DNT Cell-based Immunotherapy: Progress and Applications

Cancer immunotherapy has firmly established a dominant status in recent years. Adoptive cellular immunotherapy (ACI) is the main branch of immunotherapy. Recently, the immune effector cells of ACI, such as T cells, NK cells, and genetically engineered cells, have been used to achieve significant clinical benefits in the treatment of malignant tumors. However, the clinical applications have limitations, including toxicity, unexpectedly low efficiency, high costs and strict technical requirements. More exploration is needed to optimize ACI for cancer patients. CD3+CD4-CD8- double negative T cells (DNTs) have emerged as functional antitumor effector cells, according to the definition of adoptive immunotherapy. They constitute a kind of T cell subset that mediates nontumor antigen-restricted immunity and has important immune regulatory functions. Preclinical experiments showed that DNTs had a dual effect by killing tumor cells and inhibiting graft-versus-host disease. Notably, DNTs can be acquired from healthy donors and expanded in vitro; thus, allogeneic DNTs may be provided as “off-the-shelf” cellular products that can be readily available for direct clinical application. We review the progress and application of DNTs in immunotherapy. DNTs may provide some novel perspectives on cancer immunotherapy.

[1]  R. Hoffman,et al.  Double-negative T Cells Inhibit Proliferation and Invasion of Human Pancreatic Cancer Cells in Co-culture , 2019, AntiCancer Research.

[2]  Zhongtao Zhang,et al.  Double negative T cells mediate Lag3-dependent antigen-specific protection in allergic asthma , 2019, Nature Communications.

[3]  C. Peano,et al.  Neutrophils Driving Unconventional T Cells Mediate Resistance against Murine Sarcomas and Selected Human Tumors , 2019, Cell.

[4]  Qin Li,et al.  Efficacy and safety of combination immunotherapy for malignant solid tumors: A systematic review and meta-analysis. , 2019, Critical reviews in oncology/hematology.

[5]  B. Monk,et al.  Safety and efficacy of adoptive cell transfer using autologous tumor infiltrating lymphocytes (LN-145) for treatment of recurrent, metastatic, or persistent cervical carcinoma. , 2019, Journal of Clinical Oncology.

[6]  A. Santoni,et al.  Activation of liver X receptor up‐regulates the expression of the NKG2D ligands MICA and MICB in multiple myeloma through different molecular mechanisms , 2019, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[7]  S. Jagannath,et al.  Anti‐BCMA CAR T‐Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma , 2019, The New England journal of medicine.

[8]  C. June,et al.  Emerging Cellular Therapies for Cancer. , 2019, Annual review of immunology.

[9]  Yingrui Li,et al.  Molecular Mechanisms and Countermeasures of Immunotherapy Resistance in Malignant Tumor , 2019, Journal of Cancer.

[10]  M. Sadelain,et al.  CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape , 2019, Nature.

[11]  M. Tsao,et al.  Targeting late-stage non-small cell lung cancer with a combination of DNT cellular therapy and PD-1 checkpoint blockade , 2019, Journal of Experimental & Clinical Cancer Research.

[12]  Xiaoying Wang,et al.  Double-negative T cells remarkably promote neuroinflammation after ischemic stroke , 2019, Proceedings of the National Academy of Sciences.

[13]  L. Nguyen,et al.  Phase II clinical trial of adoptive cell therapy for patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and low-dose interleukin-2 , 2019, Cancer Immunology and Immunotherapy.

[14]  G. Linette,et al.  A Single Dose of Neoadjuvant PD-1 Blockade Predicts Clinical Outcomes in Resectable Melanoma , 2019, Nature Medicine.

[15]  M. Tsao,et al.  Human double negative T cells target lung cancer via ligand-dependent mechanisms that can be enhanced by IL-15 , 2019, Journal of Immunotherapy for Cancer.

[16]  Li Zhang,et al.  Developing Allogeneic Double-Negative T Cells as a Novel Off-the-Shelf Adoptive Cellular Therapy for Cancer , 2019, Clinical Cancer Research.

[17]  P. Wen,et al.  Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma , 2018, Nature Medicine.

[18]  I. Yaylim,et al.  Effect of trail C1595T variant and gene expression on the pathogenesis of non-small cell lung cancer , 2018, The Libyan journal of medicine.

[19]  Haibo Zhou,et al.  Antipancreatic cancer effect of DNT cells and the underlying mechanism. , 2019, Pancreatology : official journal of the International Association of Pancreatology (IAP) ... [et al.].

[20]  A. Rana,et al.  TGFβ Blockade Augments PD-1 Inhibition to Promote T-Cell–Mediated Regression of Pancreatic Cancer , 2018, Molecular Cancer Therapeutics.

[21]  Ralph Weissleder,et al.  Successful Anti‐PD‐1 Cancer Immunotherapy Requires T Cell‐Dendritic Cell Crosstalk Involving the Cytokines IFN‐&ggr; and IL‐12 , 2018, Immunity.

[22]  P. Micke,et al.  The Role of TGF-β Signaling in Lung Cancer Associated with Idiopathic Pulmonary Fibrosis , 2018, International journal of molecular sciences.

[23]  A. Broeks,et al.  Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma , 2018, Nature Medicine.

[24]  G. Hampton,et al.  Differential regulation of PD-L1 expression by immune and tumor cells in NSCLC and the response to treatment with atezolizumab (anti–PD-L1) , 2018, Proceedings of the National Academy of Sciences.

[25]  Heidi Ledford,et al.  Cancer immunologists scoop medicine Nobel prize , 2018, Nature.

[26]  Kai Liu,et al.  Ox40 regulates the conversion and suppressive function of double‐negative regulatory T cells , 2018, International immunopharmacology.

[27]  D. Ly,et al.  Infusion of ex‐vivo expanded human TCR‐αβ+ double‐negative regulatory T cells delays onset of xenogeneic graft‐versus‐host disease , 2018, Clinical and experimental immunology.

[28]  D. Torigian,et al.  Activity of Mesothelin-Specific Chimeric Antigen Receptor T Cells Against Pancreatic Carcinoma Metastases in a Phase 1 Trial. , 2018, Gastroenterology.

[29]  Y. Naito,et al.  Phase I clinical trial of adoptive transfer of expanded natural killer cells in combination with IgG1 antibody in patients with gastric or colorectal cancer , 2018, International journal of cancer.

[30]  Juan Wu,et al.  Identification of α‐fetoprotein‐specific T‐cell receptors for hepatocellular carcinoma immunotherapy , 2018, Hepatology.

[31]  J. Gartner,et al.  Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer , 2018, Nature Medicine.

[32]  B. Nelson,et al.  Publisher Correction: Oncolytic viruses as engineering platforms for combination immunotherapy , 2018, Nature Reviews Cancer.

[33]  M. Minden,et al.  Targeting chemotherapy-resistant leukemia by combining DNT cellular therapy with conventional chemotherapy , 2018, Journal of experimental & clinical cancer research : CR.

[34]  P. Morel,et al.  In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: A phase I clinical trial , 2018, Oncoimmunology.

[35]  Gen Sheng Wu,et al.  Developing TRAIL/TRAIL death receptor-based cancer therapies , 2018, Cancer and Metastasis Reviews.

[36]  Yuxiao Zhang,et al.  Cervical Cancer HeLa Cell Autocrine Apoptosis Induced by Coimmobilized IFN-γ plus TNF-α Biomaterials. , 2018, ACS applied materials & interfaces.

[37]  R. Bourgon,et al.  TGF-β attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells , 2018, Nature.

[38]  M. Manns,et al.  Perioperative, Spatiotemporally Coordinated Activation of T and NK Cells Prevents Recurrence of Pancreatic Cancer. , 2018, Cancer research.

[39]  J. Jansen,et al.  Decitabine enhances targeting of AML cells by CD34+ progenitor-derived NK cells in NOD/SCID/IL2Rgnull mice. , 2018, Blood.

[40]  Xi Yang,et al.  Fusion Proteins of NKG2D/NKG2DL in Cancer Immunotherapy , 2018, International journal of molecular sciences.

[41]  T. Kidokoro,et al.  Safety and persistence of WT1-specific T-cell receptor gene-transduced lymphocytes in patients with AML and MDS. , 2017, Blood.

[42]  J. Dick,et al.  Allogeneic Human Double Negative T Cells as a Novel Immunotherapy for Acute Myeloid Leukemia and Its Underlying Mechanisms , 2017, Clinical Cancer Research.

[43]  H. Oberg,et al.  The Ambiguous Role of γδ T Lymphocytes in Antitumor Immunity. , 2017, Trends in immunology.

[44]  J. Utikal,et al.  Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer , 2017, Nature.

[45]  Charles H. Yoon,et al.  An immunogenic personal neoantigen vaccine for patients with melanoma , 2017, Nature.

[46]  Jennifer Abbasí Amid FDA Approval Filings, Another CAR-T Therapy Patient Death. , 2017, JAMA.

[47]  Xianquan Zhang,et al.  Phase I Escalating-Dose Trial of CAR-T Therapy Targeting CEA+ Metastatic Colorectal Cancers. , 2017, Molecular therapy : the journal of the American Society of Gene Therapy.

[48]  S. Parmar,et al.  Phase I study of cord blood‐derived natural killer cells combined with autologous stem cell transplantation in multiple myeloma , 2017, British journal of haematology.

[49]  H. Zhang,et al.  Interferon-γ affects leukemia cell apoptosis through regulating Fas/FasL signaling pathway. , 2017, European review for medical and pharmacological sciences.

[50]  David M. Thomas,et al.  IFN-γ is required for cytotoxic T cell-dependent cancer genome immunoediting , 2017, Nature Communications.

[51]  S. Fox,et al.  Tumour-infiltrating lymphocytes in advanced HER2-positive breast cancer treated with pertuzumab or placebo in addition to trastuzumab and docetaxel: a retrospective analysis of the CLEOPATRA study. , 2017, The Lancet. Oncology.

[52]  J. Gartner,et al.  T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. , 2016, The New England journal of medicine.

[53]  J. Jia,et al.  Interleukin-2 Enhances the Regulatory Functions of CD4(+)T Cell-Derived CD4(-)CD8(-) Double Negative T Cells. , 2016, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research.

[54]  Hiroaki Ikeda,et al.  T-cell adoptive immunotherapy using tumor-infiltrating T cells and genetically engineered TCR-T cells. , 2016, International immunology.

[55]  Peter A. Fasching,et al.  Tumor-Infiltrating Lymphocytes: A Predictive and Prognostic Biomarker in Neoadjuvant-Treated HER2-Positive Breast Cancer , 2016, Clinical Cancer Research.

[56]  K. Curran,et al.  Toxicity and management in CAR T-cell therapy , 2016, Molecular therapy oncolytics.

[57]  H. You,et al.  Combination of double negative T cells and anti-thymocyte serum reverses type 1 diabetes in NOD mice , 2016, Journal of Translational Medicine.

[58]  Jiong Chen,et al.  DNT cell inhibits the growth of pancreatic carcinoma via abnormal expressions of NKG2D and MICA in vivo. , 2016, Biochemical and biophysical research communications.

[59]  K. Zhao,et al.  MicroRNA‐181a, a potential diagnosis marker, alleviates acute graft versus host disease by regulating IFN‐γ production , 2015, American journal of hematology.

[60]  M. Mathieu,et al.  Prognostic and predictive value of tumor-infiltrating lymphocytes in two phase III randomized adjuvant breast cancer trials. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[61]  Quan-shun Wang,et al.  Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

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

[63]  S. Hodge,et al.  Lung cancer is associated with decreased expression of perforin, granzyme B and interferon (IFN)‐γ by infiltrating lung tissue T cells, natural killer (NK) T‐like and NK cells , 2014, Clinical and experimental immunology.

[64]  P. Jia,et al.  MHC Class II Restricted Innate-Like Double Negative T Cells Contribute to Optimal Primary and Secondary Immunity to Leishmania major , 2014, PLoS pathogens.

[65]  M. Tsao,et al.  NKG2D regulates production of soluble TRAIL by ex vivo expanded human γδ T cells , 2013, European journal of immunology.

[66]  A. Scott,et al.  Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[67]  A. Santoni,et al.  Chemotherapy-elicited upregulation of NKG2D and DNAM-1 ligands as a therapeutic target in multiple myeloma , 2013, Oncoimmunology.

[68]  W. Lee,et al.  The effect of alloferon on the enhancement of NK cell cytotoxicity against cancer via the up-regulation of perforin/granzyme B secretion. , 2013, Immunobiology.

[69]  I. Mellman,et al.  Oncology meets immunology: the cancer-immunity cycle. , 2013, Immunity.

[70]  P. Calabresi,et al.  Helper T cells down-regulate CD4 expression upon chronic stimulation giving rise to double-negative T cells. , 2013, Cellular immunology.

[71]  S. Lesage,et al.  A comprehensive review of the phenotype and function of antigen-specific immunoregulatory double negative T cells. , 2013, Journal of autoimmunity.

[72]  A. Jevnikar,et al.  Double negative Treg cells promote nonmyeloablative bone marrow chimerism by inducing T‐cell clonal deletion and suppressing NK cell function , 2012, European journal of immunology.

[73]  D. Olive,et al.  Human Vγ9Vδ2 T Cells Specifically Recognize and Kill Acute Myeloid Leukemic Blasts , 2012, The Journal of Immunology.

[74]  A. Singer,et al.  Clonal deletion and the fate of autoreactive thymocytes that survive negative selection , 2012, Nature Immunology.

[75]  Li Zhang,et al.  Double negative regulatory T cells in transplantation and autoimmunity: recent progress and future directions. , 2012, Journal of molecular cell biology.

[76]  M. Minden,et al.  Anti-leukemia effect of ex vivo expanded DNT cells from AML patients: a potential novel autologous T-cell adoptive immunotherapy , 2011, Leukemia.

[77]  F. D’Acquisto,et al.  CD3+CD4-CD8- (double negative) T cells: saviours or villains of the immune response? , 2011, Biochemical pharmacology.

[78]  T. Waldmann,et al.  Safety (toxicity), pharmacokinetics, immunogenicity, and impact on elements of the normal immune system of recombinant human IL-15 in rhesus macaques. , 2011, Blood.

[79]  Erik Meulmeester,et al.  The dynamic roles of TGF‐β in cancer , 2011, The Journal of pathology.

[80]  Peter Chen,et al.  Chemotherapy-induced genotoxic stress promotes sensitivity to natural killer cell cytotoxicity by enabling missing-self recognition. , 2010, Cancer research.

[81]  C. Bona,et al.  Double Negative (CD3+4−8−) TCRαβ Splenic Cells from Young NOD Mice Provide Long-Lasting Protection against Type 1 Diabetes , 2010, PloS one.

[82]  L. Lanier,et al.  Effect of NKG2D ligand expression on host immune responses , 2010, Immunological reviews.

[83]  M. Colonna,et al.  DNAM-1/CD155 Interactions Promote Cytokine and NK Cell-Mediated Suppression of Poorly Immunogenic Melanoma Metastases , 2009, The Journal of Immunology.

[84]  G. Tsokos,et al.  Human TCR-αβ+ CD4− CD8− T Cells Can Derive from CD8+ T Cells and Display an Inflammatory Effector Phenotype1 , 2009, The Journal of Immunology.

[85]  R. Foà,et al.  ATM-ATR-dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype. , 2009, Blood.

[86]  M. Rehli,et al.  Characterization of MHC class-I restricted TCRαβ+ CD4− CD8− double negative T cells recognizing the gp100 antigen from a melanoma patient after gp100 vaccination , 2008, Cancer Immunology, Immunotherapy.

[87]  K. Gollob,et al.  Immunoregulatory mechanisms and CD4-CD8- (double negative) T cell subpopulations in human cutaneous leishmaniasis: a balancing act between protection and pathology. , 2008, International immunopharmacology.

[88]  G. Morgan,et al.  The requirement for DNAM-1, NKG2D, and NKp46 in the natural killer cell-mediated killing of myeloma cells. , 2007, Cancer research.

[89]  M. Eberl,et al.  Targeting human {gamma}delta} T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. , 2007, Cancer research.

[90]  Wei Yang,et al.  New differentiation pathway for double-negative regulatory T cells that regulates the magnitude of immune responses. , 2007, Blood.

[91]  H. Petrie,et al.  Zoned out: functional mapping of stromal signaling microenvironments in the thymus. , 2007, Annual review of immunology.

[92]  D. Kabelitz,et al.  Perspectives of gammadelta T cells in tumor immunology. , 2007, Cancer research.

[93]  Li Zhang,et al.  Double-Negative T Regulatory Cells Can Develop Outside the Thymus and Do Not Mature from CD8+ T Cell Precursors1 , 2006, The Journal of Immunology.

[94]  R. Oliveira,et al.  Disparate immunoregulatory potentials for double-negative (CD4- CD8-) alpha beta and gamma delta T cells from human patients with cutaneous leishmaniasis. , 2006, Infection and immunity.

[95]  Dick de Ridder,et al.  New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling , 2005, The Journal of experimental medicine.

[96]  Andreas Mackensen,et al.  Isolation and characterization of human antigen-specific TCR alpha beta+ CD4(-)CD8- double-negative regulatory T cells. , 2005, Blood.

[97]  Li Zhang,et al.  Antitumor activity mediated by double-negative T cells. , 2003, Cancer research.

[98]  H. Grey,et al.  Interactions between double positive thymocytes and high affinity ligands presented by cortical epithelial cells generate double negative thymocytes with T cell regulatory activity , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[99]  S. Miller,et al.  Epitope spreading in immune-mediated diseases: implications for immunotherapy , 2002, Nature Reviews Immunology.

[100]  Hergen Spits,et al.  Development of alphabeta T cells in the human thymus. , 2002, Nature reviews. Immunology.

[101]  J. Priatel,et al.  TCR/Self-Antigen Interactions Drive Double-Negative T Cell Peripheral Expansion and Differentiation into Suppressor Cells1 , 2001, The Journal of Immunology.

[102]  E. Wisse,et al.  Perforin and granzyme B induce apoptosis in FasL-resistant colon carcinoma cells , 2001, Cancer Immunology, Immunotherapy.

[103]  R. Negrin,et al.  Expansion of cytolytic CD8(+) natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon gamma production. , 2001, Blood.

[104]  H C Clevers,et al.  Transcriptional Control of T Lymphocyte Differentiation , 2001, Stem cells.

[105]  G. Levy,et al.  CD4(-)CD8(-) regulatory T cells implicated in preventing graft-versus-host and promoting graft-versus-leukemia responses. , 2001, Transplantation proceedings.

[106]  K. Young,et al.  CD3+CD4-CD8- alphabeta-TCR+ T cell as immune regulatory cell. , 2001, Journal of molecular medicine.

[107]  Li Zhang,et al.  Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression , 2000, Nature Medicine.

[108]  R. Budd,et al.  A model for the origin of TCR-alphabeta+ CD4-CD8- B220+ cells based on high affinity TCR signals. , 1999, Journal of immunology.

[109]  C. Rauch,et al.  Tumoricidal activity of tumor necrosis factor–related apoptosis–inducing ligand in vivo , 1999, Nature Medicine.

[110]  B. Fowlkes,et al.  Early T lymphocytes. Differentiation in vivo of adult intrathymic precursor cells , 1985, The Journal of experimental medicine.