Network


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

Hotspot


Dive into the research topics where Ashley Orillion is active.

Publication


Featured researches published by Ashley Orillion.


Cancer immunology research | 2015

Tasquinimod modulates suppressive myeloid cells and enhances cancer immunotherapies in murine models

Li Shen; Anette Sundstedt; Michael J. Ciesielski; Kiersten Marie Miles; Mona Celander; Remi Adelaiye; Ashley Orillion; Eric Ciamporcero; Swathi Ramakrishnan; Leigh Ellis; Robert A. Fenstermaker; Scott I. Abrams; Helena Eriksson; Tomas Leanderson; Anders Olsson; Roberto Pili

Shen, Sundstedt, and colleagues show in murine models that tasquinimod enhanced the antitumor effects of SurVaxM tumor vaccine for prostate cancer and of 5T4Fab-SEA tumor-targeted superantigen for melanoma by inhibiting the accumulation and function of tumor-infiltrating suppressive myeloid cells. A major barrier for cancer immunotherapy is the presence of suppressive cell populations in patients with cancer, such as myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM), which contribute to the immunosuppressive microenvironment that promotes tumor growth and metastasis. Tasquinimod is a novel antitumor agent that is currently at an advanced stage of clinical development for treatment of castration-resistant prostate cancer. A target of tasquinimod is the inflammatory protein S100A9, which has been demonstrated to affect the accumulation and function of tumor-suppressive myeloid cells. Here, we report that tasquinimod provided a significant enhancement to the antitumor effects of two different immunotherapeutics in mouse models of cancer: a tumor vaccine (SurVaxM) for prostate cancer and a tumor-targeted superantigen (TTS) for melanoma. In the combination strategies, tasquinimod inhibited distinct MDSC populations and TAMs of the M2-polarized phenotype (CD206+). CD11b+ myeloid cells isolated from tumors of treated mice expressed lower levels of arginase-1 and higher levels of inducible nitric oxide synthase (iNOS), and were less immunosuppressive ex vivo, which translated into a significantly reduced tumor-promoting capacity in vivo when these cells were coinjected with tumor cells. Tumor-specific CD8+ T cells were increased markedly in the circulation and in tumors. Furthermore, T-cell effector functions, including cell-mediated cytotoxicity and IFNγ production, were potentiated. Taken together, these data suggest that pharmacologic targeting of suppressive myeloid cells by tasquinimod induces therapeutic benefit and provide the rationale for clinical testing of tasquinimod in combination with cancer immunotherapies. Cancer Immunol Res; 3(2); 136–48. ©2014 AACR.


Molecular Cancer Therapeutics | 2015

Sunitinib Dose Escalation Overcomes Transient Resistance in Clear Cell Renal Cell Carcinoma and Is Associated with Epigenetic Modifications

Remi Adelaiye; Eric Ciamporcero; Kiersten Marie Miles; Paula Sotomayor; Jonathan Bard; Maria Tsompana; Dylan Conroy; Li Shen; Swathi Ramakrishnan; Sheng-Yu Ku; Ashley Orillion; Joshua Prey; Gerald J. Fetterly; Michael J. Buck; Sreenivasulu Chintala; Georg A. Bjarnason; Roberto Pili

Sunitinib is considered a first-line therapeutic option for patients with advanced clear cell renal cell carcinoma (ccRCC). Despite sunitinibs clinical efficacy, patients eventually develop drug resistance and disease progression. Herein, we tested the hypothesis whether initial sunitinib resistance may be transient and could be overcome by dose increase. In selected patients initially treated with 50 mg sunitinib and presenting with minimal toxicities, sunitinib dose was escalated to 62.5 mg and/or 75 mg at the time of tumor progression. Mice bearing two different patient-derived ccRCC xenografts (PDX) were treated 5 days per week with a dose-escalation schema (40–60–80 mg/kg sunitinib). Tumor tissues were collected before dose increments for immunohistochemistry analyses and drug levels. Selected intrapatient sunitinib dose escalation was safe and several patients had added progression-free survival. In parallel, our preclinical results showed that PDXs, although initially responsive to sunitinib at 40 mg/kg, eventually developed resistance. When the dose was incrementally increased, again we observed tumor response to sunitinib. A resistant phenotype was associated with transient increase of tumor vasculature despite intratumor sunitinib accumulation at higher dose. In addition, we observed associated changes in the expression of the methyltransferase EZH2 and histone marks at the time of resistance. Furthermore, specific EZH2 inhibition resulted in increased in vitro antitumor effect of sunitinib. Overall, our results suggest that initial sunitinib-induced resistance may be overcome, in part, by increasing the dose, and highlight the potential role of epigenetic changes associated with sunitinib resistance that can represent new targets for therapeutic intervention. Mol Cancer Ther; 14(2); 513–22. ©2014 AACR.


Journal of Biological Chemistry | 2014

PTPN14 forms a complex with Kibra and LATS1 proteins and negatively regulates the YAP oncogenic function.

Kayla E. Wilson; Ying-Wei Li; Nuo Yang; He Shen; Ashley Orillion; Jianmin Zhang

Background: Transcriptional co-activators YAP/TAZ are pivotal effectors of the Hippo pathway and their dysfunction promote epithelial-to-mesenchymal transition (EMT) and malignant transformation. Results: PTPN14 interacts with Kibra and activates LATS1 (upstream negative regulator of YAP). Conclusion: PTPN14 and Kibra activate LATS1 and negatively regulate the YAP oncogenic function. Significance: Study of the YAP regulatory mechanism is crucial for understanding its role in the physiological and pathological processes. The Hippo signaling pathway regulates cellular proliferation and survival, thus exerting profound effects on normal cell fate and tumorigenesis. Pivotal effectors of this pathway are YAP/TAZ, transcriptional co-activators whose dysfunction contributes to epithelial-to-mesenchymal transition and malignant transformation. Therefore, it is of great importance to decipher the mechanisms underlying the regulations of YAP/TAZ at various levels. Here we report that non-receptor tyrosine phosphatase 14 (PTPN14) interacts with the Kibra protein. The interaction between PTPN14 and Kibra is through the PPXY domain of PTPN14 and WW domain of Kibra. PTPN14 and Kibra can induce the LATS1 activation independently and cooperatively. Interestingly, activation of LATS1 by PTPN14 is dependent on the C terminus of PTPN14 and independent of the upstream mammalian STE20-like kinase (MST) proteins. Furthermore, we demonstrate that PTPN14 increases the LAST1 protein stability. Last, overexpression of Kibra rescues the increased cell migration and aberrant three-dimensional morphogenesis induced by knockdown of PTPN14, and this rescue is mediated through the activation of the upstream LATS1 kinase and subsequent cytoplasmic sequestration of YAP. In summary, our results indicate a potential regulatory role of PTPN14 in the Hippo pathway and demonstrate another layer of regulation in the YAP oncogenic function.


Clinical Cancer Research | 2017

Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma

Ashley Orillion; Ayumi Hashimoto; Nur P. Damayanti; Li Shen; Remi Adelaiye-Ogala; Sreevani Arisa; Sreenivasulu Chintala; Peter Ordentlich; Chinghai Kao; Bennett D. Elzey; Dmitry I. Gabrilovich; Roberto Pili

Purpose: Recent advances in immunotherapy highlight the antitumor effects of immune checkpoint inhibition despite a relatively limited subset of patients receiving clinical benefit. The selective class I histone deacetylase inhibitor entinostat has been reported to have immunomodulatory activity including targeting of immune suppressor cells in the tumor microenvironment. Thus, we decided to assess whether entinostat could enhance anti–PD-1 treatment and investigate those alterations in the immunosuppressive tumor microenvironment that contribute to the combined antitumor activity. Experimental Design: We utilized syngeneic mouse models of lung (LLC) and renal cell (RENCA) carcinoma and assessed immune correlates, tumor growth, and survival following treatment with entinostat (5 or 10 mg/kg, p.o.) and a PD-1 inhibitor (10 and 20 mg/kg, s.c.). Results: Entinostat enhanced the antitumor effect of PD-1 inhibition in two syngeneic mouse tumor models by reducing tumor growth and increasing survival. Entinostat inhibited the immunosuppressive function of both polymorphonuclear (PMN)- and monocytic-myeloid derived suppressor cell (M-MDSC) populations. Analysis of MDSC response to entinostat revealed significantly reduced arginase-1, iNOS, and COX-2 levels, suggesting potential mechanisms for the altered function. We also observed significant alterations in cytokine/chemokine release in vivo with a shift toward a tumor-suppressive microenvironment. Conclusions: Our results demonstrate that entinostat enhances the antitumor effect of PD-1 targeting through functional inhibition of MDSCs and a transition away from an immune-suppressive tumor microenvironment. These data provide a mechanistic rationale for the clinical testing and potential markers of response of this novel combination in solid tumor patients. Clin Cancer Res; 23(17); 5187–201. ©2017 AACR.


Epigenomics | 2016

Histone deacetylase inhibitors as immunomodulators in cancer therapeutics

Li Shen; Ashley Orillion; Roberto Pili

HDAC inhibitors (HDACIs) are anticancer agents being developed in preclinical and clinical settings due to their capacity to modulate gene expression involved in cell growth, differentiation and apoptosis, through modification of both chromatin histone and nonhistone proteins. Most HDACIs in clinical development have cytotoxic or cytostatic properties and their direct inhibitory effects on tumor cells are well documented. Numerous studies have revealed that HDACIs have potent immunomodulatory activity in tumor-bearing animals and cancer patients, providing guidance to apply these agents in cancer immunotherapies. Here, we summarize recent reports addressing the effects of HDACIs on tumor cell immunogenicity, and on different components of the host immune system. In addition, we discuss the complexity of the immunomodulatory activity of these agents, which depends on the class specificity of the HDACIs, different experimental settings and the target immune cell populations.


BMC Cancer | 2016

HDAC 1 and 6 modulate cell invasion and migration in clear cell renal cell carcinoma

Swathi Ramakrishnan; Sheng Yu Ku; Eric Ciamporcero; Kiersten Marie Miles; Kris Attwood; Sreenivasulu Chintala; Li Shen; Leigh Ellis; Paula Sotomayor; Wendy M. Swetzig; Ray Huang; Dylan Conroy; Ashley Orillion; Gokul Das; Roberto Pili

BackgroundClass I histone deacetylases (HDACs) have been reported to be overexpressed in clear cell renal cell carcinoma (ccRCC), whereas the expression of class II HDACs is unknown.MethodsFour isogenic cell lines C2/C2VHL and 786-O/786-OVHL with differential VHL expression are used in our studies. Cobalt chloride is used to mimic hypoxia in vitro. HIF-2α knockdowns in C2 and 786-O cells is used to evaluate the effect on HDAC 1 expression and activity. Invasion and migration assays are used to investigate the role of HDAC 1 and HDAC 6 expression in ccRCC cells. Comparisons are made between experimental groups using the paired T-test, the two-sample Student’s T-test or one-way ANOVA, as appropriate. ccRCC and the TCGA dataset are used to observe the clinical correlation between HDAC 1 and HDAC 6 overexpression and overall and progression free survival.ResultsOur analysis of tumor and matched non-tumor tissues from radical nephrectomies showed overexpression of class I and II HDACs (HDAC6 only in a subset of patients). In vitro, both HDAC1 and HDAC6 over-expression increased cell invasion and motility, respectively, in ccRCC cells. HDAC1 regulated invasiveness by increasing matrix metalloproteinase (MMP) expression. Furthermore, hypoxia stimulation in VHL-reconstituted cell lines increased HIF isoforms and HDAC1 expression. Presence of hypoxia response elements in the HDAC1 promoter along with chromatin immunoprecipitation data suggests that HIF-2α is a transcriptional regulator of HDAC1 gene. Conversely, HDAC6 and estrogen receptor alpha (ERα) were co-localized in cytoplasm of ccRCC cells and HDAC6 enhanced cell motility by decreasing acetylated α-tubulin expression, and this biological effect was attenuated by either biochemical or pharmacological inhibition. Finally, analysis of human ccRCC specimens revealed positive correlation between HIF isoforms and HDAC. HDAC1 mRNA upregulation was associated with worse overall survival in the TCGA dataset.ConclusionsTaking together, these results suggest that HDAC1 and HDAC6 may play a role in ccRCC biology and could represent rational therapeutic targets.


Clinical Cancer Research | 2017

Immunomodulation by entinostat in renal cell carcinoma patients receiving high-dose interleukin 2: A multicenter, single-arm, phase I/II trial (NCI-CTEP#7870)

Roberto Pili; David I. Quinn; Hans J. Hammers; J. Paul Monk; Saby George; Tanya B. Dorff; Thomas Olencki; Li Shen; Ashley Orillion; Dominick Lamonica; Roberto Andres Salas Fragomeni; Zsolt Szabo; Alan D. Hutson; Adrienne Groman; Susan M. Perkins; Richard Piekarz; Michael A. Carducci

Purpose: On the basis of preclinical data suggesting that the class I selective HDAC inhibitor entinostat exerts a synergistic antitumor effect in combination with high-dose IL2 in a renal cell carcinoma model by downregulating Foxp3 expression and function of regulatory T cells (Treg), we conducted a phase I/II clinical study with entinostat and high-dose IL2 in patients with metastatic clear cell renal cell carcinoma (ccRCC). Experimental Design: Clear cell histology, no prior treatments, and being sufficiently fit to receive high-dose IL2 were the main eligibility criteria. The phase I portion consisted of two dose levels of entinostat (3 and 5 mg, orally every 14 days) and a fixed standard dose of IL2 (600,000 U/kg i.v.). Each cycle was 85 days. The primary endpoint was objective response rate and toxicity. Secondary endpoints included progression-free survival and overall survival. Results: Forty-seven patients were enrolled. At a median follow-up of 21.9 months, the objective response rate was 37% [95% confidence interval (CI), 22%–53%], the median progression-free survival was 13.8 months (95% CI, 6.0–18.8), and the median overall survival was 65.3 months (95% CI, 52.6.-65.3). The most common grade 3/4 toxicities were hypophosphatemia (16%), lymphopenia (15%), and hypocalcemia (7%), and all were transient. Decreased Tregs were observed following treatment with entinostat, and lower numbers were associated with response (P = 0.03). Conclusions: This trial suggests a promising clinical activity for entinostat in combination with high-dose IL2 in ccRCC patients and provides the first example of an epigenetic agent being rationally combined with immunotherapy. Clin Cancer Res; 23(23); 7199–208. ©2017 AACR.


Cancer Research | 2017

EZH2 Modifies Sunitinib Resistance in Renal Cell Carcinoma by Kinome Reprogramming

Remi Adelaiye-Ogala; Justin Budka; Nur P. Damayanti; Justine V. Arrington; Mary W. Ferris; Chuan-Chih Hsu; Sreenivasulu Chintala; Ashley Orillion; Kiersten Marie Miles; Li Shen; May Elbanna; Eric Ciamporcero; Sreevani Arisa; Piergiorgio Pettazzoni; Giulio Draetta; Mukund Seshadri; Bradley A. Hancock; Milan Radovich; Janaiah Kota; Michael J. Buck; Heike Keilhack; Brian P. McCarthy; Scott Persohn; Paul R. Territo; Yong Zang; Joseph Irudayaraj; Andy Tao; Peter C. Hollenhorst; Roberto Pili

Acquired and intrinsic resistance to receptor tyrosine kinase inhibitors (RTKi) represents a major hurdle in improving the management of clear cell renal cell carcinoma (ccRCC). Recent reports suggest that drug resistance is driven by tumor adaptation via epigenetic mechanisms that activate alternative survival pathways. The histone methyl transferase EZH2 is frequently altered in many cancers, including ccRCC. To evaluate its role in ccRCC resistance to RTKi, we established and characterized a spontaneously metastatic, patient-derived xenograft model that is intrinsically resistant to the RTKi sunitinib, but not to the VEGF therapeutic antibody bevacizumab. Sunitinib maintained its antiangiogenic and antimetastatic activity but lost its direct antitumor effects due to kinome reprogramming, which resulted in suppression of proapoptotic and cell-cycle-regulatory target genes. Modulating EZH2 expression or activity suppressed phosphorylation of certain RTKs, restoring the antitumor effects of sunitinib in models of acquired or intrinsically resistant ccRCC. Overall, our results highlight EZH2 as a rational target for therapeutic intervention in sunitinib-resistant ccRCC as well as a predictive marker for RTKi response in this disease. Cancer Res; 77(23); 6651-66. ©2017 AACR.


Cancer Research | 2015

Abstract 3508: Inhibition of EZH2 overcomes resistance to sunitinib in clear cell renal cell carcinoma models

Remi M. Adelaiye-Ogala; Sreenivasulu Chintala; Li Shen; Ashley Orillion; Eric Ciamporcero; May Elbanna; Kiersten Marie Miles; Bryan M. Gillard; Michael Buck; Roberto Pili

Background: Alterations in epigenetic mechanisms including histone modification and hyper-methylation at gene promoter regions have been implicated as mechanisms of drug resistance in cancer. Alternation of epigenetic regulators such histone methyltransferase, EZH2, has been reported in numerous cancer types including advanced renal cell carcinoma (RCC). Previous studies suggest that sunitinib may have a direct anti-tumor effect and that acquired sunitinib resistance may be induced in tumor cells rather than just in endothelial cells. In our study, we investigated the role of EZH2 in sunitinib resistance in clear cell renal cell carcinoma. Methods: Human RCC cell lines 786-0 were treated and exposed to increasing concentrations of sunitinib to develop a resistant cell line, 786-0R. Parental and resistant cell lines were treat with either sunitinib, GSK126 (EZH2 inhibitor) or both. In parallel, EZH2 was knocked down in 786-0 cells and exposed to increasing concentrations of sunitinib. Cell viability was quantitated by absorbance of crystal violet stained cells using a spectrometer at 570nm. In a second set of experiments, control and treated cells were collected for western analysis. Mice bearing human ccRCC patient derived xenograft (PDXs); RP-R-01, RP-R-02 and RP-R-02LM (a metastatic ccRCC model established from RP-R-02) were implanted into SCID mice either subcutaneously or orthotopic in the kidney (sub-renal). When tumors reached an average volume of 50mm3, mice were randomly grouped into 2 arms; control and sunitinib treatment (40mg/kg, 5days/week). Tumors volumes and body weight were assessed once per week. Tumor tissues and lungs were collected for immunohistochemistry analysis. All assessments and quantification were done blindly. Results: Our in vitro and in vivo data showed an increased expression of EZH2 with resistance to sunitinib. Furthermore, inhibition of EZH2 in our in vitro studies correlated with a significant decrease in the anti-tumor effect of sunitinib in both parental and resistant cell lines. Conclusion: Overall our data suggest the potential role of epigenetic alterations, specifically EZH2 overexpression and its association with resistance to sunitinib. We are currently assessing the effect of EZH2 inhibition with sunitinib resistance in our in vivo system using metastatic ccRCC PDX models. Citation Format: Remi M. Adelaiye-Ogala, Sreenivasulu Chintala, Li Shen, Ashley Orillion, Eric Ciamporcero, May Elbanna, Kiersten Marie Miles, Bryan Gillard, Michael Buck, Roberto Pili. Inhibition of EZH2 overcomes resistance to sunitinib in clear cell renal cell carcinoma 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 3508. doi:10.1158/1538-7445.AM2015-3508


Cancer Research | 2014

Abstract 4061: Evidence for hdac6 and er-α association in a subset of clear cell renal cell carcinoma

Swathi Ramakrishnan; Sheng-Yu Ku; Wendy M. Swetzig; Dylan Conroy; Li Shen; Sreenivasulu Chintala; Paula Sotomayor; Kiersten Marie Miles; Remi Adelaiye; Eric Ciamporcero; Ashley Orillion; Leigh Ellis; Gokul Das; Roberto Pili

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Introduction: Histone deacetylases are overexpressed in several tumors including prostate, breast and clear cell renal cell carcinoma (ccRCC). Our group has previously reported that class II HDACs, HDAC4 and HDAC6 regulate HIF-α stability in ccRCC cell lines. Interestingly, HDAC6 overexpression in ER-α positive breast cancer has been shown to correlate with overall and cancer specific survival in response to tamoxifen treatment. In addition, HDAC6 increases cell motility by deacetylating α-tubulin, and HDAC6 interaction with ER-α on the cell membrane increases its deacetylating activity. The objective of our study was to assess whether HDAC6 associates with ER-α in a subset of ccRCC and whether this association can be targeted therapeutically. Methods: Radical nephrectomy tumor samples (n=14) with matched adjacent non tumor tissues were collected and analyzed for HDAC6 expression by Western blot analysis. HDAC6 expression was also assessed in C2, C2VHL and 786-O ccRCC cell lines by Western blot and immunofluorescence analysis. HDAC6 and ER-α colocalization was examined by immunoprecipitation and immunofluorescence. HDAC 6 was overexpressed in cell lines and investigated for cell motility by scratch assay. Cell lines were also treated with hydroxy tamoxifen in short term (4 hours) and long term (24, 48 and 72 hours) culture experiments for evaluation of effects on acetylated α-tubulin and cell proliferation, respectively. Results: Analysis of matched patient tumor samples revealed that a subset of ccRCC had higher HDAC6 expression as compared to the adjacent non tumor tissue. HDAC6 and ER-α examined in ccRCC tumors (n=44) by immunofluorescence showed overexpression in 10% of tumor samples. Immunoprecipitation of HDAC 6 in ccRCC cell lines showed that ER-α is present in the same complex as HDAC 6 as confirmed also by fluorescence microscopy. HDAC6 overexpression in ccRCC cell lines increased cell motility, although overexpression did not affect cell proliferation. Cells treated for short term experiments with hydroxy tamoxifen showed an increase in acetylated α-tubulin when examined by immunofluorescence. Upon long term hydroxy tamoxifen treatment in regular DMEM medium with serum, ccRCC cell proliferation was affected at high concentrations (10-20µM), similar to MCF 7 cells treated under similar conditions. Conclusions: HDAC 6 and ER-α are overexpressed in a subset of ccRCC. HDAC6 overexpression affects cell motility but not proliferation. HDAC6 and ER-α are present in the same immunocomplex and this association may be targeted with therapeutic interventions. Ongoing studies are testing concomitant, either genetic or pharmacological, inhibition of both HDAC6 and ER-α in ccRCC and will provide the rationale for novel targeted therapies for a selected group of patients with ccRCC. Citation Format: Swathi Ramakrishnan, Sheng-Yu Ku, Wendy Swetzig, Dylan Conroy, Li Shen, Sreenivasulu Chintala, Paula Sotomayor, Kiersten M. Miles, Remi Adelaiye, Eric Ciamporcero, Ashley Orillion, Leigh Ellis, Gokul Das, Roberto Pili. Evidence for hdac6 and er-α association in a subset of clear cell renal cell carcinoma. [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 4061. doi:10.1158/1538-7445.AM2014-4061

Collaboration


Dive into the Ashley Orillion's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li Shen

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

May Elbanna

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Kiersten Marie Miles

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Remi Adelaiye

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Swathi Ramakrishnan

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge