scholarly journals Immune checkpoint blockade impairs immunosuppressive mechanisms of regulatory T cells in B-cell lymphoma

2021 ◽  
Vol 14 (9) ◽  
pp. 101170
Author(s):  
Vera Bauer ◽  
Fatima Ahmetlić ◽  
Nadine Hömberg ◽  
Albert Geishauser ◽  
Martin Röcken ◽  
...  
Blood ◽  
2020 ◽  
Vol 135 (8) ◽  
pp. 523-533 ◽  
Author(s):  
Justin Kline ◽  
James Godfrey ◽  
Stephen M. Ansell

Abstract The clinical development of effective cancer immunotherapies, along with advances in genomic analysis, has led to the identification of tumor environmental features that predict for sensitivity to immune checkpoint blockade therapy (CBT). Early-phase clinical trial results have demonstrated the remarkable effectiveness of CBT in specific lymphoma subtypes, including classical Hodgkin lymphoma and primary mediastinal B-cell lymphoma. Conversely, CBT has been relatively disappointing in follicular lymphoma and diffuse large B-cell lymphoma. These clinical observations, coupled with important scientific discoveries, have uncovered salient features of the lymphoma microenvironment that correlate with immunotherapy response in patients. For example, classical Hodgkin lymphoma is characterized by an inflammatory environment, genetic alterations that facilitate escape from immune attack, and sensitivity to PD-1 blockade therapy. On the other hand, for lymphomas in which measures of immune surveillance are lacking, including follicular lymphoma and most diffuse large B-cell lymphomas, anti-PD-1 therapy has been less effective. An improved understanding of the immune landscapes of these lymphomas is needed to define subsets that might benefit from CBT. In this article, we describe the immune environments associated with major B-cell lymphomas with an emphasis on the immune escape pathways orchestrated by these diseases. We also discuss how oncogenic alterations in lymphoma cells may affect the cellular composition of the immune environment and ultimately, vulnerability to CBT. Finally, we highlight key areas for future investigation, including the need for the development of biomarkers that predict for sensitivity to CBT in lymphoma patients.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A829-A829
Author(s):  
Vera Bauer ◽  
Fatima Ahmetlic ◽  
Martin Roecken ◽  
Ralph Mocikat ◽  
Ralph Mocikat

BackgroundAntibodies against programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have become established part of anti-cancer therapy. However, the mechanisms contributing to the therapeutic success have not been entirely uncovered by now. Here we focus on the impact of PD-1/CTLA-4-blocking antibodies on regulatory T cells (Tregs), which are known to be involved in tumor immune evasion in many cancer types.MethodsTo evaluate how Tregs are affected by anti-PD-1/CTLA-4 therapy, we used a MYC-transgenic mouse model of spontaneously arising B-cell lymphoma, which can be effectively treated by immune checkpoint inhibition. Data were acquired by flow cytometry.ResultsAs earlier shown, Tregs were involved in immune escape of MYC tumors. The Treg to effector T cell (Teff) ratio was elevated within the CD4-positive cell compartment. Tumor-infiltrating Tregs were predominantly thymic Tregs, which recognized overexpressed tumor-derived self-peptides in an MHC class II-restricted manner and showed upregulated expression of activation markers, Foxp3, CD25 and IL-10. To examine whether these phenotypic alterations correlated with the immunosuppressive capability of Tregs, an in vitro suppression assay was established. In this setting, MYC Tregs turned out to suppress proliferation and IFN-γ release of Teff cells more effectively than wildtype Tregs. The suppression observed in vitro was mediated by cell contacts and IL-10. Further suppressive mechanisms are likely to play a role, such as competition for MHC-II ligands and a consumption of IL-2. To investigate if immune checkpoint blockade interferes with these Treg-dependent immunosuppressive pathways, MYC mice were treated with a combination of anti-PD-1 and anti-CTLA-4 antibodies. Tregs from treated MYC mice showed decreased expression of CD69, CD137, Foxp3, CD25 and IL-10 as compared to Tregs from untreated MYC mice. This correlated with a lower suppressive capacity of Tregs from treated animals in the in vitro suppression assay.ConclusionsTaken together, the data show that immune checkpoint blockade impairs the development of the suppressive phenotype of intratumoral Tregs. Thus, apart from the initially described Teff reactivation, other mechanisms are also relevant for unfolding the therapeutic effect of immune checkpoint inhibitors.Ethics ApprovalAll animal experiments were approved by Regierung von Oberbayern, approval number 55.2-1-54.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A5.1-A5
Author(s):  
A Martinez-Usatorre ◽  
E Kadioglu ◽  
C Cianciaruso ◽  
B Torchia ◽  
J Faget ◽  
...  

BackgroundImmune checkpoint blockade (ICB) with antibodies against PD-1 or PD-L1 may provide therapeutic benefits in patients with non-small cell lung cancer (NSCLC). However, most tumours are resistant and cases of disease hyper-progression have also been reported.Materials and MethodsGenetically engineered mouse models of KrasG12Dp53null NSCLC were treated with cisplatin along with antibodies against angiopoietin-2/VEGFA, PD-1 and CSF1R. Tumour growth was monitored by micro-computed tomography and the tumour vasculature and immune cell infiltrates were assessed by immunofluorescence staining and flow cytometry.ResultsCombined angiopoietin-2/VEGFA blockade by a bispecific antibody (A2V) modulated the vasculature and abated immunosuppressive macrophages while increasing CD8+effector T cells in the tumours, achieving disease stabilization comparable or superior to cisplatin-based chemotherapy. However, these immunological responses were unexpectedly limited by the addition of a PD-1 antibody, which paradoxically enhanced progression of a fraction of the tumours through a mechanism involving regulatory T cells and macrophages. Elimination of tumour-associated macrophages with a CSF1R-blocking antibody induced NSCLC regression in combination with PD-1 blockade and cisplatin.ConclusionsThe immune cell composition of the tumour determines the outcome of PD-1 blockade. In NSCLC, high infiltration of regulatory T cells and immunosuppressive macrophages may account for tumour hyper-progression upon ICB.Disclosure InformationA. Martinez-Usatorre: None. E. Kadioglu: None. C. Cianciaruso: None. B. Torchia: None. J. Faget: None. E. Meylan: None. M. Schmittnaegel: None. I. Keklikoglou: None. M. De Palma: None.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e19040-e19040
Author(s):  
Kalyan Kusum Mukherjee ◽  
Sukanya Dhar ◽  
Mohona Chakraborty ◽  
Rajib Bhattacharjee ◽  
Shayani Bhanja ◽  
...  

e19040 Background: NHL is a type of lymphoma either B or T cell origin. However, 85% is of B cell type, especially diffuse large B cell lymphoma (DLBCL) with CD20+ in nature. Standard of care of CD20+DLBCL is R-CHOP 6 to 8 cycles and 66% patients generally respond to this treatment. Remaining 34% is still unresponsive to R-CHOP. Thus, establishment of a biomarker is required to intensify the treatment.Out of different scope of biomarker development, alterations in immune cellular components within tumor microenvironment may be tried as a potential biomarker to assess the possibility of occurrence of residual disease and relapse within 2 years for the DLBCL patients with 6-8 cycles of R-CHOP. Methods: 51 of selected pt.CD20+ DLBCL were treated with 6-8 cycles of R-CHOP and included in the present study. A panel of immune cells CD4+, CD8+ T cells, CD4+CD25+FoxP3 regulatory T cells, CD33+CD11b+CD14-/+ MDSCs and CD8+CD45RO+ Memory T cells were studied by flow-cytometry at different phases of treatment. Results: Within 51selected patients, 9 were disease free and 11patients exhibited stable disease for 2years (stable, non-relapsed, n = 20) following the completion of treatment. Rest of the patients (n = 31) showed relapse in different time periods within 2 years. Among several immune cells, CD33+CD11b+MDSCs were remarkably elevated in high grade residual and relapsed DLBCL patients compared to non-relapsed patients and normal healthy individuals. CD33+CD14- granulocytic, but not CD33+CD14+ monocytic MDSCs are mostly increased in relapsed patients than those having stable disease. CD4+CD25+FoxP3 regulatory T cells are also elevated in relapsed DLBCL patients, but increment is not comparable as MDSCs. No significant alteration was noticed in CD4+ and CD8+ T cells. Among relapsed patients CD8+CD45RO+ Memory T cells are increased, those are mostly corrupted in nature. Conclusions: Observed correlation between increased granulocytic MDSCs with the occurrence of residual disease and/or relapse suggests granulocytic MDSCs might be a potential biomarker for prediction of residual and relapse for DLBCL patients.


2008 ◽  
Vol 68 (13) ◽  
pp. 5439-5449 ◽  
Author(s):  
Paolo Serafini ◽  
Stephanie Mgebroff ◽  
Kimberly Noonan ◽  
Ivan Borrello

Author(s):  
Anne Scheuerpflug ◽  
Fatima Ahmetlić ◽  
Vera Bauer ◽  
Tanja Riedel ◽  
Martin Röcken ◽  
...  

Abstract Immune checkpoint blocking (ICB) is a promising new tool of cancer treatment. Yet, the underlying therapeutic mechanisms are not fully understood. Here we investigated the role of dendritic cells (DCs) for the therapeutic effect of ICB in a λ-MYC-transgenic mouse model of endogenously arising B-cell lymphoma. The growth of these tumors can be effectively delayed by antibodies against CTLA-4 and PD-1. Tumor-infiltrating DCs from mice having received therapy showed an upregulation of costimulatory molecules as well as an augmented IL-12/IL-10 ratio as compared to untreated controls. Both alterations seemed to be induced by interferon-γ (IFN-γ), which is upregulated in T cells and natural killer cells upon ICB. Furthermore, the enhanced IL-12/IL-10 ratio, which favors Th1-prone antitumor T-cell responses, was a consequence of direct interaction of ICB antibodies with DCs. Importantly, the capability of tumor-infiltrating DCs of stimulating peptide-specific or allogeneic T-cell responses in vitro was improved when DCs were derived from ICB-treated mice. The data indicate that ICB therapy is not only effective by directly activating T cells, but also by triggering a complex network, in which DCs play a pivotal role at the interface between innate and adaptive antitumor responses.


2015 ◽  
Vol 144 (6) ◽  
pp. 935-944 ◽  
Author(s):  
Chen Chang ◽  
Shang-Yin Wu ◽  
Yu-Wei Kang ◽  
Kun-Piao Lin ◽  
Tsai-Yun Chen ◽  
...  

2008 ◽  
Vol 152 (2) ◽  
pp. 381-387 ◽  
Author(s):  
I. Heier ◽  
P. O. Hofgaard ◽  
P. Brandtzaeg ◽  
F. L. Jahnsen ◽  
M. Karlsson

2007 ◽  
Vol 137 (4) ◽  
pp. 364-373 ◽  
Author(s):  
Sverker Hasselblom ◽  
Margret Sigurdadottir ◽  
Ulrika Hansson ◽  
Herman Nilsson-Ehle ◽  
Börje Ridell ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1565-1565
Author(s):  
Jae-Yong Kwak ◽  
Na-Ri Lee ◽  
Eun-Kee Song ◽  
Kyu Yun Jang ◽  
Ha Na Choi ◽  
...  

Abstract Tumor infiltrating immune cells play an important role in host immune reactions against diffuse large B-cell lymphoma (DLBCL). In the present study, we have identified a subset of tumor infiltrating FOXP3-positive regulatory T cells (Tregs) in the initial DLBCL biopsy specimens and evaluated its prognostic significance. Ninety-six patients with DLBCL were retrospectively evaluated. Expression pattern of FOXP3 protein was examined using standard immunohistochemistry in paraffin-embedded tissue samples. The median overall survival (OS) was 28 months. Compared to the others, the patients with higher percentage of FOXP3-positive Tregs in the initial tumor biopsy, showed a significantly longer OS (p=0.003). When prognostic factors were evaluated in a multivariate model, the international prognostic index and the percentage of infiltrating FOXP3-positive Tregs in the initial biopsy were shown to be independent predictors of OS. In conclusion, the presence of increased percentage of FOXP3-positive Tregs in DLBCL predicts a better prognosis. Figure Figure


Sign in / Sign up

Export Citation Format

Share Document