Network


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

Hotspot


Dive into the research topics where Courtney Smith is active.

Publication


Featured researches published by Courtney Smith.


Cancer Immunology, Immunotherapy | 2014

Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer.

George C. Prendergast; Courtney Smith; Sunil Thomas; Laura Mandik-Nayak; Lisa Laury-Kleintop; Richard Metz; Alexander J. Muller

Abstract Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-β signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.


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.


International Immunology | 2014

IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation

Richard Metz; Courtney Smith; James B. DuHadaway; Phillip Chandler; Babak Baban; Lauren M.F. Merlo; Elizabeth Pigott; Martin P. Keough; Sonja Rust; Andrew L. Mellor; Laura Mandik-Nayak; Alexander J. Muller; George C. Prendergast

IDO2 is implicated in tryptophan catabolism and immunity but its physiological functions are not well established. Here we report the characterization of mice genetically deficient in IDO2, which develop normally but exhibit defects in IDO-mediated T-cell regulation and inflammatory responses. Construction of this strain was prompted in part by our discovery that IDO2 function is attenuated in macrophages from Ido1 (-/-) mice due to altered message splicing, generating a functional mosaic with implications for interpreting findings in Ido1 (-/-) mice. No apparent defects were observed in Ido2 (-/-) mice in embryonic development or hematopoietic differentiation, with wild-type profiles documented for kynurenine in blood serum and for immune cells in spleen, lymph nodes, peritoneum, thymus and bone marrow of naive mice. In contrast, upon immune stimulation we determined that IDO1-dependent T regulatory cell generation was defective in Ido2 (-/-) mice, supporting Ido1-Ido2 genetic interaction and establishing a functional role for Ido2 in immune modulation. Pathophysiologically, both Ido1 (-/-) and Ido2 (-/-) mice displayed reduced skin contact hypersensitivity responses, but mechanistic distinctions were apparent, with only Ido2 deficiency associated with a suppression of immune regulatory cytokines that included GM-CSF, G-CSF, IFN-γ, TNF-α, IL-6 and MCP-1/CCL2. Different contributions to inflammation were likewise indicated by the finding that Ido2 (-/-) mice did not phenocopy Ido1 (-/-) mice in the reduced susceptibility of the latter to inflammatory skin cancer. Taken together, our results offer an initial glimpse into immune modulation by IDO2, revealing its genetic interaction with IDO1 and distinguishing its non-redundant contributions to inflammation.


Cancer Biology & Therapy | 2011

Cardiac and gastrointestinal liabilities caused by deficiency in the immune modulatory enzyme indoleamine 2,3-dioxygenase

Mee Young Chang; Courtney Smith; James B. DuHadaway; Pyle; Janette Boulden; Alejandro Peralta Soler; Alexander J. Muller; Lisa Laury-Kleintop; George C. Prendergast

Indoleamine 2,3-dioxygenase (IDO) modifies adaptive immunity, in part by determining the character of inflammatory responses in the tissue microenvironment. Small molecule inhibitors of IDO are being developed to treat cancer, chronic infections and other diseases, so the systemic effects of IDO disruption on inflammatory phenomena may influence the design and conduct of early phase clinical investigations of this new class of therapeutic agents. Here, we report cardiac and gastrointestinal phenotypes observed in IDO deficient mice that warrant consideration in planned assessments of the safety risks involved in clinical development of IDO inhibitors. Calcification of the cardiac endometrium proximal to the right ventricle was a sexually dimorphic strain-specific phenotype with ~30% penetrance in BALB/c mice lacking IDO. Administration of complete Freund’s adjuvant containing Toll-like receptor ligands known to induce IDO caused acute pancreatitis in IDO deficient mice, with implications for the design of planned combination studies of IDO inhibitors with cancer vaccines. In an established model of hyperlipidemia, IDO deficiency caused a dramatic elevation in levels of serum triglycerides. In the large intestine, IDO loss only slightly increased sensitivity to induction of acute colitis, but it markedly elevated tumor incidence, multiplicity and staging during inflammatory colon carcinogenesis. Together, our findings suggest potential cardiac and gastrointestinal risks of IDO inhibitors that should be monitored in patients as this new class of drugs enter early clinical development.


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.


Archive | 2014

IDO in Inflammatory Programming and Immune Suppression in Cancer

George C. Prendergast; Courtney Smith; Sunil Thomas; Laura Mandik-Nayak; Lisa Laury-Kleintop; Richard Metz; Alexander J. Muller

Tryptophan catabolism by indoleamine 2,3-dioxygenase (IDO) contributes to immune tolerance and inflammatory programming in a variety of tissue microenvironments. In cancer, IDO is overexpressed in both tumor cells and stromal cells where it promotes malignant development and progression by sustaining supportive inflammatory processes and engendering tolerance to tumor antigens. Genetic and pharmacological studies in mice indicate that IDO activity is crucial for cancer development and progression, particularly in settings where inflammatory drivers are essential. IDO is critical for myeloid suppressor functions that contribute to angiogenesis and metastasis. Mechanistic investigations have defined the aryl hydrocarbon receptor (AhR), the master metabolic regulator mammalian target of rapamycin complex 1 (mTORC1), and the stress kinase general control non-repressed 2 (GCN2) as key effector signaling targets of IDO, which also displays a non-catalytic function in transforming growth factor beta (TGF-β) signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with radiotherapy, immunotherapy, or chemotherapy to trigger regression of aggressive tumors otherwise largely resistant to treatment. IDO inhibitors that block catalytic activity or selectively reverse IDO-mediated suppression of mTORC1 are being evaluated now in clinical trials. Interestingly, the dramatic antitumor activity of certain targeted therapeutics such as imatinib can be traced, in part, to IDO downregulation. After presenting a historical background on its discovery and early study, this chapter focuses on work that defines IDO as an important mediator of pathogenic inflammation in cancer and summarizes the development of IDO inhibitors as potential anticancer modalities.


Atlas of genetics and cytogenetics in oncology and haematology | 2013

Inflammatory programming and immune modulation in cancer by IDO

Courtney Smith; George C. Prendergast

Immune dysregulation is one of the hallmarks of tumor growth and progression, a key event that allows for tumor evasion of the host immune system. More recent cancer modalities are embracing combinations incorporat ing immunotherapy with more traditional chemotherapy and radiotherapy. Traditional approaches are difficul t to tolerate for the patient and become less effective as tumors evo lve to survive these treatments. Immunotherapy has the benefit of reduced toxicity as it utilizes the patients own i mmune system to identify and eliminate tumor cells. One mechanism manipulated by tumors is upregulation of the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO). In this review, we focus on the mechanism by which tumors use IDO to evade detection by T cell immunity, as we ll as on novel small molecules that inhibit it as a cancer t herapeutic strategy.


Archive | 2011

Indoleamine 2,3-Dioxygenase Amino Acid Metabolism and Tumour-Associated Macrophages: Regulation in Cancer-Associated Inflammation and Immune Escape

George C. Prendergast; Richard Metz; Mee Young Chang; Courtney Smith; Alexander J. Muller; Suzanne Ostrand-Rosenberg

Indoleamine 2,3-dioxygenases (IDO) enzymes that catabolize tryptophan are expressed in tumor, stromal, and immune cells in the tumor microenvironment, including in monocytic cells such as macrophages (TAMs). Recent mouse genetic studies define a crucial role for IDO in supporting inflammatory carcinogenesis. Key gaps remain in elucidating the immunoregulatory pathways that modify progression versus dormancy in early stage tumors, an important clinical concern. Within the tumor microenvironment, chronic inflammation and immune escape are critical contributors to progression. However, there is still limited knowledge about the molecular mechanisms which underlie cancer-associated inflammation and immune escape in cancer. Both the IDO1 and IDO2 enzymes that catabolize tryptophan have been implicated in immune escape (pathogenic immune tolerance). Contributions from different parts of the tumor microenvironment have yet to be fully defined, but an important implication of these recent studies is that pathways of immune escape and pathogenic inflammation in cancer may be genetically synonymous. IDO enzymes are generally regarded as being immunosuppressive, however, emerging evidence suggests that this view is too narrow and that IDO enzymes may function more broadly to modify or ‘flavor’ the nature of an inflammatory microenvironment, rendering it supportive to tumor progression. In this review, we summarize work on IDO in immune escape and pathogenic inflammation in cancer, discussing potential roles for IDO pathways and other amino acid catabolism pathways in TAMs and other immune cells that may contribute to shaping tissue microenvironments that nurture the development and progression of malignancy.


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


Cancer Research | 2014

Abstract 3665: IDO1 is an integrative determinant of tumor-promoting, pathogenic inflammation

Alexander J. Muller; Courtney Smith; Mee Young Chang; James B. DuHadaway; Arpita Mondal; Hollie Flick; Katherine H. Parker; Suzanne Ostrand-Rosenberg; George C. Prendergast

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The tryptophan-catabolizing enzyme IDO1 (indoleamine 2,3-dioxygenase 1) has been implicated as a mediator of immune tolerance in the reproductively essential process of shielding the ‘foreign’ fetus from maternal immunity. By extrapolation, we and others hypothesized that tumors might elevate IDO1 under selective pressure by the immune system. Our finding that loss of the Bin1 tumor suppressor gene potentiates the superinduction of IDO1 provided the first discreet example of a molecular pathway through which this immune escape process can occur. However, the involvement of IDO1 in tumorigenesis has turned out to be far more complex. Tumors that inherently lack IDO1 expression have been demonstrated to induce IDO1 expression in antigen presenting cells of the host, providing an alternative mechanism for immune escape. We have also found from studies in the classical DMBA/TPA skin carcinogenesis model that IDO1 can be induced by the inflammatory tumor-promoting process itself independent of the presence of an initiated tumor. Thus, IDO1 can be a factor in tumor promotion throughout the entire immunoediting process. With the rapid pace of development of IDO inhibitors, which are currently being evaluated in clinical trials, we are interested in determining whether mouse tumor models might provide additional insight into the optimal therapeutic application of these agents based on the underlying biology. In current studies, we have found that IDO1-nullizygous mice are resistant to both KRAS-induced lung adenocarcinomas and pulmonary breast carcinoma metastases. Micro-computed tomographic imaging confirmed that lung tumor burden was correspondingly lower in IDO1-nullizygous mice. Surprisingly, this analysis also revealed a significantly reduced pulmonary blood vessel density in IDO1-nullizygous mice. Elevation of the inflammatory cytokine IL6 (interleukin 6) was greatly attenuated in conjunction with the loss of IDO1, consistent with in vitro evidence that IDO1 potentiates IL6 production. MDSCs (myeloid derived suppressor cells) from IDO1-nullizygous animals exhibited reduced T cell suppressive activity that could be rescued by IL6. IL6 could likewise reverse the pulmonary metastasis resistance exhibited by IDO1-nullizygous mice. Together, our findings provide support for the emerging concept of IDO1 as a prototypical, integrative immune modifier that bridges inflammation, vascularization and immune escape to foster the establishment of a pathogenic, tumor-promoting environment. Citation Format: Alexander J. Muller, Courtney Smith, Mee Young Chang, James DuHadaway, Arpita Mondal, Hollie Flick, Katherine Parker, Daniel Beury, Suzanne Ostrand-Rosenberg, George C. Prendergast. IDO1 is an integrative determinant of tumor-promoting, pathogenic inflammation. [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 3665. doi:10.1158/1538-7445.AM2014-3665

Collaboration


Dive into the Courtney Smith's collaboration.

Top Co-Authors

Avatar

Alexander J. Muller

Lankenau Institute for Medical Research

View shared research outputs
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

Richard Metz

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
Top Co-Authors

Avatar

Lisa Laury-Kleintop

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

Mee Young Chang

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arturo Bravo-Nuevo

Lankenau Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge