301
|
Señalización asociada al receptor del factor de crecimiento similar a la insulina de tipo I en una línea celular colombiana de carcinoma mamario. BIOMEDICA 2011. [DOI: 10.7705/biomedica.v30i4.293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
302
|
Andreopoulou E. The PI3K/AKT/mTOR Signaling Pathway: Implications in the Treatment of Breast Cancer. CURRENT BREAST CANCER REPORTS 2011. [DOI: 10.1007/s12609-010-0038-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
303
|
Insulin-Like Growth Factor Pathway–Targeted Therapy in Breast Cancer. CURRENT BREAST CANCER REPORTS 2011. [DOI: 10.1007/s12609-010-0030-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
304
|
Davies E, Hiscox S. New therapeutic approaches in breast cancer. Maturitas 2011; 68:121-8. [DOI: 10.1016/j.maturitas.2010.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 10/29/2010] [Accepted: 10/29/2010] [Indexed: 01/29/2023]
|
305
|
Somatic expression of PyMT or activated ErbB2 induces estrogen-independent mammary tumorigenesis. Neoplasia 2011; 12:718-26. [PMID: 20824048 DOI: 10.1593/neo.10516] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 06/04/2010] [Accepted: 06/07/2010] [Indexed: 11/18/2022] Open
Abstract
Estrogen signaling is required for the proliferation of normal breast epithelial cells. However, prophylactic inhibition of estrogen signaling fails to prevent 56% of human breast cancer cases. The underlying mechanism is not well understood. Aberrant activation of growth factor signaling is known to provide alternative proliferation pathways in breast cells that are fully transformed, but it is not known whether activation of growth factor signaling can substitute for estrogen signaling in causing aberrant proliferation in the normal breast epithelium. Here, we report that in a retrovirus-based somatic mouse model (replication-competent ALV-LTR splice acceptor/tumor virus A) that closely mimics the evolution of sporadic human breast cancers, mammary epithelial cells harboring PyMT or activated ErbB2 evolve into tumors independent of estrogen or other ovarian functions in contrast to previous observations of estrogen-dependent cancer formation in germ line mouse models of ErbB2 activation. Importantly, ErbB2 activation in normal mammary cells causes estrogen-independent proliferation in both estrogen receptor (ER)-negative cells as well as in normally quiescent ER-positive cells. Therefore, aberrant activation of growth factor signaling contributes to estrogen-independent proliferation of both preneoplastic and cancerous mammary cells, and prophylactic therapy against both growth factor signaling and estrogen signaling may need to be considered in women with increased risk of breast cancer.
Collapse
|
306
|
Osborne CK, Neven P, Dirix LY, Mackey JR, Robert J, Underhill C, Schiff R, Gutierrez C, Migliaccio I, Anagnostou VK, Rimm DL, Magill P, Sellers M. Gefitinib or placebo in combination with tamoxifen in patients with hormone receptor-positive metastatic breast cancer: a randomized phase II study. Clin Cancer Res 2011; 17:1147-59. [PMID: 21220480 DOI: 10.1158/1078-0432.ccr-10-1869] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Increased growth factor signaling may contribute to tamoxifen resistance. This randomized phase II trial assessed tamoxifen plus placebo or the epidermal growth factor receptor inhibitor gefitinib in estrogen receptor (ER)-positive metastatic breast cancer. EXPERIMENTAL DESIGN Patients with newly metastatic disease or recurred after adjuvant tamoxifen (stratum 1), or recurred during/after adjuvant aromatase inhibitor (AI) or after failed first-line AI (stratum 2), were eligible. Primary variables were progression-free survival (PFS; stratum 1) and clinical benefit rate (CBR; stratum 2). A 5% or more improvement in response variables with gefitinib was considered to warrant further investigation. Outcome was correlated with biomarkers measured on the primary tumor. RESULTS In stratum 1 (n = 206), the PFS HR (gefitinib:placebo) was 0.84 (95% CI, 0.59-1.18; median PFS 10.9 versus 8.8 months). In the stratum 1 endocrine therapy-naïve subset (n = 158) the HR was 0.78 (95% CI, 0.52-1.15), and the prior endocrine-treated subgroup (n = 48) 1.47 (95% CI, 0.63-3.45). In stratum 1, CBRs were 50.5% with gefitinib and 45.5% with placebo. In stratum 2 (n = 84), CBRs were 29.2% with gefitinib and 31.4% with placebo. Biomarker analysis suggested that in stratum 1 there was greater benefit with gefitinib in patients who were ER-negative or had lower levels of ER protein. CONCLUSIONS In stratum 1, the improved PFS with gefitinib plus tamoxifen met the protocol criteria to warrant further investigation of this strategy. In stratum 2, there was a numerical disadvantage for gefitinib; additional investigation after AI therapy is not warranted. Studies of predictive biomarkers are needed to subset appropriate patients.
Collapse
Affiliation(s)
- C Kent Osborne
- Lester and Sue Smith Breast Center and Department of Pathology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
307
|
Abstract
The estrogen receptor (ER) pathway plays a pivotal role in breast cancer development and progression. Endocrine therapy to block the ER pathway is highly effective, but its usefulness is limited by common intrinsic and acquired resistance. Multiple mechanisms responsible for endocrine resistance have been proposed and include deregulation of various components of the ER pathway itself, alterations in cell cycle and cell survival signaling molecules, and the activation of escape pathways that can provide tumors with alternative proliferative and survival stimuli. Among these, increased expression or signaling of growth factor receptor pathways, especially the EGFR/HER2 pathway, has been associated with both experimental and clinical endocrine therapy resistance. New treatment combinations targeting both ER and growth factor receptor signaling to block the crosstalk between these pathways and eliminate escape routes have been proven highly effective in preclinical models. Results of recent clinical studies, while partly supporting this approach, also highlight the need to better identify a priori the patients whose tumors are most likely to benefit from these specific cotargeting strategies.
Collapse
Affiliation(s)
- C Kent Osborne
- Dan L. Duncan Cancer Center, Lester & Sue Smith Breast Center, Departments of Medicine and Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Rachel Schiff
- Dan L. Duncan Cancer Center, Lester & Sue Smith Breast Center, Departments of Medicine and Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| |
Collapse
|
308
|
Nardone A, Corvigno S, Brescia A, D'Andrea D, Limite G, Veneziani BM. Long-term cultures of stem/progenitor cells from lobular and ductal breast carcinomas under non-adherent conditions. Cytotechnology 2010; 63:67-80. [PMID: 21188518 DOI: 10.1007/s10616-010-9328-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 12/06/2010] [Indexed: 01/16/2023] Open
Abstract
A small subpopulation of stem/progenitor cells can give rise to the diversity of differentiated cells that comprise the bulk of the tumor. Are proliferating cells, within the bulk of tumor, few cells with uncommon features? The cell biological approach provides a limitless model for studying the hierarchical organization of progenitor subpopulation and identifying potential therapeutic targets. Aim of the study was to expand patients' breast cancer cells for evaluating functional cell properties, and to characterize the protein expression profile of selected cells to be compared with that of primary tumors. Breast cancer cells from estrogen receptor (ERα) positive, HER2 negative lobular (LoBS cells) and ductal (DuBS cells) histotype were cultured under non-adherent conditions to form mammospheres. Sorting of the cells by their surface expression of CD24 and CD44 gave rise to subpopulations which were propagated, enriched and characterized for the expression of epithelial and stromal markers. We found that non-adherent culture conditions generate mammospheres of slowly proliferating cells; single cells, dissociated from mammospheres, grow in soft agar; long-term cultured LoBS and DuBS cells, CD44+/CD24low, express cytokeratin 5 (CK5), α-smooth muscle actin (α-sma) and vimentin, known as markers of basal/myoepithelial cells; and ERα (only DuBS cells), HER1 (EGF-Receptor), activated HER2, and cyclinD1 as markers of luminal epithelial cell. Isolates of cells from breast cancer patients may be a tool for a marker-driven testing of targeted therapies.
Collapse
Affiliation(s)
- Agostina Nardone
- Dipartimento di Biologia e Patologia Cellulare e Molecolare "L. Califano", Università di Napoli Federico II, via Pansini 5, 80131, Naples, Italy
| | | | | | | | | | | |
Collapse
|
309
|
Gu F, Hsu HK, Hsu PY, Wu J, Ma Y, Parvin J, Huang THM, Jin VX. Inference of hierarchical regulatory network of estrogen-dependent breast cancer through ChIP-based data. BMC SYSTEMS BIOLOGY 2010; 4:170. [PMID: 21167036 PMCID: PMC3012048 DOI: 10.1186/1752-0509-4-170] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 12/17/2010] [Indexed: 12/16/2022]
Abstract
Background Global profiling of in vivo protein-DNA interactions using ChIP-based technologies has evolved rapidly in recent years. Although many genome-wide studies have identified thousands of ERα binding sites and have revealed the associated transcription factor (TF) partners, such as AP1, FOXA1 and CEBP, little is known about ERα associated hierarchical transcriptional regulatory networks. Results In this study, we applied computational approaches to analyze three public available ChIP-based datasets: ChIP-seq, ChIP-PET and ChIP-chip, and to investigate the hierarchical regulatory network for ERα and ERα partner TFs regulation in estrogen-dependent breast cancer MCF7 cells. 16 common TFs and two common new TF partners (RORA and PITX2) were found among ChIP-seq, ChIP-chip and ChIP-PET datasets. The regulatory networks were constructed by scanning the ChIP-peak region with TF specific position weight matrix (PWM). A permutation test was performed to test the reliability of each connection of the network. We then used DREM software to perform gene ontology function analysis on the common genes. We found that FOS, PITX2, RORA and FOXA1 were involved in the up-regulated genes. We also conducted the ERα and Pol-II ChIP-seq experiments in tamoxifen resistance MCF7 cells (denoted as MCF7-T in this study) and compared the difference between MCF7 and MCF7-T cells. The result showed very little overlap between these two cells in terms of targeted genes (21.2% of common genes) and targeted TFs (25% of common TFs). The significant dissimilarity may indicate totally different transcriptional regulatory mechanisms between these two cancer cells. Conclusions Our study uncovers new estrogen-mediated regulatory networks by mining three ChIP-based data in MCF7 cells and ChIP-seq data in MCF7-T cells. We compared the different ChIP-based technologies as well as different breast cancer cells. Our computational analytical approach may guide biologists to further study the underlying mechanisms in breast cancer cells or other human diseases.
Collapse
Affiliation(s)
- Fei Gu
- Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| | | | | | | | | | | | | | | |
Collapse
|
310
|
Oyama M, Nagashima T, Suzuki T, Kozuka-Hata H, Yumoto N, Shiraishi Y, Ikeda K, Kuroki Y, Gotoh N, Ishida T, Inoue S, Kitano H, Okada-Hatakeyama M. Integrated quantitative analysis of the phosphoproteome and transcriptome in tamoxifen-resistant breast cancer. J Biol Chem 2010; 286:818-29. [PMID: 21044952 DOI: 10.1074/jbc.m110.156877] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Quantitative phosphoproteome and transcriptome analysis of ligand-stimulated MCF-7 human breast cancer cells was performed to understand the mechanisms of tamoxifen resistance at a system level. Phosphoproteome data revealed that WT cells were more enriched with phospho-proteins than tamoxifen-resistant cells after stimulation with ligands. Surprisingly, decreased phosphorylation after ligand perturbation was more common than increased phosphorylation. In particular, 17β-estradiol induced down-regulation in WT cells at a very high rate. 17β-Estradiol and the ErbB ligand heregulin induced almost equal numbers of up-regulated phospho-proteins in WT cells. Pathway and motif activity analyses using transcriptome data additionally suggested that deregulated activation of GSK3β (glycogen-synthase kinase 3β) and MAPK1/3 signaling might be associated with altered activation of cAMP-responsive element-binding protein and AP-1 transcription factors in tamoxifen-resistant cells, and this hypothesis was validated by reporter assays. An examination of clinical samples revealed that inhibitory phosphorylation of GSK3β at serine 9 was significantly lower in tamoxifen-treated breast cancer patients that eventually had relapses, implying that activation of GSK3β may be associated with the tamoxifen-resistant phenotype. Thus, the combined phosphoproteome and transcriptome data set analyses revealed distinct signal transcription programs in tumor cells and provided a novel molecular target to understand tamoxifen resistance.
Collapse
Affiliation(s)
- Masaaki Oyama
- Medical Proteomics Laboratory, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
311
|
Hsia EY, Goodson ML, Zou JX, Privalsky ML, Chen HW. Nuclear receptor coregulators as a new paradigm for therapeutic targeting. Adv Drug Deliv Rev 2010; 62:1227-37. [PMID: 20933027 DOI: 10.1016/j.addr.2010.09.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 09/24/2010] [Accepted: 09/30/2010] [Indexed: 02/06/2023]
Abstract
The complex function and regulation of nuclear receptors cannot be fully understood without a thorough knowledge of the receptor-associated coregulators that either enhance (coactivators) or inhibit (corepressors) transcription. While nuclear receptors themselves have garnered much attention as therapeutic targets, the clinical and etiological relevance of the coregulators to human diseases is increasingly recognized. Aberrant expression or function of coactivators and corepressors has been associated with malignant and metabolic disease development. Many of them are key epigenetic regulators and utilize enzymatic activities to modify chromatin through histone acetylation/deacetylation, histone methylation/demethylation or chromatin remodeling. In this review, we showcase and evaluate coregulators--such as SRCs and ANCCA--with the most promising therapeutic potential based on their physiological roles and involvement in various diseases that are revealed thus far. We also describe the structural features of the coactivator and corepressor functional domains and highlight areas that can be further explored for molecular targeting.
Collapse
|
312
|
Cittelly DM, Das PM, Salvo VA, Fonseca JP, Burow ME, Jones FE. Oncogenic HER2{Delta}16 suppresses miR-15a/16 and deregulates BCL-2 to promote endocrine resistance of breast tumors. Carcinogenesis 2010; 31:2049-57. [PMID: 20876285 PMCID: PMC2994280 DOI: 10.1093/carcin/bgq192] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tamoxifen is the most commonly prescribed therapy for patients with estrogen receptor (ER)α-positive breast tumors. Tumor resistance to tamoxifen remains a serious clinical problem especially in patients with tumors that also overexpress human epidermal growth factor receptor 2 (HER2). Current preclinical models of HER2 overexpression fail to recapitulate the clinical spectrum of endocrine resistance associated with HER2/ER-positive tumors. Here, we show that ectopic expression of a clinically important oncogenic isoform of HER2, HER2Δ16, which is expressed in >30% of ER-positive breast tumors, promotes tamoxifen resistance and estrogen independence of MCF-7 xenografts. MCF-7/HER2Δ16 cells evade tamoxifen through upregulation of BCL-2, whereas mediated suppression of BCL-2 expression or treatment of MCF-7/HER2Δ16 cells with the BCL-2 family pharmacological inhibitor ABT-737 restores tamoxifen sensitivity. Tamoxifen-resistant MCF-7/HER2Δ16 cells upregulate BCL-2 protein levels in response to suppressed ERα signaling mediated by estrogen withdrawal, tamoxifen treatment or fulvestrant treatment. In addition, HER2Δ16 expression results in suppression of BCL-2-targeting microRNAs miR-15a and miR-16. Reintroduction of miR-15a/16 reduced tamoxifen-induced BCL-2 expression and sensitized MCF-7/HER2Δ16 to tamoxifen. Conversely, inhibition of miR-15a/16 in tamoxifen-sensitive cells activated BCL-2 expression and promoted tamoxifen resistance. Our results suggest that HER2Δ16 expression promotes endocrine-resistant HER2/ERα-positive breast tumors and in contrast to wild-type HER2, preclinical models of HER2Δ16 overexpression recapitulate multiple phenotypes of endocrine-resistant human breast tumors. The mechanism of HER2Δ16 therapeutic evasion, involving tamoxifen-induced upregulation of BCL-2 and suppression of miR-15a/16, provides a template for unique therapeutic interventions combining tamoxifen with modulation of microRNAs and/or ABT-737-mediated BCL-2 inhibition and apoptosis.
Collapse
Affiliation(s)
- Diana M Cittelly
- Department of Pathology, School of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | |
Collapse
|
313
|
Jin K, He K, Li G, Teng L. Personalized cancer therapy using a patient-derived tumor tissue xenograft model: a translational field worthy of exploring further? Per Med 2010; 7:597-606. [PMID: 29776245 DOI: 10.2217/pme.10.48] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
It has long been observed that interpatient variability in response to anticancer drugs is associated with different outcomes. Oncologists continually hold the desire of matching the right therapeutic regimen with the right cancer patient, which is termed ‘personalized cancer therapy’. Rapid advances in genetics, genomics and related technologies are promising a new era of personalized cancer therapy based on individual molecular biomarkers. However, these molecular predictors of tumor response are far from perfect. Because of the inherent limitations in the current approaches for anticancer drugs response prediction, the need for new techniques to predict tumor response to therapy is urgent. Using a patient-derived human tumor tissue (PDTT) xenograft model to predict tumor response to therapy might be an ideal candidate method to choose. This article provides an overview of the achievements and limitations of genetic, genomic and proteomic molecular markers for personalized cancer therapy, and further discusses the potentials of using a PDTT xenograft model as a candidate strategy for personalized cancer therapy.
Collapse
Affiliation(s)
- Ketao Jin
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of General Surgery, Zhejiang University Teaching Hospital Zhuji Hospital, Zhuji, Zhejiang, China
| | - Kuifeng He
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangliang Li
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | |
Collapse
|
314
|
Plaza-Menacho I, Morandi A, Robertson D, Pancholi S, Drury S, Dowsett M, Martin LA, Isacke CM. Targeting the receptor tyrosine kinase RET sensitizes breast cancer cells to tamoxifen treatment and reveals a role for RET in endocrine resistance. Oncogene 2010; 29:4648-57. [PMID: 20531297 DOI: 10.1038/onc.2010.209] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 04/23/2010] [Accepted: 04/27/2010] [Indexed: 12/11/2022]
Abstract
Endocrine therapy is the main therapeutic option for patients with estrogen receptor (ERalpha)-positive breast cancer. Resistance to this treatment is often associated with estrogen-independent activation of ERalpha. In this study, we show that in ERalpha-positive breast cancer cells, activation of the receptor tyrosine kinase RET (REarranged during Transfection) by its ligand GDNF results in increased ERalpha phosphorylation on Ser118 and Ser167 and estrogen-independent activation of ERalpha transcriptional activity. Further, we identify mTOR as a key component in this downstream signaling pathway. In tamoxifen response experiments, RET downregulation resulted in 6.2-fold increase in sensitivity of MCF7 cells to antiproliferative effects of tamoxifen, whereas GDNF stimulation had a protective effect against the drug. In tamoxifen-resistant (TAM(R)-1) MCF7 cells, targeting RET restored tamoxifen sensitivity. Finally, examination of two independent tissue microarrays of primary human breast cancers revealed that expression of RET protein was significantly associated with ERalpha-positive tumors and that in primary tumors from patients who subsequently developed invasive recurrence after adjuvant tamoxifen treatment, there was a twofold increase in the number of RET-positive tumors. Together these findings identify RET as a potentially important therapeutic target in ERalpha-positive breast cancers and in particular in tamoxifen-resistant tumors.
Collapse
Affiliation(s)
- I Plaza-Menacho
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
315
|
Kabos P, Haughian JM, Wang X, Dye WW, Finlayson C, Elias A, Horwitz KB, Sartorius CA. Cytokeratin 5 positive cells represent a steroid receptor negative and therapy resistant subpopulation in luminal breast cancers. Breast Cancer Res Treat 2010; 128:45-55. [PMID: 20665103 DOI: 10.1007/s10549-010-1078-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 07/17/2010] [Indexed: 12/29/2022]
Abstract
A majority of breast cancers are estrogen receptor (ER) positive and have a luminal epithelial phenotype. However, these ER⁺ tumors often contain heterogeneous subpopulations of ER⁻ tumor cells. We previously identified a population of cytokeratin 5 (CK5) positive cells within ER⁺ and progesterone receptor positive (PR⁺) tumors that is both ER⁻PR⁻ and CD44⁺, a marker of breast tumor-initiating cells (TICs). These CK5⁺ cells have properties of TICs in luminal tumor xenografts, and we speculated that they are more resistant to chemo- and anti-ER-targeted therapies than their ER⁺ neighbors. To test this, we used ER⁺PR⁺ T47D and MCF7 breast cancer cells. CK5⁺ cells had lower proliferative indices than CK5⁻ cells, were less sensitive to 5-fluorouracil and docetaxel, and cultures became enriched for CK5⁺ cells after treatments. CK5⁺ cells were less prone to drug-induced apoptosis than CK5⁻ cells. In cells treated with 17β-estradiol (E) plus anti-estrogens tamoxifen or fulvestrant, ER protein levels decreased, and CK5 protein levels increased, compared to controls treated with E alone. In ER⁺ tumors from patients treated with neoadjuvant endocrine therapies ER gene expression decreased, and CK5 gene expression increased in post compared to pre-treatment tumors. The number of CK5⁺ cells in tumors also increased in post- compared to pre-treatment tumors. We conclude that an ER⁻PR⁻CK5⁺ subpopulation found in many luminal tumors is resistant to standard endocrine and chemotherapies, relative to the majority ER⁺PR⁺CK5⁻ cells. Compounds that effectively target these cells are needed to improve outcome in luminal breast cancers.
Collapse
Affiliation(s)
- Peter Kabos
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|
316
|
Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, Hicks DG, Lester S, Love R, Mangu PB, McShane L, Miller K, Osborne CK, Paik S, Perlmutter J, Rhodes A, Sasano H, Schwartz JN, Sweep FCG, Taube S, Torlakovic EE, Valenstein P, Viale G, Visscher D, Wheeler T, Williams RB, Wittliff JL, Wolff AC. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer (unabridged version). Arch Pathol Lab Med 2010; 134:e48-72. [PMID: 20586616 DOI: 10.5858/134.7.e48] [Citation(s) in RCA: 782] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE To develop a guideline to improve the accuracy of immunohistochemical (IHC) estrogen receptor (ER) and progesterone receptor (PgR) testing in breast cancer and the utility of these receptors as predictive markers. METHODS The American Society of Clinical Oncology and the College of American Pathologists convened an international Expert Panel that conducted a systematic review and evaluation of the literature in partnership with Cancer Care Ontario and developed recommendations for optimal IHC ER/PgR testing performance. RESULTS Up to 20% of current IHC determinations of ER and PgR testing worldwide may be inaccurate (false negative or false positive). Most of the issues with testing have occurred because of variation in pre-analytic variables, thresholds for positivity, and interpretation criteria. RECOMMENDATIONS The Panel recommends that ER and PgR status be determined on all invasive breast cancers and breast cancer recurrences. A testing algorithm that relies on accurate, reproducible assay performance is proposed. Elements to reliably reduce assay variation are specified. It is recommended that ER and PgR assays be considered positive if there are at least 1% positive tumor nuclei in the sample on testing in the presence of expected reactivity of internal (normal epithelial elements) and external controls. The absence of benefit from endocrine therapy for women with ER-negative invasive breast cancers has been confirmed in large overviews of randomized clinical trials.
Collapse
Affiliation(s)
- M Elizabeth H Hammond
- Intermountain Healthcare, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
317
|
Cavaliere C, Corvigno S, Galgani M, Limite G, Nardone A, Veneziani BM. Combined inhibitory effect of formestane and herceptin on a subpopulation of CD44+/CD24low breast cancer cells. Cancer Sci 2010; 101:1661-9. [PMID: 20491779 PMCID: PMC11159050 DOI: 10.1111/j.1349-7006.2010.01593.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In breast cancer, stromal cells surrounding cancer epithelial cells can influence phenotype by producing paracrine factors. Among many mediators of epithelial-stromal interactions, aromatase activity is perhaps one of the best studied. Clinical data suggest that estrogen-independent signaling leads to increased proliferation even during therapy with aromatase inhibitors (AIs). Molecular mechanism of crosstalk between the estrogen receptor (ER) and the epidermal growth factor receptor (HER) family have been implicated in resistance to endocrine therapy, but this interaction is unclear. The ability of aromatase to induce estradiol biosynthesis provides a molecular rationale to combine agents that target aromatase activity and the HER pathway. We targeted stromal-epithelial interactions using formestane, which exerts antiaromatase activity, combined with the monoclonal anti-HER2 antibody herceptin, in a subpopulation of CD44+/CD24low cells sorted from epithelial-mesenchymal co-cultures of breast cancer tissues. The growth inhibition was respectively 16% (P < 0.01) in the response to herceptin, 25% to formestane (P < 0.01), and 50% (P < 0.001) with the combination of the two drugs, suggesting that herceptin cooperates with formestane-induced inhibition of aromatase and this effect could be mediated through HER family receptors. In cells which expressed ERalpha, formestane/herceptin combination suppressed the mRNA expression of aromatase and HER2 and decreased cyclin D1 expression. These results show that combination therapies involving AIs and anti-HER2 can be efficacious for the treatment of cancer in experimental models and suggest that subtyping breast tumors gives useful information about response to treatment.
Collapse
Affiliation(s)
- Carla Cavaliere
- Department of Cellular and Molecular Biology and Pathology L. Califano, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | |
Collapse
|
318
|
Miller TW, Hennessy BT, González-Angulo AM, Fox EM, Mills GB, Chen H, Higham C, García-Echeverría C, Shyr Y, Arteaga CL. Hyperactivation of phosphatidylinositol-3 kinase promotes escape from hormone dependence in estrogen receptor-positive human breast cancer. J Clin Invest 2010; 120:2406-13. [PMID: 20530877 PMCID: PMC2898598 DOI: 10.1172/jci41680] [Citation(s) in RCA: 441] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 04/14/2010] [Indexed: 02/03/2023] Open
Abstract
Many breast cancers exhibit a degree of dependence on estrogen for tumor growth. Although several therapies have been developed to treat individuals with estrogen-dependent breast cancers, some tumors show de novo or acquired resistance, rendering them particularly elusive to current therapeutic strategies. Understanding the mechanisms by which these cancers develop resistance would enable the development of new and effective therapeutics. In order to determine mechanisms of escape from hormone dependence in estrogen receptor-positive (ER-positive) breast cancer, we established 4 human breast cancer cell lines after long-term estrogen deprivation (LTED). LTED cells showed variable changes in ER levels and sensitivity to 17beta-estradiol. Proteomic profiling of LTED cells revealed increased phosphorylation of the mammalian target of rapamycin (mTOR) substrates p70S6 kinase and p85S6 kinase as well as the PI3K substrate AKT. Inhibition of PI3K and mTOR induced LTED cell apoptosis and prevented the emergence of hormone-independent cells. Using reverse-phase protein microarrays, we identified a breast tumor protein signature of PI3K pathway activation that predicted poor outcome after adjuvant endocrine therapy in patients. Our data suggest that upon adaptation to hormone deprivation, breast cancer cells rely heavily on PI3K signaling. Our findings also imply that acquired resistance to endocrine therapy in breast cancer may be abrogated by combination therapies targeting both ER and PI3K pathways.
Collapse
Affiliation(s)
- Todd W. Miller
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Bryan T. Hennessy
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Ana M. González-Angulo
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Emily M. Fox
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Gordon B. Mills
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Heidi Chen
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Catherine Higham
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Carlos García-Echeverría
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Yu Shyr
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Carlos L. Arteaga
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Department of Gynecology Medical Oncology,
Department of Systems Biology, and
Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Department of Medicine and
Department of Biostatistics, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA.
Institute for Oncology Research, Novartis Pharma AG, Basel, Switzerland.
Breast Cancer Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
319
|
Spears M, Kenicer J, Munro AF, Bartlett JMS. Type I receptor tyrosine kinases as predictive or prognostic markers in early breast cancer. Biomark Med 2010; 2:397-407. [PMID: 20477393 DOI: 10.2217/17520363.2.4.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The type I receptor tyrosine kinase (RTK) family of proteins play an essential role in the progression of early breast cancer. Our understanding of the role of these proteins has increased over the last 20 years, however, as yet, there are still a number of unanswered questions regarding their position in endocrine resistance, chemotherapy resistance and in the biology of breast cancer. There have been, and are currently, a number of clinical trials that have examined the use of anticancer therapy such as cytotoxic drugs, and treatments that target the RTKs and signaling pathways that have been identified. There is clear evidence that molecular subtypes of cancer respond differently to different therapeutic options, which challenges the 'one size fits all' approach to chemotherapy. Here we review the human epidermal growth factor receptor family of proteins and their potential predictive or prognostic role in early breast cancer.
Collapse
Affiliation(s)
- M Spears
- Endocrine Cancer Group, Edinburgh Cancer Research Centre, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, Scotland
| | | | | | | |
Collapse
|
320
|
PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer. Proc Natl Acad Sci U S A 2010; 107:10208-13. [PMID: 20479250 PMCID: PMC2890442 DOI: 10.1073/pnas.0907011107] [Citation(s) in RCA: 303] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PIK3CA mutations are reported to be present in approximately 25% of breast cancer (BC), particularly the estrogen receptor-positive (ER+) and HER2-overexpressing (HER2+) subtypes, making them one of the most common genetic aberrations in BC. In experimental models, these mutations have been shown to activate AKT and induce oncogenic transformation, and hence these lesions have been hypothesized to render tumors highly sensitive to therapeutic PI3K/mTOR inhibition. By analyzing gene expression and protein data from nearly 1,800 human BCs, we report that a PIK3CA mutation-associated gene signature (PIK3CA-GS) derived from exon 20 (kinase domain) mutations was able to predict PIK3CA mutation status in two independent datasets, strongly suggesting a characteristic set of gene expression-induced changes. However, in ER+/HER2- BC despite pathway activation, PIK3CA mutations were associated with a phenotype of relatively low mTORC1 signaling and a good prognosis with tamoxifen monotherapy. The relationship between clinical outcome and the PIK3CA-GS was also assessed. Although the PIK3CA-GS was not associated with prognosis in ER- and HER2+ BC, it could identify better clinical outcomes in ER+/HER2- disease. In ER+ BC cell lines, PIK3CA mutations were also associated with sensitivity to tamoxifen. These findings could have important implications for the treatment of PIK3CA-mutant BCs and the development of PI3K/mTOR inhibitors.
Collapse
|
321
|
Endocrine Therapy for Metastatic Disease: Reversing Resistance. CURRENT BREAST CANCER REPORTS 2010. [DOI: 10.1007/s12609-010-0002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
322
|
Saggar JK, Chen J, Corey P, Thompson LU. The Effect of Secoisolariciresinol Diglucoside and Flaxseed Oil, Alone and in Combination, on MCF-7 Tumor Growth and Signaling Pathways. Nutr Cancer 2010; 62:533-42. [DOI: 10.1080/01635580903532440] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
323
|
EGFR/HER2 inhibitor AEE788 increases ER-mediated transcription in HER2/ER-positive breast cancer cells but functions synergistically with endocrine therapy. Br J Cancer 2010; 102:1235-43. [PMID: 20386540 PMCID: PMC2856013 DOI: 10.1038/sj.bjc.6605641] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background: Cross-talk between receptor tyrosine kinases and the oestrogen receptor (ER) is implicated in resistance to endocrine therapy. We investigated whether AEE788 (a combined inhibitor of EGFR, HER2 and VEGFR) plus tamoxifen or letrozole enhanced the individual anti-tumour effects of these agents. Methods: Breast cancer cell lines modelling endocrine-resistant and -sensitive disease were engineered to express aromatase (A) and examined using proliferation, western blotting and ER-α transcription assays. Results: AEE788 enhanced the anti-proliferative effect of tamoxifen and letrozole in ER+ cell lines (MCF-7 2A, ZR75.1 A3 and BT474 A3). This associated with an elevated G1 arrest and nuclear accumulation of p27. It is noteworthy that AEE788 alone or in combination with endocrine therapy increased the expression of progesterone receptor (PGR) and TFF1 in BT474 A3 cells. This may indicate a mechanism of resistance to AEE788 in ER+/HER2+ breast cancers. In a ZR75.1 A3 xenograft, AEE788 alone or in combination with tamoxifen provided no further benefit compared with letrozole. However, letrozole plus AEE788 produced a significantly greater inhibition of tumour growth compared with letrozole alone. Conclusion: These data suggest that AEE788 plus letrozole in breast cancer overexpressing HER2 may provide superior anti-tumour activity, compared with single agents.
Collapse
|
324
|
Haagenson KK, Wu GS. The role of MAP kinases and MAP kinase phosphatase-1 in resistance to breast cancer treatment. Cancer Metastasis Rev 2010; 29:143-9. [PMID: 20111893 DOI: 10.1007/s10555-010-9208-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chemotherapy resistance is an important problem often encountered during the course of breast cancer treatment. In order to design rational and efficacious therapies, the molecular mechanisms used by cells to develop resistance must be investigated. One mechanism employed by cancer cells is to alter cell signaling. This review examines the role of mitogen-activated protein kinases (MAPKs) and their endogenous negative regulators, mitogen-activated protein kinase phosphatases (MKPs), in chemotherapy resistance in breast cancer. MAPK signaling is activated in response to both growth factors and cellular stress. MKPs dephosphorylate MAPKs and are part of the dual-specificity family of phosphatases. MAPKs have been shown to be involved in resistance to tamoxifen, and MKPs have been linked to resistance to treatment with doxorubicin, mechlorethamine, paclitaxel, proteasome inhibitors, and oxidative-stress-induced cell death in breast cancer. The role of MKPs in tamoxifen resistance and the elucidation of the mechanisms involved with resistance to standard chemotherapy agents need to be investigated further. Growing evidence suggests that modulating MKP-1 activity could be a viable option to make breast cancer chemotherapy more effective.
Collapse
Affiliation(s)
- Kelly K Haagenson
- Graduate Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
325
|
Artemin is estrogen regulated and mediates antiestrogen resistance in mammary carcinoma. Oncogene 2010; 29:3228-40. [PMID: 20305694 DOI: 10.1038/onc.2010.71] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have previously identified an oncogenic role of artemin (ARTN), a member of glial cell derived neurotrophic factor family of ligands, in mammary carcinoma. We herein report that ARTN is an estrogen-inducible gene. Meta-analysis of gene expression data sets showed that ARTN expression is positively correlated to estrogen receptor (ER) status in human mammary carcinoma. Furthermore, in patients with ER-positive mammary carcinoma treated with tamoxifen, high ARTN expression is significantly correlated with decreased survival. Forced expression of ARTN in ER-positive human mammary carcinoma cells increased ER transcriptional activity, promoted estrogen-independent growth and produced resistance to tamoxifen and fulvestrant in vitro and to tamoxifen in xenograft models. ARTN-stimulated resistance to tamoxifen and fulvestrant is mediated by increased BCL-2 expression. Conversely, depletion of endogenous ARTN by small-interfering RNA or functional antagonism of ARTN by antibody enhanced the efficacy of antiestrogens. Tamoxifen decreased ARTN expression in tamoxifen-sensitive mammary carcinoma cells whereas ARTN expression was increased in tamoxifen-resistant cells and not affected by tamoxifen treatment. Antibody inhibition of ARTN in tamoxifen-resistant cells improved tamoxifen sensitivity. Functional antagonism of ARTN therefore warrants consideration as an adjuvant therapy to enhance antiestrogen efficacy in ER-positive mammary carcinoma.
Collapse
|
326
|
Abstract
The IGF pathway plays a major role in cancer cell proliferation, survival and resistance to antineoplastic therapies in many human malignancies. As such, interference with this pathway is the target of many investigational pharmacologic agents. Cixutumumab, a monoclonal antibody to IGF-1R, utilizes this concept. In this review, we summarize preclinical, pharmacologic and early clinical data regarding this agent and discuss the impact this drug might have on the future treatment of human cancers.
Collapse
Affiliation(s)
- Kevin P McKian
- Mayo Clinic College of Medicine, Division of Medical Oncology, 200 First St. SW Rochester, MN 55905, USA
| | | |
Collapse
|
327
|
Rimawi MF, Shetty PB, Weiss HL, Schiff R, Osborne CK, Chamness GC, Elledge RM. Epidermal growth factor receptor expression in breast cancer association with biologic phenotype and clinical outcomes. Cancer 2010; 116:1234-42. [PMID: 20082448 PMCID: PMC2829330 DOI: 10.1002/cncr.24816] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) expression is associated with aggressive phenotypes in preclinical breast cancer models, but in clinical studies, EGFR has been inconsistently linked to poor outcome. We hypothesized that EGFR expression in human breast tumors, when centrally and uniformly assessed, is associated with an aggressive phenotype and resistance to systemic therapy. METHODS In a database of 47,286 patients with breast cancer, EGFR status was known on 2567 tumors. EGFR levels were measured centrally by ligand binding assay, and tumors with > or =10 fmol/mg were prospectively deemed positive. Clinical and biological features of EGFR-positive and EGFR-negative tumors were compared. Clinical outcomes were assessed by systemic therapy status. RESULTS Of 2567 tumors, 475 (18%) were EGFR positive. EGFR-positive tumors were more common in younger and in black women, were larger, had a higher S-phase fraction, and were more likely to be aneuploid. EGFR-positive tumors were more likely to be HER2-positive (26% vs 16%, P < .0001), but less likely to be estrogen receptor-positive (60% vs 88%, P < .0001) or progesterone receptor-positive (26% vs 65%, P < .0001). In multivariate analyses, EGFR expression independently correlated with worse disease-free survival (hazard ratio [HR] = 1.66; 95% confidence interval [CI], 1.4-2.41, P = .007) and overall survival (HR = 1.98, 95% CI, 1.36-2.88, P = .0004) in treated patients, but not in untreated patients. CONCLUSIONS EGFR expression is more common in breast tumors in younger and black women. It is associated with lower hormone receptor levels, higher proliferation, genomic instability, and HER2 overexpression. It is correlated with higher risk of relapse in patients receiving adjuvant treatment. Blocking EGFR may improve outcome in selected patients.
Collapse
|
328
|
Schwartzberg LS, Franco SX, Florance A, O'Rourke L, Maltzman J, Johnston S. Lapatinib plus letrozole as first-line therapy for HER-2+ hormone receptor-positive metastatic breast cancer. Oncologist 2010; 15:122-9. [PMID: 20156908 PMCID: PMC3227947 DOI: 10.1634/theoncologist.2009-0240] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Reported are results from a subgroup analysis of postmenopausal women with hormone receptor–positive human epidermal growth factor receptor 2–positive metastatic breast cancer from a phase III trial of letrozole plus placebo versus letrozole plus lapatinib. The combination was well tolerated and more efficacious than letrozole alone. Objective. To evaluate the efficacy and tolerability of letrozole plus lapatinib versus letrozole plus placebo in women with hormone receptor (HR)+ human epidermal growth factor receptor (HER)-2+ tumors receiving first-line therapy for metastatic breast cancer (MBC). Patients and Methods. Postmenopausal women (n = 1,286) with HR+ MBC were randomized to daily oral treatment with letrozole (2.5 mg) plus lapatinib (1,500 mg) versus letrozole (2.5 mg) plus placebo. Of the 1,286 patients enrolled in the phase III study, 219 had HER-2+ tumors. The primary endpoint was progression-free survival (PFS) in HER-2+ patients. Results. Results in the HR+ HER-2+ population (n = 219) are presented. The addition of lapatinib to letrozole resulted in a significantly lower risk for disease progression than with letrozole alone (hazard ratio, 0.71; 95% confidence interval, 0.53–0.96). The PFS time was 8.2 months, versus 3.0 months. The objective response rate (ORR) (28% versus 15%) and clinical benefit rate (CBR) (48% versus 29%) were also significantly greater in lapatinib-treated women. The most common adverse events in the lapatinib group were diarrhea (68%) and rash (46%), primarily grade 1 and 2. Conclusions. The addition of lapatinib to letrozole is well tolerated and leads to a significantly greater PFS time, ORR, and CBR than with letrozole alone in women with MBC who coexpress HR and HER-2.
Collapse
|
329
|
Lin SL, Yan LY, Zhang XT, Yuan J, Li M, Qiao J, Wang ZY, Sun QY. ER-alpha36, a variant of ER-alpha, promotes tamoxifen agonist action in endometrial cancer cells via the MAPK/ERK and PI3K/Akt pathways. PLoS One 2010; 5:e9013. [PMID: 20126312 PMCID: PMC2814868 DOI: 10.1371/journal.pone.0009013] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 01/13/2010] [Indexed: 11/18/2022] Open
Abstract
Background Recently, a novel variant of ER-α, ER-α36 was identified and cloned. ER-α36 lacks intrinsic transcription activity and mainly mediates nongenomic estrogen signaling. Here, we studied the role of nongenomic estrogen signaling pathways mediated by ER-α36 in tamoxifen resistance and agonist action. Methodology The cellular localization of ER-α36 was examined by immunofluorescence in MCF-7 cells and Hec1A cells. MCF-7 breast cancer cells, MCF-7 cells expressing recombinant ER-α36 (MCF-7/ER36), Hec1A endometrial cancer cells and Hec1A cells with siRNA knockdown of ER-α36 (Hec1A/RNAiER36) were treated with 17β-estradial (E2) and tamoxifen (TAM) in the absence and presence of kinase inhibitor U0126 and LY294002. We examined phosphorylation of signaling molecules and the expression of c-Myc by immunoblotting, and tumor cell growth by MTT assay. Conclusions ER variant ER-α36 enhances TAM agonist activity through activation of the membrane-initiated signaling pathways in endometrial cancer, and that ER-α36 is involved in de novo and acquired TAM resistance in breast cancer.
Collapse
Affiliation(s)
- Sheng-Li Lin
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences and Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Li-Ying Yan
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Xin-Tian Zhang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, Omaha, Nebraska, United States of America
| | - Ju Yuan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences and Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Mo Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences and Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Jie Qiao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- * E-mail: (JQ); (Z-YW); (Q-YS)
| | - Zhao-Yi Wang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, Omaha, Nebraska, United States of America
- * E-mail: (JQ); (Z-YW); (Q-YS)
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences and Graduate School, Chinese Academy of Sciences, Beijing, China
- * E-mail: (JQ); (Z-YW); (Q-YS)
| |
Collapse
|
330
|
Suba Z. Common soil of smoking-associated and hormone-related cancers: estrogen deficiency. Oncol Rev 2010. [DOI: 10.1007/s12156-010-0036-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
331
|
Carver KC, Piazza TM, Schuler LA. Prolactin enhances insulin-like growth factor I receptor phosphorylation by decreasing its association with the tyrosine phosphatase SHP-2 in MCF-7 breast cancer cells. J Biol Chem 2010; 285:8003-12. [PMID: 20080972 DOI: 10.1074/jbc.m109.066480] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Normal mammary development requires coordinated interactions of numerous factors, including prolactin (PRL) and insulin-like growth factor I (IGF-I), both of which have also been implicated in breast cancer pathogenesis and progression. We previously reported that PRL and IGF-I synergize in breast cancer cells to activate ERK1/2 and AKT, leading to increased proliferation, survival, and invasion. Intriguingly, PRL co-treatment with IGF-I augments IGF-I receptor (IGF-IR) phosphorylation 2-fold higher than IGF-I alone. Here, we showed the importance of the tyrosine phosphatase SHP-2 in this cross-talk using pharmacological inhibition and small interfering RNA. SHP-2 recruitment to IGF-IR was significantly attenuated by PRL co-treatment. Src family kinase activity was required for IGF-IR association with SHP-2, ligand-induced IGF-IR internalization, and PRL-enhanced IGF-IR phosphorylation. Inhibition of internalization, via knockdown of the GTPase, dynamin-2, prevented not only IGF-IR dephosphorylation, but also PRL-enhanced IGF-IR phosphorylation. Consistently, PRL diminished IGF-I-induced IGF-IR internalization, which may result from reduced SHP-2 association with IGF-IR, because we demonstrated an essential role for SHP-2 in IGF-IR internalization. Together, these findings describe a novel mechanism of cross-talk between PRL and IGF-I in breast cancer cells, with implications for our understanding of tumor progression and potential therapeutic strategies.
Collapse
Affiliation(s)
- Kristopher C Carver
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
332
|
Ghayad SE, Vendrell JA, Ben Larbi S, Dumontet C, Bieche I, Cohen PA. Endocrine resistance associated with activated ErbB system in breast cancer cells is reversed by inhibiting MAPK or PI3K/Akt signaling pathways. Int J Cancer 2010; 126:545-62. [PMID: 19609946 DOI: 10.1002/ijc.24750] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endocrine therapy resistance is one of the main challenges in the treatment of estrogen receptor positive (ER+) breast cancer patients. This study showed that two ER+ human breast carcinoma cell lines derived from MCF-7 (MVLN cells) that have acquired under OH-Tamoxifen selection two distinct phenotypes of endocrine resistance both displayed constitutive activation of the PI3K/Akt and MAPK pathways. Aberrant expression and activation of the ErbB system (phospho-EGFR, phospho-ErbB2, phospho-ErbB3, over-expression of ErbB4 and over-expression of several ErbB ligands) were also observed in the two resistant cell lines, suggesting the existence of an autocrine loop leading to constitutive activation of MAPK and PI3K/Akt survival pathways. The recent clinical use of specific signal transduction inhibitors is one of the most promising therapeutic approaches in breast cancers. The MEK inhibitor PD98059 and the PI3K inhibitor LY294002 were both able to enhance the cytostatic effect of OH-Tamoxifen or fulvestrant on MVLN sensitive cells. In the two resistant cell lines, inhibition of the MAPK or the PI3K/Akt pathways associated with endocrine therapy was sufficient to reverse OH-Tamoxifen or fulvestrant resistance. Investigating the effect of a combination of both inhibitors on the reversion of OH-Tamoxifen and fulvestrant resistance in the two resistant cell lines suggested that, in clinical practice, a strategy combining the two inhibitors would be the best approach to target the different endocrine resistance phenotypes possibly present in a tumor. In conclusion, the combination of MAPK and PI3K inhibitors represents a promising strategy to overcome endocrine therapy resistance in ER+ breast cancer patients.
Collapse
|
333
|
McCune K, Bhat-Nakshatri P, Thorat MA, Nephew KP, Badve S, Nakshatri H. Prognosis of hormone-dependent breast cancers: implications of the presence of dysfunctional transcriptional networks activated by insulin via the immune transcription factor T-bet. Cancer Res 2010; 70:685-96. [PMID: 20068169 DOI: 10.1158/0008-5472.can-09-1530] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Estrogen receptor alpha (ERalpha)-positive breast cancers that co-express transcription factors GATA-3 and FOXA1 have a favorable prognosis. These transcription factors form an autoregulatory hormonal network that influences estrogen responsiveness and sensitivity to hormonal therapy. Disruption of this network may be a mechanism whereby ERalpha-positive breast cancers become resistant to therapy. The transcription factor T-bet is a negative regulator of GATA-3 in the immune system. In this study, we report that insulin increases the expression of T-bet in breast cancer cells, which correlates with reduced expression of GATA-3, FOXA1, and the ERalpha:FOXA1:GATA-3 target gene GREB-1. The effects of insulin on GATA-3 and FOXA1 could be recapitulated through overexpression of T-bet in MCF-7 cells (MCF-7-T-bet). Chromatin immunoprecipitation assays revealed reduced ERalpha binding to GREB-1 enhancer regions in MCF-7-T-bet cells and in insulin-treated MCF-7 cells. MCF-7-T-bet cells were resistant to tamoxifen in the presence of insulin and displayed prolonged extracellular signal-regulated kinase and AKT activation in response to epidermal growth factor treatment. ERalpha-positive cells with intrinsic tamoxifen resistance as well as MCF-7 cells with acquired tamoxifen and fulvestrant resistance expressed elevated levels of T-bet and/or reduced levels of FOXA1 and GATA-3. Analysis of publicly available databases revealed ERalpha-positive/T-bet-positive breast cancers expressing lower levels of FOXA1 (P = 0.0137) and GATA-3 (P = 0.0063) compared with ERalpha-positive/T-bet-negative breast cancers. Thus, T-bet expression in primary tumors and circulating insulin levels may serve as surrogate biomarkers to identify ERalpha-positive breast cancers with a dysfunctional hormonal network, enhanced growth factor signaling, and resistance to hormonal therapy.
Collapse
Affiliation(s)
- Kasi McCune
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
334
|
Masri S, Liu Z, Phung S, Wang E, Yuan YC, Chen S. The role of microRNA-128a in regulating TGFbeta signaling in letrozole-resistant breast cancer cells. Breast Cancer Res Treat 2010; 124:89-99. [PMID: 20054641 DOI: 10.1007/s10549-009-0716-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 12/23/2009] [Indexed: 12/19/2022]
Abstract
Resistance to endocrine therapy agents has presented a clinical obstacle in the treatment of hormone-dependent breast cancer. Our laboratory has initiated a study of microRNA regulation of signaling pathways that may result in breast cancer progression on aromatase inhibitors (AI). Microarray analysis of hormone refractory cell lines identified 115 differentially regulated microRNAs, of which 49 microRNAs were believed to be hormone-responsive. A group of microRNAs were inversely expressed in the AI-resistant lines versus LTEDaro and tamoxifen-resistant. We focused our work on hsa-miR-128a which was hormone-responsive and selectively up-regulated in the letrozole-resistant cell lines. Human miR-128a was predicted to target the TGFβ signaling pathway and indeed sensitivity to TGFβ was compromised in the letrozole-resistant cells, as compared to parental MCF-7aro. Human miR-128a was shown to negatively target TGFβRI protein expression by binding to the 3'UTR region of the gene. Inhibition of endogenous miR-128a resulted in resensitization of the letrozole-resistant lines to TGFβ growth inhibitory effects. These data suggest that the hormone-responsive miR-128a can modulate TGFβ signaling and survival of the letrozole-resistant cell lines. To our knowledge, this is the first study to address the role of microRNA regulation as well as TGFβ signaling in AI-resistant breast cancer cell lines. We believe that in addition to estrogen-modulation of gene expression, hormone-regulated microRNAs may provide an additional level of post-transcriptional regulation of signaling pathways critically involved in breast cancer progression and AI-resistance.
Collapse
Affiliation(s)
- Selma Masri
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | | | | | | | | | | |
Collapse
|
335
|
Sabnis G, Goloubeva O, Gilani R, Macedo L, Brodie A. Sensitivity to the aromatase inhibitor letrozole is prolonged after a "break" in treatment. Mol Cancer Ther 2010; 9:46-56. [PMID: 20053764 DOI: 10.1158/1535-7163.mct-09-0696] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Using a hormone-dependent xenograft model, we established that loss of response to letrozole was accompanied by upregulation of the Her-2/mitogen-activated protein kinase (MAPK) pathway and downregulation of estrogen receptor alpha (ERalpha) and aromatase activity. In our previous study, we showed that stopping letrozole treatment or adding trastuzumab could reverse acquired resistance. In this study, we compared the effects of intermittent letrozole treatment and switching treatment between letrozole and trastuzumab on tumor growth in an attempt to optimize discontinuous letrozole treatment. The mice were treated with letrozole until the tumors developed resistance and then were divided into three groups: (a) letrozole, (b) trastuzumab, and (c) "off" (Delta(4)A supplement only); tumors were collected every week to examine changes in tumor protein expression and activity. In off group tumors, Her-2/p-MAPK activation gradually decreased and ERalpha and aromatase protein (and activity) increased. Within the first week of trastuzumab treatment, Her-2 and MAPK were downregulated and ERalpha was upregulated. When letrozole-resistant MCF-7Ca tumors were taken off treatment for 4 weeks, the second course of letrozole treatment provided a much longer duration of response (P = 0.02). However, switching treatment to trastuzumab for 4 weeks did not provide any inhibition of tumor growth. Our studies revealed that the adaptation of cells to a low-estrogen environment by upregulation of Her-2/MAPK and downregulation of ERalpha/aromatase was reversed on letrozole withdrawal. The tumors once again became responsive to letrozole for a significant period. These results suggest that response to letrozole can be prolonged by a short "break" in the treatment.
Collapse
Affiliation(s)
- Gauri Sabnis
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | |
Collapse
|
336
|
Creighton CJ, Fu X, Hennessy BT, Casa AJ, Zhang Y, Gonzalez-Angulo AM, Lluch A, Gray JW, Brown PH, Hilsenbeck SG, Osborne CK, Mills GB, Lee AV, Schiff R. Proteomic and transcriptomic profiling reveals a link between the PI3K pathway and lower estrogen-receptor (ER) levels and activity in ER+ breast cancer. Breast Cancer Res 2010; 12:R40. [PMID: 20569503 PMCID: PMC2917035 DOI: 10.1186/bcr2594] [Citation(s) in RCA: 197] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 06/05/2010] [Accepted: 06/22/2010] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Accumulating evidence suggests that both levels and activity of the estrogen receptor (ER) and the progesterone receptor (PR) are dramatically influenced by growth-factor receptor (GFR) signaling pathways, and that this crosstalk is a major determinant of both breast cancer progression and response to therapy. The phosphatidylinositol 3-kinase (PI3K) pathway, a key mediator of GFR signaling, is one of the most altered pathways in breast cancer. We thus examined whether deregulated PI3K signaling in luminal ER+ breast tumors is associated with ER level and activity and intrinsic molecular subtype. METHODS We defined two independent molecular signatures of the PI3K pathway: a proteomic (reverse-phase proteomic array) PI3K signature, based on protein measurement for PI3K signaling intermediates, and a PI3K transcriptional (mRNA) signature based on the set of genes either induced or repressed by PI3K inhibitors. By using these signatures, we scored each ER+ breast tumor represented in multiple independent expression-profiling datasets (four mRNA, n = 915; one protein, n = 429) for activation of the PI3K pathway. Effects of PI3K inhibitor BEZ-235 on ER expression and activity levels and cell growth were tested by quantitative real-time PCR and cell proliferation assays. RESULTS Within ER+ tumors, ER levels were negatively correlated with the PI3K activation scores, both at the proteomic and transcriptional levels, in all datasets examined. PI3K signature scores were also higher in ER+ tumors and cell lines of the more aggressive luminal B molecular subtype versus those of the less aggressive luminal A subtype. Notably, BEZ-235 treatment in four different ER+ cell lines increased expression of ER and ER target genes including PR, and treatment with IGF-I (which signals via PI3K) decreased expression of ER and target genes, thus further establishing an inverse functional relation between ER and PI3K. BEZ-235 had an additional effect on tamoxifen in inhibiting the growth of a number of ER+ cell lines. CONCLUSIONS Our data suggest that luminal B tumors have hyperactive GFR/PI3K signaling associated with lower ER levels, which has been correlated with resistance to endocrine therapy. Targeting PI3K in these tumors might reverse loss of ER expression and signaling and restore hormonal sensitivity.
Collapse
Affiliation(s)
- Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Xiaoyong Fu
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Bryan T Hennessy
- Department of Systems Biology, M.D. Anderson, Houston, TX 77030, USA
| | - Angelo J Casa
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Yiqun Zhang
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Ana Maria Gonzalez-Angulo
- Department of Systems Biology, M.D. Anderson, Houston, TX 77030, USA,Department of Breast Medical Oncology, M.D. Anderson, Houston, TX 77030, USA
| | - Ana Lluch
- Department of Hematology Oncology, Hospital Clinico Universitario de Valencia, Av de Vicente Blasco Ibáñez, 17, 46010, Valencia, Spain
| | - Joe W Gray
- Life Sciences Division, Lawrence Berkeley National Laboratory, One Cyclotron Road, Berkeley, CA 94720, USA,Department of Laboratory Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, 1600 Divisadero Street, San Francisco, CA 94143, USA
| | - Powell H Brown
- Department of Clinical Cancer Prevention, Division of OVP, Cancer Prevention and Population Sciences, M.D. Anderson, Houston, TX 77030, USA
| | - Susan G Hilsenbeck
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - C Kent Osborne
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Gordon B Mills
- Department of Systems Biology, M.D. Anderson, Houston, TX 77030, USA
| | - Adrian V Lee
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| | - Rachel Schiff
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA
| |
Collapse
|
337
|
Saggar JK, Chen J, Corey P, Thompson LU. Dietary flaxseed lignan or oil combined with tamoxifen treatment affects MCF-7 tumor growth through estrogen receptor- and growth factor-signaling pathways. Mol Nutr Food Res 2009; 54:415-25. [DOI: 10.1002/mnfr.200900068] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
338
|
Wong ST, Goodin S. Overcoming drug resistance in patients with metastatic breast cancer. Pharmacotherapy 2009; 29:954-65. [PMID: 19637949 DOI: 10.1592/phco.29.8.954] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Metastatic breast cancer is generally considered to be incurable, with response rates and duration of response progressively declining with subsequent lines of treatment. Tumors are either intrinsically resistant to systemic therapy or acquire resistance at some point during multiple courses of therapy. Mechanisms of drug resistance are numerous and include accelerated drug efflux, drug activation and inactivation, alterations in drug target, processing of drug-induced damage, and evasion of apoptosis. Targeted anticancer agents for the treatment of breast cancer, such as hormonal agents or the more recently approved epidermal growth factor receptor inhibitors, are also associated with intrinsic and acquired resistance. A variety of strategies have been devised to prevent or overcome resistance to systemic anticancer therapy, including drug combinations and sequential regimens. However, it appears that resistance to established cytotoxic and targeted agents is inevitable. Novel agents with reduced susceptibility to resistance may prevent or delay the emergence of resistance and improve survival in patients with common solid tumors, including metastatic breast cancer. We are hopeful that further elucidation of the cellular and molecular processes that allow tumor cells to develop resistance and the use of new agents to combat these mechanisms will improve outcomes for patients with metastatic breast cancer.
Collapse
Affiliation(s)
- Serena T Wong
- Department of Medicine, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, New Jersey 08903, USA
| | | |
Collapse
|
339
|
Sukocheva O, Wang L, Verrier E, Vadas MA, Xia P. Restoring endocrine response in breast cancer cells by inhibition of the sphingosine kinase-1 signaling pathway. Endocrinology 2009; 150:4484-4492. [PMID: 19706837 DOI: 10.1210/en.2009-0391] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We previously demonstrated that sphingosine kinase-1 (SphK1) is an important mediator in the cytoplasmic signaling of estrogens, including Ca(2+) mobilization, ERK1/2 activation, and the epidermal growth factor receptor transactivation. Here we report for the first time that SphK1 activity is causally associated with endocrine resistance in MCF-7 human breast cancer cells. Enforced overexpression of human SphK1 in MCF-7 cells resulted in enhanced cell proliferation and resistance to tamoxifen-induced cell growth arrest and apoptosis. Tamoxifen-resistant (TamR) MCF-7 cells selected by prolonged exposure to 4-hydroxytamoxifen, exhibited higher levels in SphK1 expression and activity, compared with the control cells. Inhibition of SphK1 activity by either specific pharmaceutical inhibitors or the dominant-negative mutant SphK1(G82D) restored the antiproliferative and proapoptotic effects of tamoxifen in the TamR cells. Furthermore, silencing of SphK1, but not SphK2, expression by the specific small interference RNA also restored the tamoxifen responsiveness in the TamR cells. Thus, blockade of the SphK1 signaling pathway may reprogram cellular responsiveness to tamoxifen and abrogate antiestrogen resistance in human breast cancer cells.
Collapse
Affiliation(s)
- Olga Sukocheva
- Signal Transduction Laboratory, Centenary Institute, Newtown, New South Wales 2042, Australia
| | | | | | | | | |
Collapse
|
340
|
Implication of protein tyrosine phosphatase 1B in MCF-7 cell proliferation and resistance to 4-OH tamoxifen. Biochem Biophys Res Commun 2009; 387:748-53. [DOI: 10.1016/j.bbrc.2009.07.105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 07/22/2009] [Indexed: 12/12/2022]
|
341
|
Johnston S, Pippen J, Pivot X, Lichinitser M, Sadeghi S, Dieras V, Gomez HL, Romieu G, Manikhas A, Kennedy MJ, Press MF, Maltzman J, Florance A, O'Rourke L, Oliva C, Stein S, Pegram M. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor-positive metastatic breast cancer. J Clin Oncol 2009; 27:5538-46. [PMID: 19786658 DOI: 10.1200/jco.2009.23.3734] [Citation(s) in RCA: 745] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Cross-talk between human epidermal growth factor receptors and hormone receptor pathways may cause endocrine resistance in breast cancer. This trial evaluated the effect of adding lapatinib, a dual tyrosine kinase inhibitor blocking epidermal growth factor receptor and human epidermal growth factor receptor 2 (HER2), to the aromatase inhibitor letrozole as first-line treatment of hormone receptor (HR) -positive metastatic breast cancer (MBC). PATIENTS AND METHODS Postmenopausal women with HR-positive MBC were randomly assigned to daily letrozole (2.5 mg orally) plus lapatinib (1,500 mg orally) or letrozole and placebo. The primary end point was progression-free survival (PFS) in the HER2-positive population. Results In HR-positive, HER2-positive patients (n = 219), addition of lapatinib to letrozole significantly reduced the risk of disease progression versus letrozole-placebo (hazard ratio [HR] = 0.71; 95% CI, 0.53 to 0.96; P = .019); median PFS was 8.2 v 3.0 months, respectively. Clinical benefit (responsive or stable disease >or= 6 months) was significantly greater for lapatinib-letrozole versus letrozole-placebo (48% v 29%, respectively; odds ratio [OR] = 0.4; 95% CI, 0.2 to 0.8; P = .003). Patients with centrally confirmed HR-positive, HER2-negative tumors (n = 952) had no improvement in PFS. A preplanned Cox regression analysis identified prior antiestrogen therapy as a significant factor in the HER2-negative population; a nonsignificant trend toward prolonged PFS for lapatinib-letrozole was seen in patients who experienced relapse less than 6 months since prior tamoxifen discontinuation (HR = 0.78; 95% CI, 0.57 to 1.07; P = .117). Grade 3 or 4 adverse events were more common in the lapatinib-letrozole arm versus letrozole-placebo arm (diarrhea, 10% v 1%; rash, 1% v 0%, respectively), but they were manageable. CONCLUSION This trial demonstrated that a combined targeted strategy with letrozole and lapatinib significantly enhances PFS and clinical benefit rates in patients with MBC that coexpresses HR and HER2.
Collapse
|
342
|
Vivacqua A, Lappano R, De Marco P, Sisci D, Aquila S, De Amicis F, Fuqua SAW, Andò S, Maggiolini M. G protein-coupled receptor 30 expression is up-regulated by EGF and TGF alpha in estrogen receptor alpha-positive cancer cells. Mol Endocrinol 2009; 23:1815-26. [PMID: 19749156 DOI: 10.1210/me.2009-0120] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In the present study, we evaluated the regulation of G protein-coupled receptor (GPR)30 expression in estrogen receptor (ER)-positive endometrial, ovarian, and estrogen-sensitive, as well as tamoxifen-resistant breast cancer cells. We demonstrate that epidermal growth factor (EGF) and TGF alpha transactivate the GPR30 promoter and accordingly up-regulate GPR30 mRNA and protein levels only in endometrial and tamoxifen-resistant breast cancer cells. These effects exerted by EGF and TGF alpha were dependent on EGF receptor (EGFR) expression and activation and involved phosphorylation of the Tyr(1045) and Tyr(1173) EGFR sites. Using gene-silencing experiments and specific pharmacological inhibitors, we have ascertained that EGF and TGF alpha induce GPR30 expression through the EGFR/ERK transduction pathway, and the recruitment of c-fos to the activator protein-1 site located within GPR30 promoter sequence. Interestingly, we show that functional cross talk of GPR30 with both activated EGFR and ER alpha relies on a physical interaction among these receptors, further extending the potential of estrogen to trigger a complex stimulatory signaling network in hormone-sensitive tumors. Given that EGFR/HER2 overexpression is associated with tamoxifen resistance, our data may suggest that ligand-activated EGFR could contribute to the failure of tamoxifen therapy also by up-regulating GPR30, which in turn could facilitates the action of estrogen. In addition, important for resistance is the ability of tamoxifen to bind to and activate GPR30, the expression of which is up-regulated by EGFR activation. Our results emphasize the need for new endocrine agents able to block widespread actions of estrogen without exerting any stimulatory activity on transduction pathways shared by the steroid and growth factor-signaling networks.
Collapse
Affiliation(s)
- Adele Vivacqua
- Department of Pharmaco-Biology, University of Calabria, 87030 Rende, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Carver KC, Arendt LM, Schuler LA. Complex prolactin crosstalk in breast cancer: new therapeutic implications. Mol Cell Endocrinol 2009; 307:1-7. [PMID: 19524120 PMCID: PMC3190192 DOI: 10.1016/j.mce.2009.03.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 03/23/2009] [Indexed: 12/27/2022]
Abstract
The contributions of prolactin (PRL) to breast cancer are becoming increasingly recognized. To better understand the role for PRL in this disease, its interactions with other oncogenic growth factors and hormones must be characterized. Here, we review our current understanding of PRL crosstalk with other mammary oncogenic factors, including estrogen, epidermal growth factor (EGF) family members, and insulin-like growth factor-I (IGF-I). The ability of PRL to potentiate the actions of these targets of highly successful endocrine and molecular therapies suggests that PRL and/or its receptor (PRLR) may be an attractive therapeutic target(s). We discuss the potential benefit of PRL/PRLR-targeted therapy in combination with established therapies and implications for de novo and acquired resistance to treatment.
Collapse
Affiliation(s)
- Kristopher C. Carver
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, United States
- Biotechnology Training Program, University of Wisconsin-Madison, Madison, WI 53706, United States
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Lisa M. Arendt
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, United States
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Linda A. Schuler
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, United States
- Biotechnology Training Program, University of Wisconsin-Madison, Madison, WI 53706, United States
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, United States
- Corresponding author at: Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, United States. Tel.: +1 608 263 9825; fax: +1 608 263 3926. (L.A. Schuler)
| |
Collapse
|
344
|
Mauro L, Pellegrino M, Lappano R, Vivacqua A, Giordano F, Palma MG, Andò S, Maggiolini M. E-cadherin mediates the aggregation of breast cancer cells induced by tamoxifen and epidermal growth factor. Breast Cancer Res Treat 2009; 121:79-89. [PMID: 19593637 DOI: 10.1007/s10549-009-0456-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Accepted: 06/25/2009] [Indexed: 01/03/2023]
Abstract
In the present study, we evaluated the ability of 4-hydroxytamoxifen (OHT) and epidermal growth factor (EGF) to regulate homotypic adhesion in MCF7 breast cancer cells. Our results demonstrate that OHT and EGF activate the E-cadherin promoter, increase E-cadherin mRNA and protein expression and enhance homotypic aggregation of MCF7 cells. Interestingly, an ERalpha and EGFR cross-talk is involved in the E-cadherin expression by OHT and EGF, as demonstrated by knocking down either receptor. On the basis of our findings, the well-established cross-talk between ERalpha and EGFR could be extended to the modulation of E-cadherin expression by OHT and EGF. Thus, the potential ability of tamoxifen to induce cell-cell aggregation may contribute to the biologic response of pharmacologic intervention in patients with breast cancer.
Collapse
Affiliation(s)
- Loredana Mauro
- Department Cellular Biology, University of Calabria, Cosenza, Arcavacata-Rende (CS), 87030, Italy
| | | | | | | | | | | | | | | |
Collapse
|
345
|
Meacham WD, Antoon JW, Burow ME, Struckhoff AP, Beckman BS. Sphingolipids as determinants of apoptosis and chemoresistance in the MCF-7 cell model system. Exp Biol Med (Maywood) 2009; 234:1253-63. [PMID: 19546354 DOI: 10.3181/0902-mr-77] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
An estimated 182,640 women and 1,990 men were diagnosed with breast cancer in 2008, and approximately 40,480 women and 450 men died from the disease. Thus, continued mechanistic studies are needed to understand the causes and develop additional therapeutics for this complicated disease. The MCF-7 cell system is one of the most recognized models for estrogen receptor (ER)-positive breast cancer and has generated approximately 13,000 publications cited in PubMed to date. A number of clues for biological mechanisms related to apoptotic/anti-apoptotic pathways and chemoresistance were elucidated and summarized in our previous review. The focus of this review is new knowledge of the central role of sphingolipid signaling in apoptotic mechanisms in estrogen receptor-positive breast cancer. The ultimate goal is to target crucial steps in survival signaling pathways that may ultimately provide additional translational solutions to the successful pharmacologic treatment of breast cancer.
Collapse
Affiliation(s)
- William D Meacham
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
346
|
Abstract
Earlier, estrogens were considered simply the most important hormones involved in female physiology and reproduction. Nowadays it has become familiar that they have pivotal roles in gene regulation of cell differentiation and proliferation. There are many contradictions concerning the associations of female sexual steroids and cancer. Cancers of the highly estrogen dependent organs are in the forefront of tumors as they are regarded as hormone associated ones. However, re-evaluation of earlier results supporting the carcinogenic capacity of estrogen exhibited many shortcomings and controversies. Recently, the clinical studies on hormone replacement therapy in postmenopausal women justified beneficial anticancer effects in several organs even in the female breast. The newly revealed association between estrogen deficiency and oral cancer risk also means a contradiction of the traditional concept of estrogen-induced cancer. Distinction between cancers of moderately and highly estrogen dependent tumors can be based on their different epidemiological features. The vast majority of the so-called smoking associated malignancies of the moderately estrogen dependent organs occur typically in the late postmenopausal life of women when the ovarian estrogen production is fairly decreased. However cancers of the highly estrogen dependent organs such as breast, endometrium and ovary exhibit both premenopausal and postmenopausal occurrence. In spite of the different epidemiological data of these two groups of cancers the mechanism of gene regulation disorder in the background of tumor initiation cannot act through quite opposite pathways. This suggests that in moderately estrogen sensitive organs a serious, in the highly estrogen dependent sites even a mild estrogen deficiency is enough to provoke gene regulation disorders. The new findings both on smoking associated and hormone related cancers might lead to the same conversion; not estrogen but rather its deficiency may provoke cancer initiation.
Collapse
Affiliation(s)
- Zsuzsanna Suba
- Semmelweis Egyetem, Fogorvostudományi Kar Arc-, Allcsont-, Szájsebészeti és Fogászati Klinika, Orálpatológiai Részleg Budapest.
| |
Collapse
|
347
|
Johnston SR. Enhancing the Efficacy of Hormonal Agents with Selected Targeted Agents. Clin Breast Cancer 2009; 9 Suppl 1:S28-36. [DOI: 10.3816/cbc.2009.s.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
348
|
Santen RJ, Brodie H, Simpson ER, Siiteri PK, Brodie A. History of aromatase: saga of an important biological mediator and therapeutic target. Endocr Rev 2009; 30:343-75. [PMID: 19389994 DOI: 10.1210/er.2008-0016] [Citation(s) in RCA: 301] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Aromatase is the enzyme that catalyzes the conversion of androgens to estrogens. Initial studies of its enzymatic activity and function took place in an environment focused on estrogen as a component of the birth control pill. At an early stage, investigators recognized that inhibition of this enzyme could have major practical applications for treatment of hormone-dependent breast cancer, alterations of ovarian and endometrial function, and treatment of benign disorders such as gynecomastia. Two general approaches ultimately led to the development of potent and selective aromatase inhibitors. One targeted the enzyme using analogs of natural steroidal substrates to work out the relationships between structure and function. The other approach initially sought to block adrenal function as a treatment for breast cancer but led to the serendipitous finding that a nonsteroidal P450 steroidogenesis inhibitor, aminoglutethimide, served as a potent but nonselective aromatase inhibitor. Proof of the therapeutic concept of aromatase inhibition involved a variety of studies with aminoglutethimide and the selective steroidal inhibitor, formestane. The requirement for even more potent and selective inhibitors led to intensive molecular studies to identify the structure of aromatase, to development of high-sensitivity estrogen assays, and to "mega" clinical trials of the third-generation aromatase inhibitors, letrozole, anastrozole, and exemestane, which are now in clinical use in breast cancer. During these studies, unexpected findings led investigators to appreciate the important role of estrogens in males as well as in females and in multiple organs, particularly the bone and brain. These studies identified the important regulatory properties of aromatase acting in an autocrine, paracrine, intracrine, neurocrine, and juxtacrine fashion and the organ-specific enhancers and promoters controlling its transcription. The saga of these studies of aromatase and the ultimate utilization of inhibitors as highly effective treatments of breast cancer and for use in reproductive disorders serves as the basis for this first Endocrine Reviews history manuscript.
Collapse
Affiliation(s)
- R J Santen
- University of Virginia Health System, Division of Endocrinology, P.O. Box 801416, Charlottesville, Virginia 22908, USA.
| | | | | | | | | |
Collapse
|
349
|
Spears M, Bartlett J. The potential role of estrogen receptors and the SRC family as targets for the treatment of breast cancer. Expert Opin Ther Targets 2009; 13:665-74. [DOI: 10.1517/14728220902911509] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
350
|
Nierth-Simpson EN, Martin MM, Chiang TC, Melnik LI, Rhodes LV, Muir SE, Burow ME, McLachlan JA. Human uterine smooth muscle and leiomyoma cells differ in their rapid 17beta-estradiol signaling: implications for proliferation. Endocrinology 2009; 150:2436-45. [PMID: 19179429 PMCID: PMC2671893 DOI: 10.1210/en.2008-0224] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Uterine leiomyomas, benign uterine smooth muscle tumors that affect 30% of reproductive-aged women, are a significant health concern. The initiation event for these tumors is unclear, but 17beta-estradiol (E2) is an established promoter of leiomyoma growth. E2 not only alters transcription of E2-regulated genes but also can rapidly activate signaling pathways. The aim of our study is to investigate the role of rapid E2-activated cytoplasmic signaling events in the promotion of leiomyomas. Western blot analysis revealed that E2 rapidly increases levels of phosphorylated protein kinase C alpha (PKC alpha) in both immortalized uterine smooth muscle (UtSM) and leiomyoma (UtLM) cell lines, but increases levels of phosphorylated ERK1/2 only in UtLM cells. Our studies demonstrate a paradoxical effect of molecular and pharmacological inhibition of PKC alpha on ERK1/2 activation and cellular proliferation in UtLM and UtSM cells. PKC alpha inhibition decreases levels of phosphorylated ERK1/2 and proliferation in UtLM cells but raises these levels in UtSM cells. cAMP-PKA signaling is rapidly activated only in UtSM cells with E2 and inhibits ERK1/2 activation and proliferation. We therefore propose a model whereby E2's rapid activation of PKC alpha and cAMP-PKA signaling plays a central role in the maintenance of a low proliferative index in normal uterine smooth muscle via its inhibition of the MAPK cascade and these pathways are altered in leiomyomas to promote MAPK activation and proliferation. These studies demonstrate that rapid E2-signaling pathways contribute to the promotion of leiomyomas.
Collapse
|