Biological Stratification of Invasive and Advanced Urothelial Carcinoma

Ferlay J. Soerjomataram I. Dikshit R. et al.

Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012.

Int J Cancer. 136: E359-E386https://doi.org/10.1002/ijc.29210Jemal A. Bray F. Center M.M. et al.

Global cancer statistics.

CA Cancer J Clin. 61: 69-90https://doi.org/10.3322/caac.20107Shah J.B. McConkey D.J. Dinney C.P.

New strategies in muscle-invasive bladder cancer: on the road to personalized medicine.

Clin Cancer Res. 17: 2608-2612https://doi.org/10.1158/1078-0432.CCR-10-2770Grossman H.B. Natale R.B. Tangen C.M. et al.

Neoadjuvant Chemotherapy plus Cystectomy Compared with Cystectomy Alone for Locally Advanced Bladder Cancer.

New Engl J Med. 349: 859-866https://doi.org/10.1056/NEJMoa022148Witjes J.A. Bruins H.M. Cathomas R. et al.

European Association of Urology Guidelines on Muscle-invasive and Metastatic Bladder Cancer: Summary of the 2020 Guidelines.

Eur Urol. 79: 82-104https://doi.org/10.1016/j.eururo.2020.03.055Sylvester R.J. Rodriguez O. Hernandez V. et al.

European Association of Urology (EAU) Prognostic Factor Risk Groups for Non-muscle-invasive Bladder Cancer (NMIBC) Incorporating the WHO 2004/2016 and WHO 1973 Classification Systems for Grade: An Update from the EAU NMIBC Guidelines Panel.

Eur Urol. 79: 480-488https://doi.org/10.1016/j.eururo.2020.12.033Babjuk M. Burger M. Capoun O. et al.

European Association of Urology Guidelines on Non-muscle-invasive Bladder Cancer (Ta, T1, and Carcinoma in Situ).

Eur Urol. 81: 75-94https://doi.org/10.1016/j.eururo.2021.08.010Lindskrog S.V. Prip F. Lamy P. et al.

An integrated multi-omics analysis identifies prognostic molecular subtypes of non-muscle-invasive bladder cancer.

Nat Commun. 12: 2301https://doi.org/10.1038/s41467-021-22465-wSjodahl G. Lauss M. Lovgren K. et al.

A molecular taxonomy for urothelial carcinoma.

Clin Cancer Res. 18: 3377-3386https://doi.org/10.1158/1078-0432.CCR-12-0077-TSjodahl G. Eriksson P. Liedberg F. et al.

Molecular classification of urothelial carcinoma: global mRNA classification versus tumour-cell phenotype classification.

J Pathol. 242: 113-125https://doi.org/10.1002/path.4886Patschan O. Sjodahl G. Chebil G. et al.

A Molecular Pathologic Framework for Risk Stratification of Stage T1 Urothelial Carcinoma.

Eur Urol. 68 (): 824-832https://doi.org/10.1016/j.eururo.2015.02.021Hurst C.D. Alder O. Platt F.M. et al.

Genomic Subtypes of Non-invasive Bladder Cancer with Distinct Metabolic Profile and Female Gender Bias in KDM6A Mutation Frequency.

Cancer Cell. 32: 701-715 e7https://doi.org/10.1016/j.ccell.2017.08.005Hurst C.D. Cheng G. Platt F.M. et al.

Stage-stratified molecular profiling of non-muscle-invasive bladder cancer enhances biological, clinical, and therapeutic insight.

Cell Rep Med. 2https://doi.org/10.1016/j.xcrm.2021.100472Robertson A.G. Groeneveld C.S. Jordan B. et al.

Identification of Differential Tumor Subtypes of T1 Bladder Cancer.

Eur Urol. 78: 533-537https://doi.org/10.1016/j.eururo.2020.06.048Bellmunt J. Kim J. Reardon B. et al.

Genomic Predictors of Good Outcome, Recurrence, or Progression in High-Grade T1 Non-Muscle-Invasive Bladder Cancer.

Cancer Res. 80: 4476-4486https://doi.org/10.1158/0008-5472.CAN-20-0977Pietzak E.J. Bagrodia A. Cha E.K. et al.

Next-generation Sequencing of Nonmuscle Invasive Bladder Cancer Reveals Potential Biomarkers and Rational Therapeutic Targets.

Eur Urol. 72: 952-959https://doi.org/10.1016/j.eururo.2017.05.032Bacon J.V.W. Muller D.C. Ritch E. et al.

Somatic Features of Response and Relapse in Non-muscle-invasive Bladder Cancer Treated with Bacillus Calmette-Guerin Immunotherapy.

Eur Urol Oncol. https://doi.org/10.1016/j.euo.2021.11.002Balbas-Martinez C. Rodriguez-Pinilla M. Casanova A. et al.

ARID1A alterations are associated with FGFR3-wild type, poor-prognosis, urothelial bladder tumors.

PLoS One. 8: e62483https://doi.org/10.1371/journal.pone.0062483Robertson A.G. Kim J. Al-Ahmadie H. et al.

Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer.

Cell. 171: 540-556 e25https://doi.org/10.1016/j.cell.2017.09.007Liu D. Plimack E.R. Hoffman-Censits J. et al.

Clinical Validation of Chemotherapy Response Biomarker ERCC2 in Muscle-Invasive Urothelial Bladder Carcinoma.

JAMA Oncol. 2: 1094-1096https://doi.org/10.1001/jamaoncol.2016.1056Van Allen E.M. Mouw K.W. Kim P. et al.

Somatic ERCC2 Mutations Correlate with Cisplatin Sensitivity in Muscle-Invasive Urothelial Carcinoma.

Cancer Discov. 4: 1140-1153https://doi.org/10.1158/2159-8290.Cd-14-0623Pietzak E.J. Zabor E.C. Bagrodia A. et al.

Genomic Differences Between "Primary" and "Secondary" Muscle-invasive Bladder Cancer as a Basis for Disparate Outcomes to Cisplatin-based Neoadjuvant Chemotherapy.

Eur Urol. 75: 231-239https://doi.org/10.1016/j.eururo.2018.09.002Taber A. Prip F. Lamy P. et al.

Immune Contexture and Differentiation Features Predict Outcome in Bladder Cancer.

Eur Urol Oncol. 5: 203-213https://doi.org/10.1016/j.euo.2022.01.008Heide T. Maurer A. Eipel M. et al.

Multiregion human bladder cancer sequencing reveals tumour evolution, bladder cancer phenotypes and implications for targeted therapy.

J Pathol. 248: 230-242https://doi.org/10.1002/path.5250Sjodahl G. Eriksson P. Patschan O. et al.

Molecular changes during progression from nonmuscle invasive to advanced urothelial carcinoma.

Int J Cancer. 146: 2636-2647https://doi.org/10.1002/ijc.32737Mo Q. Nikolos F. Chen F. et al.

Prognostic Power of a Tumor Differentiation Gene Signature for Bladder Urothelial Carcinomas.

J Natl Cancer Inst. 110: 448-459https://doi.org/10.1093/jnci/djx243Damrauer J.S. Hoadley K.A. Chism D.D. et al.

Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology.

Proc Natl Acad Sci U S A. 111: 3110-3115https://doi.org/10.1073/pnas.1318376111Choi W. Porten S. Kim S. et al.

Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy.

Cancer Cell. 25: 152-165https://doi.org/10.1016/j.ccr.2014.01.009Rebouissou S. Bernard-Pierrot I. Reyniès Ad et al.

EGFR as a potential therapeutic target for a subset of muscle-invasive bladder cancers presenting a basal-like phenotype.

Sci Translational Med. 6: 244ra91https://doi.org/10.1126/scitranslmed.3008970Marzouka N.A. Eriksson P. Rovira C. et al.

A validation and extended description of the Lund taxonomy for urothelial carcinoma using the TCGA cohort.

Sci Rep. 8: 3737https://doi.org/10.1038/s41598-018-22126-xKamoun A. de Reynies A. Allory Y. et al.

A Consensus Molecular Classification of Muscle-invasive Bladder Cancer.

Eur Urol. 77: 420-433https://doi.org/10.1016/j.eururo.2019.09.006Batista da Costa J. Gibb E.A. Bivalacqua T.J. et al.

Molecular Characterization of Neuroendocrine-like Bladder Cancer.

Clin Cancer Res. 25: 3908-3920https://doi.org/10.1158/1078-0432.CCR-18-3558Seiler R. Ashab H.A.D. Erho N. et al.

Impact of Molecular Subtypes in Muscle-invasive Bladder Cancer on Predicting Response and Survival after Neoadjuvant Chemotherapy.

Eur Urol. 72: 544-554https://doi.org/10.1016/j.eururo.2017.03.030McConkey D.J. Choi W. Shen Y. et al.

A Prognostic Gene Expression Signature in the Molecular Classification of Chemotherapy-naive Urothelial Cancer is Predictive of Clinical Outcomes from Neoadjuvant Chemotherapy: A Phase 2 Trial of Dose-dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin with Bevacizumab in Urothelial Cancer.

Eur Urol. 69: 855-862https://doi.org/10.1016/j.eururo.2015.08.034Lotan Y. de Jong J.J. Liu V.Y.T. et al.

Patients with Muscle-Invasive Bladder Cancer with Nonluminal Subtype Derive Greatest Benefit from Platinum Based Neoadjuvant Chemotherapy.

J Urol. ()https://doi.org/10.1097/JU.0000000000002261Taber A. Christensen E. Lamy P. et al.

Molecular correlates of cisplatin-based chemotherapy response in muscle invasive bladder cancer by integrated multi-omics analysis.

Nat Commun. 11: 4858https://doi.org/10.1038/s41467-020-18640-0Roumiguie M. Contreras-Sanz A. Kumar G. et al.

Reconciling differences in impact of molecular subtyping on response to cisplatin-based chemotherapy.

Nat Commun. 12: 4833https://doi.org/10.1038/s41467-021-24837-8Sjödahl G. Abrahamsson J. Holmsten K. et al.

Different Responses to Neoadjuvant Chemotherapy in Urothelial Carcinoma Molecular Subtypes.

Eur Urol. https://doi.org/10.1016/j.eururo.2021.10.035

De Jong JJ, Gibb EA. Re: Gottfrid Sjödahl, Johan Abrahamsson, Karin Holmsten, et al. Different Responses to Neoadjuvant Chemotherapy in Urothelial Carcinoma Molecular Subtypes. Eur Urol. 2022;81:316-7.: Neoadjuvant Chemotherapy Response in Muscle-invasive Bladder Cancer: Differences in Intrinsic Biology or Subtyping Nomenclature? Eur Urol. Nov 12 2021;81(4):e90-e91. https://doi.org/10.1016/j.eururo.2021.12.033.

Flaig T.W. Tangen C.M. Daneshmand S. et al.

A Randomized Phase II Study of Coexpression Extrapolation (COXEN) with Neoadjuvant Chemotherapy for Bladder Cancer (SWOG S1314; NCT02177695).

Clin Cancer Res. 27: 2435-2441https://doi.org/10.1158/1078-0432.CCR-20-2409Teo M.Y. Bambury R.M. Zabor E.C. et al.

DNA Damage Response and Repair Gene Alterations Are Associated with Improved Survival in Patients with Platinum-Treated Advanced Urothelial Carcinoma.

Clin Cancer Res. 23: 3610-3618https://doi.org/10.1158/1078-0432.CCR-16-2520Li Q. Damish A.W. Frazier Z. et al.

ERCC2 Helicase Domain Mutations Confer Nucleotide Excision Repair Deficiency and Drive Cisplatin Sensitivity in Muscle-Invasive Bladder Cancer.

Clin Cancer Res. 25: 977-988https://doi.org/10.1158/1078-0432.CCR-18-1001Plimack E.R. Dunbrack R.L. Brennan T.A. et al.

Defects in DNA Repair Genes Predict Response to Neoadjuvant Cisplatin-based Chemotherapy in Muscle-invasive Bladder Cancer.

Eur Urol. 68: 959-967https://doi.org/10.1016/j.eururo.2015.07.009Miron B. Hoffman-Censits J.H. Anari F. et al.

Defects in DNA Repair Genes Confer Improved Long-term Survival after Cisplatin-based Neoadjuvant Chemotherapy for Muscle-invasive Bladder Cancer.

Eur Urol Oncol. 3: 544-547https://doi.org/10.1016/j.euo.2020.02.003Geynisman D.M. Abbosh P. Ross E.A. et al.

A phase II trial of risk enabled therapy after initiating neoadjuvant chemotherapy for bladder cancer (RETAIN BLADDER): Interim analysis.

J Clin Oncol. 39: 397https://doi.org/10.1200/JCO.2021.39.6_suppl.397Galsky M.D. Daneshmand S. Chan K.G. et al.

Phase 2 trial of gemcitabine, cisplatin, plus nivolumab with selective bladder sparing in patients with muscle- invasive bladder cancer (MIBC): HCRN GU 16-257.

J Clin Oncol. 39: 4503https://doi.org/10.1200/JCO.2021.39.15_suppl.4503Efstathiou J.A. Mouw K.W. Gibb E.A. et al.

Impact of Immune and Stromal Infiltration on Outcomes Following Bladder-Sparing Trimodality Therapy for Muscle-Invasive Bladder Cancer.

Eur Urol. 76: 59-68https://doi.org/10.1016/j.eururo.2019.01.011Sharma P. Retz M. Siefker-Radtke A. et al.

Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial.

Lancet Oncol. 18: 312-322https://doi.org/10.1016/s1470-2045(17)30065-7Rosenberg J.E. Hoffman-Censits J. Powles T. et al.

Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial.

Lancet. 387: 1909-1920https://doi.org/10.1016/s0140-6736(16)00561-4Mariathasan S. Turley S.J. Nickles D. et al.

TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells.

Nature. 554: 544-548https://doi.org/10.1038/nature25501Szabados B. Kockx M. Assaf Z.J. et al.

Final Results of Neoadjuvant Atezolizumab in Cisplatin-ineligible Patients with Muscle-invasive Urothelial Cancer of the Bladder.

Eur Urol. https://doi.org/10.1016/j.eururo.2022.04.013Powles T. Kockx M. Rodriguez-Vida A. et al.

Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial.

Nat Med. 25: 1706-1714https://doi.org/10.1038/s41591-019-0628-7Kim J. Kwiatkowski D. McConkey D.J. et al.

The Cancer Genome Atlas Expression Subtypes Stratify Response to Checkpoint Inhibition in Advanced Urothelial Cancer and Identify a Subset of Patients with High Survival Probability.

Eur Urol. 75: 961-964https://doi.org/10.1016/j.eururo.2019.02.017Necchi A. Raggi D. Gallina A. et al.

Impact of Molecular Subtyping and Immune Infiltration on Pathological Response and Outcome Following Neoadjuvant Pembrolizumab in Muscle-invasive Bladder Cancer.

Eur Urol. https://doi.org/10.1016/j.eururo.2020.02.028Nakauma-González J.A. Rijnders M. van Riet J. et al.

Comprehensive Molecular Characterization Reveals Genomic and Transcriptomic Subtypes of Metastatic Urothelial Carcinoma.

Eur Urol. https://doi.org/10.1016/j.eururo.2022.01.026Galsky M.D. Saci A. Szabo P.M. et al.

Nivolumab in Patients with Advanced Platinum-resistant Urothelial Carcinoma: Efficacy, Safety, and Biomarker Analyses with Extended Follow-up from CheckMate 275.

Clin Cancer Res. 26: 5120-5128https://doi.org/10.1158/1078-0432.CCR-19-4162Powles T. Assaf Z.J. Davarpanah N. et al.

ctDNA guiding adjuvant immunotherapy in urothelial carcinoma.

Nature. 595: 432-437https://doi.org/10.1038/s41586-021-03642-9Sjodahl G. Eriksson P. Lovgren K. et al.

Discordant molecular subtype classification in the basal-squamous subtype of bladder tumors and matched lymph-node metastases.

Mod Pathol. 31: 1869-1881https://doi.org/10.1038/s41379-018-0096-5Vandekerkhove G. Lavoie J.M. Annala M. et al.

Plasma ctDNA is a tumor tissue surrogate and enables clinical-genomic stratification of metastatic bladder cancer.

Nat Commun. 12: 184https://doi.org/10.1038/s41467-020-20493-6Powles T. Durán I. van der Heijden M.S. et al.

Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial.

Lancet. 391: 748-757https://doi.org/10.1016/s0140-6736(17)33297-xSnyder A. Nathanson T. Funt S.A. et al.

Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis.

Plos Med. 14e1002309https://doi.org/10.1371/journal.pmed.1002309Balar A.V. Castellano D. O'Donnell P.H. et al.

First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study.

Lancet Oncol. 18: 1483-1492https://doi.org/10.1016/s1470-2045(17)30616-2Bellmunt J. de Wit R. Vaughn D.J. et al.

Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma.

N Engl J Med. 376: 1015-1026https://doi.org/10.1056/NEJMoa1613683Powles T. Csőszi T. Özgüroğlu M. et al.

Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): a randomised, open-label, phase 3 trial.

Lancet Oncol. 22: 931-945https://doi.org/10.1016/s1470-2045(21)00152-2da Costa J.B. Gibb E.A. Nykopp T.K. et al.

Molecular tumor heterogeneity in muscle invasive bladder cancer: Biomarkers, subtypes, and implications for therapy.

Urol Oncol. https://doi.org/10.1016/j.urolonc.2018.11.015Faltas B.M. Prandi D. Tagawa S.T. et al.

Clonal evolution of chemotherapy-resistant urothelial carcinoma.

Nat Genet. 48: 1490-1499https://doi.org/10.1038/ng.3692Thomsen M.B. Nordentoft I. Lamy P. et al.

Spatial and temporal clonal evolution during development of metastatic urothelial carcinoma.

Mol Oncol. 10: 1450-1460https://doi.org/10.1016/j.molonc.2016.08.003Thomsen M.B.H. Nordentoft I. Lamy P. et al.

Comprehensive multiregional analysis of molecular heterogeneity in bladder cancer.

Sci Rep. 711702https://doi.org/10.1038/s41598-017-11291-0Bettegowda C. Sausen M. Leary R.J. et al.

Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies.

Sci Translational Med. 6: 224ra24https://doi.org/10.1126/scitranslmed.3007094Birkenkamp-Demtroder K. Nordentoft I. Christensen E. et al.

Genomic Alterations in Liquid Biopsies from Patients with Bladder Cancer.

Eur Urol. 70: 75-82https://doi.org/10.1016/j.eururo.2016.01.007Vandekerkhove G. Todenhofer T. Annala M. et al.

Circulating Tumor DNA Reveals Clinically Actionable Somatic Genome of Metastatic Bladder Cancer.

Clin Cancer Res. 23: 6487-6497https://doi.org/10.1158/1078-0432.CCR-17-1140Christensen E. Birkenkamp-Demtroder K. Sethi H. et al.

Early Detection of Metastatic Relapse and Monitoring of Therapeutic Efficacy by Ultra-Deep Sequencing of Plasma Cell-Free DNA in Patients With Urothelial Bladder Carcinoma.

J Clin Oncol. 37: 1547-1557https://doi.org/10.1200/JCO.18.02052Raja R. Kuziora M. Brohawn P.Z. et al.

Early Reduction in ctDNA Predicts Survival in Patients with Lung and Bladder Cancer Treated with Durvalumab.

Clin Cancer Res. 24: 6212-6222https://doi.org/10.1158/1078-0432.CCR-18-0386Loriot Y. Necchi A. Park S.H. et al.

Erdafitinib in Locally Advanced or Metastatic Urothelial Carcinoma.

N Engl J Med. 381: 338-348https://doi.org/10.1056/NEJMoa1817323Siefker-Radtke A.O. Loriot Y. Necchi A. et al.

751P Analysis of circulating tumor DNA (ctDNA) from the phase II BLC2001 trial of erdafitinib in locally advanced or metastatic urothelial carcinoma (mUC) to identify markers of intrinsic resistance to fibroblast growth factor receptor (FGFR)-targeted therapy.

Ann Oncol. : 31doihttps://doi.org/10.1016/j.annonc.2020.08.823Crowley E. Di Nicolantonio F. Loupakis F. et al.

Liquid biopsy: monitoring cancer-genetics in the blood.

Nat Rev Clin Oncol. 10: 472-484https://doi.org/10.1038/nrclinonc.2013.110Teo M.Y. Mota J.M. Whiting K.A. et al.

Fibroblast Growth Factor Receptor 3 Alteration Status is Associated with Differential Sensitivity to Platinum-based Chemotherapy in Locally Advanced and Metastatic Urothelial Carcinoma.

Eur Urol. 78: 907-915https://doi.org/10.1016/j.eururo.2020.07.018Turo R. Harnden P. Thygesen H. et al.

FGFR3 expression in primary invasive bladder cancers and matched lymph node metastases.

J Urol. 193: 325-330https://doi.org/10.1016/j.juro.2014.06.026Tully K.H. Jutte H. Wirtz R.M. et al.

Prognostic Role of FGFR Alterations and FGFR mRNA Expression in Metastatic Urothelial Cancer Undergoing Checkpoint Inhibitor Therapy.

Urology. 157: 93-101https://doi.org/10.1016/j.urology.2021.05.055Wang L. Gong Y. Saci A. et al.

Fibroblast Growth Factor Receptor 3 Alterations and Response to PD-1/PD-L1 Blockade in Patients with Metastatic Urothelial Cancer.

Eur Urol. 76: 599-603https://doi.org/10.1016/j.eururo.2019.06.025Powles T. Sridhar S.S. Loriot Y. et al.

Avelumab maintenance in advanced urothelial carcinoma: biomarker analysis of the phase 3 JAVELIN Bladder 100 trial.

Nat Med. 27: 2200-2211https://doi.org/10.1038/s41591-021-01579-0

留言 (0)

沒有登入
gif