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

Ann Taber, Emil Christensen, Philippe Lamy, Iver Nordentoft, Frederik Prip, Sia Viborg Lindskrog, Karin Birkenkamp-Demtröder, Trine Line Hauge Okholm, Michael Knudsen, Jakob Skou Pedersen, Torben Steiniche, Mads Agerbæk, Jørgen Bjerggaard Jensen, Lars Dyrskjøt, Ann Taber, Emil Christensen, Philippe Lamy, Iver Nordentoft, Frederik Prip, Sia Viborg Lindskrog, Karin Birkenkamp-Demtröder, Trine Line Hauge Okholm, Michael Knudsen, Jakob Skou Pedersen, Torben Steiniche, Mads Agerbæk, Jørgen Bjerggaard Jensen, Lars Dyrskjøt

Abstract

Overtreatment with cisplatin-based chemotherapy is a major issue in the management of muscle-invasive bladder cancer (MIBC), and currently none of the reported biomarkers for predicting response have been implemented in the clinic. Here we perform a comprehensive multi-omics analysis (genomics, transcriptomics, epigenomics and proteomics) of 300 MIBC patients treated with chemotherapy (neoadjuvant or first-line) to identify molecular changes associated with treatment response. DNA-based associations with response converge on genomic instability driven by a high number of chromosomal alterations, indels, signature 5 mutations and/or BRCA2 mutations. Expression data identifies the basal/squamous gene expression subtype to be associated with poor response. Immune cell infiltration and high PD-1 protein expression are associated with treatment response. Through integration of genomic and transcriptomic data, we demonstrate patient stratification to groups of low and high likelihood of cisplatin-based response. This could pave the way for future patient selection following validation in prospective clinical trials.

Conflict of interest statement

L.D. has sponsored research agreements with C2i genomics, AstraZeneca, Natera and Ferring, and has an advisory/consulting role at Ferring. J.B.J. is proctor for Intuitive Surgery, member of advisory board for Olympus Europe, Cephaid and Ferring, and has sponsored research agreements with Medac, Photocure ASA, Cephaid and Ferring. The following authors declare no competing interests: A.T., E.C., P.L., I.N., F.F.P., S.V.L., K.B., T.L.H.O., M.K., J.S.P., M.A., and T.S.

Figures

Fig. 1. Overview of the genomic alterations…
Fig. 1. Overview of the genomic alterations correlated to chemotherapy response.
a Oncoplot showing the significantly mutated genes in 165 tumors annotated by exome coverage, mutation load stratified by impact (as defined by SnpEff) and mutational signature deconvolution (top panels) and by clinical response, neoantigen load, number of damaging mutations in DDR genes, percentage of genome in allelic imbalance, expression subtypes, regulon cluster, RNA immune score, hypermethylation cluster and immune phenotype (bottom panel). be Violin plots showing the total number of SNVs, indels, neoantigens or the percentage of the genome in allelic imbalance compared to chemotherapy response. f Presence of damaging DDR mutations compared to chemotherapy response. gi Violin plots showing the total number of SNVs, indels or the percentage of the genome in allelic imbalance compared to the two main mutational subtypes. j Presence of DDR mutations compared to the two main mutational subtypes. k Mutational subtypes compared to chemotherapy response. All p-values were calculated using a Wilcoxon rank-sum test for continuous variables and a Fisher’s exact test for categorical variables. Source data are provided as a Source data file.
Fig. 2. BRCA2 mutated tumors are associated…
Fig. 2. BRCA2 mutated tumors are associated with mutations in an SBS5 context and response to chemotherapy.
a Number of mutations in an SBS5 context in relation to response to chemotherapy. b Mutation status of ERCC2 compared to chemotherapy response. c, d Volcano plots showing the difference between the median number of mutations for mutated tumors and the median number of mutations for wild-type tumors for all genes mutated in more than 5% of the patient cohort (only mutations with moderate- or high protein impact are considered). The red dashed lines indicate significance levels at p = 0.05. c Represents the number of mutations in an SBS5 context and d represents the number of mutations in an SBS2+13 (APOBEC) context. P-values were calculated using a permutation test (n = 100,000) that controls for mutation burden per sample and gene. e Mutation status of BRCA2 compared to chemotherapy response. f Kaplan–Meier survival analysis depicting the probability of overall survival stratified by BRCA2 mutation status. gj Based on TCGA data. g Number of mutations in an SBS5 context for ERCC2 and BRCA2 wild-type and mutant samples in TCGA data. h Number of mutations in an SBS2+13 (APOBEC) context for ERCC2 and BRCA2 wild-type and mutant samples in TCGA data. i, j Mutation status of ERCC2 and BRCA2 compared to response to chemotherapy in TCGA data. P-values were calculated using a Fisher’s exact test for categorical variables, a Wilcoxon rank-sum test for continuous variables and a log-rank test for comparing survival curves. For all boxplots, the center line represents the median, box hinges represent first and third quartiles, whiskers represent ±1.5 × interquartile range (IQR) and points represent outliers. Source data are provided as a Source data file.
Fig. 3. Delineation of metastatic evolution before…
Fig. 3. Delineation of metastatic evolution before chemotherapy.
All six patients were treated with cisplatin-based chemotherapy upon detection of metastatic disease. a Location of metastasis and the time to recurrence (mo = months). P = primary tumor, M = metastasis. The images were created using Biorender.com. b Clonal relationships between primary tumor samples and metastatic lesions depicted by phylogenetic trees. Trunk/branch lengths correspond to the number of SNVs. Mutations in genes involved in DDR, frequently mutated in TCGA or identified as drivers in BC (IntOGen) are indicated. Green = trunk, yellow = branch, blue = primary tumors, pink = metastatic lesions. c Variant allele frequencies for mutations identified in either of the available samples per patient. Identified mutations were subjected to read-counting in processed bam files to enable identification of mutations called in one sample, and present, but not called in another sample. The required read depth for identifying a given mutation was calculated for every position based on the lowest observed allele frequency. Only positions with sufficient read depth in all investigated samples were included. d Box plots depicting the observed allele frequencies for trunk and branch mutations. Asterisks indicate p-values below 2.2e−16. e Relative contribution of mutational signatures in the trunks (left circles) and branches (right circles). P-values were calculated using a Wilcoxon rank-sum test. For all boxplots, the center line represents the median, box hinges represent first and third quartiles, whiskers represent ±1.5 × interquartile range (IQR) and points represent outliers. Source data are provided as a Source data file.
Fig. 4. Gene expression characteristics and relation…
Fig. 4. Gene expression characteristics and relation to chemotherapy response.
a Visualisation of four identified consensus gene expression subtypes by selected gene sets. b Gene expression subtypes compared to chemotherapy response. c Kaplan–Meier survival analysis showing the probability of overall survival for patients with and without Ba/Sq gene expression subtype. d Number of mutations for gene expression subtypes. e Heatmap showing relative expression values for identified regulons and deconvoluted immune cells. f Regulon clusters compared to gene expression subtypes. g Regulon clusters in relation to response to chemotherapy. h Immune score across the identified gene expression subtypes. i Immune score compared to response to chemotherapy. j Summarized expression of the antigen-presenting machinery for immune hot (above median immune score) samples stratified by RECIST 1.1 response values. Only one tumor was classified as NE-like and was therefore omitted from this figure. Missing data is depicted in gray. P-values were calculated using a Fisher’s exact test for categorical variables, a Wilcoxon rank-sum test for continuous variables and a log-rank test for comparing survival curves. For all boxplots, the center line represents the median, box hinges represent first and third quartiles, whiskers represent ±1.5 × interquartile range (IQR) and points represent outliers. Source data are provided as a Source data file.
Fig. 5. DNA methylation subtypes based on…
Fig. 5. DNA methylation subtypes based on hypermethylated cancer-specific CpG sites.
a Clustering of samples based on hypermethylation events (n = 5000). Heatmap shows beta values and the right panel shows normal bladder and leukocyte beta values for comparison. b Methylation clusters compared to gene expression subtypes. c Gene set scores calculated using xCell and stratified by methylation clusters (HMC2: n = 17; HMC1: n = 31; HMC3: n = 16). d RECIST 1.1 response measurements stratified by methylation clusters. MEscore = Microenvironment score. P-values were calculated using a Wilcoxon rank-sum test. For all boxplots, the center line represents the median, box hinges represent first and third quartiles, whiskers represent ±1.5 × interquartile range (IQR) and points represent outliers. Source data are provided as a Source data file.
Fig. 6. Immune tumor microenvironment analysis by…
Fig. 6. Immune tumor microenvironment analysis by spatial proteomics.
Immunostaining performed on bladder cancer tissue microarrays from 184 patients. All protein measurements were performed once for each distinct sample. a Staining results shown for multiplex immunofluorescence (mIF) panel 1 and PD-1 with corresponding image analysis application (APP) of four tumors representing each immune subtype and high PD-1 expression, respectively. Red dashed lines divide tissue into intratumoral and peritumoral regions of interest. Scale bar: 20 µm. b Spatial organisation of immune cells and immune evasion mechanisms stratified by immune subtypes. Heatmap shows z-scores and the barplot the mean + SD immune scores (IS), cell percentages, or H-score [1 × (% cells low intensity) + 2 × (% cells moderate intensity) + 3 × (% cells high intensity)] for MHC-expression on carcinoma cells. Asterisks denote the barplot representing the H-score. Points represent the corresponding data points. c Immune subtypes compared to chemotherapy response. d Kaplan–Meier survival curves showing overall survival (OS) stratified by immune subtypes. eg Intratumoral and peritumoral fractions of immune evasion mechanisms (PD-1, PD-L1, MHC) compared to chemotherapy response. h Intratumoral combined PD-1/PD-L1 expression stratified by chemotherapy response. PD-1 high/PD-L1 low was compared to PD-1 high/PD-L1 high, PD-1 low/ PD-L1 low and PD-1 low/PD-L1 high combined. ik Relationship between immune evasion mechanisms and immune subtypes. Statistical significance was assessed using a chi-square test for categorical variables, a Wilcoxon rank-sum test for continuous variables and a log-rank test for comparing survival curves. For all boxplots, the center line represents the median, box hinges represent first and third quartiles, whiskers represent ±1.5 × interquartile range (IQR) and points represent outliers. Source data are provided as a Source data file.
Fig. 7. Integrative analysis.
Fig. 7. Integrative analysis.
a Chemotherapy response stratified by patient groups of high genomic instability (HGI) or low genomic instability (LGI). b Integration of genomic, transcriptomic and proteomic data for all patients with genomic data available and a gene expression subtype assigned (n = 121), stratified by high and low genomic instability. c Chemotherapy response for patients with Ba/Sq gene expression subtype and patients with non-Ba/Sq gene expression subtype (i.e., LumP, LumU, NE-like or Stroma-rich). d Overview of odds ratios (OR) calculated for molecular and clinical features for all patients (black), NAC (green) and first-line treated patients (yellow). Continuous variables were dichotomized based on the median and high vs. low is presented. Dots indicate odds ratios and horizontal lines show 95% confidence intervals (CI). Open lines indicate that the full range of the 95% CI is not shown. Asterisks denote p-values below 0.05. Currency signs denote p-values below 0.05 in a logistic regression model using continuous data. e Likelihood of cisplatin-based chemotherapy response stratified by genomic instability and gene expression subtypes. P-values were calculated using a chi-square test. Source data are provided as a Source data file.

References

    1. Antoni S, et al. Bladder cancer incidence and mortality: a global overview and recent trends. Eur. Urol. 2017;71:96–108.
    1. Bellmunt J, Petrylak DP. New therapeutic challenges in advanced bladder cancer. Semin. Oncol. 2012;39:598–607.
    1. Petrelli F, et al. Correlation of pathologic complete response with survival after neoadjuvant chemotherapy in bladder cancer treated with cystectomy: a meta-analysis. Eur. Urol. 2014;65:350–357.
    1. von der Maase H, et al. Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J. Clin. Oncol. 2005;23:4602–4608.
    1. Robertson AG, et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell. 2017;171:540–556.e25.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
    1. Teo MY, 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. 2017;23:3610–3618.
    1. Van Allen EM, et al. Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma. Cancer Discov. 2014;4:1140–1153.
    1. Plimack ER, et al. Defects in DNA repair genes predict response to neoadjuvant cisplatin-based chemotherapy in muscle-invasive bladder cancer. Eur. Urol. 2015;68:959–967.
    1. Li Q, et al. ERCC2 helicase domain mutations confer nucleotide excision repair deficiency and drive cisplatin sensitivity in muscle-invasive bladder cancer. Clin. Cancer Res. 2019;25:977–988.
    1. Groenendijk FH, et al. ERBB2 mutations characterize a subgroup of muscle-invasive bladder cancers with excellent response to neoadjuvant chemotherapy. Eur. Urol. 2016;69:384–388.
    1. Christensen, E. 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, 1547–1557 (2019).
    1. Choi W, et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell. 2014;25:152–165.
    1. Seiler R, et al. Impact of molecular subtypes in muscle-invasive bladder cancer on predicting response and survival after neoadjuvant chemotherapy. Eur. Urol. 2017;72:544–554.
    1. Kamoun, A. et al. A consensus molecular classification of muscle-invasive bladder cancer. Eur. Urol. 10.1016/j.eururo.2019.09.006 (2019).
    1. Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 2014;21:15–25.
    1. Krantz D, et al. Neoadjuvant chemotherapy reinforces antitumour T cell response in urothelial urinary bladder cancer. Eur. Urol. 2018;74:688–692.
    1. Adzhubei IA, et al. A method and server for predicting damaging missense mutations. Nat. Methods. 2010;7:248–249.
    1. Reva B, Antipin Y, Sander C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 2011;39:e118.
    1. Kim J, et al. Somatic ERCC2 mutations are associated with a distinct genomic signature in urothelial tumors. Nat. Genet. 2016;48:600–606.
    1. Cingolani P, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly. 2012;6:80–92.
    1. Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25:198–213.
    1. Şenbabaoğlu Y, et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 2016;17:231.
    1. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–330.
    1. Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567.
    1. Sawant A, et al. Differential role of base excision repair proteins in mediating cisplatin cytotoxicity. DNA Repair. 2017;51:46–59.
    1. Yang D, et al. Association of BRCA1 and BRCA2 mutations with survival, chemotherapy sensitivity, and gene mutator phenotype in patients with ovarian cancer. JAMA. 2011;306:1557–1565.
    1. Carlo, M. I. et al. Cancer susceptibility mutations in patients with urothelial malignancies. J. Clin. Oncol. 38, 406–414 (2019).
    1. Thomsen MBH, et al. Spatial and temporal clonal evolution during development of metastatic urothelial carcinoma. Mol. Oncol. 2016;10:1450–1460.
    1. Faltas BM, et al. Clonal evolution of chemotherapy-resistant urothelial carcinoma. Nat. Genet. 2016;48:1490–1499.
    1. Dong Y, Sun Q, Zhang X. PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget. 2017;8:2171–2186.
    1. Rosenblatt R, et al. Pathologic downstaging is a surrogate marker for efficacy and increased survival following neoadjuvant chemotherapy and radical cystectomy for muscle-invasive urothelial bladder cancer. Eur. Urol. 2012;61:1229–1238.
    1. Witjes, J. A. et al. Neuzillet. EAU Guidelines on muscle-invasive and metastatic bladder cancer. Eur. Urol. (2018).
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247.
    1. Winquist E, et al. Neoadjuvant chemotherapy for transitional cell carcinoma of the bladder: a systematic review and meta-analysis. J. Urol. 2004;171:561–569.
    1. Grossman HB, et al. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N. Engl. J. Med. 2003;349:859–866.
    1. Dash A, et al. A role for neoadjuvant gemcitabine plus cisplatin in muscle-invasive urothelial carcinoma of the bladder: a retrospective experience. Cancer. 2008;113:2471–2477.
    1. Harris PA, et al. The REDCap consortium: Building an international community of software platform partners. J. Biomed. Inform. 2019;95:103208.
    1. MuTect2 Pitfalls—Best Practices for Processing HTS Data 0.0 documentation. .
    1. Koboldt DC, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576.
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079.
    1. Shukla SA, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 2015;33:1152–1158.
    1. Bjerregaard A-M, Nielsen M, Hadrup SR, Szallasi Z, Eklund AC. MuPeXI: prediction of neo-epitopes from tumor sequencing data. Cancer Immunol. Immunother. 2017;66:1123–1130.
    1. Nielsen M, et al. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS ONE. 2007;2:e796.
    1. Van Loo P, et al. Allele-specific copy number analysis of tumors. Proc. Natl Acad. Sci. USA. 2010;107:16910–16915.
    1. Gehring JS, Fischer B, Lawrence M, Huber W. SomaticSignatures: inferring mutational signatures from single-nucleotide variants. Bioinformatics. 2015;31:3673–3675.
    1. Blokzijl F, Janssen R, van Boxtel R, Cuppen E. MutationalPatterns: comprehensive genome-wide analysis of mutational processes. Genome Med. 2018;10:33.
    1. Robertson AG, et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell. 2017;171:540–556.e25.
    1. Strona G, Nappo D, Boccacci F, Fattorini S, San-Miguel-Ayanz J. A fast and unbiased procedure to randomize ecological binary matrices with fixed row and column totals. Nat. Commun. 2014;5:4114.
    1. Niu B, et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics. 2014;30:1015–1016.
    1. Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods. 2017;14:417–419.
    1. Soneson C, Love MI, Robinson MD. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res. 2015;4:1521.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140.
    1. Castro MAA, et al. Regulators of genetic risk of breast cancer identified by integrative network analysis. Nat. Genet. 2016;48:12–21.
    1. Breyer J, et al. ESR1, ERBB2, and Ki67 mRNA expression predicts stage and grade of non-muscle-invasive bladder carcinoma (NMIBC) Virchows Arch. 2016;469:547–552.
    1. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18:220.
    1. Danaher P, et al. Gene expression markers of tumor infiltrating leukocytes. J. Immunother. Cancer. 2017;5:18.
    1. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science355, eaaf8399 (2017).
    1. Tian Y, et al. ChAMP: updated methylation analysis pipeline for Illumina BeadChips. Bioinformatics. 2017;33:3982–3984.
    1. Harris RA, et al. Genome-wide peripheral blood leukocyte DNA methylation microarrays identified a single association with inflammatory bowel diseases. Inflamm. Bowel Dis. 2012;18:2334–2341.
    1. Wilkerson MD, Neil Hayes D. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26:1572–1573.
    1. Gonzalez-Perez A, et al. IntOGen-mutations identifies cancer drivers across tumor types. Nat. Methods. 2013;10:1081–1082.

Source: PubMed

3
Subskrybuj