Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer

Aurélie S Cazet, Mun N Hui, Benjamin L Elsworth, Sunny Z Wu, Daniel Roden, Chia-Ling Chan, Joanna N Skhinas, Raphaël Collot, Jessica Yang, Kate Harvey, M Zahied Johan, Caroline Cooper, Radhika Nair, David Herrmann, Andrea McFarland, Niantao Deng, Manuel Ruiz-Borrego, Federico Rojo, José M Trigo, Susana Bezares, Rosalía Caballero, Elgene Lim, Paul Timpson, Sandra O'Toole, D Neil Watkins, Thomas R Cox, Michael S Samuel, Miguel Martín, Alexander Swarbrick, Aurélie S Cazet, Mun N Hui, Benjamin L Elsworth, Sunny Z Wu, Daniel Roden, Chia-Ling Chan, Joanna N Skhinas, Raphaël Collot, Jessica Yang, Kate Harvey, M Zahied Johan, Caroline Cooper, Radhika Nair, David Herrmann, Andrea McFarland, Niantao Deng, Manuel Ruiz-Borrego, Federico Rojo, José M Trigo, Susana Bezares, Rosalía Caballero, Elgene Lim, Paul Timpson, Sandra O'Toole, D Neil Watkins, Thomas R Cox, Michael S Samuel, Miguel Martín, Alexander Swarbrick

Abstract

The cellular and molecular basis of stromal cell recruitment, activation and crosstalk in carcinomas is poorly understood, limiting the development of targeted anti-stromal therapies. In mouse models of triple negative breast cancer (TNBC), Hedgehog ligand produced by neoplastic cells reprograms cancer-associated fibroblasts (CAFs) to provide a supportive niche for the acquisition of a chemo-resistant, cancer stem cell (CSC) phenotype via FGF5 expression and production of fibrillar collagen. Stromal treatment of patient-derived xenografts with smoothened inhibitors (SMOi) downregulates CSC markers expression and sensitizes tumors to docetaxel, leading to markedly improved survival and reduced metastatic burden. In the phase I clinical trial EDALINE, 3 of 12 patients with metastatic TNBC derived clinical benefit from combination therapy with the SMOi Sonidegib and docetaxel chemotherapy, with one patient experiencing a complete response. These studies identify Hedgehog signaling to CAFs as a novel mediator of CSC plasticity and an exciting new therapeutic target in TNBC.

Conflict of interest statement

Novartis funded a part of the study. The authors declare no other competing interests.

Figures

Fig. 1
Fig. 1
Malignant epithelial cells with increased self-renewal properties are located adjacent to the stroma of Hh-expressing cancers. a Scheme depicting the purification of epithelial and mixed stromal populations from disaggregated M6 murine tumor models. b Expression of genes significantly downregulated (blue) or upregulated (red) by RNA-Seq analysis (cut-off log2 (Fold Change) ≥ 2; vertical lines) in the epithelium of M6-Hh tumors compared to the epithelium of M6-Ctrl or M6-Hh tumors treated with the SMOi, GDC-0449 (100 mg/kg/bid), plotted against FDR values (horizontal lines indicate −log (FDR) > 2). Each symbol represents the transcriptome from five biological replicates per treatment group. c GSEA analysis reveals significant enrichment for genes encoding stemness and invasion in M6-Hh primary cells (FDR q value < 0.05). d Heat map showing relative CSC genes expression in the epithelium of M6-Hh tumors compared to M6-Ctrl and M6-Hh tumors + SMOi. Data show normalized row Z-score (n = 5 biological replicates for each treatment group). e Representative FACS dot plots showing the expression of CSC markers CD61 and CD29 within the EpCAM+/GFP+/CD24+ population of M6-Ctrl and M6-Hh tumors (n = 3 biological replicates per group). f Relative expression of key stemness genes in M6-Ctrl and M6-Hh whole tumors ± SMOi (100 mg/kg/bid). n = 3 biological replicates per treatment group; statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. g Kaplan−Meier and h tumor penetrance curves of mice injected with 250 primary M6-Ctrl (blue) or M6-Hh cells (violet); n = 10 biological replicates. Six tumors from the M6-Hh and two tumors from the M6-Ctrl models were detectable, respectively. Statistical significance was determined using log-rank test. i Primary M6-Ctrl and M6-Hh tumor cells were isolated by FACS and transplanted at various dilutions into recipient mice. Limiting dilution analysis demonstrating higher in vivo tumor-forming capacity in M6-Hh cells compared to M6-Ctrl cells. n = 10 mice per condition. j Representative images and quantification of CK6-progenitor and phospho-Histone H3-positive cancer cells at the tumor−stromal interface of M6 tumor models. Scale bars: 100 μm for CK6 and 200 μm for phospho-Histone H3. n = 3 biological replicates per treatment group. Statistical significance was determined using unpaired two-tailed Student’s t test; *P < 0.05; **P < 0.01. Bars represent mean ± s.e.m.
Fig. 2
Fig. 2
Hh-activated stroma defines a novel transcriptional signature robustly associated with poor-prognosis in TNBC. a Expression of genes significantly downregulated (blue) or upregulated (red) (cut-off log2 (Fold Change) ≥ 2; vertical lines) in the stroma of M6-Hh tumors compared to the stroma of M6-Ctrl or M6-Hh tumors treated with SMOi (100 mg/kg/bid), plotted against FDR values (horizontal lines indicate −log (FDR) > 2). Each symbol represents the transcriptome of the stromal population from five biological replicates per treatment group. b RNA-Seq analysis reveals significant enrichment of GO groups related to ECM organization and production in M6-Hh tumor stroma (FDR q value < 0.05). c Heat maps demonstrating relative expression levels of gene-sets defined by RNA-Seq analysis. Genes highlighted in red represent those that are directly involved in ECM production. Data show normalized row Z-score (n = 5 biological replicates for each treatment group). d Kaplan−Meier curves of overall survival in unstratified breast cancer patients and in patients with basal, TNBC. Blue and red lines represent low and high Hh-stromal gene signature expression (HSGS), respectively. Statistical significance was determined using the Log-rank test; ***P < 0.001
Fig. 3
Fig. 3
Paracrine Hedgehog signaling at cellular resolution. a Freshly isolated M6-Ctrl (blue), M6-Hh (magenta) and M6-Hh tumors + SMOi (100 mg/kg/bid; pink violin plot) were captured using 10x Chromium technology and the Cell Ranger Single Cell Software Suite 2.0 was used to perform demultiplexing, barcode processing, and single-cell 3′gene counting. t-SNE plot shows the subcellular clusters present in the breast TME of M6 tumors. Unless stated, P values are highly significant (Supplementary Data 2). b Single-cell RNA-Seq analysis reveals significant enrichment of GSEA groups related to ECM organization and production specifically in the CAF population of M6-Hh tumors (ECM-related processes highlighted in red; FDR q value < 0.05)
Fig. 4
Fig. 4
High fibrillar collagen content resulting from Hh pathway activation promotes mechano-signaling and breast cancer stemness. ad Concomitant expression analysis of collagen content and organization, integrin/focal adhesion activation and CSC-like characteristics at the tumor−stromal interface of M6 tumor models ± SMOi (100 mg/kg/bid; n= 3 biological replicates). a Representative multiphoton SHG imaging (scale bars: 100 μm) and quantitative analysis of collagen abundance. Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. Corresponding graphs comparing fiber orientation (top right panel) and quantifying GLCM (bottom right panel). Unpaired two-tailed nonparametric Mann–Whitney U test was used for determining statistical significance across distributions. For GLCM analysis, statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. b Collagen I and III deposition detected and quantified by picrosirius red staining. Scale bars, 200 μm. Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. c Representative immunofluorescence images and quantification of phospho-FAK and d CK6 expression. Scale bars, 100 μm. Statistical significance was determined using Kruskal−Wallis test. e M6-Ctrl and M6-Hh single cells were embedded and grown for 12 days in 3D Alginate IPNs containing increasing concentrations of collagen. Quantification of the number of colonies was normalized to the mean colony count in 0% collagen IPNs. n = 3 biological replicates with at least six technical replicates per condition. Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. Representative phase contrast images of M6 colonies on polyacrylamide substrata after 12 days of culture. Scale bars: 100 μm. f Relative mRNA expression of the CSC markers Id3, Igtb3, and Krt6b in M6-Ctrl and M6-Hh cells cultured within 3D alginate-collagen I IPNs. n = 3 biological replicates with six technical replicates per experiment. Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Bars represent mean ± s.e.m.
Fig 5
Fig 5
Hh-activated CAFs form a reversible, chemo-resistant CSC niche via FGF pathway activity. a RT-qPCR analysis of Fgf5 expression in M6 whole tumors. n = 3 biological replicates per treatment group with three technical replicates per assay. Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. b Expression of Fgf5 at single-cell resolution in M6 tumor models. Freshly isolated M6-Ctrl (blue), M6-Hh (magenta), and M6-Hh tumors + SMOi (100 mg/kg/bid; violin dots) were captured using 10x Chromium technology. t-SNE plot represents the subcellular clusters present in the breast TME of M6 tumors. A sole subset of Hh-activated CAFs exhibits robust expression of Fgf5 in M6-Hh tumors. c Representative immunohistochemistry staining for phospho-FGFR on M6 tumors. Scale bars: 100 μm. Quantification of phospho-FGFR-positive cancer cells at the tumor−stromal interface. n = 4 biological replicates per treatment group; statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. d Relative mRNA expression of CSC markers Id3 and Sox10 in M6-Ctrl cells treated with DMSO (vehicle; blue) or recombinant FGF5 (red) in vitro. n = 3 biological replicates with three technical replicates per experiment; statistical significance was determined using unpaired two-tailed Student’s t test. e Primary and secondary tumorsphere formation of M6-Ctrl cells treated with DMSO (vehicle; blue) or recombinant FGF5 (red). Sphere Formation Efficiency (SFE) values in % are mean ± s.e.m.; n = 3 biological replicates with three technical replicates per tumorsphere assay. Representative phase contrast micrographs of M6-Ctrl spheres upon recombinant FGF5 stimulation. Scale bars: 100 μm. f Cell viability of M6-Ctrl and M6-Hh cells treated with indicated agents (n = 5 biological replicates with six technical replicates each). Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d.; *P < 0.05; **P < 0.01; ***P < 0.001. Bars represent mean ± s.e.m.
Fig. 6
Fig. 6
Efficacy of long-term SMOi-combined therapy in preclinical models. ac Concomitant expression analysis of collagen content and organization, integrin pathway activation and CSC-like characteristics at the tumor−stromal interface of the TNBC HCI-002 PDX model treated with vehicle (red) or the SMO inhibitor NVP-LDE225 (80 mg/kg/day; blue) (n = 5 biological replicates). a Representative imaging (scale bars: 100 μm) and quantitative analysis of collagen abundance (left panel). Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. Corresponding graphs comparing fiber orientation (middle) and quantifying GLCM analysis (right panel) in the vehicle and SMOi-treated models. Unpaired two-tailed nonparametric Mann–Whitney U test was used for determining statistical significance across distributions. b Collagen I and III deposition detected and quantified by picrosirius red staining. Scale bars, 200 μm. Statistical significance was determined using unpaired two-tailed Student’s t test with equal s.d. c Representative immunofluorescence images and quantification of concomitant phospho-FAK (green) and the human CSC marker ALDH1 (red) expression in the HCI-002 model. The white arrows illustrate examples of co-staining within the epithelial population. Scale bars, 100 μm. n = 3 biological replicates. Statistical significance was determined using Kruskal−Wallis test. df TNBC HCI-002 PDX model treated with vehicle (blue), SMO inhibitor (NVP-LDE225; 80 mg/kg/day; magenta), chemotherapy (docetaxel; 15 mg/kg/week; dark orange) or NVP-LDE225 (80 mg/kg/day) + docetaxel (15 mg/kg/week; orange line) (n = 7 mice per treatment group). d Tumor growth curves. Statistical significance was determined using unpaired Student’s t test. e Percentage of mice with detectable metastases in the lung, liver, and axillary lymph node in each treatment group. Statistical significance was determined using the Fisher’s exact test. f Kaplan−Meier curves of mice overall survival of each treatment group. Statistical significance was determined using Log-rank test of NVP-LDE225 + docetaxel vs. docetaxel; *P < 0.05; ***P < 0.001; ****P < 0.0001. Bars on the graphs represent mean ± s.e.m.
Fig. 7
Fig. 7
Phase I clinical trial of docetaxel and SMO inhibitor, NVP-LDE225 (EDALINE) in patients with advanced TNBC. a Representative computed tomography (CT) images from a patient with complete radiological response. The 54-year-old postmenopausal woman was diagnosed with recurrent metastatic TNBC in the lungs with one measurable lesion on the right upper lobe (red arrow) and several nonmeasurable lesions (blue arrows). Therapeutic response was evaluated according to RECIST criteria version 1.1. The remaining structure seen in the right upper lobe corresponds to the azygos vein (violet arrow). b Representative HH and GLI1 immunostaining of treatment-naive tumor specimens from patients enrolled in the EDALINE trial. The left panel is representative of patients with high HPAS (characterized by high epithelial HH ligand and high stromal GLI1 expression) while the right panel represents low/intermediate epithelial HH and low stromal GLI1 expression. Scale bars: 100 μm. c Representative immunohistochemistry staining for phospho-FGFR, collagen deposition depicted by SHG imaging and concomitant phospho-FAK and ALDH1 stem cell marker expression in treatment-naive tumor specimens with high HPAS from the EDALINE trial. The left panel represents tissue derived from the patient who experienced a complete clinical response, the middle is from the patient with stable disease and the right panel represents tissue from the patient with high HPAS who progressed on the prescribed regimen. Scale bars: 100 μm. The white arrows illustrate examples of co-staining. d Graphical summary: Paracrine Hh signaling in TNBC drives a reversible stem-like, chemo-resistant phenotype via FGF signaling and ECM remodeling. CAFs represent the primary cells of the breast TME that respond to Hh ligand stimulation. Hh-activated CAFs enhance ECM collagen deposition and express FGF5 to establish a supportive niche for chemo-resistant cancer stem cell maintenance. This study strongly highlights a novel rational approach targeting both the tumor cells and their surrounding signaling support using SMO inhibitors to overcome chemoresistance in patients with TNBC

References

    1. Perrimon N, Pitsouli C, Shilo BZ. Signaling mechanisms controlling cell fate and embryonic patterning. Cold Spring Harb. Perspect. Biol. 2012;4:a005975. doi: 10.1101/cshperspect.a005975.
    1. Wiseman BS, Werb Z. Stromal effects on mammary gland development and breast cancer. Science. 2002;296:1046–1049. doi: 10.1126/science.1067431.
    1. Chuang PT, Kawcak T, McMahon AP. Feedback control of mammalian Hedgehog signaling by the Hedgehog-binding protein, Hip1, modulates Fgf signaling during branching morphogenesis of the lung. Genes Dev. 2003;17:342–347. doi: 10.1101/gad.1026303.
    1. Hui M, et al. The Hedgehog signalling pathway in breast development, carcinogenesis and cancer therapy. Breast Cancer Res. 2013;15:203. doi: 10.1186/bcr3401.
    1. Amakye D, Jagani Z, Dorsch M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat. Med. 2013;19:1410–1422. doi: 10.1038/nm.3389.
    1. O’Toole SA, et al. Hedgehog overexpression is associated with stromal interactions and predicts for poor outcome in breast cancer. Cancer Res. 2011;71:4002–4014. doi: 10.1158/0008-5472.CAN-10-3738.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Levental KR, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139:891–906. doi: 10.1016/j.cell.2009.10.027.
    1. Laklai H, et al. Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat. Med. 2016;22:497–505. doi: 10.1038/nm.4082.
    1. Finak G, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat. Med. 2008;14:518–527. doi: 10.1038/nm1764.
    1. Farmer P, et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat. Med. 2009;15:68–74. doi: 10.1038/nm.1908.
    1. Valenti G, et al. Cancer stem cells regulate cancer-associated fibroblasts via activation of Hedgehog signaling in mammary gland tumors. Cancer Res. 2017;77:2134–2147. doi: 10.1158/0008-5472.CAN-15-3490.
    1. Heller E, et al. Hedgehog signaling inhibition blocks growth of resistant tumors through effects on tumor microenvironment. Cancer Res. 2012;72:897–907. doi: 10.1158/0008-5472.CAN-11-2681.
    1. Lim E, et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat. Med. 2009;15:907–913. doi: 10.1038/nm.2000.
    1. Vaillant F, et al. The mammary progenitor marker CD61/beta3 integrin identifies cancer stem cells in mouse models of mammary tumorigenesis. Cancer Res. 2008;68:7711–7717. doi: 10.1158/0008-5472.CAN-08-1949.
    1. Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347:70–78. doi: 10.1016/j.jim.2009.06.008.
    1. Bu W, et al. Keratin 6a marks mammary bipotential progenitor cells that can give rise to a unique tumor model resembling human normal-like breast cancer. Oncogene. 2011;30:4399–4409. doi: 10.1038/onc.2011.147.
    1. Hynes RO, Naba A. Overview of the matrisome—an inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 2012;4:a004903. doi: 10.1101/cshperspect.a004903.
    1. Cancer Genome Atlas, N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412.
    1. Peiris-Pages M, Sotgia F, Lisanti MP. Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget. 2015;6:10728–10745. doi: 10.18632/oncotarget.3828.
    1. Huo CW, et al. High mammographic density is associated with an increase in stromal collagen and immune cells within the mammary epithelium. Breast Cancer Res. 2015;17:79. doi: 10.1186/s13058-015-0592-1.
    1. Keikhosravi A, Bredfeldt JS, Sagar AK, Eliceiri KW. Second-harmonic generation imaging of cancer. Methods Cell Biol. 2014;123:531–546. doi: 10.1016/B978-0-12-420138-5.00028-8.
    1. Mayorca-Guiliani AE, et al. ISDoT: in situ decellularization of tissues for high-resolution imaging and proteomic analysis of native extracellular matrix. Nat. Med. 2017;23:890–898. doi: 10.1038/nm.4352.
    1. Cicchi R, et al. Scoring of collagen organization in healthy and diseased human dermis by multiphoton microscopy. J. Biophotonics. 2010;3:34–43. doi: 10.1002/jbio.200910062.
    1. Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials. 1999;20:45–53. doi: 10.1016/S0142-9612(98)00107-0.
    1. Branco da Cunha C, et al. Influence of the stiffness of three-dimensional alginate/collagen-I interpenetrating networks on fibroblast biology. Biomaterials. 2014;35:8927–8936. doi: 10.1016/j.biomaterials.2014.06.047.
    1. Chaudhuri O, et al. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 2014;13:970–978. doi: 10.1038/nmat4009.
    1. O’Reilly EA, et al. The fate of chemoresistance in triple negative breast cancer (TNBC) BBA Clin. 2015;3:257–275. doi: 10.1016/j.bbacli.2015.03.003.
    1. DeRose YS, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat. Med. 2011;17:1514–1520. doi: 10.1038/nm.2454.
    1. Volk-Draper L, et al. Paclitaxel therapy promotes breast cancer metastasis in a TLR4-dependent manner. Cancer Res. 2014;74:5421–5434. doi: 10.1158/0008-5472.CAN-14-0067.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Malanchi I, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011;481:85–89. doi: 10.1038/nature10694.
    1. Lawson DA, et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature. 2015;526:131–135. doi: 10.1038/nature15260.
    1. Pang R, et al. A subpopulation of CD26+cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell. 2010;6:603–615. doi: 10.1016/j.stem.2010.04.001.
    1. Chen J, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488:522–526. doi: 10.1038/nature11287.
    1. Li X, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl. Cancer Inst. 2008;100:672–679. doi: 10.1093/jnci/djn123.
    1. Yu F, et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007;131:1109–1123. doi: 10.1016/j.cell.2007.10.054.
    1. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501:328–337. doi: 10.1038/nature12624.
    1. Mani SA, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–715. doi: 10.1016/j.cell.2008.03.027.
    1. Chaffer CL, et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 2013;154:61–74. doi: 10.1016/j.cell.2013.06.005.
    1. Samuel MS, et al. Actomyosin-mediated cellular tension drives increased tissue stiffness and beta-catenin activation to induce epidermal hyperplasia and tumor growth. Cancer Cell. 2011;19:776–791. doi: 10.1016/j.ccr.2011.05.008.
    1. Ibbetson SJ, Pyne NT, Pollard AN, Olson MF, Samuel MS. Mechanotransduction pathways promoting tumor progression are activated in invasive human squamous cell carcinoma. Am. J. Pathol. 2013;183:930–937. doi: 10.1016/j.ajpath.2013.05.014.
    1. Pang MF, et al. Tissue stiffness and hypoxia modulate the integrin-linked kinase ILK to control breast cancer stem-like cells. Cancer Res. 2016;76:5277–5287. doi: 10.1158/0008-5472.CAN-16-0579.
    1. Sherratt MJ, McConnell JC, Streuli CH. Raised mammographic density: causative mechanisms and biological consequences. Breast Cancer Res. 2016;18:45. doi: 10.1186/s13058-016-0701-9.
    1. McConnell JC, et al. Increased peri-ductal collagen micro-organization may contribute to raised mammographic density. Breast Cancer Res. 2016;18:5. doi: 10.1186/s13058-015-0664-2.
    1. Alvero AB, et al. Stem-like ovarian cancer cells can serve as tumor vascular progenitors. Stem Cells. 2009;27:2405–2413. doi: 10.1002/stem.191.
    1. Spike BT, et al. A mammary stem cell population identified and characterized in late embryogenesis reveals similarities to human breast cancer. Cell Stem Cell. 2012;10:183–197. doi: 10.1016/j.stem.2011.12.018.
    1. Dravis C, et al. Sox10 regulates stem/progenitor and mesenchymal cell states in mammary epithelial cells. Cell Rep. 2015;12:2035–2048. doi: 10.1016/j.celrep.2015.08.040.
    1. Fillmore CM, et al. Estrogen expands breast cancer stem-like cells through paracrine FGF/Tbx3 signaling. Proc. Natl. Acad. Sci. USA. 2010;107:21737–21742. doi: 10.1073/pnas.1007863107.
    1. Su SC, et al. CD10(+) GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell. 2018;172:841-+. doi: 10.1016/j.cell.2018.01.009.
    1. Babina IS, Turner NC. Advances and challenges in targeting FGFR signalling in cancer. Nat. Rev. Cancer. 2017;17:318–332. doi: 10.1038/nrc.2017.8.
    1. Wilkinson SE, et al. Hedgehog signaling is active in human prostate cancer stroma and regulates proliferation and differentiation of adjacent epithelium. Prostate. 2013;73:1810–1823. doi: 10.1002/pros.22720.
    1. Lasorella A, Benezra R, Iavarone A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat. Rev. Cancer. 2014;14:77–91. doi: 10.1038/nrc3638.
    1. Bain G, et al. Regulation of the helix-loop-helix proteins, E2A and Id3, by the Ras-ERK MAPK cascade. Nat. Immunol. 2001;2:165–171. doi: 10.1038/84273.
    1. Wilgus TA. Growth factor-extracellular matrix interactions regulate wound repair. Adv. Wound Care (New Rochelle) 2012;1:249–254. doi: 10.1089/wound.2011.0344.
    1. Kouros-Mehr H, Werb Z. Candidate regulators of mammary branching morphogenesis identified by genome-wide transcript analysis. Dev. Dyn. 2006;235:3404–3412. doi: 10.1002/dvdy.20978.
    1. Lewis MT, et al. The Gli2 transcription factor is required for normal mouse mammary gland development. Dev. Biol. 2001;238:133–144. doi: 10.1006/dbio.2001.0410.
    1. Lu P, Ewald AJ, Martin GR, Werb Z. Genetic mosaic analysis reveals FGF receptor 2 function in terminal end buds during mammary gland branching morphogenesis. Dev. Biol. 2008;321:77–87. doi: 10.1016/j.ydbio.2008.06.005.
    1. Pond AC, et al. Fibroblast growth factor receptor signaling is essential for normal mammary gland development and stem cell function. Stem Cells. 2013;31:178–189. doi: 10.1002/stem.1266.
    1. Luhr I, et al. Mammary fibroblasts regulate morphogenesis of normal and tumorigenic breast epithelial cells by mechanical and paracrine signals. Cancer Lett. 2012;325:175–188. doi: 10.1016/j.canlet.2012.06.014.
    1. Rhim AD, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747. doi: 10.1016/j.ccr.2014.04.021.
    1. Shin K, et al. Hedgehog signaling restrains bladder cancer progression by eliciting stromal production of urothelial differentiation factors. Cancer Cell. 2014;26:521–533. doi: 10.1016/j.ccell.2014.09.001.
    1. Gerling M, et al. Stromal Hedgehog signalling is downregulated in colon cancer and its restoration restrains tumour growth. Nat. Commun. 2016;7:12321. doi: 10.1038/ncomms12321.
    1. Ohlund D, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017;214:579–596.
    1. Chen WJ, et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat. Commun. 2014;5:3472. doi: 10.1038/ncomms4472.
    1. Hirata E, et al. Intravital imaging reveals how BRAF inhibition generates drug-tolerant microenvironments with high integrin beta1/FAK signaling. Cancer Cell. 2015;27:574–588. doi: 10.1016/j.ccell.2015.03.008.
    1. Lotti F, et al. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A. J. Exp. Med. 2013;210:2851–2872. doi: 10.1084/jem.20131195.
    1. Holzer RG, et al. Development and characterization of a progressive series of mammary adenocarcinoma cell lines derived from the C3(1)/SV40 Large T-antigen transgenic mouse model. Breast Cancer Res. Treat. 2003;77:65–76. doi: 10.1023/A:1021175931177.
    1. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat. Biotechnol. 2014;32:896–902. doi: 10.1038/nbt.2931.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616.
    1. Macosko EZ, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015;161:1202–1214. doi: 10.1016/j.cell.2015.05.002.
    1. Gao J, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013;6:pl1. doi: 10.1126/scisignal.2004088.
    1. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat. Protoc. 2008;3:1101–1108. doi: 10.1038/nprot.2008.73.
    1. Zhu AJ, Scott MP. Incredible journey: how do developmental signals travel through tissue? Genes Dev. 2004;18:2985–2997. doi: 10.1101/gad.1233104.
    1. Marei H, et al. Differential Rac1 signalling by guanine nucleotide exchange factors implicates FLII in regulating Rac1-driven cell migration. Nat. Commun. 2016;7:10664. doi: 10.1038/ncomms10664.

Source: PubMed

3
S'abonner