Microenvironmental control of breast cancer subtype elicited through paracrine platelet-derived growth factor-CC signaling

Pernilla Roswall, Matteo Bocci, Michael Bartoschek, Hong Li, Glen Kristiansen, Sara Jansson, Sophie Lehn, Jonas Sjölund, Steven Reid, Christer Larsson, Pontus Eriksson, Charlotte Anderberg, Eliane Cortez, Lao H Saal, Christina Orsmark-Pietras, Eugenia Cordero, Bengt Kristian Haller, Jari Häkkinen, Ingrid J G Burvenich, Elgene Lim, Akira Orimo, Mattias Höglund, Lisa Rydén, Holger Moch, Andrew M Scott, Ulf Eriksson, Kristian Pietras, Pernilla Roswall, Matteo Bocci, Michael Bartoschek, Hong Li, Glen Kristiansen, Sara Jansson, Sophie Lehn, Jonas Sjölund, Steven Reid, Christer Larsson, Pontus Eriksson, Charlotte Anderberg, Eliane Cortez, Lao H Saal, Christina Orsmark-Pietras, Eugenia Cordero, Bengt Kristian Haller, Jari Häkkinen, Ingrid J G Burvenich, Elgene Lim, Akira Orimo, Mattias Höglund, Lisa Rydén, Holger Moch, Andrew M Scott, Ulf Eriksson, Kristian Pietras

Abstract

Breast tumors of the basal-like, hormone receptor-negative subtype remain an unmet clinical challenge, as there is high rate of recurrence and poor survival in patients following treatment. Coevolution of the malignant mammary epithelium and its underlying stroma instigates cancer-associated fibroblasts (CAFs) to support most, if not all, hallmarks of cancer progression. Here we delineate a previously unappreciated role for CAFs as determinants of the molecular subtype of breast cancer. We identified paracrine crosstalk between cancer cells expressing platelet-derived growth factor (PDGF)-CC and CAFs expressing the cognate receptors in human basal-like mammary carcinomas. Genetic or pharmacological intervention of PDGF-CC activity in mouse models of cancer resulted in conversion of basal-like breast cancers into a hormone receptor-positive state that enhanced sensitivity to endocrine therapy in previously resistant tumors. We conclude that specification of breast cancer to the basal-like subtype is under microenvironmental control and is therapeutically actionable.

Conflict of interest statement

Competing Financial Interest

KP, UE and PR are named inventors on PCT application# PCT/EP2016/077295 relating to the findings of the current study. KP, UE and AMS are shareholders of Paracrine Therapeutics that develop inhibitory agents to PDGF-CC.

Figures

Figure 1. Epithelial expression of PDGF-CC is…
Figure 1. Epithelial expression of PDGF-CC is associated with poor outcome in human breast carcinoma.
a-d, Immunostaining of normal breast tissue (a-b) or breast carcinoma (c-d) for PDGF-CC. Appropriate validation on >10 independent samples was performed to ensure reproducible staining pattern on human tissue. Dotted line represents the epithelium-stroma boundary and arrows identify tumor cells with particularly high expression of PDGF-CC on the invasive border. Scale bar, 40 µm. e, Kaplan-Meier analysis of breast cancer-specific survival dichotomized according to absence (negative, n=438 samples) or presence (positive, defined as a score of 1+ to 3+, n=452 samples) of PDGF-CC in tumor cells (for scoring scheme, see Supplementary Fig. 1a) of breast carcinomas from the Zürich cohort. ** p=0.002, log-rank test f, Average volume (with SEM) of tumors from 14 weeks old MMTV-PyMT mice (n=10 animals in each group). *** p=0.0001, two-sided, unpaired, equal variance Student’s t-test. g, Grading of tumors from 14 weeks old MMTV-PyMT mice (n=5 animals in each group). * Distribution of Pdgfc-/- vs Pdgfc+/- p=0.036, χ2 test. *** Distribution of Pdgfc-/- vs Pdgfc+/+ p=3*10-11 and distribution of Pdgfc+/- vs Pdgfc+/+ p=0.00052, χ2 test. h-k, Hematoxylin and eosin staining of tumors from MMTV-PyMT mice at 14 weeks of age. Scale bar, 500 µm. l, Average volume (with SEM) of tumors derived from MMTV-PyMT mice transplanted into the mammary fat pad of wt mice (n=21 animals in each group, composed of n=7 animals each from 3 independent experiments). *** p=6*10-6, two-sided, unpaired, equal variance Student’s t-test. m-n, Masson’s tri-chrome staining of tissue sections from tumors from 14 weeks old MMTV-PyMT mice. Representative pictures from the analysis of n=10 animals from each group are shown. Scale bar, 500 µm. o, Quantitative real-time PCR analysis of the average expression (with SEM) of markers for cancer-associated fibroblasts in tumor lysates of tumors from 14 weeks old MMTV-PyMT mice (n=3 animals in each group, analysis performed independently 3 times). ** Pdgfra, p=0.007; S100a4, p=0.003; Acta2, p=0.009 two-sided, unpaired, equal variance Student’s t-test.
Figure 2. Expression of PDGF-CC in breast…
Figure 2. Expression of PDGF-CC in breast carcinomas is associated with the basal-like molecular subtype and hormone receptor negativity.
a, Quantitative RT-PCR analysis of the luminal subtype marker Foxa1 in tumors from 14 weeks old MMTV-PyMT mice (n=5 animals in each group, analysis performed independently 3 times). *** p<0.001, two-sided, unpaired, equal variance Student’s t-test. b-c, Expression of FOXA1 (b) and PDGFC (c) in a panel of 51 breast cancer cell lines divided according to basal-like or luminal-like molecular subtype. d, Correlation between expression of FOXA1 and PDGFC in a panel of 51 breast cancer cell lines divided according to basal-like or luminal-like molecular subtype. Correlation coefficient and p-value from Pearson correlation analysis of n=26 basal cell lines and n=25 luminal cell lines. e, Expression of all PDGF family members in n=26 basal cell lines and n=25 luminal cell lines . Box represents the interquartile range, line represents the average expression and whiskers depict range of expression with statistical outliers indicated (>2 standard deviations from the mean). *** p=1*10-6, two-sided, unpaired Welch’s unequal variances t-test. f-k, Enrichment of luminal-defining transcription factors ESR1, FOXA1, and GATA3 binding sites, as derived from public datasets of ChIP-seq analysis in MCF7 cells, in the 1000 most up/down-regulated genes in Pdgfc-deficient PeRo-Lum1 cells compared to Pdgfc-proficient PeRo-Bas1 cells. Red, query signature; blue, n=105 randomly sampled gene lists of equal size to the query signature; green, average of the randomly sampled gene lists. The rank of the query gene list divided by the total number of re-sample instances was then used as a p-value for the probability to acquire the level of enrichment by chance. l-n, Correlation between expression of ERα and PDGF-CC in primary breast carcinomas (l, n=470 independent samples, *** p=2.1*10-14 independent samples, two-sided Jonckheere-Terpstra test for ordered alternatives), synchronous lymph nodes (m, n=132 independent samples, *** p=1.5*10-8 independent samples, two-sided Jonckheere-Terpstra test for ordered alternatives) or asynchronous distant replapses (n, n=29 independent samples, ** p=0.0015 independent samples, two-sided Jonckheere-Terpstra test for ordered alternatives) included in the Lund cohort. Box represents the interquartile range, line represents the median expression and whiskers depict 1.5x the height of the box. Circles and stars represent statistical outliers that fall outside of the whiskers and extreme outliers that fall outside of 3x the height of the box, respectively.
Figure 3. CAF-derived factors induced by PDGF-CC…
Figure 3. CAF-derived factors induced by PDGF-CC reduce the sensitivity of breast tumor cells to endocrine therapy.
a, Viability of PeRo-Lum1 luminal breast cancer cells derived from MMTV-PyMT; Pdgfc-/- mice in the presence of increasing concentrations of 4-OH-tamoxifen in medium conditioned by tumor cells (TC) or CAFs (the average (with SEM) of n=6 independent experiments is shown). ** p<0.01, *** p<0.001, (p-values for control vs STC1+HGF+IGFBP3 for each tamoxifen concentration (1-5 µM=0.0001, 0.00001, 0.00001, 0.00001, 0.005, respectively), two-way ANOVA with p-values from Bonferroni post-hoc test. b, Viability of PeRo-Lum1 cells in the presence of increasing concentrations of 4-OH-tamoxifen in medium conditioned by nothing or STC1, HGF and IGFBP3 (the average (with SEM) of n=6 independent experiments is shown). ** p<0.01, *** p<0.001 (p-values for control vs STC1+HGF+IGFBP3 for each tamoxifen concentration (1-5 µM=0.003, 0.0003, 0.00008, 0.0002, 0.005, respectively), two-way ANOVA with p-values from Bonferroni post-hoc test. c-e, Quantitative RT-PCR analysis of the average expression (with SEM) of Foxa1 (c, n=3 independent experiments), Esr1 (d, n=3 independent experiments) or Gata3 (e, n=2 independent experiments) in PeRo-Lum1 cells in the presence of combinations of STC1, HGF and IGFBP3. Foxa1 * p=0.03, ** p=0.002, *** p=0.0003, one-way ANOVA with p-values from Bonferroni post-hoc test. Esr1 * p=0.01, ** p=0.005, *** p=0.001, one-way ANOVA with p-values from Bonferroni post-hoc test. Gata3 * p=0.02, ** p=0.007, *** p=0.001, one-way ANOVA with p-values from Bonferroni post-hoc test. f-h, Immunostaining of tissue sections from tumors from 14 weeks old MMTV-PyMT mice for STC1 (f), HGF (g) or IGFBP3 (h). Immunostaining was performed n=3 independent times and representative images from n=5 tumors are shown. Scale bar, 100 µm. i, Pearson’s correlation analysis of factors denoting luminal-like (FOXA1, ESR1, GATA3) or basal-like (FOXC1, PDGFC) molecular subtype in luminal subtype breast carcinomas included in the TCGA cohort (n=1086 independent tumors included in analysis).
Figure 4. Genetic or pharmacological targeting of…
Figure 4. Genetic or pharmacological targeting of PDGF-CC induces expression of ERα and sensitizes tumors to endocrine therapy.
a-b, Treatment of wt mice transplanted orthotopically with tumors derived from 14 weeks old MMTV-PyMT; Pdgfc+/+ mice (a) or MMTV-PyMT; Pdgfc-/- mice (b) with oil vehicle (Pdgfc+/+ n=7 animals, Pdgfc-/- n=6 animals) or tamoxifen (Pdgfc-+/+ n=6 animals, Pdgfc-/- n=7 animals). Average tumor volume (with SEM) depicted. ** p=0.002, two-sided, unpaired, equal variance Student’s t-test. X-axis shows time after start of therapy. c-d, Immunostaining for ERα (brown) of tissue sections from the triple-negative patient-derived xenograft 12.58 from mice treated with control IgG or 6B3. Immunostaining performed n=3 independent times and representative images are shown. Scale bar, 40 µm. e, Quantification of the average ERα expression (with SEM) in patient-derived xenograft 12.58 from mice treated with control IgG or 6B3 (IgG, n=7 animals; 6B3, n=8 animals). *** p=7*10-7, χ2 test. f, Quantification of the average number of ERα-expressing cells (with SEM) in MDA-MB-231 tumors from mice treated with control IgG or 6B3 in combination with oil vehicle (IgG, n=10 animals; 6B3, n=12 animals) or tamoxifen (IgG, n=12 animals; 6B3, n=12 animals). ** p=0.003, two-sided, unpaired, equal variance Student’s t-test. g, Treatment of mice carrying orthotopically transplanted triple-negative patient-derived xenograft 14.32 with control IgG in combination with vehicle (n=10 animals) or tamoxifen (n=10 animals). Average tumor volume (with SEM) depicted. X-axis shows time after start of therapy. h, Treatment of mice carrying orthotopically transplanted MDA-MB-231 tumors with control IgG in combination with oil vehicle (n=10 animals) or tamoxifen (n=12 animals). Average tumor volume (with SEM) depicted. X-axis shows time after start of therapy. i, Treatment of mice carrying orthotopically transplanted triple-negative patient-derived xenograft 14.32 with the neutralizing PDGF-CC antibody 6B3 in combination with vehicle (n=10 animals) or tamoxifen (n=10 animals). Average tumor volume (with SEM) depicted. * p=0.03, two-sided, unpaired, equal variance Student’s t-test. X-axis shows time after start of therapy. j, Treatment of mice carrying orthotopically transplanted MDA-MB-231 tumors with the neutralizing PDGF-CC antibody 6B3 in combination with oil vehicle (n=12 animals) or tamoxifen (n=13 animals). Average tumor volume (with SEM) depicted. * p=0.025, two-sided, unpaired, equal variance Student’s t-test. X-axis shows time after start of therapy. k, Treatment of mice carrying orthotopically transplanted MCF7 tumors infected with lentivirus carrying empty vector or Pdgfc vector with oil vehicle (Vector, n=9 animals; Pdgfc, n=10 animals) or tamoxifen (Vector, n=10 animals, Pdgfc, n=8 animals). Average tumor volume (with SEM) depicted. * Vector + Tamoxifen vs Pdgfc + Tamoxifen, p=0.029, two-sided, unpaired, equal variance, Student’s t-test. X-axis shows time after start of therapy. l, Schematic diagram of the paracrine action of PDGF-CC in the breast tumor microenvironment. An active crosstalk between malignant cells and CAFs mediated through PDGF-CC results in specification of the molecular subtype and regulation of sensitivity to endocrine therapy.

References

    1. Perou CM, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752.
    1. Goldhirsch A, et al. Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the primary therapy of early breast cancer 2009. Ann Oncol. 2009;20:1319–1329.
    1. Sorlie T, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98:10869–10874.
    1. Goldhirsch A, et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013;24:2206–2223.
    1. Goldhirsch A, et al. Strategies for subtypes--dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol. 2011;22:1736–1747.
    1. Prat A, et al. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast. 2015;24(Suppl 2):S26–35.
    1. Ignatiadis M, Sotiriou C. Luminal breast cancer: from biology to treatment. Nat Rev Clin Oncol. 2013;10:494–506.
    1. Voduc KD, et al. Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 2010;28:1684–1691.
    1. Prat A, Perou CM. Deconstructing the molecular portraits of breast cancer. Mol Oncol. 2011;5:5–23.
    1. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309–322.
    1. Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res. 2010;316:1324–1331.
    1. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401.
    1. Cortez E, Roswall P, Pietras K. Functional subsets of mesenchymal cell types in the tumor microenvironment. Semin Cancer Biol. 2014;25:3–9.
    1. Augsten M. Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol. 2014;4:62.
    1. Kim HM, Jung WH, Koo JS. Expression of cancer-associated fibroblast related proteins in metastatic breast cancer: an immunohistochemical analysis. J Transl Med. 2015;13:222.
    1. Anderberg C, et al. Paracrine signaling by platelet-derived growth factor-CC promotes tumor growth by recruitment of cancer-associated fibroblasts. Cancer Res. 2009;69:369–378.
    1. Pietras K, Pahler J, Bergers G, Hanahan D. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med. 2008;5:e19.
    1. Li X, et al. PDGF-C is a new protease-activated ligand for the PDGF a-receptor. Nature Cell Biol. 2000;2:302–309.
    1. Cao R, et al. Angiogenesis stimulated by PDGF-CC, a novel member in the PDGF family, involves activation of PDGFR-aa and -ab receptors. FASEB J. 2002;16:1575–1583.
    1. Theurillat JP, et al. NY-ESO-1 protein expression in primary breast carcinoma and metastases: correlation with CD8+ T-cell and CD79a+ plasmacytic/B-cell infiltration. Int J Cancer. 2007;120:2411–2417.
    1. Falck AK, et al. Biomarker expression and St Gallen molecular subtype classification in primary tumours, synchronous lymph node metastases and asynchronous relapses in primary breast cancer patients with 10 years' follow-up. Breast Cancer Res Treat. 2013;140:93–104.
    1. Falck AK, et al. Analysis of and prognostic information from disseminated tumour cells in bone marrow in primary breast cancer: a prospective observational study. BMC Cancer. 2012;12:403.
    1. Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12:954–961.
    1. Lin EY, et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J Pathol. 2003;163:2113–2126.
    1. Ding H, et al. A specific requirement for PDGF-C in palate formation and PDGFR-alpha signaling. Nat Genet. 2004;36:1111–1116.
    1. Cancer Genome Atlas: N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
    1. Badve S, et al. FOXA1 expression in breast cancer--correlation with luminal subtype A and survival. Clin Cancer Res. 2007;13:4415–4421.
    1. Thorat MA, et al. Forkhead box A1 expression in breast cancer is associated with luminal subtype and good prognosis. J Clin Pathol. 2008;61:327–332.
    1. Neve RM, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10:515–527.
    1. Kong SL, Li G, Loh SL, Sung WK, Liu ET. Cellular reprogramming by the conjoint action of ERalpha, FOXA1, and GATA3 to a ligand-inducible growth state. Mol Syst Biol. 2011;7:526.
    1. Kojima Y, et al. Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci U S A. 2010;107:20009–20014.
    1. Ray PS, et al. FOXC1 is a potential prognostic biomarker with functional significance in basal-like breast cancer. Cancer Res. 2010;70:3870–3876.
    1. Chebil G, Bendahl PO, Idvall I, Ferno M. Comparison of immunohistochemical and biochemical assay of steroid receptors in primary breast cancer--clinical associations and reasons for discrepancies. Acta Oncol. 2003;42:719–725.
    1. Hammond ME, et al. American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 2010;28:2784–2795.
    1. Polyak K. Breast cancer: origins and evolution. J Clin Invest. 2007;117:3155–3163.
    1. Gupta PB, et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell. 2011;146:633–644.
    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.
    1. Molyneux G, Smalley MJ. The cell of origin of BRCA1 mutation-associated breast cancer: a cautionary tale of gene expression profiling. J Mammary Gland Biol Neoplasia. 2011;16:51–55.
    1. Liu S, et al. BRCA1 regulates human mammary stem/progenitor cell fate. Proc Natl Acad Sci U S A. 2008;105:1680–1685.
    1. Molyneux G, et al. BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell. 2010;7:403–417.
    1. Su Y, et al. Somatic Cell Fusions Reveal Extensive Heterogeneity in Basal-like Breast Cancer. Cell Rep. 2015;11:1549–1563.
    1. Yamamoto S, et al. JARID1B is a luminal lineage-driving oncogene in breast cancer. Cancer Cell. 2014;25:762–777.
    1. Bernardo GM, et al. FOXA1 represses the molecular phenotype of basal breast cancer cells. Oncogene. 2013;32:554–563.
    1. Sflomos G, et al. A Preclinical Model for ERalpha-Positive Breast Cancer Points to the Epithelial Microenvironment as Determinant of Luminal Phenotype and Hormone Response. Cancer Cell. 2016;29:407–422.
    1. Tam WL, et al. Protein kinase C alpha is a central signaling node and therapeutic target for breast cancer stem cells. Cancer Cell. 2013;24:347–364.
    1. Meng F, et al. PDGFRalpha and beta play critical roles in mediating Foxq1-driven breast cancer stemness and chemoresistance. Cancer Res. 2015;75:584–593.
    1. Jansson S, et al. The three receptor tyrosine kinases c-KIT, VEGFR2 and PDGFRalpha, closely spaced at 4q12, show increased protein expression in triple-negative breast cancer. PLoS One. 2014;9:e102176.
    1. Horikawa S, et al. PDGFRalpha plays a crucial role in connective tissue remodeling. Sci Rep. 2015;5 17948.
    1. Kim YJ, et al. MET is a potential target for use in combination therapy with EGFR inhibition in triple-negative/basal-like breast cancer. Int J Cancer. 2014;134:2424–2436.
    1. Ho-Yen CM, et al. C-Met in invasive breast cancer: is there a relationship with the basal-like subtype? Cancer. 2014;120:163–171.
    1. Ponzo MG, et al. Met induces mammary tumors with diverse histologies and is associated with poor outcome and human basal breast cancer. Proc Natl Acad Sci U S A. 2009;106:12903–12908.
    1. Graveel CR, et al. Met induces diverse mammary carcinomas in mice and is associated with human basal breast cancer. Proc Natl Acad Sci U S A. 2009;106:12909–12914.
    1. Gastaldi S, et al. Met signaling regulates growth, repopulating potential and basal cell-fate commitment of mammary luminal progenitors: implications for basal-like breast cancer. Oncogene. 2013;32:1428–1440.
    1. Marzec KA, Baxter RC, Martin JL. Targeting Insulin-Like Growth Factor Binding Protein-3 Signaling in Triple-Negative Breast Cancer. Biomed Res Int. 2015;2015 638526.
    1. Bissell MJ, Hines WC. Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med. 2011;17:320–329.
    1. Marsh T, Pietras K, McAllister SS. Fibroblasts as architects of cancer pathogenesis. Biochim Biophys Acta. 2013;1832:1070–1078.
    1. Gascard P, Tlsty TD. Carcinoma-associated fibroblasts: orchestrating the composition of malignancy. Genes Dev. 2016;30:1002–1019.
    1. Brechbuhl HM, et al. Fibroblast Subtypes Regulate Responsiveness of Luminal Breast Cancer to Estrogen. Clin Cancer Res. 2017;23:1710–1721.
    1. Ozdemir BC, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–734.
    1. Rhim AD, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747.
    1. Theurillat JP, et al. NY-ESO-1 protein expression in primary breast carcinoma and metastases: correlation with CD8+ T-cell and CD79a+ plasmacytic/B-cell infiltration. Int J Cancer. 2007;120:2411–2417.
    1. Falck AK, et al. Biomarker expression and St Gallen molecular subtype classification in primary tumours, synchronous lymph node metastases and asynchronous relapses in primary breast cancer patients with 10 years' follow-up. Breast Cancer Res Treat. 2013;140:93–104.
    1. Falck AK, et al. Analysis of and prognostic information from disseminated tumour cells in bone marrow in primary breast cancer: a prospective observational study. BMC Cancer. 2012;12:403.
    1. Chebil G, Bendahl PO, Idvall I, Ferno M. Comparison of immunohistochemical and biochemical assay of steroid receptors in primary breast cancer--clinical associations and reasons for discrepancies. Acta Oncol. 2003;42:719–725.
    1. Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12:954–961.
    1. Ding H, et al. A specific requirement for PDGF-C in palate formation and PDGFR-alpha signaling. Nat Genet. 2004;36:1111–1116.
    1. Kojima Y, et al. Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci U S A. 2010;107:20009–20014.
    1. Parker JS, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27:1160–1167.
    1. Neve RM, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10:515–527.
    1. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34:525–527.
    1. Pimentel H, Bray N, Puente S, Melsted P, Pachter L. Differential analysis of RNA-Seq incorporating quantification uncertainty. bioRxiv. 2016 doi: 10.1101/058164.
    1. Mei S, et al. Cistrome Data Browser: a data portal for ChIP-Seq and chromatin accessibility data in human and mouse. Nucleic Acids Res. 2017;45:D658–D662.
    1. Kong SL, Li G, Loh SL, Sung WK, Liu ET. Cellular reprogramming by the conjoint action of ERalpha, FOXA1, and GATA3 to a ligand-inducible growth state. Mol Syst Biol. 2011;7:526.
    1. Sikora-Wohlfeld W, Ackermann M, Christodoulou EG, Singaravelu K, Beyer A. Assessing computational methods for transcription factor target gene identification based on ChIP-seq data. PLoS Comput Biol. 2013;9:e1003342.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550.
    1. Mootha VK, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34:267–273.

Source: PubMed

3
Sottoscrivi