Generation of NK cells with chimeric-switch receptors to overcome PD1-mediated inhibition in cancer immunotherapy

[1]  P. Riese,et al.  PD-1 expression on mouse intratumoral NK cells and its effects on NK cell phenotype , 2022, iScience.

[2]  T. Cathomen,et al.  CRISPR-Cas9 based gene editing of the immune checkpoint NKG2A enhances NK cell mediated cytotoxicity against multiple myeloma , 2022, Oncoimmunology.

[3]  Jeffrey S. Miller,et al.  Low-density PD-1 expression on resting human natural killer cells is functional and upregulated after transplantation. , 2021, Blood advances.

[4]  M. Camandaroba,et al.  Anti‐PD1 versus anti‐PD‐L1 immunotherapy in first‐line therapy for advanced non‐small cell lung cancer: A systematic review and meta‐analysis , 2021, Thoracic cancer.

[5]  S. Steinberg,et al.  Avelumab, a PD‐L1 Inhibitor, in Combination with Hypofractionated Radiotherapy and the Abscopal Effect in Relapsed Refractory Multiple Myeloma , 2021, The oncologist.

[6]  Feng Xu,et al.  Immune checkpoint molecules in natural killer cells as potential targets for cancer immunotherapy , 2020, Signal Transduction and Targeted Therapy.

[7]  Jianzhu Chen,et al.  CAR-NK cells: A promising cellular immunotherapy for cancer , 2020, EBioMedicine.

[8]  J. Schlom,et al.  PD-L1 targeting high-affinity NK (t-haNK) cells induce direct antitumor effects and target suppressive MDSC populations , 2020, Journal for immunotherapy of cancer.

[9]  P. Thall,et al.  Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. , 2020, The New England journal of medicine.

[10]  Z. Zeng,et al.  Predictive biomarkers and mechanisms underlying resistance to PD1/PD-L1 blockade cancer immunotherapy , 2020, Molecular Cancer.

[11]  Y. You,et al.  EGFRvIII-CAR-T Cells with PD-1 Knockout Have Improved Anti-Glioma Activity , 2020, Pathology & Oncology Research.

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

[13]  Xiaoyin Wang,et al.  Structure-based rational design of a novel chimeric PD1-NKG2D receptor for natural killer cells. , 2019, Molecular immunology.

[14]  G. Coukos,et al.  Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance , 2019, Nature Reviews Clinical Oncology.

[15]  S. Jagannath,et al.  Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): a randomised, open-label, phase 3 trial. , 2019, The Lancet. Haematology.

[16]  Zhiqiang Wu,et al.  PD-1 silencing impairs the anti-tumor function of chimeric antigen receptor modified T cells by inhibiting proliferation activity , 2019, Journal of Immunotherapy for Cancer.

[17]  Jason J. Z. Liao,et al.  Pembrolizumab plus pomalidomide and dexamethasone for patients with relapsed or refractory multiple myeloma (KEYNOTE-183): a randomised, open-label, phase 3 trial. , 2019, The Lancet. Haematology.

[18]  S. Perner,et al.  Immune Cell Infiltration of the Primary Tumor, Not PD-L1 Status, Is Associated With Improved Response to Checkpoint Inhibition in Metastatic Melanoma , 2019, Front. Med..

[19]  O. Lantz,et al.  Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells , 2018, Cell.

[20]  R. Ferris,et al.  PD-L1 Mediates Dysfunction in Activated PD-1+ NK Cells in Head and Neck Cancer Patients , 2018, Cancer Immunology Research.

[21]  E. Levy,et al.  Avelumab, an IgG1 anti-PD-L1 Immune Checkpoint Inhibitor, Triggers NK Cell-Mediated Cytotoxicity and Cytokine Production Against Triple Negative Breast Cancer Cells , 2018, Front. Immunol..

[22]  R. Sun,et al.  Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity , 2018, Nature Immunology.

[23]  Andre H Crepaldi,et al.  Daratumumab plus Bortezomib, Melphalan, and Prednisone for Untreated Myeloma , 2017, The New England journal of medicine.

[24]  Y. Cheng,et al.  Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers , 2017, Oncogene.

[25]  J. Lunceford,et al.  PD-L2 Expression in Human Tumors: Relevance to Anti-PD-1 Therapy in Cancer , 2017, Clinical Cancer Research.

[26]  H. Ljunggren,et al.  Natural killer cell-mediated immunosurveillance of human cancer. , 2017, Seminars in immunology.

[27]  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.

[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]  G. Schackert,et al.  DAP12-Based Activating Chimeric Antigen Receptor for NK Cell Tumor Immunotherapy , 2015, The Journal of Immunology.

[30]  P. Parren,et al.  Preclinical Evidence for the Therapeutic Potential of CD38-Targeted Immuno-Chemotherapy in Multiple Myeloma Patients Refractory to Lenalidomide and Bortezomib , 2014, Clinical Cancer Research.

[31]  David A. Scott,et al.  Genome engineering using the CRISPR-Cas9 system , 2013, Nature Protocols.

[32]  E. Alici,et al.  Inhibition of intracellular antiviral defense mechanisms augments lentiviral transduction of human natural killer cells: implications for gene therapy. , 2012, Human gene therapy.

[33]  M. Caligiuri,et al.  The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. , 2010, Blood.

[34]  Eric O Long,et al.  Regulation of human NK-cell cytokine and chemokine production by target cell recognition. , 2010, Blood.

[35]  L. Lanier DAP10‐ and DAP12‐associated receptors in innate immunity , 2009, Immunological reviews.

[36]  Lewis L Lanier,et al.  Up on the tightrope: natural killer cell activation and inhibition , 2008, Nature Immunology.

[37]  B. Fehse,et al.  A multicolor panel of novel lentiviral "gene ontology" (LeGO) vectors for functional gene analysis. , 2008, Molecular Therapy.

[38]  H. Ljunggren,et al.  Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components. , 2008, Blood.

[39]  H. Ljunggren,et al.  Anti-myeloma activity of endogenous and adoptively transferred activated natural killer cells in experimental multiple myeloma model. , 2007, Experimental hematology.

[40]  K. Wucherpfennig,et al.  The activating NKG2D receptor assembles in the membrane with two signaling dimers into a hexameric structure. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[41]  William C Hahn,et al.  Lentivirus-delivered stable gene silencing by RNAi in primary cells. , 2003, RNA.

[42]  Jun Wu,et al.  Dap10 and Dap12 Form Distinct, but Functionally Cooperative, Receptor Complexes in Natural Killer Cells , 2000, The Journal of experimental medicine.

[43]  D. Trono,et al.  A Third-Generation Lentivirus Vector with a Conditional Packaging System , 1998, Journal of Virology.

[44]  R. Biassoni,et al.  Molecular Cloning of NKp46: A Novel Member of the Immunoglobulin Superfamily Involved in Triggering of Natural Cytotoxicity , 1998, The Journal of experimental medicine.