The T-cell Receptor Repertoire Influences the Tumor Microenvironment and Is Associated with Survival in Aggressive B-cell Lymphoma

Purpose: To investigate the relationship between the intra-tumoral T-cell receptor (TCR) repertoire and the tumor microenvironment (TME) in de novo diffuse large B-cell lymphoma (DLBCL) and the impact of TCR on survival. Experimental Design: We performed high-throughput unbiased TCRβ sequencing on a population-based cohort of 92 patients with DLBCL treated with conventional (i.e., non-checkpoint blockade) frontline “R-CHOP” therapy. Key immune checkpoint genes within the TME were digitally quantified by nanoString. The primary endpoints were 4-year overall survival (OS) and progression-free survival (PFS). Results: The TCR repertoire within DLBCL nodes was abnormally narrow relative to non-diseased nodal tissues (P < 0.0001). In DLBCL, a highly dominant single T-cell clone was associated with inferior 4-year OS rate of 60.0% [95% confidence interval (CI), 31.7%–79.6%], compared with 79.8% in patients with a low dominant clone (95% CI, 66.7%–88.5%; P = 0.005). A highly dominant clone also predicted inferior 4-year PFS rate of 46.6% (95% CI, 22.5%–76.6%) versus 72.6% (95% CI, 58.8%–82.4%, P = 0.008) for a low dominant clone. In keeping, clonal expansions were most pronounced in the EBV+ DLBCL subtype that is known to express immunogenic viral antigens and is associated with particularly poor outcome. Increased T-cell diversity was associated with significantly elevated PD-1, PD-L1, and PD-L2 immune checkpoint molecules. Conclusions: Put together, these findings suggest that the TCR repertoire is a key determinant of the TME. Highly dominant T-cell clonal expansions within the TME are associated with poor outcome in DLBCL treated with conventional frontline therapy. Clin Cancer Res; 23(7); 1820–8. ©2016 AACR.

[1]  A. Rickinson,et al.  Epstein–Barr virus-associated lymphomas , 2017, Philosophical Transactions of the Royal Society B: Biological Sciences.

[2]  Nicolai J. Birkbak,et al.  Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade , 2016, Science.

[3]  M. Roederer,et al.  The impact of HLA class I and EBV latency‐II antigen‐specific CD8+ T cells on the pathogenesis of EBV+ Hodgkin lymphoma , 2015, Clinical and experimental immunology.

[4]  Michael R. Green,et al.  Ratios of T-cell immune effectors and checkpoint molecules as prognostic biomarkers in diffuse large B-cell lymphoma: a population-based study. , 2015, The Lancet. Haematology.

[5]  B. Jakobsen,et al.  ImmTACs for targeted cancer therapy: Why, what, how, and which. , 2015, Molecular immunology.

[6]  R. Emerson,et al.  High-throughput pairing of T cell receptor α and β sequences , 2015, Science Translational Medicine.

[7]  L. Staudt,et al.  Prognostic Significance of Diffuse Large B-Cell Lymphoma Cell of Origin Determined by Digital Gene Expression in Formalin-Fixed Paraffin-Embedded Tissue Biopsies. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  B. Vogelstein,et al.  PD-1 blockade in tumors with mismatch repair deficiency. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[9]  R. Emerson,et al.  High-throughput pairing of T cell receptor alpha and beta sequences (TECH2P.930) , 2015, The Journal of Immunology.

[10]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[11]  Ash A. Alizadeh,et al.  Mutations in early follicular lymphoma progenitors are associated with suppressed antigen presentation , 2015, Proceedings of the National Academy of Sciences.

[12]  H. Ishwaran,et al.  Radiation and Dual Checkpoint Blockade Activates Non-Redundant Immune Mechanisms in Cancer , 2015, Nature.

[13]  J. Wolchok,et al.  Genetic basis for clinical response to CTLA-4 blockade in melanoma. , 2014, The New England journal of medicine.

[14]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[15]  D. Koning,et al.  Complex T-Cell Receptor Repertoire Dynamics Underlie the CD8+ T-Cell Response to HIV-1 , 2014, Journal of Virology.

[16]  Ryan Emerson,et al.  CTLA4 Blockade Broadens the Peripheral T-Cell Receptor Repertoire , 2014, Clinical Cancer Research.

[17]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[18]  K. Young,et al.  EBV-positive diffuse large B-cell lymphoma of the elderly. , 2013, Blood.

[19]  Peter Schirmacher,et al.  Tumor-infiltrating lymphocytes in colorectal tumors display a diversity of T cell receptor sequences that differ from the T cells in adjacent mucosal tissue , 2013, Cancer Immunology, Immunotherapy.

[20]  M. Gandhi,et al.  CD4+ Tumor infiltrating lymphocytes are prognostic and independent of R‐IPI in patients with DLBCL receiving R‐CHOP chemo‐immunotherapy , 2013, American journal of hematology.

[21]  M. Gandhi,et al.  Expression profiling of Epstein-Barr virus-encoded microRNAs from paraffin-embedded formalin-fixed primary Epstein-Barr virus-positive B-cell lymphoma samples. , 2012, Journal of virological methods.

[22]  Qianjun Zhang,et al.  Blood T-cell receptor diversity decreases during the course of HIV infection, but the potential for a diverse repertoire persists. , 2012, Blood.

[23]  P. Doherty,et al.  T Cell Receptor αβ Diversity Inversely Correlates with Pathogen-Specific Antibody Levels in Human Cytomegalovirus Infection , 2012, Science Translational Medicine.

[24]  M. Gandhi,et al.  Epstein-Barr virus-positive diffuse large B-cell lymphoma of the elderly expresses EBNA3A with conserved CD8 T-cell epitopes. , 2011, American journal of blood research.

[25]  G. D. de Bock,et al.  The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis , 2011, British Journal of Cancer.

[26]  Sang Joon Kim,et al.  A Mathematical Theory of Communication , 2006 .

[27]  R. Khanna,et al.  Human cytomegalovirus: clinical aspects, immune regulation, and emerging treatments. , 2004, The Lancet. Infectious diseases.

[28]  I. Messaoudi,et al.  The many important facets of T-cell repertoire diversity , 2004, Nature Reviews Immunology.

[29]  Marie-Paule Lefranc,et al.  IMGT/JunctionAnalysis: the first tool for the analysis of the immunoglobulin and T cell receptor complex V-J and V-D-J JUNCTIONs , 2004, ISMB/ECCB.

[30]  R. Marcus,et al.  Late diversification in the clonal composition of human cytomegalovirus-specific CD8+ T cells following allogeneic hemopoietic stem cell transplantation. , 2003, Blood.

[31]  I. Messaoudi,et al.  Direct Link Between mhc Polymorphism, T Cell Avidity, and Diversity in Immune Defense , 2002, Science.

[32]  Pierre Morel,et al.  CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. , 2002, The New England journal of medicine.

[33]  A. Hughes,et al.  The outcome of hepatitis C virus infection is predicted by escape mutations in epitopes targeted by cytotoxic T lymphocytes. , 2001, Immunity.

[34]  C. Sample,et al.  An Epstein-Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs. , 2001, Blood.

[35]  T. Habermann,et al.  Cd4+ T-cell immune response to large B-cell non-Hodgkin's lymphoma predicts patient outcome. , 2001, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[36]  M. Bevan,et al.  Selecting and maintaining a diverse T-cell repertoire , 1999, Nature.

[37]  Charles R. M. Bangham,et al.  Human immunodeficiency virus genetic variation that can escape cytotoxic T cell recognition , 1991, Nature.

[38]  R. Hardy,et al.  The role of clonal selection in the pathogenesis of an autoreactive human B cell lymphoma , 1991, The Journal of experimental medicine.