β-catenin-independent WNT signaling and Ki67 in contrast to the estrogen receptor status are prognostic and associated with poor prognosis in breast cancer liver metastases

Annalen Bleckmann, Lena-Christin Conradi, Kerstin Menck, Nadine Annette Schmick, Antonia Schubert, Eva Rietkötter, Jetcy Arackal, Peter Middel, Alexandra Schambony, Torsten Liersch, Kia Homayounfar, Tim Beißbarth, Florian Klemm, Claudia Binder, Tobias Pukrop, Annalen Bleckmann, Lena-Christin Conradi, Kerstin Menck, Nadine Annette Schmick, Antonia Schubert, Eva Rietkötter, Jetcy Arackal, Peter Middel, Alexandra Schambony, Torsten Liersch, Kia Homayounfar, Tim Beißbarth, Florian Klemm, Claudia Binder, Tobias Pukrop

Abstract

Liver metastasis development in breast cancer patients is common and confers a poor prognosis. So far, the prognostic significance of surgical resection and clinical relevance of biomarker analysis in metastatic tissue have barely been investigated. We previously demonstrated an impact of WNT signaling in breast cancer brain metastasis. This study aimed to investigate the value of established prognostic markers and WNT signaling components in liver metastases. Overall N = 34 breast cancer liver metastases (with matched primaries in 19/34 cases) were included in this retrospective study. Primaries and metastatic samples were analyzed for their expression of the estrogen (ER) and progesterone receptor, HER-2, Ki67, and various WNT signaling-components by immunohistochemistry. Furthermore, β-catenin-dependent and -independent WNT scores were generated and analyzed for their prognostic value. Additionally, the influence of the alternative WNT receptor ROR on signaling and invasiveness was analyzed in vitro. ER positivity (HR 0.09, 95 % CI 0.01-0.56) and high Ki67 (HR 3.68, 95 % CI 1.12-12.06) in the primaries had prognostic impact. However, only Ki67 remained prognostic in the metastatic tissue (HR 2.46, 95 % CI 1.11-5.44). Additionally, the β-catenin-independent WNT score correlated with reduced overall survival only in the metastasized situation (HR 2.19, 95 % CI 1.02-4.69, p = 0.0391). This is in line with the in vitro results of the alternative WNT receptors ROR1 and ROR2, which foster invasion. In breast cancer, the value of prognostic markers established in primary tumors cannot directly be translated to metastases. Our results revealed β-catenin-independent WNT signaling to be associated with poor prognosis in patients with breast cancer liver metastasis.

Keywords: Breast cancer; Metastasis; Prognostic score; WNT signaling.

Figures

Fig. 1
Fig. 1
a Immunoblot for WNT5a, Dvl3, β-catenin, and c-Jun of MCF-7, SK-BR-3 and MDA-MB-231 cell lysates. B qRT-PCR of MCF-7, SK-BR-3 and MDA-MB-231 for ROR1 and ROR2. Each dot represents one independent biological sample (means, n = 3, n.d. = not detectable). Note that in MDA-MB-231 ROR2 was only detectable in 2/3 samples. C Flow cytometry (gray isotype control, black stained cells for MCF-7, SK-BR-3 and MDA-MB-231) measuring the expression of ROR1
Fig. 2
Fig. 2
a Immunoblots showing the expression of ROR1 and ROR2 in MCF-7 and SK-BR-3 cells transfected with either an empty vector (ctl) or a ROR2 overexpression plasmid (pROR2). B In vitro microinvasion assays of ROR2-overexpressing MCF-7 and SK-BR-3 cells compared to wildtype cells (wt) or cells transfected with an empty vector (ctl) (mean ± SD, n = 3, *p < 0.001). C Immunoblot for c-Jun and HDAC of MCF-7 and SK-BR-3 empty vector (ctl) or ROR2-overexpressing cells for cytoplasmic (C) and nuclear (N) cell lysates. D Immunofluorescence staining of c-Jun and F-actin (phalloidin) in MCF-7 empty vector (ctl) and ROR2 cells as well as β-catenin and Dapi in MCF-7 wt and ROR2 cells. E Immunoblot of ROR1 in MDA-MB-231 cells transfected with nonsense control (ctl) or ROR1 (shROR1) shRNA. F In vitro microinvasion assay of MDA-MB-231 wildtype (wt), non-sense control (ctl) and ROR1 knockdown (shROR1) cells (mean ± SD, n = 3, *p < 0.001)
Fig. 3
Fig. 3
Kaplan-Meier curves illustrate that ER expression in the primary tumor but not in the liver metastases is correlated with a good prognosis (a). Positivity for the proliferation index Ki67 in the primary tumor tissue as well as in the liver metastases (b) is associated with reduced overall survival
Fig. 4
Fig. 4
Immunohistochemical stainings for all proteins analysed were performed on primary tumors and metastases respectively. Representative pictures are shown for stainings of primary tumor samples (af) and on metastatic tissue (gl). IHC-staining of membrane E-cadherin showing a positivity in primary tumor cells of >75 % in 20× (a) and 40× magnification (b); nuclear Lef1 positivity is shown at 20× (c) and 40× magnification of primary tumor tissue being positive in 30 % of tumor cells (d); β-catenin staining is shown in panel e (20×) and f without any nuclear activity. Representative examples of nuclear Phospho-c-Jun staining is shown in g (20×) and h (40×) with a positivity rate of >26 % for intrahepatic breast cancer metastases cells; nuclear c-Jun staining is represented in panels i (20×) and j (40×) showing positive stained nuclei in >76 % of metastatic tissue; nuclear Dvl3 staining is also represented in liver metastases beeing positive in <25 % of metastases cells at a magnification of 20× (k) and 40× (l). For all markers, the specific expression pattern was analyzed for primaries and metastases and classified into five groups: 0 = 0 %, 1+ = 1–25 %, 2+ = 26–50 %, 3+ = 51–75 % and 4+ = 76–100 % of the tumor cells with positive (≥1) staining. Bar: 200 μm (a, c, e, g, i and k) and 50 μm (b, d, f, h, j and l)
Fig. 5
Fig. 5
No prognostic impact could be shown for the β-catenin-dependent WNT score (a). In contrast, the β-catenin-independent WNT score has prognostic impact on survival in the metastatic setting (b). Survival rates are depicted with Kaplan–Meier-curves in months

References

    1. De Angelis R, et al. Cancer survival in Europe 1999–2007 by country and age: results of EUROCARE–5-a population-based study. Lancet Oncol. 2014;15(1):23–34. doi: 10.1016/S1470-2045(13)70546-1.
    1. Cardoso F et al. (2014) ESO-ESMO 2nd international consensus guidelines for advanced breast cancer (ABC2) dagger. Annals of oncology: Official Journal of the European Society for Medical Oncology/ESMO vol 25(10), pp 1871–88
    1. Ruiterkamp J, Ernst MF. The role of surgery in metastatic breast cancer. Eur J Cancer. 2011;47(Suppl 3):S6–S22. doi: 10.1016/S0959-8049(11)70142-3.
    1. Choti MA. Liver-directed treatments for metastatic colorectal cancer. Curr Treat Options Oncol. 2014;15(3):456–464. doi: 10.1007/s11864-014-0297-1.
    1. Elsberger B, Roxburgh CS, Horgan PG. Is there a role for surgical resections of hepatic breast cancer metastases? Hepatogastroenterology. 2014;61(129):181–186.
    1. Page AJ, et al. Advances in understanding of colorectal liver metastasis and implications for the clinic. Expert Rev Gastroenterol Hepatol. 2014;9(2):245–259. doi: 10.1586/17474124.2014.940897.
    1. Krop IE, et al. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol. 2012;30(26):3234–3241. doi: 10.1200/JCO.2011.40.5902.
    1. Emons G, Ghadimi M, Grade M. General principles of tumour biology in visceral surgery. Zentralbl Chir. 2015;140(1):74–82.
    1. Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–284. doi: 10.1038/nrc2622.
    1. Vanharanta S, Massague J. Origins of metastatic traits. Cancer Cell. 2013;24(4):410–421. doi: 10.1016/j.ccr.2013.09.007.
    1. Nguyen DX, Massague J. Genetic determinants of cancer metastasis. Nat Rev Genet. 2007;8(5):341–352. doi: 10.1038/nrg2101.
    1. Langley RR, Fidler IJ (2011) The seed and soil hypothesis revisited—the role of tumor-stroma interactions in metastasis to different organs. Int J Cancer (J Int Cancer), vol 128(11), pp 2527–2535
    1. Gerlinger M, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–892. doi: 10.1056/NEJMoa1113205.
    1. Huang Y et al (2014) Multilayered molecular profiling supported the monoclonal origin of metastatic renal cell carcinoma. Int J Cancer (J Int Cancer), vol 135(1), pp 78–87
    1. Klein CA. Parallel progression of primary tumours and metastases. Nat Rev Cancer. 2009;9(4):302–312. doi: 10.1038/nrc2627.
    1. Stoecklein NH, Klein CA (2010) Genetic disparity between primary tumours, disseminated tumour cells, and manifest metastasis. Int J Cancer (J Int Cancer), vol 126(3), pp 589–98
    1. Chambers AF, Groom AC, Macdonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–572. doi: 10.1038/nrc865.
    1. Chuang HN, et al. Carcinoma cells misuse the host tissue damage response to invade the brain. Glia. 2013;61(8):1331–1346. doi: 10.1002/glia.22518.
    1. Kienast Y, et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2009;16(1):116–122. doi: 10.1038/nm.2072.
    1. Valiente M, et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell. 2014;156(5):1002–1016. doi: 10.1016/j.cell.2014.01.040.
    1. Pukrop T, et al. Microglia promote colonization of brain tissue by breast cancer cells in a Wnt-dependent way. Glia. 2010;58(12):1477–1489.
    1. Bleckmann A, et al. Nuclear LEF1/TCF4 correlate with poor prognosis but not with nuclear beta-catenin in cerebral metastasis of lung adenocarcinomas. Clin Exp Metastasis. 2013;30(4):471–482. doi: 10.1007/s10585-012-9552-7.
    1. Klemm F, et al. beta-catenin-independent WNT signaling in basal-like breast cancer and brain metastasis. Carcinogenesis. 2011;32(3):434–442. doi: 10.1093/carcin/bgq269.
    1. Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue renewal and regeneration: wnt signaling and stem cell control. Science. 2014;346(6205):1248012. doi: 10.1126/science.1248012.
    1. Nusse R. Wnt signaling. Cold Spring Harb Perspect Biol. 2012;4(5):a011163. doi: 10.1101/cshperspect.a011163.
    1. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434(7035):843–850. doi: 10.1038/nature03319.
    1. Gessert S, Kuhl M. The multiple phases and faces of wnt signaling during cardiac differentiation and development. Circ Res. 2010;107(2):186–199. doi: 10.1161/CIRCRESAHA.110.221531.
    1. Ocana OH, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012;22(6):709–724. doi: 10.1016/j.ccr.2012.10.012.
    1. Giancotti FG. Mechanisms governing metastatic dormancy and reactivation. Cell. 2013;155(4):750–764. doi: 10.1016/j.cell.2013.10.029.
    1. Li J, Zhou BP. Activation of beta-catenin and Akt pathways by Twist are critical for the maintenance of EMT associated cancer stem cell-like characters. BMC Cancer. 2011;11:49. doi: 10.1186/1471-2407-11-49.
    1. van de Wetering M, et al. Mutant E-cadherin breast cancer cells do not display constitutive Wnt signaling. Cancer Res. 2001;61(1):278–284.
    1. Gilles C, et al. Transactivation of vimentin by beta-catenin in human breast cancer cells. Cancer Res. 2003;63(10):2658–2664.
    1. Zhang S, et al. ROR1 is expressed in human breast cancer and associated with enhanced tumor-cell growth. PLoS ONE. 2012;7(3):e31127. doi: 10.1371/journal.pone.0031127.
    1. Henry C, et al. Expression of the novel Wnt receptor ROR2 is increased in breast cancer and may regulate both beta-catenin dependent and independent Wnt signalling. J Cancer Res Clin Oncol. 2015;141(2):243–254. doi: 10.1007/s00432-014-1824-y.
    1. Kikuchi A, et al. Wnt5a: its signalling, functions and implication in diseases. Acta Physiol. 2012;204(1):17–33. doi: 10.1111/j.1748-1716.2011.02294.x.
    1. Pukrop T, Binder C. The complex pathways of Wnt 5a in cancer progression. J Mol Med. 2008;86(3):259–266. doi: 10.1007/s00109-007-0266-2.
    1. Witte F, et al. Receptor tyrosine kinase-like orphan receptor 2 (ROR2) and Indian hedgehog regulate digit outgrowth mediated by the phalanx-forming region. Proc Natl Acad Sci USA. 2010;107(32):14211–14216. doi: 10.1073/pnas.1009314107.
    1. Gentzel M, et al. Distinct functionality of dishevelled isoforms on Ca2 +/calmodulin-dependent protein kinase 2 (CamKII) in Xenopus gastrulation. Mol Biol Cell. 2015;26(5):966–977. doi: 10.1091/mbc.E14-06-1089.
    1. Hagemann T, et al. Enhanced invasiveness of breast cancer cell lines upon co-cultivation with macrophages is due to TNF-alpha dependent up-regulation of matrix metalloproteases. Carcinogenesis. 2004;25(8):1543–1549. doi: 10.1093/carcin/bgh146.
    1. van de Vijver M, et al. Chromogenic in situ hybridisation for the assessment of HER2 status in breast cancer: an international validation ring study. Breast Cancer Res. 2007;9(5):R68. doi: 10.1186/bcr1776.
    1. McShane LM, et al. REporting recommendations for tumor MARKer prognostic studies (REMARK) Breast Cancer Res Treat. 2006;100(2):229–235. doi: 10.1007/s10549-006-9242-8.
    1. Nishita M, et al. Ror2/Frizzled complex mediates Wnt5a-induced AP-1 activation by regulating Dishevelled polymerization. Mol Cell Biol. 2010;30(14):3610–3619. doi: 10.1128/MCB.00177-10.
    1. Gao S, et al. Dsh homolog DVL3 mediates resistance to IGFIR inhibition by regulating IGF-RAS signaling. Cancer Res. 2014;74(20):5866–5877. doi: 10.1158/0008-5472.CAN-14-0806.
    1. Hegazy SA, et al. Disheveled proteins promote cell growth and tumorigenicity in ALK-positive anaplastic large cell lymphoma. Cell Signal. 2013;25(1):295–307. doi: 10.1016/j.cellsig.2012.09.027.
    1. Kwan HT, et al. AMPK activators suppress cervical cancer cell growth through inhibition of DVL3 mediated Wnt/beta-catenin signaling activity. PLoS ONE. 2013;8(1):e53597. doi: 10.1371/journal.pone.0053597.
    1. Zhao Y, et al. Dishevelled-1 and dishevelled-3 affect cell invasion mainly through canonical and noncanonical Wnt pathway, respectively, and associate with poor prognosis in nonsmall cell lung cancer. Mol Carcinog. 2010;49(8):760–770.
    1. Navarro V, et al. Toothbrush-thinking seizures. Epilepsia. 2006;47(11):1971–1973. doi: 10.1111/j.1528-1167.2006.00822.x.
    1. Sabol M, et al. Surgical management of breast cancer liver metastases. Neoplasma. 2014;61(5):601–606. doi: 10.4149/neo_2014_074.
    1. Van Cutsem E, et al. Metastatic colorectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014;25(Suppl 3):1–9. doi: 10.1093/annonc/mdu260.
    1. Bachmeier B, et al. Development of resistance towards artesunate in MDA-MB-231 human breast cancer cells. PLoS ONE. 2011;6(5):e20550. doi: 10.1371/journal.pone.0020550.
    1. Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2014;25(4):198–213. doi: 10.1016/j.tcb.2014.11.006.
    1. Devaud C, et al. Tissues in different anatomical sites can sculpt and vary the tumor microenvironment to affect responses to therapy. Mol Ther. 2014;22(1):18–27. doi: 10.1038/mt.2013.219.
    1. Shah SP, et al. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature. 2009;461(7265):809–813. doi: 10.1038/nature08489.
    1. Ding L, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–510. doi: 10.1038/nature10738.
    1. Kroigard AB, et al. Clonal expansion and linear genome evolution through breast cancer progression from pre-invasive stages to asynchronous metastasis. Oncotarget. 2015;6(8):5634–5649. doi: 10.18632/oncotarget.3111.
    1. Brastianos PK, Cahill DP. Management of brain metastases in the era of targeted and immunomodulatory therapies. Oncology. 2015;29(4):261–263.
    1. Saunus JM, et al. Integrated genomic and transcriptomic analysis of human brain metastases identifies alterations of potential clinical significance. J Pathol. 2015;237(3):363–378. doi: 10.1002/path.4583.
    1. Luga V, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 2012;151(7):1542–1556. doi: 10.1016/j.cell.2012.11.024.
    1. Pukrop T, et al. Wnt 5a signaling is critical for macrophage-induced invasion of breast cancer cell lines. Proc Natl Acad Sci USA. 2006;103(14):5454–5459. doi: 10.1073/pnas.0509703103.
    1. Brastianos PK, et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 2015;5(11):1164–1177. doi: 10.1158/-15-0369.
    1. Paik PK, et al. Next-generation sequencing of stage IV squamous cell lung cancers reveals an association of PI3 K aberrations and evidence of clonal heterogeneity in patients with brain metastases. Cancer Discov. 2015;5(6):610–621. doi: 10.1158/-14-1129.

Source: PubMed

3
Subscribe