Jeffrey S. Wefel
University of Texas MD Anderson Cancer Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jeffrey S. Wefel.
The New England Journal of Medicine | 2014
Mark R. Gilbert; James J. Dignam; Terri S. Armstrong; Jeffrey S. Wefel; Deborah T. Blumenthal; Michael A. Vogelbaum; Howard Colman; Arnab Chakravarti; Stephanie L. Pugh; Minhee Won; R Jeraj; Paul D. Brown; Kurt A. Jaeckle; David Schiff; Volker W. Stieber; David Brachman; Maria Werner-Wasik; Ivo W. Tremont-Lukats; Erik P. Sulman; Kenneth D. Aldape; Walter J. Curran; Minesh P. Mehta
BACKGROUND Concurrent treatment with temozolomide and radiotherapy followed by maintenance temozolomide is the standard of care for patients with newly diagnosed glioblastoma. Bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor A, is currently approved for recurrent glioblastoma. Whether the addition of bevacizumab would improve survival among patients with newly diagnosed glioblastoma is not known. METHODS In this randomized, double-blind, placebo-controlled trial, we treated adults who had centrally confirmed glioblastoma with radiotherapy (60 Gy) and daily temozolomide. Treatment with bevacizumab or placebo began during week 4 of radiotherapy and was continued for up to 12 cycles of maintenance chemotherapy. At disease progression, the assigned treatment was revealed, and bevacizumab therapy could be initiated or continued. The trial was designed to detect a 25% reduction in the risk of death and a 30% reduction in the risk of progression or death, the two coprimary end points, with the addition of bevacizumab. RESULTS A total of 978 patients were registered, and 637 underwent randomization. There was no significant difference in the duration of overall survival between the bevacizumab group and the placebo group (median, 15.7 and 16.1 months, respectively; hazard ratio for death in the bevacizumab group, 1.13). Progression-free survival was longer in the bevacizumab group (10.7 months vs. 7.3 months; hazard ratio for progression or death, 0.79). There were modest increases in rates of hypertension, thromboembolic events, intestinal perforation, and neutropenia in the bevacizumab group. Over time, an increased symptom burden, a worse quality of life, and a decline in neurocognitive function were more frequent in the bevacizumab group. CONCLUSIONS First-line use of bevacizumab did not improve overall survival in patients with newly diagnosed glioblastoma. Progression-free survival was prolonged but did not reach the prespecified improvement target. (Funded by the National Cancer Institute; ClinicalTrials.gov number, NCT00884741.).
Journal of Clinical Oncology | 2013
Mark R. Gilbert; Meihua Wang; Kenneth D. Aldape; Roger Stupp; Monika E. Hegi; Kurt A. Jaeckle; Terri S. Armstrong; Jeffrey S. Wefel; Minhee Won; Deborah T. Blumenthal; Anita Mahajan; Christopher J. Schultz; Sara Erridge; Brigitta G. Baumert; Kristen I. Hopkins; Tzahala Tzuk-Shina; Paul D. Brown; Arnab Chakravarti; Walter J. Curran; Minesh P. Mehta
PURPOSE Radiotherapy with concomitant and adjuvant temozolomide is the standard of care for newly diagnosed glioblastoma (GBM). O(6)-methylguanine-DNA methyltransferase (MGMT) methylation status may be an important determinant of treatment response. Dose-dense (DD) temozolomide results in prolonged depletion of MGMT in blood mononuclear cells and possibly in tumor. This trial tested whether DD temozolomide improves overall survival (OS) or progression-free survival (PFS) in patients with newly diagnosed GBM. PATIENTS AND METHODS This phase III trial enrolled patients older than age 18 years with a Karnofsky performance score of ≥ 60 with adequate tissue. Stratification included clinical factors and tumor MGMT methylation status. Patients were randomly assigned to standard temozolomide (arm 1) or DD temozolomide (arm 2) for 6 to 12 cycles. The primary end point was OS. Secondary analyses evaluated the impact of MGMT status. RESULTS A total of 833 patients were randomly assigned to either arm 1 or arm 2 (1,173 registered). No statistically significant difference was observed between arms for median OS (16.6 v 14.9 months, respectively; hazard ratio [HR], 1.03; P = .63) or median PFS (5.5 v 6.7 months; HR, 0.87; P = .06). Efficacy did not differ by methylation status. MGMT methylation was associated with improved OS (21.2 v 14 months; HR, 1.74; P < .001), PFS (8.7 v 5.7 months; HR, 1.63; P < .001), and response (P = .012). There was increased grade ≥ 3 toxicity in arm 2 (34% v 53%; P < .001), mostly lymphopenia and fatigue. CONCLUSION This study did not demonstrate improved efficacy for DD temozolomide for newly diagnosed GBM, regardless of methylation status. However, it did confirm the prognostic significance of MGMT methylation. Feasibility of large-scale accrual, prospective tumor collection, and molecular stratification was demonstrated.
Neuro-oncology | 2013
Paul D. Brown; Stephanie L. Pugh; Nadia N. Laack; Jeffrey S. Wefel; Deepak Khuntia; Christina A. Meyers; Ali K. Choucair; Sherry Fox; John H. Suh; David Roberge; Vivek Kavadi; Søren M. Bentzen; Minesh P. Mehta; Deborah Watkins-Bruner
BACKGROUND To determine the protective effects of memantine on cognitive function in patients receiving whole-brain radiotherapy (WBRT). METHODS Adult patients with brain metastases received WBRT and were randomized to receive placebo or memantine (20 mg/d), within 3 days of initiating radiotherapy for 24 weeks. Serial standardized tests of cognitive function were performed. RESULTS Of 554 patients who were accrued, 508 were eligible. Grade 3 or 4 toxicities and study compliance were similar in the 2 arms. There was less decline in delayed recall in the memantine arm at 24 weeks (P = .059), but the difference was not statistically significant, possibly because there were only 149 analyzable patients at 24 weeks, resulting in only 35% statistical power. The memantine arm had significantly longer time to cognitive decline (hazard ratio 0.78, 95% confidence interval 0.62-0.99, P = .01); the probability of cognitive function failure at 24 weeks was 53.8% in the memantine arm and 64.9% in the placebo arm. Superior results were seen in the memantine arm for executive function at 8 (P = .008) and 16 weeks (P = .0041) and for processing speed (P = .0137) and delayed recognition (P = .0149) at 24 weeks. CONCLUSIONS Memantine was well tolerated and had a toxicity profile very similar to placebo. Although there was less decline in the primary endpoint of delayed recall at 24 weeks, this lacked statistical significance possibly due to significant patient loss. Overall, patients treated with memantine had better cognitive function over time; specifically, memantine delayed time to cognitive decline and reduced the rate of decline in memory, executive function, and processing speed in patients receiving WBRT. RTOG 0614, ClinicalTrials.gov number CT00566852.
International Journal of Radiation Oncology Biology Physics | 2011
Victor A. Levin; Luc Bidaut; Ping Hou; Ashok J. Kumar; Jeffrey S. Wefel; B. Nebiyou Bekele; Sujit S. Prabhu; Monica Elena Loghin; Mark R. Gilbert; Edward F. Jackson
PURPOSE To conduct a controlled trial of bevacizumab for the treatment of symptomatic radiation necrosis of the brain. METHODS AND MATERIALS A total of 14 patients were entered into a placebo-controlled randomized double-blind study of bevacizumab for the treatment of central nervous system radiation necrosis. All patients were required to have radiographic or biopsy proof of central nervous system radiation necrosis and progressive neurologic symptoms or signs. Eligible patients had undergone irradiation for head-and-neck carcinoma, meningioma, or low- to mid-grade glioma. Patients were randomized to receive intravenous saline or bevacizumab at 3-week intervals. The magnetic resonance imaging findings 3 weeks after the second treatment and clinical signs and symptoms defined the response or progression. RESULTS The volumes of necrosis estimated on T(2)-weighted fluid-attenuated inversion recovery and T(1)-weighted gadolinium-enhanced magnetic resonance imaging scans demonstrated that although no patient receiving placebo responded (0 of 7), all bevacizumab-treated patients did so (5 of 5 randomized and 7 of 7 crossover) with decreases in T(2)-weighted fluid-attenuated inversion recovery and T(1)-weighted gadolinium-enhanced volumes and a decrease in endothelial transfer constant. All bevacizumab-treated patients-and none of the placebo-treated patients-showed improvement in neurologic symptoms or signs. At a median of 10 months after the last dose of bevacizumab in patients receiving all four study doses, only 2 patients had experienced a recurrence of magnetic resonance imaging changes consistent with progressive radiation necrosis; one patient received a single additional dose of bevacizumab and the other patient received two doses. CONCLUSION The Class I evidence of bevacizumab efficacy from the present study in the treatment of central nervous system radiation necrosis justifies consideration of this treatment option for people with radiation necrosis secondary to the treatment of head-and-neck cancer and brain cancer.
Cancer | 2010
Jeffrey S. Wefel; Angele K. Saleeba; Aman U. Buzdar; Christina A. Meyers
Growing evidence supports cognitive dysfunction associated with standard dose chemotherapy in breast cancer survivors. We determined the incidence, nature, and chronicity of cognitive dysfunction in a prospective longitudinal randomized phase 3 treatment trial for patients with T1‐3, N0‐1, M0 breast cancer receiving 5‐fluorouracil, doxorubicin, and cyclophosphamide with or without paclitaxel.
Journal of Clinical Oncology | 2011
James Welsh; Ritsuko Komaki; Arya Amini; Mark F. Munsell; Wyatt Unger; Pamela K. Allen; Joe Y. Chang; Jeffrey S. Wefel; Susan L. McGovern; Linda Garland; Su S. Chen; Jamie Holt; Zhongxing Liao; Paul D. Brown; Erik P. Sulman; John V. Heymach; Edward S. Kim; Baldassarre Stea
PURPOSE Brain metastasis (BM) is a leading cause of death from non-small-cell lung cancer (NSCLC). Reasoning that activation of the epidermal growth factor receptor (EGFR) contributes to radiation resistance, we undertook a phase II trial of the EGFR inhibitor erlotinib with whole-brain radiation therapy (WBRT) in an attempt to extend survival time for patients with BM from NSCLC. Additional end points were radiologic response and safety. PATIENTS AND METHODS Eligible patients had BM from NSCLC, regardless of EGFR status. Erlotinib was given at 150 mg orally once per day for 1 week, then concurrently with WBRT (2.5 Gy per day 5 days per week, to 35 Gy), followed by maintenance. EGFR mutation status was tested by DNA sequencing at an accredited core facility. RESULTS Forty patients were enrolled and completed erlotinib plus WBRT (median age, 59 years; median diagnosis-specific graded prognostic assessment score, 1.5). The overall response rate was 86% (n = 36). No increase in neurotoxicity was detected, and no patient experienced grade ≥ 4 toxicity, but three patients required dose reduction for grade 3 rash. At a median follow-up of 28.5 months (for living patients), median survival time was 11.8 months (95% CI, 7.4 to 19.1 months). Of 17 patients with known EGFR status, median survival time was 9.3 months for those with wild-type EGFR and 19.1 months for those with EGFR mutations. CONCLUSION Erlotinib was well tolerated in combination with WBRT, with a favorable objective response rate. The higher-than-expected rate of EGFR mutations in these unselected patients raises the possibility that EGFR-mutated tumors are prone to brain dissemination.
Oncologist | 2008
Jorg Dietrich; Michelle Monje; Jeffrey S. Wefel; Christina A. Meyers
Standard oncological therapies, such as chemotherapy and cranial radiotherapy, frequently result in a spectrum of neurocognitive deficits that includes impaired learning, memory, attention, and speed of information processing. In addition to classical mechanisms of neurotoxicity associated with chemo- and radiotherapy, such as radiation necrosis and leukoencephalopathy, damage to dynamic progenitor cell populations in the brain is emerging as an important etiologic factor. Radiation- and chemotherapy-induced damage to progenitor populations responsible for maintenance of white matter integrity and adult hippocampal neurogenesis is now believed to play a major role in the neurocognitive impairment many cancer survivors experience.
Current Neurology and Neuroscience Reports | 2012
Jeffrey S. Wefel; Sanne B. Schagen
Many cancer patients develop treatment-related cognitive dysfunction that affects their quality of life and can result in diminished functional independence. There is an emerging body of transdisciplinary research demonstrating that chemotherapeutic agents can produce neurobiological changes within the brain, which are associated with a constellation of cognitive changes that can result in decreased quality of life and functional independence. The increased incidence of cancer, coupled with longer survival times, has resulted in larger numbers of cancer survivors who are struggling with this neurotoxicity. This review summarizes the neuropsychological findings in patients with breast and brain cancer who receive systemic chemotherapy as well as the recent animal and imaging research elucidating the mechanisms by which these therapies impact brain structure, function, and consequent behavior.
Neurosurgery | 2007
Eric L. Chang; Jeffrey S. Wefel; Moshe H. Maor; Samuel J. Hassenbusch; Anita Mahajan; Frederick F. Lang; Shiao Y. Woo; Leni A. Mathews; Pamela K. Allen; Almon S. Shiu; Christina A. Meyers
OBJECTIVEWhether to administer or omit adjuvant whole-brain radiation therapy in conjunction with stereotactic radiosurgery (SRS) in the initial management of patients with one to three newly diagnosed brain metastases is the subject of debate. This report provides data from a pilot study in which neurocognitive function (NCF) was prospectively measured for patients with one to three newly diagnosed brain metastases treated with initial SRS alone. METHODSFifteen patients were prospectively treated with initial SRS alone. Assessment of NCF and magnetic resonance imaging scans were performed. RESULTSAt baseline, 67% of the patients had impairment on one or more tests of NCF. The domains most frequently impaired at baseline were executive function, motor dexterity, and learning/memory with an incidence of 50, 40, and 27% respectively. Brain metastasis volume (.3 cm3) measured at the time of initial SRS treatment was associated with worse performance on a measure of attention (P < 0.05). At 1 month, declines in the learning/memory and motor dexterity domains were most common. In a subgroup of five patients still alive 200 days after enrollment, four patients (80%) demonstrated stable or improved learning/memory, three (60%) demonstrated stable or improved executive function, and three (60%) demonstrated stable or improved motor dexterity relative to their baseline evaluation. CONCLUSIONAlthough two-thirds of the brain metastasis patients had impaired NCF at baseline, the majority of five long-term survivors had stable or improved NCF performance across executive function, learning/memory, and motor dexterity.
CA: A Cancer Journal for Clinicians | 2015
Jeffrey S. Wefel; Shelli R. Kesler; Kyle R. Noll; Sanne B. Schagen
Answer questions and earn CME/CNE