Unleashing the power of immune checkpoints: Post-translational modification of novel molecules and clinical applications.

[1]  Ni Hong,et al.  Sirpα on tumor-associated myeloid cells restrains antitumor immunity in colorectal cancer independent of its interaction with CD47. , 2024, Nature cancer.

[2]  N. Tang,et al.  SUMOylation inhibitors activate anti-tumor immunity by reshaping the immune microenvironment in a preclinical model of hepatocellular carcinoma. , 2023, Cellular oncology.

[3]  S. Gardai,et al.  Nonfucosylation of an anti-TIGIT antibody enhances FcγR engagement, driving innate immune activation and antitumor activity , 2023, Frontiers in immunology.

[4]  Z. Zeng,et al.  CD155 and its receptors in cancer immune escape and immunotherapy. , 2023, Cancer letters.

[5]  Xu Qian,et al.  EGFR‐Induced and c‐Src‐Mediated CD47 Phosphorylation Inhibits TRIM21‐Dependent Polyubiquitylation and Degradation of CD47 to Promote Tumor Immune Evasion , 2023, Advanced science.

[6]  Kun-Pin Wu,et al.  SheddomeDB 2023: A Revision of an Ectodomain Shedding Database Based on a Comprehensive Literature Review and Online Resources , 2023, Journal of proteome research.

[7]  M. Cippitelli,et al.  SUMOylation and related post-translational modifications in natural killer cell anti-cancer responses , 2023, Frontiers in Cell and Developmental Biology.

[8]  H. Tawbi,et al.  The introduction of LAG-3 checkpoint blockade in melanoma: immunotherapy landscape beyond PD-1 and CTLA-4 inhibition , 2023, Therapeutic advances in medical oncology.

[9]  Mingli Xiang,et al.  B7 family protein glycosylation: Promising novel targets in tumor treatment , 2022, Frontiers in Immunology.

[10]  Cheng Xu,et al.  Checkpoint Nano‐PROTACs for Activatable Cancer Photo‐Immunotherapy , 2022, Advanced materials.

[11]  J. Welsh,et al.  Inhibition of the CD47-SIRPα axis for cancer therapy: A systematic review and meta-analysis of emerging clinical data , 2022, Frontiers in Immunology.

[12]  Yajun Zhang,et al.  Small-Molecule PROTACs for Cancer Immunotherapy , 2022, Molecules.

[13]  Liang Chen,et al.  Strategies for developing PD-1 inhibitors and future directions. , 2022, Biochemical pharmacology.

[14]  M. Kiebish,et al.  The Next Frontier: Translational Development of Ubiquitination, SUMOylation, and NEDDylation in Cancer , 2022, International journal of molecular sciences.

[15]  Kristina Xega,et al.  The SUMOylation inhibitor subasumstat potentiates rituximab activity by IFN1-dependent macrophage and NK cell stimulation , 2022, Blood.

[16]  K. Wucherpfennig,et al.  MICA/B-targeted antibody promotes NK cell–driven tumor immunity in patients with intrahepatic cholangiocarcinoma , 2022, Oncoimmunology.

[17]  Xiaolong Yan,et al.  Emerging role of ubiquitination/deubiquitination modification of PD-1/PD-L1 in cancer immunotherapy , 2022, Genes & diseases.

[18]  J. Rip,et al.  Targeting pancreatic cancer by TAK-981: a SUMOylation inhibitor that activates the immune system and blocks cancer cell cycle progression in a preclinical model , 2022, Gut.

[19]  Anson Snow,et al.  The development of pevonedistat in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML): hope or hype? , 2022, Therapeutic advances in hematology.

[20]  C. Robert LAG-3 and PD-1 blockade raises the bar for melanoma , 2021, Nature Cancer.

[21]  A. Shibuya,et al.  DNAM-1 versus TIGIT: competitive roles in tumor immunity and inflammatory responses. , 2021, International immunology.

[22]  B. Helmink,et al.  Hallmarks of response, resistance, and toxicity to immune checkpoint blockade , 2021, Cell.

[23]  Zhen Bian,et al.  SIRPα sequesters SHP-2 to promote IL-4 and IL-13 signaling and the alternative activation of macrophages , 2021, Science Signaling.

[24]  J. Wolchok,et al.  Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways , 2021, Nature Reviews Clinical Oncology.

[25]  Steven S. Langston,et al.  A small-molecule SUMOylation inhibitor activates antitumor immune responses and potentiates immune therapies in preclinical models , 2021, Science Translational Medicine.

[26]  A. Santoni,et al.  Immunomodulatory effect of NEDD8-activating enzyme inhibition in Multiple Myeloma: upregulation of NKG2D ligands and sensitization to Natural Killer cell recognition , 2021, Cell Death & Disease.

[27]  Feng Xu,et al.  Poliovirus receptor (PVR)-like protein cosignaling network: new opportunities for cancer immunotherapy , 2021, Journal of experimental & clinical cancer research : CR.

[28]  K. Wucherpfennig,et al.  MICA/B antibody induces macrophage-mediated immunity against acute myeloid leukemia. , 2021, Blood.

[29]  M. Fuertes,et al.  Leveraging NKG2D Ligands in Immuno-Oncology , 2021, Frontiers in Immunology.

[30]  D. Wainwright,et al.  Tumor-derived NKG2D ligand sMIC reprograms NK cells to an inflammatory phenotype through CBM signalosome activation , 2021, Communications biology.

[31]  P. Trzonkowski,et al.  KIR Receptors as Key Regulators of NK Cells Activity in Health and Disease , 2021, Cells.

[32]  Y. Kikushige TIM‐3 in normal and malignant hematopoiesis: Structure, function, and signaling pathways , 2021, Cancer science.

[33]  J. Lohmueller,et al.  The costimulatory activity of Tim-3 requires Akt and MAPK signaling and its recruitment to the immune synapse , 2021, Science Signaling.

[34]  A. Regev,et al.  TIM-3 restrains anti-tumour immunity by regulating inflammasome activation , 2021, Nature.

[35]  Qian Wang,et al.  Generation of TIM3 inhibitory single-domain antibodies to boost the antitumor activity of chimeric antigen receptor T cells , 2021, Oncology letters.

[36]  T. Kaisho,et al.  The inhibitory receptor TIM-3 limits activation of the cGAS-STING pathway in intra-tumoral dendritic cells by suppressing extracellular DNA uptake. , 2021, Immunity.

[37]  F. Hodi,et al.  Phase I/Ib Clinical Trial of Sabatolimab, an Anti–TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti–PD-1 Antibody, in Advanced Solid Tumors , 2021, Clinical Cancer Research.

[38]  A. Regev,et al.  Tim-3 adaptor protein Bat3 is a molecular checkpoint of T cell terminal differentiation and exhaustion , 2021, Science Advances.

[39]  Y. Lou,et al.  Next generation of immune checkpoint inhibitors and beyond , 2021, Journal of Hematology & Oncology.

[40]  H. Hang,et al.  Protein S-palmitoylation in immunity , 2021, Open Biology.

[41]  N. Vélez de Mendizábal,et al.  Blocking TIM-3 in Treatment-refractory Advanced Solid Tumors: A Phase Ia/b Study of LY3321367 with or without an Anti-PD-L1 Antibody , 2021, Clinical Cancer Research.

[42]  Jiaxin Liu,et al.  Targeting the ubiquitination/deubiquitination process to regulate immune checkpoint pathways , 2021, Signal Transduction and Targeted Therapy.

[43]  Lucas Ferrari de Andrade,et al.  NKG2D and MICA/B shedding: a ‘tag game’ between NK cells and malignant cells , 2020, Clinical & translational immunology.

[44]  F. He,et al.  Neddylation of PTEN regulates its nuclear import and promotes tumor development , 2020, Cell Research.

[45]  Domarin Khago,et al.  The Role of Conformational Dynamics in the Recognition and Regulation of Ubiquitination , 2020, Molecules.

[46]  K. Venkatakrishnan,et al.  Phase I study assessing the mass balance, pharmacokinetics, and excretion of [14C]-pevonedistat, a NEDD8-activating enzyme inhibitor in patients with advanced solid tumors , 2020, Investigational New Drugs.

[47]  Yoshiki Narimatsu,et al.  Global view of human protein glycosylation pathways and functions , 2020, Nature Reviews Molecular Cell Biology.

[48]  G. Kristiansen,et al.  CD155 on Tumor Cells Drives Resistance to Immunotherapy by Inducing the Degradation of the Activating Receptor CD226 in CD8+ T Cells. , 2020, Immunity.

[49]  M. Otsuka,et al.  The fatty-acid amide hydrolase inhibitor URB597 inhibits MICA/B shedding , 2020, Scientific Reports.

[50]  I. Bahar,et al.  Pharmacological suppression of B7-H4 glycosylation restores antitumor immunity in immune-cold breast cancers. , 2020, Cancer discovery.

[51]  T. Okazaki,et al.  LAG-3: from molecular functions to clinical applications , 2020, Journal for ImmunoTherapy of Cancer.

[52]  G. Lajoie,et al.  ADAM protease inhibition overcomes resistance of breast cancer stem-like cells to γδ T cell immunotherapy , 2020, bioRxiv.

[53]  K. Madauss,et al.  Restoration of antitumor immunity through anti-MICA antibodies elicited with a chimeric protein , 2020, Journal for immunotherapy of cancer.

[54]  J. Pruneda,et al.  The Tumour Suppressor TMEM127 Is a Nedd4-Family E3 Ligase Adaptor Required by Salmonella SteD to Ubiquitinate and Degrade MHC Class II Molecules , 2020, Cell host & microbe.

[55]  T. Schumacher,et al.  The CD47-SIRPα Immune Checkpoint. , 2020, Immunity.

[56]  Q. Jiang,et al.  K6-linked SUMOylation of BAF regulates nuclear integrity and DNA replication in mammalian cells , 2020, Proceedings of the National Academy of Sciences.

[57]  Wenyi Wei,et al.  The role of ubiquitination in tumorigenesis and targeted drug discovery , 2020, Signal Transduction and Targeted Therapy.

[58]  Lin Sun,et al.  Recent Advances in Molecular Mechanisms of the NKG2D Pathway in Hepatocellular Carcinoma , 2020, Biomolecules.

[59]  B. Schulman,et al.  NEDD8 nucleates a multivalent cullin-RING-UBE2D ubiquitin ligation assembly , 2020, Nature.

[60]  Xianqun Fan,et al.  SUMOylation homeostasis in tumorigenesis. , 2019, Cancer letters.

[61]  Kongming Wu,et al.  Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4 , 2019, Molecular Cancer.

[62]  Jia-Yu Wang,et al.  B7-H4, a promising target for immunotherapy. , 2019, Cellular immunology.

[63]  S. M. Toor,et al.  Breast Cancer Cells and PD-1/PD-L1 Blockade Upregulate the Expression of PD-1, CTLA-4, TIM-3 and LAG-3 Immune Checkpoints in CD4+ T Cells , 2019, Vaccines.

[64]  A. Gartner,et al.  The Balance between Mono- and NEDD8-Chains Controlled by NEDP1 upon DNA Damage Is a Regulatory Module of the HSP70 ATPase Activity , 2019, Cell reports.

[65]  Qiang Zhang,et al.  Neddylation inhibition upregulates PD‐L1 expression and enhances the efficacy of immune checkpoint blockade in glioblastoma , 2019, International journal of cancer.

[66]  J. Mintern,et al.  MARCH ligases in immunity. , 2019, Current opinion in immunology.

[67]  H. Puhr,et al.  New emerging targets in cancer immunotherapy: the role of LAG3 , 2019, ESMO Open.

[68]  Jean-David Fumet,et al.  Tim-3/galectin-9 pathway and mMDSC control primary and secondary resistances to PD-1 blockade in lung cancer patients , 2019, Oncoimmunology.

[69]  Z. Zeng,et al.  Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape , 2019, Molecular Cancer.

[70]  G. Zhu,et al.  Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3 , 2019, Cell.

[71]  M. Hung,et al.  Posttranslational Modifications of PD-L1 and Their Applications in Cancer Therapy. , 2018, Cancer research.

[72]  L. Macdonald,et al.  Combination cancer immunotherapy targeting PD-1 and GITR can rescue CD8+ T cell dysfunction and maintain memory phenotype , 2018, Science Immunology.

[73]  S. Dixon,et al.  Protein palmitoylation and cancer , 2018, EMBO reports.

[74]  B. Walcheck,et al.  Anti-ADAM17 monoclonal antibody MEDI3622 increases IFNγ production by human NK cells in the presence of antibody-bound tumor cells , 2018, Cancer Immunology, Immunotherapy.

[75]  D. Faller,et al.  Phase Ib study of pevonedistat, a NEDD8-activating enzyme inhibitor, in combination with docetaxel, carboplatin and paclitaxel, or gemcitabine, in patients with advanced solid tumors , 2018, Investigational new drugs.

[76]  D. Faller,et al.  Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML. , 2018, Blood.

[77]  H. Nishiura,et al.  Inhibition of Asparagine-linked Glycosylation Participates in Hypoxia-induced Down-regulation of Cell-surface MICA Expression. , 2018, Anticancer research.

[78]  Geoff S. Higgins,et al.  Oxidative Phosphorylation as an Emerging Target in Cancer Therapy , 2018, Clinical Cancer Research.

[79]  B. Shen,et al.  Negative regulation of Nod‐like receptor protein 3 inflammasome activation by T cell Ig mucin‐3 protects against peritonitis , 2018, Immunology.

[80]  V. de Turris,et al.  Innate immune activating ligand SUMOylation affects tumor cell recognition by NK cells , 2017, Scientific Reports.

[81]  Ana Magalhães,et al.  Protein glycosylation in gastric and colorectal cancers: Toward cancer detection and targeted therapeutics. , 2017, Cancer letters.

[82]  R. Kumar,et al.  Phosphorylation: Implications in Cancer , 2017, The Protein Journal.

[83]  K. Odunsi,et al.  Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer , 2016, Oncoimmunology.

[84]  N. L. La Gruta,et al.  Ubiquitin ligase MARCH 8 cooperates with CD83 to control surface MHC II expression in thymic epithelium and CD4 T cell selection , 2016, The Journal of experimental medicine.

[85]  Jian-nan Feng,et al.  Tim-3 promotes tumor-promoting M2 macrophage polarization by binding to STAT1 and suppressing the STAT1-miR-155 signaling axis , 2016, Oncoimmunology.

[86]  G. Freeman,et al.  Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family. , 2016, Immunity.

[87]  Shohei Koyama,et al.  Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints , 2016, Nature Communications.

[88]  P. Lehner,et al.  Identifying the ERAD ubiquitin E3 ligases for viral and cellular targeting of MHC class I , 2015, Molecular immunology.

[89]  M. Colonna,et al.  DNAM-1 controls NK cell activation via an ITT-like motif , 2015, The Journal of experimental medicine.

[90]  A. Legat,et al.  Vaccination with LAG-3Ig (IMP321) and Peptides Induces Specific CD4 and CD8 T-Cell Responses in Metastatic Melanoma Patients—Report of a Phase I/IIa Clinical Trial , 2015, Clinical Cancer Research.

[91]  S. Pinho,et al.  Glycosylation in cancer: mechanisms and clinical implications , 2015, Nature Reviews Cancer.

[92]  Zihai Li,et al.  Nonblocking Monoclonal Antibody Targeting Soluble MIC Revamps Endogenous Innate and Adaptive Antitumor Responses and Eliminates Primary and Metastatic Tumors , 2015, Clinical Cancer Research.

[93]  M. Maris,et al.  Pevonedistat (MLN4924), a First‐in‐Class NEDD8‐activating enzyme inhibitor, in patients with acute myeloid leukaemia and myelodysplastic syndromes: a phase 1 study , 2015, British journal of haematology.

[94]  M. Weekes,et al.  Plasma Membrane Profiling Defines an Expanded Class of Cell Surface Proteins Selectively Targeted for Degradation by HCMV US2 in Cooperation with UL141 , 2015, PLoS pathogens.

[95]  M. Peter,et al.  Protein neddylation: beyond cullin–RING ligases , 2014, Nature Reviews Molecular Cell Biology.

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

[97]  Yibing Chen,et al.  UPR decreases CD226 ligand CD155 expression and sensitivity to NK cell‐mediated cytotoxicity in hepatoma cells , 2014, European journal of immunology.

[98]  Hidde L. Ploegh,et al.  CEACAM1 regulates TIM-3-mediated tolerance and exhaustion , 2014, Nature.

[99]  A. Santoni,et al.  c‐Cbl regulates MICA‐ but not ULBP2‐induced NKG2D down‐modulation in human NK cells , 2014, European journal of immunology.

[100]  P. Altevogt,et al.  Metalloprotease-mediated tumor cell shedding of B7-H6, the ligand of the natural killer cell-activating receptor NKp30. , 2014, Cancer research.

[101]  T. K. van den Berg,et al.  The interaction between signal regulatory protein alpha (SIRPα) and CD47: structure, function, and therapeutic target. , 2014, Annual review of immunology.

[102]  P. Sun,et al.  Natural Killer Cell-Mediated Shedding of ULBP2 , 2014, PloS one.

[103]  M. Weller,et al.  A disintegrin and metalloproteinases 10 and 17 modulate the immunogenicity of glioblastoma-initiating cells. , 2014, Neuro-oncology.

[104]  M. Ostrowski,et al.  T Cell Ig and Mucin Domain–Containing Protein 3 Is Recruited to the Immune Synapse, Disrupts Stable Synapse Formation, and Associates with Receptor Phosphatases , 2014, The Journal of Immunology.

[105]  J. Scheller,et al.  A Disintegrin and Metalloprotease (ADAM) 10 and ADAM17 Are Major Sheddases of T Cell Immunoglobulin and Mucin Domain 3 (Tim-3)* , 2013, The Journal of Biological Chemistry.

[106]  A. Santoni,et al.  Inhibition of Glycogen Synthase Kinase-3 Increases NKG2D Ligand MICA Expression and Sensitivity to NK Cell–Mediated Cytotoxicity in Multiple Myeloma Cells: Role of STAT3 , 2013, The Journal of Immunology.

[107]  S. Thorne,et al.  Potential for Enhanced Therapeutic Activity of Biological Cancer Therapies with Doxycycline Combination , 2012, Gene Therapy.

[108]  C. Li,et al.  Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells , 2012, Cell Death and Differentiation.

[109]  Sheng Xiao,et al.  Bat3 promotes T cell responses and autoimmunity by repressing Tim-3–mediated cell death and exhaustion , 2012, Nature Medicine.

[110]  H. Yoshiyama,et al.  Tumor-infiltrating DCs suppress nucleic acid–mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1 , 2012, Nature Immunology.

[111]  T. Kuroishi,et al.  Histamine reduces susceptibility to natural killer cells via down‐regulation of NKG2D ligands on human monocytic leukaemia THP‐1 cells , 2012, Immunology.

[112]  Michael Hagemann-Jensen,et al.  2-Deoxy d-Glucose Prevents Cell Surface Expression of NKG2D Ligands through Inhibition of N-Linked Glycosylation , 2012, The Journal of Immunology.

[113]  T. Spies,et al.  Expression of ERp5 and GRP78 on the membrane of chronic lymphocytic leukemia cells: association with soluble MICA shedding , 2012, Cancer Immunology, Immunotherapy.

[114]  Eric O Long,et al.  Palmitoylation of MICA, a ligand for NKG2D, mediates its recruitment to membrane microdomains and promotes its shedding , 2011, European journal of immunology.

[115]  Jenna M. Sullivan,et al.  Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity , 2010, The Journal of experimental medicine.

[116]  M. Gutiérrez,et al.  First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity , 2010, Journal of Translational Medicine.

[117]  A. Santoni,et al.  Heat Shock Protein-90 Inhibitors Increase MHC Class I-Related Chain A and B Ligand Expression on Multiple Myeloma Cells and Their Ability to Trigger NK Cell Degranulation1 , 2009, The Journal of Immunology.

[118]  B. Escudier,et al.  A Phase I Pharmacokinetic and Biological Correlative Study of IMP321, a Novel MHC Class II Agonist, in Patients with Advanced Renal Cell Carcinoma , 2009, Clinical Cancer Research.

[119]  Steven C Almo,et al.  T cell immunoglobulin mucin-3 crystal structure reveals a galectin-9-independent ligand-binding surface. , 2007, Immunity.

[120]  C. Blobel,et al.  Metalloproteases regulate T‐cell proliferation and effector function via LAG‐3 , 2007, The EMBO journal.

[121]  J. Bonventre,et al.  A highly conserved tyrosine of Tim-3 is phosphorylated upon stimulation by its ligand galectin-9. , 2006, Biochemical and biophysical research communications.

[122]  F. Triebel,et al.  A soluble LAG-3 protein as an immunopotentiator for therapeutic vaccines: Preclinical evaluation of IMP321. , 2006, Vaccine.

[123]  V. Kuchroo,et al.  The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity , 2005, Nature Immunology.

[124]  D. Voelker Protein and lipid motifs regulate phosphatidylserine traffic in yeast. , 2005, Biochemical Society transactions.

[125]  A. Shibuya,et al.  Requirement of the serine at residue 329 for lipid raft recruitment of DNAM-1 (CD226). , 2005, International immunology.

[126]  S. Wildt,et al.  The humanization of N-glycosylation pathways in yeast , 2005, Nature Reviews Microbiology.

[127]  D. Vignali,et al.  Biochemical Analysis of the Regulatory T Cell Protein Lymphocyte Activation Gene-3 (LAG-3; CD223)1 , 2004, The Journal of Immunology.

[128]  A. Nomoto,et al.  Ligand stimulation of CD155alpha inhibits cell adhesion and enhances cell migration in fibroblasts. , 2004, Biochemical and biophysical research communications.

[129]  H. Nakauchi,et al.  CD226 (DNAM-1) Is Involved in Lymphocyte Function–associated Antigen 1 Costimulatory Signal for Naive T Cell Differentiation and Proliferation , 2003, The Journal of experimental medicine.

[130]  H. Rammensee,et al.  Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. , 2003, Blood.

[131]  D. Vignali,et al.  The CD4‐related molecule, LAG‐3 (CD223), regulates the expansion of activated T cells , 2003, European journal of immunology.

[132]  D. Voelker,et al.  Phosphatidylserine Transport to the Mitochondria Is Regulated by Ubiquitination* , 2002, The Journal of Biological Chemistry.

[133]  D. Vignali,et al.  Cutting Edge: Molecular Analysis of the Negative Regulatory Function of Lymphocyte Activation Gene-31 , 2002, The Journal of Immunology.

[134]  T. Spies,et al.  Broad tumor-associated expression and recognition by tumor-derived γδ T cells of MICA and MICB , 1999 .

[135]  L. Caskey,et al.  Cleavage of the C-terminus of NEDD8 by UCH-L3. , 1998, Biochemical and biophysical research communications.

[136]  L. Lanier,et al.  Protein kinase C is involved in the regulation of both signaling and adhesion mediated by DNAX accessory molecule-1 receptor. , 1998, Journal of immunology.

[137]  E. Yeh,et al.  Characterization of NEDD8, a Developmentally Down-regulated Ubiquitin-like Protein* , 1997, The Journal of Biological Chemistry.

[138]  S. Roman-Roman,et al.  LAG-3, a novel lymphocyte activation gene closely related to CD4 , 1990, The Journal of experimental medicine.

[139]  Yiting Wang,et al.  Phosphorylation: A Fast Switch For Checkpoint Signaling. , 2020, Advances in experimental medicine and biology.

[140]  Haojie Lu,et al.  Palmitoylation as a Signal for Delivery. , 2020, Advances in experimental medicine and biology.

[141]  H. Clark,et al.  The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells , 2009, Nature Immunology.