Network


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

Hotspot


Dive into the research topics where Whitney B. Pope is active.

Publication


Featured researches published by Whitney B. Pope.


Journal of Clinical Oncology | 2011

Phase II Study of Bevacizumab Plus Temozolomide During and After Radiation Therapy for Patients With Newly Diagnosed Glioblastoma Multiforme

Albert Lai; Anh Tran; Phioanh L. Nghiemphu; Whitney B. Pope; Orestes E. Solis; Michael T. Selch; Emese Filka; William H. Yong; Paul S. Mischel; Linda M. Liau; Surasak Phuphanich; Keith L. Black; Scott Peak; Richard M. Green; Cynthia Elizabeth Spier; Tatjana Kolevska; Jonathan Polikoff; Louis Fehrenbacher; Robert Elashoff; Timothy F. Cloughesy

PURPOSE This open-label, prospective, multicenter single-arm phase II study combined bevacizumab (BV) with radiation therapy (RT) and temozolomide (TMZ) for the treatment of newly diagnosed glioblastoma (GBM). The objectives were to determine the efficacy of this treatment combination and the associated toxicity. PATIENTS AND METHODS Seventy patients with newly diagnosed GBM were enrolled between August 2006 and November 2008. Patients received standard RT starting within 3 to 6 weeks after surgery with concurrent administration of daily TMZ and biweekly BV. After completion of RT, patients resumed TMZ for 5 days every 4 weeks and continued biweekly BV. MGMT promoter methylation was assessed on patient tumor tissue. A University of California, Los Angeles/Kaiser Permanente Los Angeles (KPLA) control cohort of newly diagnosed patients treated with first-line RT and TMZ who had mostly received BV at recurrence was derived for comparison. RESULTS The overall survival (OS) and progression-free survival (PFS) were 19.6 and 13.6 months, respectively, compared to 21.1 and 7.6 months in the University of California, Los Angeles/KPLA control cohort, and 14.6 and 6.9 months in the European Organisation for Research and Treatment of Cancer-National Cancer Institute of Canada cohort. Correlation of MGMT promoter methylation and improved OS and PFS was retained in the study group. Comparative subset analysis showed that poor prognosis patients (recursive partitioning analysis class V/VI) may derive an early benefit from the use of first-line BV. Toxicity attributable to RT/TMZ was similar, and additional toxicities were consistent with those reported in other BV trials. CONCLUSION Patients treated with BV and TMZ during and after RT showed improved PFS without improved OS compared to the University of California, Los Angeles/KPLA control group. Additional studies are warranted to determine if BV administered first-line improves survival compared to BV at recurrence.


Journal of Clinical Oncology | 2007

Predicting Treatment Response of Malignant Gliomas to Bevacizumab and Irinotecan by Imaging Proliferation With [18F] Fluorothymidine Positron Emission Tomography: A Pilot Study

Wei Chen; Sibylle Delaloye; Daniel H.S. Silverman; Cheri Geist; Johannes Czernin; James Sayre; Nagichettiar Satyamurthy; Whitney B. Pope; Albert Lai; Michael E. Phelps; Timothy F. Cloughesy

PURPOSE Evaluation of treatment effects in malignant brain tumors is challenging because of the lack of reliable response predictors of tumor response. This study examines the predictive value of positron emission tomography (PET) using [18F] fluorothymidine (FLT), an imaging biomarker of cell proliferation, in patients with recurrent malignant gliomas treated with bevacizumab in combination with irinotecan. PATIENTS AND METHODS Patients with recurrent malignant gliomas treated with biweekly cycles of bevacizumab and irinotecan were prospectively studied with FLT-PET at baseline, after 1 to 2 weeks, and after 6 weeks from start of treatment. A more than 25% reduction in tumor FLT uptake as measured by standardized uptake value was defined as a metabolic response. FLT responses were compared with response as shown by magnetic resonance imaging (MRI) and patient survival. RESULTS Twenty-one patients were included, and 19 were assessable for metabolic response evaluation with FLT-PET. There were nine responders (47%) and 10 nonresponders (53%). Metabolic responders survived three times as long as nonresponders (10.8 v 3.4 months; P = .003), and tended to have a prolonged progression-free survival (P = .061). Both early and later FLT-PET responses were more significant predictors of overall survival (1 to 2 weeks, P = .006; 6 weeks, P = .002), compared with the MRI responses (P = .060 for both 6-week and best responses). CONCLUSION FLT-PET as an imaging biomarker seems to be predictive of overall survival in bevacizumab and irinotecan treatment of recurrent gliomas. Whether FLT-PET performed as early as 1 to 2 week after starting treatment is as predictive as the study indicates at 6 weeks warrants further investigation.


Neurology | 2006

MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy

Whitney B. Pope; Albert Lai; Phioanh L. Nghiemphu; Paul S. Mischel; T. Cloughesy

To the Editor: Pope et al. retrospectively reviewed 14 patients with recurrent high-grade gliomas (HGG) treated with chemotherapy and bevacizumab.1 Fifty percent of patients responded to treatment determined by post-treatment MRI. Several aspects of this report merit commentary. All patients in the study previously failed primary therapy and one prior salvage therapy. The number of patients treated within 3 months of radiotherapy was unclear. Though the protocol specified at least 4 weeks elapsed since administration of radiotherapy, it is recognized that early delayed radiotherapy changes seen on MRI may simulate tumor progression and resolve over time.2 The performance of MRI to assess response to therapy in patients with recurrent HGG is customarily performed every 2 months and coordinated with chemotherapy cycles. In this report, first post-treatment MRI appears desynchronized with chemotherapy administration. As response rates were based on first post-treatment MRI, response appears not to be bevacizumab dose dependent. To this author’s knowledge, there has not been a single agent trial of bevacizumab for recurrent HGG though trials in other solid cancers indicate a less than 10% single agent response rate. The rationale for combining bevacizumab with CPT-11 was not mentioned in this article. A number of reports evaluating CPT-11 as a single agent for recurrent glioblastoma have concluded the agent has little efficacy.3 The first report by Stark-Vance indicating response to the combination of bevacizumab and CPT-11 for recurrent HGG was not cited.4 The durability of response was also not reported. Clinical trials in recurrent HGG report response duration by overall survival, time to tumor progression, and 6-month progression free survival. Fifteen percent 6-month progression free survival for patients with recurrent glioblastoma is used as the standard by which new trials are compared with respect to efficacy.5 A variety of methods have been utilized to assess response to anti-angiogenic agents including dynamic contrast enhanced MRI. These methods provide proof of principle by quantifying tumor blood volume or perfusion. Pope et al. do not report whether the observed response represents an anti-angiogenic effect on tumor vasculature or an effect on blood brain barrier disruption with normalization of peritumoral edema and diminished tumor contrast enhancement. Pope et al emphasize the complexity of treating recurrent HGG and remind neurologists that the approach to malignant gliomas continues to evolve and will increasingly utilize targeted therapies such as antiangiogenic agents.


Radiology | 2009

Recurrent Glioblastoma Multiforme: ADC Histogram Analysis Predicts Response to Bevacizumab Treatment

Whitney B. Pope; Hyun J. Kim; Jing Huo; Jeffry R. Alger; Matthew S. Brown; David W. Gjertson; Victor Sai; Jonathan R. Young; Leena Tekchandani; Timothy F. Cloughesy; Paul S. Mischel; Albert Lai; Phioanh L. Nghiemphu; Syed Rahmanuddin; Jonathan G. Goldin

PURPOSE To determine if apparent diffusion coefficient (ADC) histogram analysis can stratify progression-free survival in patients with recurrent glioblastoma multiforme (GBM) prior to bevacizumab treatment. MATERIALS AND METHODS The study was approved by the institutional review board and was HIPAA compliant; informed consent was obtained. Bevacizumab-treated and control patients (41 per cohort) diagnosed with recurrent GBM were analyzed by using whole enhancing tumor ADC histograms with a two normal distribution mixture fitting curve on baseline (pretreatment) magnetic resonance (MR) images to generate ADC classifiers, including the overall mean ADC as well as the mean ADC from the lower curve (ADC(L)). Overall and 6-month progression-free survival (as defined by the Macdonald criteria) was determined by using Cox proportional hazard ratios and the Kaplan-Meier method with log-rank test. RESULTS For bevacizumab-treated patients, the hazard ratio for progression by 6 months in patients with less than versus greater than mean ADC(L) was 4.1 (95% confidence interval: 1.6, 10.4), and there was a 2.75-fold reduction in the median time to progression. For the control patients, there was no significant difference in median time to progression for the patients with low versus high ADC(L) (hazard ratio, 1.8; 95% confidence interval: 0.9, 3.7). For bevacizumab-treated patients, pretreatment ADC more accurately stratified 6-month progression-free survival than did change in enhancing tumor volume at first follow-up (73% vs 58% accuracy, P = .034). CONCLUSION Pretreatment ADC histogram analysis can stratify progression-free survival in bevacizumab-treated patients with recurrent GBM.


Journal of Clinical Oncology | 2011

Evidence for Sequenced Molecular Evolution of IDH1 Mutant Glioblastoma From a Distinct Cell of Origin

Albert Lai; Samir Kharbanda; Whitney B. Pope; Anh Tran; Orestes E. Solis; Franklin Peale; William F. Forrest; Kanan Pujara; Jose Carrillo; Ajay Pandita; Benjamin M. Ellingson; Chauncey W. Bowers; Robert Soriano; Nils Ole Schmidt; Sankar Mohan; William H. Yong; Somasekar Seshagiri; Zora Modrusan; Zhaoshi Jiang; Kenneth D. Aldape; Paul S. Mischel; Linda M. Liau; Cameron Escovedo; Weidong Chen; Phioanh L. Nghiemphu; C. David James; Michael D. Prados; Manfred Westphal; Katrin Lamszus; Timothy F. Cloughesy

PURPOSE Mutation in isocitrate dehydrogenase 1 (IDH1) at R132 (IDH1(R132MUT)) is frequent in low-grade diffuse gliomas and, within glioblastoma (GBM), has been proposed as a marker for GBMs that arise by transformation from lower-grade gliomas, regardless of clinical history. To determine how GBMs arising with IDH1(R132MUT) differ from other GBMs, we undertook a comprehensive comparison of patients presenting clinically with primary GBM as a function of IDH1(R132) mutation status. PATIENTS AND METHODS In all, 618 treatment-naive primary GBMs and 235 lower-grade diffuse gliomas were sequenced for IDH1(R132) and analyzed for demographic, radiographic, anatomic, histologic, genomic, epigenetic, and transcriptional characteristics. RESULTS Investigation revealed a constellation of features that distinguishes IDH1(R132MUT) GBMs from other GBMs (including frontal location and lesser extent of contrast enhancement and necrosis), relates them to lower-grade IDH1(R132MUT) gliomas, and supports the concept that IDH1(R132MUT) gliomas arise from a neural precursor population that is spatially and temporally restricted in the brain. The observed patterns of DNA sequence, methylation, and copy number alterations support a model of ordered molecular evolution of IDH1(R132MUT) GBM in which the appearance of mutant IDH1 protein is an initial event, followed by production of p53 mutant protein, and finally by copy number alterations of PTEN and EGFR. CONCLUSION Although histologically similar, GBMs arising with and without IDH1(R132MUT) appear to represent distinct disease entities that arise from separate cell types of origin as the result of largely nonoverlapping sets of molecular events. Optimal clinical management should account for the distinction between these GBM disease subtypes.


Neurology | 2009

Bevacizumab and chemotherapy for recurrent glioblastoma A single-institution experience

Phioanh L. Nghiemphu; W. Liu; Yohan Lee; T. Than; Carrie Anne Graham; Albert Lai; Richard M. Green; Whitney B. Pope; Linda M. Liau; Paul S. Mischel; Stanley F. Nelson; Robert M. Elashoff; T. Cloughesy

Objective: Bevacizumab has been shown to be effective in the treatment of recurrent glioblastoma in combination with chemotherapy compared with historic controls but not in randomized trials. Methods: We conducted a retrospective analysis of patients treated for recurrent glioblastoma with bevacizumab vs a control group of patients, comparing progression-free survival (PFS) and overall survival (OS) between the two groups, and performed subgroup analysis based on age and performance status. Expression of vascular endothelial growth factor (VEGF) based on age was examined using DNA microarray analysis. We also evaluated the impact of bevacizumab on quality of life. Results: We identified 44 patients who received bevacizumab and 79 patients who had not been treated with bevacizumab. There was a significant improvement in PFS and OS in the bevacizumab-treated group. Patients of older age (≥55 years) and poor performance status (Karnofsky Performance Status ≤80) had significantly better PFS when treated with bevacizumab, and bevacizumab-treated older patients had significantly increased OS. VEGF expression was significantly higher in older glioblastoma patients (aged ≥55 years). Patients treated with bevacizumab also required less dexamethasone use and maintained their functional status longer than the control group. Conclusions: Bevacizumab in combination with chemotherapy may be a more effective treatment for recurrent glioblastoma and warrants further randomized prospective studies to determine its effect on survival. Bevacizumab also has more effect in those with older age and might reflect biologic differences in glioblastoma in different age groups as seen with the expression of vascular endothelial growth factor. GBM = glioblastoma; HR = hazard ratio; KPS = Karnofsky Performance Status; OS = overall survival; PFS = progression-free survival; VEGF = vascular endothelial growth factor.


International Journal of Radiation Oncology Biology Physics | 2008

Phase II Pilot Study of Bevacizumab in Combination with Temozolomide and Regional Radiation Therapy for Up-Front Treatment of Patients With Newly Diagnosed Glioblastoma Multiforme: Interim Analysis of Safety and Tolerability

Albert Lai; Emese Filka; Bruce McGibbon; Phioanh L. Nghiemphu; Carrie Anne Graham; William H. Yong; Paul S. Mischel; Linda M. Liau; Marvin Bergsneider; Whitney B. Pope; Michael T. Selch; T. Cloughesy

PURPOSE To assess interim safety and tolerability of a 10-patient, Phase II pilot study using bevacizumab (BV) in combination with temozolomide (TMZ) and regional radiation therapy (RT) in the up-front treatment of patients with newly diagnosed glioblastoma. METHODS AND MATERIALS All patients received standard external beam regional RT of 60.0 Gy in 30 fractions started within 3 to 5 weeks after surgery. Concurrently TMZ was given daily at 75 mg/m(2) for 42 days during RT, and BV was given every 2 weeks at 10 mg/kg starting with the first day of RT/TMZ. After a 2-week interval upon completion of RT, the post-RT phase commenced with resumption of TMZ at 150 to 200 mg/m(2) for 5 days every 4 weeks and continuation of BV every 2 weeks. RESULTS For these 10 patients, toxicities were compiled until study discontinuation or up to approximately 40 weeks from initial study treatment for those remaining on-study. In terms of serious immediate or delayed neurotoxicity, 1 patient developed presumed radiation-induced optic neuropathy. Among the toxicities that could be potentially treatment related, relatively high incidences of fatigue, myelotoxicity, wound breakdown, and deep venous thrombosis/pulmonary embolism were observed. CONCLUSION The observed toxicities were acceptable to continue enrollment toward the overall target group of 70 patients. Preliminary efficacy analysis shows encouraging mean progression-free survival. At this time data are not sufficient to encourage routine off-label use of BV combined with TMZ/RT in the setting of newly diagnosed glioblastoma without longer follow-up, enrollment of additional patients, and thorough efficacy assessment.


American Journal of Neuroradiology | 2011

Apparent Diffusion Coefficient Histogram Analysis Stratifies Progression-Free Survival in Newly Diagnosed Bevacizumab-Treated Glioblastoma

Whitney B. Pope; Albert Lai; R. Mehta; H.J. Kim; J. Qiao; Jonathan R. Young; X. Xue; J. Goldin; M.S. Brown; Phioanh L. Nghiemphu; A. Tran; Timothy F. Cloughesy

BACKGROUND AND PURPOSE: Currently it is difficult to predict tumor response to anti-angiogenic therapy in individual patients. Our aim was to determine if ADC histogram analysis can stratify progression-free and overall survival in patients with newly diagnosed GBM treated “up-front” (ie, before tumor recurrence) with bevacizumab. MATERIALS AND METHODS: Up-front bevacizumab-treated and control patients (n = 59 and 62, respectively) with newly diagnosed GBM were analyzed by using an ADC histogram approach based on enhancing tumor. Progression-free and overall survival was determined by using Cox proportional HRs and the Kaplan-Meier method with logrank and Wilcoxon tests. RESULTS: For up-front bevacizumab-treated patients, lower ADCL was associated with significantly longer progression-free survival (median, 459 days for ADCL < 1200 versus 315 days for ADCL ≥ 1200 10−6mm2/s; P = .008, logrank test) and trended with longer overall survival (581 versus 429 days, P = .055). ADC values did not stratify progression-free or overall survival for patients in the control group (P = .92 and P = .22, respectively). Tumors with MGMT promoter methylation had lower ADCL values than unmethylated tumors (mean, 1071 versus 1183 10−6mm2/s; P = .01, 2-group t test). CONCLUSIONS: Pretreatment ADC histogram analysis can stratify progression-free survival in bevacizumab-treated patients with newly diagnosed GBM. Lower ADC is associated with tumor MGMT promoter methylation, which may, in part, account for the favorable outcome associated with low ADCL tumors.


Radiology | 2008

Relationship between Gene Expression and Enhancement in Glioblastoma Multiforme: Exploratory DNA Microarray Analysis

Whitney B. Pope; Jenny H. Chen; Jun Dong; Marc Carlson; Alla Perlina; Timothy F. Cloughesy; Linda M. Liau; Paul S. Mischel; Phioanh L. Nghiemphu; Albert Lai; Stanley F. Nelson

PURPOSE To determine the difference in gene expression between completely versus incompletely enhancing glioblastoma multiforme (GBM). MATERIALS AND METHODS Gene expression was determined for 52 newly diagnosed GBMs by using DNA microarrays, and the relationship to enhancement pattern and survival was analyzed. This study was approved by the institutional review board and was HIPAA compliant; informed consent was obtained. RESULTS Thirty-eight percent (20 of 52) of GBMs were incompletely enhancing (IE). The expression of eight genes was increased more than twofold in IE GBM when compared with completely enhancing (CE) GBM. Among these were tight junction protein-2 (2.2-fold increase, P = .019), and the oligodendroglioma markers oligodendrocyte lineage transcription factor 2 (2.4-fold increase, P = .029) and Achaete-scute complex-like 1 (ASCL1; 2.7-fold increase, P = .023). The expression of 71 genes showed relative overexpression in CE when compared with IE GBM. These included several proangiogenic and edema-related genes, including vascular endothelial growth factor (2.1-fold, P = .005) and neuronal pentraxin-2 (3.0-fold, P = .029). Several genes associated with primary GBM were overexpressed in CE tumors, whereas ASCL1, which is associated with secondary GBM, was overexpressed in IE tumors. Many genes overexpressed in IE GBM were associated with longer survival, whereas several genes overexpressed in CE GBM correlated with shortened survival. CONCLUSION The enhancement pattern divides GBM in two groups with differing prognoses. By comparing gene expression between IE and CE GBMs, it was possible to identify genes that may affect magnetic resonance imaging features of edema and enhancement, and genes whose expression levels are predictive of both improved and shortened survival.


Neuro-oncology | 2016

Response Assessment in Neuro-Oncology working group and European Association for Neuro-Oncology recommendations for the clinical use of PET imaging in gliomas.

Nathalie L. Albert; Michael Weller; Bogdana Suchorska; Norbert Galldiks; Riccardo Soffietti; Michelle M. Kim; Christian la Fougère; Whitney B. Pope; Ian Law; Javier Arbizu; Marc C. Chamberlain; Michael A. Vogelbaum; Ben Ellingson; Joerg Tonn

This guideline provides recommendations for the use of PET imaging in gliomas. The review examines established clinical benefit in glioma patients of PET using glucose ((18)F-FDG) and amino acid tracers ((11)C-MET, (18)F-FET, and (18)F-FDOPA). An increasing number of studies have been published on PET imaging in the setting of diagnosis, biopsy, and resection as well radiotherapy planning, treatment monitoring, and response assessment. Recommendations are based on evidence generated from studies which validated PET findings by histology or clinical course. This guideline emphasizes the clinical value of PET imaging with superiority of amino acid PET over glucose PET and provides a framework for the use of PET to assist in the management of patients with gliomas.

Collaboration


Dive into the Whitney B. Pope's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Albert Lai

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linda M. Liau

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin Leu

University of California

View shared research outputs
Top Co-Authors

Avatar

Hyun J. Kim

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge