Elizabeth A. Harrington
AstraZeneca
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Elizabeth A. Harrington.
Cancer Research | 2008
Valeria R. Fantin; Andrey Loboda; Cloud P. Paweletz; Ronald C. Hendrickson; Jacqueline W. Pierce; Jennifer A. Roth; Lixia Li; Frank Gooden; Susan Korenchuk; Xiaoli S. Hou; Elizabeth A. Harrington; Sophia Randolph; John F. Reilly; Christopher Ware; Marshall E. Kadin; Stanley R. Frankel; Victoria M. Richon
Vorinostat is a histone deacetylase inhibitor that induces differentiation, growth arrest, and/or apoptosis of malignant cells both in vitro and in vivo and has shown clinical responses in approximately 30% of patients with advanced mycosis fungoides and Sézary syndrome cutaneous T-cell lymphoma (CTCL). The purpose of this study was to identify biomarkers predictive of vorinostat response in CTCL using preclinical model systems and to assess these biomarkers in clinical samples. The signal transducer and activator of transcription (STAT) signaling pathway was evaluated. The data indicate that persistent activation of STAT1, STAT3, and STAT5 correlate with resistance to vorinostat in lymphoma cell lines. Simultaneous treatment with a pan-Janus-activated kinase inhibitor resulted in synergistic antiproliferative effect and down-regulation of the expression of several antiapoptotic genes. Immunohistochemical analysis of STAT1 and phosphorylated tyrosine STAT3 (pSTAT3) in skin biopsies obtained from CTCL patients enrolled in the vorinostat phase IIb trial showed that nuclear accumulation of STAT1 and high levels of nuclear pSTAT3 in malignant T cells correlate with a lack of clinical response. These results suggest that deregulation of STAT activity plays a role in vorinostat resistance in CTCL, and strategies that block this pathway may improve vorinostat response. Furthermore, these findings may be of prognostic value in predicting the response of CTCL patients to vorinostat.
Cancer Research | 2015
Margaret Veldman-Jones; Roz Brant; Claire Rooney; Catherine Geh; Hollie Emery; Chris Harbron; Mark Wappett; Alan Sharpe; Michael Dymond; J. Carl Barrett; Elizabeth A. Harrington; Gayle Marshall
Analysis of clinical trial specimens such as formalin-fixed paraffin-embedded (FFPE) tissue for molecular mechanisms of disease progression or drug response is often challenging and limited to a few markers at a time. This has led to the increasing importance of highly multiplexed assays that enable profiling of many biomarkers within a single assay. Methods for gene expression analysis have undergone major advances in biomedical research, but obtaining a robust dataset from low-quality RNA samples, such as those isolated from FFPE tissue, remains a challenge. Here, we provide a detailed evaluation of the NanoString Technologies nCounter platform, which provides a direct digital readout of up to 800 mRNA targets simultaneously. We tested this system by examining a broad set of human clinical tissues for a range of technical variables, including sensitivity and limit of detection to varying RNA quantity and quality, reagent performance over time, variability between instruments, the impact of the number of fields of view sampled, and differences between probe sequence locations and overlapping genes across CodeSets. This study demonstrates that Nanostring offers several key advantages, including sensitivity, reproducibility, technical robustness, and utility for clinical application.
Clinical Cancer Research | 2015
Bristi Basu; Emma Dean; M. Puglisi; Alastair Greystoke; Michael Ong; Wendy Burke; Maria Cavallin; Graham Bigley; Christopher Womack; Elizabeth A. Harrington; Stephen Green; Elisabeth Oelmann; Johann S. de Bono; Malcolm R Ranson; Udai Banerji
Purpose: AZD2014 is a novel, oral, m-TORC 1/2 inhibitor that has shown in vitro and in vivo efficacy across a range of preclinical human cancer models. Experimental Design: A rolling six-dose escalation was performed to define an MTD (part A), and at MTD a further cohort of patients was treated to further characterize toxicities and perform pre- and posttreatment biopsies (part B). AZD2014 was administered orally twice a day continuously. Flow cytometry, ELISA, and immunohistochemistry were used to quantify pharmacodynamic biomarkers. Pharmacokinetic analysis was carried out by mass spectrometry. Results: A total of 56 patients were treated across a dose range of 25 to 100 mg. The MTD was 50 mg twice daily. The dose-limiting toxicities were fatigue and mucositis. At the MTD, the most common adverse events (AE) were fatigue (78%), nausea (51%), and mucositis (49%), but these were equal to or greater than grade 3 in only 5% of patients. Drug levels achieved at the MTD (AUCss 6686 ng·h/mL, Cmax ss 1,664 ng/mL) were consistent with activity in preclinical models. A reduction in p-S6 levels and Ki67 staining was observed in 8 of 8 and 5 of 9 evaluable paired biopsy samples. Partial responses were seen in a patient with pancreatic cancer and a patient with breast cancer, who were found to have a PDGFR and ERBB2 mutation, respectively. Conclusions: The recommended phase II dose for further evaluation of AZD2014 is 50 mg twice daily, and at this dose it has been possible to demonstrate pharmacologically relevant plasma concentrations, target inhibition in tumor, and clinical responses. Clin Cancer Res; 21(15); 3412–9. ©2015 AACR.
Clinical Cancer Research | 2015
Margaret Veldman-Jones; Zhongwu Lai; Mark Wappett; Chris Harbron; J. Carl Barrett; Elizabeth A. Harrington; Kenneth S. Thress
Purpose: Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease with distinct molecular subtypes. The most established subtyping approach, the “Cell of Origin” (COO) algorithm, categorizes DLBCL into activated B-cell (ABC) and germinal center B-cell (GCB)-like subgroups through gene expression profiling. Recently developed immunohistochemical (IHC) techniques and other established methodologies can deliver discordant results and have various technical limitations. We evaluated the NanoString nCounter gene expression system to address issues with current platforms. Experimental Design: We devised a scoring system using 145 genes from published datasets to categorize DLBCL samples. After cell line validation, clinical tissue segmentation was tested using commercially available diagnostic DLBCL samples. Finally, we profiled biopsies from patients with relapsed/refractory DLBCL enrolled in the fostamatinib phase IIb clinical trial using three independent RNA expression platforms: NanoString, Affymetrix, and qNPA. Results: Diagnostic samples showed a typical spread of subtypes with consistent gene expression profiles across matched fresh, frozen, and formalin-fixed paraffin-embedded tissues. Results from biopsy samples across platforms were remarkably consistent, in contrast to published IHC data. Interestingly, COO segmentation of longitudinal fostamatinib biopsies taken at initial diagnosis and then again at primary relapse showed 88% concordance (15/17), suggesting that COO designation remains stable over the course of disease progression. Conclusions: DLBCL segmentation of patient tumor samples is possible using a number of expression platforms. However, we found that NanoString offers the most flexibility and fewest limitations in regards to robust clinical tissue subtype characterization. These subtype distinctions should help guide disease prognosis and treatment options within DLBCL clinical practice. Clin Cancer Res; 21(10); 2367–78. ©2014 AACR. See related commentary by Rimsza, p. 2204
Molecular Cancer Therapeutics | 2015
Urs Hancox; Sabina Cosulich; Lyndsey Hanson; Cath Trigwell; Carol Lenaghan; Rebecca Ellston; Hannah Dry; Claire Crafter; Bernard Barlaam; Martina Fitzek; Paul D. Smith; Donald J. Ogilvie; Celina D'Cruz; Lillian Castriotta; Stephen R. Wedge; Lara Ward; Steve Powell; Mandy Lawson; Barry R. Davies; Elizabeth A. Harrington; Emily Foster; Marie Cumberbatch; Stephen Green; Simon T. Barry
Loss of PTEN protein results in upregulation of the PI3K/AKT pathway, which appears dependent on the PI3Kβ isoform. Inhibitors of PI3Kβ have potential to reduce growth of tumors in which loss of PTEN drives tumor progression. We have developed a small-molecule inhibitor of PI3Kβ and PI3Kδ (AZD8186) and assessed its antitumor activity across a panel of cell lines. We have then explored the antitumor effects as single agent and in combination with docetaxel in triple-negative breast (TNBC) and prostate cancer models. In vitro, AZD8186 inhibited growth of a range of cell lines. Sensitivity was associated with inhibition of the AKT pathway. Cells sensitive to AZD8186 (GI50 < 1 μmol/L) are enriched for, but not exclusively associated with, PTEN deficiency. In vivo, AZD8186 inhibits PI3K pathway biomarkers in prostate and TNBC tumors. Scheduling treatment with AZD8186 shows antitumor activity required only intermittent exposure, and that increased tumor control is achieved when AZD8186 is used in combination with docetaxel. AZD8186 is a potent inhibitor of PI3Kβ with activity against PI3Kδ signaling, and has potential to reduce growth of tumors dependent on dysregulated PTEN for growth. Moreover, AZD8186 can be combined with docetaxel, a chemotherapy commonly used to treat advanced TBNC and prostate tumors. The ability to schedule AZD8186 and maintain efficacy offers opportunity to combine AZD8186 more effectively with other drugs. Mol Cancer Ther; 14(1); 48–58. ©2014 AACR.
Clinical Cancer Research | 2017
Roz Brant; Alan Sharpe; Liptrot T; Jonathan R. Dry; Elizabeth A. Harrington; Barrett Jc; Whalley N; Womack C; Paul D. Smith; Darren Hodgson
Purpose: To develop a clinically viable gene expression assay to measure RAS/RAF/MEK/ERK (RAS–ERK) pathway output suitable for hypothesis testing in non–small cell lung cancer (NSCLC) clinical studies. Experimental Design: A published MEK functional activation signature (MEK signature) that measures RAS–ERK functional output was optimized for NSCLC in silico. NanoString assays were developed for the NSCLC optimized MEK signature and the 147-gene RAS signature. First, platform transfer from Affymetrix to NanoString, and signature modulation following treatment with KRAS siRNA and MEK inhibitor, were investigated in cell lines. Second, the association of the signatures with KRAS mutation status, dynamic range, technical reproducibility, and spatial and temporal variation was investigated in NSCLC formalin-fixed paraffin-embedded tissue (FFPET) samples. Results: We observed a strong cross-platform correlation and modulation of signatures in vitro. Technical and biological replicates showed consistent signature scores that were robust to variation in input total RNA; conservation of scores between primary and metastatic tumor was statistically significant. There were statistically significant associations between high MEK (P = 0.028) and RAS (P = 0.003) signature scores and KRAS mutation in 50 NSCLC samples. The signatures identify overlapping but distinct candidate patient populations from each other and from KRAS mutation testing. Conclusions: We developed a technically and biologically robust NanoString gene expression assay of MEK pathway output, compatible with the quantities of FFPET routinely available. The gene signatures identified a different patient population for MEK inhibitor treatment compared with KRAS mutation testing. The predictive power of the MEK signature should be studied further in clinical trials. Clin Cancer Res; 23(6); 1471–80. ©2016 AACR. See related commentary by Xue and Lito, p. 1365
PLOS ONE | 2016
Claire Rooney; Catherine Geh; Victoria Williams; Johannes M. Heuckmann; Roopika Menon; Petra Schneider; Katherine Al-Kadhimi; Michael Dymond; Neil R. Smith; Dawn Baker; Tim French; Paul D. Smith; Elizabeth A. Harrington; J. Carl Barrett; Elaine Kilgour
FGFR1 amplification occurs in ~20% of sqNSCLC and trials with FGFR inhibitors have selected FGFR1 amplified patients by FISH. Lung cancer cell lines were profiled for sensitivity to AZD4547, a potent, selective inhibitor of FGFRs 1–3. Sensitivity to FGFR inhibition was associated with but not wholly predicted by increased FGFR1 gene copy number. Additional biomarker assays evaluating expression of FGFRs and correlation between amplification and expression in clinical tissues are therefore warranted. We validated nanoString for mRNA expression analysis of 194 genes, including FGFRs, from clinical tumour tissue. In a panel of sqNSCLC tumours 14.4% (13/90) were FGFR1 amplified by FISH. Although mean FGFR1 expression was significantly higher in amplified samples, there was significant overlap in the range of expression levels between the amplified and non-amplified cohorts with several non-amplified samples expressing FGFR1 to levels equivalent to amplified samples. Statistical analysis revealed increased expression of FGFR1 neighboring genes on the 8p12 amplicon (BAG4, LSM1 and WHSC1L1) in FGFR1 amplified tumours, suggesting a broad rather than focal amplicon and raises the potential for codependencies. High resolution aCGH analysis of pre-clinical and clinical samples supported the presence of a broad and heterogeneous amplicon around the FGFR1 locus. In conclusion, the range of FGFR1 expression levels in both FGFR1 amplified and non-amplified NSCLC tissues, together with the breadth and intra-patient heterogeneity of the 8p amplicon highlights the need for gene expression analysis of clinical samples to inform the understanding of determinants of response to FGFR inhibitors. In this respect the nanoString platform provides an attractive option for RNA analysis of FFPE clinical samples.
BMC Clinical Pathology | 2016
Caroline Seiler; Alan Sharpe; J. Carl Barrett; Elizabeth A. Harrington; Emma V. Jones; Gayle Marshall
BackgroundAdvanced genomic techniques such as Next-Generation-Sequencing (NGS) and gene expression profiling, including NanoString, are vital for the development of personalised medicines, as they enable molecular disease classification. This has become increasingly important in the treatment of cancer, aiding patient selection. However, it requires efficient nucleic acid extraction often from formalin-fixed paraffin-embedded tissue (FFPE).MethodsHere we provide a comparison of several commercially available manual and automated methods for DNA and/or RNA extraction from FFPE cancer cell line samples from Qiagen, life Technologies and Promega. Differing extraction geometric mean yields were evaluated across each of the kits tested, assessing dual DNA/RNA extraction vs. specialised single extraction, manual silica column based extraction techniques vs. automated magnetic bead based methods along with a comparison of subsequent nucleic acid purity methods, providing a full evaluation of nucleic acids isolated.ResultsOut of the four RNA extraction kits evaluated the RNeasy FFPE kit, from Qiagen, gave superior geometric mean yields, whilst the Maxwell 16 automated method, from Promega, yielded the highest quality RNA by quantitative real time RT-PCR. Of the DNA extraction kits evaluated the PicoPure DNA kit, from Life Technologies, isolated 2–14× more DNA. A miniaturised qPCR assay was developed for DNA quantification and quality assessment.ConclusionsCareful consideration of an extraction kit is necessary dependent on quality or quantity of material required. Here we provide a flow diagram on the factors to consider when choosing an extraction kit as well as how to accurately quantify and QC the extracted material.
Annals of Oncology | 2018
Bristi Basu; Matthew Krebs; Raghav Sundar; Richard Wilson; James Spicer; Robert Jones; M. Brada; Denis C. Talbot; Nicola Steele; A.H. Ingles Garces; Wolfram Brugger; Elizabeth A. Harrington; J. Evans; Emma Hall; Holly Tovey; Fm de Oliveira; Suzanne Carreira; Karen E Swales; Ruth Ruddle; Florence I. Raynaud; Beth Purchase; Joanna C Dawes; Mona Parmar; Alison Turner; Nina Tunariu; Susana Banerjee; J. S. De Bono; Udai Banerji
Abstract Background We have previously shown that raised p-S6K levels correlate with resistance to chemotherapy in ovarian cancer. We hypothesised that inhibiting p-S6K signalling with the dual m-TORC1/2 inhibitor in patients receiving weekly paclitaxel could improve outcomes in such patients. Patients and methods In dose escalation, weekly paclitaxel (80 mg/m2) was given 6/7 weeks in combination with two intermittent schedules of vistusertib (dosing starting on the day of paclitaxel): schedule A, vistusertib dosed bd for 3 consecutive days per week (3/7 days) and schedule B, vistusertib dosed bd for 2 consecutive days per week (2/7 days). After establishing a recommended phase II dose (RP2D), expansion cohorts in high-grade serous ovarian cancer (HGSOC) and squamous non-small-cell lung cancer (sqNSCLC) were explored in 25 and 40 patients, respectively. Results The dose-escalation arms comprised 22 patients with advanced solid tumours. The dose-limiting toxicities were fatigue and mucositis in schedule A and rash in schedule B. On the basis of toxicity and pharmacokinetic (PK) and pharmacodynamic (PD) evaluations, the RP2D was established as 80 mg/m2 paclitaxel with 50 mg vistusertib bd 3/7 days for 6/7 weeks. In the HGSOC expansion, RECIST and GCIG CA125 response rates were 13/25 (52%) and 16/25 (64%), respectively, with median progression-free survival (mPFS) of 5.8 months (95% CI: 3.28–18.54). The RP2D was not well tolerated in the SqNSCLC expansion, but toxicities were manageable after the daily vistusertib dose was reduced to 25 mg bd for the following 23 patients. The RECIST response rate in this group was 8/23 (35%), and the mPFS was 5.8 months (95% CI: 2.76–21.25). Discussion In this phase I trial, we report a highly active and well-tolerated combination of vistusertib, administered as an intermittent schedule with weekly paclitaxel, in patients with HGSOC and SqNSCLC. Clinical trial registration ClinicialTrials.gov identifier: CNCT02193633
Cancer Research | 2015
Manish R. Patel; Erika Paige Hamilton; Patricia LoRusso; W Larry Gluck; Suzanne F. Jones; Muaiad Kittaneh; Sabina Cosulich; Elizabeth A. Harrington; Stephen Green; Wendy Burke; Donald K Strickland; Elisabeth Oelmann; Howard A. Burris
Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: Preclinical and clinical (BOLERO 2) data suggest that pts with ER+ breast cancer become less sensitive to hormonal therapy over time with greater dependency on the mTOR pathway. AZD2014 is a selective dual mTORC1 and mTORC2 inhibitor that may offer additional benefit vs mTORC1 inhibitors through suppression of AKT via mTORC2. Preclinical data demonstrate that continuous or intermittent dosing schedules are equally effective in a model of ER+ breast cancer and the latter may achieve improved tolerability in pts. This Ph I trial assesses the safety, tolerability, PK, PD and preliminary efficacy of AZD2014 administered both continuously and intermittently in combination with fulvestrant (F); data are unvalidated and subject to change. Methods: Adult pts with ER+ metastatic breast cancer were treated with continuous or intermittent AZD2014 in combination with F 500mg intramuscularly on day 1 of each 28 day cycle. Results: In the continuous schedules, 43 pts have been treated in 4 cohorts: 50mg BD = 13; 35mg BD = 6; 100mg QD = 10; and 75mg QD = 14. In the intermittent schedules 23 pts have been treated on D1 and 2 (2 days out of 7) per week in 2 cohorts: 170mg BD = 8; 125mg BD = 15. DLTs on the continuous schedule included rash/stomatitis (1 pt) and hyperglycemia (1 pt) at 50mg BD; fatigue (1 pt) and stomatitis (1 pt) at 100mg QD; and rash (1 pt) at 75mg QD. There were no DLTs at 170mg or 125mg BD (D1&2) on the intermittent schedule. Treatment-related toxicities (any grade) in 66 pts include: nausea (58%), fatigue (52%), diarrhea (46%), stomatitis (41%), vomiting (32%), decreased appetite (30%), maculo-papular rash (26%), and hyperglycemia (15%). PK data show that AZD2014 is rapidly absorbed (median tmax 1-1.75h). The mean single dose terminal elimination t1/2 was 3.3-5.6h and increased following multiple and/or high dosing. There was no evidence that co-administration of F had a clinically relevant impact on AZD2014 exposure. Inhibition of both mTORC1 and 2 was observed in both surrogate and tumor tissue. In the 49 pts with measureable disease, 9 confirmed (duration 1.8-22 months) and 3 unconfirmed PRs were observed. Overall, CBR (CR+PR+SD >24 weeks) was 29/66. Conclusion: Continuous and intermittent dosing of AZD2014 in combination with F is tolerable with clinical benefit observed in 44% of all pts with clinical activity observed with both schedules. Toxicities observed with the continuous dosing schedule are broadly consistent with AEs observed in other trials with mTOR inhibitors. Notably, the intermittent schedule had a different AE profile with a lower incidence of rash. AZD2014 inhibited both mTORC1 and mTORC2 in surrogate and tumor tissue and the PK data were broadly consistent with previous findings for AZD2014 single agent. A randomized phase II trial of the combination comparing both the continuous and intermittent dosing schedules is ongoing. Citation Format: Manish Patel, Erika Hamilton, Patricia M. LoRusso, W. Larry Gluck, Suzanne F. Jones, Muaiad Kittaneh, Sabina Cosulich, Elizabeth A. Harrington, Stephen Green, Wendy Burke, Donald K. Strickland, Elisabeth Oelmann, Howard A. Burris. A phase I study evaluating continuous and intermittent AZD2014 in combination with fulvestrant in patients with ER+ advanced metastatic breast cancer. [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 CT233. doi:10.1158/1538-7445.AM2015-CT233