401
Views
17
CrossRef citations to date
0
Altmetric
Review

Targeting Toll-like receptor 3 in dendritic cells for cancer immunotherapy

, &
Pages 937-946 | Received 10 Dec 2019, Accepted 26 Mar 2020, Published online: 07 Apr 2020

References

  • Medzhitov R, Janeway CA Jr. Innate immune recognition and control of adaptive immune responses. Semin Immunol. 1998;5:351–353. Review.
  • Janeway CA Jr, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. Review.
  • Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. Review.
  • Yoneyama M, Onomoto K, Jogi M, et al. Viral RNA detection by RIG-I-like receptors. Curr Opin Immunol. 2015;32:48–53.
  • Matsumoto M, Seya T. TLR3: interferon induction by double-stranded RNA including poly(I:C). Adv Drug Del Rev. 2008;60:805–812.
  • Coulie PG, Van den Eynde BJ, van der Bruggen P, et al. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;5:135–146.
  • Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. Review.
  • Savina A, Amigorena S. Phagocytosis and antigen presentation in dendritic cells. Immunol Rev. 2007;219:143–156. Review.
  • Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell. 2001;106:255–258. Review.
  • Heath WR, Belz GT, Behrens GM, et al. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol Rev. 2004;199:9–26.
  • Jongbloed SL, Kassianos AJ, McDonald KJ, et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med. 2010;207:1247–1260.
  • Poulin LF, Salio M, Griessinger E, et al. Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8α+ dendritic cells. J Exp Med. 2010;207:1261–1271.
  • Fuertes MB, Kacha AK, Kline J, et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J Exp Med. 2011;208:2005–2016.
  • Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (Anti–PD-1) in melanoma. N Engl J Med. 2013;369:134–144.
  • Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571.
  • Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567.
  • Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123–135.
  • Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.
  • Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.
  • Gajewski TF, Woo SR, Zha Y, et al. Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment. Curr Opin Immunol. 2013;25:268–276.
  • Joyce JA, Fearon DT. T cell exhaustion, immune privilege, and tumor microenvironment. Science. 2015;348:74–79.
  • Dudek AM, Martin S, Garg AD, et al. Immature, semi-mature, and fully mature dendritic cells: toward a DC-cancer cells interface that augments anticancer immunity. Front Immunol. 2013;4:438.
  • Schietinger A, Greenberg PD. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol. 2014;35:51–60.
  • Pauken KE, Sammons MA, Odorizzi PM, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354:1160–1165.
  • Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436–444.
  • Matsumoto M, Tatematsu M, Nishikawa F, et al. Defined TLR3-specific adjuvant that induces NK and CTL activation without significant cytokine production in vivo. Nat Commun. 2015;6:6280.
  • Ueno H, Klechevsky E, Morita R, et al. Dendritic cell subsets in health and disease. Immunol Rev. 2007;219:118–142. Review.
  • Bachem A, Güttler S, Hartung E, et al. Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J Exp Med. 2010;207:1273–1281.
  • Crozat K, Guiton R, Contreras V, et al. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8α+ dendritic cells. J Exp Med. 2010;207:1283–1292.
  • Joffre OP, Segura E, Savina A, et al. Cross-presentation by dendritic cells. Nat Rev Immunol. 2012;12:557–569. Review.
  • Hornung V, Ellegast J, Kim S, et al. 5ʹ-Triphosphate RNA is the ligand for RIG-I. Science. 2006;10(314):994–997.
  • Pichlmair A, Schulz O, Tan CP, et al. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5ʹ-phosphates. Science. 2006;314:997–1001.
  • Goubau D, Schlee M, Deddouche S, et al. Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5ʹ-diphosphates. Nature. 2014;514:372–375.
  • Malathi K, Saito T, Crochet N, et al. RNase L releases a small RNA from HCV RNA that refolds into a potent PAMP. RNA. 2010;16:2108–2119.
  • Weber F, Wagner V, Rasmussen SB, et al. Double-stranded RNA is produced by positive-stranded RNA viruses and DNA viruses but not in detectable amounts by negative-stranded RNA viruses. J Virol. 2006;80:5059–5064.
  • Kato H, Takeuchi O, Sato S, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441:101–105.
  • Brisse M, Ly H. Comparative structure and function Analysis of the RIG-I-like receptors: RIG-I and MDA5. Front Immunol. 2019;10:1586.
  • Loo YM, Gale M. Immune signaling by RIG-I-like receptors. Immunity. 2011;34:680–692.
  • Belgnaoui SM, Paz S, Hiscott J. Orchestrating the interferon antiviral response through the mitochondrial antiviral signaling (MAVS) adapter. Curr Opin Immunol. 2011;23:564–572. Review.
  • Kato H, Fujita T. RIG-I-like receptors and autoimmune diseases. Curr Opin Immunol. 2015;37:40–45.
  • Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15:760–770.
  • Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–1149.
  • Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signaling. Nature. 2008;455:674–678.
  • Liu Y, Jesus AA, Marrero B, et al. Activated STING in a vascular and pulmonary syndrome. N Engl J Med. 2014;371:507–518.
  • Ho SSW, Zhang WYL, Tan NYJ, et al. The DNA structure-specific endonuclease MUS81 mediates DNA sensor STING-dependent host rejection of prostate cancer cells. Immunity. 2016;44:1177–1189.
  • Woo SR, Fuertes MB, Corrales L, et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41:830–842.
  • Deng L, Liang H, Xu M, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced Type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41:843–852.
  • Ahn J, Xia T, Konno H, et al. Inflammation-driven carcinogenesis is mediated through STING. Nat Commun. 2014;5:5166.
  • Ng KW, Marshall EA, Bell JC, et al. cGAS–STING and cancer: dichotomous roles in tumor immunity and development. Trends Immunol. 2018;39:44–54.
  • Matsumoto M, Kikkawa S, Kohase M, et al. Establishment of a monoclonal antibody against human Toll-like receptor 3 that blocks double-stranded RNA-mediated signaling. Biochem Biophys Res Commun. 2001;293:1364–1369.
  • Matsumoto M, Funami K, Tanabe M, et al. Subcellular localization of Toll-like receptor 3 in human dendritic cells. J Immunol. 2003;171:3154–3162.
  • Alexopoulou L, Holt AC, Medzhitov R, et al. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001;413(6857):732–738.
  • Tatematsu M, Nishikawa F, Seya T, et al. Toll-like receptor 3 recognizes incomplete stem structures in single-stranded viral RNA. Nat Commun. 2013;4:1833.
  • Oshiumi H, Matsumoto M, Funami K, et al. TICAM-1, an adaptor molecule that participates in Toll-like receptor 3-mediated interferon β-induction. Nat Immunol. 2003;4:161–167.
  • Yamamoto M, Sato S, Hemmi H, et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science. 2003;301:640–643.
  • Schulz O, Diebold SS, Chen M, et al. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature. 2005;433:887–892.
  • Azuma M, Ebihara T, Oshiumi H, et al. Cross-priming for antitumor CTL induced by soluble Ag + polyI:C depends on the TICAM-1 pathway in mouse CD11c(+)/CD8α(+) dendritic cells. Oncoimmunol. 2012;1:581–592.
  • Kobayashi KS. van den Elsen PJ. NLRC5: a key regulator of MHC class-I-dependent immune responses. Nat Rev Immunol. 2012;12:813–820.
  • Zehner M, Marschall AL, Bos E, et al. The translocon protein Sec61 mediates antigen transport from endosomes in the cytosol for cross-presentation to CD8+ T cells. Immunity. 2015;42:850–863.
  • Gerner MY, Heltemes-Harris LM, Fife BT, et al. Cutting edge: IL-12 and type I IFN differentially program CD8 T cells for programmed death 1 re-expression levels and tumor control. J Immunol. 2013;191:1011–1015.
  • Azuma M, Takeda Y, Nakajima H, et al. Biphasic function of TLR3 adjuvant on tumor and spleen dendritic cells promotes tumor T cell infiltration and regression in a vaccine model. Oncoimmunol. 2016;5:e1188244.
  • Matsumoto M, Oshiumi H, Seya T. Antiviral responses induced by the TLR3 pathway. Rev Med Virol. 2011;21:67–77.
  • Leonard JN, Ghirlando R, Askins J, et al. The TLR3 signaling complex forms by cooperative receptor dimerization. Proc Natl Acad Sci USA. 2008;105:258–263.
  • Jelinek I, Leonard JN, Price GE, et al. TLR3-specific double-stranded RNA oligonucleotide adjuvants induce dendritic cell cross-presentation, CTL responses, and antiviral protection. J Immunol. 2011;186:2422–2429.
  • Gitlin L, Barchet W, Gilfillan S, et al. Essential role of mda-5 in type I IFN responses to polyriboinosinic: polyribocytidylicacid and encephalomyocarditis picornavirus. Proc Natl Acad Sci USA. 2006;103:8459–8464.
  • Kato H, Takeuchi O, Mikamo-Satoh E, et al. Length-dependent recognition of double-stranded ribonucleic acids by retinoic acid-inducible gene-I and melanoma differentiation-associated gene 5. J Exp Med. 2008;205:1601–1610.
  • Itoh K, Watanabe A, Funami K, et al. The clathrin-mediated endocytic pathway participates in dsRNA-induced IFN-beta production. J Immunol. 2008;181:5522–5529.
  • Watanabe A, Tatematsu M, Saeki K, et al. Raftlin is involved in the nucleocapture complex to induce poly(I:C)-mediated TLR3 activation. J Biol Chem. 2011;286:10702–10711.
  • Nguyen TA, Smith BRC, Tate MD, et al. SIDT2 transports extracellular dsRNA into the cytoplasm for innate immune recognition. Immunity. 2017;47:498–509.e6.
  • Lee HK, Dunzendorfer S, Soldau K, et al. Double-stranded RNA-mediated TLR3 activation is enhanced by CD14. Immunity. 2006;24:153–163.
  • DeWitte-Orr SJ, Collins SE, Bauer CM, et al. An accessory to the ‘Trinity’: SR-As are essential pathogen sensors of extracellular dsRNA, mediating entry and leading to subsequent type I IFN responses. PLoS Pathog. 2010;6:e1000829.
  • Zhou H, Liao J, Aloor J, et al. CD11b/CD18 (Mac-1) is a novel surface receptor for extracellular double-stranded RNA to mediate cellular inflammatory responses. J Immunol. 2013;190:115–125.
  • Shingai M, Ebihara T, Begum NA, et al. Differential type I interferon (IFN) inducing abilities of wild-type vs. vaccine strains of measles virus. J Immunol. 2007;179:6123–6133.
  • Takeda Y, Kataoka K, Yamagishi J, et al. A TLR3-specific adjuvant relieves innate resistance to PD-L1 blockade without cytokine toxicity in tumor vaccine immunotherapy. Cell Rep. 2017;19:1874–1887.
  • Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–915.
  • Mbow ML, De Gregorio E, Valiante NM, et al. New adjuvants for human vaccines. Curr Opin Immunol. 2010;22:411–416.
  • Seya T, Azuma M, Matsumoto M. Targeting TLR3 with no RIG-I/MDA5 activation is effective in immunotherapy for cancer. Expert Opin Ther Targets. 2013;17:533–544. Review.
  • Sabbatini P, Tsuji T, Ferran L, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res. 2012;18:6497–6508.
  • Smith M, Garcia-Martinez E, Pitter MR, et al. Trial watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunol. 2018;7:e1526250.
  • Levine AS, Sivulich M, Wiernik PH, et al. Initial clinical trials in cancer patients of polyriboinosinic-polyribocytidylic acid stabilized with poly-l-lysine, in carboxymethylcellulose [Poly(ICLC)], a highly effective interferon inducer. Cancer Res. 1979;395:1645–1650.
  • Lampkin BC, Levine AS, Levy H, et al. Phase II trial of a complex polyriboinosinic-polyribocytidylic acid with poly-L-lysine and carboxymethyl cellulose in the treatment of children with acute leukemia and neuroblastoma: a report from the children’s cancer study group. Cancer Res. 1985;45:5904–5909.
  • Seya T, Takeda Y, Matsumoto M. A Toll-like receptor 3 (TLR3) agonist ARNAX for therapeutic immunotherapy. Adv Drug Deliv Rev. 2019 July 11;147:37–43.
  • Takeda Y, Yoshida S, Takashima K, et al. Vaccine immunotherapy with ARNAX induces tumor-specific memory T cells and durable anti-tumor immunity in mouse models. Cancer Sci. 2018;1097:2119–2129.
  • Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–221.
  • Zhang JG, Czabotar PE, Policheni AN, et al. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity. 2012;36:646–657.
  • Kroemer G, Galluzzi L, Keppo O, et al. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72.
  • Yatim N, Cullen S, Albert ML. Dying cells actively regulate adaptive immune responses. Nat Rev Immunol. 2017;17:262–275.
  • Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14:1014–1022.
  • Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165:35–44.
  • Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214.
  • De Henau O, Rausch M, Winkler D, et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells. Nature. 2016;539:443–447.
  • DeNardo D, Ruffell B. Macrophages as regulators of tumor immunity and immunotherapy. Nat Rev Immunol. 2019;19:369–382. Review.
  • Rizvi N, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science. 2015;348:124–128.
  • Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413.
  • Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Science. 2016;8:328rv4.
  • Kataoka K, Shiraishi Y, Takeda Y, et al. Aberrant PD-L1 expression via 3ʹ-UTR disruption in multiple cancers. Nature. 2016;534:402–406.
  • Curiel TJ, Wei S, Dong H, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9:562–567.
  • Kleinovink JW, van Hall T, Ossendorp F, et al. PD-L1 immune suppression in cancer: tumor cells or host cells? Oncoimmunol. 2017;6:e1325982.
  • Matsumoto M, Takeda Y, Tatematsu M, et al. TLR3 signal in dendritic cells benefits cancer immunotherapy. Front Immunol. 2017;8:1897. Review.
  • Shime H, Matsumoto M, Oshiumi H, et al. Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors. Proc Natl Acad Sci USA. 2012;109:2066–2071.
  • Shime H, Matsumoto M, Seya T. Double-stranded RNA promotes CTL-independent tumor cytolysis mediated by CD11b+Ly6G+ intratumor myeloid cells through the TICAM-1 signaling pathway. Cell Death Diff. 2016;24:385–396.
  • van der Burg SH, Arens R, Ossendorp F, et al. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer. 2016;16:219–233.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.