Abstract
Although type I IFNs were initially described based on their anti-viral properties, it was quickly realized that these cytokines had anti-proliferative and anti-cancer activities. These observations ultimately led to the clinical development and utility of IFN-α2b for the treatment of patients with melanoma, renal cell carcinoma, and chronic myelogenous leukemia, among others. However, the mechanism of action of type I IFNs in vivo was never fully elucidated, and the pleiotropic effects of IFNs on multiple cell types had made it challenging to decipher. Advancement of genetically engineered mouse models has provided new tools for interrogating these mechanisms. Recent evidence has indicated that spontaneous innate immune sensing of cancers that leads to adaptive immune responses is dependent on host type I IFN production and signaling. The major innate immune receptor pathway that leads to type I IFN production in response to a growing tumor appears to be the STING pathway of cytosolic DNA sensing. STING agonists drive type I IFN production and are impressively therapeutic in mouse tumor models. Targeting low doses of type I IFNs to the tumor microenvironment also promotes anti-tumor activity via host adaptive immunity that is T cell-dependent. However, high doses of intratumoral type I IFNs largely function via an anti-angiogenic effect. Understanding these mechanistic details should enable improved clinical manipulation of the type I IFN system in cancer.
1. Endogenous innate immune sensing of cancer involves host type I IFN signaling
A major subset of human cancer patients shows evidence for a spontaneous T cell response against their tumor as evidence by a T cell-inflamed tumor microenvironment gene expression signature and the presence of CD8+ T cells by immunohistochemistry (1–3). Tumor antigen-specific T cells have been identified among this infiltrate, arguing that at least a component of this T cell populations is directly tumor-reactive (4–6). The positive prognostic import of this phenotype (7, 8) suggests that this smoldering immune response is attempting to control the tumor, but without the ultimate success of tumor elimination. In fact, recent evidence suggests that this subset of tumors is dominated by immune inhibitory pathways that restrain T cell function and ultimately allow tumor outgrowth (9, 10). Targeting these immune inhibitory pathways has led to a new class of cancer immunotherapies, including anti-CTLA-4 and anti-PD-1/PD-L1 mAbs (11–13). As such, understanding the underlying molecular mechanisms that control the presence or absence of this spontaneous T cell-inflamed tumor microenvironment phenotype has evolved into an active area of investigation.
Productive T cell activation and differentiation into effector cells is thought to depend upon proper innate immune signaling upstream, particularly at the level of dendritic cells (DCs). However, how a sterile tumor could potentially lead to T cell priming in vivo in the absence of exogenous pathogen-associated molecular patterns (PAMPs) had been elusive. Interrogation of melanoma gene expression profiles for evidence of innate immune activation pathways that might be associated with the presence of T cell transcripts revealed evidence for a positive correlation with a type I IFN gene signature (14, 15). Based on this observation, preclinical mouse model experiments were performed and indeed revealed that type I IFN signaling was required upstream for spontaneous T cell priming against tumor-associated antigens in vivo (14, 15). Similarly, host type I IFN signaling was required for spontaneous regression of immunogenic tumors. IFN-β was found to be rapidly induced upon tumor implantation in vivo, largely by CD11c+ DCs. Detailed mapping using mixed bone marrow chimeras and conditional type I IFNR−/− mice demonstrated that type I IFN signaling had to occur within the Batf3-lineage CD8α+ subset of DCs (16). Thus, like for most viral infections, host type I IFN signaling is crucial for an adaptive immune response against tumors in vivo. Knowledge of this requirement for natural immunity against tumors has provided new insights to guide therapeutic considerations for type I IFNs in the cancer context.
2. A major mechanism of innate immune sensing that leads to type I IFN production is through the STING pathway
The observation that type I IFN production was induced in response to a growing tumor in vivo raised the next level question of which innate immune pathway might be “sensing” the presence of tumor and thereby promoting induction of type I IFN gene expression. From the infectious disease context, several distinct receptor and signaling systems have been identified that could ultimately lead to type I IFN transcription. These are the TLR pathways that signal via MyD88 and/or TRIF (15), the cytosolic RNA sensing pathways that signal via MAVS, and the cytosolic DNA sensing pathway that signals through STING (17). Gene-targeted mice lacking these individual pathways were employed in order to evaluate whether each of these might be required for induction of type I IFN production and spontaneous T cell responses against tumor-associated antigens in vivo. Interestingly, no defect was seen in mice deficient in MyD88, TRIF, or MAVs, nor was any defect observed in specific TLR−/− mice. However, a major defect in both type I IFN induction and T cell priming was observed in STING−/− mice, as well is in mice lacking the downstream transcription factor IRF3. In vitro, tumor-derived DNA was capable of inducing STING pathway activation and type I IFN production when introduced into the cytosol of DCs in vitro. Moreover, labeled tumor-associated DNA was identified within the cytosol of host DCs within the tumor microenvironment in vivo. Together, these data argue that a major mechanism for innate immune sensing of tumors occurs via cytosolic DNA sensing through the STING pathway within tumor-infiltrating DCs (Woo, Gajewski et al., Immunity, In Press 2014).
Recent data have indicated that radiation therapy for cancer also involves induction of type I IFNs by host DCs and facilitation of an adaptive immune response that contributes to tumor control in vivo (18). Thus, it was of interest to determine whether the STING pathway was also important for the therapeutic effect of radiation. Indeed, the efficacy of radiation was largely ablated in STING−/− hosts, whereas no defect was observed using mice lacking specific TLR signaling pathways (Deng et al, Immunity, In Press 2014). Thus, it is tempting to speculate that radiation facilitates the proper acquisition of tumor-derived DNA by host DCs in the tumor microenvironment as a major mechanism of immune priming in response to tumor irradiation.
3. Therapeutic strategies to promote endogenous type I IFN production
Based on the discovered importance of the STING pathway and host type I IFN induction as a bridge to adaptive immune responses against tumors in vivo, strategies to mimic or activate host innate immune sensing pathways as a cancer therapeutic are being considered. 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) is a drug that has anti-tumor activity in mouse models but which failed to show benefit in advanced stage clinical trials in cancer patients (19). However, the molecular target of DMXAA had not been defined at the time of those studies. Recently, it was reported that DMXAA directly binds to murine STING, and a crystal structure of the interaction has been solved (20). Importantly, this compound fails to bind human STING, which probably explains the failure of this agent clinically (21). Using mouse transplantable tumor models, we have observed that intratumoral administration of DMXAA exerts profound anti-tumor activity that is completely dependent on host STING. Treatment is associated with a marked increase in systemic anti-tumor T cell responses, which is capable of regressing non-injected distant tumors. Novel cyclic dinucleotides have been synthesized that do indeed bind human STING and induce type I IFNs from human dendritic cells in vitro (Corrales, Gajewski et al., manuscript submitted). Thus, it is hoped that clinical grade drug will be developed for future translation into human phase I trials.
4. Therapeutic strategies to target type I IFNs to the tumor microenvironment
As an alternative to eliciting type I IFN production from host DCs in vivo, targeting of type I IFNs directly into the tumor site also is being explored. Interestingly, the mechanism of anti-tumor activity seems to vary depending on the specific strategy utilized, which likely is related to the dose and duration of type I IFN presence within the tumor microenvironment. We found that transfection of tumor cells to express high levels of IFN-β led to tumor regression that was largely independent of host adaptive immunity. Rather, this approach led to an elimination of the tumor vasculature, consistent with an anti-angiogenic effect. Most of the therapeutic effect was preserved in RAG−/− and NK cell-depleted mice. Surprisingly, bone marrow chimera experiments revealed a major role for type I IFN signaling on non-hematopoietic cells. Moreover, conditional type I IFNR−/− mice lacking type I IFN signaling exclusively on Tie2+ cells lost the therapeutic effect of high-dose IFN-β, arguing for a direct effect on the tumor vasculature (22).
In contrast, an alternative approach to introduce IFN-β into the tumor microenvironment is by the use of tumor-targeting monoclonal antibodies (mAbs) coupled to IFN-β protein as a payload. Indeed, coupling of IFN-β either to anti-Her2 or anti-EGFR mAbs led to a synergistic therapeutic effect in the appropriate models, that was completely T cell-dependent. In this case, type I IFN signaling was required on bone marrow-derived cells. Conditional type I IFNR−/− mice lacking type I IFN signaling specifically on CD11c+ cells lost the therapeutic effect with this strategy (23). Thus, transient expression of lower doses of IFN-β within the tumor microenvironment may facilitate adaptive immunity to tumors. The distinction between these in vivo therapeutic effects is summarized in Figure 1.
Figure 1. Therapeutic mechanisms of intratumoral type I IFNs appears to vary with dose.
Low doses of endogenous IFN-β produced in response to innate immune sensing of cancer is upstream from T cell priming and drives an adaptive immune response. Targeting of low doses of IFN-β into tumor sites with mAb conjugation also facilitates induction of T cell responses against tumor antigens. However, high sustained doses of IFN-β, which also has a therapeutic effect, largely works via a T cell-independent mechanism. Instead, a direct anti-angiogenic effect is observed, which is mediated through the type I IFN receptor (IFNAR) expressed by endothelial cells.
5. Implications and future directions
The identification of the host STING pathway as a critical mechanism of innate immune sensing of cancers, that drives type I IFN production and facilitates anti-tumor immunity, raises the question of whether non-T cell-inflamed tumors lack activation of the STING pathway in vivo. It will thus be critical to phenotype DC subsets within human tumors, and understand if Batf3-lineage DCs are present and activated within the non-T cell-inflamed tumors. Understanding the level of block in tumors of this phenotype should instruct on how to intervene therapeutically to overcome the putative immune defect involved. The development of human STING agonists for clinical translation should move forward, to test whether direct STING activation within the tumor microenvironment might be therapeutic in patients. Such studies should involve careful pharmacodynamic biomarker evaluation, for example to determine whether post-treatment biopsies who upregulation of type I IFNs. The pursuit of IFN-β/mAb fusions is also ripe for clinical translation, for example with anti-Her2 mAb. The recent success of TDM1 as a mAb-chemotherapeutic toxin fusion has increased enthusiasm for such payload approaches. Ultimately, refining strategies for delivering type I IFNs directly within the tumor microenvironment should improve the therapeutic window for these cytokines and increase effectiveness against cancer.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14:1014–1022. doi: 10.1038/ni.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69:3077–3085. doi: 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gajewski TF, Louahed J, Brichard VG. Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy. Cancer J. 2010;16:399–403. doi: 10.1097/PPO.0b013e3181eacbd8. [DOI] [PubMed] [Google Scholar]
- 4.Harlin H, Kuna TV, Peterson AC, Meng Y, Gajewski TF. Tumor progression despite massive influx of activated CD8(+) T cells in a patient with malignant melanoma ascites. Cancer Immunol Immunother. 2006;55:1185–1197. doi: 10.1007/s00262-005-0118-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Appay V, Jandus C, Voelter V, Reynard S, Coupland SE, Rimoldi D, Lienard D, Guillaume P, Krieg AM, Cerottini JC, Romero P, Leyvraz S, Rufer N, Speiser DE. New generation vaccine induces effective melanoma-specific CD8+ T cells in the circulation but not in the tumor site. J Immunol. 2006;177:1670–1678. doi: 10.4049/jimmunol.177.3.1670. [DOI] [PubMed] [Google Scholar]
- 6.Mortarini R, Piris A, Maurichi A, Molla A, Bersani I, Bono A, Bartoli C, Santinami M, Lombardo C, Ravagnani F, Cascinelli N, Parmiani G, Anichini A. Lack of terminally differentiated tumor-specific CD8+ T cells at tumor site in spite of antitumor immunity to self-antigens in human metastatic melanoma. Cancer Res. 2003;63:2535–2545. [PubMed] [Google Scholar]
- 7.Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, Tosolini M, Camus M, Berger A, Wind P, Zinzindohoue F, Bruneval P, Cugnenc PH, Trajanoski Z, Fridman WH, Pages F. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
- 8.Galon J, Pages F, Marincola FM, Thurin M, Trinchieri G, Fox BA, Gajewski TF, Ascierto PA. The immune score as a new possible approach for the classification of cancer. J Transl Med. 2012;10:1. doi: 10.1186/1479-5876-10-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, Gajewski TF. Up-Regulation of PD-L1, IDO, and Tregs in the Melanoma Tumor Microenvironment Is Driven by CD8+ T Cells. Sci Transl Med. 2013;5:200ra116. doi: 10.1126/scitranslmed.3006504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gajewski TF, Woo SR, Zha Y, Spaapen R, Zheng Y, Corrales L, Spranger S. Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment. Curr Opin Immunol. 2013;25:268–276. doi: 10.1016/j.coi.2013.02.009. [DOI] [PubMed] [Google Scholar]
- 11.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med. 2010 doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N Engl J Med. 2012 doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, Alaparthy S, Berman D, Jure-Kunkel M, Siemers NO, Jackson JR, Shahabi V. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother. 2011 doi: 10.1007/s00262-011-1172-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, Gajewski TF. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. J Exp Med. 2011 doi: 10.1084/jem.20101159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fuertes MB, Woo SR, Burnett B, Fu YX, Gajewski TF. Type I interferon response and innate immune sensing of cancer. Trends Immunol. 2012 doi: 10.1016/j.it.2012.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn GP, Archambault JM, Lee H, Arthur CD, White JM, Kalinke U, Murphy KM, Schreiber RD. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208:1989–2003. doi: 10.1084/jem.20101158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009;461:788–792. doi: 10.1038/nature08476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Burnette BC, Liang H, Lee Y, Chlewicki L, Khodarev NN, Weichselbaum RR, Fu YX, Auh SL. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011;71:2488–2496. doi: 10.1158/0008-5472.CAN-10-2820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Head M, Jameson MB. The development of the tumor vascular-disrupting agent ASA404 (vadimezan, DMXAA): current status and future opportunities. Expert Opin Investig Drugs. 2010;19:295–304. doi: 10.1517/13543780903540214. [DOI] [PubMed] [Google Scholar]
- 20.Gao P, Ascano M, Zillinger T, Wang W, Dai P, Serganov AA, Gaffney BL, Shuman S, Jones RA, Deng L, Hartmann G, Barchet W, Tuschl T, Patel DJ. Structure-function analysis of STING activation by c[G(2′,5′)pA(3′,5′)p] and targeting by antiviral DMXAA. Cell. 2013;154:748–762. doi: 10.1016/j.cell.2013.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Conlon J, Burdette DL, Sharma S, Bhat N, Thompson M, Jiang Z, Rathinam VA, Monks B, Jin T, Xiao TS, Vogel SN, Vance RE, Fitzgerald KA. Mouse, but not Human STING, Binds and Signals in Response to the Vascular Disrupting Agent 5,6-Dimethylxanthenone-4-Acetic Acid. J Immunol. 2013;190:5216–5225. doi: 10.4049/jimmunol.1300097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Spaapen RM, Leung MY, Fuertes MB, Kline JP, Zhang L, Zheng Y, Fu YX, Luo X, Cohen KS, Gajewski TF. Therapeutic Activity of High-Dose Intratumoral IFN-beta Requires Direct Effect on the Tumor Vasculature. J Immunol. 2014;193:4254–4260. doi: 10.4049/jimmunol.1401109. [DOI] [PubMed] [Google Scholar]
- 23.Yang X, Zhang X, Fu ML, Weichselbaum RR, Gajewski TF, Guo Y, Fu YX. Targeting the tumor microenvironment with interferon-beta bridges innate and adaptive immune responses. Cancer Cell. 2014;25:37–48. doi: 10.1016/j.ccr.2013.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]