Evaluation of PD-L1 expression on circulating tumor cells (CTCs) in patients with advanced urothelial carcinoma (UC)

Sonja Bergmann, Anja Coym, Leonie Ott, Armin Soave, Michael Rink, Melanie Janning, Malgorzata Stoupiec, Cornelia Coith, Sven Peine, Gunhild von Amsberg, Klaus Pantel, Sabine Riethdorf, Sonja Bergmann, Anja Coym, Leonie Ott, Armin Soave, Michael Rink, Melanie Janning, Malgorzata Stoupiec, Cornelia Coith, Sven Peine, Gunhild von Amsberg, Klaus Pantel, Sabine Riethdorf

Abstract

Immune checkpoint inhibition (ICI) of the PD-1/PD-L1 axis shows durable responses in a subset of patients with metastatic urothelial carcinoma (UC). However, PD-L1 expression in tumor biopsies does not necessarily correlate with response to PD-1/PD-L1 inhibitors. Thus, a reliable predictive biomarker is urgently needed. Here, the expression of PD-L1 on circulating tumor cells (CTCs) in blood from patients with advanced UC was analyzed. For this purpose, an assay to test PD-L1 expression on CTCs using the CellSearch® system was established using cells of five UC cell lines spiked into blood samples from healthy donors and applied to a heterogeneous cohort of UC patients. Enumeration of CTCs was performed in blood samples from 49 patients with advanced UC. PD-L1 expression in ≥1 CTC was found in 10 of 16 CTC-positive samples (63%). Both intra- and inter-patient heterogeneity regarding PD-L1 expression of CTCs were observed. Furthermore, vimentin-expressing CTCs were detected in 4 of 15 CTC-positive samples (27%), independently of PD-L1 analysis. Both CTC detection and presence of CTCs with moderate or strong PD-L1 expression correlated with worse overall survival. Analyses during disease course of three individual patients receiving ICI suggest that apart from CTC numbers also PD-L1 expression on CTCs might potentially indicate disease progression. This is the first study demonstrating the feasibility to detect CTC-PD-L1 expression in patients with advanced UC using the CellSearch® system. This assay is readily available for clinical application and could be implemented in future clinical trials to evaluate its relevance for predicting and monitoring response to ICI.

Keywords: PD-L1; cellsearch; circulating tumor cells; immune checkpoint inhibition; urothelial carcinoma; vimentin.

© 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.

Figures

Figure 1.
Figure 1.
Characterization of the anti-PD-L1 antibody clone E1L3N®. (a) Western blot analysis of PD-L1, EpCAM, pan-keratin and vimentin expression in UC cell lines (RT-4, 647V, 5637, T24, and TCC-SUP) and in the breast cancer cell line MDA-MB-231. Protein loading control: HSC70 (Heat shock cognate 71 kDa protein). The bar chart indicates EMT scores according to Tan et al. (b) Specificity of the anti-PD-L1 antibody was confirmed by Western blot analysis of 5637 cells with the retroviral transfer of the CD274 gene encoding for PD-L1 or the empty vector (EV). Protein loading control: HSC70. (c) FACS (fluorescence activated cell sorting) analysis of PD-L1 expression in UC cell lines (RT-4, 647V, 5637, T24, and TCC-SUP). Cells were stained with the PE-conjugated anti-PD-L1 antibody clone E1L3N® (blue) in comparison to the respective isotype control clone DA1E (gray). Mean fluorescence intensities (MFI) were determined. (d) IF (immunofluorescence) analysis of PD-L1 expression in UC cell line cells (RT-4: PD-L1-negative, 647V: PD-L1-positive). Cells were spiked into whole blood from healthy donors prior to centrifugation. PD-L1 protein was detected by the PE-conjugated anti-PD-L1 antibody clone E1L3N®. The cells were additionally stained with the AlexaFluor488 (AF488)-conjugated anti-keratin antibodies (clones AE1/AE3 and C11) and the APC-conjugated anti-CD45 (clone REA747) antibody. Nuclei were stained by DAPI (4‘,6-Diamidin-2-phenylindol).
Figure 2.
Figure 2.
CellSearch® CXC kit analysis of UC cells spiked into the blood from healthy donors. Cells were enriched by anti-EpCAM magnetic beads using the CXC kit. Keratin+ (KER-FLU), CD45- (CD45-APC), DAPI+ cells were identified as tumor cells. (a) PD-L1 expression of RT-4 cells and (b) 647V cells were detected by the PE-conjugated anti-PD-L1 antibody clone E1L3N® (PD-L1-PE, right panel) in comparison to the isotype control clone DA1E (isotype-PE, left panel). Images of PD-L1-specific fluorescent signals were generated using the indicated exposure time. Furthermore, PD-L1 expression of (c) 5637, (d) T24 and (e) TCC-SUP cells were detected by the PE-conjugated anti-PD-L1 antibody clone E1L3N® (PD-L1-PE) using an exposure time of 1.6 sec. Identification of TCC-SUP cells was performed by manually evaluating fluorescent signals of stained cells in the CellSelect mode because of the low keratin expression impeding automatic selection. (f) TCC-SUP identity of the reported cells was confirmed by single-cell mutational analysis of the PIK3CA gene exon 9. Representative sequencing results showed a heterozygous mutation (G1633A) previously reported by Platt et al. that was detected in a recovered single TCC-SUP cell but was absent in a leukocyte.
Figure 3.
Figure 3.
Determination of PD-L1 expression on CTCs. (a) CellSearch® analysis of CTCs detected in the blood of an advanced UC patient. Cells were enriched by anti-EpCAM magnetic beads using the CXC kit. Keratin+ (KER-FLU), CD45- (CD45-APC), DAPI+ cells were identified as tumor cells. PD-L1 expression of individual CTCs was detected by the PE-conjugated anti-PD-L1 antibody clone E1L3N® (PD-L1-PE) and categorized into negative (0), weakly positive (1+), moderately positive (2+) and strongly positive (3+). Images of PD-L1-specific fluorescent signals were generated using an exposure time of 1.6 sec. (b) IF analysis of simultaneous binding of the therapeutic anti-PD-L1 antibody atezolizumab and the diagnostic anti-PD-L1 antibody E1L3N® to PD-L1 on tumor cells. PD-L1-negative RT-4 cells and PD-L1-positive 647V cells were spun onto glass slides and incubated in the presence of 1.2 µg/mL atezolizumab, which was detected by AlexaFluor488-labeled secondary antibody (AF488). Cells were additionally incubated with PE-conjugated anti-PD-L1 antibody. Nuclei were stained by DAPI. (c) Detection of atezolizumab in human blood samples. PD-L1-negative RT-4 cells and PD-L1-positive 647V cells spiked into the blood from healthy donors were spun onto glass slides and incubated in presence of 1.2 µg/mL atezolizumab, which was detected by anti-atezolizumab antibody and AF546-labeled secondary anti-mouse antibody. Mouse IgG served as an isotype control to anti-atezolizumab antibody. Keratins, CD45, and nuclei were stained by manually applying reagents of the CellSearch® CXC kit.
Figure 4.
Figure 4.
CellSearch® CTC kit analysis of vimentin expression. Cells were enriched by anti-EpCAM magnetic beads using the CTC kit. Keratin+ (KER-PE), CD45- (CD45-APC), DAPI+ cells were identified as tumor cells. Vimentin expression of (a) RT-4, 5637 and MDA-MB-231 cells and of (b) CTCs detected in the blood of advanced UC patients was analyzed by the AlexaFluor488-conjugated anti-vimentin antibody clone V9 (VIM-AF488). Images of vimentin-specific fluorescent signals were generated using an exposure time of 0.8 sec. Vimentin expression of individual CTCs was categorized into negative (0), weakly positive (1+) and moderately positive (2+).
Figure 5.
Figure 5.
Disease courses and clinical outcome of patients with advanced UC in relation to CTC-PD-L1 expression. (a) Kaplan-Meier plot and Log Rank test for overall survival of UC patients according to CTC-PD-L1 expression (0/1+ compared to 2+/3+) of CTC-positive samples using the CXC kit. Values of p < .05 were considered statistically significant. (b, c) Disease courses of two individual patients with metastatic UC who progressed under ICI (immune checkpoint inhibition) (B, pretreated; C, first-line) and (d) one patient who responded to ICI (pretreated) accompanied by CTC numbers as well as CTC-vimentin (VIM) and CTC-PD-L1 expression levels analyzed using the CellSearch® system. For each sample (1 and 2), metastatic sites are indicated. CTC analyses were performed using the CTC kit and/or the CXC kit (A and B, respectively, with N/A indicating that the analysis was not applied). Time spans between samples and maximum follow-up times as well as survival information are given. Arrows depict the duration of ICI therapy in relation to the acquisition of samples.

References

    1. Bellmunt J, Powles T, Vogelzang NJ.. A review on the evolution of PD-1/PD-L1 immunotherapy for bladder cancer: the future is now. Cancer Treat Rev. 2017;54:58–13. doi:10.1016/j.ctrv.2017.01.007.
    1. Powles T, Necchi A, Rosen G, Hariharan S, Apolo AB. Anti-programmed cell death 1/ligand 1 (PD-1/PD-L1) antibodies for the treatment of urothelial carcinoma: state of the art and future development. Clin Genitourin Cancer. 2018;16:117–129. doi:10.1016/j.clgc.2017.11.002.
    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. Carbognin L, Pilotto S, Milella M, Vaccaro V, Brunelli M, Calio A, Cuppone F, Sperduti I, Giannarelli D, Chilosi M, et al. Differential activity of nivolumab, pembrolizumab and MPDL3280A according to the tumor expression of programmed death-ligand-1 (PD-L1): sensitivity analysis of trials in melanoma, lung and genitourinary cancers. PLoS One. 2015;10:e0130142. doi:10.1371/journal.pone.0130142.
    1. Madore J, Vilain RE, Menzies AM, Kakavand H, Wilmott JS, Hyman J, Yearley JH, Kefford RF, Thompson JF, Long GV, et al. PD-L1 expression in melanoma shows marked heterogeneity within and between patients: implications for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res. 2015;28:245–253. doi:10.1111/pcmr.12340.
    1. Kerr KM, Tsao MS, Nicholson AG, Yatabe Y, Wistuba II, Hirsch FR, Committee IP. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol. 2015;10:985–989. doi:10.1097/JTO.0000000000000526.
    1. Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol. 2016;27:409–416. doi:10.1093/annonc/mdv615.
    1. Shi Y. Regulatory mechanisms of PD-L1 expression in cancer cells. Cancer Immunol Immunother. 2018;67:1481–1489. doi:10.1007/s00262-018-2226-9.
    1. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, Zaretsky JM, Sun L, Hugo W, Wang X, et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 2017;19:1189–1201. doi:10.1016/j.celrep.2017.04.031.
    1. Moon JW, Kong SK, Kim BS, Kim HJ, Lim H, Noh K, Kim Y, Choi JW, Lee JH, Kim YS. IFNgamma induces PD-L1 overexpression by JAK2/STAT1/IRF-1 signaling in EBV-positive gastric carcinoma. Sci Rep. 2017;7:17810. doi:10.1038/s41598-017-18132-0.
    1. Tretiakova M, Fulton R, Kocherginsky M, Long T, Ussakli C, Antic T, Gown A. Concordance study of PD-L1 expression in primary and metastatic bladder carcinomas: comparison of four commonly used antibodies and RNA expression. Mod Pathol. 2018;31:623–632. doi:10.1038/modpathol.2017.188.
    1. Pantel K, Alix-Panabieres C. Liquid biopsy: potential and challenges. Mol Oncol. 2016;10:371–373. doi:10.1016/j.molonc.2016.01.009.
    1. Pantel K, Speicher MR. The biology of circulating tumor cells. Oncogene. 2016;35:1216–1224. doi:10.1038/onc.2015.192.
    1. Rink M, Chun FK, Dahlem R, Soave A, Minner S, Hansen J, Stoupiec M, Coith C, Kluth LA, Ahyai SA, et al. Prognostic role and HER2 expression of circulating tumor cells in peripheral blood of patients prior to radical cystectomy: a prospective study. Eur Urol. 2012;61:810–817. doi:10.1016/j.eururo.2012.01.017.
    1. Soave A, Riethdorf S, Dahlem R, von Amsberg G, Minner S, Weisbach L, Engel O, Fisch M, Pantel K, Rink M. A nonrandomized, prospective, clinical study on the impact of circulating tumor cells on outcomes of urothelial carcinoma of the bladder patients treated with radical cystectomy with or without adjuvant chemotherapy. Int J Cancer. 2017;140:381–389. doi:10.1002/ijc.30445.
    1. Okegawa T, Itaya N, Hara H, Tambo M, Nutahara K. Epidermal growth factor receptor status in circulating tumor cells as a predictive biomarker of sensitivity in castration-resistant prostate cancer patients treated with docetaxel chemotherapy. Int J Mol Sci. 2016:17. doi:10.3390/ijms17122008.
    1. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442–454. doi:10.1038/nrc822.
    1. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–890. doi:10.1016/j.cell.2009.11.007.
    1. Alix-Panabieres C, Pantel K. Circulating tumor cells: liquid biopsy of cancer. Clin Chem. 2013;59:110–118. doi:10.1373/clinchem.2012.194258.
    1. Mak MP, Tong P, Diao L, Cardnell RJ, Gibbons DL, William WN, Skoulidis F, Parra ER, Rodriguez-Canales J, Wistuba II, et al. Pan-cancer EMT signature identifies global molecular alterations and immune target enrichment following epithelial-to-mesenchymal transition. Clin Cancer Res. 2016;22:609–620. doi:10.1158/1078-0432.CCR-15-0876.
    1. Boorjian SA, Sheinin Y, Crispen PL, Farmer SA, Lohse CM, Kuntz SM, Leibovich BC, Kwon ED, Frank I. T-cell coregulatory molecule expression in urothelial cell carcinoma: clinicopathologic correlations and association with survival. Clin Cancer Res. 2008;14:4800–4808. doi:10.1158/1078-0432.CCR-08-0731.
    1. Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S. Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother. 2007;56:1173–1182. doi:10.1007/s00262-006-0266-z.
    1. Brock G, Castellanos-Rizaldos E, Hu L, Coticchia C, Skog J. Liquid biopsy for cancer screening, patient stratification and monitoring. Transl Cancer Res. 2015;4:280–290.
    1. Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, Khoo KH, Chang SS, Cha JH, Kim T, et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat Commun. 2016;7:12632. doi:10.1038/ncomms12632.
    1. Mahoney KM, Sun H, Liao X, Hua P, Callea M, Greenfield EA, Hodi FS, Sharpe AH, Signoretti S, Rodig SJ, et al. PD-L1 antibodies to its cytoplasmic domain most clearly delineate cell membranes in immunohistochemical staining of tumor cells. Cancer Immunol Res. 2015;3:1308–1315. doi:10.1158/2326-6066.CIR-15-0116.
    1. Warrick JI, Walter V, Yamashita H, Chung E, Shuman L, Amponsa VO, Zheng Z, Chan W, Whitcomb TL, Yue F, et al. FOXA1, GATA3 and PPAR cooperate to drive luminal subtype in bladder cancer: a molecular analysis of established human cell lines. Sci Rep. 2016;6:38531. doi:10.1038/srep38531.
    1. Tan TZ, Miow QH, Miki Y, Noda T, Mori S, Huang RY, Thiery JP. Epithelial-mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO Mol Med. 2014;6:1279–1293. doi:10.15252/emmm.201404208.
    1. Platt FM, Hurst CD, Taylor CF, Gregory WM, Harnden P, Knowles MA. Spectrum of phosphatidylinositol 3-kinase pathway gene alterations in bladder cancer. Clin Cancer Res. 2009;15:6008–6017. doi:10.1158/1078-0432.CCR-09-0898.
    1. Miller MC, Doyle GV, Terstappen LW. Significance of circulating tumor cells detected by the cellsearch system in patients with metastatic breast colorectal and prostate cancer. J Oncol. 2010;2010:617421. doi:10.1155/2010/617421.
    1. Gazzaniga P, de Berardinis E, Raimondi C, Gradilone A, Busetto GM, De Falco E, Nicolazzo C, Giovannone R, Gentile V, Cortesi E, et al. Circulating tumor cells detection has independent prognostic impact in high-risk non-muscle invasive bladder cancer. Int J Cancer. 2014;135:1978–1982. doi:10.1002/ijc.28830.
    1. Abrahamsson J, Aaltonen K, Engilbertsson H, Liedberg F, Patschan O, Ryden L, Sjodahl G, Gudjonsson S. Circulating tumor cells in patients with advanced urothelial carcinoma of the bladder: association with tumor stage, lymph node metastases, FDG-PET findings, and survival. Urol Oncol. 2017;35:606 e609–606 e616. doi:10.1016/j.urolonc.2017.05.021.
    1. Gallagher DJ, Milowsky MI, Ishill N, Trout A, Boyle MG, Riches J, Fleisher M, Bajorin DF. Detection of circulating tumor cells in patients with urothelial cancer. Ann Oncol. 2009;20:305–308. doi:10.1093/annonc/mdn627.
    1. Flaig TW, Wilson S, van Bokhoven A, Varella-Garcia M, Wolfe P, Maroni P, Genova EE, Morales D, Lucia MS. Detection of circulating tumor cells in metastatic and clinically localized urothelial carcinoma. Urology. 2011;78:863–867. doi:10.1016/j.urology.2011.05.045.
    1. Okegawa T, Hayashi K, Hara H, Nutahara K, Higashihara E. Immunomagnetic quantification of circulating tumor cells in patients with urothelial cancer. Int J Urol. 2010;17:254–258. doi:10.1111/j.1442-2042.2010.02454.x.
    1. Naoe M, Ogawa Y, Morita J, Omori K, Takeshita K, Shichijyo T, Okumura T, Igarashi A, Yanaihara A, Iwamoto S, et al. Detection of circulating urothelial cancer cells in the blood using the cellsearch system. Cancer. 2007;109:1439–1445. doi:10.1002/cncr.22543.
    1. Riethdorf S, Soave A, Rink M. The current status and clinical value of circulating tumor cells and circulating cell-free tumor DNA in bladder cancer. Transl Androl Urol. 2017;6:1090–1110. doi:10.21037/tau.2017.09.16.
    1. Alix-Panabieres C, Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer. 2014;14:623–631. doi:10.1038/nrc3820.
    1. Jolly MK, Tripathi SC, Jia D, Mooney SM, Celiktas M, Hanash SM, Mani SA, Pienta KJ, Ben-Jacob E, Levine H. Stability of the hybrid epithelial/mesenchymal phenotype. Oncotarget. 2016;7:27067–27084. doi:10.18632/oncotarget.8166.
    1. Lindsay CR, Faugeroux V, Michiels S, Pailler E, Facchinetti F, Ou D, Bluthgen MV, Pannet C, Ngo-Camus M, Bescher G, et al. A prospective examination of circulating tumor cell profiles in non-small-cell lung cancer molecular subgroups. Ann Oncol. 2017;28:1523–1531. doi:10.1093/annonc/mdx156.
    1. Lindsay CR, Le Moulec S, Billiot F, Loriot Y, Ngo-Camus M, Vielh P, Fizazi K, Massard C, Farace F. Vimentin and Ki67 expression in circulating tumour cells derived from castrate-resistant prostate cancer. BMC Cancer. 2016;16:168. doi:10.1186/s12885-016-2192-6.
    1. Raimondi C, Carpino G, Nicolazzo C, Gradilone A, Gianni W, Gelibter A, Gaudio E, Cortesi E, Gazzaniga P. PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system? Oncoimmunology. 2017;6:e1315488. doi:10.1080/2162402X.2017.1315488.
    1. Hodgson A, Liu SK, Vesprini D, Xu B, Downes MR. Basal-subtype bladder tumours show a ‘hot’ immunophenotype. Histopathology. 2018;73:748–757. doi:10.1111/his.13696.
    1. Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, Hinoue T, Laird PW, Hoadley KA, Akbani R, et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell. 2018;174:1033. doi:10.1016/j.cell.2018.07.036.
    1. Anantharaman A, Friedlander T, Lu D, Krupa R, Premasekharan G, Hough J, Edwards M, Paz R, Lindquist K, Graf R, et al. Programmed death-ligand 1 (PD-L1) characterization of circulating tumor cells (CTCs) in muscle invasive and metastatic bladder cancer patients. BMC Cancer. 2016;16:744. doi:10.1186/s12885-016-2758-3.
    1. Mazel M, Jacot W, Pantel K, Bartkowiak K, Topart D, Cayrefourcq L, Rossille D, Maudelonde T, Fest T, Alix-Panabieres C. Frequent expression of PD-L1 on circulating breast cancer cells. Mol Oncol. 2015;9:1773–1782. doi:10.1016/j.molonc.2015.05.009.
    1. Nicolazzo C, Raimondi C, Mancini M, Caponnetto S, Gradilone A, Gandini O, Mastromartino M, Del Bene G, Prete A, Longo F, et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Sci Rep. 2016;6:31726. doi:10.1038/srep31726.
    1. Janning M, Kobus F, Babayan A, Wikman H, Velthaus JL, Bergmann S, Schatz S, Falk M, Berger LA, Bottcher LM, et al. Determination of PD-L1 expression in circulating tumor cells of NSCLC patients and correlation with response to PD-1/PD-L1 inhibitors. Cancers (Basel). 2019;11:835. doi:10.3390/cancers11060835.
    1. Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells - mechanisms of immune surveillance and escape. Nat Rev Clin Oncol. 2017;14:155–167. doi:10.1038/nrclinonc.2016.144.
    1. Werner S, Frey S, Riethdorf S, Schulze C, Alawi M, Kling L, Vafaizadeh V, Sauter G, Terracciano L, Schumacher U, et al. Dual roles of the transcription factor grainyhead-like 2 (GRHL2) in breast cancer. J Biol Chem. 2013;288:22993–23008. doi:10.1074/jbc.M113.456293.
    1. Assmann V, Marshall JF, Fieber C, Hofmann M, Hart IR. The human hyaluronan receptor RHAMM is expressed as an intracellular protein in breast cancer cells. J Cell Sci. 1998;111:1685–1694.

Source: PubMed

3
Předplatit