Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Pharmacological targeting of netrin-1 inhibits EMT in cancer

Abstract

Epithelial-to-mesenchymal transition (EMT) regulates tumour initiation, progression, metastasis and resistance to anti-cancer therapy1,2,3,4,5,6,7. Although great progress has been made in understanding the role of EMT and its regulatory mechanisms in cancer, no therapeutic strategy to pharmacologically target EMT has been identified. Here we found that netrin-1 is upregulated in a primary mouse model of skin squamous cell carcinoma (SCC) exhibiting spontaneous EMT. Pharmacological inhibition of netrin-1 by administration of NP137, a netrin-1-blocking monoclonal antibody currently used in clinical trials in human cancer (ClinicalTrials.gov identifier NCT02977195), decreased the proportion of EMT tumour cells in skin SCC, decreased the number of metastases and increased the sensitivity of tumour cells to chemotherapy. Single-cell RNA sequencing revealed the presence of different EMT states, including epithelial, early and late hybrid EMT, and full EMT states, in control SCC. By contrast, administration of NP137 prevented the progression of cancer cells towards a late EMT state and sustained tumour epithelial states. Short hairpin RNA knockdown of netrin-1 and its receptor UNC5B in EPCAM+ tumour cells inhibited EMT in vitro in the absence of stromal cells and regulated a common gene signature that promotes tumour epithelial state and restricts EMT. To assess the relevance of these findings to human cancers, we treated mice transplanted with the A549 human cancer cell line—which undergoes EMT following TGFβ1 administration8,9—with NP137. Netrin-1 inhibition decreased EMT in these transplanted A549 cells. Together, our results identify a pharmacological strategy for targeting EMT in cancer, opening up novel therapeutic interventions for anti-cancer therapy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Targeting netrin-1 inhibits EMT.
Fig. 2: Targeting netrin-1 reduces metastasis and sensitizes tumour cells to chemotherapy in skin SCC.
Fig. 3: Pharmacological inhibition of netrin-1 inhibits late EMT and promotes epithelial tumour states.
Fig. 4: Pharmacological inhibition of netrin-1 inhibits late EMT and promotes epithelial differentiation trajectories of tumour cells.
Fig. 5: Netrin-1 and UNCnc5B knockdown inhibits EMT and promotes the epithelial state.
Fig. 6: Anti-netrin-1 therapy inhibits EMT in human cancer cells.

Similar content being viewed by others

Data availability

All raw sequence data for mouse RNA-seq, single cell RNA-seq and 10x Visium have been deposited in the Gene Expression Omnibus under the accession number GSE234267Source data are provided with this paper.

References

  1. Ye, X. & Weinberg, R. A. Epithelial-mesenchymal plasticity: a central regulator of cancer progression. Trends Cell Biol. 25, 675–686 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Shibue, T. & Weinberg, R. A. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 14, 611–629 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Lambert, A. W. & Weinberg, R. A. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev. Cancer 21, 325–338 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Puisieux, A., Brabletz, T. & Caramel, J. Oncogenic roles of EMT-inducing transcription factors. Nat. Cell. Biol. 16, 488–494 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Brabletz, S., Schuhwerk, H., Brabletz, T. & Stemmler, M. P. Dynamic EMT: a multi-tool for tumor progression. EMBO J. 40, e108647 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pastushenko, I. & Blanpain, C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 29, 212–226 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Kasai, H., Allen, J. T., Mason, R. M., Kamimura, T. & Zhang, Z. TGF-β1 induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir Res. 6, 56 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Kim, J. H. et al. Transforming growth factor β1 induces epithelial-to-mesenchymal transition of A549 cells. J. Korean Med. Sci. 22, 898–904 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Voon, D. C., Huang, R. Y., Jackson, R. A. & Thiery, J. P. The EMT spectrum and therapeutic opportunities. Mol. Oncol. 11, 878–891 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Tan, T. Z. et al. Epithelial–mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO Mol. Med. 6, 1279–1293 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lapouge, G. et al. Skin squamous cell carcinoma propagating cells increase with tumour progression and invasiveness. EMBO J. 31, 4563–4575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Latil, M. et al. Cell-type-specific chromatin states differentially prime squamous cell carcinoma tumor-initiating cells for epithelial to mesenchymal transition. Cell Stem Cell 20, 191–204.e195 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Pastushenko, I. et al. Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Paradisi, A. et al. Combining chemotherapeutic agents and netrin-1 interference potentiates cancer cell death. EMBO Mol. Med. 5, 1821–1834 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Paradisi, A. et al. Netrin-1 up-regulation in inflammatory bowel diseases is required for colorectal cancer progression. Proc. Natl Acad. Sci. USA 106, 17146–17151 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Paradisi, A. et al. NF-κB regulates netrin-1 expression and affects the conditional tumor suppressive activity of the netrin-1 receptors. Gastroenterology 135, 1248–1257 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Fitamant, J. et al. Netrin-1 expression confers a selective advantage for tumor cell survival in metastatic breast cancer. Proc. Natl Acad. Sci. USA 105, 4850–4855 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Delloye-Bourgeois, C. et al. Netrin-1 acts as a survival factor for aggressive neuroblastoma. J. Exp. Med. 206, 833–847 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sung, P. J. et al. Cancer-associated fibroblasts produce netrin-1 to control cancer cell plasticity. Cancer Res. 79, 3651–3661 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Park, K. W. et al. The axonal attractant Netrin-1 is an angiogenic factor. Proc. Natl Acad. Sci. USA 101, 16210–16215 (2004).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Arakawa, H. Netrin-1 and its receptors in tumorigenesis. Nat. Rev. Cancer 4, 978–987 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Brisset, M., Grandin, M., Bernet, A., Mehlen, P. & Hollande, F. Dependence receptors: new targets for cancer therapy. EMBO Mol. Med. 13, e14495 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hao, W. et al. The pan-cancer landscape of netrin family reveals potential oncogenic biomarkers. Sci. Rep. 10, 5224 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dumartin, L. et al. Netrin-1 mediates early events in pancreatic adenocarcinoma progression, acting on tumor and endothelial cells. Gastroenterology 138, 1595–1606 (2010). 1606 e1591-1598.

    Article  CAS  PubMed  Google Scholar 

  26. Kefeli, U. et al. Netrin-1 in cancer: potential biomarker and therapeutic target? Tumour Biol. 39, 1010428317698388 (2017).

    Article  PubMed  Google Scholar 

  27. Haerinck, J. & Berx, G. Partial EMT takes the lead in cancer metastasis. Dev. Cell 56, 3174–3176 (2021).

    Article  CAS  PubMed  Google Scholar 

  28. Simeonov, K. P. et al. Single-cell lineage tracing of metastatic cancer reveals selection of hybrid EMT states. Cancer Cell 39, 1150–1162.e1159 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yang, J. et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 21, 341–352 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Jin, X. et al. Netrin-1 interference potentiates epithelial-to-mesenchymal transition through the PI3K/AKT pathway under the hypoxic microenvironment conditions of non-small cell lung cancer. Int. J. Oncol. 54, 1457–1465 (2019).

  31. Zhang, X. et al. Netrin-1 elicits metastatic potential of non-small cell lung carcinoma cell by enhancing cell invasion, migration and vasculogenic mimicry via EMT induction. Cancer Gene Ther. 25, 18–26 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Yan, W. et al. Netrin-1 induces epithelial-mesenchymal transition and promotes hepatocellular carcinoma invasiveness. Dig. Dis. Sci. 59, 1213–1221 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Han, P. et al. Netrin-1 promotes cell migration and invasion by down-regulation of BVES expression in human hepatocellular carcinoma. Am. J. Cancer Res. 5, 1396–1409 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. Revenco, T. et al. Context dependency of epithelial-to-mesenchymal transition for metastasis. Cell Rep. 29, 1458–1468.e1453 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. DeConti, R. C. Chemotherapy of squamous cell carcinoma of the skin. Semin. Oncol. 39, 145–149 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Khansur, T. & Kennedy, A. Cisplatin and 5-fluorouracil for advanced locoregional and metastatic squamous cell carcinoma of the skin. Cancer 67, 2030–2032 (1991).

    Article  CAS  PubMed  Google Scholar 

  37. Chen, Q. Y. et al. miR-206 regulates cisplatin resistance and EMT in human lung adenocarcinoma cells partly by targeting MET. Oncotarget 7, 24510–24526 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Grandin, M. et al. Structural decoding of the Netrin-1/UNC5 interaction and its therapeutical implications in cancers. Cancer Cell 29, 173–185 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Mak, M. P. et al. A patient-derived, pan-cancer EMT signature identifies global molecular alterations and immune target enrichment following epithelial-to-mesenchymal transition. Clin. Cancer Res. 22, 609–620 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  44. Tuveson, D. A. et al. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell 5, 375–387 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat. Genet. 29, 418–425 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Boussouar, A. et al. Netrin-1 and its receptor DCC are causally implicated in melanoma progression. Cancer Res. 80, 747–756 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zappia, L. & Oshlack, A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience 7, giy083 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Franzen, O., Gan, L. M. & Björkegren, J. L. M. PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data. Database 2019, baz046 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Buttner, M., Ostner, J., Muller, C. L., Theis, F. J. & Schubert, B. scCODA is a Bayesian model for compositional single-cell data analysis. Nat. Commun. 12, 6876 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Elyada, E. et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Saelens, W., Cannoodt, R., Todorov, H. & Saeys, Y. A comparison of single-cell trajectory inference methods. Nat. Biotechnol. 37, 547–554 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank the ULB animal facility; ULB genomic core facility (F. Libert and A. Lefort) for bulk RNA-seq and scRNA-seq; the Gilles Thomas bioinformatic platform; Centre de Recherche en Cancérologie de Lyon, Fondation Synergie Lyon cancer for the spatial transcriptomic sequencing; S. Bottieau for technical assistance; F. Lavial for Unc5b shRNA; and R. Derynck for the A549 cell line. J.L. is supported by NETRIS Pharma. I.P. is supported by FNRS and WELBIO. S.V. is supported by a PhD fellowship for Strategic Basic Research (1S93320N) from the Research Foundation Flanders (FWO). C. Decaestecker is a senior Research Associate with Fond National de la Recherche Scientifique (FNRS, Brussels, Belgium). DIAPath and the Department of Pathology are supported by the Fonds Yvonne Boël. The CMMI is supported by the European Regional Development Fund and the Walloon region (Wallonia-biomed; grant no. 411132–957270; project “CMMI-ULB” support the Center for Microscopy and Molecular Imaging and its DIAPath department). C.B. is supported by WELBIO, FNRS, TELEVIE, Fondation Contre le Cancer, ULB Foundation, Foundation Baillet Latour, FNRS/FWO EOS (40007513) and the European Research Council (AdvGrant 885093). This work was also supported by institutional grants from CNRS, University of Lyon1, Centre Léon Bérard and from the Ligue Contre le Cancer, INCA, ARC Sign’it and ANR (nos. ANR-10-LABX- 0061, ANR-17-CONV-0002 and ANR-18-RHUS-0009).

Author information

Authors and Affiliations

Authors

Contributions

J.L., I.P., S.V. and C.B. designed the experiments and performed data analysis. J.L. and I.P. performed most of the biological experiments. S.V. performed most of bioinformatic analysis for single-cell sequencing. N.R., Y.S. and A.S. helped with bioinformatic analysis. J.V.H. helped with 10x single-cell sequencing. R.M.S. helped with RNAscope analysis. V.M., A. Boinet., S.S., S.L., S.G. and S.B. helped with cell culture experiments, immunostaining, blocking antibody injection and follow-up with the mice. I.S., J.A., E.Z., C. Decaestecker. and A.C. performed immunostaining and quantification of EMT in human cancer samples. B.D., M.B. and N.B. performed biological in vivo and in vitro experiments on Ishikawa endometrial cell lines. C.S. and D.V. performed bioanalysis from TCGA. C. Dubois performed FACS sorting. T.V. helped and supervised the single-cell data analysis. P.M. and A. Bernet helped with the design of the experiments, data analysis and provided NP137 antibody. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Patrick Mehlen, Agnès Bernet or Cédric Blanpain.

Ethics declarations

Competing interests

A. Bernet and P.M. declare a conflict of interest as founders and shareholders of NETRIS Pharma. J.L., P.M., B.D., M.B. and N.B. declare a conflict of interest as employees of NETRIS Pharma. A. Bernet. and N.R. declare a conflict of interest as consultants for NETRIS. T.V. is co-inventor on licensed patents WO/2011/157846 (Methods for Haplotyping Single Cells), WO/2014/053664 (High-Throughput Genotyping by Sequencing Low Amounts of Genetic Material) and WO/2015/028576 (Haplotyping and Copy Number Typing Using Polymorphic Variant Allelic Frequencies).

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Strategy to study the impact of Netrin-1 on EMT in mouse skin SCCs.

a, Mouse model of skin SCC allowing the expression of KrasG12D, YFP, p53 deletion and overexpression of human NETRIN-1 in hair follicle stem cells and their progeny using Lgr5CreER. b, Relative mRNA expression of Ntn1 in EPCAM- control LKPR (n = 5) and LKPR-NTN1 (n = 8) skin SCC defined by qRT-PCR (data are normalized to Tbp gene, mean ± s.e.m., two tailed Mann-Whitney U test). c, Western blot analysis of Netrin-1 expression in EPCAM- control LKPR and LKPR-NTN1 skin SCC TCs. d, FACS plots showing the gating strategy used to FACS-isolate or to analyse the proportion of YFP+/EPCAM+ and EPCAM tumour cells. e, Drawing showing the experimental strategy of NP137 administration after Tamoxifen induction in Lgr5CreER/KrasLSL-G12D/p53fl/fl/Rosa26-YFP+/+ mice. IP, intraperitoneal.

Source data

Extended Data Fig. 2 Single cell analysis of the cellular composition of control and NP137-treated skin SCCs.

a,b Uniform Manifold Approximation and Projection (UMAP) plot for control (a) and NP137 -treated skin SCC (b) coloured by the identified cell types. c,d, UMAP plot for control (c) and NP137-treated skin SCC (d) coloured by the sample of origin for each cell. CAFs, cancer-associated fibroblasts.

Extended Data Fig. 3 Annotation of the cell types found by single cell RNA-seq in control and NP137-treated skin SCCs.

a, UMAP plots coloured by normalized Yfp and Epcam expression in the control tumours. Gene expression values are visualized as colour gradient with grey indicating no expression and red indicating the maximum expression. b, UMAP plots coloured by normalized Yfp and Epcam in NP137-treated samples. c, UMAP plots coloured by the activity of modules containing the mouse-specific marker genes of the different cell types including CAFs, Macrophages, Neutrophils, Endothelial cells and T cells obtained from the PanglaoDB database in control samples (left) and anti-Netrin-1 treated samples (right). Module activity visualized as a colour gradient with blue indicating no expression and yellow indicating maximum activity. d, UMAP plots coloured by normalized Pdgfra, Acta2, Pecam1, Cd3d, Ptprc, Itgam, Cd86 and Cxcr2 expression in the control samples (left) and NP137-treated samples (right). CAFs, cancer-associated fibroblasts. e, UMAP plot coloured by normalized Ntn1 expression in control condition.

Extended Data Fig. 4 Impact of anti-Netrin antibody administration on the cellular composition of skin SCCs.

a,b, Uniform Manifold Approximation and Projection (UMAP) plots coloured by the cell type labels obtained from the analysis of the microenvironment for the integration of all the samples in total (a) and split per sample (b), respectively. c, Boxplot depicting the proportions of the different cell types for the 4 samples, split by their condition. The boxplots are coloured by their condition, and the individual measurements are visualized as red dots. The centre line, top and bottom of the boxplots represent respectively the median, 25th and 75th percentile and whiskers are 1.5 × IQR. Significant proportion changes are indicated by FDR < 0.2. d, barplot depicting the relative log fold change of the relative abundance of the different cell types after NP137-treated samples compared to the pericytes. Bars are coloured according to their cell type. e,f, UMAP plot of the CAFS subclustering, coloured by the identified seven subclusters and the sample the cell originated from, respectively. g, Boxplot depicting the proportions of the different CAF subclusters for the 4 samples, split by their condition. The boxplots are coloured by their condition, and the individual measurements are visualized as red dots. The centre line, top and bottom of the boxplots represent respectively the median, 25th and 75th percentile and whiskers are 1.5 × IQR. h. barplot depicting the relative log fold change of the relative abundance of the different CAF subclusters after NP137 treatment compared to the glycolysis CAFs subcluster. i, Co-immunostaining of YFP and Vimentin in control (top) (n = 5 tumours) and NP137- treated skin SCC (bottom) (n = 5 tumours) that defines YFP-/VIM+ CAFs as cells (Scale bars, 20 μm).

Extended Data Fig. 5 Expression of markers of the different EMT states in control and NP137-treated skin SCCs.

a, b, UMAP plots coloured by normalized gene expression values for the indicated genes in the control (a) and treated samples (b). Gene expression values are visualized as colour gradient with grey indicating no expression and red indicating the maximum expression. Circles represent TCs groups with a different degree of EMT based on the expression of Epcam, Krt14, Krt8, Vim, Pdgfra (green: Epcam+/Krt14+/Vim− as epithelial state; orange: Epcam−/Krt14+/Vim+ as early hybrid EMT state; red: Epcam−/Krt14−/Krt8+/Vim+ as late hybrid EMT state; dark red: Epcam−/Krt14−/Krt8−/Vim+ as late full EMT state expressing Pdgfra and Aqp1). c, Barplot depicting the relative log fold change of the relative abundance of the different EMT states after NP137-treatment compared to the early hybrid state. Significant proportion changes are indicated by FDR < 0.2.

Extended Data Fig. 6 Histological analysis of the control and NP137-treated tumors.

a–d, Haematoxylin and Eosin staining showing the control (n = 1) (a,b) or NP137-treated (n = 1) (c,d) tumour skin SCC analysed in Visium spatial transcriptomic method. The annotated areas represent the EMT states previously defined by the expression of Epcam, Krt14, Krt8 and Vim (1: epithelial, 2: early hybrid, 3, late hybrid, 4: full late EMT) (scale bars in a, c, 500 μm, scale bars in b, 20 μm).

Extended Data Fig. 7 Analysis of NP137 treatment on tumour growth, EMT and migration in endometrial human cancer cell line.

a, Tumor growth quantification of human Ishikawa endometrial carcinoma cells grafted in nude mice treated with either control (n = 9) or NP137 (n = 9) (mean ± s.e.m., 2-way ANOVA). b, Relative mRNA expression of epithelial markers CDH1, MUC1 and HOOK1 by qRT-PCR in Ishikawa human cells grafted in nude mice treated with control (n = 7) or NP137 (n = 8) (data are normalized to HPRT gene, mean +/− s.e.m., two tailed Mann-Whitney U test). c, Percentage of migrated Ishikawa cells treated with NP137 relative to the migration of control condition through serum deprived culture medium complemented with 2.5% Matrigel between 5 and 24 h of invasion. (n = 3) (mean ± s.e.m, two tailed t test).

Source data

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lengrand, J., Pastushenko, I., Vanuytven, S. et al. Pharmacological targeting of netrin-1 inhibits EMT in cancer. Nature 620, 402–408 (2023). https://doi.org/10.1038/s41586-023-06372-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06372-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer