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.

Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy

This article has been updated

Abstract

Lymphoid organs, in which antigen presenting cells (APCs) are in close proximity to T cells, are the ideal microenvironment for efficient priming and amplification of T-cell responses1. However, the systemic delivery of vaccine antigens into dendritic cells (DCs) is hampered by various technical challenges. Here we show that DCs can be targeted precisely and effectively in vivo using intravenously administered RNA-lipoplexes (RNA-LPX) based on well-known lipid carriers by optimally adjusting net charge, without the need for functionalization of particles with molecular ligands. The LPX protects RNA from extracellular ribonucleases and mediates its efficient uptake and expression of the encoded antigen by DC populations and macrophages in various lymphoid compartments. RNA-LPX triggers interferon-α (IFNα) release by plasmacytoid DCs and macrophages. Consequently, DC maturation in situ and inflammatory immune mechanisms reminiscent of those in the early systemic phase of viral infection are activated2. We show that RNA-LPX encoding viral or mutant neo-antigens or endogenous self-antigens induce strong effector and memory T-cell responses, and mediate potent IFNα-dependent rejection of progressive tumours. A phase I dose-escalation trial testing RNA-LPX that encode shared tumour antigens is ongoing. In the first three melanoma patients treated at a low-dose level, IFNα and strong antigen-specific T-cell responses were induced, supporting the identified mode of action and potency. As any polypeptide-based antigen can be encoded as RNA3,4, RNA-LPX represent a universally applicable vaccine class for systemic DC targeting and synchronized induction of both highly potent adaptive as well as type-I-IFN-mediated innate immune mechanisms for cancer immunotherapy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: RNA-LPX of negative net charge deliver RNA-encoded antigens body-wide to lymphoid-resident DCs.
Figure 2: RNA-LPX vaccines induce TLR7-triggered IFNα production, IFNAR-dependent activation of APCs and effector cells, and strong expansion of fully functional antigen-specific T cells.
Figure 3: RNA-LPX vaccines mediate rejection of advanced, aggressively growing tumours in mice.
Figure 4: Clinically administered RNA-LPX vaccines dose-dependently induce systemic INFα and de novo priming and amplification of T cells against vaccine antigens.

Similar content being viewed by others

Change history

  • 07 June 2016

    The competing financial interests statement did not display correctly online when this paper was first published; this has been corrected and the statement is now available.

References

  1. Zinkernagel, R. M. et al. Antigen localisation regulates immune responses in a dose- and time-dependent fashion: a geographical view of immune reactivity. Immunol. Rev. 156, 199–209 (1997).

    Article  CAS  PubMed  Google Scholar 

  2. Stetson, D. B. & Medzhitov, R. Type I interferons in host defense. Immunity 25, 373–381 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Boczkowski, D., Nair, S. K., Snyder, D. & Gilboa, E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J. Exp. Med. 184, 465–472 (1996).

    Article  CAS  PubMed  Google Scholar 

  4. Sahin, U., Karikó, K. & Türeci, Ö. mRNA-based therapeutics—developing a new class of drugs. Nat. Rev. Drug Discov. 13, 759–780 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Banchereau, J. & Steinman, R. M. Dendritic cells and the control of immunity. Nature 392, 245–252 (1998).

    Article  CAS  PubMed  ADS  Google Scholar 

  6. Tacken, P. J., de Vries, I. J. M., Torensma, R. & Figdor, C. G. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat. Rev. Immunol. 7, 790–802 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Phua, K. K. L. Towards targeted delivery systems: ligand conjugation strategies for mRNA nanoparticle tumor vaccines. J. Immunol. Res. 680620 (2015).

  8. Mitragotri, S., Burke, P. A. & Langer, R. Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat. Rev. Drug Discov. 13, 655–672 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Brito, L. A. et al. A cationic nanoemulsion for the delivery of next generation RNA vaccines. Mol. Ther. 22, 2118–2129 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pollard, C. et al. Type I IFN counteracts the induction of antigen-specific immune responses by lipid-based delivery of mRNA vaccines. Mol. Ther. 21, 251–259 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Zhou, W. Z. et al. RNA melanoma vaccine: induction of antitumor immunity by human glycoprotein 100 mRNA immunization. Hum. Gene Ther. 10, 2719–2724 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Hess, P. R., Boczkowski, D., Nair, S. K., Snyder, D. & Gilboa, E. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunol. Immunother. 55, 672–683 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Perche, F. et al. Enhancement of dendritic cells transfection in vivo and of vaccination against B16F10 melanoma with mannosylated histidylated lipopolyplexes loaded with tumor antigen messenger RNA. Nanomedicine 7, 445–453 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. 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  PubMed  PubMed Central  Google Scholar 

  15. Lee, E. R. et al. Detailed analysis of structures and formulations of cationic lipids for efficient gene transfer to the lung. Hum. Gene Ther. 7, 1701–1717 (1996).

    Article  CAS  PubMed  Google Scholar 

  16. Liu, Y. et al. Factors influencing the efficiency of cationic liposome-mediated intravenous gene delivery. Nat. Biotechnol. 15, 167–173 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Diken, M. et al. Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation. Gene Ther. 18, 702–708 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Sallusto, F., Cella, M., Danieli, C. & Lanzavecchia, A. Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: downregulation by cytokines and bacterial products. J. Exp. Med. 182, 389–400 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Platt, C. D. et al. Mature dendritic cells use endocytic receptors to capture and present antigens. Proc. Natl Acad. Sci. USA 107, 4287–4292 (2010).

    Article  PubMed  ADS  PubMed Central  Google Scholar 

  20. Diebold, S. S., Kaisho, T., Hemmi, H., Akira, S. & Reis e Sousa & C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 303, 1529–1531 (2004).

    Article  CAS  PubMed  ADS  Google Scholar 

  21. Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520, 692–696 (2015).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  22. Le Bon, A. et al. Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat. Immunol. 4, 1009–1015 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Bacher, N. et al. Interferon-α suppresses cAMP to disarm human regulatory T cells. Cancer Res. 73, 5647–5656 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Zitvogel, L., Galluzzi, L., Kepp, O., Smyth, M. J. & Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 15, 405–414 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Janeway, C. A. Jr The immune system evolved to discriminate infectious nonself from noninfectious self. Immunol. Today 13, 11–16 (1992).

    Article  CAS  PubMed  Google Scholar 

  26. Janeway, C. A. Jr & Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 20, 197–216 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Swiecki, M., Gilfillan, S., Vermi, W., Wang, Y. & Colonna, M. Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8+ T cell accrual. Immunity 33, 955–966 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kamphuis, E., Junt, T., Waibler, Z., Forster, R. & Kalinke, U. Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia. Blood 108, 3253–3261 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Müller, U. et al. Functional role of type I and type II interferons in antiviral defense. Science 264, 1918–1921 (1994).

    Article  PubMed  ADS  Google Scholar 

  30. Jung, S. et al. In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity 17, 211–220 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Huang, A. Y. et al. The immunodominant major histocompatibility complex class I-restricted antigen of a murine colon tumor derives from an endogenous retroviral gene product. Proc. Natl Acad. Sci. USA 93, 9730–9735 (1996).

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  32. Lin, K.-Y. et al. Treatment of established tumors with a novel vaccine that enhances major histocompatibility class II presentation of tumor antigen. Cancer Res. 21–26 (1996).

  33. Chuang, C.-M., Monie, A., Wu, A. & Hung, C.-F. Combination of apigenin treatment with therapeutic HPV DNA vaccination generates enhanced therapeutic antitumor effects. J. Biomed. Sci. 16, 49 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Holtkamp, S. et al. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood 108, 4009–4017 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Kreiter, S. et al. Increased antigen presentation efficiency by coupling antigens to MHC class I trafficking signals. J. Immunol. 180, 309–318 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Kuhn, A. N. et al. Phosphorothioate cap analogs increase stability and translational efficiency of RNA vaccines in immature dendritic cells and induce superior immune responses in vivo. Gene Ther. 17, 961–971 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Slansky, J. E. et al. Enhanced antigen-specific antitumor immunity with altered peptide ligands that stabilize the MHC-peptide-TCR complex. Immunity 13, 529–538 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Kenter, G. G. et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N. Engl. J. Med. 361, 1838–1847 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Kreiter, S. et al. Simultaneous ex vivo quantification of antigen-specific CD4+ and CD8+ T cell responses using in vitro transcribed RNA. Cancer Immunol. Immunother. 56, 1577–1587 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Bangham, A. D., Hill, M. W. & Miller, N. G. A. in Methods in Membrane Biology (ed. Korn, E. D. ) 1–68 (Springer US, 1974).

  41. Duzgunes, N. in Methods in Enzymology (Academic Press Inc, 2009).

  42. Batzri, S. & Korn, E. D. Single bilayer liposomes prepared without sonication. Biochim. Biophys. Acta 298, 1015–1019 (1973).

    Article  CAS  PubMed  Google Scholar 

  43. Barichello, J. M., Ishida, T. & Kiwada, H. Complexation of siRNA and pDNA with cationic liposomes: the important aspects in lipoplex preparation. Methods Mol. Biol. 605, 461–472 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Racoosin, E. L. & Swanson, J. A. Macrophage colony-stimulating factor (rM-CSF) stimulates pinocytosis in bone marrow-derived macrophages. J. Exp. Med. 170, 1635–1648 (1989).

    Article  CAS  PubMed  Google Scholar 

  45. Sarkar, K., Kruhlak, M. J., Erlandsen, S. L. & Shaw, S. Selective inhibition by rottlerin of macropinocytosis in monocyte-derived dendritic cells. Immunology 116, 513–524 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Bar-On, L. & Jung, S. Defining in vivo dendritic cell functions using CD11c-DTR transgenic mice. Methods Mol. Biol. 595, 429–442 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Aichele, P. et al. Macrophages of the splenic marginal zone are essential for trapping of blood-borne particulate antigen but dispensable for induction of specific T cell responses. J. Immunol. 171, 1148–1155 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Wexler, H. Accurate identification of experimental pulmonary metastases. J. Natl. Cancer Inst. 36, 641–645 (1966).

    Article  CAS  PubMed  Google Scholar 

  49. Chen, Y. T. et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc. Natl Acad. Sci. USA 94, 1914–1918 (1997).

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  50. Brichard, V. et al. The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J. Exp. Med. 178, 489–495 (1993).

    Article  CAS  PubMed  Google Scholar 

  51. Gaugler, B. et al. Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J. Exp. Med. 179, 921–930 (1994).

    Article  CAS  PubMed  Google Scholar 

  52. Simon, P. et al. Functional TCR retrieval from single antigen-specific human T cells reveals multiple novel epitopes. Cancer Immunol. Res. 2, 1230–1244 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank M. Holzmann, R. Roth, U. Schmitt, M. Brkic, A. König, C. Worm, N. Krimmel, A.-K. Thiel, C. Bender, M. Suchan, A.-L. Popa, P. Bezerra Gomes, S. Herbert, M. Lux, D. Wintergerst, V. Bischoff, R. Krishna, Y. Hajime, J. Groß, A. Spruss, M. Erdeljan, S. Wöll, T. Bukur, H. Muramatsu and M. Baiersdörfer for technical support, NIH Tetramer Core Facility for providing gp70 MHC class I tetramer, A. Kong for critical reading, A. Kemmer-Brück, D. Schwarck, S. Bolte for clinical operations support and K. Kariko for advice. This work was supported by the technology innovation program of the Rhineland Palatinate government, the InnoTop program, the CI3 Cutting Edge Cluster Funding of the German Ministry of Technology (BMBF) and the Collaborative Research Group 1066 of Deutsche Forschungsgemeinschaft (DFG). L.M.K. was funded by the Graduate School of Immunotherapy 1043 of DFG.

Author information

Authors and Affiliations

Authors

Contributions

U.S. was responsible for conception and experimental strategy of the study. Formulation development was performed by H.H. and P.L. Design and analysis of the experiments were done by L.M.K, M.D., H.H., S.K, M.M. and D.F. supported by K.C.R. and F.V. L.M.K, M.D., M.M., D.F., K.C.R., A.S., F.V., Y.H., Ho.He, C.G. and M.V. performed the experiments and acquired the data. C.L., J. H., J.D., A.N.K., J.B., R.J., S.H., S.G., E.D., R.R. and S.A. were involved in design, implementation or laboratory analyses of the clinical study. L.M.K., M.D., Ö.T. and U.S. interpreted the data and drafted the manuscript. C.H. supported the revision of the manuscript.

Corresponding author

Correspondence to Ugur Sahin.

Ethics declarations

Competing interests

U.S., H.H, M.M., D.F., K.C.R, C.G., Ho.He, Y.H., M.V., A.N.K., J.B., E.D., J.D., R.J. and S.H. are employees at BioNTech AG (Mainz, Germany). M.D. and S.K. are working as consultants for BioNTech AG (Mainz, Germany). U.S., L.M.K., M.D., H.H., S.K., D.F., M.M. and K.C.R. are inventors on patents and patent applications related to this study. U.S. and C.H are stock owners, C.H. is advisor and supervisory board member and U.S. is management board member of BioNTech AG. All other authors have no potential conflict of interest.

Additional information

Reviewer Information: Nature thanks O. Farokhzad, C. G. Figdor, C. Melief, L. Zhang and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Physicochemical characteristics and biological activity of RNA-LPX constituted from different lipids at various charge ratios.

a, Bioluminescence imaging of Luc expression in BALB/c mice 6 h after i.v. injection of different transfection reagents and controls: PBS (n = 3), 60 μg Luc-RNA alone (n = 3), 25 μg Luc-RNA complexed with TransMessenger (Qiagen) (n = 3), 5 μg Luc-RNA complexed with Viromer RED (Lipocalyx) (n = 3). b, Cryo-TEM images of Luc-LPX constituted at a positive:negative ((+):(−)) charge ratio of 1.3:2 with DOTMA/DOPE liposomes. Scale bar, 100 nm. c, Fraction of uncomplexed RNA in Luc-LPX preparations constituted at different charge ratios with DOTMA/DOPE liposomes determined by capillary gel electrophoresis (n = 2–7). d, Particle size, polydispersity index (left) and zeta potential (right) (n = 3) of RNA-LPX constituted with Luc-RNA and differently constituted liposomes at various charge ratios. e, Bioluminescence imaging of BALB/c mice (n = 3) after i.v. injection of Luc-LPX constituted with different liposomes at various charge ratios corresponding to d. Pie charts show relative contribution of each organ to total signal. f, Relative biodistribution of Luc expression in explanted organs of BALB/c mice (n = 3) after i.v. injection of Luc-LPX constituted with DOTMA/DOPE liposomes at a charge ratio of (+):(-) of 1.3:2 or Luc-RNA alone. g, Luc expression in human immature DCs transfected with 5 μg Luc-LPX constituted freshly or stored after constitution for indicated time periods at 4 °C (left) or room temperature (right). RNA-LPX tested in duplicates (stored) or quadruplets (fresh). Each bar represents triplicates. h, Particle size (upper left) and percentage of RNA integrity (upper right) of Luc-LPX (n = 1) incubated in 50% mouse serum for indicated time periods at 37 °C. Bioluminescence imaging of Luc expression in BALB/c mice (n = 5) after i.v. injection of Luc-LPX preincubated in 50% mouse serum for 30 min at 37 °C (lower left and right). NM, not measured. Error bars, median with interquartile range (h), otherwise mean ± s.d.

Extended Data Figure 2 Biodistribution and cellular uptake mechanism of RNA-LPX vaccines.

a, Uptake of Cy5-labelled RNA in splenic cell subsets of C57BL/6 mice (n = 3) 1 h after i.v. injection of 40 μg Cy5-labelled RNA-LPX. b, Localization of CD11c and Cy3 double-positive cells in the spleen of BALB/c mice (n = 2) 1 h after i.v. injection of 40 μg Cy3-labelled RNA-LPX. Nuclear staining in blue. Scale bar,100 μm. c, Half-life of RNA-LPX in circulation analysed by quantitative RT–qPCR in male and female C57BL/6 mice (n = 5 per time-point) after injection of 60 μg RNA-LPX constituted with NY-ESO-I, tyrosinase, MAGE-A3 and TPTE RNA (15 μg each). d, Localization of Cy5+ (upper left) or Thy1.1+ cells (lower left) in spleen and liver of BALB/c mice (n = 5) determined by microscopy or flow cytometry 1 h or 20 h after i.v. injection of 40 μg Cy5-labelled RNA-LPX or 40 μg 1-methyl-pseudouridine-modified Thy1.1-LPX, respectively. Nuclear staining in blue. Scale bar, 50 μm (top), 20 μm (bottom). Biodistribution of Cy5 signal in homogenized organs of BALB/c mice (n = 2) (right). Note the signal in the liver is overestimated in this analysis owing to the strong signal in the gall bladder, probably reflecting biliary secreted free dye. e, Bioluminescence imaging of lymph nodes of BALB/c mice (n = 3) 18 h after i.v. injection of 40 μg 1-methyl-pseudouridine-modified Luc-LPX. ax, axillary; ing, inguinal; mand, mandibular. f, Flow cytometry analysis of Cy5 and Thy1.1 expression in CD11c+ cells in the bone marrow of C57BL/6 mice (n = 3) 1 h or 20 h after i.v. injection of 40 μg Cy5-labelled RNA-LPX or 40 μg 1-methyl-pseudouridine-modified Thy1.1-LPX, respectively. g, h, Localization of Cy3-labelled RNA in human immature DCs after co-transfection of 1.25 μg Cy3-labelled RNA-LPX at a charge ratio of (+):(−) of 1.3:2 and 3:1 with dextran (g) or of 1.3:2 after staining for TLR7 or EEA1 (h). Nuclear staining in blue. Scale bar, 10 μm. i, Visualization and quantification of inhibited uptake of positively as well as negatively charged Cy3-labelled RNA-LPX in human immature DCs pretreated with rottlerin or cytochalasin D. Scale bar, 10 μm. j, Bioluminescence imaging of lymph nodes of BALB/c mice (n = 3) injected intranodally with 10 μM rottlerin in 10 μl PBS 15 min before i.v. injection of 80 μg Luc-LPX. k, Luminescence assay of whole blood enriched or not enriched with human immature DCs pretreated with poly I:C or not (control) before transfection with Luc-LPX at a charge ratio of 1.3:2. WB, whole blood. l, Poly-I:C-induced maturation determined by CD86 expression (left), bioluminescence imaging (middle) and eGFP expression in splenic cDC subsets (right) upon injection of BALB/c mice (n = 3) with 50 μg poly I:C i.p. 12 h before i.v. injection of 20 μg Luc-LPX or 80 μg eGFP-LPX, respectively. Significance was determined using unpaired two-tailed Student’s t-test (d, lower left, f, l, middle) and one-way ANOVA and Tukey’s multiple comparisons test (ik, l, right). Error bars, mean ± s.e.m. (k) or mean ± s.d. otherwise.

Extended Data Figure 3 Systemic TLR7- and IFNAR-dependent activation of APCs and effector cells, IFNα production and strong expansion of fully functional antigen-specific T cells induced by RNA-LPX vaccines.

a, Localization of splenic CD11chi cells at baseline (top) and 6 h after i.v. injection of 40 μg HA-LPX (bottom) into BALB/c mice (n = 2). Nuclear staining in blue. Scale bar, 100 μm. RP, red pulp; WP, white pulp. be, Activation marker expression in splenic cell subsets and kinetics of IFNα serum levels after i.v. injection of mice (n = 3 per time point) with HA-LPX in Tlr3−/−, Tlr4−/− and Tlr9−/− mice (b), in Ifnar1−/− mice (c, d), or in BALB/c mice treated with 100 μg anti-IFNAR1 antibody or isotype i.p. 1 h before i.v. injection of HA-LPX (e). Ab, antibody. f, mRNA levels of IFNα isoforms in sorted splenic APC subsets of C57BL/6 mice (n = 3) 1 h after i.v. injection of HA-LPX determined by qRT–PCR. Data expressed as log2-fold change, as compared to control animals. g, IFNα serum levels after i.v. injection of HA-LPX in BDCA2-DTR mice (n = 3 per time point) depleted (depl) of pDCs (left) and in C57BL/6 mice (n = 3 per time point) depleted of macrophages (right). h, CFSE proliferation profile of HA-specific CD4+ T cells in lymphoid compartments of BALB/c Thy1.1+ mice (n = 3) after adoptive transfer of HA-specific Thy1.2+ HA-TCR-transgenic CD4+ T cells and subsequent immunization with HA-LPX or control (untreated). Fraction of proliferated cells indicated. tg, transgenic. i, Priming of naive HA-specific CD8+ T cells ex vivo. BALB/c (n = 3) mice were immunized with 80 μg HA-LPX, irrelevant (eGFP)-LPX or NaCl (control). Splenocytes were prepared 12 h later and co-incubated with CFSE-labelled CL4-TCR-transgenic CD8+ T cells isolated using MACS magnetic microbeads coated with CD8 antibodies at an effector:target ratio of 1:6. Four days later, proliferation profiles were analysed by flow cytometry. Numbers indicate the percentage of proliferated cells. j, Fraction of cytokine-secreting CD8+ T cells within CD8+ T cells in the spleen upon de novo priming in C57BL/6 mice (n = 5) immunized i.v. (day 0, 3, 8) with OVA-LPX after in vitro restimulation with no (none), irrelevant VSV (irrelevant) or OVA peptide and intracellular cytokine staining (top). Spleen ex vivo ELISPOT assay upon de novo priming in BALB/c mice (n = 5) immunized i.v. (day 0, 3, 8) with gp70-LPX. Stimulation with no (none), irrelevant HA (irrelevant) or gp70 peptide (lower left). gp70-specific cytotoxicity in vivo (lower right). BALB/c mice (n = 5) were immunized i.v. (day 0, 3, 8) with 40 μg gp70-LPX. Naive splenocytes were labelled with 0.5 or 5 μM CFSE and pulsed with peptide (6 μg ml−1) five days after the last immunization, and target cells (2 × 107) were adoptively transferred into immunized recipients i.v. (irrelevant HA-loaded CFSElow:gp70-loaded CFSEhigh = 1:1). Recipient splenocytes were analysed by flow cytometry 18 h after transfer, and antigen-specific lysis was determined: specific lysis (%) = (1 − (percentage of cells pulsed with gp70/percentage of cells pulsed with HA)) × 100). k, Expression of memory markers CD127 and CD62L in gp70-specific, CD44+CD8+ T cells compared to non-specific CD8+ T cells in blood (day 19) and spleen (day 67) of BALB/c mice (n = 3) after priming with gp70-LPX (day 0, 7, 14). l, Fraction of gp70-specific CD8+ T cells within total CD8+ T cells in blood, bone marrow and lymph nodes determined by MHC class I tetramer staining after de novo priming of splenectomized BALB/c mice (n = 5–7) immunized with gp70-LPX (day 0, 7) or left untreated (control). Significance was determined using unpaired two-tailed Student’s t-test (b left, c), two-way ANOVA and Bonferroni’s multiple comparisons test (b right, g) and one-way ANOVA and Tukey’s multiple comparisons test (j, l). Error bars, mean ± s.d.

Extended Data Figure 4 Potent antitumour immunity and rejection of advanced aggressively growing tumours in mice conferred by RNA-LPX vaccines.

a, B16-OVA melanoma load in lungs of C57BL/6 mice (n = 8) immunized i.v. (days 4, 7, 11) with OVA-LPX or irrelevant (eGFP)-LPX. b, Expression of activation markers measured 24 h after i.v. injection of 40 μg irrelevant (empty vector)-LPX, eGFP-LPX or OVA-LPX by flow cytometry in splenic immune cell subsets (n = 3) and IFNα serum levels (n = 3) 6 h after injection in C57BL/6 mice. c, Bioluminescence signal of tumours in different groups before immunization and on day 25 (upper left), tumour load and lung weights (upper right) and TRP-1-specific CD8+ and CD4+ T-cell responses in spleens of control (untreated), irrelevant (empty vector)-LPX and TRP-1-LPX-immunized B6 albino mice (n = 12) on day 25 detected by ELISPOT assay (bottom), depicted in Fig. 3b. d, Bioluminescence imaging of CT26-Luc carcinoma in BALB/c mice (n = 4–7) depicted in Fig. 3c (left). e, TC-1-Luc tumour growth in C57BL/6 mice (n = 10) (left), depicted in Fig. 3d, and remission of established advanced TC-1-Luc tumours in C57BL/6 mice (n = 10) immunized i.v. with 40 μg E6/E7-LPX (day 13, 20, 27) (right). f, Survival of BALB/c mice rechallenged with CT26-Luc colon carcinoma cells on day 109, depicted in Fig. 3e. Significance was determined using one-way ANOVA and Tukey’s multiple comparisons test (c), two-way ANOVA and Bonferroni’s multiple comparisons test (d), paired two-tailed Student’s t-test (f, right), unpaired two-tailed Student’s t-test (f, far right), and log-rank test (f, left). Error bars, median with interquartile range (d), mean ± s.d. otherwise.

Extended Data Figure 5 Clinical application of RNA-LPX vaccines and de novo priming and amplification of patient T-cell responses against encoded vaccine antigens.

a, Vaccination scheme and monitoring for patients 1–3. b, Antigen-specific T-cell responses against NY-ESO-1 and tyrosinase determined by restimulation with overlapping peptide mixtures in IFNγ ELISPOT for patient 1. c, Antigen-specific T-cell responses against NY-ESO-1 and MAGE-A3, determined by post-IVS IFNγ ELISPOT assay at indicated days for patient 2. Values are corrected for background (no peptide). d, Antigen-specific T-cell responses against NY-ESO-I and MAGE-A3, determined by ex vivo IFNγ ELISPOT assay at indicated days for patient 3. Numbers in ELISPOT data indicate the amino acid position of each epitope. Significance was determined using unpaired two-tailed Student’s t-test. Error bars, mean ± s.e.m.

Extended Data Figure 6 Comparison of i.v. and s.c. routes for RNA-LPX administration in the context of T-cell priming and biodistribution of RNA-LPX upon s.c. administration.

Fraction of OVA-specific CD8+ T cells within CD8+ T cells on day 13 in blood after de novo priming of C57BL/6 mice (n = 5) immunized i.v. with OVA-LPX (day 0, 3, 8) (left). Biodistribution of Luc expression 24 h after s.c. injection of Luc-LPX in BALB/c mice (n = 3) (right). Signal can only be observed at the injection site and the draining lymph node. Significance was determined using one-way ANOVA and Tukey’s multiple comparisons test. Error bars, mean ± s.d.

Extended Data Table 1 Findings of non-GLP pilot pharmacokinetics and pharmacodynamics study in cynomolgus monkeys

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kranz, L., Diken, M., Haas, H. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–401 (2016). https://doi.org/10.1038/nature18300

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18300

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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