Resistance to PD1 blockade in the absence of metalloprotease-mediated LAG3 shedding

LAG3 shedding from conventional CD4+ T cells drives responsiveness to anti-PD1 immunotherapy (see the related Focus by Seidel and Bengsch). Lending PD1 a helping hand LAG3 is an inhibitory receptor expressed on exhausted T cells that is thought to temper T cell activation by engaging peptide–MHC class II complexes. Here, Andrews et al. have engineered mice expressing a noncleavable form of LAG3 (LAG3NC) that cannot be shed from the cell surface by ADAM family proteases. By generating mouse strains that express LAG3NC ion-distinct T cell types, they found that LAG3 shedding by conventional CD4+ T cells rather than T regulatory cells (Tregs) or CD8+ T cells to be important for driving responsiveness to anti-PD1. Although both CD8+ T cells and Tregs have received considerable attention in the context of immunotherapy, this study highlights the importance of T cell help in promoting antitumor immunity. Mechanisms of resistance to cancer immunotherapy remain poorly understood. Lymphocyte activation gene–3 (LAG3) signaling is regulated by a disintegrin and metalloprotease domain-containing protein–10 (ADAM10)– and ADAM17-mediated cell surface shedding. Here, we show that mice expressing a metalloprotease-resistant, noncleavable LAG3 mutant (LAG3NC) are resistant to PD1 blockade and fail to mount an effective antitumor immune response. Expression of LAG3NC intrinsically perturbs CD4+ T conventional cells (Tconvs), limiting their capacity to provide CD8+ T cell help. Furthermore, the translational relevance for these observations is highlighted with an inverse correlation between high LAG3 and low ADAM10 expression on CD4+ Tconvs in the peripheral blood of patients with head and neck squamous cell carcinoma, which corresponded with poor prognosis. This correlation was also observed in a cohort of patients with skin cancers and was associated with increased disease progression after standard-of-care immunotherapy. These data suggest that subtle changes in LAG3 inhibitory receptor signaling can act as a resistance mechanism with a substantive effect on patient responsiveness to immunotherapy.

[1]  R. Martin Faculty Opinions recommendation of Resistance to PD1 blockade in the absence of metalloprotease-mediated LAG3 shedding. , 2020, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[2]  Steffi Oesterreich,et al.  Immune Landscape of Viral- and Carcinogen-Driven Head and Neck Cancer. , 2019, Immunity.

[3]  T. Crompton,et al.  The IFITM protein family in adaptive immunity , 2019, Immunology.

[4]  D. Vignali,et al.  Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups , 2019, Nature Immunology.

[5]  Paul J. Hoffman,et al.  Comprehensive Integration of Single-Cell Data , 2018, Cell.

[6]  M. Barenco,et al.  IFITM proteins drive type 2 T helper cell differentiation and exacerbate allergic airway inflammation , 2018, European journal of immunology.

[7]  Christoph Hafemeister,et al.  Comprehensive integration of single cell data , 2018, bioRxiv.

[8]  R. Fluhrer,et al.  Proteolytic ectodomain shedding of membrane proteins in mammals—hardware, concepts, and recent developments , 2018, The EMBO journal.

[9]  K. Harrington,et al.  Nivolumab vs investigator's choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. , 2018, Oral oncology.

[10]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[11]  Laleh Haghverdi,et al.  Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors , 2018, Nature Biotechnology.

[12]  R. Ferris,et al.  Tumor Immunology and Immunotherapy for Head and Neck Squamous Cell Carcinoma , 2018, Journal of dental research.

[13]  Y. Kluger,et al.  Efficient Algorithms for t-distributed Stochastic Neighborhood Embedding , 2017, ArXiv.

[14]  Y. Kohwi,et al.  Essential Roles of SATB1 in Specifying T Lymphocyte Subsets. , 2017, Cell reports.

[15]  C. Drake,et al.  LAG3 (CD223) as a cancer immunotherapy target , 2017, Immunological reviews.

[16]  W. Seo,et al.  Distinct requirement of Runx complexes for TCRβ enhancer activation at distinct developmental stages , 2017, Scientific Reports.

[17]  P. Verkade,et al.  PKCθ links proximal T cell and Notch signaling through localized regulation of the actin cytoskeleton , 2016, eLife.

[18]  Grace X. Y. Zheng,et al.  Massively parallel digital transcriptional profiling of single cells , 2016, Nature Communications.

[19]  K. Harrington,et al.  Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. , 2016, The New England journal of medicine.

[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]  S. Gettinger,et al.  Nivolumab Monotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer. , 2016, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  K. Tao,et al.  KLRG1 restricts memory T cell antitumor immunity , 2016, Oncotarget.

[23]  J. Radford Nivolumab for recurrent squamous-cell carcinoma of the head and neck , 2016, BDJ.

[24]  D. Vignali,et al.  Inhibitory receptors as targets for cancer immunotherapy , 2015, European journal of immunology.

[25]  Gregory L. Szeto,et al.  Nanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvants. , 2015, The Journal of clinical investigation.

[26]  David C. Smith,et al.  Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[27]  Gregory L. Szeto,et al.  Structure-based programming of lymph-node targeting in molecular vaccines , 2014, Nature.

[28]  Antoni Ribas,et al.  Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. , 2013, The New England journal of medicine.

[29]  Iannis Aifantis,et al.  Distinct T cell receptor signaling pathways drive proliferation and cytokine production in T cells , 2013, Nature Immunology.

[30]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[31]  G. Smyth,et al.  Camera: a competitive gene set test accounting for inter-gene correlation , 2012, Nucleic acids research.

[32]  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 .

[33]  L. Sherman,et al.  CD4+ T-cell help in the tumor milieu is required for recruitment and cytolytic function of CD8+ T lymphocytes. , 2010, Cancer research.

[34]  Christophe Benoist,et al.  Stability of the Regulatory T Cell Lineage in Vivo , 2010, Science.

[35]  Elizabeth A. Kruse,et al.  Membrane-bound Fas ligand only is essential for Fas-induced apoptosis , 2009, Nature.

[36]  C. Drake,et al.  LAG-3 Regulates Plasmacytoid Dendritic Cell Homeostasis1 , 2009, The Journal of Immunology.

[37]  P. Dempsey,et al.  The ADAM10 Prodomain Is a Specific Inhibitor of ADAM10 Proteolytic Activity and Inhibits Cellular Shedding Events* , 2007, Journal of Biological Chemistry.

[38]  C. Blobel,et al.  Metalloproteases regulate T‐cell proliferation and effector function via LAG‐3 , 2007, The EMBO journal.

[39]  D. Vignali,et al.  Biochemical Analysis of the Regulatory T Cell Protein Lymphocyte Activation Gene-3 (LAG-3; CD223)1 , 2004, The Journal of Immunology.

[40]  N. Copeland,et al.  A highly efficient recombineering-based method for generating conditional knockout mutations. , 2003, Genome research.

[41]  W. M. Weaver,et al.  A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. , 2001, Immunity.