Knockout of the inhibitory receptor TIGIT enhances the antitumor response of ex vivo expanded NK cells and prevents fratricide with therapeutic Fc-active TIGIT antibodies

Background Inhibitory receptor T-cell Immunoreceptor with Ig and ITIM domains (TIGIT) expressed by Natural Killer (NK) and T cells regulates cancer immunity and has been touted as the next frontier in the development of cancer immunotherapeutics. Although early results of anti-TIGIT and its combinations with antiprogrammed death-ligand 1 were highly exciting, results from an interim analysis of phase III trials are disappointing. With mixed results, there is a need to understand the effects of therapeutic anti-TIGIT on the TIGIT+ immune cells to support its clinical use. Most of the TIGIT antibodies in development have an Fc-active domain, which binds to Fc receptors on effector cells. In mouse models, Fc-active anti-TIGIT induced superior immunity, while Fc receptor engagement was required for its efficacy. NK-cell depletion compromised the antitumor immunity of anti-TIGIT indicating the essential role of NK cells in the efficacy of anti-TIGIT. Since NK cells express TIGIT and Fc-receptor CD16, Fc-active anti-TIGIT may deplete NK cells via fratricide, which has not been studied. Methods CRISPR-Cas9-based TIGIT knockout (KO) was performed in expanded NK cells. Phenotypic and transcriptomic properties of TIGIT KO and wild-type (WT) NK cells were compared with flow cytometry, CyTOF, and RNA sequencing. The effect of TIGIT KO on NK-cell cytotoxicity was determined by calcein-AM release and live cell imaging-based cytotoxicity assays. The metabolic properties of TIGIT KO and WT NK cells were compared with a Seahorse analyzer. The effect of the Fc-component of anti-TIGIT on NK-cell fratricide was determined by co-culturing WT and TIGIT KO NK cells with Fc-active and Fc-inactive anti-TIGIT. Results TIGIT KO increased the cytotoxicity of NK cells against multiple cancer cell lines including spheroids. TIGIT KO NK cells upregulated mTOR complex 1 (mTORC1) signaling and had better metabolic fitness with an increased basal glycolytic rate when co-cultured with cancer cells compared with WT NK cells. Importantly, TIGIT KO prevented NK-cell fratricide when combined with Fc-active anti-TIGIT. Conclusions TIGIT KO in ex vivo expanded NK cells increased their cytotoxicity and metabolic fitness and prevented NK-cell fratricide when combined with Fc-active anti-TIGIT antibodies. These fratricide-resistant TIGIT KO NK cells have therapeutic potential alone or in combination with Fc-active anti-TIGIT antibodies to enhance their efficacy.

[1]  A. Copik,et al.  TIGIT Expression on Activated NK Cells Correlates with Greater Anti-Tumor Activity but Promotes Functional Decline upon Lung Cancer Exposure: Implications for Adoptive Cell Therapy and TIGIT-Targeted Therapies , 2023, Cancers.

[2]  J. Gariépy,et al.  The IgV domain of the poliovirus receptor alone is immunosuppressive and binds to its receptors with comparable affinity , 2023, Scientific Reports.

[3]  S. McComb,et al.  Simultaneous engineering of natural killer cells for CAR transgenesis and CRISPR-Cas9 knockout using retroviral particles. , 2023, Molecular therapy. Methods & clinical development.

[4]  C. Bourin,et al.  497 Evaluation of the TIGIT+ immune subset depletion effect of the anti-TIGIT antibody M6223 , 2022, Regular and Young Investigator Award Abstracts.

[5]  S. Schleicher,et al.  Preclinical Evaluation of CRISPR-Edited CAR-NK-92 Cells for Off-the-Shelf Treatment of AML and B-ALL , 2022, International journal of molecular sciences.

[6]  Robert W. Hsieh,et al.  Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. , 2022, The Lancet. Oncology.

[7]  I. Garrido-Laguna,et al.  Clinical Development of Anti-TIGIT Antibodies for Immunotherapy of Cancer , 2022, Current Oncology Reports.

[8]  A. Copik,et al.  Cryopreserved PM21-Particle-Expanded Natural Killer Cells Maintain Cytotoxicity and Effector Functions In Vitro and In Vivo , 2022, Frontiers in Immunology.

[9]  I. Mellman,et al.  TIGIT-CD226-PVR axis: advancing immune checkpoint blockade for cancer immunotherapy , 2022, Journal for ImmunoTherapy of Cancer.

[10]  Lei Jiang,et al.  An Fc-Competent Anti-Human TIGIT Blocking Antibody Ociperlimab (BGB-A1217) Elicits Strong Immune Responses and Potent Anti-Tumor Efficacy in Pre-Clinical Models , 2022, Frontiers in Immunology.

[11]  F. Soriano,et al.  258 AB308 is an anti-TIGIT antibody that enhances immune activation and anti-tumor immunity alone and in combination with other I-O therapeutic agents , 2021, Journal for ImmunoTherapy of Cancer.

[12]  H. Huls,et al.  Adoptive immunotherapy with double‐bright (CD56bright/CD16bright) expanded natural killer cells in patients with relapsed or refractory acute myeloid leukaemia: a proof‐of‐concept study , 2021, British journal of haematology.

[13]  M. Peppelenbosch,et al.  TIGIT, the Next Step Towards Successful Combination Immune Checkpoint Therapy in Cancer , 2021, Frontiers in Immunology.

[14]  S. Gardai,et al.  Abstract 1583: SEA-TGT is an empowered anti-TIGIT antibody that displays superior combinatorial activity with several therapeutic agents , 2021, Immunology.

[15]  Zhirong Shen,et al.  Abstract 1854: A Fc-competent anti-human TIGIT blocking antibody BGB-A1217 elicits strong immune responses and potent anti-tumor efficacy in pre-clinical models , 2021, Immunology.

[16]  J. Machiels,et al.  Abstract CT118: Preliminary data from Phase I first-in-human study of EOS884448, a novel potent anti-TIGIT antibody, monotherapy shows favorable tolerability profile and early signsof clinical activity in immune-resistant advanced cancers , 2021, Clinical Trials.

[17]  D. Davar,et al.  SGNTGT-001: A phase 1 study of SEA-TGT, an effector-function enhanced monoclonal antibody (mAb), in advanced malignancies (trial in progress). , 2021 .

[18]  A. Copik,et al.  Natural Killer Cells: The Linchpin for Successful Cancer Immunotherapy , 2021, Frontiers in Immunology.

[19]  Valentina Perri,et al.  Increased sCD163 and sCD14 Plasmatic Levels and Depletion of Peripheral Blood Pro-Inflammatory Monocytes, Myeloid and Plasmacytoid Dendritic Cells in Patients With Severe COVID-19 Pneumonia , 2021, Frontiers in Immunology.

[20]  Steven Lin,et al.  A robust platform for expansion and genome editing of primary human natural killer cells , 2021, The Journal of experimental medicine.

[21]  Kimberley Burt,et al.  TIGIT blockade enhances NK cell activity against autologous HIV‐1‐infected CD4+ T cells , 2021, Clinical & translational immunology.

[22]  N. Wald,et al.  Restoration of T-cell Effector Function, Depletion of Tregs, and Direct Killing of Tumor Cells: The Multiple Mechanisms of Action of a-TIGIT Antagonist Antibodies , 2020, Molecular Cancer Therapeutics.

[23]  S. Thorpe,et al.  Analysis of tumor-infiltrating NK and T cells highlights IL-15 stimulation and TIGIT blockade as a combination immunotherapy strategy for soft tissue sarcomas , 2020, Journal for ImmunoTherapy of Cancer.

[24]  E. Paltrinieri,et al.  253 Anti-TIGIT antibodies require enhanced FcγR co-engagement for optimal T and NK cell-dependent anti-tumor immunity , 2020 .

[25]  W. Seghezzi,et al.  Effective Anti-tumor Response by TIGIT Blockade Associated With FcγR Engagement and Myeloid Cell Activation , 2020, Frontiers in Immunology.

[26]  Jeffrey S. Miller,et al.  Exploring the NK cell platform for cancer immunotherapy , 2020, Nature Reviews Clinical Oncology.

[27]  J. Sui,et al.  A Cross-Species Reactive TIGIT-Blocking Antibody Fc Dependently Confers Potent Antitumor Effects , 2020, The Journal of Immunology.

[28]  E. Dolgin Antibody engineers seek optimal drug targeting TIGIT checkpoint , 2020, Nature Biotechnology.

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

[30]  Dean Anthony Lee,et al.  CD38 deletion of human primary NK cells eliminates daratumumab-induced fratricide and boosts their effector activity. , 2020, Blood.

[31]  Anton Nekrutenko,et al.  The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2020 update , 2020, Nucleic Acids Res..

[32]  Steven Lin,et al.  Enhanced NK-92 Cytotoxicity by CRISPR Genome Engineering Using Cas9 Ribonucleoproteins , 2020, Frontiers in Immunology.

[33]  Robert W. Hsieh,et al.  Primary analysis of a randomized, double-blind, phase II study of the anti-TIGIT antibody tiragolumab (tira) plus atezolizumab (atezo) versus placebo plus atezo as first-line (1L) treatment in patients with PD-L1-selected NSCLC (CITYSCAPE). , 2020 .

[34]  Dean Anthony Lee,et al.  Evaluation of serum-free media formulations in feeder cell-stimulated expansion of natural killer cells. , 2020, Cytotherapy.

[35]  Giovanni Parmigiani,et al.  ComBat-seq: batch effect adjustment for RNA-seq count data , 2020, bioRxiv.

[36]  D. Campana,et al.  NK cells for cancer immunotherapy , 2020, Nature Reviews Drug Discovery.

[37]  Haoyu Sun,et al.  The Rise of NK Cell Checkpoints as Promising Therapeutic Targets in Cancer Immunotherapy , 2019, Front. Immunol..

[38]  Dean Anthony Lee,et al.  Expression of carcinoma, apoptosis, and cell‐death–related genes are determinants for sensitivity of pediatric cancer cell lines to lysis by natural killer cells , 2019, Pediatric blood & cancer.

[39]  T. Waldmann,et al.  First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia. , 2019, Blood advances.

[40]  Jinwoo Hong,et al.  Cryopreserved Human Natural Killer Cells Exhibit Potent Antitumor Efficacy against Orthotopic Pancreatic Cancer through Efficient Tumor-Homing and Cytolytic Ability (Running Title: Cryopreserved NK Cells Exhibit Antitumor Effect) , 2019, Cancers.

[41]  Dean Anthony Lee Cellular therapy: Adoptive immunotherapy with expanded natural killer cells , 2019, Immunological reviews.

[42]  A. Pera,et al.  DNAM-1 and the TIGIT/PVRIG/TACTILE Axis: Novel Immune Checkpoints for Natural Killer Cell-Based Cancer Immunotherapy , 2019, Cancers.

[43]  J. M. Peralta,et al.  The Advances in Molecular and New Point-of-Care (POC) Diagnosis of Schistosomiasis Pre- and Post-praziquantel Use: In the Pursuit of More Reliable Approaches for Low Endemic and Non-endemic Areas , 2019, Front. Immunol..

[44]  J. Madore,et al.  Integrated molecular and immunophenotypic analysis of NK cells in anti-PD-1 treated metastatic melanoma patients , 2018, Oncoimmunology.

[45]  Gang Li,et al.  Natural Killer Cell-Based Cancer Immunotherapy: A Review on 10 Years Completed Clinical Trials , 2018, Cancer investigation.

[46]  Hana Choi,et al.  Optimization of Large-Scale Expansion and Cryopreservation of Human Natural Killer Cells for Anti-Tumor Therapy , 2018, Immune network.

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

[48]  K. Mclaughlin,et al.  Generation of Knock-out Primary and Expanded Human NK Cells Using Cas9 Ribonucleoproteins. , 2018, Journal of visualized experiments : JoVE.

[49]  L. Swiech,et al.  Selective FcγR Co-engagement on APCs Modulates the Activity of Therapeutic Antibodies Targeting T Cell Antigens. , 2018, Cancer cell.

[50]  S. Asthana,et al.  A natural killer–dendritic cell axis defines checkpoint therapy–responsive tumor microenvironments , 2018, Nature Medicine.

[51]  Christopher M. Jackson,et al.  TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM , 2018, Oncoimmunology.

[52]  E. Sahai,et al.  NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control , 2018, Cell.

[53]  S. Lonial,et al.  Effects of daratumumab on natural killer cells and impact on clinical outcomes in relapsed or refractory multiple myeloma. , 2017, Blood advances.

[54]  K. Excoffon,et al.  Poliovirus Receptor: More than a simple viral receptor. , 2017, Virus research.

[55]  E. Kleinerman,et al.  Interferon Gamma Induces Changes in Natural Killer (NK) Cell Ligand Expression and Alters NK Cell-Mediated Lysis of Pediatric Cancer Cell Lines , 2017, Front. Immunol..

[56]  David K. Finlay,et al.  What Fuels Natural Killers? Metabolism and NK Cell Responses , 2017, Front. Immunol..

[57]  P. Kruzliak,et al.  Cryopreserved NK cells in the treatment of haematological malignancies: preclinical study , 2016, Journal of Cancer Research and Clinical Oncology.

[58]  Jeffrey T Leek,et al.  Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown , 2016, Nature Protocols.

[59]  A. Copik,et al.  Natural killer cells stimulated with PM21 particles expand and biodistribute in vivo: Clinical implications for cancer treatment. , 2016, Cytotherapy.

[60]  K. Rezvani,et al.  Infusion of Ex Vivo Expanded Allogeneic Cord Blood-Derived Natural Killer Cells in Combination with Autologous Stem Cell Transplantation for Multiple Myeloma: Results of a Phase I Study , 2015 .

[61]  Shuhua Liu Jung Cheol Shin, Gerard A. Postiglione, and Futao Huang (eds.): Mass higher education development in East Asia: strategy, quality, and challenges , 2015, Higher Education.

[62]  L. Chan,et al.  A Novel Method for Assessment of Natural Killer Cell Cytotoxicity Using Image Cytometry , 2015, PloS one.

[63]  H. Hou,et al.  TIGIT expression levels on human NK cells correlate with functional heterogeneity among healthy individuals , 2015, European journal of immunology.

[64]  A. Copik,et al.  Generation of highly cytotoxic natural killer cells for treatment of acute myelogenous leukemia using a feeder-free, particle-based approach. , 2015, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[65]  J. Hackney,et al.  The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. , 2014, Cancer cell.

[66]  Björn Usadel,et al.  Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..

[67]  L. Hurton,et al.  Membrane-Bound IL-21 Promotes Sustained Ex Vivo Proliferation of Human Natural Killer Cells , 2012, PloS one.

[68]  Erin F. Simonds,et al.  Single-Cell Mass Cytometry of Differential Immune and Drug Responses Across a Human Hematopoietic Continuum , 2011, Science.

[69]  Davis J. McCarthy,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[70]  Eric Vivier,et al.  Functions of natural killer cells , 2008, Nature Immunology.

[71]  J. Kirkwood,et al.  Natural killer–dendritic cell cross-talk in cancer immunotherapy , 2005, Expert opinion on biological therapy.

[72]  J. Mesirov,et al.  From the Cover: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005 .

[73]  C. Le,et al.  Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. , 2005, Blood.

[74]  Katia Perruccio,et al.  Effectiveness of Donor Natural Killer Cell Alloreactivity in Mismatched Hematopoietic Transplants , 2002, Science.

[75]  A. Copik,et al.  Kinetic, imaging based assay to measure NK cell cytotoxicity against adherent cells. , 2023, Methods in Cell Biology.

[76]  E. Chiang,et al.  TIGIT: A Key Inhibitor of the Cancer Immunity Cycle. , 2017, Trends in immunology.

[77]  G. Nuovo,et al.  Natural killer cells produce T cell-recruiting chemokines in response to antibody-coated tumor cells. , 2006, Cancer research.

[78]  JoVE Video Dataset , 2022 .