Enrichment of T-cell proliferation and memory gene signatures of CD79A/CD40 costimulatory domain potentiates CD19CAR-T cell functions

CD19 chimeric antigen receptor (CAR) T-cells have demonstrated remarkable outcomes in B-cell malignancies. Recently, the novel CD19CAR-T cells incorporated with B-cell costimulatory molecules of CD79A/CD40 demonstrated superior antitumor activity in the B-cell lymphoma model compared with CD28 or 4-1BB. Here, we investigated the intrinsic transcriptional gene underlying the functional advantage of CD19.79A.40z CAR-T cells following CD19 antigen exposure using transcriptome analysis compared to CD28 or 4-1BB. Notably, CD19.79A.40z CAR-T cells up-regulated genes involved in T-cell activation, T-cell proliferation, and NF-κB signaling, whereas down-regulated genes associated with T-cell exhaustion and apoptosis. Interestingly, CD19.79A.40z CAR- and CD19.BBz CAR-T cells were enriched in almost similar pathways. Furthermore, gene set enrichment analysis demonstrated the enrichment of genes, which were previously identified to correlate with T-cell proliferation, interferon signaling pathway, and naïve and memory T-cell signatures, and down-regulated T-cell exhaustion genes in CD79A/CD40, compared with the T-cell costimulatory domain. The CD19.79A.40z CAR-T cells also up-regulated genes related to glycolysis and fatty acid metabolism, which are necessary to drive T-cell proliferation and differentiation compared with conventional CD19CAR-T cells. Our study provides a comprehensive insight into the understanding of gene signatures that potentiates the superior antitumor functions by CD19CAR-T cells incorporated with the CD79A/CD40 costimulatory domain.

[1]  Xingxu Huang,et al.  IKZF3 deficiency potentiates chimeric antigen receptor T cells targeting solid tumors. , 2021, Cancer letters.

[2]  J. Leitner,et al.  Composite CD79A/CD40 costimulatory endodomain enhances CD19CAR-T cell proliferation and survival. , 2021, Molecular therapy : the journal of the American Society of Gene Therapy.

[3]  Yiping Fan,et al.  MyD88/CD40 signaling retains CAR T cells in a less differentiated state , 2020, JCI insight.

[4]  H. Einsele,et al.  BATF3 programs CD8+ T cell memory , 2020, Nature Immunology.

[5]  A. Regev,et al.  A Distinct Transcriptional Program in Human CAR T Cells Bearing the 4-1BB Signaling Domain Revealed by scRNA-Seq. , 2020, Molecular therapy : the journal of the American Society of Gene Therapy.

[6]  C. June,et al.  4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling , 2020, Science Signaling.

[7]  K. Oestreich,et al.  Ikaros Zinc Finger Transcription Factors: Regulators of Cytokine Signaling Pathways and CD4+ T Helper Cell Differentiation , 2019, Front. Immunol..

[8]  J. Vilo,et al.  g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update) , 2019, Nucleic Acids Res..

[9]  M. Milone,et al.  An introduction to chimeric antigen receptor (CAR) T‐cell immunotherapy for human cancer , 2019, American journal of hematology.

[10]  Wei-Chun Chang,et al.  Constitutively active MyD88/CD40 costimulation enhances expansion and efficacy of chimeric antigen receptor T cells targeting hematological malignancies , 2019, Leukemia.

[11]  Ke Liu,et al.  FDA Approval Summary: Tisagenlecleucel for Treatment of Patients with Relapsed or Refractory B-cell Precursor Acute Lymphoblastic Leukemia , 2018, Clinical Cancer Research.

[12]  Qing Zhang,et al.  Chimeric Antigen Receptor-T Cells with 4-1BB Co-Stimulatory Domain Present a Superior Treatment Outcome than Those with CD28 Domain Based on Bioinformatics , 2018, Acta Haematologica.

[13]  Yongliang Zhang,et al.  4-1BB enhancement of CAR T function requires NF-κB and TRAFs. , 2018, JCI insight.

[14]  Hans Bitter,et al.  Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia , 2018, Nature Medicine.

[15]  Stephen J. Schuster,et al.  Chimeric Antigen Receptor T Cells in Refractory B‐Cell Lymphomas , 2017, The New England journal of medicine.

[16]  Michel Sadelain,et al.  Therapeutic T cell engineering , 2017, Nature.

[17]  T. Sparwasser,et al.  Metabolic pathways in T cell activation and lineage differentiation. , 2016, Seminars in immunology.

[18]  D. Maloney,et al.  Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo , 2015, Leukemia.

[19]  C. June,et al.  Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. , 2016, Immunity.

[20]  M. Sadelain,et al.  Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T Cells. , 2015, Cancer cell.

[21]  Michel Sadelain,et al.  The pharmacology of second-generation chimeric antigen receptors , 2015, Nature Reviews Drug Discovery.

[22]  R. Kaplan,et al.  4-1BB Costimulation Ameliorates T Cell Exhaustion Induced by Tonic Signaling of Chimeric Antigen Receptors , 2015, Nature Medicine.

[23]  J. Rathmell,et al.  The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. , 2014, Cell metabolism.

[24]  Linda V. Sinclair,et al.  Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation , 2013, Nature Immunology.

[25]  D. Sabatini,et al.  mTOR Signaling in Growth Control and Disease , 2012, Cell.

[26]  C. Turtle,et al.  Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells. , 2012, Blood.

[27]  D. Green,et al.  The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. , 2011, Immunity.

[28]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[29]  C. Thompson,et al.  The CD28 signaling pathway regulates glucose metabolism. , 2002, Immunity.

[30]  Chi V. Dang,et al.  c-Myc Target Genes Involved in Cell Growth, Apoptosis, and Metabolism , 1999, Molecular and Cellular Biology.