LAG3: a novel immune checkpoint expressed by multiple lymphocyte subsets in diffuse large B-cell lymphoma.

Blockade of the PD-1 axis has modest efficacy in diffuse large B-cell lymphoma (DLBCL), but data regarding LAG3 are sparse. The impact of LAG3 digital gene expression was tested in 309 patients with DLBCL treated with standard chemoimmunotherapy. Cellular distribution of LAG3 protein was determined by immunohistochemistry and flow cytometry. In tumor-infiltrating lymphocytes (TILs), LAG3 expression was highest on CD4+ regulatory T cells (Tregs) and was also highly expressed on CD8+ T cells compared with CD4+ non-Tregs (both P = .008). LAG3high TILs were enriched in PD-1 and TIM-3. LAG3 was also expressed on a proportion of malignant B cells, and these patients had significantly higher LAG3 messenger RNA in their biopsies (P = .03). LAG3high gene expression was associated with inferior survival in discovery/validation cohorts, independent of cell of origin and the international prognostic index. Patients who were PD-L1high were fivefold more likely to be LAG3high (P < .0001). Patients who were LAG3high/PD-L1high had an inferior progression-free survival (P = .011) and overall survival (P = .005) compared with patients who were LAG3low/PD-L1high. Digital spatial protein analysis confirms LAG3 expression on T cells and, surprisingly, tumor-associated macrophages (TAMs) at higher levels than found on CD20+ B cells in the tumor microenvironment. LAG3 is frequently expressed on CD4+ Tregs and CD8+ TILs, typically with other immune checkpoints, and is also present in a proportion of malignant B cells in DLBCL and in areas enriched for TAMs. LAG3high expression is associated with poor outcome independent of conventional prognosticators.

[1]  Allen W. Zhang,et al.  Single cell transcriptome analysis reveals disease-defining T cell subsets in the tumor microenvironment of classic Hodgkin lymphoma. , 2019, Cancer discovery.

[2]  A. Gill,et al.  The tumour microenvironment is immuno‐tolerogenic and a principal determinant of patient outcome in EBV‐positive diffuse large B‐cell lymphoma , 2019, European journal of haematology.

[3]  K. Syrigos,et al.  High-Plex Predictive Marker Discovery for Melanoma Immunotherapy–Treated Patients Using Digital Spatial Profiling , 2019, Clinical Cancer Research.

[4]  Sonali M. Smith,et al.  PD-L1 gene alterations identify a subset of diffuse large B-cell lymphoma harboring a T-cell-inflamed phenotype. , 2019, Blood.

[5]  A. Evens,et al.  The immune checkpoint molecules PD-1, PD-L1, TIM-3 and LAG-3 in diffuse large B-cell lymphoma , 2019, Oncotarget.

[6]  M. Shipp,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. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[7]  Juliet Investigators Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma , 2019 .

[8]  T. Okazaki,et al.  LAG-3 inhibits the activation of CD4+ T cells that recognize stable pMHCII through its conformation-dependent recognition of pMHCII , 2018, Nature Immunology.

[9]  T. Mcclanahan,et al.  LAG3+ Regulatory T Cells Restrain Interleukin‐23‐Producing CX3CR1+ Gut‐Resident Macrophages during Group 3 Innate Lymphoid Cell‐Driven Colitis , 2018, Immunity.

[10]  L. Jouneau,et al.  LAG-3 Inhibitory Receptor Expression Identifies Immunosuppressive Natural Regulatory Plasma Cells , 2018, Immunity.

[11]  Xue Zhang,et al.  The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy , 2018, Genes & cancer.

[12]  J. Wilmott,et al.  Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. , 2018, The Lancet. Oncology.

[13]  M. Gandhi,et al.  Immune evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. , 2018, Blood.

[14]  S. Rodig,et al.  Checkpoint blockade in Hodgkin and non-Hodgkin lymphoma. , 2017, Blood advances.

[15]  P. Ascierto,et al.  Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. , 2017 .

[16]  Hyo Jin Kim,et al.  Expression of LAG-3 defines exhaustion of intratumoral PD-1+ T cells and correlates with poor outcome in follicular lymphoma , 2017, Oncotarget.

[17]  F. Hirsch,et al.  LAG‐3 Protein Expression in Non–Small Cell Lung Cancer and Its Relationship with PD‐1/PD‐L1 and Tumor‐Infiltrating Lymphocytes , 2017, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[18]  A. Wiestner,et al.  Lymphocyte activation gene 3: a novel therapeutic target in chronic lymphocytic leukemia , 2017, Haematologica.

[19]  A. Wiestner,et al.  Lymphocyte Activation Gene 3-a Novel Therapeutic Target in Chronic Lymphocytic Leukemia , 2016 .

[20]  A. Kulkarni,et al.  LAG-3 confers poor prognosis and its blockade reshapes antitumor response in head and neck squamous cell carcinoma , 2016, Oncoimmunology.

[21]  M. Millenson,et al.  Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. , 2016, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  S. Lewin,et al.  CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART , 2016, PLoS pathogens.

[23]  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.

[24]  R. Phillips,et al.  Immunological biomarkers predict HIV-1 viral rebound after treatment interruption , 2015, Nature Communications.

[25]  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.

[26]  P. Brousset,et al.  Several immune escape patterns in non-Hodgkin's lymphomas , 2015, Oncoimmunology.

[27]  G. Freeman,et al.  Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer , 2015, Clinical Cancer Research.

[28]  Kai Fu,et al.  Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. , 2014, Blood.

[29]  Clelia Di Serio,et al.  Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells , 2013, Nature Medicine.

[30]  Peter Vogel,et al.  Microenvironment and Immunology Immune Inhibitory Molecules Lag-3 and Pd-1 Synergistically Regulate T-cell Function to Promote Tumoral Immune Escape , 2022 .

[31]  Barbara Fazekas de St Groth,et al.  Flow cytometric detection of human regulatory T cells. , 2011, Methods in molecular biology.

[32]  R. Khanna,et al.  Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients. , 2006, Blood.

[33]  T. Gingeras,et al.  CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells , 2006, The Journal of experimental medicine.

[34]  D. Vignali,et al.  The CD4‐related molecule, LAG‐3 (CD223), regulates the expansion of activated T cells , 2003, European journal of immunology.

[35]  Torsten Hothorn,et al.  On the Exact Distribution of Maximally Selected Rank Statistics , 2002, Comput. Stat. Data Anal..

[36]  C. Kurschner,et al.  Phenotypic analysis of the murine CD4‐related glycoprotein, CD223 (LAG‐3) , 2002, European journal of immunology.

[37]  B. Maigret,et al.  Characterization of the major histocompatibility complex class II binding site on LAG-3 protein. , 1997, Proceedings of the National Academy of Sciences of the United States of America.