The BRAF-inhibitor PLX4720 inhibits CXCL8 secretion in BRAFV600E mutated and normal thyroid cells: a further anti-cancer effect of BRAF-inhibitors

Francesca Coperchini, Laura Croce, Marco Denegri, Oriana Awwad, Samuel Tata Ngnitejeu, Marina Muzza, Valentina Capelli, Francesco Latrofa, Luca Persani, Luca Chiovato, Mario Rotondi, Francesca Coperchini, Laura Croce, Marco Denegri, Oriana Awwad, Samuel Tata Ngnitejeu, Marina Muzza, Valentina Capelli, Francesco Latrofa, Luca Persani, Luca Chiovato, Mario Rotondi

Abstract

CXCL8 is a chemokine secreted by normal and thyroid cancer cells with proven tumor-promoting effects. The presence of BRAFV600E mutation is associated with a more aggressive clinical behavior and increased ability to secrete CXCL8 by papillary-thyroid-cancer cells. Aim of this study was to test the effect of the BRAF-inhibitor (PLX4720) on the basal and TNF-α-induced CXCL8 secretions in BRAFV600E mutated (BCPAP, 8305C, 8505C), in RET/PTC rearranged (TPC-1) thyroid-cancer-cell-lines and in normal-human-thyrocytes (NHT). Cells were incubated with increasing concentrations of PLX4720 alone or in combination with TNF-α for 24-hours. CXCL8 concentrations were measured in the cell supernatants. PLX4720 dose-dependently inhibited the basal and the TNF-α-induced CXCL8 secretions in BCPAP (F: 14.3, p < 0.0001 for basal and F: 12.29 p < 0.0001 for TNF-α), 8305C (F: 407.9 p < 0.0001 for basal and F: 5.76 p < 0.0001 for TNF-α) and 8505C (F:55.24 p < 0.0001 for basal and F: 42.85 p < 0.0001 for TNF-α). No effect was found in TPC-1 (F: 1.8, p = 0.134 for basal; F: 1.6, p = 0.178 for TNF-α). In NHT an inhibitory effect was found only at the highest concentration of PLX4720 (F: 13.13 p < 0.001 for basal and F: 2.5 p < 0.01 for TNF-α). Cell migration assays showed that PLX4720 reduced both basal and CXCL8-induced cell migration in BCPAP, 8305C, 8505C and NHT but not in TPC-1 cells. These results constitutes the first demonstration that PLX4720 is able to inhibit the secretion of CXCL8 in BRAFV600E mutated thyroid cancer cells indicating that, at least some, of the anti-tumor activities of PLX4720 could be exerted through a lowering of CXCL8 in the thyroid-cancer-microenvironment.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Panel A PLX4720 inhibit the basal CXCL8 secretion in BCPAP (ANOVA F: 14.3; p p < 0.001) being significant only at the higher concentration of 10 µM (Post Hoc analysis by Bonferroni *p < 0.01 vs. basal). Panel F PLX4720 inhibit the TNF-𝛼-stimulated CXCL8 secretion in BCPAP (ANOVA F: 12.29 p < 0.0001), the inhibitory effect was significant starting by 1 10 µM (3.13; t anPost Hoc analysis by Bonferroni *p < 0.001 vs. TNF-𝛼). Panel G PLX4720 inhibit the TNF-α-stimulated CXCL8 secretion in 8305C (ANOVA F: 5.76 p < 0.0001), the inhibitory effect started from 1 rom Post Hoc analysis by Bonferroni *p < 0.001 vs. TNF-𝛼). Panel H PLX4720 inhibit the TNF-𝛼-stimulated CXCL8 secretion in 8505C (ANOVA F: 42.85 p < 0.0001), the inhibitory effect started from 0.1 rom Post Hoc analysis by Bonferroni *p < 0.001 vs. TNF-𝛼, **p < 0.001 vs. 0.1 ni *p < Panel I TNF-𝛼-stimulated CXCL8 secretion was not inhibited by PLX4720 in TPC-1 cell lines at any concentrations (ANOVA F: 1.6, p = 1.78). Panel E TNF-𝛼-stimulated CXCL8 secretion was inhibited by PLX4720 in NHT (ANOVA F: 2.5; p < 0.01) being significant only at the higher concentration of10 µM (Post Hoc analysis by Bonferroni *p < 0.001 vs. TNF-α alone).
Figure 2
Figure 2
CXCL8 concentrations progressively increased throughout the 72-h time course in untreated thyroid cells. Panel A BCPAP cells (ANOVA: F = 13.39; p 

Figure 3

Time course of PLX4720 inhibitory…

Figure 3

Time course of PLX4720 inhibitory effect on CXCL8 secretion. No significant change in…

Figure 3
Time course of PLX4720 inhibitory effect on CXCL8 secretion. No significant change in the inhibitory power of PLX4720 at a given concentration was found throughout the 72-h time course. Panel A BCPAP (ANOVAs: PLX4720 0.1 µM F: 0.6; NS; 1 µM F: 1.7; NS; 2 µM F: 1.7; p = 0.2; 5 µM F: 2.7; p = 0.7; 10 µM F: 1.0, NS). Panel B NHT (ANOVAs: PLX4720 0.1 µM F:1.7, NS; 1 µM F:2.8; NS; 2 µM F: 3; NS; 5 µM F: 1; NS; 10 µM F: 2.3, NS). Panel C 8305C (ANOVAs: PLX4720 0.1 µM F:1.6, NS; 1 µM F: 2.6; NS; 2 µM F: 0.5; NS; 5 µM F: 0.4; NS; 10 µM F: 1.3, NS). Panel D 8505C (ANOVAs: PLX4720 0.1 µM F:0.02, NS; 1 µM F: 0.2; NS; 2 µM F: 0.1; NS; 5 µM F: 1.2; NS; 10 µM F: 2.9, NS).

Figure 4

Effect of PLX4720 on the…

Figure 4

Effect of PLX4720 on the basal and rh-CXCL8-induced migration in BCPAP, 8305C, 8505C,…

Figure 4
Effect of PLX4720 on the basal and rh-CXCL8-induced migration in BCPAP, 8305C, 8505C, NHT and TPC-1 thyroid cancer cells. Representative images and the respective histograms after 16 hours of migration within the trans-well migration chamber system in BCPAP, 8305C, 8505C, NHT and TPC-1 (Panels A–E respectively). Panel (A) in BCPAP the treatment with PLX4720 10 µM reduced the basal migration (ANOVA F = 27.9, p Post Hoc °p < 0.005 vs. basal). The incubation of BCPAP with rh-CXCL8 produced a significant increase of BCPAP migration (Post Hoc *p < 0.0001 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the BCPAP migration induced by rh-CXCL8 (Post Hoc#p < 0.001 vs. CXCL8). Panel (B) in 8305C, the treatment with PLX4720 10 µM reduced the basal migration of 8305C (ANOVA F = 161.7, p < 0.0001; Post Hoc °p < 0.005 vs. basal). The incubation of 8305C with rh-CXCL8 produced a significant increase of 8305C migration (Post Hoc *p < 0.0001 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the 8305C migration induced by rh-CXCL8 (Post Hoc#p < 0.0001 vs. CXCL8). Panel (C) in 8505C the treatment with PLX4720 10 µM reduced the basal migration of 8505C (ANOVA F = 21.6, p < 0.0001; Post Hoc °p < 0.005 vs. basal). The incubation of 8505C with rh-CXCL8 produced a significant increase of 8505C migration (Post Hoc *p < 0.0001 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the 8505C migration induced by rh-CXCL8 (Post Hoc#p < 0.0001 vs. CXCL8). Panel (D) in NHT, the treatment with PLX4720 10 µM reduced the basal migration of NHT (ANOVA F = 25.3, p < 0.0001; Post Hoc °p < 0.005 vs. basal). The incubation of NHT with rh-CXCL8 produced a significant increase of NHT migration (Post Hoc *p < 0.05 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the NHT migration induced by rh-CXCL8 (Post Hoc#p < 0.0001 vs. CXCL8). Panel (E) in TPC-1 the treatment with PLX4720 10 µM did not reduce the basal migration of TPC-1 (ANOVA F = 4.4, p < 0.01), Post Hoc NS vs. basal). The incubation of TPC-1 with rh-CXCL8 produced a significant increase of TPC-1 migration (Post Hoc *p < 0.05 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM did not reduce the TPC-1 migration induced by rh-CXCL8 (Post Hoc # NS vs. CXCL8). Bar graphs show the corresponding analysis of migrated cells on the lower side of the transwell filter. Basal migration was conventionally estimated as 100%.
Figure 3
Figure 3
Time course of PLX4720 inhibitory effect on CXCL8 secretion. No significant change in the inhibitory power of PLX4720 at a given concentration was found throughout the 72-h time course. Panel A BCPAP (ANOVAs: PLX4720 0.1 µM F: 0.6; NS; 1 µM F: 1.7; NS; 2 µM F: 1.7; p = 0.2; 5 µM F: 2.7; p = 0.7; 10 µM F: 1.0, NS). Panel B NHT (ANOVAs: PLX4720 0.1 µM F:1.7, NS; 1 µM F:2.8; NS; 2 µM F: 3; NS; 5 µM F: 1; NS; 10 µM F: 2.3, NS). Panel C 8305C (ANOVAs: PLX4720 0.1 µM F:1.6, NS; 1 µM F: 2.6; NS; 2 µM F: 0.5; NS; 5 µM F: 0.4; NS; 10 µM F: 1.3, NS). Panel D 8505C (ANOVAs: PLX4720 0.1 µM F:0.02, NS; 1 µM F: 0.2; NS; 2 µM F: 0.1; NS; 5 µM F: 1.2; NS; 10 µM F: 2.9, NS).
Figure 4
Figure 4
Effect of PLX4720 on the basal and rh-CXCL8-induced migration in BCPAP, 8305C, 8505C, NHT and TPC-1 thyroid cancer cells. Representative images and the respective histograms after 16 hours of migration within the trans-well migration chamber system in BCPAP, 8305C, 8505C, NHT and TPC-1 (Panels A–E respectively). Panel (A) in BCPAP the treatment with PLX4720 10 µM reduced the basal migration (ANOVA F = 27.9, p Post Hoc °p < 0.005 vs. basal). The incubation of BCPAP with rh-CXCL8 produced a significant increase of BCPAP migration (Post Hoc *p < 0.0001 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the BCPAP migration induced by rh-CXCL8 (Post Hoc#p < 0.001 vs. CXCL8). Panel (B) in 8305C, the treatment with PLX4720 10 µM reduced the basal migration of 8305C (ANOVA F = 161.7, p < 0.0001; Post Hoc °p < 0.005 vs. basal). The incubation of 8305C with rh-CXCL8 produced a significant increase of 8305C migration (Post Hoc *p < 0.0001 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the 8305C migration induced by rh-CXCL8 (Post Hoc#p < 0.0001 vs. CXCL8). Panel (C) in 8505C the treatment with PLX4720 10 µM reduced the basal migration of 8505C (ANOVA F = 21.6, p < 0.0001; Post Hoc °p < 0.005 vs. basal). The incubation of 8505C with rh-CXCL8 produced a significant increase of 8505C migration (Post Hoc *p < 0.0001 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the 8505C migration induced by rh-CXCL8 (Post Hoc#p < 0.0001 vs. CXCL8). Panel (D) in NHT, the treatment with PLX4720 10 µM reduced the basal migration of NHT (ANOVA F = 25.3, p < 0.0001; Post Hoc °p < 0.005 vs. basal). The incubation of NHT with rh-CXCL8 produced a significant increase of NHT migration (Post Hoc *p < 0.05 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM significantly inhibited the NHT migration induced by rh-CXCL8 (Post Hoc#p < 0.0001 vs. CXCL8). Panel (E) in TPC-1 the treatment with PLX4720 10 µM did not reduce the basal migration of TPC-1 (ANOVA F = 4.4, p < 0.01), Post Hoc NS vs. basal). The incubation of TPC-1 with rh-CXCL8 produced a significant increase of TPC-1 migration (Post Hoc *p < 0.05 vs. basal). The co-incubation with rh-CXCL8 and PLX4720 10 µM did not reduce the TPC-1 migration induced by rh-CXCL8 (Post Hoc # NS vs. CXCL8). Bar graphs show the corresponding analysis of migrated cells on the lower side of the transwell filter. Basal migration was conventionally estimated as 100%.

References

    1. Davies H, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766.
    1. Crawford S, et al. A novel B-RAF inhibitor blocks interleukin-8 (IL-8) synthesis in human melanoma xenografts, revealing IL-8 as a potential pharmacodynamic biomarker. Mol Cancer Ther. 2008;7:492–499. doi: 10.1158/1535-7163.MCT-07-0307.
    1. Elisei R, et al. BRAF(V600E) mutation and outcome of patients with papillary thyroid carcinoma: a 15-year median follow-up study. J Clin Endocrinol Metab. 2008;93:3943–3949. doi: 10.1210/jc.2008-0607.
    1. Fugazzola L, et al. BRAF mutations in an Italian cohort of thyroid cancers. Clin Endocrinol (Oxf) 2004;61:239–243. doi: 10.1111/j.1365-2265.2004.02089.x.
    1. Nucera C, Lawler J, Parangi S. BRAF(V600E) and microenvironment in thyroid cancer: a functional link to drive cancer progression. Cancer Res. 2011;71:2417–2422. doi: 10.1158/0008-5472.CAN-10-3844.
    1. Xing M. BRAF mutation in papillary thyroid cancer: pathogenic role, molecular bases, and clinical implications. Endocr Rev. 2007;28:742–762. doi: 10.1210/er.2007-0007.
    1. Kondo T, et al. Enhanced B-Raf protein expression is independent of V600E mutant status in thyroid carcinomas. Hum Pathol. 2007;38:1810–1818. doi: 10.1016/j.humpath.2007.04.014.
    1. Frasca F, et al. BRAF(V600E) mutation and the biology of papillary thyroid cancer. Endocr Relat Cancer. 2008;15:191–205. doi: 10.1677/ERC-07-0212.
    1. Xing M, et al. Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA. 2013;309:1493–1501. doi: 10.1001/jama.2013.3190.
    1. Ikenoue T, et al. Functional analysis of mutations within the kinase activation segment of B-Raf in human colorectal tumors. Cancer Res. 2003;63:8132–8137.
    1. Michaloglou C, Vredeveld LC, Mooi WJ, Peeper DS. BRAF(E600) in benign and malignant human tumours. Oncogene. 2008;27:877–895. doi: 10.1038/sj.onc.1210704.
    1. Liu D, Liu Z, Condouris S, Xing M. BRAF V600E maintains proliferation, transformation, and tumorigenicity of BRAF-mutant papillary thyroid cancer cells. J Clin Endocrinol Metab. 2007;92:2264–2271. doi: 10.1210/jc.2006-1613.
    1. Cunha LL, Marcello MA, Ward LS. The role of the inflammatory microenvironment in thyroid carcinogenesis. Endocr Relat Cancer. 2014;21:R85–R103. doi: 10.1530/ERC-13-0431.
    1. French JD, et al. Tumor-associated lymphocytes and increased FoxP3+ regulatory T cell frequency correlate with more aggressive papillary thyroid cancer. J Clin Endocrinol Metab. 2010;95:2325–2333. doi: 10.1210/jc.2009-2564.
    1. Modi J, Patel A, Terrell R, Tuttle RM, Francis GL. Papillary thyroid carcinomas from young adults and children contain a mixture of lymphocytes. J Clin Endocrinol Metab. 2003;88:4418–4425. doi: 10.1210/jc.2003-030342.
    1. Rotondi M, Coperchini F, Chiovato L. CXCL8 in thyroid disease: from basic notions to potential applications in clinical practice. Cytokine Growth Factor Rev. 2013;24:539–546. doi: 10.1016/j.cytogfr.2013.08.001.
    1. Coperchini F, et al. Normal human thyroid cells, BCPAP, and TPC-1 thyroid tumor cell lines display different profile in both basal and TNF-α-induced CXCL8 secretion. Endocrine. 2016;54:123–128. doi: 10.1007/s12020-015-0764-x.
    1. Rotondi M, Coperchini F, Latrofa F, Chiovato L. Role of Chemokines in Thyroid Cancer Microenvironment: Is CXCL8 the Main Player? Front Endocrinol (Lausanne) 2018;9:314. doi: 10.3389/fendo.2018.00314.
    1. Fang W, et al. Tumor-associated macrophages promote the metastatic potential of thyroid papillary cancer by releasing CXCL8. Carcinogenesis. 2014;35:1780–1787. doi: 10.1093/carcin/bgu060.
    1. Liotti F, et al. Interleukin-8, but not the Related Chemokine CXCL1, Sustains an Autocrine Circuit Necessary for the Properties and Functions of Thyroid Cancer Stem Cells. Stem Cells. 2017;35:135–146. doi: 10.1002/stem.2492.
    1. Liotti F, Visciano C, Melillo RM. Inflammation in thyroid oncogenesis. Am J Cancer Res. 2012;2:286–297.
    1. Rotondi M, et al. Effect of Interferon-γ on the Basal and the TNFα-Stimulated Secretion of CXCL8 in Thyroid Cancer Cell Lines Bearing Either the RET/PTC Rearrangement Or the BRAF V600e Mutation. Mediators Inflamm. 2016;2016:8512417. doi: 10.1155/2016/8512417.
    1. Visciano C, et al. Mast cells induce epithelial-to-mesenchymal transition and stem cell features in human thyroid cancer cells through an IL-8-Akt-Slug pathway. Oncogene. 2015;34:5175–5186. doi: 10.1038/onc.2014.441.
    1. Rotondi M, et al. Type I and type II interferons inhibit both basal and tumor necrosis factor-α-induced CXCL8 secretion in primary cultures of human thyrocytes. J Interferon Cytokine Res. 2013;33:508–513. doi: 10.1089/jir.2012.0080.
    1. Rotondi M, Coperchini F, Pignatti P, Magri F, Chiovato L. Metformin reverts the secretion of CXCL8 induced by TNF-α in primary cultures of human thyroid cells: an additional indirect anti-tumor effect of the drug. J Clin Endocrinol Metab. 2015;100:E427–432. doi: 10.1210/jc.2014-3045.
    1. Nucera C, et al. B-Raf(V600E) and thrombospondin-1 promote thyroid cancer progression. Proc Natl Acad Sci USA. 2010;107:10649–10654. doi: 10.1073/pnas.1004934107.
    1. Tsai J, et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci USA. 2008;105:3041–3046. doi: 10.1073/pnas.0711741105.
    1. Cartlidge RA, et al. Oncogenic BRAF(V600E) inhibits BIM expression to promote melanoma cell survival. Pigment Cell Melanoma Res. 2008;21:534–544. doi: 10.1111/j.1755-148X.2008.00491.x.
    1. Salerno P, et al. Cytostatic activity of adenosine triphosphate-competitive kinase inhibitors in BRAF mutant thyroid carcinoma cells. J Clin Endocrinol Metab. 2010;95:450–455. doi: 10.1210/jc.2009-0373.
    1. Nucera C, et al. A novel orthotopic mouse model of human anaplastic thyroid carcinoma. Thyroid. 2009;19:1077–1084. doi: 10.1089/thy.2009.0055.
    1. Rotondi M, et al. Interferon-γ and tumor necrosis factor-α sustain secretion of specific CXC chemokines in human thyrocytes: a first step toward a differentiation between autoimmune and tumor-related inflammation? J Clin Endocrinol Metab. 2013;98:308–313. doi: 10.1210/jc.2012-2555.
    1. Bae YH, Park K. Targeted drug delivery to tumors: myths, reality and possibility. J Control Release. 2011;153:198–205. doi: 10.1016/j.jconrel.2011.06.001.
    1. Nucera C, et al. Targeting BRAFV600E with PLX4720 displays potent antimigratory and anti-invasive activity in preclinical models of human thyroid cancer. Oncologist. 2011;16:296–309. doi: 10.1634/theoncologist.2010-0317.
    1. Montero-Conde C, et al. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov. 2013;3:520–533. doi: 10.1158/-12-0531.
    1. Fernando RI, Castillo MD, Litzinger M, Hamilton DH, Palena C. IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Res. 2011;71:5296–5306. doi: 10.1158/0008-5472.CAN-11-0156.
    1. David, J. M., Dominguez, C., Hamilton, D. H. & Palena, C. The IL-8/IL-8R Axis: A Double Agent in Tumor Immune Resistance. Vaccines (Basel)4, 10.3390/vaccines4030022 (2016).
    1. Coperchini F, et al. TNF-α increases the membrane expression of the chemokine receptor CCR6 in thyroid tumor cells, but not in normal thyrocytes: potential role in the metastatic spread of thyroid cancer. Tumour Biol. 2016;37:5569–5575. doi: 10.1007/s13277-015-4418-7.
    1. Abbonante V, et al. Discoidin domain receptor 1 protein is a novel modulator of megakaryocyte-collagen interactions. J Biol Chem. 2013;288:16738–16746. doi: 10.1074/jbc.M112.431528.

Source: PubMed

Подписаться