GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
  • 2
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2020
    In:  Breast Cancer Research and Treatment Vol. 179, No. 3 ( 2020-02), p. 577-584
    In: Breast Cancer Research and Treatment, Springer Science and Business Media LLC, Vol. 179, No. 3 ( 2020-02), p. 577-584
    Type of Medium: Online Resource
    ISSN: 0167-6806 , 1573-7217
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2004077-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Nucleic Acids Research, Oxford University Press (OUP), Vol. 40, No. 10 ( 2012-5), p. 4368-4384
    Type of Medium: Online Resource
    ISSN: 1362-4962 , 0305-1048
    RVK:
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2012
    detail.hit.zdb_id: 1472175-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Research Vol. 11, No. 10_Supplement ( 2013-10-01), p. B108-B108
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 11, No. 10_Supplement ( 2013-10-01), p. B108-B108
    Abstract: The tumor microenvironment (TME) has been shown to play a vital role in tumor development and progression. Carcinoma associated fibroblasts (CAFs) form a very essential component of the TME. In fact, the TME in different types of carcinomas, particularly carcinomas of the breast, is comprised mainly of fibroblasts. CAFs in breast cancer TME secrete cytokines and growth factors that are known to activate a multitude of signaling pathways in breast cancer cells. One of these pathways is the MAPK pathway, a signaling pathway activated downstream of ERBB receptor tyrosine kinase family members. We have previously demonstrated that activation of the MAPK pathway represses estrogen receptor (ER) expression leading to an ER-negative phenotype in breast cancer. We have generated dissociated CAF cell cultures from ER+, ER-/Her2+, and triple negative (TN) primary breast tumors. We have characterized these CAFs on the basis of fibroblast markers, epithelial markers, microarray analysis, soft agar assay and in vivo tumorigenecity studies. Our results show that these CAF populations from primary breast tumors are pure populations of CAF cell cultures. These CAFs provide a model that allows us to examine the role of CAF interaction with ER+ breast cancer cells regarding activation of MAPK and subsequent repression of ER expression. We demonstrate that treatment of the ER+ MCF-7 cell line with conditioned media (CM) from the CAFs results in transient activation of MAPK signaling and subsequent repression of ER. Continuous exposure of ER+ breast cancer cells to soluble factors from CAFs also results in activation of MAPK and down-regulation of ER expression. Importantly, CM from human mammary fibroblasts (HMFs) and CAFs generated from an ER+ tumor do not down-regulate ER expression. Gene expression analysis and cytokine arrays indicate key differences in gene and cytokine expression between HMFs and the ER+ tumor CAFs compared to the two CAFs generated from ER- tumors. We have previously identified a miRNA signature associated with hyperactivation of MAPK signaling (hMAPK). This signature identifies the majority of ER- breast cancers as well as a population of ER+ breast cancers that express lower levels of ER. Like the ER- breast cancers identified by this signature, ER+ cancers identified by this signature exhibit gene expression patterns indicative of activated MAPK signaling, and exhibit significantly increased disease recurrence and significantly reduced disease survival. Here we show that breast tumors cancers bearing this hMAPK-miRNA signature have elevated expression of numerous stromal markers associated with poor clinical outcome, as well as a number of microRNAs that are differentially expressed between normal human mammary fibroblasts (HMFs) and mammary tumor carcinoma CAFs. Some of these miRNAs have been established as targeting ER. Using reporter constructs to investigate microRNA regulation of ER, we have observed that treatment of ER+ breast cancer cells with CM from ER- CAFs results in enhanced microRNA-dependent repression of ER. These data are suggestive of a role for CAF secreted factors in activating MAPK in breast cancer cells leading to repression of ER. In addition, CAFs are able to increase the expression of hMAPK-miRNAs that can down-regulate ER expression either via the activation of MAPK in the cancer cell or secreted directly from the CAFs. We hypothesize that CAFs may contribute to an ER-negative phenotype in breast cancer cells not only by activating MAPK signaling in the tumor cells via secretion of growth factors, but also potentially by transfer of MAPK-regulated miRNAs to the breast cancer cells as well. Citation Format: Sanket H. Shah, Philip Miller, Katherine Drews-Elger, Joeli Brinkman, Stefania Lairet, Alana Steinberg, Dorraya El-Ashry. Cancer-associated fibroblasts contribute to establishment of ER-negative breast cancer phenotype through secreted factors and miRNAs. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research: Genetics, Biology, and Clinical Applications; Oct 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2013;11(10 Suppl):Abstract nr B108.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2392-2392
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2392-2392
    Abstract: Approximately 35% of breast cancers lack expression of estrogen receptor (ER) protein. ER-negative (ER-) breast cancer carries a worse prognosis than ER positive (ER+) breast cancer, but more importantly, it precludes use of less toxic endocrine therapies. Our objective was to establish primary cultures from dissociation of ER- breast tumors in order to provide an alternative cellular model that can be used as a valuable tool for both in vitro and in vivo studies of ER- breast cancer. A total of eight ER- tumors were successfully dissociated into primary cultures: four from ER-/PR-/H2N- and four from ER-/PR-/H2N+ primary breast tumors with a 100% success rate. The epithelial-enriched cell pellet was placed in culture and for experiments carried for a maximum of 30 passages. These cells will be hereafter referred to as “dissociated tumor” (DT) cultures. Cells were grown in 2D culture and their in vitro morphology, proliferation rates, mammosphere and soft agar colony formation ability, and CD44/CD24 surface marker expression (tumor initiating cell (TIC) content) are determined. In addition we have performed gene expression profiling and established their tumorigenic potential in NOD/SCID female mice. Each culture exhibited its own relatively unique morphology; two of the eight cultures grew mainly as suspensions while the remaining six DT cultures grew with a higher percentage of attached, mesenchymal phenotype cells. Proliferation rates ranged from 38 to 60 hours, and while all of the DT cultures had the ability to form mammospheres, five of the eight DTs (DT13, DT16, DT22, DT25 and DT28) formed colonies in soft agar. Analysis of CD44 and CD24 surface markers expression showed that while all DT cultures were CD44 positive, expression of CD24 varied among DTs. Of the eight DT cultures, we found five of them to have a high ( & gt;85%) CD44+/CD24-/lo cell content, one the CD44+/CD24high phenotype and two of eight have populations with increasing levels of CD24 expression ranging from -/lo to medium. This CD44+/CD24-/lo phenotype is stable when examined over several passages. Microarray analysis comparing the DT cultures to cancer cell lines showed that they clustered with each other and with several breast cancer cell lines of known ER- status and EGFR or Her2 overexpression status. In agreement with the soft agar assay, DT16, DT22, DT25 and DT28 had the ability to form tumors when injected into the mammary fat pad of female NOD/SCID mice. In summary, our study describes primary cultures of dissociated ER negative breast cancer cells that provide an alternative, primary cell based model that allows both in vitro as well as in vivo experimental approaches. The use of these types of cellular models may lead to a better understanding of ER- breast cancer biology as well as being a valuable tool for screening potential therapeutic options for ER- breast cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2392. doi:10.1158/1538-7445.AM2011-2392
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3335-3335
    Abstract: Primary human breast cancer subpopulations with a CD44+ CD24NEG/LOW ESA+ phenotype exhibit self-renewal and generate xenograft tumors with as few as 100 cells in immunodeficient mice. While CD44 expression is strongly associated with self renewal in several cancers, the relative contributions of CD24 negative versus low subpopulations to stemness is poorly defined. Early reports have not distinguished negative from low CD24 expression in human lines and primary tumors, and both, together with CD44+ status have been linked to breast cancer stem cell phenotypes. In contrast, certain reports have associated CD24 expression with tumor progression and metastasis. Here, we show increased mitogenic kinase activation, expression of ESC and EMT genes in the CD24LOW subpopulation of CD44+ cells in both triple negative MDA-MB-231 breast cancer cells and primary dissociated breast cancers (DTs) compared to the CD24NEG cells. Moreover, tumorigenicity was increased and metastasis arose exclusively from orthotopic xeno-implantation of the CD44+CD24LOW cells. MDA-MB-231 and two different primary DTs were live sorted into CD44+CD24NEG and CD44+CD24LOW enriched subpopulations. Cells expressing other putative stem cell markers, ALDH1 and ESA were almost entirely CD44+CD24LOW. CD44+CD24LOWcells showed higher PI3-kinase/Akt, MAPK, and Src activites compared to CD44+ CD24NEG cells. While both subpopulations formed soft agar colonies and/or mammospheres, those arising from CD44+CD24LOW were greater in number and size than from CD44+CD24NEG cells. When cultured in 2D after sorting, CD44+ CD24LOW cells gave rise to both CD44+ CD24LOW and CD44+ CD24NEG progeny while CD44+ CD24NEG yielded only CD44+ CD24NEG progeny. CD44+CD24LOW expressed higher levels of ESC genes and EMT-associated genes than CD44+CD24NEG cells and uniquely expressed stem cell-associated miRNAs. On orthotopic injection into BalbC nude mice, both subpopulations initiated tumors with as few as 100 cells, however tumors arising from CD44+CD24LOW cells had a shorter latency and grew more rapidly than those arising from CD44+CD24NEG cells. In vivo imaging of luciferase positive cells showed CD44+CD24LOW cells yield metastasis while CD44+CD24NEG cells did not. Our data demonstrate, for the first time, distinct biological properties between the CD44+CD24LOW and CD44+CD24NEG enriched subpopulations of MDA-MB-231 and primary human breast cancer DTs. Low surface expression of CD24 is associated with ALDH1 and ESA+ status, increased self-renewal as shown by mammosphere and colony formation, tumorigenicity and metastatic potential of CD44+ cells. The ability to isolate and characterize such TSC-enriched subpopulations will be necessary for development of therapeutic strategies that preferentially target breast cancer stem cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3335. doi:10.1158/1538-7445.AM2011-3335
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 5_Supplement ( 2023-03-01), p. P4-08-10-P4-08-10
    Abstract: Background: Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype with a high metastatic rate. Despite significant advances in breast cancer therapeutics, due to the lack of specific therapeutic targets in TNBC, cytotoxic chemotherapy is still the mainstay of treatment for this BC subtype. Preclinical studies have shown that the Receptor for Advanced Glycation End-products (RAGE) drives the progression and metastasis of aggressive cancer subtypes, including TNBC. RAGE plays a multifaceted role in driving tumorigenesis and metastasis through tumor cell-intrinsic mechanisms, such as cancer cell invasion, migration and epithelial-mesenchymal transition, and tumor cell-extrinsic mechanisms. This multifaceted role in cancer progression and metastasis makes RAGE a promising therapeutic target in the prevention and treatment of breast cancer. Here we tested the preclinical anti-metastasic efficacy of two small molecule RAGE inhibitors; TTP48 (Azeliragon) and FPS-ZM1. Importantly, TTP488 displays a high safety profile in human trials and has previously undergone Phase 3 clinical trials for Alzheimer’s disease. While FPS-ZM1 is a well-known RAGE inhibitor in preclinical cancer models, TTP488 has not been tested for its anti-cancer activity in breast cancer. Methods: We tested the in vitro anti-metastatic effect of TTP488 and FPS-ZM1 on cancer cell migration and invasion in Boyden chamber assays with TNBC cell lines (MDA-MB-231 and 4T-1). We used the 4175 highly metastatic MDA-MB-231 variant in xenograft studies in NSG mice to test the efficacy of the RAGE inhibitors in vivo on tumor progression and metastasis. Experimental metastasis assays were performed with tail-vein injection of 4T-1 cells into BALBc mice. We performed bulk RNA sequencing on the MDA-MB-231/4175/NSG tumors to unveil and compare the mechanism of action of the two small molecule RAGE inhibitors. Results: Our results showed that TTP488 and FPS-ZM1 impaired mechanisms of metastasis in vitro with both MDA-MB231/4175 and 4T-1 cells. TTP488 and FPS-ZM1 significantly inhibited MDA-MB231/4175 cell metastasis from the orthotopic site in NSG mice without displaying any deleterious effects on mouse health. In the syngeneic 4T-1/BALBc model, both TTP488 and FPS-ZM1 impaired metastasis in tail-vein injected experimental metastasis assays. Transcriptomic analysis of primary xenograft tumors from NSG mice revealed that TTP488 and FPS-ZM1 displayed high concordance in gene expression changes. Pathway enrichment analysis showed that both RAGE inhibitors affected metastatic pathways, including focal adhesion, ECM-receptor interaction, cell cycle, and DNA replication. Conclusions: These results show that TTP488 impairs metastasis of multiple highly aggressive TNBC models for the first time. Importantly, as TTP488 displays a high safety profile in human trials, this study provides the rationale for evaluating TTP488 in clinical trials to treat or prevent metastatic breast cancer. Citation Format: Melinda Magna, Gyong Ha Hwang, Alec McIntosh, Katherine Drews-Elger, Masaru Takabatake, Barbara Mera, Taekyoung Kwak, Philip Miller, Marc Lippman, Barry I. Hudson. Azeliragon (TTP488), an orally- available small molecule RAGE inhibitor, reduces metastasis in preclinical mouse models of breast cancer [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P4-08-10.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2270-2270
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2270-2270
    Abstract: Background: Breast cancer is most common malignant state in women and 20% of these patients will develop metastasis during the course of their disease. Further understanding is needed of the process and mechanisms of metastasis. Our lab and others have been shown that Receptor of Advanced-Glycation End-products (RAGE) plays a role in tumorigenesis and metastasis. RAGE is highly expressed in various cancers including breast cancer and its protein levels correlate with poor patient outcome in breast cancer and other cancers. Activation of RAGE results in increased proliferation, migration and invasion of cancer cells. Further studies in mice have shown its blockade may be a therapeutic target to reduce tumor growth and the resulting metastasis. Methods: Using the breast cancer cell model (MDA-MB-231) and its organotropic sister cells lines selected in vivo for increased metastasis to lung (4175) and bone (1833), we tested the role of RAGE in driving mechanisms of metastasis in vitro and in vivo. Results: First, we demonstrated that the highly metastatic variant of 231 cells (4175 and 1833) have increased expression level of RAGE compared to MDA-MB-231 parental cells. Moreover, RAGE knockdown by shRNA in 4175 and 231 parental cells showed decreased cell invasion in transwell assays compared to control scramble shRNA. Ectopic over-expression of RAGE in parental 231 cells led to increased migratory and invasive properties compared to vector control cells, without affecting cell proliferation or viability. To explore the underlying mechanisms we probed for differences in RAGE signaling pathways including MEK/ERK, p38, AKT and SAPK/JNK; with MEK/ERK and Akt showing activation. Using chemical inhibitors of these pathways we demonstrated that the MEK/ERK, but not other pathways inhibited RAGE-driven cell invasion in transwell assays. To validate our data in vivo, we performed mammary fat pad injection of 4175 cells (RAGE and scr shRNA) in NSG mice. Tumor growth and weight was impaired in RAGE gene knockdown 4175 cells compared to scramble (scr) controls. Analysis of lung tissue retrieved from mice revealed RAGE knockdown in 4175 cells resulted in less metastasis compared to 4175 scr control cells. Conclusion: We have shown that higher RAGE levels are seen in metastatic human breast cancer BC cells. RAGE gene knockdown in metastatic BC cells reduces their invasive properties in vitro and reduces tumor progression and metastasis in vivo in NSG mice. These data highlight RAGE as a novel therapeutic target for metastatic disease in breast and other cancers. Citation Format: Taekyoung Kwak, Katherine Drews-Elger, Dekuang Zhao, Alexander Besser, Ayse Ergonul, Joyce M. Slingerland, Marc E. Lippman, Barry I. Hudson. RAGE-ligand signaling drives breast cancer invasion and metastasis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2270. doi:10.1158/1538-7445.AM2015-2270
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: npj Breast Cancer, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2023-07-13)
    Abstract: Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic cancer subtype, which is generally untreatable once it metastasizes. We hypothesized that interfering with the Receptor for Advanced Glycation End-products (RAGE) signaling with the small molecule RAGE inhibitors (TTP488/Azeliragon and FPS-ZM1) would impair TNBC metastasis and impair fundamental mechanisms underlying tumor progression and metastasis. Both TTP488 and FPS-ZM1 impaired spontaneous and experimental metastasis of TNBC models, with TTP488 reducing metastasis to a greater degree than FPS-ZM1. Transcriptomic analysis of primary xenograft tumor and metastatic tissue revealed high concordance in gene and protein changes with both drugs, with TTP488 showing greater potency against metastatic driver pathways. Phenotypic validation of transcriptomic analysis by functional cell assays revealed that RAGE inhibition impaired TNBC cell adhesion to multiple extracellular matrix proteins (including collagens, laminins, and fibronectin), migration, and invasion. Neither RAGE inhibitor impaired cellular viability, proliferation, or cell cycle in vitro. Proteomic analysis of serum from tumor-bearing mice revealed RAGE inhibition affected metastatic driver mechanisms, including multiple cytokines and growth factors. Further mechanistic studies by phospho-proteomic analysis of tumors revealed RAGE inhibition led to decreased signaling through critical BC metastatic driver mechanisms, including Pyk2, STAT3, and Akt. These results show that TTP488 impairs metastasis of TNBC and further clarifies the signaling and cellular mechanisms through which RAGE mediates metastasis. Importantly, as TTP488 displays a favorable safety profile in human studies, our study provides the rationale for evaluating TTP488 in clinical trials to treat or prevent metastatic TNBC.
    Type of Medium: Online Resource
    ISSN: 2374-4677
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2843288-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...