Network


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

Hotspot


Dive into the research topics where Mildred Felder is active.

Publication


Featured researches published by Mildred Felder.


Molecular Cancer | 2014

MUC16 (CA125): tumor biomarker to cancer therapy, a work in progress

Mildred Felder; Arvinder Kapur; Jesus Gonzalez-Bosquet; Sachi Horibata; Joseph A. Heintz; Ralph M. Albrecht; Lucas Fass; Justanjyot Kaur; Kevin Hu; Hadi Shojaei; Rebecca J. Whelan; Manish S. Patankar

Over three decades have passed since the first report on the expression of CA125 by ovarian tumors. Since that time our understanding of ovarian cancer biology has changed significantly to the point that these tumors are now classified based on molecular phenotype and not purely on histological attributes. However, CA125 continues to be, with the recent exception of HE4, the only clinically reliable diagnostic marker for ovarian cancer. Many large-scale clinical trials have been conducted or are underway to determine potential use of serum CA125 levels as a screening modality or to distinguish between benign and malignant pelvic masses. CA125 is a peptide epitope of a 3–5 million Da mucin, MUC16. Here we provide an in-depth review of the literature to highlight the importance of CA125 as a prognostic and diagnostic marker for ovarian cancer. We focus on the increasing body of literature describing the biological role of MUC16 in the progression and metastasis of ovarian tumors. Finally, we consider previous and on-going efforts to develop therapeutic approaches to eradicate ovarian tumors by targeting MUC16. Even though CA125 is a crucial marker for ovarian cancer, the exact structural definition of this antigen continues to be elusive. The importance of MUC16/CA125 in the diagnosis, progression and therapy of ovarian cancer warrants the need for in-depth research on the biochemistry and biology of this mucin. A renewed focus on MUC16 is likely to culminate in novel and more efficient strategies for the detection and treatment of ovarian cancer.


Molecular Cancer | 2010

MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells

Jennifer A. A. Gubbels; Mildred Felder; Sachi Horibata; Jennifer A. Belisle; Arvinder Kapur; Helen Holden; Sarah Petrie; Martine Migneault; Claudine Rancourt; Joseph P. Connor; Manish S. Patankar

BackgroundCancer cells utilize a variety of mechanisms to evade immune detection and attack. Effective immune detection largely relies on the formation of an immune synapse which requires close contact between immune cells and their targets. Here, we show that MUC16, a heavily glycosylated 3-5 million Da mucin expressed on the surface of ovarian tumor cells, inhibits the formation of immune synapses between NK cells and ovarian tumor targets. Our results indicate that MUC16-mediated inhibition of immune synapse formation is an effective mechanism employed by ovarian tumors to evade immune recognition.ResultsExpression of low levels of MUC16 strongly correlated with an increased number of conjugates and activating immune synapses between ovarian tumor cells and primary naïve NK cells. MUC16-knockdown ovarian tumor cells were more susceptible to lysis by primary NK cells than MUC16 expressing controls. This increased lysis was not due to differences in the expression levels of the ligands for the activating receptors DNAM-1 and NKG2D. The NK cell leukemia cell line (NKL), which does not express KIRs but are positive for DNAM-1 and NKG2D, also conjugated and lysed MUC16-knockdown cells more efficiently than MUC16 expressing controls. Tumor cells that survived the NKL challenge expressed higher levels of MUC16 indicating selective lysis of MUC16low targets. The higher csMUC16 levels on the NKL resistant tumor cells correlated with more protection from lysis as compared to target cells that were never exposed to the effectors.ConclusionMUC16, a carrier of the tumor marker CA125, has previously been shown to facilitate ovarian tumor metastasis and inhibits NK cell mediated lysis of tumor targets. Our data now demonstrates that MUC16 expressing ovarian cancer cells are protected from recognition by NK cells. The immune protection provided by MUC16 may lead to selective survival of ovarian cancer cells that are more efficient in metastasizing within the peritoneal cavity and also at overcoming anti-tumor innate immune responses.


Journal of Immunotherapy | 2004

Ex vivo evaluation of anti-EpCAM immunocytokine huKS-IL2 in ovarian cancer.

Joseph P. Connor; Mildred Felder; Jacquelyn A. Hank; Josephine Harter; Jacek Gan; Stephen D. Gillies; Paul M. Sondel

Abstract: Despite encouraging responses to treatment, 70% to 80% of women with ovarian cancer will recur due to subclinical residual disease. One experimental agent that merits testing in this setting is the immunocytokine huKS-IL2. Immunocytokines are fusion proteins consisting of a humanized monoclonal antibody linked to IL-2 (or other cytokines). The humanized monoclonal antibody (mAb) huKS, which recognizes the epithelial cell adhesion molecule (EpCAM), has been used to construct the immunocytokine huKS-IL2. To determine the potential therapeutic use of huKS-IL2 in ovarian cancer, the authors evaluated the expression of EpCAM in these cancers and investigated the effects of huKS-IL2 on peritoneal white blood cells and peripheral blood mononuclear cells from women with ovarian cancer. EpCAM expression was determined by immunohistochemistry using both huKS-IL2 and the parent KS1/4 antibody. Ascites fluid was collected and the cellular fraction cultured with or without huKS-IL2 to evaluate the cellular content and potential anti-tumor effects of the peritoneal effector cells (PECs). Peritoneal cells were incubated with FITC-conjugated KS antibody to determine the relative amount of EpCAM-positive cells. Nonadherent cells were analyzed by flow cytometry for hematopoietic origin with CD45 mAb and for CD69 expression as an indication of immune cell activation. EpCAM-positive NIH:OVCAR-3 cells were radiolabeled as targets in a chromium release assay with either PECs or PBMCs as effector cells in the presence or absence of 0.25 mcg/mL huKS-IL2. Differences between treatments were determined by t test. Thirty-two of thirty-three (97%) ovarian cancers were found to express EpCAM via immunohistochemistry. Eleven cases were stained using both KS1/4 and huKS-IL2, and identical patterns of staining were seen. All ascites samples tested had EpCAM-positive cells by flow cytometry. The mean fluorescence intensity of CD69 expression on peritoneal WBCs was increased from 20.7 to 43.9 as a result of culturing with huKS-IL2, indicating effector cell activation. In chromium release assays, KS-IL2 facilitated cell lysis of NIH:OVCAR-3 by PBMCs from both healthy controls and patients with ovarian cancer. PECs from all cases tested showed significant cell lysis induced by huKS-IL2 compared with untreated control cultures. Based on these findings, huKS-IL2 warrants further investigation as a potential immunotherapy for patients with epithelial ovarian cancer, preferably in a minimal disease setting as seen after complete cytoreductive surgery, after a complete clinical response to primary therapy, or when elevated CA-125 levels predict recurrent disease prior to clinical relapse.


PLOS ONE | 2012

Terpenoids from Zingiber officinale (Ginger) Induce Apoptosis in Endometrial Cancer Cells through the Activation of p53

Yang Liu; Rebecca J. Whelan; Bikash R. Pattnaik; Kai D. Ludwig; Enkateswar Subudhi; Helen Rowland; Nick Claussen; Noah Zucker; Shitanshu Uppal; David M. Kushner; Mildred Felder; Manish S. Patankar; Arvinder Kapur

Novel strategies are necessary to improve chemotherapy response in advanced and recurrent endometrial cancer. Here, we demonstrate that terpenoids present in the Steam Distilled Extract of Ginger (SDGE) are potent inhibitors of proliferation of endometrial cancer cells. SDGE, isolated from six different batches of ginger rhizomes, consistently inhibited proliferation of the endometrial cancer cell lines Ishikawa and ECC-1 at IC50 of 1.25 µg/ml. SDGE also enhanced the anti-proliferative effect of radiation and cisplatin. Decreased proliferation of Ishikawa and ECC-1 cells was a direct result of SDGE-induced apoptosis as demonstrated by FITC-Annexin V staining and expression of cleaved caspase 3. GC/MS analysis identified a total of 22 different terpenoid compounds in SDGE, with the isomers neral and geranial constituting 30–40%. Citral, a mixture of neral and geranial inhibited the proliferation of Ishikawa and ECC-1 cells at an IC50 10 µM (2.3 µg/ml). Phenolic compounds such as gingerol and shogaol were not detected in SDGE and 6-gingerol was a weaker inhibitor of the proliferation of the endometrial cancer cells. SDGE was more effective in inducing cancer cell death than citral, suggesting that other terpenes present in SDGE were also contributing to endometrial cancer cell death. SDGE treatment resulted in a rapid and strong increase in intracellular calcium and a 20–40% decrease in the mitochondrial membrane potential. Ser-15 of p53 was phosphorylated after 15 min treatment of the cancer cells with SDGE. This increase in p53 was associated with 90% decrease in Bcl2 whereas no effect was observed on Bax. Inhibitor of p53, pifithrin-α, attenuated the anti-cancer effects of SDGE and apoptosis was also not observed in the p53neg SKOV-3 cells. Our studies demonstrate that terpenoids from SDGE mediate apoptosis by activating p53 and should be therefore be investigated as agents for the treatment of endometrial cancer.


Gynecologic Oncology | 2008

Ascites from epithelial ovarian cancer contain high levels of functional decoy receptor 3 (DcR3) and is associated with platinum resistance

Joseph P. Connor; Mildred Felder

OBJECTIVE Decoy receptor 3 (DcR3), a soluble tumor necrosis factor receptor is a known binding partner of multiple apoptotic ligands inhibiting apoptosis. The expression of DcR3 by cancers has been reported in gastrointestinal cancers yet it has not been described in ovarian cancer. Abnormalities in apoptosis pathways are seen in ovarian cancer and we theorized that the presence of DcR3 is a component of the dysregulation. METHODS Ascites samples from 44 women with advanced ovarian cancer were tested for DcR3 by ELISA. The ability of ascites to inhibit Fas-ligand mediated apoptosis was determined by chromium release assays using cell surface or soluble Fas-ligand. Clinical parameters including, response to platinum and progression free and overall survival were compared between patients with high or low levels of DcR3. RESULTS DcR3 was found in all 44 cases by ELISA. Ascites fluid significantly inhibited Fas-ligand mediate apoptosis using both surface Fas-ligand (KFL-9 cells) and soluble Fas-ligand. Blocking DcR3 with antibodies restores the cytolytic effects in both assays. HIGH DcR3 level was associated with stage IV disease and more than double the incidence of platinum resistant disease. In this modest sample size Low DcR3 cases had longer PFI and overall survival however neither difference was statistically significant. CONCLUSIONS DcR3 is expressed by epithelial ovarian cancers, concentrated in ascites and inhibits Fas-ligand mediated apoptosis. Together with a trend toward poor patient outcome these results indicate that expression of DcR3 by ovarian cancers is worthy of further investigation in a larger population to allow multivariate analysis.


Journal of Immunotherapy | 2013

Intratumoral delivery of low doses of anti-CD40 mAb combined with monophosphoryl lipid a induces local and systemic antitumor effects in immunocompetent and T cell-deficient mice.

Tyler J. Van De Voort; Mildred Felder; Richard K. Yang; Paul M. Sondel; Alexander L. Rakhmilevich

In this study, an agonistic anti-CD40 monoclonal antibody was combined with monophosphoryl lipid A (MPL), a nontoxic derivative of lipopolysaccharide and agonist of toll-like receptor-4, to assess the immunomodulatory and antitumor synergy between the 2 agents in mice. Anti-CD40 was capable of priming macrophages to subsequent ex vivo activation by MPL in immunocompetent and T-cell–depleted mice. Intraperitoneal injections of anti-CD40+MPL induced additive to synergistic suppression of poorly immunogenic B16-F10 melanoma growing subcutaneously in syngeneic mice. When anti-CD40+MPL were injected directly into the subcutaneous tumor, the combination treatment was more effective, even with a 25-fold reduction in dose. Low-dose intratumoral treatment also slowed the growth of a secondary tumor growing simultaneously at a distant, untreated site. Antitumor effects were also induced in severe combined immunodeficiency mice and in T-cell–depleted C57BL/6 mice. Taken together, our results show that the antitumor effects of anti-CD40 are enhanced by subsequent treatment with MPL, even in T-cell–deficient hosts. These preclinical data suggest that an anti-CD40+MPL combined regimen is appropriate for clinical testing in human patients, including cancer patients who may be immunosuppressed from prior chemotherapy.


Journal of Immunotherapy | 2011

Enhanced T-cell-independent Antitumor Effect of Cyclophosphamide Combined With Anti-cd40 mab and Cpg in Mice

Erik E. Johnson; Ilia N. Buhtoiarov; Mark J. Baldeshwiler; Mildred Felder; Nico van Rooijen; Paul M. Sondel; Alexander L. Rakhmilevich

We have earlier demonstrated T-cell-independent antitumor effects of a combination of anti-CD40 monoclonal antibody (mAb) and CpG oligodeoxynucleotides (CpG) which involved macrophages. As some immunotherapeutic treatments can be potentiated by chemotherapy, we tested if cyclophosphamide (CY) would enhance the antitumor effect of anti-CD40 mAb+CpG. Treatment of B16 melanoma-bearing mice with CY and anti-CD40 mAb+CpG resulted in a significant reduction in tumor growth in immunocompetent mice compared with either CY alone or anti-CD40 mAb with CpG. This enhanced antitumor effect was maintained in severe combined immunodeficiency mice, as measured by both tumor growth and overall survival. Natural killer cells were not required for this antitumor effect as it was also observed in severe combined immunodeficiency/beige mice. Moreover, although CY treatment of immunocompetent mice suppressed natural killer cell activity, it did not negatively affect the antitumor activity of their macrophages when assayed in vitro. Depletion of macrophages in vivo reduced the antitumor effect of CY and anti-CD40 mAb+CpG. These results suggest that therapeutic strategies to activate macrophages may have potential for clinical application in cancer patients receiving chemotherapy.


Scientific Reports | 2016

Modulation of oxidative stress and subsequent induction of apoptosis and endoplasmic reticulum stress allows citral to decrease cancer cell proliferation.

Arvinder Kapur; Mildred Felder; Lucas Fass; Justanjot Kaur; Austin Czarnecki; Kavya Rathi; San Zeng; Kathryn Kalady Osowski; Colin Howell; May P. Xiong; Rebecca J. Whelan; Manish S. Patankar

The monoterpenoid, citral, when delivered through PEG-b-PCL nanoparticles inhibits in vivo growth of 4T1 breast tumors. Here, we show that citral inhibits proliferation of multiple human cancer cell lines. In p53 expressing ECC-1 and OVCAR-3 but not in p53-deficient SKOV-3 cells, citral induces G1/S cell cycle arrest and apoptosis as determined by Annexin V staining and increased cleaved caspase3 and Bax and decreased Bcl-2. In SKOV-3 cells, citral induces the ER stress markers CHOP, GADD45, EDEM, ATF4, Hsp90, ATG5, and phospho-eIF2α. The molecular chaperone 4-phenylbutyric acid attenuates citral activity in SKOV-3 but not in ECC-1 and OVCAR-3 cells. In p53-expressing cells, citral increases phosphorylation of serine-15 of p53. Activation of p53 increases Bax, PUMA, and NOXA expression. Inhibition of p53 by pifithrin-α, attenuates citral-mediated apoptosis. Citral increases intracellular oxygen radicals and this leads to activation of p53. Inhibition of glutathione synthesis by L-buthionine sulfoxamine increases potency of citral. Pretreatment with N-acetylcysteine decreases phosphorylation of p53 in citral-treated ECC-1 and OVCAR-3. These results define a p53-dependent, and in the absence of p53, ER stress-dependent mode of action of citral. This study indicates that citral in PEG-b-PCL nanoparticle formulation should be considered for treatment of breast and other tumors.


BMC Cancer | 2012

DcR3 binds to ovarian cancer via heparan sulfate proteoglycans and modulates tumor cells response to platinum with corresponding alteration in the expression of BRCA1

Joseph P. Connor; Mildred Felder; Arvinder Kapur; Nonyem Onujiogu

BackgroundOvercoming platinum resistance is a major obstacle in the treatment of Epithelial Ovarian Cancer (EOC). In our previous work Decoy Receptor 3 (DcR3) was found to be related to platinum resistance. The major objective of this work was to define the cellular interaction of DcR3 with EOC and to explore its effects on platinum responsiveness.MethodsWe studied cell lines and primary cultures for the expression of and the cells ability to bind DcR3. Cells were cultured with DcR3 and then exposed to platinum. Cell viability was determined by MTT assay. Finally, the cells molecular response to DcR3 was studied using real time RT-PCR based differential expression arrays, standard RT-PCR, and Western blot.ResultsHigh DcR3 in the peritoneal cavity of women with EOC is associated with significantly shorter time to first recurrence after platinum based therapy (p = 0.02). None-malignant cells contribute DcR3 in the peritoneal cavity. The cell lines studied do not secrete DcR3; however they all bind exogenous DcR3 to their surface implying that they can be effected by DcR3 from other sources. DcR3s protein binding partners are minimally expressed or negative, however, all cells expressed the DcR3 binding Heparan Sulfate Proteoglycans (HSPGs) Syndecans-2, and CD44v3. DcR3 binding was inhibited by heparin and heparinase. After DcR3 exposure both SKOV-3 and OVCAR-3 became more resistant to platinum with 15% more cells surviving at high doses. On the contrary CaOV3 became more sensitive to platinum with 20–25% more cell death. PCR array analysis showed increase expression of BRCA1 mRNA in SKOV-3 and OVCAR-3 and decreased BRCA1 expression in CaOV-3 after exposure to DcR3. This was confirmed by gene specific real time PCR and Western blot analysis.ConclusionsNon-malignant cells contribute to the high levels of DcR3 in ovarian cancer. DcR3 binds readily to EOC cells via HSPGs and alter their responsiveness to platinum chemotherapy. The paradoxical responses seen were related to the expression pattern of HSPGs available on the cells surface to interact with. Although the mechanism behind this is not completely known alterations in DNA repair pathways including the expression of BRCA1 appear to be involved.


American Journal of Reproductive Immunology | 2012

The mucin MUC16 (CA125) binds to NK cells and monocytes from peripheral blood of women with healthy pregnancy and preeclampsia

Chanel T. Tyler; Arvinder Kapur; Mildred Felder; Jennifer A. Belisle; Christine Trautman; Jennifer A. A. Gubbels; Joseph P. Connor; Manish S. Patankar

MUC16 (CA125) released from ovarian tumors binds to NK cells and monocytes via the inhibitory receptor Siglec‐9. Here, we investigate whether MUC16 also binds to circulating immune cells during pregnancy and in women with preeclampsia.

Collaboration


Dive into the Mildred Felder's collaboration.

Top Co-Authors

Avatar

Manish S. Patankar

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Arvinder Kapur

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Joseph P. Connor

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul M. Sondel

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer A. Belisle

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kavya Rathi

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lucas Fass

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Tyler J. Van De Voort

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge