2025
Sex-specific effects of exogenous asparagine on colorectal tumor growth, 17β-estradiol levels, and aromatase
Aladelokun O, Benitez K, Wang Y, Jain A, Berardi D, Maroun G, Shen X, Roper J, Gibson J, Sumigray K, Khan S, Johnson C. Sex-specific effects of exogenous asparagine on colorectal tumor growth, 17β-estradiol levels, and aromatase. Pharmacological Research 2025, 107736. PMID: 40228761, DOI: 10.1016/j.phrs.2025.107736.Peer-Reviewed Original ResearchTumor-specific survivalColorectal cancerTumor growthR2G2 miceIncreased serum estradiol levelsSerum estradiol levelsSub-populations of macrophagesAssociated with cancer prognosisSuppressed tumor growthColorectal tumor growthExogenous asparagineColorectal cancer developmentColorectal cancer cellsNegative feed-back effectEstradiol levelsGlutamate levelsSex-related differencesSex-specific effectsMale miceCancer prognosisAsparagine supplementationCancer progressionMiceDecreased numberTumorMIF as an oncogenic driver of low‐heterogeneity melanomas
Tran T, Sánchez‐Zuno G, Kulkarni R, Kluger H, Bucala R. MIF as an oncogenic driver of low‐heterogeneity melanomas. Molecular Oncology 2025, 19: 1295-1298. PMID: 40131169, PMCID: PMC12077282, DOI: 10.1002/1878-0261.70031.Peer-Reviewed Original ResearchConceptsMacrophage migration inhibitory factorImproving therapeutic outcomesMigration inhibitory factorPotential therapeutic utilityImmune landscapeMelanoma clonesImmune escapeT cellsImmunoregulatory cytokinesTumor heterogeneityTumor progressionOncogenic driversPathway inhibitorTherapeutic outcomesTumor evolutionInhibitory factorCell proliferationMelanomaTumorCellsAntagonistCytokinesThe Association of Gender in the Management and Prognosis of Vertebral and Sacral Chordoma: A SEER Analysis
Elsamadicy A, Sayeed S, Sherman J, Serrato P, Ghanekar S, Lo S, Sciubba D. The Association of Gender in the Management and Prognosis of Vertebral and Sacral Chordoma: A SEER Analysis. Journal Of Clinical Medicine 2025, 14: 1737. PMID: 40095791, PMCID: PMC11901273, DOI: 10.3390/jcm14051737.Peer-Reviewed Original ResearchFive-year survivalICD-O-3 codesNeo-adjuvant radiotherapyPredictors of survivalCancer Registry databaseRetrospective cohort studyAssociated with survivalICD-O-3SEER analysisTumor characteristicsSacral chordomaTumor sizeTumor locationMale patientsPatient demographicsSkull baseOsseous tumorsTreatment utilizationEnd ResultsRegistry databaseChordomaCohort studyTreatment characteristicsTumorPatientsDeterminants of response and molecular dynamics in HER2+ER+ breast cancers from the NA-PHER2 trial receiving HER2-targeted and endocrine therapies
Callari M, Dugo M, Barreca M, Győrffy B, Galbardi B, Vigano L, Locatelli A, Dall’Ara C, Ferrarini M, Bisagni G, Colleoni M, Mansutti M, Zamagni C, Del Mastro L, Zambelli S, Frassoldati A, Biasi O, Pusztai L, Valagussa P, Viale G, Gianni L, Bianchini G. Determinants of response and molecular dynamics in HER2+ER+ breast cancers from the NA-PHER2 trial receiving HER2-targeted and endocrine therapies. Nature Communications 2025, 16: 2195. PMID: 40038334, PMCID: PMC11880565, DOI: 10.1038/s41467-025-57293-9.Peer-Reviewed Original ResearchConceptsTumor-infiltrating lymphocytesBreast cancerEndocrine therapyImmune infiltrationPrediction of pCRAnti-HER2 therapyLuminal A phenotypeHigher immune infiltrationFemale breast cancerHER2 blockadeDeterminants of responseTumor responseInfiltrating lymphocytesCDK4/6 inhibitionAnti-HER2HER2 targetingClinical endpointsER signalingKi67Response groupCancerTherapyTherapeutic targetMolecular changesTumorT cell exhaustion: early or late in tumour progression?
Galluzzi L. T cell exhaustion: early or late in tumour progression? Nature Reviews Immunology 2025, 1-2. PMID: 40038448, DOI: 10.1038/s41577-025-01158-1.Peer-Reviewed Original ResearchThe development of an artificial intelligence auto-segmentation tool for 3D volumetric analysis of vestibular schwannomas
Jester N, Singh M, Lorr S, Tommasini S, Wiznia D, Buono F. The development of an artificial intelligence auto-segmentation tool for 3D volumetric analysis of vestibular schwannomas. Scientific Reports 2025, 15: 5918. PMID: 39966622, PMCID: PMC11836447, DOI: 10.1038/s41598-025-88589-x.Peer-Reviewed Original ResearchConceptsGround-truth datasetDice scoreVestibular schwannomaImage processing accuracyVolumetric analysisML-based algorithmsMeasuring tumor sizeMean dice scoreAuto-segmentation toolAccurate AIAI modelsTumor sizeTumor modelVS tumorsTumor growthTesting stageAI-LTumorImage processing softwareClinical practicePatient recruitmentProcessing softwareSchwannomaDatasetManual segmentationPilot study evaluating duplex sequencing of urinary DNA as a biomarker for recurrence in non-muscle invasive bladder cancer.
Ghali F, Blaha O, Lam H, Kennedy S, Wright J. Pilot study evaluating duplex sequencing of urinary DNA as a biomarker for recurrence in non-muscle invasive bladder cancer. Journal Of Clinical Oncology 2025, 43: 865-865. DOI: 10.1200/jco.2025.43.5_suppl.865.Peer-Reviewed Original ResearchNon-muscle invasive bladder cancerInvasive bladder cancerVariant allele fractionUrinary DNAIntravesical therapyBladder cancerPathogenic variantsTransurethral resection of bladder tumorMedian time to recurrenceResection of bladder tumorNext-generation sequencingDetection of recurrenceTime to recurrenceTime to relapseRecurrence of tumorBiomarkers of recurrencePilot studyTransurethral resectionBladder tumorsMedian ageRecurrenceAllele fractionUrine samplesTumorBiomarkersSacral tumors: a comprehensive review of imaging, diagnostic challenges, and tumor mimics
Adin M, Woolf G, Hegde R, Elsamadicy A, Mendel E, Zucconi W, Pucar D, Aygün N. Sacral tumors: a comprehensive review of imaging, diagnostic challenges, and tumor mimics. Skeletal Radiology 2025, 1-26. PMID: 39821683, DOI: 10.1007/s00256-024-04862-6.Peer-Reviewed Original ResearchPrimary tumorTumor mimicsImaging findingsAssessment of response to treatmentGerm cell tumorsEvaluation of systemic diseasesResponse to treatmentInsufficiency fracturesSacral massSecondary tumorsCell tumorsPretreatment evaluationDiagnostic challengeLymphoproliferative diseaseClinical detailsMalignant tumorsPrimary modalitySystemic diseaseEwing sarcomaTumorNotochordal remnantsDevelopmental entityEpidemiological factorsDiagnosisMetastasisSorafenib Alters Interstitial Proton and Sodium Levels in the Tumor Microenvironment: A 1H/23Na Spectroscopic Imaging Study
Khan M, Walsh J, Kurdi S, Mishra S, Mihailović J, Coman D, Hyder F. Sorafenib Alters Interstitial Proton and Sodium Levels in the Tumor Microenvironment: A 1H/23Na Spectroscopic Imaging Study. NMR In Biomedicine 2025, 38: e5319. PMID: 39764672, DOI: 10.1002/nbm.5319.Peer-Reviewed Original ResearchConceptsU87 tumorsSorafenib-treated tumorsUpregulated aerobic glycolysisSodium-potassium pumpInterstitial spaceTumor microenvironmentIntracellular NaTumor growthSpectroscopic imaging studiesTumor invasionGlioblastoma modelSodium levelsTumorGlioblastoma therapyImaging studiesPlaceboSorafenibMetabolic changesImmune functionCancer hallmarksAerobic glycolysisProliferative stateMeasure treatment effectsIonic changesProliferation rateChapter 14 Molecular biology, genetic, and epigenetics of kidney tumor
Kazemi R, Rezaeian A, Deyhimfar R, Taheri D. Chapter 14 Molecular biology, genetic, and epigenetics of kidney tumor. 2025, 209-221. DOI: 10.1016/b978-0-443-27302-5.00003-6.Peer-Reviewed Original ResearchDNA damage repair genesKidney cancer developmentRenal cell carcinomaTumor suppressor genePlanning therapeutic strategiesRenal tumorsUrothelial carcinomaWilms tumorCell carcinomaRenal sarcomaKidney tumorsClinical manifestationsUrological cancersKidney cancerPredisposing genesSuppressor geneTherapeutic strategiesCancer developmentTumorRepair genesCancerCarcinomaKidneyGenetic basisGenetic changesTrogocytosis-mediated immune evasion in the tumor microenvironment
Kim J, Park S, Kim J, Kim Y, Yoon H, Rayhan B, Jeong J, Bothwell A, Shin J. Trogocytosis-mediated immune evasion in the tumor microenvironment. Experimental & Molecular Medicine 2025, 57: 1-12. PMID: 39741180, PMCID: PMC11799389, DOI: 10.1038/s12276-024-01364-2.Peer-Reviewed Original ResearchConceptsCD4 T cellsT cellsMajor histocompatibility complexTumor microenvironmentImmune evasionMechanisms of immune evasionAnti-tumor immunityImmune regulatory moleculesAntigen-presenting cellsImmune-regulatory moleculesCTLA-4Cell-to-cell interactionsDonor cellsHistocompatibility complexTrogocytosisRecipient cellsTumorMembrane lossMembrane moleculesRegulatory moleculesMicroenvironmentSurface localizationPlasma membraneCellsTrogoptosisAdaptations of neutrophils in cancer
Ng M, Cerezo-Wallis D, Ng L, Hidalgo A. Adaptations of neutrophils in cancer. Immunity 2025, 58: 40-58. PMID: 39813993, DOI: 10.1016/j.immuni.2024.12.009.Peer-Reviewed Original Research
2024
High p16INK4A expression in glioblastoma is associated with senescence phenotype and better prognosis
Park S, Roh T, Tanaka Y, Kim Y, Park S, Kim T, Eom S, Park T, Park I, Kim S, Kim J. High p16INK4A expression in glioblastoma is associated with senescence phenotype and better prognosis. Neoplasia 2024, 60: 101116. PMID: 39724755, PMCID: PMC11729681, DOI: 10.1016/j.neo.2024.101116.Peer-Reviewed Original ResearchConceptsP16<sup>INK4a</sup> expressionImmune cell infiltrationTumor cellsCell infiltrationImmunologically active tumor microenvironmentInfiltration of T cellsActive tumor microenvironmentTERT promoter mutationsExtended overall survivalIsocitrate dehydrogenase (IDH)-wildtypeSecretion of chemokinesSenescent phenotypeMalignant brain tumorsIn vitro studiesEGFR amplificationOverall survivalTumor microenvironmentCDKN2A/2B deletionT cellsPrognostic markerImprove prognosisP16INK4a expressionPromoter mutationsTumorBrain tumorsA novel pharmacological entity toward integrated multimodal immunotherapy
Sirera R, Beltrán-Visiedo M, Galluzzi L. A novel pharmacological entity toward integrated multimodal immunotherapy. Trends In Pharmacological Sciences 2024, 46: 95-97. PMID: 39721827, DOI: 10.1016/j.tips.2024.12.001.Peer-Reviewed Original ResearchTertiary amine modification enables triterpene nanoparticles to target the mitochondria and treat glioblastoma via pyroptosis induction
Gao X, Tang X, Tu Z, Yu J, Bao Y, Long G, Sheu W, Wu H, Liu J, Zhou J. Tertiary amine modification enables triterpene nanoparticles to target the mitochondria and treat glioblastoma via pyroptosis induction. Biomaterials 2024, 317: 123035. PMID: 39731842, PMCID: PMC11827167, DOI: 10.1016/j.biomaterials.2024.123035.Peer-Reviewed Original ResearchConceptsSurvival of tumor-bearing miceBrain tumorsEffective treatmentPenetrate brain tumorsEffective treatment of glioblastomaTumor-bearing micePrimary brain tumorTreatment of glioblastomaMitochondria-targeted effectsTreating glioblastomaCancer treatmentGlioblastomaEffective killingTherapeutic targetHexokinase inhibitorGBM cellsPyroptosis inductionMitochondriaTumorGlycyrrhetinic acidTargeting effectTreatmentComparing the Accuracy and Reliability of ABC/2 and Planimetry for Vestibular Schwannoma Volume Assessment
Singh K, Abdou H, Panth N, Chiang V, Buono F, Schwartz N, Mahajan A. Comparing the Accuracy and Reliability of ABC/2 and Planimetry for Vestibular Schwannoma Volume Assessment. Otology & Neurotology 2024, 46: 196-200. PMID: 39792983, DOI: 10.1097/mao.0000000000004392.Peer-Reviewed Original ResearchConceptsTumor volume assessmentABC/2 methodVestibular schwannomaVolume assessmentPlanimetry methodTumor volumeTumor volume changesFollow-up scansOverestimate tumor volumePatients' quality of lifeTumor sizeRetrospective reviewIntracranial tumorsImprove clinical decision makingSubgroup analysisVolumetric assessmentTumorClinical decision makingQuality of lifePatients' qualityPatientsClinical settingPlanimetryImaging techniquesPositive correlationIdentification of HER2-positive breast cancer molecular subtypes with potential clinical implications in the ALTTO clinical trial
Rediti M, Venet D, Joaquin Garcia A, Maetens M, Vincent D, Majjaj S, El-Abed S, Di Cosimo S, Ueno T, Izquierdo M, Piccart M, Pusztai L, Loi S, Salgado R, Viale G, Rothé F, Sotiriou C. Identification of HER2-positive breast cancer molecular subtypes with potential clinical implications in the ALTTO clinical trial. Nature Communications 2024, 15: 10402. PMID: 39613746, PMCID: PMC11607438, DOI: 10.1038/s41467-024-54621-3.Peer-Reviewed Original ResearchMeSH KeywordsAntineoplastic Agents, ImmunologicalBiomarkers, TumorBreast NeoplasmsClinical Trials, Phase III as TopicFemaleGene Expression ProfilingGene Expression Regulation, NeoplasticHumansMiddle AgedNeoplasm Recurrence, LocalPrognosisRandomized Controlled Trials as TopicReceptor, ErbB-2TrastuzumabTumor MicroenvironmentConceptsHER2-positive breast cancerMolecular subtypesBreast cancerRate of pathological complete responseSensitive to HER2-targeted therapiesClinical trialsRisk of distant recurrenceBreast cancer molecular subtypesPathological complete responseHER2-targeted therapyCancer molecular subtypesPotential clinical implicationsNeoALTTO trialDistant recurrenceComplete responseAdjuvant trastuzumabPrognostic/predictive valueHeterogeneous biologySurvival outcomesI-SPY2Clinical outcomesMicroenvironment featuresGene expression profilesExternal cohortTumorCirculating tumor-reactive KIR+CD8+ T cells suppress anti-tumor immunity in patients with melanoma
Lu B, Lucca L, Lewis W, Wang J, Nogueira C, Heer S, Rayon-Estrada V, Axisa P, Reeves S, Buitrago-Pocasangre N, Pham G, Kojima M, Wei W, Aizenbud L, Bacchiocchi A, Zhang L, Walewski J, Chiang V, Olino K, Clune J, Halaban R, Kluger Y, Coyle A, Kisielow J, Obermair F, Kluger H, Hafler D. Circulating tumor-reactive KIR+CD8+ T cells suppress anti-tumor immunity in patients with melanoma. Nature Immunology 2024, 26: 82-91. PMID: 39609626, DOI: 10.1038/s41590-024-02023-4.Peer-Reviewed Original ResearchCD8+ T cellsAnti-tumor immunityRegulatory T cellsT cellsSubpopulation of CD8+ T cellsCytotoxic CD8+ T cellsHuman CD8+ T cellsTumor antigen-specific CD8Impaired anti-tumor immunityTumor antigen-specificPoor overall survivalTumor rejectionKIR expressionOverall survivalTumor antigensImmune evasionCellular mediatorsHuman cancersCD8MelanomaTumorTranscriptional programsFunctional heterogeneityImmunityPatientsConditional Activation of c-MYC in Distinct Catecholaminergic Cells Drives Development of Neuroblastoma or Somatostatinoma
Wang T, Liu L, Fang J, Jin H, Natarajan S, Sheppard H, Lu M, Turner G, Confer T, Johnson M, Steinberg J, Ha L, Yadak N, Jain R, Picketts D, Ma X, Murphy A, Davidoff A, Glazer E, Easton J, Chen X, Wang R, Yang J. Conditional Activation of c-MYC in Distinct Catecholaminergic Cells Drives Development of Neuroblastoma or Somatostatinoma. Cancer Research 2024, 85: 424-441. PMID: 39531507, PMCID: PMC11786959, DOI: 10.1158/0008-5472.can-24-1142.Peer-Reviewed Original ResearchPancreatic neuroendocrine tumorsC-mycCre recombinaseAnti-GD2 immunotherapyHigh-risk neuroblastomaDopamine B-hydroxylaseActivity of c-MycFDA-approved inhibitorC-Myc overexpressionC-Myc activityC-myc inductionTesting immunotherapyNeuroendocrine tumorsTargeted therapyTumor typesNeuroblastoma developmentNeuroblastoma tumorsNeuroblastoma oncogenesisEffective therapyTyrosine hydroxylaseTarget cellsTumorNeuroblastomaHuman neuroblastomaGenetic featuresTargeting T-Cell Costimulation to the Surface of Tumor Cells.
Eguren-Santamaría I, Sanmamed M, Molero-Glez P, Perez-Gracia J, Melero I. Targeting T-Cell Costimulation to the Surface of Tumor Cells. Clinical Cancer Research 2024, 31: 231-233. PMID: 39531541, DOI: 10.1158/1078-0432.ccr-24-3003.Peer-Reviewed Original ResearchT lymphocytesTumor cellsTargeting T-cell costimulationSurface of tumor cellsT cell costimulationProportion of patientsT cell activationAntitumor responseT cellsSolid tumorsSurface antigensActivator receptorSignal 2CostimulationTumorTarget specificityCellsCD137HER2Novel evidencePatientsAntigenReceptors
This site is protected by hCaptcha and its Privacy Policy and Terms of Service apply