Type I and II Interferons in the Anti-Tumor Immune Response
Abstract
:Simple Summary
Abstract
1. Introduction
2. Interferon Production and Signaling
2.1. Type I Interferons
2.2. Type II Interferon
3. Effects of Interferons on the Tumor Microenvironment
3.1. Dendritic Cells
3.2. Macrophages
3.3. Natural Killer Cells
3.4. CD4+ Helper T Cells
3.5. CD8+ Cytotoxic T Cells
3.6. B Cells
3.7. Regulatory T Cells
3.8. Myeloid Derived Suppressor Cells
3.9. Neutrophils
3.10. γδ T Cells
3.11. Tumor Cells
4. Resistance Mechanisms to Interferons
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Candeias, S.M.; Gaipl, U.S. The Immune System in Cancer Prevention, Development and Therapy. Anticancer Agents Med. Chem. 2016, 16, 101–107. [Google Scholar] [CrossRef]
- Galluzzi, L.; Humeau, J.; Buqué, A.; Zitvogel, L.; Kroemer, G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2020, 17, 725–741. [Google Scholar] [CrossRef]
- Dunn, G.P.; Bruce, T.A.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Dunn, G.P.; Koebel, C.M.; Schreiber, R.D. Interferons, immunity and cancer immunoediting. Nat. Rev. Immunol. 2006, 6, 836–848. [Google Scholar] [CrossRef] [PubMed]
- Dunn, G.P.; Bruce, A.T.; Sheehan, K.C.F.; Shankaran, V.; Uppaluri, R.; Bui, J.D.; Diamond, M.S.; Koebel, C.M.; Arthur, C.; White, J.M.; et al. A critical function for type I interferons in cancer immunoediting. Nat. Immunol. 2005, 6, 722–729. [Google Scholar] [CrossRef] [PubMed]
- Ivashkiv, L.B.; Donlin, L.T. Regulation of type I interferon responses. Nat. Rev. Immunol. 2014, 14, 36–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kursunel, M.A.; Esendagli, G. The untold story of IFN--gamma in cancer biology. Cytokine Growth Factor Rev. 2016, 31, 73–81. [Google Scholar] [CrossRef] [PubMed]
- Arimoto, K.I.; Miyauchi, S.; Stoner, S.A.; Fan, J.B.; Zhang, D.E. Negative regulation of type I IFN signaling. J. Leukoc. Biol. 2018, 103, 1099–1116. [Google Scholar] [CrossRef] [PubMed]
- Saleiro, D.; Platanias, L.C. Interferon signaling in cancer. Non-canonical pathways and control of intracellular immune checkpoints. Semin. Immunol. 2019, 43, 101299. [Google Scholar] [CrossRef]
- Platanias, L.C. Mechanisms of type-I- and type-II-interferon-mediated signaling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef]
- De Weerd, N.A.; Nguyen, T. The interferons and their receptors—Distribution and regulation. Immunol. Cell Biol. 2012, 90, 483–491. [Google Scholar] [CrossRef]
- Der, S.D.; Zhou, A.; Williams, B.R.; Silverman, R.H. Identification of genes differentially regulated by interferon α, β, or γ using oligonucleotide arrays. Proc. Natl. Acad. Sci. USA 1998, 95, 15623–15628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheon, H.; Borden, E.C.; Stark, G.R. Interferons and their stimulated genes in the tumor microenvironment. Semin. Oncol. 2014, 41, 156–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pestka, S.; Langer, J.A.; Zoon, K.C.; Samuel, C.E. Interferons and their actions. Annu. Rev. Biochem. 1987, 56, 727–777. [Google Scholar] [CrossRef] [PubMed]
- Pestka, S. The human interferon α species and hybrid proteins. Semin. Oncol. 1997, 24, S9-4–S9-17. [Google Scholar]
- Pestka, S.; Krause, C.D.; Walter, M.R. Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 2004, 202, 8–32. [Google Scholar] [CrossRef]
- Zitvogel, L.; Galluzzi, L.; Kepp, O.; Smyth, M.J.; Droemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 2015, 15, 405–414. [Google Scholar] [CrossRef]
- Fuertes, M.B.; Woo, S.R.; Burnett, B.; Fu, Y.X.; Gajewski, T.F. Type I interferon response and innate immune sensing of cancer. Trends Immunol. 2013, 34, 67–73. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.; Chen, Z.J. Innate immune sensing and signaling of cytosolic nucleic acids. Annu. Rev. Immunol. 2014, 32, 461–488. [Google Scholar] [CrossRef]
- Khoo, L.T.; Chen, L.Y. Role of the cGAS-STING pathway in cancer development and oncotherapeutic approaches. EMBO Rep. 2018, 19, e46935. [Google Scholar] [CrossRef]
- Woo, S.R.; Fuertes, M.B.; Corrales, L.; Spranger, S.; Furdyna, M.J.; Leung, M.Y.K.; Duggan, R.; Wang, Y.; Barber, G.N.; Fitzgerald, K.A.; et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 2014, 41, 830–842. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Sun, L.; Chen, Z.J. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat. Immunol. 2016, 17, 1142–1149. [Google Scholar] [CrossRef]
- Mazewski, C.; Perez, R.E.; Fish, E.N.; Platanias, L.C. Type I Interferon (IFN)-Regulated Activation of Canonical and Non-Canonical Signaling Pathways. Front. Immunol. 2020, 11, 606456. [Google Scholar] [CrossRef]
- Fish, E.N.; Platanias, L.C. Interferon receptor signaling in malignancy: A network of cellular pathways defining biological outcomes. Mol. Cancer Res. 2014, 12, 1691–1703. [Google Scholar] [CrossRef] [Green Version]
- Bernabei, P.; Coccia, E.M.; Rigamonti, L.; Bosticardo, M.; Forni, G.; Pestka, S.; Krause, C.D.; Battisttini, A.; Novelli, F. Interferon-gamma receptor 2 expression as the deciding factor in human T, B, and myeloid cell proliferation or death. J. Leukoc. Biol. 2001, 70, 950–960. [Google Scholar] [PubMed]
- Gough, D.J.; Levy, D.E.; Johnstone, R.W.; Clarke, C.J. IFNgamma signaling-does it mean JAK-STAT? Cytokine Growth Factor Rev. 2008, 19, 383–394. [Google Scholar] [CrossRef]
- Macri, C.; Pang, E.S.; Patton, T.; O’Keeffe, M. Dendritic cell subsets. Semin. Cell Dev. Biol. 2018, 84, 11–21. [Google Scholar] [CrossRef]
- Montoya, M.; Schiavoni, G.; Mattei, F.; Gresser, I.; Belardelli, F.; Borrow, P.; Tough, D.F. Type I interferons produced by dendritic cells promote their phenotypic and functional activation. Blood 2002, 99, 3263–3271. [Google Scholar] [CrossRef]
- Medrano, R.F.V.; Hunger, A.; Mendonca, S.A.; Barbuto, J.A.M.; Strauss, B.E. Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy. Oncotarget 2017, 8, 71249–71284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santini, S.M.; Lapenta, C.; Logozzi, M.; Parlato, S.; Spada, M.; Di Pucchio, T.; Belardelli, F. Type I interferon as a powerful adjuvant for monocyte-derived dendritic cell development and activity in vitro and in Hu-PBL-SCID mice. J. Exp. Med. 2000, 191, 1777–1788. [Google Scholar] [CrossRef] [PubMed]
- Schiavoni, G.; Mattei, F.; Gabriele, L. Type I Interferons as Stimulators of DC-Mediated Cross-Priming: Impact on Anti-Tumor Response. Front. Immunol. 2013, 4, 483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lattanzi, L.; Rozera, C.; Marescotti, D.; D’Agostino, G.; Santodonato, L.; Cellini, S.; Belardelli, F.; Gavioli, R.; Ferrantini, M. IFN-alpha boosts epitope cross-presentation by dendritic cells via modulation of proteasome activity. Immunobiology 2011, 216, 537–547. [Google Scholar] [CrossRef] [PubMed]
- Spadaro, F.; Lapenta, C.; Donati, S.; Abalsamo, L.; Barnaba, V.; Belardelli, F.; Santini, S.M.; Ferrantini, M. IFN-alpha enhances cross-presentation in human dendritic cells by modulating antigen survival, endocytic routing, and processing. Blood 2012, 119, 1407–1417. [Google Scholar] [CrossRef] [Green Version]
- Parlato, S.; Santini, S.M.; Lapenta, C.; Di Pucchio, T.; Logozzi, M.; Spada, M.; Giammarioli, A.M.; Malorni, W.; Fais, S.; Belardelli, F. Expression of CCR-7, MIP-3beta, and Th-1 chemokines in type I IFN-induced monocyte-derived dendritic cells: Importance for the rapid acquisition of potent migratory and functional activities. Blood 2001, 98, 3022–3029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Longhi, M.P.; Trumpfheller, C.; Idoyaga, J.; Caskey, M.; Matos, I.; Kluger, C.; Salazar, A.M.; Colonna, M.; Steinman, R.M. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J. Exp. Med. 2009, 206, 1589–1602. [Google Scholar] [CrossRef]
- Arico, E.; Castiello, L.; Capone, I.; Gabriele, L.; Belardelli, F. Type I interferons and cancer: An evolving story demanding novel clinical applications. Cancers 2019, 11, 1943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mattei, F.; Schiavoni, G.; Belardelli, F.; Tough, D.F. IL-15 is expressed by dendritic cells in response to type I IFN, double-stranded RNA, or lipopolysaccharide and promotes dendritic cell activation. J. Immunol. 2001, 167, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
- McKenna, K.; Beignon, A.S.; Bhardwaj, N. Plasmacytoid dendritic cells: Linking innate and adaptive immunity. J. Virol. 2005, 79, 17–27. [Google Scholar] [CrossRef] [Green Version]
- Dai, J.H.; Megjugorac, N.J.; Amrute, S.B.; Fitzgerald-Bocarsly, P. Regulation of IFN regulatory factor-7 and IFN-alpha production by enveloped virus and lipopolysaccharide in human plasmacytoid dendritic cells. J. Immunol. 2004, 173, 1535–1548. [Google Scholar] [CrossRef] [Green Version]
- Le Mercier, I.; Poujol, D.; Sanlaville, A.; Sisirak, V.; Gobert, M.; Durand, I.; Dubois, B.; Treilleux, I.; Marvel, J.; Vlach, J.; et al. Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment. Cancer Res. 2013, 73, 4629–4640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veglia, F.; Gabrilovich, D.I. Dendritic cells in cancer: The role revisited. Curr. Opin. Immunol. 2017, 45, 43–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sisirak, V.; Faget, J.; Gobert, M.; Goutagny, N.; Vey, N.; Treilleux, I.; Renaudineau, S.; Poyet, G.; Labidi-Galy, S.I.; Goddard-Leon, S.; et al. Impaired IFN-α production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 2012, 72, 5188–5197. [Google Scholar] [CrossRef] [Green Version]
- Garris, C.S.; Arlauckas, S.P.; Kohler, R.H.; Trefny, M.P.; Garren, S.; Piot, C.; Engblom, C.; Pfirschke, C.; Siwicki, M.; Gungabeesoon, J.; et al. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-γ and IL-12. Immunity 2018, 49, 1148–1161.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lövgren, T.; Sarhan, D.; Truxová, I.; Choudhary, B.; Maas, R.; Melief, J.; Nyström, M.; Edbäck, U.; Vermeij, R.; Scurti, G.; et al. Enhanced stimulation of human tumor-specific T cells by dendritic cells matured in the presence of interferon-γ and multiple toll-like receptor agonists. Cancer Immunol. Immunother. 2017, 66, 1333–1344. [Google Scholar] [CrossRef] [Green Version]
- Alspach, E.; Lussier, D.M.; Schrieber, R.D. Interferon gamma and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harb. Perspect. Biol. 2019, 11, a028480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheng, K.C.; Day, S.; Wright, M.D.; Stojanovska, L.; Apostolopoulos, V. Enhanced dendritic cell-mediated antigen-specific CD4+ T cell responses: IFN-gamma aids TLR stimulation. J. Drug Deliv. 2013, 2013, 516749. [Google Scholar] [CrossRef] [PubMed]
- Sprooten, J.; Agostinis, P.; Garg, A.D. Type I interferons and dendritic cells in cancer immunotherapy. Int. Rev. Cell Mol. Biol. 2019, 348, 217–262. [Google Scholar] [PubMed]
- Hervas-Stubbs, S.; Perez-Gracia, J.L.; Rouzaut, A.; Sanmamed, M.F.; Le Bon, A.; Melero, I. Direct effects of type I interferons on cells of the immune system. Clin. Cancer Res. 2011, 17, 2619–2627. [Google Scholar] [CrossRef] [Green Version]
- Schreiner, B.; Mitsdoerffer, M.; Kieseier, B.C.; Chen, L.; Hartung, H.P.; Weller, M.; Wiendl, H. Interferon-beta enhances monocyte and dendritic cell expression of B7-H1 (PD-L1), a strong inhibitor of autologous T-cell activation: Relevance for the immune modulatory effect in multiple sclerosis. J. Neuroimmunol. 2004, 155, 172–182. [Google Scholar] [CrossRef]
- Snell, L.M.; McGaha, T.L.; Brooks, D.G. Type I Interferon in Chronic Virus Infection and Cancer. Trends Immunol. 2017, 38, 542–557. [Google Scholar] [CrossRef]
- Nirschl, C.J.; Suarez-Farinas, M.; Izar, B.; Prakadan, S.; Dannenfelser, R.; Tirosh, I.; Liu, Y.; Zhu, Q.; Devi, K.S.P.; Carroll, S.L.; et al. IFNγ-Dependent Tissue-Immune Homeostasis Is Co-opted in the Tumor Microenvironment. Cell 2017, 170, 127–141.e115. [Google Scholar] [CrossRef] [Green Version]
- Jurgens, B.; Hainz, U.; Fuchs, D.; Felzmann, T.; Heitger, A. Interferon-γ-triggered indoleamine 2,3-dioxygenase competence in human monocyte-derived dendritic cells induces regulatory activity in allogeneic T cells. Blood 2009, 114, 3235–3243. [Google Scholar] [CrossRef] [Green Version]
- Noh, K.T.; Son, K.H.; Jung, I.D.; Kang, T.H.; Choi, C.H.; Park, Y.M. Glycogen Synthase Kinase-3β (GSK-3β) Inhibition Enhances Dendritic Cell-based Cancer Vaccine Potency via Suppression of Interferon-γ-induced Indoleamine 2,3-Dioxygenase Expression. J. Biol. Chem. 2015, 290, 12394–12402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sabado, R.L.; Balan, S.; Bhardwaj, N. Dendritic cell-based immunotherapy. Cell Res. 2017, 27, 74–95. [Google Scholar] [CrossRef] [Green Version]
- Bialek-Waldmann, J.K.; Heuser, M.; Ganser, A.; Stripecke, R. Monocytes reprogrammed with lentiviral vectors co-expressing GM-CSF, IFN-α2 and antigens for personalized immune therapy of acute leukemia pre- or post-stem cell transplantation. Cancer Immunol. Immunother. 2019, 68, 1891–1899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zanker, D.J.; Spurling, A.J.; Brockwell, N.K.; Owen, K.L.; Zakhour, J.M.; Robinson, T.; Duivenvoorden, H.M.; Hertzog, P.J.; Mullins, S.R.; Wilkinson, R.W.; et al. Intratumoral administration of the Toll-like receptor 7/8 agonist 3M-052 enhances interferon-driven tumor immunogenicity and suppresses metastatic spread in preclinical triple-negative breast cancer. Clin. Transl. Immunol. 2020, 9, e1177. [Google Scholar] [CrossRef]
- Graham, J.P.; Authie, P.; Palucka, A.K.; Zurawski, G. Targeting interferon-alpha to dendritic cells enhances a CD8+ T cell response to a human CD40-targeted cancer vaccine. Vaccine 2017, 35 Pt B, 4532–4539. [Google Scholar] [CrossRef]
- Sköld, A.E.; Mathan, T.S.M.; van Beek, J.J.P.; Flórez-Grau, G.; van den Beukel, M.D.; Sittig, S.P.; Wimmers, F.; Bakdash, G.; Schreibelt, G.; de Vries, I.J.M. Naturally produced type I IFNs enhance human myeloid dendritic cell maturation and IL-12p70 production and mediate elevated effector functions in innate and adaptive immune cells. Cancer Immunol. Immunother. 2018, 67, 1425–1436. [Google Scholar] [CrossRef] [Green Version]
- Gordy, J.T.; Luo, K.; Kapoor, A.; Kim, E.S.; Ayeh, S.K.; Karakousis, P.C.; Markham, R.B. Treatment with an immature dendritic cell-targeting vaccine supplemented with IFN-α and an inhibitor of DNA methylation markedly enhances survival in a murine melanoma model. Cancer Immunol. Immunother. 2020, 69, 569–580. [Google Scholar] [CrossRef]
- Sheng, L.; Chen, X.; Wang, Q.; Lyu, S.; Li, P. Interferon-α2b enhances survival and modulates transcriptional profiles and the immune response in melanoma patients treated with dendritic cell vaccines. Biomed. Pharmacother. 2020, 125, 109966. [Google Scholar] [CrossRef] [PubMed]
- Le Naour, J.; Zitvogel, L.; Galluzzi, L.; Vacchelli, E.; Kroemer, G. Trial watch: STING agonists in cancer therapy. Oncoimmunology 2020, 9, 1777624. [Google Scholar] [CrossRef]
- Zhu, Y.; An, X.; Zhang, X.; Qiao, Y.; Zheng, T.; Li, X. STING: A master regulator in the cancer-immunity cycle. Mol. Cancer 2019, 18, 152. [Google Scholar] [CrossRef] [Green Version]
- Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
- Parker, B.S.; Rautela, J.; Hertzog, P.J. Antitumour actions of interferons: Implications for cancer therapy. Nat. Rev. Cancer 2016, 16, 131–144. [Google Scholar] [CrossRef]
- Müller, E.; Speth, M.; Christopoulos, P.F.; Lunde, A.; Avdagic, A.; Øynebråten, I.; Corthay, A. Both Type I and Type II Interferons Can Activate Antitumor M1 Macrophages When Combined with TLR Stimulation. Front. Immunol. 2018, 9, 2520. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Chakravarty, S.D.; Ivashkiv, L.B. Regulation of interferon and Toll-like receptor signaling during macrophage activation by opposing feedforward and feedback inhibition mechanisms. Immunol. Rev. 2008, 226, 41–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeong, S.K.; Yang, K.; Park, Y.S.; Choi, Y.J.; Oh, S.J.; Lee, C.W.; Lee, K.Y.; Jeong, M.H.; Jo, W.S. Interferon gamma induced by resveratrol analog, HS--1793, reverses the properties of tumor associated macrophages. Int. Immunopharmacol. 2014, 22, 303–310. [Google Scholar] [CrossRef] [PubMed]
- Su, X.; Yu, Y.; Zhong, Y.; Giannopoulou, E.G.; Hu, X.; Liu, H.; Cross, J.R.; Rätsch, G.; Rice, C.M.; Ivashkiv, L.B. Interferon-γ regulates cellular metabolism and mRNA translation to potentiate macrophage activation. Nat. Immunol. 2015, 16, 838–849. [Google Scholar] [CrossRef] [Green Version]
- Trost, M.; English, L.; Lemieux, S.; Courcelles, M.; Desjardins, M.; Thibault, P. The phagosomal proteome in interferon-gamma-activated macrophages. Immunity 2009, 30, 143–154. [Google Scholar] [CrossRef] [Green Version]
- Siveen, K.S.; Kuttan, G. Role of macrophages in tumour progression. Immunol. Lett. 2009, 123, 97–102. [Google Scholar] [CrossRef]
- Escobar, G.; Moi, D.; Ranghetti, A.; Ozkal-Baydin, P.; Squadrito, M.L.; Kajaste-Rudnitski, A.; Bondanza, A.; Gentner, B.; De Palma, M.; Mazzieri, R.; et al. Genetic engineering of hematopoiesis for targeted IFN-alpha delivery inhibits breast cancer progression. Sci. Transl. Med. 2014, 6, 217ra3. [Google Scholar] [CrossRef] [PubMed]
- Salvagno, C.; Ciampricotti, M.; Tuit, S.; Hau, C.S.; van Weverwijk, A.; Coffelt, S.B.; Kersten, K.; Vrijland, K.; Kos, K.; Ulas, T.; et al. Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response. Nat. Cell Biol. 2019, 21, 511–521. [Google Scholar] [CrossRef] [PubMed]
- Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, M.P.; Weissman, I.L.; Majeti, R. The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 2012, 24, 225–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Silva, S.; Fromm, G.; Shuptrine, C.W.; Johannes, K.; Patel, A.; Yoo, K.J.; Huang, K.; Schreiber, T.H. CD40 Enhances Type I Interferon Responses Downstream of CD47 Blockade, Bridging Innate and Adaptive Immunity. Cancer Immunol. Res. 2020, 8, 230–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haabeth, O.A.; Lorvik, K.B.; Hammarström, C.; Donaldson, I.M.; Haraldsen, G.; Bogen, B.; Corthay, A. Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer. Nat. Commun. 2011, 2, 240. [Google Scholar] [CrossRef] [Green Version]
- Spear, P.; Barber, A.; Rynda-Apple, A.; Sentman, C.L. Chimeric antigen receptor T cells shape myeloid cell function within the tumor microenvironment through IFN-gamma and GM-CSF. J. Immunol. 2012, 188, 6389–6398. [Google Scholar] [CrossRef]
- Muller, L.; Aigner, P.; Stoiber, D. Type I interferons and natural killer cell regulation in cancer. Front. Immunol. 2017, 8, 304. [Google Scholar] [CrossRef] [Green Version]
- Stetson, D.B.; Mohrs, M.; Reinhardt, R.L.; Baron, J.L.; Wang, Z.E.; Gapin, L.; Kronenberg, M.; Locksley, R.M. Constitutive cytokine mRNAs mark natural killer (NK) and NK T cells poised for rapid effector function. J. Exp. Med. 2003, 198, 1069–1076. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, K.B.; Salazar-Mather, T.P.; Dalod, M.Y.; Van Deusen, J.B.; Wei, X.Q.; Liew, F.Y.; Caligiuri, M.A.; Durbin, J.E.; Biron, C.A. Coordinated and distinct roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J. Immunol. 2002, 169, 4279–4287. [Google Scholar] [CrossRef] [Green Version]
- Degli-Esposti, M.A.; Smyth, M.J. Close encounters of different kinds: Dendritic cells and NK cells take centre stage. Nat. Rev. Immunol. 2005, 5, 112–124. [Google Scholar] [CrossRef]
- Takashima, K.; Takeda, Y.; Oshiumi, H.; Shime, H.; Okabe, M.; Ikawa, M.; Matsumoto, M.; Seya, T. STING in tumor and host cells cooperatively work for NK cell-mediated tumor growth retardation. Biochem. Biophys. Res. Commun. 2016, 478, 1764–1771. [Google Scholar] [CrossRef]
- Nicolai, C.J.; Wolf, N.; Chang, I.C.; Kirn, G.; Marcus, A.; Ndubaku, C.O.; McWhirter, S.M.; Raulet, D.H. NK cells mediate clearance of CD8+ T cell-resistant tumors in response to STING agonists. Sci. Immunol. 2020, 5, eaaz2738. [Google Scholar] [CrossRef]
- Zanker, D.J.; Owen, K.L.; Baschuk, N.; Spurling, A.J.; Parker, B.S. Loss of type I IFN responsiveness impairs natural killer cell antitumor activity in breast cancer. Cancer Immunol. Immunother. 2021. online ahead of print. [Google Scholar] [CrossRef]
- Zhao, Y.; Chen, W.; Zhu, W.; Meng, H.; Chen, J.; Zhang, J. Overexpression of interferon regulatory factor 7 (IRF7) reduces bone metastasis of prostate cancer cells in mice. Oncol. Res. 2017, 25, 511–522. [Google Scholar] [CrossRef]
- Swann, J.B.; Hayakawa, Y.; Zerafa, N.; Sheehan, K.C.; Scott, B.; Schreiber, R.D.; Hertzog, P.; Smyth, M.J. Type I IFN contributes to NK cell homeostasis, activation, and antitumor function. J. Immunol. 2007, 178, 7540–7549. [Google Scholar] [CrossRef]
- Rautela, J.; Baschuk, N.; Slaney, C.Y.; Jayatilleke, K.M.; Xiao, K.; Bidwell, B.N.; Lucas, E.C.; Hawkins, E.D.; Lock, P.; Wong, C.S.; et al. Loss of host type-I IFN signaling accelerates metastasis and impairs NK-cell antitumor function in multiple models of breast cancer. Cancer Immunol. Res. 2015, 3, 1207–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bidwell, B.N.; Slaney, C.Y.; Withana, N.P.; Forster, S.; Cao, Y.; Loi, S.; Andrews, D.; Mikeska, T.; Mangan, N.E.; Samarajiwa, S.A.; et al. Silencing of IRF7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 2012, 18, 1224–1231. [Google Scholar] [CrossRef] [PubMed]
- Oh, H.; Kim, M.H.; Choi, S.J.; Ban, Y.H.; Lee, H.K.; Shin, E.C.; Lee, K.M.; Ha, S.J. Sustained type I interferon reinforces NK cell-mediated cancer immunosurveillance during chronic virus infection. Cancer Immunol. Res. 2019, 7, 584–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wendel, M.; Galani, I.E.; Sur-Payer, E.; Cerwenka, A. Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res. 2008, 68, 8437–8445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellucci, R.; Martin, A.; Bommarito, D.; Wang, K.; Hansen, S.H.; Freeman, G.J.; Ritz, J. Interferon-γ-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology 2015, 4, e1008824. [Google Scholar] [CrossRef] [PubMed]
- Aquino-Lopez, A.; Senyukov, V.V.; Vlasic, Z.; Kleinerman, E.S.; Lee, D.A. Interferon gamma induces changes in natural killer (NK) cell ligand expression and alters NK cell-mediated lysis of pediatric cancer cell lines. Front. Immunol. 2017, 8, 391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luckheeram, R.V.; Zhou, R.; Verma, A.D.; Xia, B. CD4+T cells: Differentiation and functions. Clin. Dev. Immunol. 2012, 2012, 925135. [Google Scholar] [CrossRef] [Green Version]
- Brinkmann, V.; Geiger, T.; Alkan, S.; Heusser, C.H. Interferon alpha increases the frequency of interferon gamma-producing human CD4+ T cells. J. Exp. Med. 1993, 178, 1655–1663. [Google Scholar] [CrossRef]
- Schulz, E.G.; Mariani, L.; Radbruch, A.; Hofer, T. Sequential polarization and imprinting of type 1 T helper lymphocytes by interferon-gamma and interleukin-12. Immunity 2009, 30, 673–683. [Google Scholar] [CrossRef] [Green Version]
- Dagenais-Lussier, X.; Loucif, H.; Murira, A.; Laulhé, X.; Stäger, S.; Lamarre, A.; van Grevenynghe, J. Sustained IFN-I Expression during Established Persistent Viral Infection: A “Bad Seed” for Protective Immunity. Viruses 2017, 10, 12. [Google Scholar] [CrossRef] [Green Version]
- Zhang, N.; Bevan, M.J. CD8+ T cells: Foot soldiers of the immune system. Immunity 2011, 35, 161–168. [Google Scholar] [CrossRef] [Green Version]
- Agarwal, P.; Raghavan, A.; Nandiwada, S.L.; Curtsinger, J.M.; Bohjanen, P.R.; Mueller, D.L.; Mescher, M.F. Gene regulation and chromatin remodeling by IL-12 and type I IFN in programming for CD8 T cell effector function and memory. J. Immunol. 2009, 183, 1695–1704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fuertes, M.B.; Kacha, A.K.; Kline, J.; Woo, S.R.; Kranz, D.M.; Murphy, K.M.; Gajewski, T.F. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J. Exp. Med. 2011, 208, 2005–2016. [Google Scholar] [CrossRef] [Green Version]
- Curtsinger, J.M.; Mescher, M.F. Inflammatory cytokines as a third signal for T cell activation. Curr. Opin. Immunol. 2010, 22, 333–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef] [Green Version]
- Hiroishi, K.; Tüting, T.; Lotze, M.T. IFN-alpha-expressing tumor cells enhance generation and promote survival of tumor-specific CTLs. J. Immunol. 2000, 164, 567–572. [Google Scholar] [CrossRef] [Green Version]
- Katlinski, K.V.; Gui, J.; Katlinskaya, Y.V.; Ortiz, A.; Chakraborty, R.; Bhattacharya, S.; Carbone, C.J.; Beiting, D.P.; Girondo, M.A.; Peck, A.R.; et al. Inactivation of Interferon Receptor Promotes the Establishment of Immune Privileged Tumor Microenvironment. Cancer Cell 2017, 31, 194–207. [Google Scholar] [CrossRef] [Green Version]
- Lu, C.; Klement, J.D.; Ibrahim, M.L.; Xiao, W.; Redd, P.S.; Nayak-Kapoor, A.; Zhou, G.; Liu, K. Type I interferon suppresses tumor growth through activating the STAT3-granzyme B pathway in tumor infiltrating cytotoxic T lymphocytes. J. Immunother. Cancer 2019, 7, 157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Campos, J.; Gallotta, M.; Gong, M.; Crain, C.; Naik, E.; Coffman, R.L.; Guiducci, C. Intratumoral injection of a CpG oligonucleotide reverts resistance to PD-1 blockade by expanding multifunctional CD8+ T cells. Proc. Natl. Acad. Sci. USA 2016, 113, E7240–E7249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sumida, T.S.; Dulberg, S.; Schupp, J.; Stillwell, H.A.; Axisa, P.P.; Comi, M.; Lincoln, M.; Unterman, A.; Kaminski, N.; Madi, A.; et al. Type I Interferon Transcriptional Network Regulates Expression of Coinhibitory Receptors in Human T cells. bioRxiv 2020. [Google Scholar] [CrossRef]
- Zimmerman, M.; Yang, D.; Hu, X.; Liu, F.; Singh, N.; Browning, D.; Ganapathy, V.; Chandler, P.; Choubey, D.; Abrams, S.I.; et al. IFN-gamma upregulates survivin and Ifi202 expression to induce survival and proliferation of tumor-specific T cells. PLoS ONE 2010, 5, e14076. [Google Scholar] [CrossRef]
- Castro, F.; Cardoso, A.P.; Goncalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-gamma at the crossroads of tumor immune surveillance or evasion. Front. Immunol. 2018, 9, 847. [Google Scholar] [CrossRef] [Green Version]
- Pai, C.S.; Huang, J.T.; Lu, X.; Simons, D.M.; Park, C.; Chang, A.; Tamaki, W.; Liu, E.; Roybal, K.T.; Seagal, J.; et al. Clonal deletion of tumor-specific T cells by interferon-gamma confers therapeutic resistance to combination immune checkpoint blockade. Immunity 2019, 50, 477–492.e478. [Google Scholar] [CrossRef] [Green Version]
- Gujer, C.; Sandgren, K.J.; Douagi, I.; Adams, W.C.; Sundling, C.; Smed-Sorensen, A.; Seder, R.A.; Karlsson Hedestam, G.B.; Lore, K. IFN-alpha produced by human plasmacytoid dendritic cells enhances T cell-dependent naive B cell differentiation. J. Leukoc. Biol. 2011, 89, 811–821. [Google Scholar] [CrossRef] [Green Version]
- Braun, D.; Caramalho, I.; Demengeot, J. IFN-alpha/beta enhances BCR-dependent B cell responses. Int. Immunol. 2002, 14, 411–419. [Google Scholar] [CrossRef]
- Le Bon, A.; Thompson, C.; Kamphuis, E.; Durand, V.; Rossmann, C.; Kalinke, U.; Tough, D.F. Cutting edge: Enhancement of antibody responses through direct stimulation of B and T cells by type I IFN. J. Immunol. 2006, 176, 2074–2078. [Google Scholar] [CrossRef] [Green Version]
- Schleimann, M.H.; Kobberø, M.L.; Vibholm, L.K.; Kjær, K.; Giron, L.B.; Busman-Sahay, K.; Chan, C.N.; Nekorchuk, M.; Schmidt, M.; Wittig, B.; et al. TLR9 agonist MGN1703 enhances B cell differentiation and function in lymph nodes. EBioMedicine 2019, 45, 328–340. [Google Scholar] [CrossRef]
- Smith, M.; García-Martínez, E.; Pitter, M.R.; Fucikova, J.; Spisek, R.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology 2018, 7, e1526250. [Google Scholar] [CrossRef] [PubMed]
- Bao, Y.; Liu, X.; Han, C.; Xu, S.; Xie, B.; Zhang, Q.; Gu, Y.; Hou, J.; Qian, L.; Qian, C.; et al. Identification of IFN-γ-producing innate B cells. Cell Res. 2014, 24, 161–176. [Google Scholar] [CrossRef] [PubMed]
- Yun, S.O.; Shin, H.Y.; Kang, C.Y.; Kang, H.J. Generation of antigen-specific cytotoxic T lymphocytes with activated B cells. Cytotherapy 2017, 19, 119–127. [Google Scholar] [CrossRef]
- Wennhold, K.; Simabukuro-Vornhagen, A.; von Bergwelt-Baildon, M. B cell-based cancer immunotherapy. Transfus. Med. Hemother. 2019, 46, 36–46. [Google Scholar] [CrossRef]
- Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
- Pace, L.; Vitale, S.; Dettori, B.; Palombi, C.; La Sorsa, V.; Belardelli, F.; Proietti, E.; Doria, G. APC activation by IFN-alpha decreases regulatory T cell and enhances Th cell functions. J. Immunol. 2010, 184, 5969–5979. [Google Scholar] [CrossRef] [Green Version]
- Hashimoto, H.; Ueda, R.; Narumi, K.; Heike, Y.; Yoshida, T.; Aoki, K. Type I IFN gene delivery suppresses regulatory T cells within tumors. Cancer Gene Ther. 2014, 21, 532–541. [Google Scholar] [CrossRef]
- Hirata, A.; Hashimoto, H.; Shibasaki, C.; Narumi, K.; Aoki, K. Intratumoral IFN-α gene delivery reduces tumor-infiltrating regulatory T cells through the downregulation of tumor CCL17 expression. Cancer Gene Ther. 2019, 26, 334–343. [Google Scholar] [CrossRef]
- Gangaplara, A.; Martens, C.; Dahlstrom, E.; Metidji, A.; Gokhale, A.S.; Glass, D.D.; Lopez-Ocasio, M.; Baur, R.; Kanakabandi, K.; Porcella, S.F.; et al. Type I interferon signaling attenuates regulatory T cell function in viral infection and in the tumor microenvironment. PLoS Pathog. 2018, 14, e1006985. [Google Scholar] [CrossRef] [Green Version]
- Anz, D.; Rapp, M.; Eiber, S.; Koelzer, V.H.; Thaler, R.; Haubner, S.; Knott, M.; Nagel, S.; Golic, M.; Wiedemann, G.M.; et al. Suppression of intratumoral CCL22 by type I interferon inhibits migration of regulatory T cells and blocks cancer progression. Cancer Res. 2015, 75, 4483–4493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.E.; Li, X.; Kim, J.C.K.; Lee, J.; González-Navajas, J.M.; Hong, S.H.; Park, I.K.; Rhee, J.H.; Raz, E. Type I interferons maintain Foxp3 expression and T-regulatory cell functions under inflammatory conditions in mice. Gastroenterology 2012, 143, 145–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Metidji, A.; Rieder, S.A.; Glass, D.D.; Cremer, I.; Punkosdy, G.A.; Shevach, E.M. IFN-α/β receptor signaling promotes regulatory T cell development and function under stress conditions. J. Immunol. 2015, 194, 4265–4276. [Google Scholar] [CrossRef] [Green Version]
- Overacre-Delgoffe, A.E.; Chikina, M.; Dadey, R.E.; Yano, H.; Brunazzi, E.A.; Shayan, G.; Horne, W.; Moskovitz, J.M.; Kolls, J.K.; Sander, C.; et al. Interferon-gamma drives Treg fragility to promote anti-tumor immunity. Cell 2017, 169, 1130–1141.e1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, X.; Leonard, K.; Collins, L.I.; Cai, S.F.; Mayer, J.C.; Payton, J.E.; Walter, M.J.; Piwnica-Worms, D.; Schreiber, R.D.; Ley, T.J. Interleukin 12 stimulates IFN-gamma-mediated inhibition of tumor-induced regulatory T-cell proliferation and enhances tumor clearance. Cancer Res. 2009, 69, 8700–8709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nishikawa, H.; Kato, T.; Tawara, I.; Ikeda, H.; Kuribayashi, K.; Allen, P.M.; Schreiber, R.D.; Old, L.J.; Shiku, H. IFN-γ controls the generation/activation of CD4+CD25+ regulatory T cells in antitumor immune response. J. Immunol. 2005, 175, 4433–4440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, Q.; Jin, J.; Xiao, Y.; Zhou, X.; Hu, H.; Cheng, X.; Kazimi, N.; Ullrich, S.E.; Sun, S.C. T Cell Intrinsic USP15 deficiency promotes excessive IFN-gamma production and an immunosuppressive tumor microenvironment in MCA-induced fibrosarcoma. Cell Rep. 2015, 13, 2470–2479. [Google Scholar] [CrossRef] [Green Version]
- Ohue, Y.; Nishikawa, H. Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 2019, 110, 2080–2089. [Google Scholar] [CrossRef]
- Wang, D.; Quiros, J.; Mahuron, K.; Pai, C.C.; Ranzani, V.; Young, A.; Silveria, S.; Harwin, T.; Abnousian, A.; Pagani, M.; et al. Targeting EZH2 Reprograms Intratumoral Regulatory T Cells to Enhance Cancer Immunity. Cell Rep. 2018, 23, 3262–3274. [Google Scholar] [CrossRef]
- Mundy-Bosse, B.L.; Lesinski, G.B.; Jaime-Ramirez, A.C.; Benninger, K.; Khan, M.; Kuppusamy, P.; Guenterberg, K.; Kondadasula, S.V.; Chaudhury, A.R.; La Perle, K.M.; et al. Myeloid-derived suppressor cell inhibition of the IFN response in tumor-bearing mice. Cancer Res. 2011, 71, 5101–5110. [Google Scholar] [CrossRef] [Green Version]
- Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef] [Green Version]
- Lu, C.; Redd, P.S.; Lee, J.R.; Savage, N.; Liu, K. The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells. Oncoimmunology 2016, 5, e1247135. [Google Scholar] [CrossRef] [Green Version]
- Xiao, W.; Klement, J.D.; Lu, C.; Ibrahim, M.L.; Liu, K. IFNAR1 controls autocrine type I IFN regulation of PD-L1 expression in myeloid-derived suppressor cells. J. Immunol. 2018, 201, 264–277. [Google Scholar] [CrossRef] [Green Version]
- Liang, H.; Deng, L.; Hou, Y.; Meng, X.; Huang, X.; Rao, E.; Zheng, W.; Mauceri, H.; Mack, M.; Xu, M.; et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat. Commun. 2017, 8, 1736. [Google Scholar] [CrossRef] [Green Version]
- Hong, E.H.; Cho, J.; Ahn, J.H.; Kwon, B.E.; Kweon, M.N.; Seo, S.U.; Yoon, B.I.; Chang, S.Y.; Ko, H.J. Plasmacytoid dendritic cells regulate colitis-associated tumorigenesis by controlling myeloid-derived suppressor cell infiltration. Cancer Lett. 2020, 493, 102–112. [Google Scholar] [CrossRef] [PubMed]
- Zoglmeier, C.; Bauer, H.; Moerenberg, D.; Wedekind, G.; Bittner, P.; Sandholzer, N.; Rapp, M.; Anz, D.; Endres, S.; Bourquin, C. CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice. Clin. Cancer Res. 2011, 17, 1765–1775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shime, H.; Maruyama, A.; Yoshida, S.; Takeda, Y.; Matsumoto, M.; Seya, T. Toll-like receptor 2 ligand and interferon-gamma suppress anti-tumor T cell responses by enhancing the immunosuppressive activity of monocytic myeloid-derived suppressor cells. Oncoimmunology 2017, 7, e1373231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Greifenberg, V.; Ribechini, E.; Rossner, S.; Lutz, M.B. Myeloid-derived suppressor cell activation by combined LPS and IFN-gamma treatment impairs DC development. Eur. J. Immunol. 2009, 39, 2865–2876. [Google Scholar] [CrossRef] [PubMed]
- Danelli, L.; Frossi, B.; Gri, G.; Mion, F.; Guarnotta, C.; Bongiovanni, L.; Tripodo, C.; Mariuzzi, L.; Marzinotto, S.; Rigoni, A.; et al. Mast cells boost myeloid-derived suppressor cell activity and contribute to the development of tumor-favoring microenvironment. Cancer Immunol. Res. 2015, 3, 85–95. [Google Scholar] [CrossRef] [Green Version]
- Huang, B.; Pan, P.Y.; Li, Q.; Sato, A.I.; Levy, D.E.; Bromberg, J.; Divino, C.M.; Chen, S.H. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006, 66, 1123–1131. [Google Scholar] [CrossRef] [Green Version]
- Burke, J.D.; Young, H.A. IFN-γ: A cytokine at the right time, is in the right place. Semin. Immunol. 2019, 43, 101280. [Google Scholar] [CrossRef] [PubMed]
- Medina-Echeverz, J.; Haile, L.A.; Zhao, F.; Gamrekelashvili, J.; Ma, C.; Métais, J.Y.; Dunbar, C.E.; Kapoor, V.; Manns, M.P.; Korangy, F.; et al. IFN-gamma regulates survival and function of tumor-induced CD11b+ Gr-1high myeloid derived suppressor cells by modulating the anti-apoptotic molecule Bcl2a1. Eur. J. Immunol. 2014, 44, 2457–2467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andzinski, L.; Kasnitz, N.; Stahnke, S.; Wu, C.F.; Gereke, M.; von Köckritz-Blickwede, M.; Schilling, B.; Brandau, S.; Weiss, S.; Jablonska, J. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int. J. Cancer 2016, 138, 1982–1993. [Google Scholar] [CrossRef] [PubMed]
- Kalafati, L.; Kourtzelis, I.; Schulte-Schrepping, J.; Li, X.; Hatzioannou, A.; Grinenko, T.; Hagag, E.; Sinha, A.; Has, C.; Dietz, S.; et al. Innate Immune Training of Granulopoiesis Promotes Anti-tumor Activity. Cell 2020, 183, 771–785.e12. [Google Scholar] [CrossRef]
- Jablonska, J.; Wu, C.F.; Andzinski, L.; Leschner, S.; Weiss, S. CXCR2-mediated tumor-associated neutrophil recruitment is regulated by IFN-β. Int. J. Cancer 2014, 134, 1346–1358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andzinski, L.; Wu, C.F.; Lienenklaus, S.; Kröger, A.; Weiss, S.; Jablonska, J. Delayed apoptosis of tumor associated neutrophils in the absence of endogenous IFN-β. Int. J. Cancer 2015, 136, 572–583. [Google Scholar] [CrossRef] [PubMed]
- Wu, C.F.; Andzinski, L.; Kasnitz, N.; Kröger, A.; Klawonn, F.; Lienenklaus, S.; Weiss, S.; Jablonska, J. The lack of type I interferon induces neutrophil-mediated pre-metastatic niche formation in the mouse lung. Int. J. Cancer 2015, 137, 837–847. [Google Scholar] [CrossRef] [PubMed]
- Raverdeau, M.; Cunningham, S.P.; Harmon, C.; Lynch, L. γδ T cells in cancer: A small population of lymphocytes with big implications. Clin. Transl. Immunol. 2019, 8, e01080. [Google Scholar] [CrossRef] [PubMed]
- Lawand, M.; Déchanet-Merville, J.; Dieu-Nosjean, M.C. Key Features of Gamma-Delta T-Cell Subsets in Human Diseases and Their Immunotherapeutic Implications. Front. Immunol. 2017, 8, 761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, Y.; Yang, W.; Pan, M.; Scully, E.; Girardi, M.; Augenlicht, L.H.; Craft, J.; Yin, Z. γδ T cells provide an early source of interferon γ in tumor immunity. J. Exp. Med. 2003, 198, 433–442. [Google Scholar] [CrossRef] [Green Version]
- He, W.; Hao, J.; Dong, S.; Gao, Y.; Tao, J.; Chi, H.; Flavell, R.; O’Brien, R.L.; Born, W.K.; Craft, J.; et al. Naturally activated V gamma 4 gamma delta T cells play a protective role in tumor immunity through expression of eomesodermin. J. Immunol. 2010, 185, 126–133. [Google Scholar] [CrossRef] [Green Version]
- Takaoka, A.; Hayakawa, S.; Yanai, H.; Stoiber, D.; Negishi, H.; Kikuchi, H.; Sasaki, S.; Imai, K.; Shibue, T.; Honda, K.; et al. Integration of interferon-α/β signalling to p53 responses in tumour suppression and antiviral defence. Nature 2003, 424, 516–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kosciuczuk, E.M.; Mehrotra, S.; Saleiro, D.; Kroczynska, B.; Majchrzak-Kita, B.; Lisowski, P.; Driehaus, C.; Rogalska, A.; Turner, A.; Lienhoop, T.; et al. Sirtuin 2-mediated deacetylation of cyclin-dependent kinase 9 promotes STAT1 signaling in type I interferon responses. J. Biol. Chem. 2019, 294, 827–837. [Google Scholar] [CrossRef] [Green Version]
- Saleiro, D.; Mehrotra, S.; Kroczynska, B.; Beauchamp, E.M.; Lisowski, P.; Majchrzak-Kita, B.; Bhagat, T.D.; Stein, B.L.; McMahon, B.; Altman, J.K.; et al. Central role of ULK1 in type I interferon signaling. Cell Rep. 2015, 11, 605–617. [Google Scholar] [CrossRef] [Green Version]
- Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.-D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-dependent cytosolic DNA sensing promotes radiation-induced Type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
- Ranoa, D.R.E.; Parekh, A.D.; Pitroda, S.P.; Huang, X.; Darga, T.; Wong, A.C.; Huang, L.; Andrade, J.; Staley, J.P.; Satoh, T.; et al. Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs. Oncotarget 2016, 7, 26496–26515. [Google Scholar] [CrossRef] [PubMed]
- Weichselbaum, R.R.; Ishwaran, H.; Yoon, T.; Nuyten, D.S.A.; Baker, S.W.; Khodarev, N.; Su, A.W.; Shaikh, A.Y.; Roach, P.; Kreike, B.; et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc. Natl. Acad. Sci. USA 2008, 105, 18490–18495. [Google Scholar] [CrossRef] [Green Version]
- Odnokoz, O.; Yu, P.; Peck, A.R.; Sun, Y.; Kovatich, A.J.; Hooke, J.A.; Hu, H.; Mitchell, E.P.; Rui, H.; Fuchs, S.Y. Malignant cell-specific pro-tumorigenic role of type I interferon receptor in breast cancers. Cancer Biol. Ther. 2020, 21, 629–636. [Google Scholar] [CrossRef]
- Arslan, A.D.; Sassano, A.; Saleiro, D.; Lisowski, P.; Kosciuczuk, E.M.; Fischietti, M.; Eckerdt, F.; Fish, E.N.; Platanias, L.C. Human SLFN5 is a transcriptional co-repressor of STAT1-mediated interferon responses and promotes the malignant phenotype in glioblastoma. Oncogene 2017, 36, 6006–6019. [Google Scholar] [CrossRef] [Green Version]
- Mojic, M.; Takeda, K.; Hayakawa, Y. The dark side of IFN-γ: Its role in promoting cancer immunoevasion. Int. J. Mol. Sci. 2018, 19, 89. [Google Scholar] [CrossRef] [Green Version]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef]
- Lüth, S.; Schrader, J.; Zander, S.; Carambia, A.; Buchkremer, J.; Huber, S.; Reifenberg, K.; Yamamura, K.I.; Schirmacher, P.; Lohse, A.W.; et al. Chronic inflammatory IFN-γ signaling suppresses hepatocarcinogenesis in mice by sensitizing hepatocytes for apoptosis. Cancer Res. 2011, 71, 3763–3771. [Google Scholar]
- Wang, W.; Green, M.; Choi, J.E.; Gijón, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.S.; Shen, S.Q.; Sun, H.W.; Xing, Z.X.; Yang, H.L. Interferon-gamma induces autophagy-associated apoptosis through induction of cPLA2-dependent mitochondrial ROS generation in colorectal cancer cells. Biochem. Biophys. Res. Commun. 2018, 498, 1058–1065. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Huang, L.; Ding, G.; Huang, H.; Cao, G.; Sun, X.; Lou, N.; Wei, Q.; Shen, T.; Xu, X.; et al. Interferon gamma inhibits CXCL8-CXCR2 axis mediated tumor-associated macrophages tumor trafficking and enhances anti-PD1 efficacy in pancreatic cancer. J. Immunother. Cancer 2020, 8, e000308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Glasner, A.; Levi, A.; Enk, J.; Isaacson, B.; Viukov, S.; Orlanski, S.; Scope, A.; Neuman, T.; Enk, C.D.; Hanna, J.H.; et al. NKp46 Receptor-Mediated Interferon-γ Production by Natural Killer Cells Increases Fibronectin 1 to Alter Tumor Architecture and Control Metastasis. Immunity 2018, 48, 107–119.e4. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Hao, X.; Sun, R.; Wei, H.; Tian, Z. Natural Killer Cell-Derived Interferon-Gamma Promotes Hepatocellular Carcinoma Through the Epithelial Cell Adhesion Molecule-Epithelial-to-Mesenchymal Transition Axis in Hepatitis B Virus Transgenic Mice. Hepatology 2019, 69, 1735–1750. [Google Scholar] [CrossRef] [PubMed]
- Zhuang, X.; Veltri, D.P.; Long, E.O. Genome-Wide CRISPR Screen Reveals Cancer Cell Resistance to NK Cells Induced by NK-Derived IFN-γ. Front. Immunol. 2019, 10, 2879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freeman, A.J.; Vervoort, S.J.; Ramsbottom, K.M.; Kelly, M.J.; Michie, J.; Pijpers, L.; Johnstone, R.W.; Kearney, C.J.; Oliaro, J. Natural Killer Cells Suppress T Cell-Associated Tumor Immune Evasion. Cell Rep. 2019, 28, 2784–2794.e5. [Google Scholar] [CrossRef] [Green Version]
- Takeda, K.; Nakayama, M.; Hayakawa, Y.; Kojima, Y.; Ikeda, H.; Imai, N.; Ogasawara, K.; Okumura, K.; Thomas, D.M.; Smyth, M.J. IFN-γ is required for cytotoxic T cell-dependent cancer genome immunoediting. Nat. Commun. 2017, 8, 14607. [Google Scholar] [CrossRef]
- Singh, S.; Kumar, S.; Srivastava, R.K.; Nandi, A.; Thacker, G.; Murali, H.; Kim, S.; Baldeon, M.; Tobias, J.; Blanco, M.A.; et al. Loss of ELF5-FBXW7 stabilizes IFNGR1 to promote the growth and metastasis of triple-negative breast cancer through interferon-γ signalling. Nat. Cell Biol. 2020, 22, 591–602. [Google Scholar] [CrossRef] [PubMed]
- Thibaut, R.; Bost, P.; Milo, I.; Cazaux, M.; Lemaitre, F.; Garcia, Z.; Amit, I.; Breart, B.; Cornuot, C.; Schwikowski, B.; et al. Bystander IFN-γ activity promotes widespread and sustained cytokine signaling altering the tumor microenvironment. Nature Cancer 2020, 1, 302–314. [Google Scholar] [CrossRef] [Green Version]
- Zaidi, M.R.; Davis, S.; Noonan, F.P.; Graff-Cherry, C.; Hawley, T.S.; Walker, R.L.; Feigenbaum, L.; Fuchs, E.; Lyakh, L.; Young, H.A.; et al. Interferon-γ links ultraviolet radiation to melanomagenesis in mice. Nature 2011, 469, 548–553. [Google Scholar] [CrossRef] [Green Version]
- Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
- Benci, J.L.; Xu, B.; Qiu, Y.; Wu, T.J.; Dada, H.; Twyman-Saint Victor, C.; Cucolo, L.; Lee, D.S.M.; Pauken, K.E.; Huang, A.C.; et al. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell 2016, 167, 1540–1554. [Google Scholar] [CrossRef] [Green Version]
- Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mo, X.; Zhang, H.; Preston, S.; Martin, K.; Zhou, B.; Vadalia, N.; Gamero, A.M.; Soboloff, J.; Tempera, I.; Zaidi, M.R. Interferon-γ signaling in melanocytes and melanoma cells regulates expression of CTLA-4. Cancer Res. 2018, 78, 436–450. [Google Scholar] [CrossRef] [Green Version]
- Spranger, S.; Spaapen, R.M.; Zha, Y.; Williams, J.; Meng, Y.; Ha, T.T.; Gajewski, T.F. Up- regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci. Transl. Med. 2013, 5, 200ra116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; Zeng, Y.; Qu, Q.; Zhu, J.; Liu, Z.; Ning, W.; Zeng, H.; Zhang, N.; Du, W.; Chen, C.; et al. PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int. J. Clin. Oncol. 2017, 22, 1026–1033. [Google Scholar] [CrossRef]
- Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 2017, 19, 1189–1201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoekstra, M.E.; Bornes, L.; Dijkgraaf, F.E.; Philips, D.; Pardieck, I.N.; Toebes, M.; Thommen, D.S.; van Rheenen, J.; Schumacker, T.N.M. Long-distance modulation of bystander tumor cells by CD8+ T-cell-secreted IFN-gamma. Nat. Cancer 2020, 1, 291–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Supplitt, S.; Karpinski, P.; Sasiadek, M.; Laczmanska, I. Current Achievements and Applications of Transcriptomics in Personalized Cancer Medicine. Int J. Mol. Sci. 2021, 22, 1422. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.; Wallace, T.A.; Yi, M.; Magi-Galluzzi, C.; Dorsey, T.H.; Onabajo, O.O.; Obajemu, A.; Jordan, S.V.; Loffredo, C.A.; Stephens, R.M.; et al. IFNL4-ΔG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with Prostate Cancer. Clin. Cancer Res. 2018, 24, 5471–5481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, D.N.; Boersma, B.J.; Yi, M.; Reimers, M.; Howe, T.M.; Yfantis, H.G.; Tsai, Y.C.; Williams, E.H.; Lee, D.H.; Stephens, R.M.; et al. Differences in the tumor microenvironment between African-American and European-American breast cancer patients. PLoS ONE 2009, 4, e4531. [Google Scholar] [CrossRef] [Green Version]
- Budhwani, M.; Mazzieri, R.; Dolcetti, R. Plasticity of Type I interferon-mediated responses in cancer therapy: From anti-tumor immunity to resistance. Front. Oncol. 2018, 8, 322. [Google Scholar] [CrossRef] [Green Version]
- Patel, S.J.; Sanjana, N.E.; Kishton, R.J.; Eidizadeh, A.; Vodnala, S.K.; Cam, M.; Gartner, J.J.; Jia, L.; Steinberg, S.M.; Yamamoto, T.N.; et al. Identification of essential genes for cancer immunotherapy. Nature 2017, 548, 537–542. [Google Scholar] [CrossRef] [Green Version]
- Hamid, M.A.; Yao, X.; Waugh, C.; Rosendo-Machado, S.; Li, C.; Rostron, T.; Frankland, J.; Peng, Y.; Dong, T. Defective interferon gamma production by tumor-specific CD8+ T cells is associated with 5′methylcytosine-guanine hypermethylation of interferon gamma promoter. Front. Immunol. 2020, 11, 310. [Google Scholar] [CrossRef] [PubMed]
- Wong, L.H.; Krauer, K.G.; Hatzinisiriou, I.; Estcourt, M.J.; Hersey, P.; Tam, N.D.; Edmondson, S.; Devenish, R.J.; Ralph, S.J. Interferon-resistant human melanoma cells are deficient in ISGF3 components, STAT1, STAT2, and p48-ISGF3γ. J. Biol. Chem. 1997, 272, 28779–28785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xi, S.; Dyer, K.F.; Kimak, M.; Zhang, Q.; Gooding, W.E.; Chaillet, J.R.; Chai, R.L.; Ferrell, R.E.; Zamboni, B.; Hunt, J.; et al. Decreased STAT1 expression by promoter methylation in squamous cell carcinogenesis. J. Natl. Cancer Inst. 2006, 98, 181–189. [Google Scholar] [CrossRef] [Green Version]
- Salerno, F.; Guislain, A.; Freen-Van Heeren, J.J.; Nicolet, B.P.; Young, H.A.; Wolkers, M.C. Critical role of post-transcriptional regulation for IFN-γ in tumor-infiltrating T cells. Oncoimmunology 2018, 8, e1532762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhattacharya, S.; HuangFu, W.C.; Dong, G.; Qian, J.; Baker, D.P.; Karar, J.; Koumenis, C.; Diehl, J.A.; Fuchs, S.Y. Anti-tumorigenic effects of Type 1 interferon are subdued by integrated stress responses. Oncogene 2013, 32, 4214–4221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huangfu, W.C.; Qian, J.; Liu, C.; Liu, J.; Lokshin, A.E.; Baker, D.P.; Rui, H.; Fuchs, S.Y. Inflammatory signaling compromises cell responses to interferon alpha. Oncogene 2012, 31, 161–172. [Google Scholar] [CrossRef] [Green Version]
- Ortiz, A.; Gui, J.; Zahedi, F.; Yu, P.; Cho, C.; Bhattacharya, S.; Carbone, C.J.; Yu, Q.; Katlinski, K.V.; Katlinskaya, Y.V.; et al. An Interferon-Driven Oxysterol-Based Defense against Tumor-Derived Extracellular Vesicles. Cancer Cell 2019, 35, 33–45.e6. [Google Scholar] [CrossRef] [Green Version]
- Katlinskaya, Y.V.; Katlinski, K.V.; Yu, Q.; Ortiz, A.; Beiting, D.P.; Brice, A.; Davar, D.; Sanders, C.; Kirkwood, J.M.; Rui, H.; et al. Suppression of Type I Interferon Signaling Overcomes Oncogene-Induced Senescence and Mediates Melanoma Development and Progression. Cell Rep. 2016, 15, 171–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, J.; Shi, L.Z.; Zhao, H.; Chen, J.; Xiong, L.; He, Q.; Chen, T.; Roszik, J.; Bernatchez, C.; Woodman, S.E.; et al. Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell 2016, 167, 397–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef]
- Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 2017, 8, 1136. [Google Scholar] [CrossRef]
- Pötzl, J.; Roser, D.; Bankel, L.; Hömberg, N.; Geishauser, A.; Brenner, C.D.; Weigand, M.; Röcken, M.; Mocikat, R. Reversal of tumor acidosis by systemic buffering reactivates NK cells to express IFN-gamma and induces NK cell-dependent lymphoma control without other immunotherapies. Int. J. Cancer 2017, 140, 2125–2133. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Fenton, S.E.; Saleiro, D.; Platanias, L.C. Type I and II Interferons in the Anti-Tumor Immune Response. Cancers 2021, 13, 1037. https://doi.org/10.3390/cancers13051037
Fenton SE, Saleiro D, Platanias LC. Type I and II Interferons in the Anti-Tumor Immune Response. Cancers. 2021; 13(5):1037. https://doi.org/10.3390/cancers13051037
Chicago/Turabian StyleFenton, Sarah E., Diana Saleiro, and Leonidas C. Platanias. 2021. "Type I and II Interferons in the Anti-Tumor Immune Response" Cancers 13, no. 5: 1037. https://doi.org/10.3390/cancers13051037
APA StyleFenton, S. E., Saleiro, D., & Platanias, L. C. (2021). Type I and II Interferons in the Anti-Tumor Immune Response. Cancers, 13(5), 1037. https://doi.org/10.3390/cancers13051037