Myeloid-derived suppressor cell impact on endogenous and adoptively transferred T cells

Myeloid-derived suppressor cell impact on endogenous and adoptively transferred T cells

Available online at www.sciencedirect.com ScienceDirect Myeloid-derived suppressor cell impact on endogenous and adoptively transferred T cells Ainho...

633KB Sizes 0 Downloads 45 Views

Available online at www.sciencedirect.com

ScienceDirect Myeloid-derived suppressor cell impact on endogenous and adoptively transferred T cells Ainhoa Arina1 and Vincenzo Bronte2 Novel models of autochthonous tumorigenesis and adoptive T cell therapy (ATT) are providing new clues regarding the pro-tumorigenic and immunosuppressive effects of myeloidderived suppressor cells (MDSC), and their interaction with T cells. New findings are shifting the perception of the main level at which MDSC act, from direct cell-to-cell suppression to others, such as limiting T cell infiltration. Adoptively transferred, high-avidity T cells recognizing peptides with high-affinity for MHC-I eliminated large tumors. However, low-avidity T cells or low-affinity peptides resulted in failure to eradicate tumors. Manipulation of intratumoral myeloid cells improved the outcome of otherwise unsuccessful ATT. Therefore, therapeutic intervention directed at the tumor stroma might be required when using suboptimal T cells for ATT. Addresses 1 Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA 2 Verona University Hospital, Department of Pathology and Diagnostics, 37134 Verona, Italy Corresponding authors: Arina, Ainhoa ([email protected]) and Bronte, Vincenzo ([email protected])

Current Opinion in Immunology 2015, 33:120–125 This review comes from a themed issue on Tumour immunology Edited by Philip D Greenberg and Hans Schreiber

http://dx.doi.org/10.1016/j.coi.2015.02.006 0952-7915/# 2015 Published by Elsevier Ltd.

Introduction

Oncogene-induced autochthonous cancers [1,2,3,4] have confirmed the critical role of cancer-secreted GM-CSF in driving accrual and function of CD11b+Gr1+ myeloid-derived suppressor cells (MDSC) [5,6], which counteract T cell responses against tumors [7], as already shown in transplantable models [5,6]. Findings from other transgenic tumor models suggest that oncogene-driven tumors can be immunogenic, and generate T cell responses that delay tumor onset [1,8,9], although a few transformed cells escaping immune control are enough to allow cancer progression [9]. Introduction of strong antigens was required for epigenetic selection of low-immunogenicity/antigen-loss variants in Kras/p53 oncogene-driven sarcomas [1], suggesting that these tumors Current Opinion in Immunology 2015, 33:120–125

are less immunogenic than tumors induced by UV light and 3-methylcholanthrene, where immune selection occurred without exogenous introduction of model antigens [10–12], or viral SV40 oncogene-driven tumors [9], where SV40 oncogene/antigen was retained and caused local T cell tolerization. These different mechanisms to evade the immune response may represent different clinical scenarios (e.g., viral vs. non-viral oncogene activation, or mutagen-induced cancer). Indeed cancer patients show different degrees of activation of spontaneous T cell responses [13]. Some tumors, for example, melanomas, present a high number of potentially immunogenic mutations [14]. Additional evidence is needed to understand whether patients with weaker spontaneous responses correspond to those with tumors presenting fewer mutations, or those endowed with more potent cancer-related immune suppression. Since ATT will become a major therapeutic modality, it is important to determine whether MDSC in the tumor microenvironment can suppress cancer eradication by the adoptively transferred T cells. Previous studies using live microscopy of surgically exposed tumors in the EL4/EG7 thymoma model showed cancer antigen-dependent T cell arrest [15] and specific killing of antigen (Ag)+ cells that were interspersed among Ag cancer cells [16]. The time required for killing a single cancer cell was estimated to be 6 h, but the observed bystander killing of Ag cancer cells was minimal [16]. Since most tumors contain at least some Ag cancer cells, achieving some level of bystander killing of these malignant cells that cause relapse is essential for tumor eradication. In the surgically exposed tumors analyzed in the earlier studies, the animal needed to be sacrificed after the imaging session; therefore, the subsequent changes in the imaged tumor could not be followed up. A recent study using a window chamber-based imaging technique [17] allowed, for the first time, real-time longitudinal analysis of the same tumor before, during and after ATT until full eradication or relapse. In this system, significant bystander killing of Ag cells occurred, consistent with earlier experiments [18]. Massive vessel destruction was observed using transgenic T cells targeting a model antigen [17], or natural tumor-specific T cells targeting autochthonous tumor-specific antigens [19]. In contrast to IFNg and TNF [20], perforin was not required for the eradication of the large solid tumors established for several weeks [17]. Stable, cognate antigen-dependent interactions of T cells with stromal cells resulted in the production of higher amounts of IFNg compared to interactions with www.sciencedirect.com

MDSC impact on endogenous and transferred T cells Arina and Bronte 121

cancer cells [17]. This higher relevance of IFNg than perforin in vivo is consistent with previous studies [21,22] and it might underlie the reported 6 h-requirement for cancer cell apoptosis. More studies are required to determine whether the effects on vessels are mediated by IFNg, either directly or indirectly, through the induction of fas/fasL [22,23], or other mechanisms. We will therefore review the most recent advances in understanding the impact of cells of myeloid lineage, and in particular of MDSC, on anti-tumor T cells.

Impact of MDSC on endogenous T cell responses Initial studies reported the ability of cells recognized by the anti-CD11b (Mac-1) and anti-Gr1 monoclonal antibodies and induced by growing tumors [5,24–30] to suppress T cell priming in vivo [5,7,28–30] and effector function in vitro [5,7,27,29,30]. The in vitro assays to test inhibition of T cell function (most frequently proliferation or IFN-g release) by MDSC, were adopted as surrogate for MDSC’s ability to suppress T cell responses. Recently, however, it has become clear that in vitro suppression does not always correlate with in vivo inhibition of T cell responses. For example, tumors growing in immunodeficient hosts induced systemic MDSC and tumorinfiltrating macrophages that were strongly suppressive in vitro; however, those same cells were not able to prevent tumor eradication by adoptively transferred tumor-specific T cells [19]. Similarly, MDSC showed the ability to suppress CTL function in vitro without reflecting the immune status of mice, which could be as diverse as tumor-induced tolerance, neonatal tolerance to tumor antigens, or even systemic immunity against the tumor [4]. Experimental evidence to explain the differences between in vivo and in vitro suppression by MDSC principally indicates three factors: cytokines secreted by tumors, the immune-independent, tumor-promoting effects of MDSC, and a different sensitivity of naı¨ve vs. effector T cells to suppression by MDSC. Whereas other cytokines might expand MDSC from precursors, GM-CSF can license them for full immunosuppressive function [4,5,6,27]. This role is further strengthened by evidence from pancreatic autochthonous tumor models, in which GMCSF produced by the Kras oncogene-transformed cancer cells mediated development and recruitment of MDSC, necessary for tumor progression from pre-invasive stage. The presence of MDSC in tumor was required to counteract prevention of tumor growth by tumor-specific T cells [2,3]. It remains unclear at what level the unique suppressive effect of GMCSF-induced MDSC takes place, but pancreatic model studies suggest an effect in restraining tumor infiltration by T cells. Alternatively, the tumor-promoting functions of MDSC, which are distinct and independent from T cell suppression, www.sciencedirect.com

are well documented and might be more important than immunoregulatory activity for many cancers. Indeed, MDSC promote tumor growth in the absence of an adaptive immune system [24–26] and can facilitate angiogenesis [31] and metastasis [32,33]. Finally, activation status of T cells seems to play an important role in sensitivity to suppression by MDSC. Most studies showing suppression of T cell responses use naı¨ve T cells as responders. An explanation for increased sensitivity of naı¨ve T cells to suppression by MDSC might relate to MDSC’s ability to down-regulate L-selectin levels on T cells; this resulted in decreased homing to lymph nodes where T cells would be activated [34]. Indeed, the function of primed, tumor-specific T cells isolated from lymph nodes was suppressed by MDSC in vitro but not in vivo [29]. In vitro studies suggest that tumor cells, but not MDSC, can directly suppress the effector, lytic machinery of T cells isolated from tumors [35]. Consistent with these findings, adoptively transferred tumor-specific memory T cells but not naı¨ve T cells eliminated tumors established in immunodeficient mice that had systemic and intratumoral MDSC but not Tregs [19]. In addition, pre-existing endogenous T cell memory responses were not affected by systemic immune suppression in tumor-bearing mice harboring MDSC as well as Tregs [19].

Impact of MDSC on ATT Three possible outcomes have been described for tumorinfiltrating myeloid cells after ATT, in tumors that responded to therapy: 1. Elimination: an increase in non-viable stromal cells was observed in tumors treated with T cells recognizing CD11b+ cells that cross-present antigen from neighboring cancer cells [18,36,37]. 2. Change: in many cases, the anti-tumoral effects of ATT required a change in either subset distribution or phenotype of MDSC, typically toward a more proinflammatory and less suppressive phenotype [38,39]. Such change can be pursued experimentally with the aim of altering MDSC differentiation and hence increase efficacy of ATT. In fact, the ability of lowdose chemotherapy to function as adjuvant for poorly effective CD8T cells recognizing mouse telomerase antigen was related to long-term elimination of proliferating, Ly6Chi suppressive cells in the marginal zone of the spleen of tumor-bearing hosts [40,41]. 3. No elimination or detectable phenotypic changes: Under certain conditions, ATT can achieve tumor eradication without detectable changes in the composition or phenotype of intratumoral myeloid cells [19]. Differences in the tumor microenvironment or in the ‘quality’ of T cells could explain these varying scenarios. Current Opinion in Immunology 2015, 33:120–125

122 Tumour immunology

Tumor microenvironment can determine the level of immune suppression: key factors GM-CSF

GM-CSF controls acquisition of the most suppressive phenotype by MDSC. Consistently, MDSC induced by GM-CSF-overexpressing B16 tumors impaired the antitumor effect of adoptively transferred, melanoma-specific pmel T cells, by limiting T cell infiltration [42]. CSF-1/M-CSF

CSF-1/M-CSF is a critical regulator of the Ly6C+ monocytes and their differentiation into tumor-associated macrophages [43]. Several female reproductive cancers secrete CSF-1 [44,45]. A small molecule inhibitor of CSF-1, PLX3397, reduced the number of tumor-associated macrophages and polarized the remaining, leading to increased infiltration by adoptively transferred T cells and enhanced antitumor effects [46]. Concurring with these results, treatment with anti-CSF-1-receptor mAb caused reduction in tumor-infiltrating macrophages in mouse and human tumors, and clinical objective responses in diffusetype giant cell tumors [47]. G-CSF

G-CSF is critical for the production of neutrophils that, by producing elastase and TIMP-free MMP9, play a pivotal role in the activation of latent TGF-b and in freeing stromal progenitors from their reservoirs [48]. Molecular programs in tumor-infiltrating myeloid cells

Control of cEBPb transcription factor by microRNA 1423p and the stress-response protein CHOP is key to drive the immunosuppressive program in MDSC within the tumor environment, in part by modulating their response to locally produced IL-6 [49–51]. Interfering with this molecular pathway enhances the therapeutic activity of otherwise ineffective antitumor CD8+ T cells [49]. HMGB1

HMGB1, produced by intratumoral macrophages, downregulates L-selectin on T cells while attracting MDSC progenitors [52]. Effects of the microbiota

In a recent study, gut microbiota ‘primed’ myeloid cells to produce TNF and ROS that mediated antitumor effects of CpG immunotherapy and platinum chemotherapy, respectively, in the absence of adaptive immunity [53]. However, whether microbiota dysbiosis can negatively influence ATT requires further investigation. Immunosuppressive effects of non-myeloid tumor stroma

Stromal cells expressing FAP (Fibroblast Activation Protein) suppress T cell responses against tumors [54]. Recent studies indicate the dense extracellular matrix and fibroblast-derived CXCL12 as key factors ‘sequestering’ T cells away from tumor cells [55,56,57]. Current Opinion in Immunology 2015, 33:120–125

Tumor burden

Immune suppression and/or T cell dysfunction induced by tumors impair T cell-mediated tumor destruction [58,59]. In a murine lymphoma model, high tumor burdens induced tumor-Ag-specific T cell dysfunction, which was not mediated by MDSC, but by cancer cells presenting antigen [60]. A low non-lymphoablative dose of chemotherapy before the ATT led to an antitumoral effect, through reduction of tumor burden below a critical threshold.

The quality of T cells can determine the outcome of ATT Not all T cells are equal

As discussed, antigen-experienced effector or memory T cells seem to be more intrinsically resistant to suppression by MDSC than naı¨ve T cells. Retrospective analysis of clinical trials using adoptively transferred TILs for melanoma patients show that the parameters correlating with tumor regression include the absolute numbers of infused T cells, short in vitro culture duration, CD27 expression in vivo on transferred CD8+ T cells, and cells with a higher capacity to proliferate and persist following ATT [61]. Preclinical studies suggest that T cell donor age can critically determine an optimal T cell quality for ATT as well [62]. A high functional avidity of TCR is also desirable. Avidity is closely related to affinity, although the contribution of affinity to avidity seems to reach a plateau around 10 mM [63]. Interestingly, six TCRs found in human melanomas, with affinities ranging between 1.4 and 60 mM, produced similar antitumoral effects in vivo. A TCR with affinity of 99 nM, however, conferred poorer antitumoral activity. Since the affinity range of natural TCR is about 1–100 nM, these results suggest that TCRs with affinities in the lower side of the natural range should be avoided for ATT. However, TCRs with high avidity for self-antigens might expose the T cell bearing them to a more robust tolerization [64]. Not all targets are equal

After multiple clinical trials using T cells that targeted antigens expressed by the tumor and normal tissues, it is increasingly clear that the efficacy of such shared Ags will always be limited by sometimes life-threatening autoimmunity, because antitumoral effect and autoimmunity go ‘hand in hand’ [65]. Consistently, in human melanoma, the TCR affinity threshold for antitumor effects and autoimmunity is the same [63]. Therefore, the choice of target antigens should be ideally directed to unique tumor-specific antigens, generated by either somatic mutations or viral infection. As an acceptable alternative, antigens shared by tumors and non-essential tissues can also be used (such as CD19). Another important aspect of target selection is the affinity for MHC-I. Contrary to TCR affinity, which has a relatively minor contribution to the outcome of ATT, the affinity of the target peptide for presenting MHC class I molecules can predict either www.sciencedirect.com

MDSC impact on endogenous and transferred T cells Arina and Bronte 123

Figure 1

Chemotherapy Radiotherapy MDSC blockade

Cancer cells High-affinity Ag

Low-affinity Ag

IFN-γ

High-avidity T cell

Low-avidity T cell

Ag crosspressentation

Myeloid cell

No other therapy required

Combination therapy required Current Opinion in Immunology

Two distinct scenarios following ATT. Left panel: high-avidity interactions between TCR and MHC-I/tumor-antigen peptide complexes on stromal cells are sufficient to allow tumor eradication by release of cytokines, such as IFN-g, affecting vascular and stromal compartment including MDSC. Right panel: below a certain avidity threshold, ATT is ineffective unless combined with other treatments capable to alter the immunosuppressive microenvironment. These include, chemotherapy and radiotherapy, which can affect both cancer and stromal cells, or more selective treatments specifically targeting MDSC, here representing the myeloid component with immune regulatory function within the tumor. In this diagram, only myeloid cells are shown for simplicity, but other stromal cell components might also contribute to the outcome of ATT, as detailed in the article.

tumor eradication or relapse [66]. High affinities (less than 10 nM) were required for tumor eradication, which is a narrow window considering that the affinity range measured for natural peptides was between 1 and 20 000 nM. Consistently, TILs from patients who had objective responses to ATT recognized high-affinity mutant tumor-specific peptides [67,68]. Recent advances in wholeexome sequencing and class I MHC binding-prediction now enable the identification of tumor-specific neoantigens for individual patients [69]. Therefore, choosing tumor-specific neoantigens with the highest MHC class I molecule affinity will be critical for optimal ATT results.

Conclusions Three elements determine the success of ATT: tumor microenvironment, quality of T cells and quality of the antigen targeted. Based on available evidence, we propose that proficient, ‘high-quality T cells’ targeting tumor-specific peptides with high affinity to the presenting MHC molecule could be sufficient therapy for cancer. ‘Suboptimal T cells’ could require additional therapeutic intervention directed at modifying the tumor microenvironment, to help T cells overcome immune suppression posed by myeloid cells, cancer cells and fibroblastic stroma (Figure 1).

Acknowledgments This work was supported by grants from the Italian Ministry of Health; Italian Ministry of Education (FIRB cup: B31J11000420001), Universities,

www.sciencedirect.com

and Research; Italian Association for Cancer Research (AIRC, grants 6599, 12182 and 14103) to V.B, as well as the Ludwig Foundation (A.A.). We especially thank Leticia Corrales for artwork design and Amy Huser for editorial help.

References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest 1.

DuPage M, Mazumdar C, Schmidt LM, Cheung AF, Jacks T: Expression of tumour-specific antigens underlies cancer immunoediting. Nature 2012, 482:405-409.

2. 

Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, Vonderheide RH: Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012, 21:822-835. The first demonstration that GM-CSF, produced by transformed cells under the action of Kras oncogene, is important to activate MDSC that inhibit antitumor T cells and allow the progression from intraepithelial to invasive pancreatic cancer.

3. 

Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D: Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 2012, 21:836-847. See annotation to [2].

4. 

Schmidt K, Zilio S, Schmollinger JC, Bronte V, Blankenstein T, Willimsky G: Differently immunogenic cancers in mice induce immature myeloid cells that suppress CTL in vitro but not in vivo following transfer. Blood 2013, 121:1740-1748. While cells with MDSC phenotype and in vitro immune suppressive functions can be generated in different autochtonous tumor models without exerting immune regulatory activity in vivo, GM-CSF is a key factor to allow a full maturation of the suppressive program. Current Opinion in Immunology 2015, 33:120–125

124 Tumour immunology

5.

6.

Dolcetti L, Peranzoni E, Ugel S, Marigo I, Fernandez Gomez A, Mesa C, Geilich M, Winkels G, Traggiai E, Casati A et al.: Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur J Immunol 2010, 40:22-35. Fu YX, Watson G, Jimenez JJ, Wang Y, Lopez DM: Expansion of immunoregulatory macrophages by granulocyte-macrophage colony-stimulating factor derived from a murine mammary tumor. Cancer Res 1990, 50:227-234.

cancer eradication. Proc Natl Acad Sci U S A 2013, 110:2276-2281. 23. Kerkar SP, Leonardi AJ, van Panhuys N, Zhang L, Yu Z, Crompton JG, Pan JH, Palmer DC, Morgan RA, Rosenberg SA et al.: Collapse of the tumor stroma is triggered by IL-12 induction of Fas. Mol Ther 2013, 21:1369-1377. 24. Pekarek LA, Starr BA, Toledano AY, Schreiber H: Inhibition of tumor growth by elimination of granulocytes. J Exp Med 1995, 181:435-440.

7.

Bronte V, Wang M, Overwijk WW, Surman DR, Pericle F, Rosenberg SA, Restifo NP: Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1+/Gr-1+ cells. J Immunol 1998, 161:5313-5320.

25. Seung LP, Rowley DA, Dubey P, Schreiber H: Synergy between T-cell immunity and inhibition of paracrine stimulation causes tumor rejection. Proc Natl Acad Sci U S A 1995, 92:6254-6258.

8.

Willimsky G, Blankenstein T: Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature 2005, 437:141-146.

26. Seung LP, Seung SK, Schreiber H: Antigenic cancer cells that escape immune destruction are stimulated by host cells. Cancer Res 1995, 55:5094-5100.

9.

Willimsky G, Schmidt K, Loddenkemper C, Gellermann J, Blankenstein T: Virus-induced hepatocellular carcinomas cause antigen-specific local tolerance. J Clin Invest 2013, 123:1032-1043.

27. Bronte V, Chappell DB, Apolloni E, Cabrelle A, Wang M, Hwu P, Restifo NP: Unopposed production of granulocytemacrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation. J Immunol 1999, 162:5728-5737.

10. Ward PL, Koeppen H, Hurteau T, Schreiber H: Tumor antigens defined by cloned immunological probes are highly polymorphic and are not detected on autologous normal cells. J Exp Med 1989, 170:217-232. 11. Dubey P, Hendrickson RC, Meredith SC, Siegel CT, Shabanowitz J, Skipper JC, Engelhard VH, Hunt DF, Schreiber H: The immunodominant antigen of an ultraviolet-induced regressor tumor is generated by a somatic point mutation in the DEAD box helicase p68. J Exp Med 1997, 185:695-705. 12. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, Schreiber RD: IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410:1107-1111.

28. Kusmartsev S, Nagaraj S, Gabrilovich DI: Tumor-associated CD8+ T cell tolerance induced by bone marrow-derived immature myeloid cells. J Immunol 2005, 175:4583-4592. 29. Watanabe S, Deguchi K, Zheng R, Tamai H, Wang LX, Cohen PA, Shu S: Tumor-induced CD11b+Gr-1+ myeloid cells suppress T cell sensitization in tumor-draining lymph nodes. J Immunol 2008, 181:3291-3300. 30. Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI: Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol 2010, 184:3106-3116.

13. Gajewski TF, Schreiber H, Fu YX: Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013, 14:1014-1022.

31. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, Matrisian LM, Carbone DP, Lin PC: Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 2004, 6:409-421.

14. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Borresen-Dale AL et al.: Signatures of mutational processes in human cancer. Nature 2013, 500:415-421.

32. Mauti LA, Le Bitoux MA, Baumer K, Stehle JC, Golshayan D, Provero P, Stamenkovic I: Myeloid-derived suppressor cells are implicated in regulating permissiveness for tumor metastasis during mouse gestation. J Clin Invest 2011, 121:2794-2807.

15. Boissonnas A, Fetler L, Zeelenberg IS, Hugues S, Amigorena S: In vivo imaging of cytotoxic T cell infiltration and elimination of a solid tumor. J Exp Med 2007, 204:345-356.

33. Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, Carbone DP, Matrisian LM, Richmond A, Lin PC et al.: Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell 2008, 13:23-35.

16. Breart B, Lemaitre F, Celli S, Bousso P: Two-photon imaging of intratumoral CD8+ T cell cytotoxic activity during adoptive T cell therapy in mice. J Clin Invest 2008, 118:1390-1397. 17. Schietinger A, Arina A, Liu RB, Wells S, Huang J, Engels B, Bindokas V, Bartkowiak T, Lee D, Herrmann A et al.: Longitudinal confocal microscopy imaging of solid tumor destruction following adoptive T cell transfer. Oncoimmunology 2013, 2:e26677. 18. Spiotto MT, Rowley DA, Schreiber H: Bystander elimination of antigen loss variants in established tumors. Nat Med 2004, 10:294-298. 19. Arina A, Schreiber K, Binder DC, Karrison TG, Liu RB, Schreiber H: Adoptively transferred immune T cells eradicate established tumors despite cancer-induced immune suppression. J Immunol 2014, 192:1286-1293. 20. Zhang B, Karrison T, Rowley DA, Schreiber H: IFN-gamma- and TNF-dependent bystander eradication of antigen-loss variants in established mouse cancers. J Clin Invest 2008, 118:1398-1404. 21. Hollenbaugh JA, Reome J, Dobrzanski M, Dutton RW: The rate of the CD8-dependent initial reduction in tumor volume is not limited by contact-dependent perforin, Fas ligand, or TNF-mediated cytolysis. J Immunol 2004, 173:1738-1743. 22. Listopad JJ, Kammertoens T, Anders K, Silkenstedt B, Willimsky G, Schmidt K, Kuehl AA, Loddenkemper C, Blankenstein T: Fas expression by tumor stroma is required for

Current Opinion in Immunology 2015, 33:120–125

34. Hanson EM, Clements VK, Sinha P, Ilkovitch D, OstrandRosenberg S: Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J Immunol 2009, 183:937-944. 35. Monu NR, Frey AB: Myeloid-derived suppressor cells and antitumor T cells: a complex relationship. Immunol Invest 2012, 41:595-613. 36. Zhang B, Zhang Y, Bowerman NA, Schietinger A, Fu YX, Kranz DM, Rowley DA, Schreiber H: Equilibrium between host and cancer caused by effector T cells killing tumor stroma. Cancer Res 2008, 68:1563-1571. 37. Thomas DL, Kim M, Bowerman NA, Narayanan S, Kranz DM, Schreiber H, Roy EJ: Recurrence of intracranial tumors following adoptive T cell therapy can be prevented by direct and indirect killing aided by high levels of tumor antigen crosspresented on stromal cells. J Immunol 2009, 183:1828-1837. 38. Garcia-Hernandez Mde L, Hamada H, Reome JB, Misra SK, Tighe MP, Dutton RW: Adoptive transfer of tumor-specific Tc17 effector T cells controls the growth of B16 melanoma in mice. J Immunol 2010, 184:4215-4227. 39. Kerkar SP, Goldszmid RS, Muranski P, Chinnasamy D, Yu Z, Reger RN, Leonardi AJ, Morgan RA, Wang E, Marincola FM et al.: IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors. J Clin Invest 2011, 121:4746-4757.

www.sciencedirect.com

MDSC impact on endogenous and transferred T cells Arina and Bronte 125

40. Ugel S, Peranzoni E, Desantis G, Chioda M, Walter S, Weinschenk T, Ochando JC, Cabrelle A, Mandruzzato S, Bronte V: Immune tolerance to tumor antigens occurs in a specialized environment of the spleen. Cell Rep 2012, 2:628-639. 41. Bronte V, Pittet MJ: The spleen in local and systemic regulation of immunity. Immunity 2013, 39:806-818. 42. Lesokhin AM, Hohl TM, Kitano S, Cortez C, HirschhornCymerman D, Avogadri F, Rizzuto GA, Lazarus JJ, Pamer EG, Houghton AN et al.: Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment. Cancer Res 2012, 72:876-886. 43. Hume DA, MacDonald KP: Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 2012, 119:1810-1820. 44. Ramakrishnan S, Xu FJ, Brandt SJ, Niedel JE, Bast RC Jr, Brown EL: Constitutive production of macrophage colonystimulating factor by human ovarian and breast cancer cell lines. J Clin Invest 1989, 83:921-926. 45. Kacinski BM: CSF-1 and its receptor in ovarian, endometrial and breast cancer. Ann Med 1995, 27:79-85. 46. Mok S, Koya RC, Tsui C, Xu J, Robert L, Wu L, Graeber TG, West BL, Bollag G, Ribas A: Inhibition of CSF-1 receptor improves the antitumor efficacy of adoptive cell transfer immunotherapy. Cancer Res 2014, 74:153-161. 47. Ries CH, Cannarile MA, Hoves S, Benz J, Wartha K, Runza V, Rey-Giraud F, Pradel LP, Feuerhake F, Klaman I et al.: Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014, 25:846-859. 48. Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L, Ponomaryov T, Taichman RS, Arenzana-Seisdedos F, Fujii N et al.: G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol 2002, 3:687-694. 49. Sonda N, Simonato F, Peranzoni E, Cali B, Bortoluzzi S, Bisognin A, Wang E, Marincola FM, Naldini L, Gentner B et al.: miR-142-3p prevents macrophage differentiation during cancer-induced myelopoiesis. Immunity 2013, 38:1236-1249. 50. Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, Ugel S, Sonda N, Bicciato S, Falisi E et al.: Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 2010, 32:790-802. 51. Thevenot PT, Sierra RA, Raber PL, Al-Khami AA, Trillo-Tinoco J, Zarreii P, Ochoa AC, Cui Y, Del Valle L, Rodriguez PC: The stressresponse sensor chop regulates the function and accumulation of myeloid-derived suppressor cells in tumors. Immunity 2014, 41:389-401.

and migration of T cells into the stroma of human lung tumors. J Clin Invest 2012, 122:899-910. Collagen distribution within the extracellular matrix in tumor influences antitumor immunity by regulating the positioning and migration of T lymphocytes. 56. Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, Marshall JF, Chin-Aleong J, Chelala C, Gribben JG et al.: Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013, 145:1121-1132. 57. Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS,  Connell CM, Roberts EW, Zhao Q, Caballero OL et al.: Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2013, 110:20212-20217. This study suggests a scenario in which tumor-associated fibroblast release CXCL12 that is then dysplayed on cancer cells to restrain the migration of T lymphocytes in pancreatic cancer. Fibroblast elimination or chemokine receptor antagonists synergized with immune checkpoint blockade therapy. 58. Mizoguchi H, O’Shea JJ, Longo DL, Loeffler CM, McVicar DW, Ochoa AC: Alterations in signal transduction molecules in T lymphocytes from tumor-bearing mice. Science 1992, 258:1795-1798. 59. Mullen CA, Urban JL, Van Waes C, Rowley DA, Schreiber H: Multiple cancers. Tumor burden permits the outgrowth of other cancers. J Exp Med 1985, 162:1665-1682. 60. Prato S, Zhan Y, Mintern JD, Villadangos JA: Rapid deletion and inactivation of CTLs upon recognition of a number of target cells over a critical threshold. J Immunol 2013, 191:3534-3544. 61. Crompton JG, Sukumar M, Restifo NP: Uncoupling T-cell expansion from effector differentiation in cell-based immunotherapy. Immunol Rev 2014, 257:264-276. 62. Schreiber K, Arina A, Engels B, Spiotto MT, Sidney J, Sette A, Karrison TG, Weichselbaum RR, Rowley DA, Schreiber H: Spleen cells from young but not old immunized mice eradicate large established cancers. Clin Cancer Res 2012, 18:2526-2533. 63. Zhong S, Malecek K, Johnson LA, Yu Z, Vega-Saenz de Miera E, Darvishian F, McGary K, Huang K, Boyer J, Corse E et al.: T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci U S A 2013, 110:6973-6978. 64. Zhu Z, Singh V, Watkins SK, Bronte V, Shoe JL, Feigenbaum L, Hurwitz AA: High-avidity T cells are preferentially tolerized in the tumor microenvironment. Cancer Res 2013, 73:595-604. 65. Hinrichs CS, Rosenberg SA: Exploiting the curative potential of adoptive T-cell therapy for cancer. Immunol Rev 2014, 257:56-71.

52. Parker KH, Sinha P, Horn LA, Clements VK, Yang H, Li J, Tracey KJ, Ostrand-Rosenberg S: HMGB1 enhances immune suppression by facilitating the differentiation and suppressive activity of myeloid-derived suppressor cells. Cancer Res 2014, 74:5723-5733.

66. Engels B, Engelhard VH, Sidney J, Sette A, Binder DC, Liu RB, Kranz DM, Meredith SC, Rowley DA, Schreiber H: Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. Cancer Cell 2013, 23:516-526.

53. Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N,  Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S et al.: Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342:967-970. This pionering study suggests that therapeutic efficacy of both chemotherapy and immunotherapy rely on a tonic signal delivered from gut microbiota to myeloid cells infiltrating the tumor.

67. Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E et al.: Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013, 19:747-752.

54. Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, Gopinathan A, Tuveson DA, Fearon DT: Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010, 330:827-830. 55. Salmon H, Franciszkiewicz K, Damotte D, Dieu-Nosjean MC,  Validire P, Trautmann A, Mami-Chouaib F, Donnadieu E: Matrix architecture defines the preferential localization

www.sciencedirect.com

68. van Rooij N, van Buuren MM, Philips D, Velds A, Toebes M, Heemskerk B, van Dijk LJ, Behjati S, Hilkmann H, El Atmioui D et al.: Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013, 31:e439-e442. 69. Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E, Zhang W, Sougnez C, Cibulskis K, Sidney J et al.: Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood 2014, 124:453-462.

Current Opinion in Immunology 2015, 33:120–125