Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
editorial
. 2015 Mar 9;17(6):769–770. doi: 10.1093/neuonc/nov034

Tregs in gliomas – the jury is still out

Michael Platten 1, Wolfgang Wick 1
PMCID: PMC4483128  PMID: 25754090

Regulatory T cells (Tregs) promote tumor growth chiefly by suppressing tumor-specific T cell responses in the tumor tissue. This is the common notion that provides the rational for targeting Tregs in cancer with the aim at enhancing antitumor immune responses. If this rational is true and if we think that there is a natural – albeit insufficient - antitumor immune response, then tumor-infiltrating and/or circulating Tregs should influence the natural course of tumor disease with high numbers predicting poor outcome. Conversely, the number of intratumoral T cells with antitumor properties, particularly CD8+ cytotoxic T lymphocytes, ought to predict a favorable outcome. While most studies analysing tumor-infiltrating T cells in gliomas support the latter, there has been ambiguous data on the relevance of tumor-infiltrating Tregs in human gliomas.16

Tregs are classically viewed synonymous with FoxP3 + CD25 + CD4+ T cells. These cells are usually identified by flow cytometry from peripheral blood or preparations of mononuclear cell suspensions from fresh tumor specimens. In the current study by Thomas and colleagues (this issue) tumor-infiltrating Tregs and CD3+ T cells were measured not by flow cytometry but by epigenetic qPCR and by immunohistochemistry. This approach offers the significant advantage of performing analyses on archival tissue but at a risk of losing specificity. Although FoxP3 transcripts and methylation patterns are generally considered to be specific for Tregs, expression has been detected in CD8+ T cells7 and non-lymphoid CNS tissue.8 Hence, Treg quantifications based on FoxP3 expression without single-cell resolution have to be viewed with caution. With their methods Thomas and colleagues did not find peripheral or tumor-infiltrating Tregs to be predictive of outcome in their small study cohort.

Does this and the negative results of most previous studies mean that Tregs are not important in glioma biology? FoxP3 + CD4+ Tregs in humans comprise several subsets, which are heterogeneous both with respect to phenotype and function. For instance, FoxP3loCD45RA + CD25lo cells are naive or resting Treg (rTreg) cells, which differentiate into highly suppressive FoxP3hiCD45RA-CD25hi effector Treg (eTreg) cells upon antigenic stimulation. In contrast, FoxP3loCD45RA-CD25lo non-Treg cells do not possess suppressive activity but can secrete pro-inflammatory cytokines.9 In addition, there is an increasing arsenal of additional markers subdividing the Treg family including CD127, Helios, CTLA-4 and CD39. Furthermore, the diversity of T cells extends well into the CD8+ compartment including FoxP3 expression.7 Future studies ought to incorporate the complexity of Treg development and differentiation including antigen-specificity to answer this question.

Is this a worthwhile effort or just l'art pour l'art? With the increasing arsenal of checkpoint inhibitors spilling over into the glioma arena we need to provide answers to the central questions: Is there a meaningful natural (antigen-specific) anti-glioma T cell immunity and if so is this immunity suppressed by cellular mediators, such as Tregs, resulting in a net poorly immunogenic tumor phenotype? Because only the presence of a meaningful natural anti-glioma T cell immunity would then provide the rational to release the break on this anti-glioma T cell immunity by introducing checkpoint inhibitors or other strategies to inhibit tumor-associated immune suppression (e.g. Tregs) as single agents. This has been demonstrated for melanoma,10 but solid evidence for gliomas is lacking. Consequently, clinical studies testing checkpoint inhibitors in gliomas clearly should be backed by an in-depth analysis of pre- and post-treatment tumor tissue including analyses of tumor-infiltrating Tregs and the repertoire of antigen-specific effector T cells. In addition to the relevance of Tregs for suppressing the natural anti-glioma immunity, this immune cell population may constitute a significant barrier for antigen-specific peptide vaccines, which are on the verge of entering the clinical arena.1115 If tumor-infiltrating Tregs (or even circulating Tregs) counteracted this induced antigen-specific anti-glioma T- and B cell immunity, then this would again constitute a strong rationale for combining peptide vaccines with checkpoint inhibitors, agents depleting Tregs or simply temozolomide chemotherapy upfront. Also here, clinical trials evaluating the efficacy of antigen-specific vaccines ought to incorporate pre- and post-treatment analyses of mechanisms preventing the induction of an effective anti-glioma immune response including Tregs. Importantly, these analyses need to take into account the heterogeneity and tissue-specificity of Tregs, which extends well beyond the classic CD4 + CD25 + FoxP3+ phenotype, which most studies including the study by Thomas and colleagues are restricted to.

Funding

This work was supported by grants of the Helmholtz Foundation (VH-NG-306) to M.P. and the German Research Foundation (SFB938 TPK) to M.P. and W.W.

Conflict of interest statement. The authors hold patents on IDH1R132H as an immunotherapeutic target.

References

  • 1.Sayour EJ, McLendon P, McLendon R, et al. Increased proportion of FoxP3+ regulatory T cells in tumor infiltrating lymphocytes is associated with tumor recurrence and reduced survival in patients with glioblastoma. Cancer Immunol Immunother. 2015. Jan 3 [Epub]. [DOI] [PMC free article] [PubMed]
  • 2.Han S, Zhang C, Li Q, et al. Tumour-infiltrating CD4(+) and CD8(+) lymphocytes as predictors of clinical outcome in glioma. Br J Cancer. 2014;110:2560–2568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lohr J, Ratliff T, Huppertz A, et al. Effector T-cell infiltration positively impacts survival of glioblastoma patients and is impaired by tumor-derived TGF-beta. Clin Cancer Res. 2011;17:4296–4308. [DOI] [PubMed] [Google Scholar]
  • 4.Jacobs JF, Idema AJ, Bol KF, et al. Prognostic significance and mechanism of Treg infiltration in human brain tumors. J Neuroimmunol. 2010;225:195–199. [DOI] [PubMed] [Google Scholar]
  • 5.Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, et al. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. 2008;14:5166–5172. [DOI] [PubMed] [Google Scholar]
  • 6.El Andaloussi A, Lesniak MS. An increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro Oncol. 2006;8:234–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Suzuki M, Jagger AL, Konya C, et al. CD8+CD45RA+CCR7+FOXP3+ T cells with immunosuppressive properties: a novel subset of inducible human regulatory T cells. J Immunol. 2012;189:2118–2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Frattini V, Pisati F, Speranza MC, et al. FOXP3, a novel glioblastoma oncosuppressor, affects proliferation and migration. Oncotarget. 2012;3:1146–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Miyara M, Yoshioka Y, Kitoh A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30:899–911. [DOI] [PubMed] [Google Scholar]
  • 10.Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. The New England Journal of Medicine. 2014;371:2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Schuster J, Lai RK, Recht LD, et al. A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: the ACT III study. Neuro Oncol. 2015;17:854–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Schumacher T, Bunse L, Pusch S, et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature. 2014;512:324–327. [DOI] [PubMed] [Google Scholar]
  • 13.Bunse L, Schumacher T, Sahm F, et al. Proximity ligation assay evaluates IDH1R132H presentation in gliomas. The Journal of Clinical Investigation. 2015;125:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Phuphanich S, Wheeler CJ, Rudnick JD, et al. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother. 2013;62:125–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dutoit V, Herold-Mende C, Hilf N, et al. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. Brain. 2012;135:1042–1054. [DOI] [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press

RESOURCES