首页 | 本学科首页   官方微博 | 高级检索  
     


An Absence of Stromal Caveolin-1 Expression Predicts Early Tumor Recurrence and Poor Clinical Outcome in Human Breast Cancers
Authors:Agnieszka K. Witkiewicz  Abhijit Dasgupta  Federica Sotgia  Isabelle Mercier  Richard G. Pestell  Michael Sabel  Celina G. Kleer  Jonathan R. Brody  Michael P. Lisanti
Affiliation:2. Jefferson Center for Pancreatic, Biliary, and Related Cancers, the Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania;3. Departments of Cancer Biology and Medical Oncology, the Division of Biostatistics, Thomas Jefferson University, Philadelphia, Pennsylvania;4. Department of Pharmacology and Experimental Therapeutics, the Stem Cell Biology and Regenerative Medicine Center, Thomas Jefferson University, Philadelphia, Pennsylvania;5. Department of Pathology, and/or Surgery, University of Michigan Medical School, Ann Arbor, Michigan
Abstract:Previously, we showed that caveolin-1 (Cav-1) expression is down-regulated in human breast cancer-associated fibroblasts. However, it remains unknown whether loss of Cav-1 occurs in the breast tumor stroma in vivo. Here, we immunostained a well-annotated breast cancer tissue microarray with antibodies against Cav-1 and scored its stromal expression. An absence of stromal Cav-1 was associated with early disease recurrence, advanced tumor stage, and lymph node metastasis, resulting in a 3.6-fold reduction in progression-free survival. When tamoxifen-treated patients were selected, an absence of stromal Cav-1 was a strong predictor of poor clinical outcome, suggestive of tamoxifen resistance. Interestingly, in lymph node-positive patients, an absence of stromal Cav-1 predicted an 11.5-fold reduction in 5-year progression-free survival. Clinical outcomes among patients positive for HER2, and patients triple-negative for estrogen receptor, progesterone receptor and HER2, were also strictly dependent on stromal Cav-1 levels. When our results were adjusted for tumor and nodal staging, an absence of stromal Cav-1 remained an independent predictor of poor outcome. Thus, stromal Cav-1 expression can be used to stratify human breast cancer patients into low-risk and high-risk groups, and to predict their risk of early disease recurrence at diagnosis. Based on related mechanistic studies, we suggest that breast cancer patients lacking stromal Cav-1 might benefit from anti-angiogenic therapy in addition to standard regimens. We conclude that Cav-1 functions as a tumor suppressor in the stromal microenvironment.Carcinoma cells grow in a complex tumor microenvironment composed of (i) non-epithelial cells (including fibroblasts, pericytes, endothelial, and inflammatory cells), (ii) extracellular matrix, and (iii) secreted diffusible growth factors/cytokines.1,2,3 Although under normal physiological conditions the stroma serves as an important barrier to malignant transformation, its role changes during neoplastic transformation. Instead, the stroma plays a key role in driving cancer cell invasiveness and progression.4 Recently, it was demonstrated that fibroblasts isolated from tumor stroma can promote tumor growth.1,2,3,5 This population of tissue fibroblasts termed “cancer associated fibroblasts” (CAFs) is characterized by a hyperproliferative phenotype, and these cells secrete increased amounts of growth factors, extracellular matrix components, and matrix metalloproteinases.5,6 CAFs also show an ability to prevent cancer cell apoptosis, induce cancer cell proliferation, and stimulate tumor angiogenesis.7 In vitro studies of breast carcinomas showed that CAFs mixed with epithelial carcinoma cells are more proficient than normal fibroblasts at enhancing tumor growth and give rise to highly vascularized tumors.8 To date, the mechanisms that govern the conversion of benign mammary stromal fibroblasts to tumor-associated fibroblasts are poorly understood, and their relationship to disease outcome has not been addressed.Down-regulation of caveolin-1 (Cav-1) is one of the mechanisms implicated in the oncogenic transformation of fibroblasts. Caveolins are the principal protein component of caveolae, which are located at the cell surface in most cell types.9 One of the caveolins, Cav-1, plays a major role in tumorigenesis through its various functions such as lipid transport, membrane trafficking, gene regulation, and signal transduction.10 In cell culture, the transformation of NIH-3T3 fibroblasts with various activated oncogenes, such as H-Ras (G12V), Bcr-Abl, or v-Abl, causes dramatic reductions in Cav-1 protein expression.11,12Furthermore, knock-down of endogenous Cav-1 in NIH-3T3 fibroblasts promotes anchorage-independent growth in soft agar and tumor formation in nude mice, which could be reversed by Cav-1 re-expression.13 Finally, Cav-1−/− null fibroblasts have a hyperproliferative phenotype (similar to CAFs) and Cav-1 re-expression drives their arrest in the G0/G1 phase of the cell cycle.14 Taken together, these data suggest that loss of Cav-1 leads to the oncogenic transformation of fibroblasts, where Cav-1 normally functions as a transformation suppressor that prevents cell cycle progression.Using primary cell cultures established from surgically excised breast tumors, we recently demonstrated that Cav-1 is down-regulated in human breast CAFs when compared with matching normal fibroblasts isolated from the same patient.15 In addition, orthotopic transplantation of Cav-1+/+ tumor tissue into the mammary stroma of Cav-1−/− null mice results in up to a ∼twofold increase in tumor mass, functionally demonstrating that the mammary stroma of Cav-1−/− mice behaves as a tumor promoter.16 However, to date, there is no study addressing the clinical significance of stromal Cav-1 expression in invasive carcinoma of the breast in vivo.The aim of this study was to evaluate the in vivo stromal expression of Cav-1 in a large series of invasive breast carcinomas and to examine the association between stromal Cav-1 expression, clinicopathological variables, and patient outcome. Our results indicate that loss of stromal caveolin-1 is a novel breast cancer biomarker that predicts early disease recurrence, metastasis, survival, and tamoxifen-resistance. Clinical outcome in HER2(+) and triple-negative (estrogen receptor [ER]−/progesterone receptor [PR]−/HER2−) patients was also strictly dependent on stromal Cav-1 levels. Remarkably, in lymph node-positive [LN(+)] patients, an absence of stromal Cav-1 was associated with an ∼11.5-fold reduction in 5-year progression-free survival. As such, Cav-1 may function as a critical tumor/metastasis suppressor in the mammary stromal compartment.
Keywords:
本文献已被 ScienceDirect 等数据库收录!
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号