Abstract
Cancer immunotherapy offers substantive benefit to patients with various tumour types, in some cases leading to complete tumour clearance. However, many patients do not respond to immunotherapy, galvanizing the field to define the mechanisms of pre-existing and acquired resistance. Interferon-γ (IFNγ) is a cytokine that has both protumour and antitumour activities, suggesting that it may serve as a nexus for responsiveness to immunotherapy. Many cancer immunotherapies and chemotherapies induce IFNγ production by various cell types, including activated T cells and natural killer cells. Patients resistant to these therapies commonly have molecular aberrations in the IFNγ signalling pathway or express resistance molecules driven by IFNγ. Given that all nucleated cells can respond to IFNγ, the functional consequences of IFNγ production need to be carefully dissected on a cell-by-cell basis. Here, we review the cells that produce IFNγ and the different effects of IFNγ in the tumour microenvironment, highlighting the pleiotropic nature of this multifunctional and abundant cytokine.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
£14.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
£139.00 per year
only £11.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Wheelock, E. F. Interferon-like virus-inhibitor induced in human leukocytes by phytohemagglutinin. Science 149, 310–311 (1965).
Dighe, A. S., Richards, E., Old, L. J. & Schreiber, R. D. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors. Immunity 1, 447–456 (1994).
Nastala, C. L. et al. Recombinant IL-12 administration induces tumor regression in association with IFN-gamma production. J. Immunol. 153, 1697–1706 (1994).
Young, H. A., Dray, J. F. & Farrar, W. L. Expression of transfected human interferon-gamma DNA: evidence for cell-specific regulation. J. Immunol. 136, 4700–4703 (1986).
Soutto, M., Zhou, W. & Aune, T. M. Cutting edge: distal regulatory elements are required to achieve selective expression of IFN-gamma in Th1/Tc1 effector cells. J. Immunol. 169, 6664–6667 (2002).
Fields, P. E., Kim, S. T. & Flavell, R. A. Cutting edge: changes in histone acetylation at the IL-4 and IFN-gamma loci accompany Th1/Th2 differentiation. J. Immunol. 169, 647–650 (2002).
Gomez, J. A. et al. The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell 152, 743–754 (2013).
Shnyreva, M. et al. Evolutionarily conserved sequence elements that positively regulate IFN-gamma expression in T cells. Proc. Natl Acad. Sci. USA 101, 12622–12627 (2004).
Kiani, A. et al. Regulation of interferon-gamma gene expression by nuclear factor of activated T cells. Blood 98, 1480–1488 (2001).
Beals, C. R., Sheridan, C. M., Turck, C. W., Gardner, P. & Crabtree, G. R. Nuclear export of NF-ATc enhanced by glycogen synthase kinase-3. Science 275, 1930–1934 (1997).
Dong, C., Davis, R. J. & Flavell, R. A. MAP kinases in the immune response. Annu. Rev. Immunol. 20, 55–72 (2002).
Park, W.-R. et al. A mechanism underlying STAT4-mediated up-regulation of IFN-gamma induction in TCR-triggered T cells. Int. Immunol. 16, 295–302 (2004).
Johnson, H. M. & Torres, B. A. Leukotrienes: positive signals for regulation of gamma-interferon production. J. Immunol. 132, 413–416 (1984).
Kasahara, T., Hooks, J. J., Dougherty, S. F. & Oppenheim, J. J. Interleukin 2-mediated immune interferon (IFN-gamma) production by human T cells and T cell subsets. J. Immunol. 130, 1784–1789 (1983).
Johnson, H. M., Russell, J. K. & Torres, B. A. Second messenger role of arachidonic acid and its metabolites in interferon-gamma production. J. Immunol. 137, 3053–3056 (1986).
Vignali, D. A. A. & Kuchroo, V. K. IL-12 family cytokines: immunological playmakers. Nat. Immunol. 13, 722–728 (2012).
Rex, D. A. B. et al. A comprehensive pathway map of IL-18-mediated signalling. J. Cell Commun. Signal. 14, 257–266 (2020).
Okamura, H. et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature 378, 88–91 (1995). The authors cloned IL-18 (also known as IGIF) and showed in vitro induction of IFNγ with recombinant IL-18 via an IL-12-independent pathway.
Nakahira, M. et al. Synergy of IL-12 and IL-18 for IFN-gamma gene expression: IL-12-induced STAT4 contributes to IFN-gamma promoter activation by up-regulating the binding activity of IL-18-induced activator protein 1. J. Immunol. 168, 1146–1153 (2002).
Mavropoulos, A., Sully, G., Cope, A. P. & Clark, A. R. Stabilization of IFN-gamma mRNA by MAPK p38 in IL-12- and IL-18-stimulated human NK cells. Blood 105, 282–288 (2005).
Hodge, D. L., Martinez, A., Julias, J. G., Taylor, L. S. & Young, H. A. Regulation of nuclear gamma interferon gene expression by interleukin 12 (IL-12) and IL-2 represents a novel form of posttranscriptional control. Mol. Cell. Biol. 22, 1742–1753 (2002).
Steiner, D. F. et al. MicroRNA-29 regulates T-box transcription factors and interferon-γ production in helper T cells. Immunity 35, 169–181 (2011).
Wang, H. et al. Regulation of human natural killer cell IFN-γ production by microRNA-146a via targeting the NF-κB signaling pathway. Front. Immunol. 9, 293 (2018).
Ma, N. et al. MicroRNA-142-3p inhibits IFN-γ production via targeting of RICTOR in Aspergillus fumigatus activated CD4+ T cells. Ann. Transl. Med. 7, 649 (2019).
Jiang, H., Zhang, G., Wu, J.-H. & Jiang, C.-P. Diverse roles of miR-29 in cancer (review). Oncol. Rep. 31, 1509–1516 (2014).
Shahriar, A. et al. The dual role of mir-146a in metastasis and disease progression. Biomed. Pharmacother. 126, 110099 (2020).
Ealick, S. E. et al. Three-dimensional structure of recombinant human interferon-gamma. Science 252, 698–702 (1991).
Miyakawa, N. et al. Prolonged circulation half-life of interferon γ activity by gene delivery of interferon γ-serum albumin fusion protein in mice. J. Pharm. Sci. 100, 2350–2357 (2011).
Charych, D. et al. Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy. PLoS ONE 12, e0179431 (2017).
Molinas, F. C., Wietzerbin, J. & Falcoff, E. Human platelets possess receptors for a lymphokine: demonstration of high specific receptors for HuIFN-gamma. J. Immunol. 138, 802–806 (1987).
Dummer, R. et al. Phase II clinical trial of intratumoral application of TG1042 (adenovirus-interferon-gamma) in patients with advanced cutaneous T-cell lymphomas and multilesional cutaneous B-cell lymphomas. Mol. Ther. 18, 1244–1247 (2010).
Altan-Bonnet, G. & Mukherjee, R. Cytokine-mediated communication: a quantitative appraisal of immune complexity. Nat. Rev. Immunol. 19, 205–217 (2019).
Pearce, E. L. et al. Control of effector CD8+ T cell function by the transcription factor eomesodermin. Science 302, 1041–1043 (2003).
Szabo, S. J. et al. Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science 295, 338–342 (2002). This study shows the requirement of T-bet expression for IFNγ production in CD4+ TH cells and NK cells but not in CD8+ T cells, indicating that distinct transcription factors control IFNγ in immune cell subsets.
Sanderson, N. S. R. et al. Cytotoxic immunological synapses do not restrict the action of interferon-γ to antigenic target cells. Proc. Natl Acad. Sci. USA 109, 7835–7840 (2012).
Gérard, A. et al. Secondary T cell-T cell synaptic interactions drive the differentiation of protective CD8+ T cells. Nat. Immunol. 14, 356–363 (2013).
Thibaut, R. et al. Bystander IFN-γ activity promotes widespread and sustained cytokine signaling altering the tumor microenvironment. Nat. Cancer 1, 302–314 (2020). This study shows that IFNγ diffuses extensively throughout the TME to produce a widespread field of IFNγ. Sustained IFNγ signalling via STAT1 and IRF8 was required to modulate tumour cell expression of MHC class I and PDL1.
Hoekstra, M. E. et al. Long-distance modulation of bystander tumor cells by CD8+ T cell-secreted IFNγ. Nat. Cancer 1, 291–301 (2020). This study visualizes the long-range diffusion of IFNγ produced by low-frequency activated CD8+ T cells in the TME.
Liu, C. et al. Treg cells promote the SREBP1-dependent metabolic fitness of tumor-promoting macrophages via repression of CD8+ T cell-derived interferon-γ. Immunity 51, 381–397.e6 (2019).
Kalathil, S. G., Hutson, A., Barbi, J., Iyer, R. & Thanavala, Y. Augmentation of IFN-γ+CD8+ T cell responses correlates with survival of HCC patients on sorafenib therapy. JCI Insight 4, e130116 (2019).
Huse, M., Lillemeier, B. F., Kuhns, M. S., Chen, D. S. & Davis, M. M. T cells use two directionally distinct pathways for cytokine secretion. Nat. Immunol. 7, 247–255 (2006).
Dai, M., Hellstrom, I., Yip, Y. Y., Sjögren, H. O. & Hellstrom, K. E. Tumor regression and cure depends on sustained th1 responses. J. Immunother. 41, 369–378 (2018).
Kryczek, I. et al. Human TH17 cells are long-lived effector memory cells. Sci. Transl Med. 3, 104ra100 (2011).
Kryczek, I. et al. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood 114, 1141–1149 (2009).
Kubota, A., Lian, R. H., Lohwasser, S., Salcedo, M. & Takei, F. IFN-gamma production and cytotoxicity of IL-2-activated murine NK cells are differentially regulated by MHC class I molecules. J. Immunol. 163, 6488–6493 (1999).
Almishri, W. et al. TNFα augments cytokine-induced NK cell IFNγ production through TNFR2. J. Innate Immun. 8, 617–629 (2016).
Reefman, E. et al. Cytokine secretion is distinct from secretion of cytotoxic granules in NK cells. J. Immunol. 184, 4852–4862 (2010).
Villegas, F. R. et al. Prognostic significance of tumor infiltrating natural killer cells subset CD57 in patients with squamous cell lung cancer. Lung Cancer 35, 23–28 (2002).
Lee, S. et al. A high-throughput assay of NK cell activity in whole blood and its clinical application. Biochem. Biophys. Res. Commun. 445, 584–590 (2014).
Henriksen, J. R. et al. Favorable prognostic impact of natural killer cells and T cells in high-grade serous ovarian carcinoma. Acta Oncol. 59, 652–659 (2020).
Lee, J. et al. Natural killer cell activity for IFN-gamma production as a supportive diagnostic marker for gastric cancer. Oncotarget 8, 70431–70440 (2017).
Moreno, M. et al. IFN-gamma-producing human invariant NKT cells promote tumor-associated antigen-specific cytotoxic T cell responses. J. Immunol. 181, 2446–2454 (2008).
Das, R. et al. The adaptor molecule SAP plays essential roles during invariant NKT cell cytotoxicity and lytic synapse formation. Blood 121, 3386–3395 (2013).
Wolf, B. J., Choi, J. E. & Exley, M. A. Novel approaches to exploiting invariant NKT cells in cancer immunotherapy. Front. Immunol. 9, 384 (2018).
Brigl, M., Bry, L., Kent, S. C., Gumperz, J. E. & Brenner, M. B. Mechanism of CD1d-restricted natural killer T cell activation during microbial infection. Nat. Immunol. 4, 1230–1237 (2003).
Zhang, Y. et al. α-GalCer and iNKT cell-based cancer immunotherapy: realizing the therapeutic potentials. Front. Immunol. 10, 1126 (2019).
Takami, M., Ihara, F. & Motohashi, S. Clinical application of iNKT cell-mediated anti-tumor activity against lung cancer and head and neck cancer. Front. Immunol. 9, 2021 (2018).
Parekh, V. V. et al. PD-1/PD-L blockade prevents anergy induction and enhances the anti-tumor activities of glycolipid-activated invariant NKT cells. J. Immunol. 182, 2816–2826 (2009).
Sawant, D. V., Hamilton, K. & Vignali, D. A. A. Interleukin-35: expanding its job profile. J. Interferon Cytokine Res. 35, 499–512 (2015).
Vignali, D. A. A., Collison, L. W. & Workman, C. J. How regulatory T cells work. Nat. Rev. Immunol. 8, 523–532 (2008).
Ouyang, W. et al. Novel Foxo1-dependent transcriptional programs control Treg cell function. Nature 491, 554–559 (2012).
Lucca, L. E. et al. TIGIT signaling restores suppressor function of Th1 Tregs. JCI Insight 4, e124427 (2019).
Koch, M. A. et al. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat. Immunol. 10, 595–602 (2009).
Levine, A. G. et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 546, 421–425 (2017).
Overacre-Delgoffe, A. E. et al. Interferon-γ drives Treg fragility to promote anti-tumor immunity. Cell 169, 1130–1141.e11 (2017). This study identifies a novel fragile state of intratumoural Treg cells characterized by the gain of IFNγ production and loss of suppressive function. A Treg cell response to IFNγ was required for benefit from anti-PD1 therapy.
Seltmann, J., Werfel, T. & Wittmann, M. Evidence for a regulatory loop between IFN-γ and IL-33 in skin inflammation. Exp. Dermatol. 22, 102–107 (2013).
Liang, Y. et al. IL-33 promotes innate IFN-γ production and modulates dendritic cell response in LCMV-induced hepatitis in mice. Eur. J. Immunol. 45, 3052–3063 (2015).
Hatzioannou, A. et al. An intrinsic role of IL-33 in Treg cell-mediated tumor immunoevasion. Nat. Immunol. 21, 75–85 (2020).
Ameri, A. H. et al. IL-33/regulatory T cell axis triggers the development of a tumor-promoting immune environment in chronic inflammation. Proc. Natl Acad. Sci. USA 116, 2646–2651 (2019).
Gao, Y. et al. Gamma delta T cells provide an early source of interferon gamma in tumor immunity. J. Exp. Med. 198, 433–442 (2003).
Hoeres, T., Holzmann, E., Smetak, M., Birkmann, J. & Wilhelm, M. PD-1 signaling modulates interferon-γ production by gamma delta (γδ) T-cells in response to leukemia. Oncoimmunology 8, 1550618 (2019).
Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).
Girardi, M. et al. Regulation of cutaneous malignancy by gammadelta T cells. Science 294, 605–609 (2001).
Fillatreau, S. B cells and their cytokine activities implications in human diseases. Clin. Immunol. 186, 26–31 (2018).
Ballesteros-Tato, A., Stone, S. L. & Lund, F. E. Innate IFNγ-producing B cells. Cell Res. 24, 135–136 (2014).
Harris, D. P., Goodrich, S., Gerth, A. J., Peng, S. L. & Lund, F. E. Regulation of IFN-gamma production by B effector 1 cells: essential roles for T-bet and the IFN-gamma receptor. J. Immunol. 174, 6781–6790 (2005).
Vitale, L. A. et al. Development of CDX-1140, an agonist CD40 antibody for cancer immunotherapy. Cancer Immunol. Immunother. 68, 1–13 (2018).
Piechutta, M. & Berghoff, A. S. New emerging targets in cancer immunotherapy: the role of cluster of differentiation 40 (CD40/TNFR5). ESMO Open 4, e000510 (2019).
Bhat, M. Y. et al. Comprehensive network map of interferon gamma signaling. J. Cell Commun. Signal. 12, 745–751 (2018). This study develops a comprehensive IFNγ signalling network curated from the literature and organized into a publicly available browser.
Samarajiwa, S. A., Forster, S., Auchettl, K. & Hertzog, P. J. INTERFEROME: the database of interferon regulated genes. Nucleic Acids Res. 37, D852–D857 (2009).
Refaeli, Y., Van Parijs, L., Alexander, S. I. & Abbas, A. K. Interferon gamma is required for activation-induced death of T lymphocytes. J. Exp. Med. 196, 999–1005 (2002).
Prabhu, N. et al. Gamma interferon regulates contraction of the influenza virus-specific CD8 T cell response and limits the size of the memory population. J. Virol. 87, 12510–12522 (2013).
Badovinac, V. P., Porter, B. B. & Harty, J. T. CD8+ T cell contraction is controlled by early inflammation. Nat. Immunol. 5, 809–817 (2004).
Ouyang, W., Beckett, O., Flavell, R. A. & Li, M. O. An essential role of the forkhead-box transcription factor Foxo1 in control of T cell homeostasis and tolerance. Immunity 30, 358–371 (2009).
Qiang, L., Banks, A. S. & Accili, D. Uncoupling of acetylation from phosphorylation regulates FoxO1 function independent of its subcellular localization. J. Biol. Chem. 285, 27396–27401 (2010).
Stoycheva, D. et al. IFN-γ regulates CD8+memory T cell differentiation and survival in response to weak, but not strong, TCR signals. J. Immunol. 194, 553–559 (2015).
Pedicord, V. A., Montalvo, W., Leiner, I. M. & Allison, J. P. Single dose of anti-CTLA-4 enhances CD8 + T-cell memory formation, function, and maintenance. Proc. Natl Acad. Sci. USA 108, 266–271 (2011).
Tau, G. Z., Cowan, S. N., Weisburg, J., Braunstein, N. S. & Rothman, P. B. Regulation of IFN-gamma signaling is essential for the cytotoxic activity of CD8(+) T cells. J. Immunol. 167, 5574–5582 (2001).
Pai, C.-C. S. et al. Clonal deletion of tumor-specific T cells by interferon-γ confers therapeutic resistance to combination immune checkpoint blockade. Immunity 50, 477–492.e8 (2019). This study shows that tumour-specific CD8+ T cells express higher levels of IFNGR than T cells, thus inducing apoptosis, an immune-intrinsic mechanism of resistance to checkpoint blockade.
Colvin, R. A., Campanella, G. S. V., Sun, J. & Luster, A. D. Intracellular domains of CXCR3 that mediate CXCL9, CXCL10, and CXCL11 function. J. Biol. Chem. 279, 30219–30227 (2004).
Liu, Z. et al. CXCL11-armed oncolytic poxvirus elicits potent antitumor immunity and shows enhanced therapeutic efficacy. Oncoimmunology 5, e1091554 (2016).
Bhat, P., Leggatt, G., Waterhouse, N. & Frazer, I. H. Interferon-γ derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 8, e2836 (2017).
Bonaventura, P. et al. Cold tumors: a therapeutic challenge for immunotherapy. Front. Immunol. 10, 168 (2019).
Pernis, A. et al. Lack of interferon gamma receptor beta chain and the prevention of interferon gamma signaling in TH1 cells. Science 269, 245–247 (1995).
Holzer, U., Reinhardt, K., Lang, P., Handgretinger, R. & Fischer, N. Influence of a mutation in IFN-γ receptor 2 (IFNGR2) in human cells on the generation of Th17 cells in memory T cells. Hum. Immunol. 74, 693–700 (2013).
Lazarevic, V. et al. T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORγt. Nat. Immunol. 12, 96–104 (2011).
Naka, T. et al. SOCS-1/SSI-1-deficient NKT cells participate in severe hepatitis through dysregulated cross-talk inhibition of IFN-gamma and IL-4 signaling in vivo. Immunity 14, 535–545 (2001).
Hwang, E. S., Szabo, S. J., Schwartzberg, P. L. & Glimcher, L. H. T helper cell fate specified by kinase-mediated interaction of T-bet with GATA-3. Science 307, 430–433 (2005).
Diskin, B. et al. PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat. Immunol. 21, 442–454 (2020). This study shows that PDL1 ‘back-signalling’ in CD4+ T cells decreased activation and TH1 cell polarization and induced anergy in CD8+ T cells. PDL1+ T cells restrained PD1+ effector T cells and macrophages and induced a protumorigenic microenvironment.
Zhou, Y., Weyman, C. M., Liu, H., Almasan, A. & Zhou, A. IFN-gamma induces apoptosis in HL-60 cells through decreased Bcl-2 and increased Bak expression. J. Interferon Cytokine Res. 28, 65–72 (2008).
Xu, X., Fu, X. Y., Plate, J. & Chong, A. S. IFN-gamma induces cell growth inhibition by Fas-mediated apoptosis: requirement of STAT1 protein for up-regulation of Fas and FasL expression. Cancer Res. 58, 2832–2837 (1998).
Rakshit, S. et al. Interferon-gamma induced cell death: regulation and contributions of nitric oxide, cJun N-terminal kinase, reactive oxygen species and peroxynitrite. Biochim. Biophys. Acta 1843, 2645–2661 (2014).
Overacre, A. E. & Vignali, D. A. Treg stability: to be or not to be. Curr. Opin. Immunol. 39, 39–43 (2016).
Halim, L. et al. An atlas of human regulatory T helper-like cells reveals features of Th2-like Tregs that support a tumorigenic environment. Cell Rep. 20, 757–770 (2017).
Wu, S.-P. et al. Stromal PD-L1-positive regulatory T cells and PD-1-positive CD8-positive T cells define the response of different subsets of non-small cell lung cancer to PD-1/PD-L1 blockade immunotherapy. J. Thorac. Oncol. 13, 521–532 (2018).
Koch, M. A. et al. T-bet+ Treg cells undergo abortive Th1 cell differentiation due to impaired expression of IL-12 receptor β2. Immunity 37, 501–510 (2012).
Fallarino, F. et al. T cell apoptosis by tryptophan catabolism. Cell Death Differ. 9, 1069–1077 (2002).
Chevolet, I. et al. Characterization of the in vivo immune network of IDO, tryptophan metabolism, PD-L1, and CTLA-4 in circulating immune cells in melanoma. Oncoimmunology 4, e982382 (2015).
Prendergast, G. C., Malachowski, W. P., DuHadaway, J. B. & Muller, A. J. Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res. 77, 6795–6811 (2017).
Mitchell, T. C. et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase I results from a multicenter, open-label phase I/II trial (ECHO-202/KEYNOTE-037). J. Clin. Oncol. 36, 3223–3230 (2018).
Carnaud, C. et al. Cutting edge: cross-talk between cells of the innate immune system: NKT cells rapidly activate NK cells. J. Immunol. 163, 4647–4650 (1999).
Putz, E. M. et al. Novel non-canonical role of STAT1 in natural killer cell cytotoxicity. Oncoimmunology 5, e1186314 (2016).
Wendel, M., Galani, I. E., Suri-Payer, E. & Cerwenka, A. Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res. 68, 8437–8445 (2008).
Park, S.-Y. et al. IFN-gamma enhances TRAIL-induced apoptosis through IRF-1. Eur. J. Biochem. 271, 4222–4228 (2004).
Putz, E. M., Gotthardt, D. & Sexl, V. STAT1-S727 - the license to kill. Oncoimmunology 3, e955441 (2014).
Früh, K. & Yang, Y. Antigen presentation by MHC class I and its regulation by interferon gamma. Curr. Opin. Immunol. 11, 76–81 (1999).
Steimle, V., Siegrist, C. A., Mottet, A., Lisowska-Grospierre, B. & Mach, B. Regulation of MHC class II expression by interferon-gamma mediated by the transactivator gene CIITA. Science 265, 106–109 (1994).
Meissner, T. B. et al. NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc. Natl Acad. Sci. USA 107, 13794–13799 (2010).
Li, J., Yang, Y., Inoue, H., Mori, M. & Akiyoshi, T. The expression of costimulatory molecules CD80 and CD86 in human carcinoma cell lines: its regulation by interferon gamma and interleukin-10. Cancer Immunol. Immunother. 43, 213–219 (1996).
Collins, A. V. et al. The interaction properties of costimulatory molecules revisited. Immunity 17, 201–210 (2002).
Pan, J. et al. Interferon-gamma is an autocrine mediator for dendritic cell maturation. Immunol. Lett. 94, 141–151 (2004).
Garris, C. S. et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity 49, 1148–1161.e7 (2018).
Jackson, S. W. et al. B cell IFN-γ receptor signaling promotes autoimmune germinal centers via cell-intrinsic induction of BCL-6. J. Exp. Med. 213, 733–750 (2016).
Metzger, D. W. et al. Interleukin-12 acts as an adjuvant for humoral immunity through interferon-gamma-dependent and -independent mechanisms. Eur. J. Immunol. 27, 1958–1965 (1997).
Cillo, A. R. et al. Immune landscape of viral- and carcinogen-driven head and neck cancer. Immunity 52, 183–199.e9 (2020).
Bruno, T. C. New predictors for immunotherapy responses sharpen our view of the tumour microenvironment. Nature 577, 474–476 (2020).
Wu, C. et al. IFN-γ primes macrophage activation by increasing phosphatase and tensin homolog via downregulation of miR-3473b. J. Immunol. 193, 3036–3044 (2014).
Sadlik, J. R. et al. Lymphocyte supernatant-induced human monocyte tumoricidal activity: dependence on the presence of gamma-interferon. Cancer Res. 45, 1940–1945 (1985).
Haabeth, O. A. W. et al. Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer. Nat. Commun. 2, 240 (2011).
Liu, M. et al. Targeting the IDO1 pathway in cancer: from bench to bedside. J. Hematol. Oncol. 11, 100 (2018).
Yan, Y. et al. IDO upregulates regulatory T cells via tryptophan catabolite and suppresses encephalitogenic T cell responses in experimental autoimmune encephalomyelitis. J. Immunol. 185, 5953–5961 (2010).
Vannini, F., Kashfi, K. & Nath, N. The dual role of iNOS in cancer. Redox Biol. 6, 334–343 (2015).
Grzywa, T. M. et al. Myeloid cell-derived arginase in cancer immune response. Front. Immunol. 11, 938 (2020).
Strieter, R. M. et al. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J. Biol. Chem. 270, 27348–27357 (1995).
Tokunaga, R. et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - a target for novel cancer therapy. Cancer Treat. Rev. 63, 40–47 (2018).
Karin, N., Wildbaum, G. & Thelen, M. Biased signaling pathways via CXCR3 control the development and function of CD4+ T cell subsets. J. Leukoc. Biol. 99, 857–862 (2016).
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).
Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404.e9 (2016). This study shows that copy-number loss of IFNγ pathway activating genes and amplification of IFNγ pathway inhibitors are associated with clinical resistance to anti-CTLA4 therapy.
Benci, J. L. et al. Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell 178, 933–948.e14 (2019). This study shows adaptive immune resistance within the TME by which protumorigenic IFNγ signalling in tumour cells opposes antitumorigenic IFNγ signalling in immune cells to limit tumour killing by adaptive and innate immune cells.
Watcharanurak, K. et al. Effects of upregulated indoleamine 2, 3-dioxygenase 1 by interferon γ gene transfer on interferon γ-mediated antitumor activity. Gene Ther. 21, 794–801 (2014).
Kostourou, V. et al. The role of tumour-derived iNOS in tumour progression and angiogenesis. Br. J. Cancer 104, 83–90 (2011).
Teleanu, R. I., Chircov, C., Grumezescu, A. M. & Teleanu, D. M. Tumor angiogenesis and anti-angiogenic strategies for cancer treatment. J. Clin. Med. 9, 84 (2019).
Kataru, R. P. et al. T lymphocytes negatively regulate lymph node lymphatic vessel formation. Immunity 34, 96–107 (2011).
Zampell, J. C. et al. Lymphatic function is regulated by a coordinated expression of lymphangiogenic and anti-lymphangiogenic cytokines. Am. J. Physiol. Cell Physiol. 302, C392–C404 (2012).
Lane, R. S. et al. IFNγ-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J. Exp. Med. 215, 3057–3074 (2018).
Mondal, A. et al. IDO1 is an integral mediator of inflammatory neovascularization. EBioMedicine 14, 74–82 (2016).
Sun, T. et al. Inhibition of tumor angiogenesis by interferon-γ by suppression of tumor-associated macrophage differentiation. Oncol. Res. 21, 227–235 (2014).
Xu, H.-M. Th1 cytokine-based immunotherapy for cancer. Hepatobiliary Pancreat. Dis. Int. 13, 482–494 (2014).
Benmebarek, M.-R. et al. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int. J. Mol. Sci. 20, 1283 (2019).
Müller, E. et al. Toll-like receptor ligands and interferon-γ synergize for induction of antitumor M1 macrophages. Front. Immunol. 8, 1383 (2017).
Ni, L. & Lu, J. Interferon gamma in cancer immunotherapy. Cancer Med. 7, 4509–4516 (2018).
Razaghi, A., Owens, L. & Heimann, K. Review of the recombinant human interferon gamma as an immunotherapeutic: Impacts of production platforms and glycosylation. J. Biotechnol. 240, 48–60 (2016).
Gleave, M. E. et al. Interferon gamma-1b compared with placebo in metastatic renal-cell carcinoma. N. Engl. J. Med. 338, 1265–1271 (1998).
Ando, M. et al. Prevention of adverse events of interferon γ gene therapy by gene delivery of interferon γ-heparin-binding domain fusion protein in mice. Mol. Ther. Methods Clin. Dev. 1, 14023 (2014).
Hemmerle, T. & Neri, D. The dose-dependent tumor targeting of antibody-IFNγ fusion proteins reveals an unexpected receptor-trapping mechanism in vivo. Cancer Immunol. Res. 2, 559–567 (2014).
Lee, C. S. et al. Adenovirus-mediated gene delivery: potential applications for gene and cell-based therapies in the new era of personalized medicine. Genes Dis. 4, 43–63 (2017).
Bourgeois-Daigneault, M.-C. et al. Oncolytic vesicular stomatitis virus expressing interferon-γ has enhanced therapeutic activity. Mol. Ther. Oncolytics 3, 16001 (2016).
Oh, E., Choi, I.-K., Hong, J. & Yun, C.-O. Oncolytic adenovirus coexpressing interleukin-12 and decorin overcomes Treg-mediated immunosuppression inducing potent antitumor effects in a weakly immunogenic tumor model. Oncotarget 8, 4730–4746 (2017).
Ando, M., Takahashi, Y., Nishikawa, M., Watanabe, Y. & Takakura, Y. Constant and steady transgene expression of interferon-γ by optimization of plasmid construct for safe and effective interferon-γ gene therapy. J. Gene Med. 14, 288–295 (2012).
Yuba, E. et al. pH-sensitive polymer-liposome-based antigen delivery systems potentiated with interferon-γ gene lipoplex for efficient cancer immunotherapy. Biomaterials 67, 214–224 (2015).
Mendoza, J. L. et al. Structure of the IFNγ receptor complex guides design of biased agonists. Nature 567, 56–60 (2019). The authors solve the crystal structure of the hexameric IFNγ–IFNGR1–IFNGR2 signalling complex, which is then used as a blueprint to develop antitumorigenic biased ligands that induce MHC class I expression but not PDL1 expression.
Taube, J. M. et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci. Transl Med. 4, 127ra37 (2012).
Gubin, M. M. et al. High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell 175, 1014–1030.e19 (2018).
Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017). This study analyses RNA expression profiles of patients treated with anti-PD1 (pembrolizumab) and finds that IFNγ-responsive genes involved in antigen presentation, chemokine expression, cytotoxic activity and adaptive immune resistance were necessary for a clinical benefit from therapy.
Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016).
Shin, D. S. et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 7, 188–201 (2017).
Peng, D. et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 527, 249–253 (2015).
Li, J. et al. Epigenetic driver mutations in ARID1A shape cancer immune phenotype and immunotherapy. J. Clin. Invest. 130, 2712–2726 (2020).
Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).
Zhai, L. et al. IDO1 in cancer: a Gemini of immune checkpoints. Cell Mol. Immunol. 15, 447–457 (2018).
Oyler-Yaniv, J. et al. Catch and release of cytokines mediated by tumor phosphatidylserine converts transient exposure into long-lived inflammation. Mol. Cell 66, 635–647.e7 (2017). This study discovers that IFNγ is captured by phosphatidylserine on the surface of cells to mediate slow IFNγ release driving long-term transcriptional effects.
Benci, J. L. et al. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell 167, 1540–1554.e12 (2016).
Farrar, M. A. & Schreiber, R. D. The molecular cell biology of interferon-gamma and its receptor. Annu. Rev. Immunol. 11, 571–611 (1993).
Valente, G. et al. Distribution of interferon-gamma receptor in human tissues. Eur. J. Immunol. 22, 2403–2412 (1992).
Schmiedel, B. J. et al. Impact of genetic polymorphisms on human immune cell gene expression. Cell 175, 1701–1715.e16 (2018).
Monaco, G. et al. RNA-seq signatures normalized by mRNA abundance allow absolute deconvolution of human immune cell types. Cell Rep. 26, 1627–1640.e7 (2019).
Ebensperger, C. et al. Genomic organization and promoter analysis of the gene Ifngr2 encoding the second chain of the mouse interferon-gamma receptor. Scand. J. Immunol. 44, 599–606 (1996).
Green, D. S., Young, H. A. & Valencia, J. C. Current prospects of type II interferon γ signaling and autoimmunity. J. Biol. Chem. 292, 13925–13933 (2017).
Larkin, J., Johnson, H. M. & Subramaniam, P. S. Differential nuclear localization of the IFNGR-1 and IFNGR-2 subunits of the IFN-gamma receptor complex following activation by IFN-gamma. J. Interferon Cytokine Res. 20, 565–576 (2000).
Gough, D. J., Levy, D. E., Johnstone, R. W. & Clarke, C. J. IFNgamma signaling — does it mean JAK-STAT? Cytokine Growth Factor Rev. 19, 383–394 (2008).
Melen, K. et al. Importin alpha nuclear localization signal binding sites for STAT1, STAT2, and influenza A virus nucleoprotein. J. Biol. Chem. 278, 28193–28200 (2003).
Decker, T., Kovarik, P. & Meinke, A. GAS elements: a few nucleotides with a major impact on cytokine-induced gene expression. J. Interferon Cytokine Res. 17, 121–134 (1997).
Ramana, C. V. et al. Stat1-independent regulation of gene expression in response to IFN-gamma. Proc. Natl Acad. Sci. USA 98, 6674–6679 (2001). This study determines that the IFNγ-induced immediate-early genes Myc, Jun and Jund, the transcription factor C/EBPβ and the chemokines CCL3 and CCL7 are induced via a non-canonical STAT1-independent mechanism.
Stephens, J. M., Lumpkin, S. J. & Fishman, J. B. Activation of signal transducers and activators of transcription 1 and 3 by leukemia inhibitory factor, oncostatin-M, and interferon-gamma in adipocytes. J. Biol. Chem. 273, 31408–31416 (1998).
Greenlund, A. C. et al. STAT recruitment by tyrosine-phosphorylated cytokine receptors: an ordered reversible affinity-driven process. Immunity 2, 677–687 (1995).
Qi, Y. et al. Elucidating the crosstalk mechanism between IFN-gamma and IL-6 via mathematical modelling. BMC Bioinformatics 14, 41 (2013).
Qing, Y., Costa-Pereira, A. P., Watling, D. & Stark, G. R. Role of tyrosine 441 of interferon-gamma receptor subunit 1 in SOCS-1-mediated attenuation of STAT1 activation. J. Biol. Chem. 280, 1849–1853 (2005).
Gresser, I., Tovey, M. G., Maury, C. & Chouroulinkov, I. Lethality of interferon preparations for newborn mice. Nature 258, 76–78 (1975).
Madonna, S. et al. The IFN-gamma-dependent suppressor of cytokine signaling 1 promoter activity is positively regulated by IFN regulatory factor-1 and Sp1 but repressed by growth factor independence-1b and Krüppel-like factor-4, and it is dysregulated in psoriatic keratinocytes. J. Immunol. 185, 2467–2481 (2010).
Liau, N. P. D. et al. The molecular basis of JAK/STAT inhibition by SOCS1. Nat. Commun. 9, 1558 (2018).
Eshhar, Z., Waks, T., Gross, G. & Schindler, D. G. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc. Natl Acad. Sci. USA 90, 720–724 (1993).
Maude, S. L., Teachey, D. T., Porter, D. L. & Grupp, S. A. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 125, 4017–4023 (2015).
Mauldin, I. S. et al. Intratumoral interferon-gamma increases chemokine production but fails to increase T cell infiltration of human melanoma metastases. Cancer Immunol. Immunother. 65, 1189–1199 (2016).
Strauss, J. et al. First-in-human phase i trial of a tumor-targeted cytokine (NHS-IL12) in subjects with metastatic solid tumors. Clin. Cancer Res. 25, 99–109 (2019).
Lyerly, H. K., Osada, T. & Hartman, Z. C. Right time and place for IL12: targeted delivery stimulates immune therapy. Clin. Cancer Res. 25, 9–11 (2019).
Chen, F. et al. Neoantigen identification strategies enable personalized immunotherapy in refractory solid tumors. J. Clin. Invest. 129, 2056–2070 (2019).
Hollingsworth, R. E. & Jansen, K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 4, 7 (2019).
Kawai, T. & Akira, S. TLR signaling. Cell Death Differ. 13, 816–825 (2006).
Hart, O. M., Athie-Morales, V., O’Connor, G. M. & Gardiner, C. M. TLR7/8-mediated activation of human NK cells results in accessory cell-dependent IFN-gamma production. J. Immunol. 175, 1636–1642 (2005).
Zobywalski, A. et al. Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70. J. Transl. Med. 5, 18 (2007).
Wagner, T. L. et al. Modulation of TH1 and TH2 cytokine production with the immune response modifiers, R-848 and imiquimod. Cell Immunol. 191, 10–19 (1999).
Rodell, C. B. et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat. Biomed. Eng. 2, 578–588 (2018).
Chuang, Y.-C. et al. Adjuvant effect of Toll-like receptor 9 activation on cancer immunotherapy using checkpoint blockade. Front. Immunol. 11, 1075 (2020).
Zhang, H. et al. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J. Control. Release 330, 1080–1094 (2021).
Acknowledgements
The authors thank everyone in the Vignali laboratory for all their constructive comments and advice. This work was supported by the US National Institutes of Health (F32 CA247004-01 and T32 CA082084 to A.M.G.; P01 AI108545, R01 CA203689 and P30 CA047904 to D.A.A.V.).
Author information
Authors and Affiliations
Contributions
The authors contributed equally to all aspects of the article.
Corresponding author
Ethics declarations
Competing interests
D.A.A.V. is a co-founder and shareholder of Novasenta and Tizona, a shareholder of Oncorus and Werewolf, has patents licensed and receives royalties from Astellas and Bristol Myers Squibb, is a scientific advisory board member for Tizona, Werewolf, F-Star and Bicara, is a consultant for Astellas, Bristol Myers Squibb, Almirall and Incyte, and receives research funding from Bristol Myers Squibb, Astellas and Novasenta. The other authors declare no competing interests.
Additional information
Peer review information
Nature Reviews Immunology thanks A. Ribas and the other, anonymous, reviewers for their contribution to the peer review of this work.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Glossary
- Programmed cell death 1 ligand 1
-
(PDL1). A ligand that binds to programmed cell death 1 (PD1) on T cells to inhibit their activation, proliferation and cytokine production. PDL1 is also known as CD274 and B7-H1, and PDL2 is also known as CD273 or B7-C.
- Epigenetic regulation
-
Control of gene expression through phenotypic changes that do not alter the DNA sequence. Examples include DNA methylation, histone modifications, microRNAs, long non-coding RNAs and nucleosome positioning.
- Invariant NK T cells
-
(iNKT cells). Innate-like T cells that express a T cell receptor α-chain that recognizes lipid antigens presented by the non-classical MHC molecule CD1d expressed on dendritic cells.
- γδ T cells
-
T cells that express T cell receptor γ and δ chains and represent 1–4% of the T cell population. They produce interferon-γ (IFNγ) rapidly following activation in a non-MHC-restricted manner by tumour-derived lipids, glycoproteins and phosphorus-containing compounds.
- Adaptive immune resistance
-
The upregulation of immunosuppressive mechanisms in response to chronic proinflammatory stimuli.
Rights and permissions
About this article
Cite this article
Gocher, A.M., Workman, C.J. & Vignali, D.A.A. Interferon-γ: teammate or opponent in the tumour microenvironment?. Nat Rev Immunol 22, 158–172 (2022). https://doi.org/10.1038/s41577-021-00566-3
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41577-021-00566-3
This article is cited by
-
ATLAS-seq: a microfluidic single-cell TCR screen for antigen-reactive TCRs
Nature Communications (2025)
-
Single-cell profiling of response to neoadjuvant chemo-immunotherapy in surgically resectable esophageal squamous cell carcinoma
Genome Medicine (2024)
-
Characterizing and forecasting neoantigens-resulting from MUC mutations in COAD
Journal of Translational Medicine (2024)
-
Integrated network pharmacology and bioinformatics to identify therapeutic targets and molecular mechanisms of Huangkui Lianchang Decoction for ulcerative colitis treatment
BMC Complementary Medicine and Therapies (2024)
-
CTLs heterogeneity and plasticity: implications for cancer immunotherapy
Molecular Cancer (2024)