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:

Precision immunomodulation with synthetic nucleic acid technologies

Abstract

DNA and RNA of both natural and synthetic origin can elicit strong immune responses in higher organisms. Computer-aided design in conjunction with high-throughput sequencing has improved our ability to build tuneable synthetic DNA and RNA architectures with high structural and functional precision. The natural biological roles of nucleic acids endow these synthetic assemblies with the ability to specifically engage cellular components. This makes DNA and RNA particularly powerful materials with which to probe or programme immune cells. In this Perspective, we discuss the potential of designer nucleic acid assemblies to control and modulate the immune response for biomedical applications.

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

Fig. 1: Synthetic nucleic acid technologies for quantitative immunology.
Fig. 2: Strategies for immunomodulation.
Fig. 3: Established receptors and cytokine targets for immunomodulation.
Fig. 4: Hybrid strategies for immunomodulation.
Fig. 5: Synthetic RNA circuits in immune cells.

Similar content being viewed by others

References

  1. Rothemund, P. W. K. Folding DNA to create nanoscale shapes and patterns. Nature 440, 297–302 (2006).

    Article  CAS  Google Scholar 

  2. Douglas, S. M. et al. Self-assembly of DNA into nanoscale three-dimensional shapes. Nature 459, 414–418 (2009).

    Article  CAS  Google Scholar 

  3. Bhatia, D. et al. Quantum dot-loaded monofunctionalized DNA icosahedra for single-particle tracking of endocytic pathways. Nat. Nanotechnol. 11, 1112–1119 (2016).

    Article  CAS  Google Scholar 

  4. Lee, H. et al. Molecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNA delivery. Nat. Nanotechnol. 7, 389–393 (2012).

    Article  CAS  Google Scholar 

  5. Krishnan, Y. & Bathe, M. Designer nucleic acids to probe and program the cell. Trends Cell Biol. 22, 624–633 (2012).

    Article  CAS  Google Scholar 

  6. Paludan, S. R. & Bowie, A. G. Immune sensing of DNA. Immunity 38, 870–880 (2013).

    Article  CAS  Google Scholar 

  7. Surana, S., Shenoy, A. R. & Krishnan, Y. Designing DNA nanodevices for compatibility with the immune system of higher organisms. Nat. Nanotechnol. 10, 741–747 (2015).

    Article  CAS  Google Scholar 

  8. Khalil, D. N., Smith, E. L., Brentjens, R. J. & Wolchok, J. D. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 13, 273–290 (2016).

    Article  CAS  Google Scholar 

  9. DeFrancesco, L. Landmark approval for Dendreon’s cancer vaccine. Nat. Biotechnol. 28, 531–532 (2010).

    Article  CAS  Google Scholar 

  10. Shaughnessy, A. F. Monoclonal antibodies: magic bullets with a hefty price tag. BMJ 345, e8346 (2012).

    Article  CAS  Google Scholar 

  11. Shang, N. et al. Dendritic cells based immunotherapy. Am. J. Cancer Res. 7, 2091–2102 (2017).

    CAS  Google Scholar 

  12. Oldenburg, M. et al. TLR13 recognizes bacterial 23S rRNA devoid of erythromycin resistance-forming modification. Science 337, 1111–1115 (2012).

    Article  CAS  Google Scholar 

  13. Bauer, S. et al. Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. Proc. Natl Acad. Sci. USA 98, 9237–9242 (2001).

    Article  CAS  Google Scholar 

  14. Ballas, Z. K. et al. Divergent therapeutic and immunologic effects of oligodeoxynucleotides with distinct CpG motifs. J. Immunol. 167, 4878–4886 (2001).

    Article  CAS  Google Scholar 

  15. Chakraborty, K., Veetil, A. T., Jaffrey, S. R. & Krishnan, Y. Nucleic acid-based nanodevices in biological imaging. Annu. Rev. Biochem. 85, 349–373 (2016).

    Article  CAS  Google Scholar 

  16. Rusconi, C. P. et al. RNA aptamers as reversible antagonists of coagulation factor IXa. Nature 419, 90–94 (2002).

    Article  CAS  Google Scholar 

  17. Groff, K., Brown, J. & Clippinger, A. J. Modern affinity reagents: recombinant antibodies and aptamers. Biotechnol. Adv. 33, 1787–1798 (2015).

    Article  CAS  Google Scholar 

  18. Ruckman, J. et al. 2′-Fluoropyrimidine RNA-based aptamers to the 165-amino acid form of vascular endothelial growth factor (VEGF165). Inhibition of receptor binding and VEGF-induced vascular permeability through interactions requiring the exon 7-encoded domain. J. Biol. Chem. 273, 20556–20567 (1998).

    Article  CAS  Google Scholar 

  19. Rohloff, J. C. et al. Nucleic acid ligands with protein-like side chains: modified aptamers and their use as diagnostic and therapeutic agents. Mol. Ther. Nucleic Acids 3, e201 (2014).

    Article  CAS  Google Scholar 

  20. Klussmann, S., Nolte, A., Bald, R., Erdmann, V. A. & Fürste, J. P. Mirror-image RNA that binds D-adenosine. Nat. Biotechnol. 14, 1112–1115 (1996).

    Article  CAS  Google Scholar 

  21. Pastor, F. et al. An RNA toolbox for cancer immunotherapy. Nat. Rev. Drug Discov. 17, 751–767 (2018).

    Article  CAS  Google Scholar 

  22. Chaplin, D. D. Overview of the immune response. J. Allergy Clin. Immunol. 125, S3–23 (2010).

    Article  Google Scholar 

  23. Arazi, A., Pendergraft, W. F., Ribeiro, R. M., Perelson, A. S. & Hacohen, N. Human systems immunology: hypothesis-based modeling and unbiased data-driven approaches. Semin. Immunol. 25, 193–200 (2013).

    Article  CAS  Google Scholar 

  24. Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science 361, eaat5691 (2018).

    Article  CAS  Google Scholar 

  25. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    Article  CAS  Google Scholar 

  26. Iyengar, R. Why we need quantitative dynamic models. Sci. Signal. 2, eg3 (2009).

    Article  Google Scholar 

  27. Jungmann, R. et al. Single-molecule kinetics and super-resolution microscopy by fluorescence imaging of transient binding on DNA origami. Nano Lett. 10, 4756–4761 (2010).

    Article  CAS  Google Scholar 

  28. Jungmann, R. et al. Quantitative super-resolution imaging with qPAINT. Nat. Methods 13, 439–442 (2016).

    Article  CAS  Google Scholar 

  29. Strauss, S. et al. Modified aptamers enable quantitative sub-10-nm cellular DNA-PAINT imaging. Nat. Methods 15, 685–688 (2018).

    Article  CAS  Google Scholar 

  30. Satija, R. & Shalek, A. K. Heterogeneity in immune responses: from populations to single cells. Trends Immunol. 35, 219–229 (2014).

    Article  CAS  Google Scholar 

  31. Avraham, R. et al. Pathogen cell-to-cell variability drives heterogeneity in host immune responses. Cell 162, 1309–1321 (2015).

    Article  CAS  Google Scholar 

  32. Prakash, V. et al. Quantitative maps of endosomal DNA processing by single molecule counting. Angew. Chem. Int. Ed Engl. 58, 3073–3076 (2019).

    Article  CAS  Google Scholar 

  33. Domenyuk, V. et al. Plasma exosome profiling of cancer patients by a next generation systems biology approach. Sci. Rep. 7, 42741 (2017).

    Article  CAS  Google Scholar 

  34. Domenyuk, V. et al. Poly-ligand profiling differentiates trastuzumab-treated breast cancer patients according to their outcomes. Nat. Commun. 9, 1219 (2018).

    Article  CAS  Google Scholar 

  35. Dan, K., Veetil, A. T., Chakraborty, K. & Krishnan, Y. DNA nanodevices map enzymatic activity in organelles. Nat. Nanotechnol. 14, 252–259 (2019).

    Article  CAS  Google Scholar 

  36. Thekkan, S. et al. A DNA-based fluorescent reporter maps HOCl production in the maturing phagosome. Nat. Chem. Biol. https://doi.org/10.1038/s41589-018-0176-3 (2018).

    Article  Google Scholar 

  37. Surana, S., Bhat, J. M., Koushika, S. P. & Krishnan, Y. An autonomous DNA nanomachine maps spatiotemporal pH changes in a multicellular living organism. Nat. Commun. 2, 340 (2011).

    Article  CAS  Google Scholar 

  38. Modi, S. et al. A DNA nanomachine that maps spatial and temporal pH changes inside living cells. Nat. Nanotechnol. 4, 325–330 (2009).

    Article  CAS  Google Scholar 

  39. Kawasaki, T. & Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 5, 461 (2014).

    Article  CAS  Google Scholar 

  40. Bogdan, C. Nitric oxide and the immune response. Nat. Immunol. 2, 907–916 (2001).

    Article  CAS  Google Scholar 

  41. Tan, R. S. T., Ho, B., Leung, B. P. & Ding, J. L. TLR cross-talk confers specificity to innate immunity. Int. Rev. Immunol. 33, 443–453 (2014).

    Article  CAS  Google Scholar 

  42. Jayasena, S. D. Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin. Chem. 45, 1628–1650 (1999).

    CAS  Google Scholar 

  43. Ellington, A. D. & Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 346, 818–822 (1990).

    Article  CAS  Google Scholar 

  44. Tuerk, C. & Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249, 505–510 (1990).

    Article  CAS  Google Scholar 

  45. Alam, K. K., Chang, J. L. & Burke, D. H. FASTAptamer: a bioinformatic toolkit for high-throughput sequence analysis of combinatorial selections. Mol. Ther. Nucleic Acids 4, e230 (2015).

    Article  CAS  Google Scholar 

  46. Cho, M. et al. Quantitative selection of DNA aptamers through microfluidic selection and high-throughput sequencing. Proc. Natl Acad. Sci. USA 107, 15373–15378 (2010).

    Article  CAS  Google Scholar 

  47. Soldevilla, M. M., Villanueva, H. & Pastor, F. Aptamers: a feasible technology in cancer immunotherapy. J. Immunol. Res. 2016, 1083738 (2016).

    Article  CAS  Google Scholar 

  48. Sun, H. et al. Oligonucleotide aptamers: new tools for targeted cancer therapy. Mol. Ther. Nucleic Acids 3, e182 (2014).

    Article  CAS  Google Scholar 

  49. Moynihan, K. D. & Irvine, D. J. Roles for innate immunity in combination immunotherapies. Cancer Res. 77, 5215–5221 (2017).

    Article  CAS  Google Scholar 

  50. Gajewski, T. F., Schreiber, H. & Fu, Y.-X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14, 1014–1022 (2013).

    Article  CAS  Google Scholar 

  51. Kärre, K., Ljunggren, H. G., Piontek, G. & Kiessling, R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature 319, 675–678 (1986).

    Article  Google Scholar 

  52. Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996).

    Article  CAS  Google Scholar 

  53. Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. USA 107, 4275–4280 (2010).

    Article  CAS  Google Scholar 

  54. Ishida, Y., Agata, Y., Shibahara, K. & Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 11, 3887–3895 (1992).

    Article  CAS  Google Scholar 

  55. Buchbinder, E. I. & Desai, A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. 39, 98–106 (2016).

    Article  CAS  Google Scholar 

  56. Santulli-Marotto, S., Nair, S. K., Rusconi, C., Sullenger, B. & Gilboa, E. Multivalent RNA aptamers that inhibit CTLA-4 and enhance tumor immunity. Cancer Res. 63, 7483–7489 (2003).

    CAS  Google Scholar 

  57. Devaraj, S. G. T., Lokesh, G. L. R., Zu, Y., Chang, J. & Iyer, S. P. DNA aptamer against anti- programmed cell death-1 (Anti-PD1-Apt) induces robust anti-leukemic activity in vitro and in vivo humanized NSG mice with myeloid leukemia xenografts. Blood 130 (Suppl. 1), 1373 (2017).

    Google Scholar 

  58. Moore, K. W., de Waal Malefyt, R., Coffman, R. L. & O’Garra, A. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 19, 683–765 (2001).

    Article  CAS  Google Scholar 

  59. Zheng, L. M. et al. Interleukin-10 inhibits tumor metastasis through an NK cell-dependent mechanism. J. Exp. Med. 184, 579–584 (1996).

    Article  CAS  Google Scholar 

  60. Berezhnoy, A. et al. Isolation and optimization of murine IL-10 receptor blocking oligonucleotide aptamers using high-throughput sequencing. Mol. Ther. 20, 1242–1250 (2012).

    Article  CAS  Google Scholar 

  61. Lee, S.-W., Salek-Ardakani, S., Mittler, R. S. & Croft, M. Hypercostimulation through 4-1BB distorts homeostasis of immune cells. J. Immunol. 182, 6753–6762 (2009).

    Article  CAS  Google Scholar 

  62. Pastor, F., Kolonias, D., McNamara, J. O. & Gilboa, E. Targeting 4-1BB costimulation to disseminated tumor lesions with bi-specific oligonucleotide aptamers. Mol. Ther. 19, 1878–1886 (2011).

    Article  CAS  Google Scholar 

  63. Tsou, P., Katayama, H., Ostrin, E. J. & Hanash, S. M. The emerging role of B cells in tumor immunity. Cancer Res. 76, 5597–5601 (2016).

    Article  CAS  Google Scholar 

  64. Von Bergwelt-Baildon, M. S. et al. Human primary and memory cytotoxic T lymphocyte responses are efficiently induced by means of CD40-activated B cells as antigen-presenting cells: potential for clinical application. Blood 99, 3319–3325 (2002).

    Article  Google Scholar 

  65. Soldevilla, M. M. et al. 2-Fluoro-RNA oligonucleotide CD40 targeted aptamers for the control of B lymphoma and bone-marrow aplasia. Biomaterials 67, 274–285 (2015).

    Article  CAS  Google Scholar 

  66. Raddatz, M.-S. L. et al. Enrichment of cell-targeting and population-specific aptamers by fluorescence-activated cell sorting. Angew. Chem. Int. Ed Engl. 47, 5190–5193 (2008).

    Article  CAS  Google Scholar 

  67. Mayer, G. et al. Fluorescence-activated cell sorting for aptamer SELEX with cell mixtures. Nat. Protoc. 5, 1993–2004 (2010).

    Article  CAS  Google Scholar 

  68. Opazo, F. et al. Modular assembly of cell-targeting devices based on an uncommon G-quadruplex aptamer. Mol. Ther. Nucleic Acids 4, e251 (2015).

    Article  CAS  Google Scholar 

  69. Thompson, J. S. et al. BAFF-R, a newly identified TNF receptor that specifically interacts with BAFF. Science 293, 2108–2111 (2001).

    Article  CAS  Google Scholar 

  70. Zhou, J. et al. Dual functional BAFF receptor aptamers inhibit ligand-induced proliferation and deliver siRNAs to NHL cells. Nucleic Acids Res. 41, 4266–4283 (2013).

    Article  CAS  Google Scholar 

  71. Datta, S. K. Production of pathogenic antibodies: cognate interactions between autoimmune T and B cells. Lupus 7, 591–596 (1998).

    Article  CAS  Google Scholar 

  72. Hardy, I. R. et al. Anti-CD79 antibody induces B cell anergy that protects against autoimmunity. J. Immunol. 192, 1641–1650 (2014).

    Article  CAS  Google Scholar 

  73. Dörner, T. et al. Initial clinical trial of epratuzumab (humanized anti-CD22 antibody) for immunotherapy of systemic lupus erythematosus. Arthritis Res. Ther. 8, R74 (2006).

    Article  CAS  Google Scholar 

  74. Palucka, K. & Banchereau, J. Dendritic cells: a link between innate and adaptive immunity. J. Clin. Immunol. 19, 12–25 (1999).

    Article  CAS  Google Scholar 

  75. Sallusto, F. & Lanzavecchia, A. The instructive role of dendritic cells on T cell responses. Arthritis Res. 4, (Suppl. 3), S127–S132 (2002).

    Article  Google Scholar 

  76. De Saint-Vis, B. et al. The cytokine profile expressed by human dendritic cells is dependent on cell subtype and mode of activation. J. Immunol. 160, 1666–1676 (1998).

    Google Scholar 

  77. Blanco, P., Palucka, A. K., Pascual, V. & Banchereau, J. Dendritic cells and cytokines in human inflammatory and autoimmune diseases. Cytokine Growth Factor Rev. 19, 41–52 (2008).

    Article  CAS  Google Scholar 

  78. Lai, Y. & Dong, C. Therapeutic antibodies that target inflammatory cytokines in autoimmune diseases. Int. Immunol. 28, 181–188 (2016).

    Article  CAS  Google Scholar 

  79. Boshtam, M., Asgary, S., Kouhpayeh, S., Shariati, L. & Khanahmad, H. Aptamers against pro- and anti-inflammatory cytokines: a review. Inflammation 40, 340–349 (2017).

    Article  CAS  Google Scholar 

  80. Takai, T., Li, M., Sylvestre, D., Clynes, R. & Ravetch, J. V. FcR γ chain deletion results in pleiotrophic effector cell defects. Cell 76, 519–529 (1994).

    Article  CAS  Google Scholar 

  81. Nathan, C. F., Murray, H. W. & Cohn, Z. A. The macrophage as an effector cell. N. Engl. J. Med. 303, 622–626 (1980).

    Article  CAS  Google Scholar 

  82. Desjarlais, J. R., Lazar, G. A., Zhukovsky, E. A. & Chu, S. Y. Optimizing engagement of the immune system by anti-tumor antibodies: an engineer’s perspective. Drug Discov. Today 12, 898–910 (2007).

    Article  CAS  Google Scholar 

  83. Boltz, A. et al. Bi-specific aptamers mediating tumor cell lysis. J. Biol. Chem. 286, 21896–21905 (2011).

    Article  CAS  Google Scholar 

  84. Hsi, E. D. et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin. Cancer Res. 14, 2775–2784 (2008).

    Article  CAS  Google Scholar 

  85. Thielens, A., Vivier, E. & Romagné, F. NK cell MHC class I specific receptors (KIR): from biology to clinical intervention. Curr. Opin. Immunol. 24, 239–245 (2012).

    Article  CAS  Google Scholar 

  86. Kohrt, H. E. et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood 123, 678–686 (2014).

    Article  CAS  Google Scholar 

  87. Godal, R. et al. Natural killer cell killing of acute myelogenous leukemia and acute lymphoblastic leukemia blasts by killer cell immunoglobulin-like receptor-negative natural killer cells after NKG2A and LIR-1 blockade. Biol. Blood Marrow Transplant. 16, 612–621 (2010).

    Article  CAS  Google Scholar 

  88. Cohen, R. et al. Phase II study of monalizumab, a first-in-class NKG2A monoclonal antibody, in combination with cetuximab in previously treated recurrent or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN): preliminary assessment of safety and efficacy [abstract]. Cancer Res. 78, (Suppl. 13), CT158 (2018).

    Google Scholar 

  89. Cassetta, L. & Kitamura, T. Macrophage targeting: opening new possibilities for cancer immunotherapy. Immunology 155, 285–293 (2018).

    Article  CAS  Google Scholar 

  90. Brown, J. M., Recht, L. & Strober, S. The promise of targeting macrophages in cancer therapy. Clin. Cancer Res. 23, 3241–3250 (2017).

    Article  Google Scholar 

  91. Biswas, S. K. & Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat. Immunol. 11, 889–896 (2010).

    Article  CAS  Google Scholar 

  92. Georgoudaki, A.-M. et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 15, 2000–2011 (2016).

    Article  CAS  Google Scholar 

  93. Hume, D. A. & MacDonald, K. P. A. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 119, 1810–1820 (2012).

    Article  CAS  Google Scholar 

  94. Ries, C. H. et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 25, 846–859 (2014).

    Article  CAS  Google Scholar 

  95. Cannarile, M. A. et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 5, 53 (2017).

    Article  Google Scholar 

  96. Fujioka, Y. et al. A novel membrane glycoprotein, SHPS-1, that binds the SH2-domain-containing protein tyrosine phosphatase SHP-2 in response to mitogens and cell adhesion. Mol. Cell. Biol. 16, 6887–6899 (1996).

    Article  CAS  Google Scholar 

  97. Ring, N. G. et al. Anti-SIRPα antibody immunotherapy enhances neutrophil and macrophage antitumor activity. Proc. Natl Acad. Sci. USA 114, E10578–E10585 (2017).

    Article  CAS  Google Scholar 

  98. Wang, R. et al. Automated modular synthesis of aptamer-drug conjugates for targeted drug delivery. J. Am. Chem. Soc. 136, 2731–2734 (2014).

    Article  CAS  Google Scholar 

  99. Alley, S. C., Okeley, N. M. & Senter, P. D. Antibody-drug conjugates: targeted drug delivery for cancer. Curr. Opin. Chem. Biol. 14, 529–537 (2010).

    Article  CAS  Google Scholar 

  100. Diamantis, N. & Banerji, U. Antibody-drug conjugates—an emerging class of cancer treatment. Br. J. Cancer 114, 362–367 (2016).

    Article  CAS  Google Scholar 

  101. Sassoon, I. & Blanc, V. Antibody-drug conjugate (ADC) clinical pipeline: a review. Methods Mol. Biol. 1045, 1–27 (2013).

    Article  Google Scholar 

  102. Ohta, A. & Sitkovsky, M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414, 916–920 (2001).

    Article  CAS  Google Scholar 

  103. De Lera Ruiz, M., Lim, Y.-H. & Zheng, J. Adenosine A2A receptor as a drug discovery target. J. Med. Chem. 57, 3623–3650 (2014).

    Article  CAS  Google Scholar 

  104. Chiang, M.-J. et al. An Fc domain protein-small molecule conjugate as an enhanced immunomodulator. J. Am. Chem. Soc. 136, 3370–3373 (2014).

    Article  CAS  Google Scholar 

  105. Differding, E. AUNP-12 - a novel peptide therapeutic targeting PD-1 immune checkpoint pathway for cancer immunotherapy – structure activity relationships & peptide / peptidomimetic analogs. Differding.com http://www.differding.com/data/AUNP_12_A_novel_peptide_therapeutic_targeting_PD_1_immune_checkpoint_pathway_for_cancer_immunotherapy.pdf (2014).

  106. Shimizu-Sato, S., Huq, E., Tepperman, J. M. & Quail, P. H. A light-switchable gene promoter system. Nat. Biotechnol. 20, 1041–1044 (2002).

    Article  CAS  Google Scholar 

  107. Gossen, M. & Bujard, H. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc. Natl Acad. Sci. USA 89, 5547–5551 (1992).

    Article  CAS  Google Scholar 

  108. Liang, J. C., Bloom, R. J. & Smolke, C. D. Engineering biological systems with synthetic RNA molecules. Mol. Cell 43, 915–926 (2011).

    Article  CAS  Google Scholar 

  109. Vinkenborg, J. L., Karnowski, N. & Famulok, M. Aptamers for allosteric regulation. Nat. Chem. Biol. 7, 519–527 (2011).

    Article  CAS  Google Scholar 

  110. Mandal, M. & Breaker, R. R. Gene regulation by riboswitches. Nat. Rev. Mol. Cell Biol. 5, 451–463 (2004).

    Article  CAS  Google Scholar 

  111. Beisel, C. L., Chen, Y. Y., Culler, S. J., Hoff, K. G. & Smolke, C. D. Design of small molecule-responsive microRNAs based on structural requirements for Drosha processing. Nucleic Acids Res. 39, 2981–2994 (2011).

    Article  CAS  Google Scholar 

  112. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    Article  CAS  Google Scholar 

  113. An, C.-I., Trinh, V. B. & Yokobayashi, Y. Artificial control of gene expression in mammalian cells by modulating RNA interference through aptamer-small molecule interaction. RNA 12, 710–716 (2006).

    Article  CAS  Google Scholar 

  114. Rosenberg, S. A. IL-2: the first effective immunotherapy for human cancer. J. Immunol. 192, 5451–5458 (2014).

    Article  CAS  Google Scholar 

  115. Fyfe, G. et al. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J. Clin. Oncol. 13, 688–696 (1995).

    Article  CAS  Google Scholar 

  116. Atkins, M. B. et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J. Clin. Oncol. 17, 2105–2116 (1999).

    Article  CAS  Google Scholar 

  117. Kammula, U. S., White, D. E. & Rosenberg, S. A. Trends in the safety of high dose bolus interleukin-2 administration in patients with metastatic cancer. Cancer 83, 797–805 (1998).

    Article  CAS  Google Scholar 

  118. Dutcher, J. P. et al. Interleukin-2-based therapy for metastatic renal cell cancer: the Cytokine Working Group experience, 1989–1997. Cancer J. Sci. Am. 3 (Suppl. 1), S73–S78 (1997).

    Google Scholar 

  119. Chen, Y. Y., Jensen, M. C. & Smolke, C. D. Genetic control of mammalian T cell proliferation with synthetic RNA regulatory systems. Proc. Natl Acad. Sci. USA 107, 8531–8536 (2010).

    Article  CAS  Google Scholar 

  120. Ji, Y., Hocker, J. D. & Gattinoni, L. Enhancing adoptive T cell immunotherapy with microRNA therapeutics. Semin. Immunol. 28, 45–53 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank J. Kuriyan (University of California, Berkeley) and V. Kumar (The University of Chicago) for comments and suggestions. This work was supported by a research grant from the University of Pennsylvania Orphan Disease Center in partnership with the Andrew Coppola Foundation, the University of Chicago Women’s Board; a Pilot and Feasibility award from National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Center grant no. P30DK42086 to the University of Chicago Digestive Diseases Research Core Center; the Chicago Biomedical Consortium, with support from the Searle Funds at The Chicago Community Trust, C-084; and University of Chicago start-up funds to Y.K. Y.K. is a Brain Research Foundation Fellow. Our sincere apologies to those colleagues whose work could not be included owing to space limitations.

Author information

Authors and Affiliations

Authors

Contributions

M.S.J., A.T.V. and Y.K. developed the concepts and framework of the review and translated these into figures and text.

Corresponding author

Correspondence to Yamuna Krishnan.

Ethics declarations

Competing interests

The authors declare no competing interests

Additional information

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jani, M.S., Veetil, A.T. & Krishnan, Y. Precision immunomodulation with synthetic nucleic acid technologies. Nat Rev Mater 4, 451–458 (2019). https://doi.org/10.1038/s41578-019-0105-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41578-019-0105-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing