Immune cell constitution in the tumor microenvironment predicts the outcome in diffuse large B-cell lymphoma

Matias Autio, Suvi-Katri Leivonen, Oscar Brück, Satu Mustjoki, Judit Mészáros Jørgensen, Marja-Liisa Karjalainen-Lindsberg, Klaus Beiske, Harald Holte, Teijo Pellinen, Sirpa Leppä, Matias Autio, Suvi-Katri Leivonen, Oscar Brück, Satu Mustjoki, Judit Mészáros Jørgensen, Marja-Liisa Karjalainen-Lindsberg, Klaus Beiske, Harald Holte, Teijo Pellinen, Sirpa Leppä

Abstract

The tumor microenvironment (TME) and limited immune surveillance play important roles in lymphoma pathogenesis. Here we aimed to characterize immunological profiles of diffuse large B-cell lymphoma (DLBCL) and predict the outcome in response to immunochemotherapy. We profiled the expression of 730 immune-related genes in tumor tissues of 81 patients with DLBCL utilizing the Nanostring platform, and used multiplex immunohistochemistry to characterize T-cell phenotypes, including cytotoxic T cells (CD8, Granzyme B, OX40, Ki67), T-cell immune checkpoint (CD3, CD4, CD8, PD1, TIM3, LAG3), as well as regulatory T-cells and Th1 effector cells (CD3, CD4, FOXP3, TBET) in 188 patients. We observed a high degree of heterogeneity at the transcriptome level. Correlation matrix analysis identified gene expression signatures with highly correlating genes, the main cluster containing genes for cytolytic factors, immune checkpoint molecules, T cells and macrophages, together named a TME immune cell signature. Immunophenotyping of the distinct cell subsets revealed that a high proportion of immune checkpoint positive T cells translated to unfavorable survival. Together, our results demonstrate that the immunological profile of DLBCL TME is heterogeneous and clinically meaningful. This highlights the potential impact of T-cell immune checkpoint in regulating survival and resistance to immunochemotherapy. (Registered at clinicaltrials.gov identifiers: NCT01502982 and NCT01325194.)

Figures

Figure 1.
Figure 1.
Digital multiplexed gene expression profiling analysis reveals distinct gene expression signatures in diffuse large B-cell lymphoma (DLBCL). The expression of PanCancer Immune Profiling panel genes was assayed by Nanostring nCounter from 81 DLBCL samples. Correlation matrix analysis was performed for the most variable genes (standard deviation >1.0; n=335). Signatures with highly correlating genes are depicted in the heatmap. Selected genes are highlighted in the right-hand panel. ECM: extracellular matrix; TME: tumor microenvironment;
Figure 2.
Figure 2.
Multiplex immunohistochemistry (mIHC) reveals significant heterogeneity in the diffuse large B-cell lymphoma (DLBCL) tumor microenvironment (TME). (A) Representative images from the 4-plex mIHC analyses performed on tissue microarrays (TMA) from the Helsinki diffuse large B-cell lymphoma study (HEL-DLBCL) group cohort. CD8+ T-cell immune checkpoints: CD8=cyan, TIM3=red, LAG3=blue, PD1=green, DAPI=gray. CD4+ T-cell immune checkpoints: CD3=green, CD4=cyan, TIM3=red, LAG3=blue, DAPI=gray. Cytotoxicity panel: CD8=cyan, Granzyme B=green, Ki67=red, OX40=blue, DAPI=gray. Regulatory T cells (Tregs) and Th1 panel: CD3=green, CD4=cyan, FOXP3=red, TBET=blue, DAPI=gray. Scale bar 10 μm. (B) Unsupervised hierarchical clustering based on the expression of CD3+, CD4+ and CD8+ T cells. (C) Proportions of distinct immune cells from all cells, from CD4+ T cells and from CD8+ cells. (D) Heatmap visualizing all quantified immune cells and their immunophenotypes organized by unsupervised hierarchical clustering. Full annotation with all phenotypes is provided in the Online Supplementary Figure S3.
Figure 3.
Figure 3.
Diffuse large B-cell lymphoma (DLBCL) can be dived into T-cell high and low phenotypes.In silico immunophenotyping with CIBERSORTx was used to deconvolute T-cell proportions based on gene expression in four publicly available datasets.6-8,28 Gene expression datasets were uploaded to the CIBERSORTx web portal and the algorithm run using the 547-gene Leukocyte gene signature matrix (LM22) at 100 permutations. T-cell data were z-score transformed and visualized by unsupervised hierarchical clustering.
Figure 4.
Figure 4.
Membranous expression of HLA-ABC and β2 microglobulin (B2M) correlates with increased T-cell infiltration. (A) Representative images from B2M, HLAABC, and HLA-DR immunohistochemical (IHC) stainings. Scale bar 10 mm. (B) Results of the B2M, HLA-ABC, and HLA-DR IHC scoring. (C and D) Box plots visualizing the association of CD3+ T cells with HLA-ABC (C) and B2M (D) scores. P-values were determined by Kruskall-Wallis H (C) and Mann-Whitney U tests (D).
Figure 5.
Figure 5.
A high proportion of T cells expressing immune checkpoint molecules in the tumor microenvironment (TME) correlates with worse outcome in patients with diffuse large B-cell lymphoma (DLBCL). (A) The multiplex immunohistochemistry (mIHC) data from the Nordic Lymphoma Group (NLG) Trial cohort was clustered according to the proportions of TIM3+ T cells and LAG3 expressing CD8+ T cells. (B) Kaplan-Meier (log-rank test) survival plots depict overall survival (OS) in months (mo) in the groups with high and low amounts of these immune checkpoint molecule-expressing T cells in the NLG Trial cohort. (C) Unsupervised hierarchical clustering of T cells expressing immune checkpoint molecules in the Helsinki diffuse large B-cell lymphoma (HEL-DLBCL) cohort. (D) Kaplan-Meier (log-rank test) survival plots depict OS in the groups with high and low amounts of T cells expressing TIM3, LAG3 and PD1 in the HEL-DLBCL cohort. (E and F) Kaplan-Meier (log-rank test) survival plots depict OS in the groups with a high and low expression of immune checkpoint molecules in patients with an International Prognostic Index (IPI) score over 1 (E) and in patients with non-GCB type DLBCL (F) in the HEL-DLBCL cohort. N=46 for (A and B) and N=119 for (C-F). (Samples having tissue microarrays spots with poor quality for any of the phenotypes were removed from the clustering and survival analyses).
Figure 6.
Figure 6.
The impact of immune checkpoint molecules and distinct T-cell subtypes on survival. (A and B) Forest plots visualizing the impact of T cells and their immunophenotypes on overall survival (OS) in months (mo) in the Nordic Lymphoma Group (NLG) Trial (A) and Helsinki diffuse large B-cell lymphoma study (HELDLBCL) (B) cohorts, as evaluated using Cox univariate tests with continuous variables. (C-H) Kaplan-Meier plots (log-rank test) visualizing survival associations of distinct TIM3+ cell subpopulations. Cut-off was set at the highest expressing one-third versus the lowest expressing two-thirds.

References

    1. Coiffier B, Lepage E, Briere J, et al. . CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med. 2002;346(4):235-242.
    1. Pfreundschuh M, Trumper L, Osterborg A, et al. . CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial (MInT) Group. Lancet Oncol. 2006;7(5):379-391.
    1. Alizadeh AA, Eisen MB, Davis RE, et al. . Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503-511.
    1. Rosenwald A, Wright G, Chan WC, et al. . The use of molecular profiling to predict survival after chemotherapy for diffuse large-Bcell lymphoma. N Engl J Med. 2002;346(25):1937-1947.
    1. Lenz G, Wright G, Dave SS, et al. . Stromal gene signatures in large-B-cell lymphomas. N Engl J Med. 2008;359(22):2313-2323.
    1. Reddy A, Zhang J, Davis NS, et al. . Genetic and functional drivers of diffuse large B-cell lymphoma. Cell. 2017;171(2):481-494.e415.
    1. Schmitz R, Wright GW, Huang DW, et al. . Genetics and pathogenesis of diffuse large Bcell lymphoma. N Engl J Med. 2018;378(15):1396-1407.
    1. Chapuy B, Stewart C, Dunford AJ, et al. . Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24(5):679-690.
    1. Scott DW, Gascoyne RD. The tumour microenvironment in B cell lymphomas. Nat Rev Cancer. 2014;14(8):517-534.
    1. Nicholas NS, Apollonio B, Ramsay AG. Tumor microenvironment (TME)-driven immune suppression in B cell malignancy. Biochim Biophys Acta. 2016;1863(3):471-482.
    1. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12(4):298-306.
    1. Keane C, Gill D, Vari F, Cross D, Griffiths L, Gandhi M. CD4(+) tumor infiltrating lymphocytes are prognostic and independent of R-IPI in patients with DLBCL receiving RCHOP chemo-immunotherapy. Am J Hematol. 2013;88(4):273-276.
    1. Keane C, Vari F, Hertzberg M, et al. . Ratios of T-cell immune effectors and checkpoint molecules as prognostic biomarkers in diffuse large B-cell lymphoma: a populationbased study. Lancet Haematol. 2015; 2(10):e445-455.
    1. Xu-Monette ZY, Xiao M, Au Q, et al. . Immune profiling and quantitative analysis decipher the clinical role of immune-checkpoint expression in the tumor immune microenvironment of DLBCL. Cancer Immunol Res. 2019;7(4):644-657.
    1. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486-499.
    1. Booman M, Douwes J, Glas AM, et al. . Mechanisms and effects of loss of human leukocyte antigen class II expression in immune-privileged site-associated B-cell lymphoma. Clin Cancer Res. 2006;12(9): 2698-2705.
    1. Challa-Malladi M, Lieu YK, Califano O, et al. . Combined genetic inactivation of beta2- Microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell. 2011;20(6):728-740.
    1. Nijland M, Veenstra RN, Visser L, et al. . HLA dependent immune escape mechanisms in B-cell lymphomas: Implications for immune checkpoint inhibitor therapy? Oncoimmunology. 2017;6(4):e1295202.
    1. Riemersma SA, Jordanova ES, Schop RF, et al. . Extensive genetic alterations of the HLA region, including homozygous deletions of HLA class II genes in B-cell lymphomas arising in immune-privileged sites. Blood. 2000;96(10):3569-3577.
    1. Rimsza LM, Roberts RA, Miller TP, et al. . Loss of MHC class II gene and protein expression in diffuse large B-cell lymphoma is related to decreased tumor immunosurveillance and poor patient survival regardless of other prognostic factors: a follow-up study from the Leukemia and Lymphoma Molecular Profiling Project. Blood. 2004;103(11):4251-4258.
    1. Holte H, Leppa S, Bjorkholm M, et al. . Dosedensified chemoimmunotherapy followed by systemic central nervous system prophylaxis for younger high-risk diffuse large Bcell/ follicular grade 3 lymphoma patients: results of a phase II Nordic Lymphoma Group study. Ann Oncol. 2013;24(5):1385-1392.
    1. Leppa S, Joergensen J, Tierens A, et al. . Dosedense chemoimmunotherapy including early CNS prophylaxis for high-risk DLBCL. Final analysis from a Nordic Phase II study (the CHIC trial). Blood. 2016;128(22):1854.
    1. Leivonen SK, Pollari M, Bruck O, et al. . T-cell inflamed tumor microenvironment predicts favorable prognosis in primary testicular lymphoma. Haematologica. 2019;104(2): 338-346.
    1. Vandesompele J, De Preter K, Pattyn F, et al. . Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3(7):Research0034.
    1. Carpenter AE, Jones TR, Lamprecht MR, et al. . CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 2006;7(10):R100.
    1. Hans CP, Weisenburger DD, Greiner TC, et al. . Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004;103(1):275-282.
    1. Newman AM, Steen CB, Liu CL, et al. . Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol. 2019;37(7):773-782.
    1. Monti S, Savage KJ, Kutok JL, et al. . Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood. 2005;105(5):1851-1861.
    1. Dysvik B, Jonassen I. J-Express: exploring gene expression data using Java. Bioinformatics. 2001;17(4):369-370.
    1. Xu-Monette ZY, Zhou J, Young KH. PD-1 expression and clinical PD-1 blockade in Bcell lymphomas. Blood. 2018;131(1):68-83.
    1. Josefsson SE, Beiske K, Blaker YN, et al. . TIGIT and PD-1 mark intratumoral T cells with reduced effector function in B-cell non- Hodgkin lymphoma. Cancer Immunol Res. 2019;7(3):355-362.
    1. Chen BJ, Dashnamoorthy R, Galera P, et al. . The immune checkpoint molecules PD-1, PD-L1, TIM-3 and LAG-3 in diffuse large Bcell lymphoma. Oncotarget. 2019; 10(21):2030-2040.
    1. Xiong H, Mittman S, Rodriguez R, et al. . Coexpression of inhibitory receptors enriches for activated and functional CD8(+) T cells in murine syngeneic tumor models. Cancer Immunol Res. 2019;7(6):963-976.
    1. Lesokhin AM, Ansell SM, Armand P, et al. . Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a Phase Ib study. J Clin Oncol. 2016;34(23):2698-2704.
    1. Ansell SM, Hurvitz SA, Koenig PA, et al. . Phase I study of ipilimumab, an anti-CTLA- 4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2009; 15(20):6446-6453.
    1. Ansell SM, Minnema MC, Johnson P, et al. . Nivolumab for relapsed/refractory diffuse large B-cell lymphoma in patients ineligible for or having failed autologous transplantation: a single-arm, Phase II study. J Clin Oncol. 2019;37(6):481-489.
    1. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207(10):2187-2194.
    1. Koyama S, Akbay EA, Li YY, et al. . Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7:10501.
    1. Fourcade J, Sun Z, Benallaoua M, et al. . Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207(10):2175-2186.
    1. Zhou Q, Munger ME, Veenstra RG, et al. . Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood. 2011;117(17):4501-4510.
    1. Hill BT, Roberts ZJ, Rossi JM, Smith MR. Marked re-expansion of chimeric antigen receptor (CAR) T cells and tumor regression following nivolumab treatment in a patient treated with axicabtagene ciloleucel (axi-cel; KTE-C19) for refractory diffuse large B cell lymphoma (DLBCL). Blood. 2017;130(Suppl 1):2825.
    1. Filion LG, Izaguirre CA, Garber GE, Huebsh L, Aye MT. Detection of surface and cytoplasmic CD4 on blood monocytes from normal and HIV-1 infected individuals. J Immunol Methods. 1990;135(1-2):59-69.
    1. O'Doherty U, Steinman RM, Peng M, et al. . Dendritic cells freshly isolated from human blood express CD4 and mature into typical immunostimulatory dendritic cells after culture in monocyte-conditioned medium. J Exp Med. 1993;178(3):1067-1076.
    1. Valentin A, Rosati M, Patenaude DJ, et al. . Persistent HIV-1 infection of natural killer cells in patients receiving highly active antiretroviral therapy. Proc Natl Acad Sci U S A. 2002;99(10):7015-7020.
    1. Alvaro T, Lejeune M, Salvado MT, et al. . Outcome in Hodgkin's lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res. 2005;11(4):1467-1473.
    1. Koreishi AF, Saenz AJ, Persky DO, et al. . The role of cytotoxic and regulatory T cells in relapsed/refractory Hodgkin lymphoma. Appl Immunohistochem Mol Morphol. 2010;18(3):206-211.
    1. Oudejans JJ, Jiwa NM, Kummer JA, et al. . Activated cytotoxic T cells as prognostic marker in Hodgkin's disease. Blood. 1997;89(4):1376-1382.
    1. Muris JJ, Meijer CJ, Cillessen SA, et al. . Prognostic significance of activated cytotoxic T-lymphocytes in primary nodal diffuse large B-cell lymphomas. Leukemia. 2004; 18(3):589-596.
    1. Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8(1):59-73.
    1. Carreras J, Lopez-Guillermo A, Fox BC, et al. . High numbers of tumor-infiltrating FOXP3- positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood. 2006;108(9):2957-2964.
    1. Tzankov A, Meier C, Hirschmann P, Went P, Pileri SA, Dirnhofer S. Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin's lymphoma. Haematologica. 2008;93(2):193-200.

Source: PubMed

3
Sottoscrivi