Commun. 9, 6 (2018).

    PubMed  PubMed Central  Google Scholar 

  152. Bentebibel, S.-E. et al. A first-in-human study and biomarker analysis of NKTR-214, a novel IL2Rβγ-biased cytokine, in patients with advanced or metastatic solid tumors. Cancer Discov. 9, 711–721 (2019).

    PubMed  Google Scholar 

  153. Ma, L. et al. Enhanced CAR–T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science 365, 162–168 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Smith, T. T. et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat. Nanotechnol. 12, 813–820 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Feig, C. et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl Acad. Sci. USA 110, 20212–20217 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Harlin, H. et al. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 69, 3077–3085 (2009).

    CAS  PubMed  Google Scholar 

  157. Rolny, C. et al. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF. Cancer Cell 19, 31–44 (2011).

    CAS  PubMed  Google Scholar 

  158. Taggart, D. et al. Anti-PD-1/anti-CTLA-4 efficacy in melanoma brain metastases depends on extracranial disease and augmentation of CD8+ T cell trafficking. Proc. Natl Acad. Sci. USA 115, E1540–E1549 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Melder, R. J. et al. During angiogenesis, vascular endothelial growth factor and basic fibroblast growth factor regulate natural killer cell adhesion to tumor endothelium. Nat. Med. 2, 992–997 (1996).

    CAS  PubMed  Google Scholar 

  160. Hamzah, J. et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 453, 410–414 (2008).

    CAS  PubMed  Google Scholar 

  161. Pinter, M., Kwanten, W. J. & Jain, R. K. Renin-angiotensin system inhibitors to mitigate cancer treatment-related adverse events. Clin. Cancer Res. 24, 3803–3812 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Zhu, Y. et al. Inhibition of tumor-promoting stroma to enforce subsequently targeting AT1R on tumor cells by pathological inspired micelles. Biomaterials 161, 33–46 (2018).

    CAS  PubMed  Google Scholar 

  163. Golder, M. R. et al. Reduction of liver fibrosis by rationally designed macromolecular telmisartan prodrugs. Nat. Biomed. Eng. 2, 822–830 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Han, X. et al. Reversal of pancreatic desmoplasia by re-educating stellate cells with a tumour microenvironment-activated nanosystem. Nat. Commun. 9, 3390 (2018).

    PubMed  PubMed Central  Google Scholar 

  165. 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).

    CAS  Google Scholar 

  166. Topp, M. S. et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 16, 57–66 (2015).

    CAS  PubMed  Google Scholar 

  167. Heiss, M. M. et al. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: results of a prospective randomized phase II/III trial. Int. J. Cancer 127, 2209–2221 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 9, eaaa0984 (2017).

    PubMed  PubMed Central  Google Scholar 

  169. Choi, B. D. et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat. Biotechnol. 37, 1049–1058 (2019).

    CAS  PubMed  Google Scholar 

  170. Yuan, H. et al. Multivalent bi-specific nanobioconjugate engager for targeted cancer immunotherapy. Nat. Nanotechnol. 12, 763–769 (2017).

    CAS  PubMed  Google Scholar 

  171. Siemens, D. R. et al. Hypoxia increases tumor cell shedding of MHC class I chain-related molecule: role of nitric oxide. Cancer Res. 68, 4746–4753 (2008).

    CAS  PubMed  Google Scholar 

  172. Sethumadhavan, S. et al. Hypoxia and hypoxia-inducible factor (HIF) downregulate antigen-presenting MHC class I molecules limiting tumor cell recognition by T cells. PLoS One 12, e0187314 (2017).

    PubMed  PubMed Central  Google Scholar 

  173. Long, G. V. et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 20, 1083–1097 (2019).

    CAS  PubMed  Google Scholar 

  174. Krähenbühl, L. et al. A longitudinal analysis of IDO and PDL1 expression during immune- or targeted therapy in advanced melanoma. Neoplasia 20, 218–225 (2018).

    PubMed  PubMed Central  Google Scholar 

  175. Cheng, K. et al. Sequentially responsive therapeutic peptide assembling nanoparticles for dual-targeted cancer immunotherapy. Nano Lett. 18, 3250–3258 (2018).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  177. Zheng, Y., Tang, L., Mabardi, L., Kumari, S. & Irvine, D. J. Enhancing adoptive cell therapy of cancer through targeted delivery of small-molecule immunomodulators to internalizing or noninternalizing receptors. ACS Nano 11, 3089–3100 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Xu, Z., Wang, Y., Zhang, L. & Huang, L. Nanoparticle-delivered transforming growth factor-β siRNA enhances vaccination against advanced melanoma by modifying tumor microenvironment. ACS Nano 8, 3636–3645 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Stephan, M. T., Moon, J. J., Um, S. H., Bershteyn, A. & Irvine, D. J. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat. Med. 16, 1035–1041 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Tang, L. et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 36, 707–716 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Siriwon, N. et al. CAR-T cells surface-engineered with drug-encapsulated nanoparticles can ameliorate intratumoral T-cell hypofunction. Cancer Immunol. Res. 6, 812–824 (2018).

    CAS  PubMed  Google Scholar 

  182. Terry, S. et al. Acquisition of tumor cell phenotypic diversity along the EMT spectrum under hypoxic pressure: consequences on susceptibility to cell-mediated cytotoxicity. Oncoimmunology 6, e1271858 (2017).

    PubMed  PubMed Central  Google Scholar 

  183. Karasaki, T. et al. An immunogram for the cancer-immunity cycle: towards personalized immunotherapy of lung cancer. J. Thorac. Oncol. 12, 791–803 (2017).

    PubMed  Google Scholar 

  184. Nam, J. et al. Cancer nanomedicine for combination cancer immunotherapy. Nat. Rev. Mater. 4, 398–414 (2019).

    Google Scholar 

  185. Kinoh, H. et al. Nanomedicines eradicating cancer stem-like cells in vivo by pH-triggered intracellular cooperative action of loaded drugs. ACS Nano 10, 5643–5655 (2016).

    CAS  PubMed  Google Scholar 

  186. Cabral, H., Miyata, K., Osada, K. & Kataoka, K. Block copolymer micelles in nanomedicine applications. Chem. Rev. 118, 6844–6892 (2018).

    CAS  PubMed  Google Scholar 

  187. Duan, X. et al. Immunostimulatory nanomedicines synergize with checkpoint blockade immunotherapy to eradicate colorectal tumors. Nat. Commun. 10, 1899 (2019).

    PubMed  PubMed Central  Google Scholar 

  188. Yang, G. et al. Hollow MnO2 as a tumor-microenvironment-responsive biodegradable nano-platform for combination therapy favoring antitumor immune responses. Nat. Commun. 8, 902 (2017).

    PubMed  PubMed Central  Google Scholar 

  189. Moynihan, K. D. et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat. Med. 22, 1402–1410 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Chiang, C.-S. et al. Combination of fucoidan-based magnetic nanoparticles and immunomodulators enhances tumour-localized immunotherapy. Nat. Nanotechnol. 13, 746–754 (2018).

    CAS  PubMed  Google Scholar 

  191. Zhang, F. et al. Nanoparticles that reshape the tumor milieu create a therapeutic window for effective T cell therapy in solid malignancies. Cancer Res. 78, 3718–3730 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Miura, Y. et al. Cyclic RGD-linked polymeric micelles for targeted delivery of platinum anticancer drugs to glioblastoma through the blood–brain tumor barrier. ACS Nano 7, 8583–8592 (2013).

    CAS  PubMed  Google Scholar 

  193. Quader, S. et al. cRGD peptide-installed epirubicin-loaded polymeric micelles for effective targeted therapy against brain tumors. J. Control. Release 258, 56–66 (2017).

    CAS  PubMed  Google Scholar 

  194. Wong, C. et al. Multistage nanoparticle delivery system for deep penetration into tumor tissue. Proc. Natl Acad. Sci. USA 108, 2426–2431 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Mura, S., Nicolas, J. & Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 12, 991–1003 (2013).

    CAS  PubMed  Google Scholar 

  196. Arvanitis, C. D. et al. Mechanisms of enhanced drug delivery in brain metastases with focused ultrasound-induced blood–tumor barrier disruption. Proc. Natl Acad. Sci. USA 115, E8717–E8726 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Mi, P. et al. A pH-activatable nanoparticle with signal-amplification capabilities for non-invasive imaging of tumour malignancy. Nat. Nanotechnol. 11, 724–730 (2016).

    CAS  PubMed  Google Scholar 

  198. Kulkarni, A. et al. Reporter nanoparticle that monitors its anticancer efficacy in real time. Proc. Natl Acad. Sci. USA 113, E2104–E2113 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Uchida, S. et al. Systemic delivery of messenger RNA for the treatment of pancreatic cancer using polyplex nanomicelles with a cholesterol moiety. Biomaterials 82, 221–228 (2016).

    CAS  PubMed  Google Scholar 

  200. Wang, H.-X. et al. Nonviral gene editing via CRISPR/Cas9 delivery by membrane-disruptive and endosomolytic helical polypeptide. Proc. Natl Acad. Sci. USA 115, 4903–4908 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Charlesworth, C. T. et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat. Med. 25, 249–254 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Glass, Z., Lee, M., Li, Y. & Xu, Q. Engineering the delivery system for CRISPR-based genome editing. Trends Biotechnol. 36, 173–185 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Song, W. et al. Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap. Nat. Commun. 9, 2237 (2018).

    PubMed  PubMed Central  Google Scholar 

  204. Jadidi-Niaragh, F. et al. CD73 specific siRNA loaded chitosan lactate nanoparticles potentiate the antitumor effect of a dendritic cell vaccine in 4T1 breast cancer bearing mice. J. Control. Release 246, 46–59 (2017).

    CAS  PubMed  Google Scholar 

  205. Deng, Z. J. et al. Layer-by-layer nanoparticles for systemic codelivery of an anticancer drug and siRNA for potential triple-negative breast cancer treatment. ACS Nano 7, 9571–9584 (2013).

    CAS  PubMed  Google Scholar 

  206. Zhao, Y. et al. Polymetformin combines carrier and anticancer activities for in vivo siRNA delivery. Nat. Commun. 7, 11822 (2016).

    PubMed  PubMed Central  Google Scholar 

  207. Kataoka, K. et al. Spontaneous formation of polyion complex micelles with narrow distribution from antisense oligonucleotide and cationic block copolymer in physiological saline. Macromolecules 29, 8556–8557 (1996).

    CAS  Google Scholar 

  208. Christie, R. J. et al. Targeted polymeric micelles for siRNA treatment of experimental cancer by intravenous injection. ACS Nano 6, 5174–5189 (2012).

    CAS  PubMed  Google Scholar 

  209. Kim, H. J. et al. Multifunctional polyion complex micelle featuring enhanced stability, targetability, and endosome escapability for systemic siRNA delivery to subcutaneous model of lung cancer. Drug. Deliv. Transl. Res. 4, 50–60 (2014).

    CAS  PubMed  Google Scholar 

  210. Nishida, H. et al. Systemic delivery of siRNA by actively targeted polyion complex micelles for silencing the E6 and E7 human papillomavirus oncogenes. J. Control. Release 231, 29–37 (2016).

    CAS  PubMed  Google Scholar 

  211. Watanabe, S. et al. In vivo rendezvous of small nucleic acid drugs with charge-matched block catiomers to target cancers. Nat. Commun. 10, 1894 (2019).

    PubMed  PubMed Central  Google Scholar 

  212. Jiang, H. et al. New path to treating pancreatic cancer: TRAIL gene delivery targeting the fibroblast-enriched tumor microenvironment. J. Control. Release 286, 254–263 (2018).

    CAS  PubMed  Google Scholar 

  213. Adams, D. et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N. Engl. J. Med. 379, 11–21 (2018).

    CAS  PubMed  Google Scholar 

  214. Setten, R. L., Rossi, J. J. & Han, S. P. The current state and future directions of RNAi-based therapeutics. Nat. Rev. Drug Discov. 18, 421–446 (2019).

    CAS  PubMed  Google Scholar 

  215. Singh, A., Trivedi, P. & Jain, N. K. Advances in siRNA delivery in cancer therapy. Artif. Cells Nanomed. Biotechnol. 46, 274–283 (2018).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors apologize to authors whose work could not be cited owing to space constraints. In general, the authors focused on systemically and locally administered particle-based therapies, so the scope of this article does not include certain nanotechnologies developed for the delivery of immunotherapies, such as injectable scaffolds, which are currently under clinical investigation (NCT01753089). The authors thank M. Kalli (University of Cyprus) for assistance with the preparation of the figures, K. Kakimi (University of Tokyo) for helpful discussions and A. Osada (NanoCarrier Co., Ltd), Y. Huang (Cyrus Tang Haematology Center), M. R. Martin (University of Tokyo), V. Melo (University of Tokyo) and Z. Amoozgar and D. Fukumura (Massachusetts General Hospital) for critical input into the manuscript. The research leading to these results has received funding from the National Foundation for Cancer Research, the Ludwig Center at Harvard, the Jane’s Trust Foundation, the Advanced Medical Research Foundation, the US National Cancer Institute grants P01-CA080124, R01-CA098706, R01-CA208205 and U01-CA224348 and the US Department of Defense Breast Cancer Research Program Innovator Award W81XWH-10-1-0016 (to R.K.J.), the European Research Council grant 838414 and the INFRASTRUCTURE/1216/0052 grant co-financed by the European Regional Development Fund and the Republic of Cyprus through the Research Promotion Foundation (to T.S.), and the Japan Society for the Promotion of Science (JSPS) Grants-in-Aid for Scientific Research B (JP16H03179) and Young Scientists B (JP25750172) to H.C. R.K.J. is a recipient of an Outstanding Investigator Award R35-CA197743 from the U.S. National Cancer Institute. J.D.M was supported by a JSPS Postdoctoral Fellowship, P16731.

Author information

Authors and Affiliations

Authors

Contributions

J.D.M. researched data for this article. All authors made a substantial contribution to the discussion of content, writing the manuscript and reviewing and/or editing the manuscript prior to submission.

Corresponding authors

Correspondence to Triantafyllos Stylianopoulos or Rakesh K. Jain.

Ethics declarations

Competing interests

J.D.M. became a full-time employee of NanoCarrier during the preparation of this manuscript. R.K.J. has received honoraria from Amgen, has acted as a consultant for Chugai, Merck, Ophthotech, Pfizer, SPARC, SynDevRx and XTuit, owns equity in Enlight, Ophthotech and SynDevRx and serves on the Boards of Trustees of Tekla Healthcare Investors, Tekla Life Sciences Investors, Tekla Healthcare Opportunities Fund and Tekla World Healthcare Fund. H.C. and T.S. declare no competing interests.

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

Martin, J.D., Cabral, H., Stylianopoulos, T. et al. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nat Rev Clin Oncol 17, 251–266 (2020). https://doi.org/10.1038/s41571-019-0308-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-019-0308-z

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