Evaluation of PD-L1 and other immune markers in bladder urothelial carcinoma stratified by histologic variants and molecular subtypes

Huili Li, Qingzhao Zhang, Lauren Shuman, Matthew Kaag, Jay D Raman, Suzanne Merrill, David J DeGraff, Joshua I Warrick, Guoli Chen, Huili Li, Qingzhao Zhang, Lauren Shuman, Matthew Kaag, Jay D Raman, Suzanne Merrill, David J DeGraff, Joshua I Warrick, Guoli Chen

Abstract

Although advanced bladder cancer overall has a poor prognosis, a subset of patients demonstrate durable response to immune checkpoint inhibitors. Evidence shows that the response to checkpoint inhibitors may be associated with type and degree of immune infiltration in the tumor microenvironment. Here, we evaluated immune markers stratified by molecular subtypes and histologic variants. The study utilized a series of urothelial carcinomas (UCs) by tissue microarray, on which histologic variants and molecular subtypes had previously been established. PD1, CD3, CD8 and CD68 expression was evaluated by immunohistochemistry in tumor infiltrating immune cells, while PD-L1 expression in the tumor microenvironment was assessed. Each marker was scored semi-quantitatively (score 0-3). Tumors were clustered by marker scores using agglomerative methods, and associations among markers, histologies, and molecular subtypes were analyzed. PD-L1 expression in the tumor microenvironment significantly correlated with presence of CD3, CD8 and chronic inflammation. Urothelial carcinoma may be classified as either immune high or low based on marker expression. The immune high group is enriched in higher CD3, PD-L1, and genomically-unstable molecular subtype, suggesting it may respond to checkpoint inhibitors. We also identified a degree of intratumoral heterogeneity in immune markers in bladder cancer.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Immune marker score analysis in association with histological variants and molecular subtypes. (a) Unsupervised hierarchical clustering of all UC cases. Each row is a marker and each column is a patient. Top bar indicates histological variants and molecular subtypes. (b) Distribution of histological variants (CIS, NIPUC, and invasive UC) and immune high and immune low clusters (Chi-square test, p < 0.01); (c) NIPUC is significantly associated with immune low cluster (Fisher’s exact test, p < 0.01); (d) Invasive UC is significantly associated with immune high cluster (Fisher’s exact test, p < 0.01); (e) Distribution of molecular subtypes (urothelial-like, basal-squamous, nontype, genomically-unstable, mesenchymal) in immune high and immune low clusters (Chi-square test, p < 0.05); (f) Genomically-unstable subtype is significantly associated with immune high cluster (Fisher’s exact test, p < 0.01).
Figure 2
Figure 2
Supervised hierarchical clustering with immunohistochemistry staining of 13 proteins representing molecular subtyping genes demonstrates that the immune high and immune low clusters are not associated with the individual gene signatures. Only specimens with available data of the immunohistochemical stains are used for the hierarchical clustering. The dendrogram is determined by unsupervised hierarchical clustering of the immune markers (top panel). Expression levels of the 13 genes are plotted in the bottom panel following the same dendrogram (supervised).
Figure 3
Figure 3
Comparisons of immune marker scores in CIS, NIPUC and invasive UC. (a,b) CIS has significantly higher chronic inflammation and PD1 scores as compared to NIPUC and invasive UC; (ce) Invasive UC has significantly higher CD3, CD8 and PD-L1 scores as compared to NIPUC and invasive UC. All statistical significance was calculated by one with ANOVA with posttest as described in methods and materials and plotted as Mean ± SD. p < 0.05 is considered significant as *.
Figure 4
Figure 4
Immune scores of PD-L1 and CD3 among invasive UC variants. (a) Sarcomatoid variant shows significantly higher PD-L1 score as compared to the nested, glandular, and conventional UC. (b) Squamous, sarcomatoid, and plasmacytoid variants tend to have higher intra-tumoral CD3 scores. All statistical significance was calculated by one with ANOVA with posttest as described in methods and materials, and plotted as Mean ± SD. p < 0.05 is considered significant as *.
Figure 5
Figure 5
Intra-tumoral immune heterogeneity in invasive urothalial carcinoma. Each row represents a patient and each column is a histologic variant. (a) Distribution of immune low and immune high cluster profile in different histologic variants from the same patient. (b) Distribution of PD-L1 scores in different histologic variants from the same patient. (c) Distribution of molecular subtypes in different histologic variants from the same patient. (d) Distribution of intra-tumoral CD3 scores in different histologic variants from the same patient.

References

    1. Bellmunt J, et al. Bladder cancer: ESMO Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2014;25(Suppl 3):iii40–48. doi: 10.1093/annonc/mdu223.
    1. Milowsky MI, et al. Guideline on Muscle-Invasive and Metastatic Bladder Cancer (European Association of Urology Guideline): American Society of Clinical Oncology Clinical Practice Guideline Endorsement. J. Clin. Oncol. 2016;34:1945–1952. doi: 10.1200/JCO.2015.65.9797.
    1. Zhu J, et al. Biomarkers of immunotherapy in urothelial and renal cell carcinoma: PD-L1, tumor mutational burden, and beyond. J. Immunother. Cancer. 2018;6:4. doi: 10.1186/s40425-018-0314-1.
    1. Massard C, et al. Safety and Efficacy of Durvalumab (MEDI4736), an Anti-Programmed Cell Death Ligand-1 Immune Checkpoint Inhibitor, in Patients With Advanced Urothelial Bladder Cancer. J. Clin. Oncol. 2016;34:3119–3125. doi: 10.1200/JCO.2016.67.9761.
    1. Hahn NM, et al. A Phase II Trial of Dovitinib in BCG-Unresponsive Urothelial Carcinoma with. Clin. Cancer Res. 2017;23:3003–3011. doi: 10.1158/1078-0432.CCR-16-2267.
    1. Mann SA, et al. Targeting the Programmed Cell Death-1 Pathway in Genitourinary Tumors: Current Progress and Future Perspectives. Curr. Drug. Metab. 2017;18:700–711. doi: 10.2174/1389200218666170518162500.
    1. Rosenberg JE, 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. 2016;387:1909–1920. doi: 10.1016/S0140-6736(16)00561-4.
    1. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17:e542–e551. doi: 10.1016/S1470-2045(16)30406-5.
    1. Necchi, A. et al. Pembrolizumab as Neoadjuvant Therapy Before Radical Cystectomy in Patients With Muscle-Invasive Urothelial Bladder Carcinoma (PURE-01): An Open-Label, Single-Arm, Phase II Study. J Clin Oncol, JCO1801148 (2018).
    1. Galon J, et al. Cancer classification using the Immunoscore: a worldwide task force. J. Transl. Med. 2012;10:205. doi: 10.1186/1479-5876-10-205.
    1. Paulsen EE, et al. Assessing PDL-1 and PD-1 in Non-Small Cell Lung Cancer: A Novel Immunoscore Approach. Clin. Lung Cancer. 2017;18:220–233.e228. doi: 10.1016/j.cllc.2016.09.009.
    1. Kwak Y, et al. Immunoscore encompassing CD3+ and CD8+ T cell densities in distant metastasis is a robust prognostic marker for advanced colorectal cancer. Oncotarget. 2016;7:81778–81790.
    1. Ford TF, Grant DA, Austen BM, Hermon-Taylor J. Intramucosal activation of pepsinogens in the pathogenesis of acute gastric erosions and their prevention by the potent semisynthetic amphipathic inhibitor pepstatinyl-glycyl-lysyl-lysine. Clin. Chim. Acta. 1985;145:37–47. doi: 10.1016/0009-8981(85)90017-8.
    1. Lopez-Beltran A, et al. Variants and new entities of bladder cancer. Histopathology. 2019;74:77–96. doi: 10.1111/his.13752.
    1. McConkey DJ, Choi W. Molecular Subtypes of Bladder Cancer. Curr. Oncol. Rep. 2018;20:77. doi: 10.1007/s11912-018-0727-5.
    1. Volkmer JP, et al. Three differentiation states risk-stratify bladder cancer into distinct subtypes. Proc. Natl Acad. Sci. USA. 2012;109:2078–2083. doi: 10.1073/pnas.1120605109.
    1. Damrauer JS, et al. Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc. Natl Acad. Sci. USA. 2014;111:3110–3115. doi: 10.1073/pnas.1318376111.
    1. Robertson AG, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder. Cancer. Cell. 2018;174:1033.
    1. Kamoun, A. et al. (Cold Spring Harbor Laboratory, BioXiv, 2018).
    1. Sjödahl, G., Eriksson, P., Liedberg, F. & Höglund, M. Molecular classification of urothelial carcinoma: global mRNA classification versus tumour-cell phenotype classification. J Pathol242 (2017).
    1. Network CGAR. Comprehensive molecular characterization of urothelial bladder carcinoma. Nat. 2014;507:315–322. doi: 10.1038/nature12965.
    1. Aggen DH, Drake CG. Biomarkers for immunotherapy in bladder cancer: a moving target. J. Immunother. Cancer. 2017;5:94. doi: 10.1186/s40425-017-0299-1.
    1. Mariathasan S, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nat. 2018;554:544–548. doi: 10.1038/nature25501.
    1. Warrick JI, et al. Squamous Dysplasia of the Urinary Bladder: A Consecutive Cystectomy Series. Int. J. Surg. Pathol. 2016;24:306–314. doi: 10.1177/1066896916629783.
    1. Warrick JI, et al. FOXA1 and CK14 as markers of luminal and basal subtypes in histologic variants of bladder cancer and their associated conventional urothelial carcinoma. Virchows Arch. 2017;471:337–345. doi: 10.1007/s00428-017-2190-3.
    1. Warrick, J. I. et al. Intratumoral Heterogeneity of Bladder Cancer by Molecular Subtypes and Histologic Variants. Eur Urol (2018).
    1. Sjodahl G, et al. A molecular taxonomy for urothelial carcinoma. Clin. Cancer Res. 2012;18:3377–3386. doi: 10.1158/1078-0432.CCR-12-0077-T.
    1. Tripathi A, Plimack ER. Immunotherapy for Urothelial Carcinoma: Current Evidence and Future Directions. Curr. Urol. Rep. 2018;19:109. doi: 10.1007/s11934-018-0851-7.
    1. Rentsch CA, et al. Building on a Solid Foundation: Enhancing Bacillus Calmette-Guérin Therapy. Eur. Urol. Focus. 2018;4:485–493. doi: 10.1016/j.euf.2018.10.010.
    1. Faraj SF, et al. Assessment of tumoral PD-L1 expression and intratumoral CD8+ T cells in urothelial carcinoma. Urol. 2015;85(703):e701–706.
    1. Yao S, Chen L. Adaptive resistance: a tumor strategy to evade immune attack. Eur. J. Immunol. 2013;43:576–579. doi: 10.1002/eji.201243275.
    1. Cheng W, Fu D, Xu F, Zhang Z. Unwrapping the genomic characteristics of urothelial bladder cancer and successes with immune checkpoint blockade therapy. Oncogenesis. 2018;7:2. doi: 10.1038/s41389-017-0013-7.
    1. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immun. 2004;21:137–148. doi: 10.1016/j.immuni.2004.07.017.
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Sci. 2015;348:56–61. doi: 10.1126/science.aaa8172.
    1. Chiappinelli KB, et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell. 2015;162:974–986. doi: 10.1016/j.cell.2015.07.011.
    1. Li H, et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5:587–598.
    1. Stone, M. L. et al. Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden. Proc Natl Acad Sci USA114 (2017).
    1. Topper MJ, et al. Epigenetic Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung Cancer. Cell. 2017;171:1284–1300.e1221. doi: 10.1016/j.cell.2017.10.022.
    1. Connolly RM, et al. Combination Epigenetic Therapy in Advanced Breast Cancer with 5-Azacitidine and Entinostat: A Phase II National Cancer Institute/Stand Up to Cancer Study. Clin. Cancer Res. 2017;23:2691–2701. doi: 10.1158/1078-0432.CCR-16-1729.
    1. Lipponen PK, Eskelinen MJ, Jauhiainen K, Harju E, Terho R. Tumour infiltrating lymphocytes as an independent prognostic factor in transitional cell bladder cancer. Eur. J. Cancer. 1992;29A:69–75.
    1. Yu A, et al. Presence of lymphocytic infiltrate cytotoxic T lymphocyte CD3+, CD8+, and immunoscore as prognostic marker in patients after radical cystectomy. PLoS One. 2018;13:e0205746. doi: 10.1371/journal.pone.0205746.
    1. Sjödahl G, et al. Infiltration of CD3+ and CD68+ cells in bladder cancer is subtype specific and affects the outcome of patients with muscle-invasive tumors. Urol. Oncol. 2014;32:791–797. doi: 10.1016/j.urolonc.2014.02.007.
    1. Puzio-Kuter AM, et al. Inactivation of p53 and Pten promotes invasive bladder cancer. Genes. Dev. 2009;23:675–680. doi: 10.1101/gad.1772909.
    1. Jakobsson L, Chebil G, Marzouka NA, Liedberg F, Sjödahl G. Low Frequency of Intratumor Heterogeneity in Bladder Cancer Tissue Microarrays. Bladder Cancer. 2018;4:327–337. doi: 10.3233/BLC-180176.
    1. Reddy OL, et al. Loss of FOXA1 Drives Sexually Dimorphic Changes in Urothelial Differentiation and Is an Independent Predictor of Poor Prognosis in Bladder Cancer. Am. J. Pathol. 2015;185:1385–1395. doi: 10.1016/j.ajpath.2015.01.014.

Source: PubMed

3
購読する