Long-term persistence and functionality of adoptively transferred antigen-specific T cells with genetically ablated PD-1 expression

Significance Adoptive transfer of antigen-specific T cells has shown impressive clinical success in cancer and infectious diseases. However, due to constant stimulation, the transferred cells can switch into dysfunctional states marked by the expression of programmed cell death protein 1 (PD-1). Recent breakthroughs in cell engineering enabled the precise genetic ablation of this inhibitory receptor via CRISPR/Cas9 technology. First studies have shown benefits of PD-1 knockout for T cell functionality in short-term experiments but potential loss of fitness at later stages. Here, we could show that PD-1 ablation is not necessarily detrimental for long-term functionality and persistence of engineered T cells. These findings are of particular importance since the first PD-1 KO T cell products have recently entered into the clinics.

[1]  Abhishek K. Jha,et al.  Metabolic regulation by PD-1 signaling promotes long-lived quiescent CD8 T cell memory in mice , 2021, Science Translational Medicine.

[2]  Jesse M. Platt,et al.  Inhibitory signaling sustains a distinct early memory CD8+ T cell precursor that is resistant to DNA damage , 2021, Science Immunology.

[3]  V. Buchholz,et al.  Early emergence of T central memory precursors programs clonal dominance during chronic viral infection , 2020, Nature Immunology.

[4]  M. Delorenzi,et al.  Central memory CD8+ T cells derive from stem-like Tcf7hi effector cells in the absence of cytotoxic differentiation. , 2020, Immunity.

[5]  Loise M. Francisco,et al.  The PD-1 Pathway Regulates Development and Function of Memory CD8+ T Cells following Respiratory Viral Infection. , 2020, Cell reports.

[6]  K. Tsumoto,et al.  GPC1 specific CAR-T cells eradicate established solid tumor without adverse effects and synergize with anti-PD-1 Ab , 2020, eLife.

[7]  Howard Y. Chang,et al.  CRISPR-engineered T cells in patients with refractory cancer , 2020, Science.

[8]  Matthew J. Frigault,et al.  CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma , 2019, Journal of Immunotherapy for Cancer.

[9]  E. McGowan,et al.  PD-1 disrupted CAR-T cells in the treatment of solid tumors: Promises and challenges. , 2019, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[10]  D. Busch,et al.  T cell engineering for adoptive T cell therapy: safety and receptor avidity , 2019, Cancer Immunology, Immunotherapy.

[11]  Meijuan Huang,et al.  Engineered triple inhibitory receptor resistance improves anti-tumor CAR-T cell performance via CD56 , 2019, Nature Communications.

[12]  Christian Stemberger,et al.  Orthotopic replacement of T-cell receptor α- and β-chains with preservation of near-physiological T-cell function , 2019, Nature Biomedical Engineering.

[13]  Xiaojing Ma,et al.  CRISPR/Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell effector functions , 2018, Cancer Immunology, Immunotherapy.

[14]  J. Szer,et al.  Nivolumab for Relapsed or Residual Haematological Malignancies after Allogeneic Haematopoietic Stem Cell Transplantation (NIVALLO) , 2018, Blood.

[15]  Hua Jiang,et al.  Disruption of PD-1 Enhanced the Anti-tumor Activity of Chimeric Antigen Receptor T Cells Against Hepatocellular Carcinoma , 2018, Front. Pharmacol..

[16]  G. Freeman,et al.  Role of PD-1 during effector CD8 T cell differentiation , 2018, Proceedings of the National Academy of Sciences.

[17]  Wei Zhang,et al.  CRISPR knock out of programmed cell death protein 1 enhances anti-tumor activity of cytotoxic T lymphocytes , 2017, Oncotarget.

[18]  L. Čičin-Šain,et al.  Exhaustion and Inflation at Antipodes of T Cell Responses to Chronic Virus Infection. , 2017, Trends in microbiology.

[19]  Junnian Zheng,et al.  Current status and future prospects of the strategy of combining CAR‑T with PD‑1 blockade for antitumor therapy (Review). , 2017, Molecular medicine reports.

[20]  R. Rad,et al.  PD-1 is a haploinsufficient suppressor of T cell lymphomagenesis , 2017, Nature.

[21]  Emily R. Levy,et al.  Donor CD19 CAR T cells exert potent graft-versus-lymphoma activity with diminished graft-versus-host activity , 2017, Nature Medicine.

[22]  A. Cheng,et al.  CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells , 2016, Cell Research.

[23]  Carl H. June,et al.  Multiplex Genome Editing to Generate Universal CAR T Cells Resistant to PD1 Inhibition , 2016, Clinical Cancer Research.

[24]  D. Busch,et al.  Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. , 2016, The Journal of clinical investigation.

[25]  M. Ebinger,et al.  T-cell responses against CD19+ pediatric acute lymphoblastic leukemia mediated by bispecific T-cell engager (BiTE) are regulated contrarily by PD-L1 and CD80/CD86 on leukemic blasts , 2016, Oncotarget.

[26]  Matthieu Texier,et al.  Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination , 2016, Nature Reviews Clinical Oncology.

[27]  E. Wherry,et al.  Costimulatory and Coinhibitory Receptor Pathways in Infectious Disease. , 2016, Immunity.

[28]  Chun Jimmie Ye,et al.  CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells , 2016, Scientific Reports.

[29]  J. Soria,et al.  Immune-related adverse events with immune checkpoint blockade: a comprehensive review. , 2016, European journal of cancer.

[30]  E. Wherry,et al.  Molecular and cellular insights into T cell exhaustion , 2015, Nature Reviews Immunology.

[31]  E. Wherry,et al.  Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells , 2015, The Journal of experimental medicine.

[32]  D. Fearon,et al.  T cell exclusion, immune privilege, and the tumor microenvironment , 2015, Science.

[33]  Emmanuel C. Alozie,et al.  Promises and Challenges , 2015 .

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

[35]  Pamela A Shaw,et al.  Chimeric antigen receptor T cells for sustained remissions in leukemia. , 2014, The New England journal of medicine.

[36]  Christian Stemberger,et al.  Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8(+) central memory T cells. , 2014, Immunity.

[37]  D. Dolfi,et al.  Multifactorial T-cell Hypofunction That Is Reversible Can Limit the Efficacy of Chimeric Antigen Receptor–Transduced Human T cells in Solid Tumors , 2014, Clinical Cancer Research.

[38]  M. Smyth,et al.  Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells , 2013, Clinical Cancer Research.

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

[40]  Michel Sadelain,et al.  CD19 CAR-Targeted T Cells Induce Long-Term Remission and B Cell Aplasia in an Immunocompetent Mouse Model of B Cell Acute Lymphoblastic Leukemia , 2013, PloS one.

[41]  Han-Hsuan Fu,et al.  A Potential New Pathway for PD-L1 Costimulation of the CD8-T Cell Response to Listeria monocytogenes Infection , 2013, PloS one.

[42]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

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

[44]  Alison P. Klein,et al.  Colocalization of Inflammatory Response with B7-H1 Expression in Human Melanocytic Lesions Supports an Adaptive Resistance Mechanism of Immune Escape , 2012, Science Translational Medicine.

[45]  S. Riddell,et al.  A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. , 2011, Blood.

[46]  E. Usherwood,et al.  Programmed Death 1 Regulates Development of Central Memory CD8 T Cells after Acute Viral Infection , 2011, The Journal of Immunology.

[47]  Kristen E. Pauken,et al.  The role of the PD‐1 pathway in autoimmunity and peripheral tolerance , 2011, Annals of the New York Academy of Sciences.

[48]  W. Wilson,et al.  Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. , 2010, Blood.

[49]  Scott N. Mueller,et al.  High antigen levels are the cause of T cell exhaustion during chronic viral infection , 2009, Proceedings of the National Academy of Sciences.

[50]  A. Bertoletti,et al.  Characterization of Hepatitis B Virus (HBV)-Specific T-Cell Dysfunction in Chronic HBV Infection , 2007, Journal of Virology.

[51]  Philip J. R. Goulder,et al.  PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression , 2006, Nature.

[52]  M. Massari,et al.  PD-1 Expression in Acute Hepatitis C Virus (HCV) Infection Is Associated with HCV-Specific CD8 Exhaustion , 2006, Journal of Virology.

[53]  D. McGavern,et al.  Reprogramming of antiviral T cells prevents inactivation and restores T cell activity during persistent viral infection. , 2006, The Journal of clinical investigation.

[54]  Yoshimasa Tanaka,et al.  Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[55]  T. Okazaki,et al.  Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. , 2001, Science.

[56]  T. Honjo,et al.  Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. , 1999, Immunity.

[57]  Tony Travers,et al.  A Comprehensive Review , 1998 .

[58]  T. Honjo,et al.  Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. , 1996, International immunology.