Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katherine Drews-Elger is active.

Publication


Featured researches published by Katherine Drews-Elger.


Oncogene | 2015

VEGF drives cancer-initiating stem cells through VEGFR-2/Stat3 signaling to upregulate Myc and Sox2

Dekuang Zhao; Chendong Pan; Jun Sun; Candace A. Gilbert; Katherine Drews-Elger; Diana J. Azzam; Manuel Picon-Ruiz; Minsoon Kim; W. Ullmer; Dorraya El-Ashry; Chad J. Creighton; Joyce M. Slingerland

Vascular endothelial growth factor-A (VEGF), a potent angiogenic factor, is also implicated in self-renewal in several normal tissue types. VEGF has been shown to drive malignant stem cells but mechanisms thereof and tumor types affected are not fully characterized. Here, we show VEGF promotes breast and lung cancer stem cell (CSC) self-renewal via VEGF receptor-2 (VEGFR-2)/STAT3-mediated upregulation of Myc and Sox2. VEGF increased tumor spheres and aldehyde dehydrogenase activity, both proxies for stem cell function in vitro, in triple-negative breast cancer (TNBC) lines and dissociated primary cancers, and in lung cancer lines. VEGF exposure before injection increased breast cancer-initiating cell abundance in vivo yielding increased orthotopic tumors, and increased metastasis from orthotopic primaries and following tail vein injection without further VEGF treatment. VEGF rapidly stimulated VEGFR-2/JAK2/STAT3 binding and activated STAT3 to bind MYC and SOX2 promoters and induce their expression. VEGFR-2 knockdown or inhibition abrogated VEGF-mediated STAT3 activation, MYC and SOX2 induction and sphere formation. Notably, knockdown of either STAT3, MYC or SOX2 impaired VEGF-upregulation of pSTAT3, MYC and SOX2 expression and sphere formation. Each transcription factor, once upregulated, appears to promote sustained activation of the others, creating a feed-forward loop to drive self-renewal. Thus, in addition to angiogenic effects, VEGF promotes tumor-initiating cell self-renewal through VEGFR-2/STAT3 signaling. Analysis of primary breast and lung cancers (>1300 each) showed high VEGF expression, was prognostic of poor outcome and strongly associated with STAT3 and MYC expression, supporting the link between VEGF and CSC self-renewal. High-VEGF tumors may be most likely to escape anti-angiogenics by upregulating VEGF, driving CSC self-renewal to re-populate post-treatment. Our work highlights the need to better define VEGF-driven cancer subsets and supports further investigation of combined therapeutic blockade of VEGF or VEGFR-2 and JAK2/STAT3.


PLOS ONE | 2012

A New Mouse Model for the Study of Human Breast Cancer Metastasis

Elizabeth Iorns; Katherine Drews-Elger; Toby M. Ward; Sonja Dean; Jennifer Clarke; Deborah L. Berry; Dorraya El Ashry; Marc E. Lippman

Breast cancer is the most common cancer in women, and this prevalence has a major impact on health worldwide. Localized breast cancer has an excellent prognosis, with a 5-year relative survival rate of 85%. However, the survival rate drops to only 23% for women with distant metastases. To date, the study of breast cancer metastasis has been hampered by a lack of reliable metastatic models. Here we describe a novel in vivo model using human breast cancer xenografts in NOD scid gamma (NSG) mice; in this model human breast cancer cells reliably metastasize to distant organs from primary tumors grown within the mammary fat pad. This model enables the study of the entire metastatic process from the proper anatomical site, providing an important new approach to examine the mechanisms underlying breast cancer metastasis. We used this model to identify gene expression changes that occur at metastatic sites relative to the primary mammary fat pad tumor. By comparing multiple metastatic sites and independent cell lines, we have identified several gene expression changes that may be important for tumor growth at distant sites.


Cancer Research | 2016

Interactions between Adipocytes and Breast Cancer Cells Stimulate Cytokine Production and Drive Src/Sox2/miR-302b–Mediated Malignant Progression

Manuel Picon-Ruiz; Chendong Pan; Katherine Drews-Elger; Kibeom Jang; Alexandra H. Besser; Dekuang Zhao; Cynthia Morata-Tarifa; Minsoon Kim; Tan A. Ince; Diana J. Azzam; Seth A. Wander; Bin Wang; Burcu Ergonul; Ram H. Datar; Richard J. Cote; Guy A. Howard; Dorraya El-Ashry; Pablo Torné-Poyatos; Juan A. Marchal; Joyce M. Slingerland

Consequences of the obesity epidemic on cancer morbidity and mortality are not fully appreciated. Obesity is a risk factor for many cancers, but the mechanisms by which it contributes to cancer development and patient outcome have yet to be fully elucidated. Here, we examined the effects of coculturing human-derived adipocytes with established and primary breast cancer cells on tumorigenic potential. We found that the interaction between adipocytes and cancer cells increased the secretion of proinflammatory cytokines. Prolonged culture of cancer cells with adipocytes or cytokines increased the proportion of mammosphere-forming cells and of cells expressing stem-like markers in vitro. Furthermore, contact with immature adipocytes increased the abundance of cancer cells with tumor-forming and metastatic potential in vivo. Mechanistic investigations demonstrated that cancer cells cultured with immature adipocytes or cytokines activated Src, thus promoting Sox2, c-Myc, and Nanog upregulation. Moreover, Sox2-dependent induction of miR-302b further stimulated cMYC and SOX2 expression and potentiated the cytokine-induced cancer stem cell-like properties. Finally, we found that Src inhibitors decreased cytokine production after coculture, indicating that Src is not only activated by adipocyte or cytokine exposures, but is also required to sustain cytokine induction. These data support a model in which cancer cell invasion into local fat would establish feed-forward loops to activate Src, maintain proinflammatory cytokine production, and increase tumor-initiating cell abundance and metastatic progression. Collectively, our findings reveal new insights underlying increased breast cancer mortality in obese individuals and provide a novel preclinical rationale to test the efficacy of Src inhibitors for breast cancer treatment.


Embo Molecular Medicine | 2013

Triple negative breast cancer initiating cell subsets differ in functional and molecular characteristics and in γ-secretase inhibitor drug responses

Diana J. Azzam; Dekuang Zhao; Jun-Jun Sun; Andy J. Minn; Prathibha Ranganathan; Katherine Drews-Elger; Xiaoqing Han; Manuel Picon-Ruiz; Candace A. Gilbert; Seth A. Wander; Anthony J. Capobianco; Dorraya El-Ashry; Joyce M. Slingerland

Increasing evidence suggests that stem‐like cells mediate cancer therapy resistance and metastasis. Breast tumour‐initiating stem cells (T‐ISC) are known to be enriched in CD44+CD24neg/low cells. Here, we identify two T‐ISC subsets within this population in triple negative breast cancer (TNBC) lines and dissociated primary breast cancer cultures: CD44+CD24low+ subpopulation generates CD44+CD24neg progeny with reduced sphere formation and tumourigenicity. CD44+CD24low+ populations contain subsets of ALDH1+ and ESA+ cells, yield more frequent spheres and/or T‐ISC in limiting dilution assays, preferentially express metastatic gene signatures and show greater motility, invasion and, in the MDA‐MB‐231 model, metastatic potential. CD44+CD24low+ but not CD44+CD24neg express activated Notch1 intracellular domain (N1‐ICD) and Notch target genes. We show N1‐ICD transactivates SOX2 to increase sphere formation, ALDH1+ and CD44+CD24low+cells. Gamma secretase inhibitors (GSI) reduced sphere formation and xenograft growth from CD44+CD24low+ cells, but CD44+CD24neg were resistant. While GSI hold promise for targeting T‐ISC, stem cell heterogeneity as observed herein, could limit GSI efficacy. These data suggest a breast T‐ISC hierarchy in which distinct pathways drive developmentally related subpopulations with different anti‐cancer drug responsiveness.


Breast Cancer Research and Treatment | 2014

Primary breast tumor-derived cellular models: characterization of tumorigenic, metastatic, and cancer-associated fibroblasts in dissociated tumor (DT) cultures

Katherine Drews-Elger; Joeli Brinkman; Philip Miller; Sanket H. Shah; J. Chuck Harrell; Thiago G. da Silva; Zheng Ao; Amy Schlater; Diana J. Azzam; Kathleen M. Diehl; Dafydd G. Thomas; Joyce M. Slingerland; Charles M. Perou; Marc E. Lippman; Dorraya El-Ashry

Our goal was to establish primary cultures from dissociation of breast tumors in order to provide cellular models that may better recapitulate breast cancer pathogenesis and the metastatic process. Here, we report the characterization of six cellular models derived from the dissociation of primary breast tumor specimens, referred to as “dissociated tumor (DT) cells.” In vitro, DT cells were characterized by proliferation assays, colony formation assays, protein, and gene expression profiling, including PAM50 predictor analysis. In vivo, tumorigenic and metastatic potential of DT cultures was assessed in NOD/SCID and NSG mice. These cellular models differ from recently developed patient-derived xenograft models in that they can be used for both in vitro and in vivo studies. PAM50 predictor analysis showed DT cultures similar to their paired primary tumor and as belonging to the basal and Her2-enriched subtypes. In vivo, three DT cultures are tumorigenic in NOD/SCID and NSG mice, and one of these is metastatic to lymph nodes and lung after orthotopic inoculation into the mammary fat pad, without excision of the primary tumor. Three DT cultures comprised of cancer-associated fibroblasts (CAFs) were isolated from luminal A, Her2-enriched, and basal primary tumors. Among the DT cells are those that are tumorigenic and metastatic in immunosuppressed mice, offering novel cellular models of ER-negative breast cancer subtypes. A group of CAFs provide tumor subtype-specific components of the tumor microenvironment (TME). Altogether, these DT cultures provide closer-to-primary cellular models for the study of breast cancer pathogenesis, metastasis, and TME.


Breast Cancer Research and Treatment | 2014

Infiltrating S100A8+ myeloid cells promote metastatic spread of human breast cancer and predict poor clinical outcome

Katherine Drews-Elger; Elizabeth Iorns; Alexandra Dias; Philip Miller; Toby M. Ward; Sonja Dean; Jennifer Clarke; Adriana Campion-Flora; Daniel Nava Rodrigues; Jorge S. Reis-Filho; James M. Rae; Dafydd G. Thomas; Deborah L. Berry; Dorraya El-Ashry; Marc E. Lippman

The mechanisms by which breast cancer (BrC) can successfully metastasize are complex and not yet fully understood. Our goal was to identify tumor-induced stromal changes that influence metastatic cell behavior, and may serve as better targets for therapy. To identify stromal changes in cancer-bearing tissue, dual-species gene expression analysis was performed for three different metastatic BrC xenograft models. Results were confirmed by immunohistochemistry, flow cytometry, and protein knockdown. These results were validated in human clinical samples at the mRNA and protein level by retrospective analysis of cohorts of human BrC specimens. In pre-clinical models of BrC, systemic recruitment of S100A8+ myeloid cells—including myeloid-derived suppressor cells (MDSCs)—was promoted by tumor-derived factors. Recruitment of S100A8+ myeloid cells was diminished by inhibition of tumor-derived factors or depletion of MDSCs, resulting in fewer metastases and smaller primary tumors. Importantly, these MDSCs retain their ability to suppress T cell proliferation upon co-culture. Secretion of macrophage inhibitory factor (MIF) activated the recruitment of S100A8+ myeloid cells systemically. Inhibition of MIF, or depletion of MDSCs resulted in delayed tumor growth and lower metastatic burden. In human BrC specimens, increased mRNA and protein levels of S100A8+ infiltrating cells are highly associated with poor overall survival and shorter metastasis free survival of BrC patients, respectively. Furthermore, analysis of nine different human gene expression datasets confirms the association of increased levels of S100A8 transcripts with an increased risk of death. Recruitment of S100A8+ myeloid cells to primary tumors and secondary sites in xenograft models of BrC enhances cancer progression independent of their suppressive activity on T cells. In clinical samples, infiltrating S100A8+ cells are associated with poor overall survival. Targeting these molecules or associated pathways in cells of the tumor microenvironment may translate into novel therapeutic interventions and benefit patient outcome.


Breast Cancer Research and Treatment | 2013

Paget’s disease of the nipple

Ana Sandoval-Leon; Katherine Drews-Elger; Carmen Gomez-Fernandez; Monica M. Yepes; Marc E. Lippman

Paget’s disease of the breast is a disorder of the nipple–areola complex that, while rare, is often associated with an underlying carcinoma. It is characterized by eczematoid changes of the nipple. Two theories have been proposed to explain the pathogenesis of Paget’s disease. The Epidermotropic, which is the most accepted theory, suggests that Paget’s cells originate from ductal cancer cells that had migrated from the underlying breast parenchyma. It is supported by the predominance of breast cancer markers found in Paget’s disease. This article provides an overview of Paget’s disease of the breast with special attention to immunohistochemistry and raises the question of new therapeutic approaches.


Oncogene | 2017

Targeting of RAGE-ligand signaling impairs breast cancer cell invasion and metastasis.

Taekyoung Kwak; Katherine Drews-Elger; Ayse Ergonul; Philip Miller; A. Braley; G. H. Hwang; Dekuang Zhao; Alexandra H. Besser; Y. Yamamoto; H. Yamamoto; Dorraya El-Ashry; Joyce M. Slingerland; Marc E. Lippman; Barry I. Hudson

The receptor for advanced glycation end products (RAGE) is highly expressed in various cancers and is correlated with poorer outcome in breast and other cancers. Here we tested the role of targeting RAGE by multiple approaches in the tumor and tumor microenvironment, to inhibit the metastatic process. We first tested how RAGE impacts tumor cell-intrinsic mechanisms using either RAGE overexpression or knockdown with short hairpin RNAs (shRNAs). RAGE ectopic overexpression in breast cancer cells increased MEK-EMT (MEK-epithelial-to-mesenchymal transition) signaling, transwell invasion and soft agar colony formation, and in vivo promoted lung metastasis independent of tumor growth. RAGE knockdown with multiple independent shRNAs in breast cancer cells led to decreased transwell invasion and soft agar colony formation, without affecting proliferation. In vivo, targeting RAGE shRNA knockdown in human and mouse breast cancer cells, decreased orthotopic tumor growth, reduced tumor angiogenesis and recruitment of inflammatory cells, and markedly decreased metastasis to the lung and liver in multiple xenograft and syngeneic mouse models. To test the non-tumor cell microenvironment role of RAGE, we performed syngeneic studies with orthotopically injected breast cancer cells in wild-type and RAGE-knockout C57BL6 mice. RAGE-knockout mice displayed striking impairment of tumor cell growth compared with wild-type mice, along with decreased mitogen-activated protein kinase signaling, tumor angiogenesis and inflammatory cell recruitment. To test the combined inhibition of RAGE in both tumor cell-intrinsic and non-tumor cells of the microenvironment, we performed in vivo treatment of xenografted tumors with FPS-ZM1 (1 mg/kg, two times per week). Compared with vehicle, FPS-ZM1 inhibited primary tumor growth, inhibited tumor angiogenesis and inflammatory cell recruitment and, most importantly, prevented metastasis to the lung and liver. These data demonstrate that RAGE drives tumor progression and metastasis through distinct tumor cell-intrinsic and -extrinsic mechanisms, and may represent a novel and therapeutically viable approach for treating metastatic cancers.


Breast Cancer Research and Treatment | 2011

Simultaneous measurement of ERα, HER2, and PhosphoERK1/2 in breast cancer cell lines by flow cytometry

Ulas Darda Bayraktar; Tae Kon Kim; Katherine Drews-Elger; Cara L. Benjamin; Dorraya El-Ashry; Eric Wieder; Krishna V. Komanduri

The activation of human epidermal growth factor receptor-2 (HER2) results in the activation of the mitogen-activated protein kinase (MAPK) cascade that may lead to the resistance to anti-estrogen therapy in estrogen receptor (ERα) expressing breast cancer by means of phosphorylation of ERα in the N-terminal region by phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) and by means of decreasing ERα expression. Immunohistochemistry is the most widely used technique for the detection of ERα and HER2 in breast cancer specimens, however, is inadequate in its ability to assess the relationship between ERα, HER2, and MAPK cascade at the single cell level. To clear this major hurdle, we devised a novel flow cytometric method to quantify the expression of ERα, HER2, and the activation of MAPK cascade simultaneously in single cells. The method was validated by concurrent Western blotting in established cell lines: MDA-231 (ERα and HER2-negative), MCF-7 (ERα-positive, HER2-negative), MCF-7 cells overexpressing ERα after long-term incubation in estrogen-free medium, and HER2 transfected MCF7 cells. Using the flow cytometry method, we confirmed the previous finding that ERα expression is down-regulated upon epidermal growth factor mediated ERK1/2 phosphorylation in EGFR/MCF-7 cells. To our knowledge, this is the first such assay to incorporate simultaneous single cell measurement for all of these pathways, which may prove useful to determine the intratumoral heterogeneity in breast tumors or the receptor status in circulating tumor cells.


Cancer Research | 2016

Abstract P2-05-07: RAGE-ligand signaling drives breast cancer metastasis through affecting cells of the tumor and microenvironment

Taekyoung Kwak; Katherine Drews-Elger; Ayse Ergonul; Dekuang Zhao; Alexandra H. Besser; Joyce M. Slingerland; Marc E. Lippman; Barry I. Hudson

Breast cancer is most common malignant state in women, with 20% of these patients developing 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 it may be a therapeutic target to reduce tumor growth and the resulting metastasis. Further understanding is needed of the role of RAGE in driving metastasis through affecting cells of both the tumor and tumor stroma to design novel therapeutics. 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 tumor metastasis in vitro and in vivo with xenograft mouse models. To test the role of RAGE in the tumor microenvironment we used the AT-3 syngeneic breast cancer cell model in C57BL6 wild-type and RAGE knockout mice. 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. 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. RAGE knockdown by shRNA in 4175 and 231 parental cells showed decreased cell invasion in transwell assays compared to control scramble shRNA. To validate our data in vivo, we performed mammary fat pad injection of 4175 cells (RAGE and scr shRNA) in NOD SCID gamma mice. Tumor growth and weight was impaired in RAGE gene knockdown 4175 cells compared to scramble (scr) controls. Analysis of lung and liver tissue retrieved from mice revealed RAGE knockdown in 4175 cells prevented metastasis compared to 4175 scr control cells. To test the role of RAGE on non-tumor cells of the breast stroma we next performed syngeneic studies with AT-3 cells (MMTV-PyMT spontaneous BC cell model), by injection into the mammary fat pad of wild-type and RAGE knockout C57BL6 immunocompetent mice. RAGE knockout mice (RAGE -/-) displayed striking impairment of tumor cell growth compared to wild-type (RAGE +/+) mice. We are currently testing whether novel RAGE inhibitors impact breast cancer progression and metastasis. These data highlight RAGE drives breast cancer progression and metastasis through affecting both tumor cell intrinsic and non-tumor cell microenvironment effects. Future studies will demonstrate the potential of RAGE inhibition as a novel therapeutic approach for preventing and treating metastatic disease in breast and other cancers. Citation Format: Kwak T, Drews-Elger K, Ergonul A, Zhao D, Besser A, Slingerland JM, Lippman ME, Hudson BI. RAGE-ligand signaling drives breast cancer metastasis through affecting cells of the tumor and microenvironment. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-05-07.

Collaboration


Dive into the Katherine Drews-Elger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge