Network


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

Hotspot


Dive into the research topics where Qiuyu Wu is active.

Publication


Featured researches published by Qiuyu Wu.


Cancer Cell | 2008

Survival of Cancer Cells Is Maintained by EGFR Independent of Its Kinase Activity

Zhang Weihua; Rachel Tsan; Wei Chien Huang; Qiuyu Wu; Chao Hua Chiu; Isaiah J. Fidler; Mien Chie Hung

Expression of the epidermal growth factor receptor (EGFR), a receptor tyrosine kinase associated with cell proliferation and survival, is overactive in many tumors of epithelial origin. Blockade of the kinase activity of EGFR has been used for cancer therapy; however, by itself, it does not seem to reach maximum therapeutic efficacy. We report here that in human cancer cells, the function of kinase-independent EGFR is to prevent autophagic cell death by maintaining intracellular glucose level through interaction and stabilization of the sodium/glucose cotransporter 1 (SGLT1).


Neuro-oncology | 2014

Role of the endothelin axis in astrocyte- and endothelial cell-mediated chemoprotection of cancer cells

Seung Wook Kim; Hyun Jin Choi; Ho Jeong Lee; Junqin He; Qiuyu Wu; Robert R. Langley; Isaiah J. Fidler; Sun Jin Kim

BACKGROUND Recent evidence suggests that astrocytes protect cancer cells from chemotherapy by stimulating upregulation of anti-apoptotic genes in those cells. We investigated the possibility that activation of the endothelin axis orchestrates survival gene expression and chemoprotection in MDA-MB-231 breast cancer cells and H226 lung cancer cells. METHODS Cancer cells, murine astrocytes, and murine fibroblasts were grown in isolation, and expression of endothelin (ET) peptides and ET receptors (ETAR and ETBR) compared with expression on cancer cells and astrocytes (or cancer cells and fibroblasts) that were co-incubated for 48 hours. Type-specific endothelin receptor antagonists were used to evaluate the contribution of ETAR and ETBR to astrocyte-induced activation of the protein kinase B (AKT)/mitogen-activated protein kinase (MAPK) signal transduction pathways, anti-apoptotic gene expression, and chemoprotection of cancer cells. We also investigated the chemoprotective potential of brain endothelial cells and microglial cells. RESULTS Gap junction signaling between MDA-MB-231 cancer cells and astrocytes stimulates upregulation of interleukin 6 (IL-6) and IL-8 expression in cancer cells, which increases ET-1 production from astrocytes and ET receptor expression on cancer cells. ET-1 signals for activation of AKT/MAPK and upregulation of survival proteins that protect cancer cells from taxol. Brain endothelial cell-mediated chemoprotection of cancer cells also involves endothelin signaling. Dual antagonism of ETAR and ETBR is required to abolish astrocyte- and endothelial cell-mediated chemoprotection. CONCLUSIONS Bidirectional signaling between astrocytes and cancer cells involves upregulation and activation of the endothelin axis, which protects cancer cells from cytotoxicity induced by chemotherapeutic drugs.


Clinical Cancer Research | 2015

Macitentan, a Dual Endothelin Receptor Antagonist, in Combination with Temozolomide Leads to Glioblastoma Regression and Long-term Survival in Mice

Sun Jin Kim; Ho Jeong Lee; Mark Seungwook Kim; Hyun Jin Choi; Junqin He; Qiuyu Wu; Kenneth D. Aldape; Jeffrey S. Weinberg; W. K. Alfred Yung; Charles A. Conrad; Robert R. Langley; Francois Lehembre; Urs Regenass; Isaiah J. Fidler

Purpose: The objective of the study was to determine whether astrocytes and brain endothelial cells protect glioma cells from temozolomide through an endothelin-dependent signaling mechanism and to examine the therapeutic efficacy of the dual endothelin receptor antagonist, macitentan, in orthotopic models of human glioblastoma. Experimental Design: We evaluated several endothelin receptor antagonists for their ability to inhibit astrocyte- and brain endothelial cell–induced protection of glioma cells from temozolomide in chemoprotection assays. We compared survival in nude mice bearing orthotopically implanted LN-229 glioblastomas or temozolomide-resistant (LN-229Res and D54Res) glioblastomas that were treated with macitentan, temozolomide, or both. Tumor burden was monitored weekly with bioluminescence imaging. The effect of therapy on cell division, apoptosis, tumor-associated vasculature, and pathways associated with cell survival was assessed by immunofluorescent microscopy. Results: Only dual endothelin receptor antagonism abolished astrocyte- and brain endothelial cell–mediated protection of glioma cells from temozolomide. In five independent survival studies, including temozolomide-resistant glioblastomas, 46 of 48 (96%) mice treated with macitentan plus temozolomide had no evidence of disease (P < 0.0001), whereas all mice in other groups died. In another analysis, macitentan plus temozolomide therapy was stopped in 16 mice after other groups had died. Only 3 of 16 mice eventually developed recurrent disease, 2 of which responded to additional cycles of macitentan plus temozolomide. Macitentan downregulated proteins associated with cell division and survival in glioma cells and associated endothelial cells, which enhanced their sensitivity to temozolomide. Conclusions: Macitentan plus temozolomide are well tolerated, produce durable responses, and warrant clinical evaluation in glioblastoma patients. Clin Cancer Res; 21(20); 4630–41. ©2015 AACR.


Cancer Research | 2011

Abstract 601: Macitentan (ACT-064992), a tissue-targeting endothelin receptor antagonist enhances therapeutic efficacy of paclitaxel by modulating survival pathways in orthotopic models of metastatic human ovarian cancer

Sun Jin Kim; Jang Seong Kim; Seung Wook Kim; Emily C. Brantley; Seok Joong Yun; Junqin He; Maya K. Marva; Fahao Zhang; Qiuyu Wu; Francois Lehembre; Urs Regenass; Isaiah J. Fidler

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Introduction: In 2009, ovarian cancer was the leading cause of death from gynecologic cancer in the United States. Efforts to develop tools for early detection and therapeutic regimens to overcome drug resistance of the ovarian cancer have not made a significant achievement to prolong the median survial of the patients. Potential treatments for ovarian cancers that have become resistant to standard chemotherapies include modulators of tumor cell survival, such as endothelin receptor (ETR) antagonist. We investigated the therapeutic efficacy of the dual ETR antagonist, macitentan, on human ovarian cancer cells, SKOV3ip1 and IGROV1, growing orthotopically in nude mice. Materials and Methods: Mice were injected with one million cells of SKOV3ip1 or IGROV1. Ten days later, mice with established disease were randomized into vehicle (control), paclitaxel (weekly, intraperitoneal injections), macitentan (daily oral administrations) or a combination of paclitaxel and macitentan treatment groups. After 4 weeks of treatment, mice were necropsied and tumor incidence, tumor weight, and incidence of ascites were recorded. Tumor tissues were processed for immunohistochemical analyses. Results: Treatment with paclitaxel decreased tumor weight and volume of ascites. Combination therapy with macitentan and paclitaxel reduced tumor incidence and further reduced tumor weight and volume of ascites when compared with paclitaxel alone. Macitentan alone occasionally reduced tumor weight but alone had no effect on tumor incidence or ascites. Immunohistochemical analyses revealed that treatment with macitentan and macitentan plus paclitaxel inhibited the phosphorylation of ETRs and suppressed the survival pathways of tumor cells by decreasing the levels of pVEGFR2, pAkt, and pMAPK. The dose of macitentan necessary for inhibition of phosphorylation correlated with the dose required to increase antitumor efficacy of paclitaxel. Conclusion: Treatment with macitentan enhanced the cytotoxicity mediated by paclitaxel as measured by the degree of apoptosis in tumor cells and tumor-associated endothelial cells. Collectively, these results show that administration of macitentan in combination with paclitaxel prevents the progression of ovarian cancer in the peritoneal cavity of nude mice in part by inhibiting survival pathways of both tumor cells and tumor-associated endothelial 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 601. doi:10.1158/1538-7445.AM2011-601


Cancer Research | 2016

Abstract 2943: Hypoxia induced resistance to chemotherapy is regulated by the endothelin receptor axis

Hyun Kyung Yu; Ho Jeong Lee; Fahao Zhang; Qiuyu Wu; Isaiah J. Fidler; Sun Jin Kim

Introduction Tumor cells growing in the brain are resistant to chemotherapy. We have recently demonstrated that the activation of the endothelin receptors A and B axis is involved in the induction of resistance to chemotherapy by primary brain tumor cells and by brain metastases. Treatment with a dual endothelin receptor antagonist in combination with chemotherapy produced significant therapy of orthotopically growing glioblastoma cells as well as breast cancer and lung cancer brain metastases. Since the milieu of tumors in the brain is hypoxic (0.5% - 1% O2), we investigated whether the chemo-resistance of the tumor cells is linked to hypoxia mediated activation of the endothelin receptor axis. Materials and Methods Human glioma (LN229), breast cancer (MDA231), and lung adenocarcinoma (PC14) cell lines were cultured in DMEM supplemented with 5% FBS. The endothelin receptor A (ETAR) and/or B (ETBR) of cancer cells were knocked down by shRNA. ETAR and ETBR were activated by incubating the tumor cells with exogenous endothelin-1 for 15 minutes or blocked by treating the parental or engineered cells with ETAR-specific antagonist, BQ123 (100nM), and/or ETBR-specific antagonist, BQ788 (100nM), for 2 hours prior to the addition of paclitaxel (5ng/ml), temozolomide (100μg/ml) or cisplatinum (5μg/ml). The cultures were placed into incubators with an atmosphere of 20%, 6% or 1% oxygen for 48 hours (MDA231 cells) or 72 hours (LN229 and PC14 cells). Tumor cells were then plated into 96-well plates to determine chemosensitivity by the MTT assay or into 6-well plates for western blots, or into chamber slides for immunohistochemical analyses. Expression of survival-related proteins such as pETAR, pETBR, pAkt, pMAPK, pNFκB, GSTA5, TWIST1 or Bcl2L1, were then determined. Results Stimulation of parental MDA-231, PC-14, or LN229 cells with exogenous ET-1 induced significant resistance against all tested chemotherapeutic agents. The resistance was abolished only when both the ETAR and ETBR were antagonized by a combination of BQ123 and BQ788. Parental cells cultured under hypoxia were also significantly resistant to chemotherapy and treatment of these cells with BQ123 and BQ788 reversed this resistance. The effects of exogenous ET-1 on the induction of chemo-resistance or BQ123 and BQ788 on restoration of chemo-sensitivity were not found in any cancer cells devoid of ETAR and ETBR. In all cases, the chemo-resistance of tumor cells was associated with increased level of expression of proteins related to cell survival. Conclusion These data suggest that the endothelin receptor axis plays a role in hypoxia induced resistance of cancer cells to chemotherapy. Additional studies are warranted to determine the functional changes of the endothelin receptor axis in hypoxia. Citation Format: Hyun Kyung Yu, Ho Jeong Lee, Fahao Zhang, Qiuyu Wu, Isaiah J. Fidler, Sun Jin Kim. Hypoxia induced resistance to chemotherapy is regulated by the endothelin receptor axis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2943.


Cancer Research | 2015

Abstract 5154: Diagnostic serum markers for metastatic brain tumors identified by cross-species hybridization of microarrays in animal models

Ho Jeong Lee; Sun Jin Kim; Hyun Kyung Yu; Seung Wook Kim; Qiuyu Wu; Junqin He; Isaiah J. Fidler

Introduction: Cancer-related mortality of many tumors is due to development of metastasis to the brain with limited response to conventional therapies. Diagnosis of metastasis to the brain depends on symptoms or imaging techniques, which often fail to detect small brain lesions. Serologic markers enabling early diagnosis of metastatic brain tumors would therefore significantly improve clinical outcome. Using the cross-species hybridization of microarrays differentiating human from mouse gene signatures, we identified proteins significantly increased in brain metastases were detected in the sera of mice bearing human cancer metastases to the brain and determined whether these can be potentially diagnostic. Materials and Methods: Human breast cancer (MDA-MB-231), lung cancer (PC-14), melanoma (A375), colon cancer (KM12), prostate cancer (PC-3P) and renal cancer (SN12) cells were implanted into relative primary and metastatic organs of nude mice. RNAs were extracted from tumor tissues and processed for differential hybridization of microarray. To identify genes differentially expressed in brain lesions, the random variance t test or one-way ANOVA was performed for the two-group or multi-group analysis, respectively. In the next set of experiments, we determined whether coded proteins differentially expressed in brain metastasis can be detected in the sera. MDA-MB-231 and PC-14 cells were implanted into mammary fat pad, lung or brain. Tumor tissues and sera were collected for immunohistochemical (IHC) analyses and antibody array with 13 available antibodies detecting proteins coded by genes differentially expressed in the brain metastases. Results: In the first set of experiments, common 210 genes were identified to be differentially expressed in metastatic brain tumors, as compared with tumors growing in other organs such as mammary fat pad, lung, skin, cecum, prostate, kidney, and bone. Among the proteins coded by the 210 genes, 13 with commercially available antibodies were selected for validation. In the second set of experiments, IHC revealed that all these 13 proteins were expressed in metastasis produced by MDA-MB-231 and PC-14. Compared to sera of mice bearing primary tumors, elevated level of 10 proteins was detected in sera collected from mice with brain metastases. Conclusion: These data demonstrated that cross-species differential hybridization of microarray with class comparison of gene expression using orthotopic animal is a novel approach to identify potential diagnostic serum markers for brain metastases. Studies to detect these proteins in the sera of patients with brain metastases are now planned. Citation Format: Ho Jeong Lee, Sun Jin Kim, Hyun Kyung Yu, Seung Wook Kim, Qiuyu Wu, Junqin He, Isaiah J. Fidler. Diagnostic serum markers for metastatic brain tumors identified by cross-species hybridization of microarrays in animal models. [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 5154. doi:10.1158/1538-7445.AM2015-5154


Cancer Research | 2014

Abstract 4878: Modification of gene expression in tumor cells by the culture microenvironment

Seung Wook Kim; Sun Jin Kim; Ho-Jeong Lee; Junqin He; Qiuyu Wu; Erica J. Lawson; Isaiah J. Fidler

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Purpose: Since cell culture techniques were developed in 1950s, in vitro assays using monolayer cell cultures have played a major role in cancer research. Despite the absence of cross-talk between tumor cells and their respective host organ-microenvironment, data produced by in vitro assays have often been used to address challenge. However, the potential discrepancy between in vitro and in vivo mircoenvironments should not be overlooked. We have investigated the influence of culture conditions of the biology of tumor cells by comparing the genetic profiles of tumor cells growing in different culture conditions, such as culture media, sera, concentration of sera, and cell confluence. Materials and Methods: We profiled the gene expression of human breast cancer tumor cells (MDA-MB-231) that were cultured for 48 hours under various culture conditions; in different media (MEM, DMEM, RPMI-1640), different sera (FBS, horse serum), different concentrations of FBS (10%, 0.1%), and different cell confluence (90%, 50%) using microarray analysis of total cellular RNA using an Illumina platform (human HT-12v12 expression beadchip) Results: Upon analysis, 2,234 genes were differentially expressed among cultures with varied cell confluence, 2,981 genes differentially expressed in cultures with various FBS concentrations, 422 genes were differentially expressed in cells cultured with different sera, and 8,925 genes were differentially expressed in cells cultured with different media. Of 8,925 genes, 10 (ARHGEF6, CAPN3, COPG, DAMM2, GSTK1, PGM1, QPCT, QPRT, SLC1A4, and VAT1) were identified as commonly upregulated in confluent cultures, regardless of the media, as compared with sub-confluent cultures. In addition, 5 genes (IL7R, MPP4, NOP56, G3BP1 and IFP38) were identified as commonly downregulated in confluent cultures. Collectively, these data indicate that the gene expression profiles varied significantly across different cultures conditions. Conclusion:These results demonstrated that even under commonly “simplified” culture conditions, gene expression profiles can significantly vary across cells exposed to different culture conditions. Some of these conditions parallel those found in vivo, i.e., sub-confluent versus confluent cultures, where cell division is more active in the former than the latter condition. The significant differences in gene expression by cells exposed to fetal bovine serum as compared to horse serum are likely caused by different levels of growth factors within the sera. Likewise, the different gene expressions in tumor cells growing in different media can be attributed to different levels of glucose and salts present. Collectively, our data indicate that any change in culture conditions can lead to significant changes in the gene expression of tumor cells. Hence, to obtain reproducible data, one must strictly adhere to specified experimental details. Citation Format: Seung Wook Kim, Sun Jin Kim, Ho-jeong Lee, Junqin He, Qiuyu Wu, Erica J. Lawson, Isaiah J. Fidler. Modification of gene expression in tumor cells by the culture microenvironment. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4878. doi:10.1158/1538-7445.AM2014-4878


Cancer Research | 2014

Abstract 3779: Treatment of experimental brain metastasis of human breast cancer by macitentan, a dual antagonist of endothelin receptors combined with paclitaxel

Ho Jeong Lee; Sun Jin Kim; Seung Wook Kim; Junqin He; Qiuyu Wu; Erica J. Lawson; Francois Lehembre; Urs Regenass; Isaiah J. Fidler

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Purpose: Breast cancer is a leading cause of cancer-related mortality among women in the United States. The major cause of death is due to metastasis that is resistant to conventional therapy. The median survival of women with brain metastases is measured in months due to resistance to all standard therapies. Thus, the development of effective treatment options for these patients is mandatory. We have reported that astrocytes can protect tumor cells from chemotherapeutic drugs by a mechanism involving the phosphorylation of endothelin receptors on tumor cells (Kim et al, AACR, 2012). Moreover, we observed a heterogeneous expression of endothelin receptors A and B in clinical samples of breast cancer brain metastasis. We therefore examined whether blockade of the endothelin axis by using macitentan, a dual endothelin antagonist, combined with paclitaxel can produce therapeutic effects in an orthotopic model of human breast cancer brain metastasis. Materials and Methods: Twenty thousand MDA-MB-231 human breast cancer cells were injected into the internal carotid artery of female nude mice to produce orthotopic brain metastasis. Two weeks later, the mice were randomized into 4 groups (n=10): (1) control group - mice receiving vehicle (p.o., daily; i.p. injection once per week); (2) paclitaxel group - mice receiving paclitaxel (8 mg/kg; i.p. injection once per week) and vehicle (p.o.; daily); (3) macitentan group - mice receiving macitentan (10 mg/kg; p.o.; daily) and vehicle (i.p. injection, once per week); and (4) paclitaxel plus macitentan group. Moribund mice were euthanized, days of survival were recorded, and the brains were harvested and processed for histology and immunohistochemical analyses. The study was repeated with luciferase-labeled MDA-MB-231 cells and produced similar therapeutic results supported by IVIS imaging. Results: After 130-140 days of treatment, the survival was significantly higher in mice treated with the combination of paclitaxel and macitentan (80%) as compared with control (20%), paclitaxel (20%), and macitentan alone (40%) (p<0.0001, log-rank test). Immunohistochemical analyses revealed that the phosphorylation of endothelin receptors A and B on tumor cells and tumor-associated endothelial cells were inhibited by macitentan. Treatment with macitentan also suppressed the expression of survival proteins such as pAKT, pMAPK, BCL2L1, TWIST1, and GSTA5 in tumor cells. More importantly, only the combination therapy induced significant apoptosis of tumor cells and tumor-associated endothelial cells and reduced the number of KI67 positive tumor cells. IVIS imaging confirmed regression of brain lesions. Conclusion: Treatment with macitentan, a dual endothelin receptor antagonist, combined with paclitaxel leads to regression of experimental breast cancer brain metastasis and therefore should be considered for clinical development. Citation Format: Ho Jeong Lee, Sun Jin Kim, Seung Wook Kim, Junqin He, Qiuyu Wu, Erica J. Lawson, Francois Lehembre, Urs Regenass, Isaiah J. Fidler. Treatment of experimental brain metastasis of human breast cancer by macitentan, a dual antagonist of endothelin receptors combined with paclitaxel. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3779. doi:10.1158/1538-7445.AM2014-3779


Cancer Research | 2013

Abstract 4951: The role of endothelin axis signaling pathway in astrocyte mediated chemoprotection of tumor cells.

Seung Wook Kim; Sun Jin Kim; Hyun Jin Choi; Ho-Jeong Lee; Junqin He; Qiuyu Wu; Erica J. Lawson; Isaiah J. Fidler

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Purpose: More than 40% of patients with the breast cancer develop brain metastasis that are resistant to conventional chemotherapeutic agents. We have reported that reactive astrocytes establish gap junction communication channels with adjacent tumor cells and protect tumor cells from chemotherapy by up-regulation of survival genes in the tumor cells. We now report on the mechanism by which astrocytes up-regulate expression of the survival genes in tumor cells. Materials and Methods: Human breast cancer cells, MDA-MB-231 cultured with murine astrocytes produce endothelin 1 (ET-1) leading to expression and activation of endothelin receptors (ETRs). Using small interfering RNA (siRNA) of ETAR and ETBR, and ETRs specific antagonists (BQ123, BQ788, and [PD145065][1]) demonstrated the correlation between the activation of the endothelin axis in tumor cells and the induction of survival genes (BCL2L1, GSTA5, and TWIST1) leading to chemoresistance. Results: Co-culture of MDA-MB-231 cells and murine astrocytes induced up-regulation of ETRs (ETAR and ETBR) and activation of endothelin receptors axis in tumor cells as well as increased production of ET-1 by murine astrocytes. Activation of the ETRs by exogenous ET-1 up-regulated the expression of survival genes, such as BCL2L1, GSTA5 and TWIST1 via activation of phosphorylated AKT/MAPKinase signal transduction pathways, leading to resistance of tumor cells to several chemotherapeutic agents. Knockdown of both ETAR and ETBR by siRNAs or treatment of tumor cells with dual ETR inhibitor abolished the chemoprotection induced by astrocytes. Conclusion: ET axis signaling pathway plays an essential role in astrocytes-mediated induction of chemoresistance of tumor cells by up-regulation of survival genes in tumor cells. Simultaneous blockade of both ETR can prevent tumor cells from developing resistance to chemotherapeutic agents. Citation Format: Seung Wook Kim, Sun Jin Kim, Hyun Jin Choi, Ho-Jeong Lee, Junqin He, Qiuyu Wu, Erica J. Lawson, Isaiah J. Fidler. The role of endothelin axis signaling pathway in astrocyte mediated chemoprotection of tumor cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4951. doi:10.1158/1538-7445.AM2013-4951 [1]: /lookup/external-ref?link_type=GEN&access_num=PD145065&atom=%2Fcanres%2F73%2F8_Supplement%2F4951.atom


Cancer Research | 2013

Abstract P1-06-07: Immune-induced epithelial to mesenchymal transition in inflammatory breast cancer induces unique increases in E-cadherin, adhesion and migration through TNF-a, IL-6 and TGF-b

Evan N. Cohen; Hui Gao; Simone Anfossi; Antonio Giordano; Sanda Tin; Qiuyu Wu; B-N Lee; R Luthra; Savitri Krishnamurthy; Gabriel N. Hortobagyi; Nt Ueno; Wendy A. Woodward; Jm Reuben

BACKGROUND AND RATIONALE Inflammatory breast cancer (IBC) is the most aggressive form of breast cancer and patients frequently present with metastases at the time of their diagnosis. Although a robust IBC-specific molecular signature remains elusive, the disease is frequently characterized by persistent expression of the adhesion molecule, E-cadherin. This is highly counterintuitive as epithelial to mesenchymal transition (EMT), frequently associated with metastasis, results in decreased E-cadherin expression and highly aggressive cancers frequently express low levels of E-cadherin. We hypothesized that persistent inflammation, mediated by immune activation, increases the plasticity of IBC cells, inducing EMT and allowing the re-acquisition of epithelia characteristics once removed from the inflammatory foci. In support of this hypothesis, previous in vitro work showed that soluble factors from activated immune cells induce EMT-related transcripts in both IBC and non-IBC cell lines. However, uniquely in 3 of 4 IBC cell lines but none of the non-IBC cell lines, this program included an increase of E-cadherin expression. RESULTS We used real-time cell analysis (RTCA) from Acea Biosciences (San Diego, CA) to probe the effect of immune conditioned media, produced by stimulating healthy donor peripheral blood mononuclear cells through the T-cell receptor or through toll-like receptor-4, on SUM149 inflammatory breast cancer cells. Consistent with the increased expression of E-cadherin, we observed rapid and strong increases in cellular adhesion as measured by the RCTA cell-index following culture with immune inflammatory factors. However, using the CIM chip, the same cells also showed strong increases in invasion and migration. To determine the inflammatory factors involved in this process, we screened the immune conditioned media using a Luminex array (Millipore, Billerica, MA). TGF-b, TNF-α, and IL-6, previously shown to induce EMT, were all found at elevated levels. In 5 culture supernatants of healthy donor PBMC activated for 48h with anti-CD3 antibody, TGF-β had a modest 1.6-fold increase; TNF-α had an average 101-fold increase; while IL-6 had an average 347-fold increase. When added to cultures of SUM149 cells, these factors recapitulated the EMT gene expression signature in SUM149 including the increase in E-cadherin expression. Furthermore, the addition of neutralizing antibodies against TNF-α, TGF-β, and IL-6 to immune conditioned media prior to exposure to SUM149 cells resulted in less EMT. CONCLUSIONS Inflammatory factors may induce both the migratory ability and the characteristic persistent E-cadherin expression of IBC cells. This is mediated in part by TNF-α, TGF-β, and IL-6. However, the molecular basis for this unique IBC response requires further study hindering the development of optimal therapies. Ongoing studies at MD Anderson are exploring both the tumor and stromal components of inflammatory breast cancer. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P1-06-07.

Collaboration


Dive into the Qiuyu Wu's collaboration.

Top Co-Authors

Avatar

Isaiah J. Fidler

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sun Jin Kim

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Junqin He

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Seung Wook Kim

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ho Jeong Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erica J. Lawson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Fahao Zhang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Robert R. Langley

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge