2023
Clinical relevance of PD-1 positive CD8 T-cells in gastric cancer
Choo J, Kua L, Soe M, Asuncion B, Tan B, Teo C, Tay R, So J, Shabbir A, Guowei K, Tan H, Chan G, Ma H, Ramachandran G, Lum J, Chee C, Sridharan S, Tan P, Sundar R, Yong W. Clinical relevance of PD-1 positive CD8 T-cells in gastric cancer. Gastric Cancer 2023, 26: 393-404. PMID: 36781556, PMCID: PMC10115710, DOI: 10.1007/s10120-023-01364-7.Peer-Reviewed Original ResearchMeSH KeywordsB7-H1 AntigenCD8-Positive T-LymphocytesClinical RelevanceGranzymesHumansKi-67 AntigenLymphocytes, Tumor-InfiltratingPrognosisProgrammed Cell Death 1 ReceptorStomach NeoplasmsTumor MicroenvironmentConceptsCD8 T cellsPD-1+CD8+ T cellsT cellsTumor microenvironmentPD-1Overall survivalGastric cancerMultiplex immunohistochemistryT cell-inflamed tumor microenvironmentAssociated with improved OSIncreased PD-1 expressionClinical relevanceGranzyme-B expressionInflamed tumor microenvironmentPD-1 expressionInfluence overall survivalNK cell proportionTreated with immunotherapyPhase 2 trialAssociated with chemotherapyCox proportional hazards modelsProportional hazards modelImproved OSImmunotherapy sensitivityNK cells
2019
Epigenomic promoter alterations predict for benefit from immune checkpoint inhibition in metastatic gastric cancer
Sundar R, Huang K, Qamra A, Kim K, Kim S, Kang W, Tan A, Lee J, Tan P. Epigenomic promoter alterations predict for benefit from immune checkpoint inhibition in metastatic gastric cancer. Annals Of Oncology 2019, 30: 424-430. PMID: 30624548, PMCID: PMC6442650, DOI: 10.1093/annonc/mdy550.Peer-Reviewed Original ResearchConceptsImmune checkpoint inhibitionMetastatic gastric cancerT cell cytolytic activityResistance to immune checkpoint inhibitionCheckpoint inhibitionGastric cancerCytolytic activityImmune evasionMedian progression-free survivalPhase II clinical trial of patientsCancer treated with immunotherapyClinical trials of patientsMechanisms of immune evasionResponse rateTreated with pembrolizumabPhase II clinical trialProgression-free survivalFresh tumor biopsiesPost-treatment biopsiesCohort of patientsTrial of patientsEarly gastric cancerArchival tissue samplesClinical responseTumor biopsies
2018
Transcriptional analysis of immune genes in Epstein–Barr virus-associated gastric cancer and association with clinical outcomes
Sundar R, Qamra A, Tan A, Zhang S, Ng C, Teh B, Lee J, Kim K, Tan P. Transcriptional analysis of immune genes in Epstein–Barr virus-associated gastric cancer and association with clinical outcomes. Gastric Cancer 2018, 21: 1064-1070. PMID: 29915957, DOI: 10.1007/s10120-018-0851-9.Peer-Reviewed Original ResearchMeSH KeywordsAdultAgedB7-H1 AntigenBiomarkers, TumorCD8 AntigensCohort StudiesDisease-Free SurvivalEpstein-Barr Virus InfectionsFemaleGene Expression ProfilingGene Expression Regulation, NeoplasticHerpesvirus 4, HumanHumansImmunohistochemistryLymphocytes, Tumor-InfiltratingMaleMiddle AgedPerforinProgrammed Cell Death 1 ReceptorStomach NeoplasmsConceptsPD-L1PD-L1highPD-L1lowEBVaGC samplesImmune-related genesAssociated with worse disease-free survivalGastric cancerPD-L1 immunohistochemistry expressionPD-L1 transcript expressionExpression of PD-L1Levels of PD-L1Epstein-Barr virus-associated gastric cancerTumour-infiltrating lymphocyte patternPD-L1 expressionPD-L1 immunohistochemistryPrimary tumor resectionDisease-free survivalAssociated with higher expressionCases of EBVaGCGastric cancer patientsEBV-negative samplesSamsung Medical CentreAsian Cancer Research GroupImmune genesCancer Research Group
2017
Combining DNA damaging therapeutics with immunotherapy: more haste, less speed
Brown J, Sundar R, Lopez J. Combining DNA damaging therapeutics with immunotherapy: more haste, less speed. British Journal Of Cancer 2017, 118: 312-324. PMID: 29123260, PMCID: PMC5808021, DOI: 10.1038/bjc.2017.376.Peer-Reviewed Original ResearchConceptsImmunogenic cell deathDNA-damaging therapeuticsPromote immunogenic cell deathPhase I-III trialsDurable response rateDNA-damaging agentsAntitumour immune responseRegimens to patientsProperties of malignant cellsChoice of combinationsCell deathAntitumour immunitySequence of agentsNeoantigen productionTumor microenvironmentMalignant cellsNeoantigen repertoireInflammatory milieuPreclinical workImmune cellsCombination trialsDamaging agentsImmunological memoryMinimal toxicityHypothesis-driven trial