Network


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

Hotspot


Dive into the research topics where Alicia Purkey is active.

Publication


Featured researches published by Alicia Purkey.


International Journal of Cancer | 2015

Potent antitumor activity of cabozantinib, a c-MET and VEGFR2 inhibitor, in a colorectal cancer patient-derived tumor explant model.

Eun Kee Song; W. M. Tai; Wells A. Messersmith; Stacey Bagby; Alicia Purkey; Kevin Quackenbush; Todd M. Pitts; Guoliang Wang; Patrick J. Blatchford; Rachel Yahn; Jeffrey Kaplan; Aik Choon Tan; Chloe Evelyn Atreya; Gail Eckhardt; Alan P. Venook; Eunice L. Kwak; David P. Ryan; John J. Arcaroli

Antiangiogenic therapy is commonly used for the treatment of colorectal cancer (CRC). Although patients derive some clinical benefit, treatment resistance inevitably occurs. The MET signaling pathway has been proposed to be a major contributor of resistance to antiangiogenic therapy. MET is upregulated in response to vascular endothelial growth factor pathway inhibition and plays an essential role in tumorigenesis and progression of tumors. In this study, we set out to determine the efficacy of cabozantinib in a preclinical CRC patient‐derived tumor xenograft model. We demonstrate potent inhibitory effects on tumor growth in 80% of tumors treated. The greatest antitumor effects were observed in tumors that possess a mutation in the PIK3CA gene. The underlying antitumor mechanisms of cabozantinib consisted of inhibition of angiogenesis and Akt activation and significantly decreased expression of genes involved in the PI3K pathway. These findings support further evaluation of cabozantinib in patients with CRC. PIK3CA mutation as a predictive biomarker of sensitivity is intriguing and warrants further elucidation. A clinical trial of cabozantinib in refractory metastatic CRC is being activated.


PLOS ONE | 2014

Dual pharmacological targeting of the MAP kinase and PI3K/mTOR pathway in preclinical models of colorectal cancer.

Todd M. Pitts; Timothy P. Newton; Erica L. Bradshaw-Pierce; Rebecca Addison; John J. Arcaroli; Peter J. Klauck; Stacey Bagby; Stephanie L. Hyatt; Alicia Purkey; John J. Tentler; Aik Choon Tan; Wells A. Messersmith; S. Gail Eckhardt; Stephen Leong

Background The activation of the MAPK and PI3K/AKT/mTOR pathways is implicated in the majority of cancers. Activating mutations in both of these pathways has been described in colorectal cancer (CRC), thus indicating their potential as therapeutic targets. This study evaluated the combination of a PI3K/mTOR inhibitor (PF-04691502/PF-502) in combination with a MEK inhibitor (PD-0325901/PD-901) in CRC cell lines and patient-derived CRC tumor xenograft models (PDTX). Materials and Methods The anti-proliferative effects of PF-502 and PD-901 were assessed as single agents and in combination against a panel of CRC cell lines with various molecular backgrounds. Synergy was evaluated using the Bliss Additivity method. In selected cell lines, we investigated the combination effects on downstream effectors by immunoblotting. The combination was then evaluated in several fully genetically annotated CRC PDTX models. Results The in vitro experiments demonstrated a wide range of IC50 values for both agents against a cell line panel. The combination of PF-502 and PD-901 demonstrated synergistic anti-proliferative activity with Bliss values in the additive range. As expected, p-AKT and p-ERK were downregulated by PF-502 and PD-901, respectively. In PDTX models, following a 30-day exposure to PF-502, PD-901 or the combination, the combination demonstrated enhanced reduction in tumor growth as compared to either single agent regardless of KRAS or PI3K mutational status. Conclusions The combination of a PI3K/mTOR and a MEK inhibitor demonstrated enhanced anti-proliferative effects against CRC cell lines and PDTX models.


British Journal of Cancer | 2013

Tumours with elevated levels of the Notch and Wnt pathways exhibit efficacy to PF-03084014, a γ-secretase inhibitor, in a preclinical colorectal explant model.

John J. Arcaroli; Kevin Quackenbush; Alicia Purkey; Powell Rw; Todd M. Pitts; Stacey Bagby; Aik Choon Tan; Benjamin Cross; Kelly McPhillips; Eun Kee Song; Tai Wm; Robert A. Winn; Bikkavilli K; Michelle Vanscoyk; Eckhardt Sg; Wells A. Messersmith

Background:Dysregulation of the Notch pathway has been identified to play an important role in the development and progression of colorectal cancer (CRC). In this study, we used a patient-derived CRC explant model to investigate the efficacy of the clinical γ-secretase inhibitor (GSI) PF-03084014.Methods:A total of 16 CRC explants were treated with PF-03084014. Knockdown of RBPjκ gene was used to determine the specificity of PF-03084014. Evaluation of the Notch and Wnt pathways in CRC explant tumours was performed by gene array and immunoblotting.Results:We identified a subset of CRC tumours that exhibited elevations of the Notch and Wnt pathways sensitive to PF-03084014. Treatment with the GSI resulted in a significant reduction in cleaved Notch, Axin2 (Wnt-dependent gene) and active β-catenin. In addition, knockdown of the RBPjκ gene showed that PF-03084014 has specificity for the Notch pathway in an HCT116 cell line xenograft model. Finally, an increase in apoptosis was observed in CRC001- and CRC021-sensitive tumours.Conclusion:This study provides evidence that inhibition of γ-secretase may be beneficial in a subset of patients with elevated levels of the Wnt and Notch pathways.


International Journal of Cancer | 2016

A NOTCH1 gene copy number gain is a prognostic indicator of worse survival and a predictive biomarker to a Notch1 targeting antibody in colorectal cancer

John J. Arcaroli; W. M. Tai; Ryan Mcwilliams; Stacey Bagby; Patrick J. Blatchford; Marileila Varella-Garcia; Alicia Purkey; Kevin Quackenbush; Eun Kee Song; Todd M. Pitts; Dexiang Gao; Christopher Hanyoung Lieu; Martine McManus; Aik Choon Tan; Xianxian Zheng; Qin Zhang; Mark Ozeck; Peter Olson; Zhi Qin Jiang; Scott Kopetz; Antonio Jimeno; Stephen B. Keysar; Gail Eckhardt; Wells A. Messersmith

Dysregulation of the Notch1 receptor has been shown to facilitate the development and progression of colorectal cancer (CRC) and has been identified as an independent predictor of disease progression and worse survival. Although mutations in the NOTCH1 receptor have not been described in CRC, we have previously discovered a NOTCH1 gene copy number gain in a portion of CRC tumor samples. Here, we demonstrated that a NOTCH1 gene copy number gain is significantly associated with worse survival and a high percentage of gene duplication in a cohort of patients with advanced CRC. In our CRC patient‐derived tumor xenograft (PDTX) model, tumors harboring a NOTCH1 gain exhibited significant elevation of the Notch1 receptor, JAG1 ligand and cleaved Notch1 activity. In addition, a significant association was identified between a gain in NOTCH1 gene copy number and sensitivity to a Notch1‐targeting antibody. These findings suggest that patients with metastatic CRC that harbor a gain in NOTCH1 gene copy number have worse survival and that targeting this patient population with a Notch1 antibody may yield improved outcomes.


Oncotarget | 2016

The novel tankyrase inhibitor (AZ1366) enhances irinotecan activity in tumors that exhibit elevated tankyrase and irinotecan resistance

Kevin Quackenbush; Stacey Bagby; Wai Meng Tai; Wells A. Messersmith; Anna Schreiber; Justin Greene; Jihye Kim; Guoliang Wang; Alicia Purkey; Todd M. Pitts; Anna Nguyen; Dexiang Gao; Patrick J. Blatchford; Anna Capasso; Alwin Schuller; S. Gail Eckhardt; John J. Arcaroli

Background Dysregulation of the canonical Wnt signaling pathway has been implicated in colorectal cancer (CRC) development as well as incipient stages of malignant transformation. In this study, we investigated the antitumor effects of AZ1366 (a novel tankyrase inhibitor) as a single agent and in combination with irinotecan in our patient derived CRC explant xenograft models. Results Six out of 18 CRC explants displayed a significant growth reduction to AZ1366. There was one CRC explant (CRC040) that reached the threshold of sensitivity (TGII ≤ 20%) in this study. In addition, the combination of AZ1366 + irinotecan demonstrated efficacy in 4 out of 18 CRC explants. Treatment effects on the WNT pathway revealed that tankyrase inhibition was ineffective at reducing WNT dependent signaling. However, the anti-tumor effects observed in this study were likely a result of alternative tankyrase effects whereby tankyrase inhibition reduced NuMA levels. Materials and Methods Eighteen CRC explants were treated with AZ1366 single agent or in combination for 28 days and treatment responses were assessed. Pharmacokinetic (AZ1366 drug concentrations) and pharmacodynamic effects (Axin2 levels) were investigated over 48 hours. Immunohistochemistry of nuclear β-catenin levels as well as western blot was employed to examine the treatment effects on the WNT pathway as well as NuMA. Conclusions Combination AZ1366 and irinotecan achieved greater anti-tumor effects compared to monotherapy. Activity was limited to CRC explants that displayed irinotecan resistance and increased protein levels of tankyrase and NuMA.


Oncotarget | 2016

Antitumor activity of the aurora a selective kinase inhibitor, alisertib, against preclinical models of colorectal cancer

Todd M. Pitts; Erica L. Bradshaw-Pierce; Stacey Bagby; Stephanie L. Hyatt; Heather M. Selby; Anna Spreafico; John J. Tentler; Kelly McPhillips; Peter J. Klauck; Anna Capasso; Jennifer R. Diamond; Lindsey S. Davis; Aik Choon Tan; John J. Arcaroli; Alicia Purkey; Wells A. Messersmith; Jeffery A. Ecsedy; Gail Eckhardt

Background The Aurora kinases are a family of serine/threonine kinases comprised of Aurora A, B, and C which execute critical steps in mitotic and meiotic progression. Alisertib (MLN8237) is an investigational Aurora A selective inhibitor that has demonstrated activity against a wide variety of tumor types in vitro and in vivo, including CRC. Results CRC cell lines demonstrated varying sensitivity to alisertib with IC50 values ranging from 0.06 to > 5 umol/L. Following exposure to alisertib we observed a decrease in pAurora A, B and C in four CRC cell lines. We also observed an increase in p53 and p21 in a sensitive p53 wildtype cell line in contrast to the p53 mutant cell line or the resistant cell lines. The addition of alisertib to standard CRC treatments demonstrated improvement over single agent arms; however, the benefit was largely less than additive, but not antagonistic. Methods Forty-seven CRC cell lines were exposed to alisertib and IC50s were calculated. Twenty-one PDX models were treated with alisertib and the Tumor Growth Inhibition Index was assessed. Additionally, 5 KRAS wildtype and mutant PDX models were treated with alisertib as single agent or in combination with cetuximab or irinotecan, respectively. Conclusion Alisertib demonstrated anti-proliferative effects against CRC cell lines and PDX models. Our data suggest that the addition of alisertib to standard therapies in colorectal cancer if pursued clinically, will require further investigation of patient selection strategies and these combinations may facilitate future clinical studies.


Journal of Biological Chemistry | 2015

A-Kinase Anchoring Protein 79/150 recruits Protein Kinase C to phosphorylate Roundabout receptors

Bret K. Samelson; Bryan B. Gore; Jennifer L. Whiting; Patrick J. Nygren; Alicia Purkey; Marcie Colledge; Lorene K. Langeberg; Mark L. Dell'Acqua; Larry S. Zweifel; John D. Scott

Background: A-kinase anchoring proteins position signaling enzymes to control neuronal phosphorylation events. Results: Biochemical and cellular approaches confirm that the AKAP79/150 signaling complex interfaces with the cytoplasmic tail of Roundabout (Robo) receptors. Conclusion: AKAP79/150-associated protein kinase C facilitates the phosphorylation of Ser-1330 on the Robo3.1 isoform. Significance: Kinase anchoring is a mechanism to control the phosphorylation of Robo3.1 within macromolecular assemblies. Anchoring proteins direct protein kinases and phosphoprotein phosphatases toward selected substrates to control the efficacy, context, and duration of neuronal phosphorylation events. The A-kinase anchoring protein AKAP79/150 interacts with protein kinase A (PKA), protein kinase C (PKC), and protein phosphatase 2B (calcineurin) to modulate second messenger signaling events. In a mass spectrometry-based screen for additional AKAP79/150 binding partners, we have identified the Roundabout axonal guidance receptor Robo2 and its ligands Slit2 and Slit3. Biochemical and cellular approaches confirm that a linear sequence located in the cytoplasmic tail of Robo2 (residues 991–1070) interfaces directly with sites on the anchoring protein. Parallel studies show that AKAP79/150 interacts with the Robo3 receptor in a similar manner. Immunofluorescent staining detects overlapping expression patterns for murine AKAP150, Robo2, and Robo3 in a variety of brain regions, including hippocampal region CA1 and the islands of Calleja. In vitro kinase assays, peptide spot array mapping, and proximity ligation assay staining approaches establish that human AKAP79-anchored PKC selectively phosphorylates the Robo3.1 receptor subtype on serine 1330. These findings imply that anchored PKC locally modulates the phosphorylation status of Robo3.1 in brain regions governing learning and memory and reward.


Pancreas | 2016

Aldehyde Dehydrogenase 1B1 as a Modulator of Pancreatic Adenocarcinoma.

Surendra Singh; John J. Arcaroli; David J. Orlicky; Ying Chen; Wells A. Messersmith; Stacey Bagby; Alicia Purkey; Kevin Quackenbush; David C. Thompson; Vasilis Vasiliou

Objectives The aim of the current study was to examine expression and the role, if any, of aldehyde dehydrogenase (ALDH)1B1 in pancreatic adenocarcinoma. Methods A tissue microarray of 61 pancreatic cancer patients were evaluated for protein expression of ALDH1B1 by immunohistochemistry. The ALDH1B1 small interfering (RNA) was used to assess the contribution of ALDH1B1 on proliferation of pancreatic cancer cells. Results In normal human pancreas, ALDH1B1 is abundantly expressed in glandular cells, but sparsely in the ducts (ALDH1B1 immunopositivity = 16.7 ± 1.7). In pancreatic ductal carcinoma, we found high ALDH1B1 expression in ductal cancerous tissues (ALDH1B1 immunopositivity = 197.2 ± 29.4). Analysis of ALDH1B1 expression in a human pancreatic adenocarcinoma tissue microarray showed the greatest expression in tumors that were more invasive. A variation in ALDH1B1 expression was also observed in 16 human pancreatic cancer cell lines. Knockdown of ALDH1B1 caused a 35% reduction in cell growth in the high ALDH1B1-expressing cell lines. Conclusions Our data show for the first time that ALDH1B1 is expressed at very high levels in human pancreatic cancer, and it contributes to proliferation in these tumor cells. These data suggest a potential modulatory role for ALDH1B1 in pancreatic cancer.


Cancer Research | 2013

Abstract LB-302: Potent antitumor activity of XL184 (cabozantinib), a c-MET and VEGFR2 inhibitor, in colorectal cancer patient-derived tumor explant models.

Chloe Evelyn Atreya; Eun-Kee Song; Wells A. Messersmith; Alicia Purkey; Stacey Bagby; Kevin Quackenbush; E. L. Kwak; David P. Ryan; Alan P. Venook; John J. Arcaroli

Background: The c-MET signaling pathway plays an essential role in tumorigenesis and is recognized as an important mediator of angiogenesis by inducing the expression of VEGF. In colorectal cancer (CRC), c-MET overexpression is a marker of local migration and invasion and may have prognostic significance. In addition, anti-VEGF therapy has been shown to promote tumor cell metastasis by a c-MET dependent mechanism. The objective of this study was to evaluate the activity of XL184 (cabozantinib), a potent inhibitor of both c-MET and VEGFR2, in CRC patient-derived tumor explant (PDTX) models. Experimental Design: We used CRC PDTX models to investigate the efficacy of XL184 in spheroid cultures and in vivo in athymic nude mice. Spheroids with retained cell-cell contact were formed by culturing incompletely digested PDTXs in suspension. Spheroids were incubated for 3 days with 1-10 μM XL184. Spheroid area was measured by microscopy. In the mouse models, a total of 10 CRC PDTXs (3 KRAS wild-type and 7 KRAS mutant) were treated with XL184 (30 mg/kg daily, oral gavage) for 28 days. Mice were monitored daily for signs of toxicity and tumor size was evaluated twice per week by caliper measurements. Results: Spheroids from PDTXs were entirely composed of tumor cells but, unlike cell lines, spheroids recruited stroma when implanted in mice. Although 10 μM XL184 did not inhibit spheroid formation, spheroid area significantly decreased following incubation for 3 days with 10 μM or 5 μM XL184 (p= 0.0012 and p= 0.0056, respectively). In our CRC PDTX in vivo models, XL184 was well tolerated and resulted in a significant decrease in tumor growth in CRC explants from all 10 patients, independent of KRAS status. Strikingly, two of the CRC explants exhibited tumor regression after 28 days of treatment with XL184. Conclusion: XL184 showed potent antitumor activity in PDTX spheroid and mouse models. PDTX models may be particularly valuable for preclinical testing of inhibitors of stroma-modulated targets such as XL184, where activity is not reflected in cell lines. These promising data have contributed to the rationale and design of a clinical trial of XL184 in patients with advanced CRC. Our upcoming studies will focus on identifying predictive biomarkers of sensitivity or resistance to XL184. Citation Format: Chloe E. Atreya, Eun-Kee Song, Wells Messersmith, Alicia Purkey, Stacey Bagby, Kevin Quackenbush, Robin K. Kelley, Eunice Kwak, David Ryan, Alan Venook, John J. Arcaroli. Potent antitumor activity of XL184 (cabozantinib), a c-MET and VEGFR2 inhibitor, in colorectal cancer patient-derived tumor explant models. [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 LB-302. doi:10.1158/1538-7445.AM2013-LB-302


PLOS ONE | 2017

Evaluation of the efficacy of dasatinib, a Src/Abl inhibitor, in colorectal cancer cell lines and explant mouse model

Aaron James Scott; Eun Kee Song; Stacey Bagby; Alicia Purkey; Martin D. McCarter; Csaba Gajdos; Kevin Quackenbush; Benjamin Cross; Todd M. Pitts; Aik Choon Tan; S. Gail Eckhardt; Hubert Fenton; John J. Arcaroli; Wells A. Messersmith

Background Dysregulation of the Src pathway has been shown to be important at various stages of cancer. Dasatinib is a potent Src/Abl inhibitor and has demonstrated to have anti-proliferative and anti-invasive activity in many preclinical models. The objective of this study was to determine the anti-tumor activity of dasatinib using in vitro and in vivo preclinical colorectal (CRC) models. Methods CRC cell lines and patient-derived tumor explant (PDX) models were used to investigate the efficacy of dasatinib. We treated 50 CRC cell lines with dasatinib for 72 hours and proliferation was assayed by a sulforhodamine B (SRB) assay; an IC50 ≤ 0.08 μmol/L was considered sensitive. We treated 17 patient-derived CRC explants with dasatinib (50 mg/kg/day, administered once-daily) for 28 days to determine in vivo efficacy. Tumor growth inhibition (TGI) ≥ 50% was considered sensitive. Results We found that 8 out of 50 CRC cell lines reached an IC50 ≤ 0.08 μmol/L with dasatinib treatment. In addition, of 17 CRC explants grown in the xenograft mouse model, 2 showed sensitivity to dasatinib. The anti-tumor effects observed in this study were a result of G1 cell cycle arrest as the dasatinib sensitive CRC cell lines exhibited G1 inhibition. Moreover, those CRC cell lines that were responsive (0.08 μmol/L) to treatment demonstrated a significant baseline increase in Src and FAK gene expression. Conclusion Dasatinib demonstrated significant anti-proliferative activity in a subset of CRC cell lines in vitro, especially in those with increased Src expression at baseline, but only showed modest efficacy in CRC explants. Dasatinib is currently being studied in combination with chemotherapy in patients with advanced CRC, as its use as a single agent appears limited.

Collaboration


Dive into the Alicia Purkey's collaboration.

Top Co-Authors

Avatar

John J. Arcaroli

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Stacey Bagby

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Wells A. Messersmith

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Kevin Quackenbush

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Todd M. Pitts

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eun Kee Song

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

S. Gail Eckhardt

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Anna Capasso

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Benjamin Cross

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge