Network


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

Hotspot


Dive into the research topics where Erika Sutanto-Ward is active.

Publication


Featured researches published by Erika Sutanto-Ward.


Nature Medicine | 2005

Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy.

Alexander J. Muller; James B. DuHadaway; P. Scott Donover; Erika Sutanto-Ward; George C. Prendergast

Immune escape is a crucial feature of cancer progression about which little is known. Elevation of the immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO) in tumor cells can facilitate immune escape. Not known is how IDO becomes elevated or whether IDO inhibitors will be useful for cancer treatment. Here we show that IDO is under genetic control of Bin1, which is attenuated in many human malignancies. Mouse knockout studies indicate that Bin1 loss elevates the STAT1- and NF-κB-dependent expression of IDO, driving escape of oncogenically transformed cells from T cell–dependent antitumor immunity. In MMTV-Neu mice, an established breast cancer model, we show that small-molecule inhibitors of IDO cooperate with cytotoxic agents to elicit regression of established tumors refractory to single-agent therapy. Our findings suggest that Bin1 loss promotes immune escape in cancer by deregulating IDO and that IDO inhibitors may improve responses to cancer chemotherapy.


Cancer Discovery | 2012

IDO is a nodal pathogenic driver of lung cancer and metastasis development

Courtney Smith; Mee Young Chang; Katherine H. Parker; James B. DuHadaway; Hollie Flick; Janette Boulden; Erika Sutanto-Ward; Alejandro Peralta Soler; Lisa Laury-Kleintop; Laura Mandik-Nayak; Richard Metz; Suzanne Ostrand-Rosenberg; George C. Prendergast; Alexander J. Muller

UNLABELLED Indoleamine 2,3-dioxygenase (IDO) enzyme inhibitors have entered clinical trials for cancer treatment based on preclinical studies, indicating that they can defeat immune escape and broadly enhance other therapeutic modalities. However, clear genetic evidence of the impact of IDO on tumorigenesis in physiologic models of primary or metastatic disease is lacking. Investigating the impact of Ido1 gene disruption in mouse models of oncogenic KRAS-induced lung carcinoma and breast carcinoma-derived pulmonary metastasis, we have found that IDO deficiency resulted in reduced lung tumor burden and improved survival in both models. Micro-computed tomographic (CT) imaging further revealed that the density of the underlying pulmonary blood vessels was significantly reduced in Ido1-nullizygous mice. During lung tumor and metastasis outgrowth, interleukin (IL)-6 induction was greatly attenuated in conjunction with the loss of IDO. Biologically, this resulted in a consequential impairment of protumorigenic myeloid-derived suppressor cells (MDSC), as restoration of IL-6 recovered both MDSC suppressor function and metastasis susceptibility in Ido1-nullizygous mice. Together, our findings define IDO as a prototypical integrative modifier that bridges inflammation, vascularization, and immune escape to license primary and metastatic tumor outgrowth. SIGNIFICANCE This study provides preclinical, genetic proof-of-concept that the immunoregulatory enzyme IDO contributes to autochthonous carcinoma progression and to the creation of a metastatic niche. IDO deficiency in vivo negatively impacted both vascularization and IL-6–dependent, MDSC-driven immune escape, establishing IDO as an overarching factor directing the establishment of a protumorigenic environment.


Cancer Immunology, Immunotherapy | 2010

Non-hematopoietic expression of IDO is integrally required for inflammatory tumor promotion

Alexander J. Muller; James B. DuHadaway; Mee Young Chang; Arivudinambi Ramalingam; Erika Sutanto-Ward; Janette Boulden; Alejandro Peralta Soler; Laura Mandik-Nayak; Susan K. Gilmour; George C. Prendergast

Indoleamine 2,3-dioxygenase (IDO) is generally considered to be immunosuppressive but recent findings suggest this characterization oversimplifies its role in disease pathogenesis. Recently, we showed that IDO is essential for tumor outgrowth in the classical two-stage model of inflammatory skin carcinogenesis. Here, we report that IDO loss did not exacerbate classical inflammatory responses. Rather, IDO induction could be elicited by environmental signals and tumor promoters as an integral component of the inflammatory tissue microenvironment even in the absence of cancer. IDO loss had limited impact on tumor outgrowth in carcinogenesis models that lacked an explicit inflammatory tumor promoter. In the context of inflammatory carcinogenesis where IDO was critical to tumor development, the most important source of IDO was radiation-resistant non-hematopoietic cells, consistent with evidence that loss of the IDO regulatory tumor suppressor gene Bin1 in transformed skin cells facilitates IDO-mediated immune escape by a cell autonomous mechanism. Taken together, our results identify IDO as an integral component of ‘cancer-associated’ inflammation that tilts the immune system toward tumor support. More generally, they promote the concept that mediators of immune escape and cancer-associated inflammation may be genetically synonymous.


Cancer Research | 2007

Bin1 ablation in mammary gland delays tissue remodeling and drives cancer progression

Mee Young Chang; Janette Boulden; Erika Sutanto-Ward; James B. DuHadaway; Alejandro Peralta Soler; Alexander J. Muller; George C. Prendergast

Genes that modify oncogenesis may influence dormancy versus progression in cancer, thereby affecting clinical outcomes. The Bin1 gene encodes a nucleocytosolic adapter protein that interacts with and suppresses the cell transforming activity of Myc. Bin1 is often attenuated in breast cancer but its ability to negatively modify oncogenesis or progression in this context has not been gauged directly. In this study, we investigated the effects of mammary gland-specific deletion of Bin1 on initiation and progression of breast cancer in mice. Bin1 loss delayed the outgrowth and involution of the glandular ductal network during pregnancy but had no effect on tumor susceptibility. In contrast, in mice where tumors were initiated by the ras-activating carcinogen 7,12-dimethylbenz(a)anthracene, Bin1 loss strongly accentuated the formation of poorly differentiated tumors characterized by increased proliferation, survival, and motility. This effect was specific as Bin1 loss did not accentuate progression of tumors initiated by an overexpressed mouse mammary tumor virus-c-myc transgene, which on its own produced poorly differentiated and aggressive tumors. These findings suggest that Bin1 loss cooperates with ras activation to drive progression, establishing a role for Bin1 as a negative modifier of oncogenicity and progression in breast cancer.


Cancer Research | 2008

Bin3 Deletion Causes Cataracts and Increased Susceptibility to Lymphoma during Aging

Arivudainambi Ramalingam; James B. DuHadaway; Erika Sutanto-Ward; Yan Wang; Joseph E. Dinchuk; Minzhou Huang; Preston S. Donover; Janette Boulden; Lois M. McNally; Alejandro Peralta Soler; Alexander J. Muller; Melinda K. Duncan; George C. Prendergast

Bin3 encodes an evolutionarily conserved and ubiquitously expressed member of the BAR superfamily of curved membrane and GTPase-binding proteins, which includes the BAR, PCH/F-BAR, and I-BAR adapter proteins implicated in signal transduction and vesicular trafficking. In humans, Bin3 maps to chromosome 8p21.3, a region widely implicated in cancer suppression that is often deleted in non-Hodgkins lymphomas and various epithelial tumors. Yeast studies have suggested roles for this gene in filamentous actin (F-actin) organization and cell division but its physiologic functions in mammals have not been investigated. Here we report that homozygous inactivation of Bin3 in the mouse causes cataracts and an increased susceptibility to lymphomas during aging. The cataract phenotype was marked by multiple morphologic defects in lens fibers, including the development of vacuoles in cortical fibers and a near total loss of F-actin in lens fiber cells but not epithelial cells. Through 1 year of age, no other phenotypes were apparent; however, by 18 months of age, Bin3(-/-) mice exhibited a significantly increased incidence of lymphoma. Bin3 loss did not affect normal cell proliferation, F-actin organization, or susceptibility to oncogenic transformation. In contrast, it increased the proliferation and invasive motility of cells transformed by SV40 large T antigen plus activated ras. Our findings establish functions for Bin3 in lens development and cancer suppression during aging. Further, they define Bin3 as a candidate for an unidentified tumor suppressor that exists at the human chromosome 8p21.3 locus.


Cancer Biology & Therapy | 2004

Targeted deletion of the suppressor gene bin1/amphiphysin2 accentuates the neoplastic character of transformed mouse fibroblasts.

Alexander J. Muller; James B. DuHadaway; P. Scott Donover; Erika Sutanto-Ward; George C. Prendergast

The Bin1/Amphiphysin2 gene encodes several alternately spliced BAR adapter proteins that have been implicated in membrane-associated and nuclear processes. Bin1 expression is often attenuated during tumor progression and Bin1 splice isoforms that localize to the nucleus display tumor suppressor properties. While these properties may reflect the ability of these isoforms to interact with and suppress the cell transforming activity of c-Myc


EBioMedicine | 2016

IDO1 is an Integral Mediator of Inflammatory Neovascularization

Arpita Mondal; Courtney Smith; James B. DuHadaway; Erika Sutanto-Ward; George C. Prendergast; Arturo Bravo-Nuevo; Alexander J. Muller

The immune tolerogenic effects of IDO1 (indoleamine 2,3-dioxygenase 1) have been well documented and genetic studies in mice have clearly established the significance of IDO1 in tumor promotion. Dichotomously, the primary inducer of IDO1, the inflammatory cytokine IFNγ (interferon-γ), is a key mediator of immune-based tumor suppression. One means by which IFNγ can exert an anti-cancer effect is by decreasing tumor neovascularization. We speculated that IDO1 might contribute to cancer promotion by countering this anti-neovascular effect of IFNγ, possibly through IDO1-potentiated elevation of the pro-tumorigenic inflammatory cytokine IL6 (interleukin-6). In this study, we investigated how genetic loss of IDO1 affects neovascularization in mouse models of oxygen-induced retinopathy and lung metastasis. Neovascularization in both models was significantly reduced in mice lacking IDO1, was similarly reduced with loss of IL6, and was restored in both cases by concomitant loss of IFNγ. Likewise, the lack of IDO1 or IL6 resulted in reduced metastatic tumor burden and increased survival, which the concomitant loss of IFNγ abrogated. This insight into IDO1s involvement in pro-tumorigenic inflammatory neovascularization may have important ramifications for IDO1 inhibitor development, not only in cancer where clinical trials are currently ongoing, but in other disease indications associated with neovascularization as well.


Cancer Research | 2015

Abstract 5223: A novel pro-angiogenic role for IDO1 in inflammatory tumor promotion

Arpita Mondal; James B. DuHadaway; Erika Sutanto-Ward; Courtney Smith; George C. Prendergast; Arturo Bravo-Nuevo; Alexander J. Muller

Chronic inflammation is a major contributing factor in cancer, but, due to the complex multifactorial nature of inflammation, there remains limited understanding of specific pathogenic determinants that might be targeted therapeutically. The tryptophan-catabolizing enzyme IDO1 (indoleamine 2,3-dioxygenase) has emerged as an intriguing, pro-tumorigenic regulator of immune function in this regard. Because IDO1 can be elevated in chronic inflammatory states even prior to the initiation of cancer, it may represent one of the earliest determinants directing the immune response towards supporting rather than eliminating tumors. Genetic studies in mice have clearly established the tumor-promoting role of IDO1, but what this actually entails remains uncertain. Here we present evidence to support the novel hypothesis that a principle means by which IDO1 facilitates tumorigenesis is by mitigating immune-based angiostasis. Tumor angiogenesis is characterized by excessive and disorganized blood vessel growth much like that induced by ischemia where immune cells have been shown to be important for limiting neovascularization. Likewise, an anti-angiogenic response may be a factor in tumor immunity. In particular, IFNγ, an inflammatory cytokine, long recognized as a major inducer of IDO1, has been shown to exert angiostatic activity against developing tumors, which was implicated in these studies as the primary mechanism for both CD4 and CD8 T cell dependent tumor rejection. Our recently reported finding that the loss of IDO1 resulted in diminished pulmonary vascularization (Smith, Cancer Discovery 2012) suggested the possibility that IDO1 might be working at cross purposes to limit IFNγ-mediated angiostasis. In this same study IDO1 loss was also associated with the attenuated induction of the pro-angiogenic inflammatory cytokine IL6. To directly investigate the role of IDO1 in pathologic angiogenesis, we have utilized an oxygen-induced retinopathy (OIR) model. As predicted, neovascularization in the OIR model was significantly reduced in Ido1-/- mice. Consistent with the hypothesis that IDO1 supports neovascularization primarily by counteracting the angiostatic activity of IFNγ, neovascularization in double knockout Ifng-/- Ido1-/- mice reverted back to wild type levels. Il6-/- mice, on the other hand, exhibited reduced neovascularization which was likewise reversed by the concurrent elimination of IFNγ. In conjunction with these angiogenesis studies, we have also examined pulmonary metastasis development by 4T1 breast carcinoma isografts. Loss of either IDO1 or IL6 resulted in resistance to pulmonary metastases that, in both cases, was abrogated by the concurrent loss of IFNγ. Taken together, these findings have led us to propose a conceptually novel working hypothesis that, in the context of an inflammatory cytokine milieu, IDO1 plays a key role in supporting tumor angiogenesis. Citation Format: Arpita Mondal, James B. DuHadaway, Erika Sutanto-Ward, Courtney Smith, George C. Prendergast, Arturo Bravo-Nuevo, Alexander J. Muller. A novel pro-angiogenic role for IDO1 in inflammatory tumor promotion. [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 5223. doi:10.1158/1538-7445.AM2015-5223


Molecular Cancer Research | 2013

Abstract B070: TIMP-4 – prognostic marker and treatment target for triple-negative breast cancers

Emily Kunkel; James B. DuHadaway; Erika Sutanto-Ward; Lauren N. Birnhak; Jenny R. Ringqvist; Zonera Ali; Paul Gilman; George C. Prendergast; U. Margaretha Wallon

Background: Tissue inhibitor of metalloproteinase-4 (TIMP-4) is a secreted multi-functional protein associated with poor survival prognosis among early-stage triple-negative breast cancers (TNBC) a . Triple-negative breast cancers (TNBC) represent a highly aggressive form of this disease with few treatment options available. Current standard chemotherapy (CTX) includes taxane or adriamycin based regiments. Extracellular TIMP-4 binds to the membrane bound tetraspanin CD63 and induces the activation of PI3K/AKT/mTOR survival pathway. Here we report that TIMP-4 induced aggressive tumor growth and metastasis can be adverted by targeting TIMP-4 either directly by sequestering extracellular pools of TIMP-4 or indirectly by blocking the activation of downstream survival pathways. Methods: Prospectively collected patient samples, in accordance with the IRB approved protocol, were tested for circulating levels of TIMP-4 using a commercially available ELISA assay in samples from time of surgery and at each treatment cycle. The medical oncology staff recommended therapy without knowledge of TIMP-4 status. The role of elevated TIMP-4 in TNBC cell behavior was tested in cell culture and animal experiments using the human breast cancer line MDA-MB-468. Cells with or without TIMP-4 added to the medium were used to determine the effects on growth, clonogenic survival and response to chemotherapeutic agents such as adriamycin, Taxol, the new anti-TIMP-4 antibody b and the PI3K/AKT/mTOR inhibitor GDC-0941. The same cell-line was used to induce tumor growth in nude mice with or without TIMP-4 containing slow-release pellets implanted into the mammary fatpad (mfp). Tumor growth and response to therapy was followed over a six-week period. Results: Results from patient samples demonstrated that circulating TIMP-4 levels in breast cancer patients remain unaffected after surgical removal of the primary tumor. Adriamycin containing regiments was the only CTX to suppress the TIMP-4 levels independent of primary tumor size and nodal status. Adding TIMP-4 to cell culture medium or the mfp of mice resulted in an 1.5-fold increased tumor growth rate. Elevated TIMP-4 in mice also resulted in liver metastasis in 25% of animals (N=8). In cell cultures, the TIMP-4 induced effects were completely adverted by addition of GDC-0941. Adding the TIMP-4 antibody, to cell culture medium or i.p. injections to mice, resulted in a decelerated growth rate and no metastasis. Conclusions: On the basis of these clinical and experimental data we suggest that TIMP-4 may represent a simple prognostic and predictive marker for TNBC patients at highest risk. The presence of TIMP-4 identifies a patient population likely to recur quickly based on the continuous activation of the PI3K/AKT/mTOR pathway. Though adriamycin therapy can reduce the TIMP-4 levels, the toxicity of this agent suggests that targeted therapy of the PI3K/AKT pathway and/or a biological therapeutic approach directed against TIMP-4 may be of benefit in this subset of pts and should be further explored. a Liss, M et.al. Am. J. Pathol. 2009 b Donover, P et.al. J. Cell. Biochem. 2010 Citation Format: Emily K. Kunkel, James DuHadaway, Erika Sutanto-Ward, Lauren N. Birnhak, Jenny R. Ringqvist, Zonera A. Ali, Paul B. Gilman, George C. Prendergast, U. Margaretha Wallon. TIMP-4 – prognostic marker and treatment target for triple-negative breast cancers. [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 B070.


Clinical Cancer Research | 2018

Host IDO2 gene status influences tumor progression and radiotherapy response in KRAS-driven sporadic pancreatic cancers

Avinoam Nevler; Alexander J. Muller; Erika Sutanto-Ward; James B. DuHadaway; Kei Nagatomo; Eric Londin; Kevin O'Hayer; Joseph A. Cozzitorto; Harish Lavu; Theresa P. Yeo; Mark T. Curtis; Tatiana Villatoro; Benjamin E. Leiby; Laura Mandik-Nayak; Jordan M. Winter; Charles J. Yeo; George C. Prendergast; Jonathan R. Brody

Collaboration


Dive into the Erika Sutanto-Ward's collaboration.

Top Co-Authors

Avatar

George C. Prendergast

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

James B. DuHadaway

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Alexander J. Muller

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Alejandro Peralta Soler

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Janette Boulden

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

P. Scott Donover

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Arpita Mondal

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Arturo Bravo-Nuevo

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Courtney Smith

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Laura Mandik-Nayak

Lankenau Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge