Network


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

Hotspot


Dive into the research topics where Elizabeth A. Blair is active.

Publication


Featured researches published by Elizabeth A. Blair.


Clinical Cancer Research | 2015

Integrative and Comparative Genomic Analysis of HPV-Positive and HPV-Negative Head and Neck Squamous Cell Carcinomas

Tanguy Y. Seiwert; Zhixiang Zuo; Michaela K. Keck; Arun Khattri; Chandra Sekhar Pedamallu; Thomas Stricker; Christopher D. Brown; Trevor J. Pugh; Petar Stojanov; Juok Cho; Michael S. Lawrence; Gad Getz; Johannes Brägelmann; Rebecca DeBoer; Ralph R. Weichselbaum; Alexander Langerman; L. Portugal; Elizabeth A. Blair; Kerstin M. Stenson; Mark W. Lingen; Ezra E.W. Cohen; Everett E. Vokes; Kevin P. White; Peter S. Hammerman

Purpose: The genetic differences between human papilloma virus (HPV)–positive and –negative head and neck squamous cell carcinomas (HNSCC) remain largely unknown. To identify differential biology and novel therapeutic targets for both entities, we determined mutations and copy-number aberrations in a large cohort of locoregionally advanced HNSCC. Experimental Design: We performed massively parallel sequencing of 617 cancer-associated genes in 120 matched tumor/normal samples (42.5% HPV-positive). Mutations and copy-number aberrations were determined and results validated with a secondary method. Results: The overall mutational burden in HPV-negative and HPV-positive HNSCC was similar with an average of 15.2 versus 14.4 somatic exonic mutations in the targeted cancer-associated genes. HPV-negative tumors showed a mutational spectrum concordant with published lung squamous cell carcinoma analyses with enrichment for mutations in TP53, CDKN2A, MLL2, CUL3, NSD1, PIK3CA, and NOTCH genes. HPV-positive tumors showed unique mutations in DDX3X, FGFR2/3 and aberrations in PIK3CA, KRAS, MLL2/3, and NOTCH1 were enriched in HPV-positive tumors. Currently targetable genomic alterations were identified in FGFR1, DDR2, EGFR, FGFR2/3, EPHA2, and PIK3CA. EGFR, CCND1, and FGFR1 amplifications occurred in HPV-negative tumors, whereas 17.6% of HPV-positive tumors harbored mutations in fibroblast growth factor receptor genes (FGFR2/3), including six recurrent FGFR3 S249C mutations. HPV-positive tumors showed a 5.8% incidence of KRAS mutations, and DNA-repair gene aberrations, including 7.8% BRCA1/2 mutations, were identified. Conclusions: The mutational makeup of HPV-positive and HPV-negative HNSCC differs significantly, including targetable genes. HNSCC harbors multiple therapeutically important genetic aberrations, including frequent aberrations in the FGFR and PI3K pathway genes. Clin Cancer Res; 21(3); 632–41. ©2014 AACR. See related commentary by Krigsfeld and Chung, p. 495


Lancet Oncology | 2009

Erlotinib and bevacizumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck: a phase I/II study

Ezra E.W. Cohen; Darren W. Davis; Theodore Karrison; Tanguy Y. Seiwert; Stuart J. Wong; Sreenivasa Nattam; Mark Kozloff; Joseph I. Clark; Duen-Hwa Yan; Wen Liu; Carolyn Pierce; Janet Dancey; Kerstin M. Stenson; Elizabeth A. Blair; Allison Dekker; Everett E. Vokes

BACKGROUND Epidermal growth factor receptor (EGFR) is a validated target in squamous-cell carcinoma of the head and neck, but in patients with recurrent or metastatic disease, EGFR targeting agents have displayed modest efficacy. Vascular endothelial growth factor (VEGF)-mediated angiogenesis has been implicated as a mechanism of resistance to anti-EGFR therapy. In this multi-institutional phase I/II study we combined an EGFR inhibitor, erlotinib, with an anti-VEGF antibody, bevacizumab. METHODS Between April 15, 2003, and Jan 27, 2005, patients with recurrent or metastatic squamous-cell carcinoma of the head and neck were enrolled from seven centres in the USA and were given erlotinib (150 mg daily) and bevacizumab in escalating dose cohorts. The primary objectives in the phase I and II sections, respectively, were to establish the maximum tolerated dose and dose-limiting toxicity of bevacizumab when administered with erlotinib and to establish the proportion of objective responses and time to disease progression. Pretreatment serum and tissues were collected and analysed by enzyme-linked immunosorbent assay and immunofluorescence quantitative laser analysis, respectively. This study was registered with ClinicalTrials.gov, number NCT00055913. FINDINGS In the phase I section of the trial, ten patients were enrolled in three successive cohorts with no dose-limiting toxic effects noted. 46 patients were enrolled in the phase II section of the trial (including three patients from the phase I section) on the highest dose of bevacizumab (15 mg/kg every 3 weeks). Two additional patients were accrued beyond the protocol-stipulated 46, leaving a total of 48 patients for the phase II assessment. The most common toxic effects of any grade were rash and diarrhoea (41 and 16 of 48 patients, respectively). Three patients had serious bleeding events of grade 3 or higher. Seven patients had a response, with four showing a complete response allowing rejection of the null hypothesis. Median time of overall survival and progression-free survival (PFS) were 7.1 months (95% CI 5.7-9.0) and 4.1 months (2.8-4.4), respectively. Higher ratios of tumour-cell phosphorylated VEGF receptor-2 (pVEGFR2) over total VEGFR2 and endothelial-cell pEGFR over total EGFR in pretreatment biopsies were associated with complete response (0.704 vs 0.386, p=0.036 and 0.949 vs 0.332, p=0.036, respectively) and tumour shrinkage (p=0.007 and p=0.008, respectively) in a subset of 11 patients with available tissue. INTERPRETATION The combination of erlotinib and bevacizumab is well tolerated in recurrent or metastatic squamous-cell carcinoma of the head and neck. A few patients seem to derive a sustained benefit and complete responses were associated with expression of putative targets in pretreatment tumour tissue.


Journal of Clinical Oncology | 2010

Epidermal Growth Factor Receptor Inhibitor Gefitinib Added to Chemoradiotherapy in Locally Advanced Head and Neck Cancer

Ezra E.W. Cohen; Daniel J. Haraf; Rangesh Kunnavakkam; Kerstin M. Stenson; Elizabeth A. Blair; Bruce Brockstein; Eric P. Lester; Joseph K. Salama; Allison Dekker; R. Williams; Mary Ellyn Witt; Tatyana A. Grushko; James J. Dignam; Mark W. Lingen; Olufunmilayo I. Olopade; Everett E. Vokes

PURPOSE Assess efficacy and toxicity of gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, added to, and in maintenance after, concurrent chemoradiotherapy (CCRT) in locally advanced head and neck cancer (LA-HNC) and correlate outcomes with EGFR gene copy number alterations. PATIENTS AND METHODS Patients with stage III to IV LA-HNC received two cycles of carboplatin/paclitaxel induction chemotherapy (IC) followed by split-course CCRT with fluorouracil, hydroxyurea, twice daily radiotherapy (FHX), and gefitinib (250 mg daily) followed by continued gefitinib for 2 years total. The primary end point was complete response (CR) rate after CCRT. EGFR gene copy number was assessed by fluorescent in situ hybridization. RESULTS Sixty-nine patients (66 with stage IV disease, 37 with oropharynx primary tumors, and 67 with performance status 0 to 1) were enrolled with a median age of 55 years. Predominant grade 3 or 4 toxicities during IC and CCRT were neutropenia (n = 20) and in-field mucositis (n = 59) and dermatitis (n = 23), respectively. CR rate after CCRT was 90%. After median follow-up of 3.5 years, 4-year overall, progression-free, and disease-specific survival rates were 74%, 72%, and 89%, respectively. To date, one patient has developed a second primary tumor in the aerodigestive tract. In 31 patients with available tissue, high EGFR gene copy number was associated with worse overall survival (P = .02). CONCLUSION Gefitinib can be administered with FHX and as maintenance therapy for at least 2 years, demonstrating CR and survival rates that compare favorably with prior experience. High EGFR gene copy number may be associated with poor outcome in patients with LA-HNC treated with this regimen.


Clinical Cancer Research | 2012

A phase II study of lapatinib in recurrent/metastatic squamous cell carcinoma of the head and neck.

Jonas A. De Souza; Darren W. Davis; Yujian Zhang; Arun Khattri; Tanguy Y. Seiwert; Serdal Aktolga; Stuart J. Wong; Mark Kozloff; Sreenivasa Nattam; Mark W. Lingen; Rangesh Kunnavakkam; Kerstin M. Stenson; Elizabeth A. Blair; Jeffrey Bozeman; Janet Dancey; Everett E. Vokes; Ezra E.W. Cohen

Purpose: This study sought to determine the efficacy and safety profile of lapatinib in patients with recurrent/metastatic squamous cell carcinoma of the head and neck (SCCHN). Experimental Design: This phase II multiinstitutional study enrolled patients with recurrent/metastatic SCCHN into two cohorts: those without (arm A) and those with (arm B) before exposure to an epidermal growth factor receptor (EGFR) inhibitor. All subjects were treated with lapatinib 1,500 mg daily. Primary endpoints were response rate (arm A) and progression-free survival (PFS; arm B). The biologic effects of lapatinib on tumor growth and survival pathways were assessed in paired tumor biopsies obtained before and after therapy. Results: Forty-five patients were enrolled, 27 in arm A and 18 in arm B. Diarrhea was the most frequent toxicity occurring in 49% of patients. Seven patients experienced related grade 3 toxicity (3 fatigue, 2 hyponatremia, 1 vomiting, and 1 diarrhea). In an intent-to-treat analysis, no complete or partial responses were observed, and stable disease was the best response observed in 41% of arm A (median duration, 50 days, range, 34–159) and 17% of arm B subjects (median, 163 days, range, 135–195). Median PFS was 52 days in both arms. Median OS was 288 (95% CI, 62–374) and 155 (95% CI, 75–242) days for arms A and B, respectively. Correlative analyses revealed an absence of EGFR inhibition in tumor tissue. Conclusion: Lapatinib as a single agent in recurrent/metastatic SCCHN, although well tolerated, appears to be inactive in either EGFR inhibitor naive or refractory subjects. Clin Cancer Res; 18(8); 2336–43. ©2012 AACR.


Laryngoscope | 2006

Planned post-chemoradiation neck dissection: significance of radiation dose.

Kerstin M. Stenson; Dezheng Huo; Elizabeth A. Blair; Ezra E.W. Cohen; Athanassios Argiris; Daniel J. Haraf; Everett E. Vokes

Objective: The increasing prominence of multimodality therapy for patients with advanced head and neck cancer reflects its high survival and functional preservation rates. We report the pathologic data on patients undergoing neck dissection (ND) after induction chemotherapy followed by concomitant chemoradiotherapy (IC‐CRT) in three similar protocols utilizing decreasing doses of radiation therapy.


Laryngoscope | 2009

Chemoradiation for Patients With Advanced Oral Cavity Cancer

Kerstin M. Stenson; Rangesh Kunnavakkam; Ezra E.W. Cohen; L. Portugal; Elizabeth A. Blair; Daniel J. Haraf; Joseph K. Salama; Everett E. Vokes

Patients with advanced oral cavity cancer (OCC) typically have not been enrolled in clinical trials utilizing contemporary multimodality strategies. There exist dogmatic expectations of inferior outcome in OCC patients secondary to ineffectiveness of treatment and unacceptable toxicity. The purpose of this study was to analyze survival, swallowing function, and incidence of osteoradionecrosis (ORN) of patients with stage III/IV OCC who have undergone primary concomitant chemoradiotherapy (CRT).


Cancer | 2011

Prior chemoradiotherapy adversely impacts outcomes of recurrent and second primary head and neck cancer treated with concurrent chemotherapy and reirradiation.

Kevin S. Choe; Daniel J. Haraf; A.A. Solanki; Ezra E.W. Cohen; Tanguy Y. Seiwert; Kerstin M. Stenson; Elizabeth A. Blair; Louis G. Portugal; Victoria M. Villaflor; Mary Ellyn Witt; Everett E. Vokes; Joseph K. Salama

It has been shown that concomitant chemotherapy (C) with reirradiation (ReRT) is feasible and effective for select patients with recurrent or second primary head and neck cancer (HNC). To examine potential prognostic factors associated with survival, the authors of this report retrospectively reviewed the outcomes of patients who received CReRT.


Annals of Oncology | 2011

A randomized phase II study of 5-fluorouracil, hydroxyurea, and twice-daily radiotherapy compared with bevacizumab plus 5-fluorouracil, hydroxyurea, and twice-daily radiotherapy for intermediate-stage and T4N0-1 head and neck cancers

Joseph K. Salama; Daniel J. Haraf; K. Stenson; Elizabeth A. Blair; M. E. Witt; R. Williams; R. Kunnavakkam; Ezra E.W. Cohen; Tanguy Y. Seiwert; Everett E. Vokes

INTRODUCTION We conducted a randomized phase II study to evaluate the impact of adding bevacizumab (B) to 5-fluorouracil (5-FU), hydroxyurea (HU), and radiotherapy (FHX) for intermediate-stage and select T4 head and neck squamous cell cancers (HNSCC). PATIENTS AND METHODS Eligible patients had newly diagnosed HNSCC. Randomization was 2:1 in favor of BFHX. All patients received 500 mg HU p.o. b.i.d., 600 mg/m(2)/day continuous infusion 5-FU, and b.i.d. radiotherapy with or without bevacizumab 10 mg/kg administered on day 1 of each 14-day cycle. Patients received five cycles consisting of chemoradiotherapy for 5 days followed by 9 days without therapy. RESULTS Twenty-six patients were enrolled (19 BFHX and 7 FHX). The study was halted following unexpected locoregional progression. Two-year survival was 68%; 89% treated with FHX and 58% (95% confidence interval 33% to 78%) treated with BFHX. Two-year locoregional control was 80% after chemoradiotherapy and 85% after surgical salvage. All locoregional progression occurred in T4 tumors randomized to BFHX. Two patients receiving BFHX died during therapy, and one died shortly after therapy. No catastrophic bleeding events were seen. CONCLUSIONS Locoregional progression seen in T4N0-1 tumors treated with BFHX was unexpected and led to study termination. The addition of bevacuzimab to chemoradiotherapy for HNSCC should be limited clinical trials.


Archives of Otolaryngology-head & Neck Surgery | 2008

Characteristics Associated With Swallowing Changes After Concurrent Chemotherapy and Radiotherapy in Patients With Head and Neck Cancer

Joseph K. Salama; Kerstin M. Stenson; Marcy A. List; Loren K. Mell; Ellen MacCracken; Ezra E.W. Cohen; Elizabeth A. Blair; Everett E. Vokes; Daniel J. Haraf

OBJECTIVE To define factors that acutely influenced swallowing function prior to and during concurrent chemotherapy and radiotherapy. DESIGN A summary score from 1 to 7 (the swallowing performance status scale [SPS]) of oral and pharyngeal impairment, aspiration, and diet, was assigned to each patient study by a single senior speech and swallow pathologist, with higher scores indicating worse swallowing. Generalized linear regression models were formulated to asses the effects of patient factors (performance status, smoking intensity, amount of alcohol ingestion, and age), tumor factors (primary site, T stage, and N stage), and treatment-related factors (radiation dose, use of intensity-modulated radiation therapy, response to induction chemotherapy, postchemoradiotherapy neck dissection, and preprotocol surgery) on the differences between SPS score before and after treatment. SETTING University hospital tertiary care referral center. PATIENTS The study included 95 patients treated under a multiple institution, phase 2 protocol who underwent a videofluorographic oropharyngeal motility (OPM) study to assess swallowing function prior to and within 1 to 2 months after the completion of concurrent chemotherapy and radiotherapy. MAIN OUTCOME MEASURES Factors associated with swallowing changes after chemoradiotherapy. RESULTS The mean pretreatment and posttreatment OPM scores were 3.09 and 3.77, respectively. Patients with T3 or T4 tumors (odds ratio [OR], 0.38; 95% confidence interval [CI], 0.15-0.95; P = .04) and a performance status of 1 or 2 (OR, 0.37; 95% CI, 0.15-0.91; P = .03) were less likely to have worsening of swallowing after chemoradiotherapy. There was a trend for worse swallowing with increasing age (OR, 1.04; 95% CI, 0.99-1.09; P = .08). Only T stage (T3 or T4) was associated with improved swallowing after treatment (OR, 8.96; 95% CI, 1.9-41.5; P < .001). CONCLUSION In patients undergoing concurrent chemotherapy and radiotherapy, improved swallowing function over baseline is associated with advanced T stage.


JAMA Oncology | 2016

Erlotinib and the Risk of Oral Cancer: The Erlotinib Prevention of Oral Cancer (EPOC) Randomized Clinical Trial

William N. William; Vassiliki Papadimitrakopoulou; J. Jack Lee; Li Mao; Ezra E.W. Cohen; Heather Lin; Ann M. Gillenwater; Jack W. Martin; Mark W. Lingen; Jay O. Boyle; Dong M. Shin; Nadarajah Vigneswaran; Nancy Shinn; John V. Heymach; Ignacio I. Wistuba; Ximing Tang; Edward S. Kim; Pierre Saintigny; Elizabeth A. Blair; Timothy Meiller; J. Silvio Gutkind; Jeffrey N. Myers; Adel K. El-Naggar; Scott M. Lippman

IMPORTANCE Standard molecularly based strategies to predict and/or prevent oral cancer development in patients with oral premalignant lesions (OPLs) are lacking. OBJECTIVE To test if the epidermal growth factor receptor inhibitor erlotinib would reduce oral cancer development in patients with high-risk OPLs defined by specific loss of heterozygosity (LOH) profiles. Secondary objectives included prospective determination of LOH as a prognostic marker in OPLs. DESIGN The Erlotinib Prevention of Oral Cancer (EPOC) study was a randomized, placebo-controlled, double-bind trial. Accrual occurred from November 2006 through July 2012, with a median follow-up time of 35 months in an ambulatory care setting in 5 US academic referral institutions. Patients with OPLs were enrolled in the protocol, and each underwent LOH profiling (N = 379); they were classified as high-risk (LOH-positive) or low-risk (LOH-negative) patients based on their LOH profiles and oral cancer history. The randomized sample consisted of 150 LOH-positive patients. INTERVENTIONS Oral erlotinib treatment (150 mg/d) or placebo for 12 months. MAIN OUTCOMES AND MEASURES Oral cancer-free survival (CFS). RESULTS A total of 395 participants were classified with LOH profiles, and 254 were classified LOH positive. Of these, 150 (59%) were randomized, 75 each to the placebo and erlotinib groups. The 3-year CFS rates in placebo- and erlotinib-treated patients were 74% and 70%, respectively (hazard ratio [HR], 1.27; 95% CI, 0.68-2.38; P = .45). The 3-year CFS was significantly lower for LOH-positive compared with LOH-negative groups (74% vs 87%, HR, 2.19; 95% CI, 1.25-3.83; P = .01). Increased EGFR gene copy number correlated with LOH-positive status (P < .001) and lower CFS (P = .01). The EGFR gene copy number was not predictive of erlotinib efficacy. Erlotinib-induced skin rash was associated with improved CFS (P = .01). CONCLUSIONS AND RELEVANCE In this trial, LOH was validated as a marker of oral cancer risk and found to be associated with increased EGFR copy number (the target of the intervention). Erlotinib did not, however, improve CFS in high-risk patients with LOH-positive or high-EGFR-gene-copy-number OPLs. These results support incorporation of LOH testing as a prognostic tool in routine clinical practice but do not support erlotinib use in this setting. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00402779.

Collaboration


Dive into the Elizabeth A. Blair's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge