Network


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

Hotspot


Dive into the research topics where Eudocia Q. Lee is active.

Publication


Featured researches published by Eudocia Q. Lee.


Current Oncology Reports | 2012

Epidemiology of Brain Metastases

Lakshmi Nayak; Eudocia Q. Lee; Patrick Y. Wen

Brain metastases are one of the most common neurologic complications of cancer. The incidence is 9%–17% based on various studies, although the exact incidence is thought to be higher. The incidence is increasing with the availability of improved imaging techniques which aid early diagnosis, and effective systemic treatment regimens which prolong life, thus allowing cancer to disseminate to the brain. Lung cancer, breast cancer, and melanoma are the most frequent to develop brain metastases, and account for 67%–80% of all cancers. Most patients with brain metastases have synchronous extracerebral metastases. Some patients present with no known primary cancer diagnosis. In children, brain metastases are rare; germ cell tumors, sarcomas, and neuroblastoma are the common offenders.


Lancet Oncology | 2015

Response assessment criteria for brain metastases: proposal from the RANO group

Nan Lin; Eudocia Q. Lee; Igor J. Barani; Daniel P. Barboriak; Brigitta G. Baumert; Martin Bendszus; Paul D. Brown; D. Ross Camidge; Susan M. Chang; Janet Dancey; Elisabeth G.E. de Vries; Laurie E. Gaspar; Gordon J. Harris; F. Stephen Hodi; Steven N. Kalkanis; Mark E. Linskey; David R. Macdonald; Kim Margolin; Minesh P. Mehta; David Schiff; Riccardo Soffietti; John H. Suh; Martin J. van den Bent; Michael A. Vogelbaum; Patrick Y. Wen

CNS metastases are the most common cause of malignant brain tumours in adults. Historically, patients with brain metastases have been excluded from most clinical trials, but their inclusion is now becoming more common. The medical literature is difficult to interpret because of substantial variation in the response and progression criteria used across clinical trials. The Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) working group is an international, multidisciplinary effort to develop standard response and progression criteria for use in clinical trials of treatment for brain metastases. Previous efforts have focused on aspects of trial design, such as patient population, variations in existing response and progression criteria, and challenges when incorporating neurological, neuro-cognitive, and quality-of-life endpoints into trials of patients with brain metastases. Here, we present our recommendations for standard response and progression criteria for the assessment of brain metastases in clinical trials. The proposed criteria will hopefully facilitate the development of novel approaches to this difficult problem by providing more uniformity in the assessment of CNS metastases across trials.


Neuro-oncology | 2012

Current clinical development of PI3K pathway inhibitors in glioblastoma

Patrick Y. Wen; Eudocia Q. Lee; David A. Reardon; Keith L. Ligon; W. K. Alfred Yung

Glioblastoma (GBM) is the most common and lethal primary malignant tumor of the central nervous system, and effective therapeutic options are lacking. The phosphatidylinositol 3-kinase (PI3K) pathway is frequently dysregulated in many human cancers, including GBM. Agents inhibiting PI3K and its effectors have demonstrated preliminary activity in various tumor types and have the potential to change the clinical treatment landscape of patients with solid tumors. In this review, we describe the activation of the PI3K pathway in GBM, explore why inhibition of this pathway may be a compelling therapeutic target for this disease, and provide an update of the data on PI3K inhibitors in clinical trials and from earlier investigation.


Neuro-oncology | 2015

Phase II trial of sunitinib for recurrent and progressive atypical and anaplastic meningioma

Thomas Kaley; Patrick Y. Wen; David Schiff; Keith L. Ligon; Sam Haidar; Sasan Karimi; Andrew B. Lassman; Craig Nolan; Lisa M. DeAngelis; Igor T. Gavrilovic; Andrew D. Norden; Jan Drappatz; Eudocia Q. Lee; Benjamin Purow; Scott R. Plotkin; Tracy T. Batchelor; Lauren E. Abrey; Antonio Omuro

BACKGROUND No proven effective medical therapy for surgery and radiation-refractory meningiomas exists. Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. METHODS This was a prospective, multicenter, investigator-initiated single-arm phase II trial. The primary cohort enrolled patients with surgery and radiation-refractory recurrent World Health Organization (WHO) grades II-III meningioma. An exploratory cohort enrolled patients with WHO grade I meningioma, hemangiopericytoma, or hemangioblastoma. Sunitinib was administered at 50 mg/d for days 1-28 of every 42-day cycle. The primary endpoint was the rate of 6-month progression-free survival (PFS6), with secondary endpoints of radiographic response rate, safety, PFS, and overall survival. Exploratory objectives include analysis of tumoral molecular markers and MR perfusion imaging. RESULTS Thirty-six patients with high-grade meningioma (30 atypical and 6 anaplastic) were enrolled. Patients were heavily pretreated (median number of 5 recurrences, range 2-10). PFS6 rate was 42%, meeting the primary endpoint. Median PFS was 5.2 months (95% CI: 2.8-8.3 mo), and median overall survival was 24.6 months (95% CI: 16.5-38.4 mo). Thirteen patients enrolled in the exploratory cohort. Overall toxicity included 1 grade 5 intratumoral hemorrhage, 2 grade 3 and 1 grade 4 CNS/intratumoral hemorrhages, 1 grade 3 and 1 grade 4 thrombotic microangiopathy, and 1 grade 3 gastrointestinal perforation. Expression of VEGFR2 predicted PFS of a median of 1.4 months in VEGFR2-negative patients versus 6.4 months in VEGFR2-positive patients (P = .005). CONCLUSION Sunitinib is active in recurrent atypical/malignant meningioma patients. A randomized trial should be performed.


Neuro-oncology | 2012

Phase I/II study of sorafenib in combination with temsirolimus for recurrent glioblastoma or gliosarcoma: North American Brain Tumor Consortium study 05-02

Eudocia Q. Lee; John G. Kuhn; Kathleen R. Lamborn; Lauren E. Abrey; Lisa M. DeAngelis; F. Lieberman; H. Ian Robins; Susan M. Chang; W. K. Alfred Yung; Jan Drappatz; Minesh P. Mehta; Victor A. Levin; Kenneth D. Aldape; Janet Dancey; John J. Wright; Michael D. Prados; Timothy F. Cloughesy; Mark R. Gilbert; Patrick Y. Wen

The activity of single-agent targeted molecular therapies in glioblastoma has been limited to date. The North American Brain Tumor Consortium examined the safety, pharmacokinetics, and efficacy of combination therapy with sorafenib, a small molecule inhibitor of Raf, vascular endothelial growth factor receptor 2, and platelet-derived growth factor receptor-β, and temsirolimus (CCI-779), an inhibitor of mammalian target of rapamycin. This was a phase I/II study. The phase I component used a standard 3 × 3 dose escalation scheme to determine the safety and tolerability of this combination therapy. The phase II component used a 2-stage design; the primary endpoint was 6-month progression-free survival (PFS6) rate. Thirteen patients enrolled in the phase I component. The maximum tolerated dosage (MTD) for combination therapy was sorafenib 800 mg daily and temsirolimus 25 mg once weekly. At the MTD, grade 3 thrombocytopenia was the dose-limiting toxicity. Eighteen patients were treated in the phase II component. At interim analysis, the study was terminated and did not proceed to the second stage. No patients remained progression free at 6 months. Median PFS was 8 weeks. The toxicity of this combination therapy resulted in a maximum tolerated dose of temsirolimus that was only one-tenth of the single-agent dose. Minimal activity in recurrent glioblastoma multiforme was seen at the MTD of the 2 combined agents.


Lancet Oncology | 2013

Challenges relating to solid tumour brain metastases in clinical trials, part 1: patient population, response, and progression. A report from the RANO group

Nan Lin; Eudocia Q. Lee; Igor J. Barani; Brigitta G. Baumert; Paul D. Brown; D. Ross Camidge; Susan M. Chang; Janet Dancey; Laurie E. Gaspar; Gordon J. Harris; F. Stephen Hodi; Steven N. Kalkanis; Kathleen R. Lamborn; Mark E. Linskey; David R. Macdonald; Kim Margolin; Minesh P. Mehta; David Schiff; Riccardo Soffietti; John H. Suh; Martin J. van den Bent; Michael A. Vogelbaum; Jeffrey S. Wefel; Patrick Y. Wen

Therapeutic outcomes for patients with brain metastases need to improve. A critical review of trials specifically addressing brain metastases shows key issues that could prevent acceptance of results by regulatory agencies, including enrolment of heterogeneous groups of patients and varying definitions of clinical endpoints. Considerations specific to disease, modality, and treatment are not consistently addressed. Additionally, the schedule of CNS imaging and consequences of detection of new or progressive brain metastases in trials mainly exploring the extra-CNS activity of systemic drugs are highly variable. The Response Assessment in Neuro-Oncology (RANO) working group is an independent, international, collaborative effort to improve the design of trials in patients with brain tumours. In this two-part series, we review the state of clinical trials of brain metastases and suggest a consensus recommendation for the development of criteria for future clinical trials.


Lancet Oncology | 2013

Challenges relating to solid tumour brain metastases in clinical trials, part 2: neurocognitive, neurological, and quality-of-life outcomes. A report from the RANO group

Nan Lin; Jeffrey S. Wefel; Eudocia Q. Lee; David Schiff; Martin J. van den Bent; Riccardo Soffietti; John H. Suh; Michael A. Vogelbaum; Minesh P. Mehta; Janet Dancey; Mark E. Linskey; D. Ross Camidge; Paul D. Brown; Susan M. Chang; Steven N. Kalkanis; Igor J. Barani; Brigitta G. Baumert; Laurie E. Gaspar; F. Stephen Hodi; David R. Macdonald; Patrick Y. Wen

Neurocognitive function, neurological symptoms, functional independence, and health-related quality of life are major concerns for patients with brain metastases. The inclusion of these endpoints in trials of brain metastases and the methods by which these measures are assessed vary substantially. If functional independence or health-related quality of life are planned as key study outcomes, then the reliability and validity of these endpoints can be crucial because methodological issues might affect the interpretation and acceptance of findings. The Response Assessment in Neuro-Oncology (RANO) working group is an independent, international, and collaborative effort to improve the design of clinical trials in patients with brain tumours. In this report, the second in a two-part series, we review clinical trials of brain metastases in relation to measures of clinical benefit and provide a framework for the design and conduct of future trials.


Neuro-oncology | 2013

Phase 2 study of dose-intense temozolomide in recurrent glioblastoma

Andrew D. Norden; Glenn J. Lesser; Jan Drappatz; Keith L. Ligon; Samantha Hammond; Eudocia Q. Lee; David Reardon; Camilo E. Fadul; Scott R. Plotkin; Tracy T. Batchelor; Jay Jiguang Zhu; Rameen Beroukhim; Alona Muzikansky; Lisa Doherty; Debra C. LaFrankie; Katrina H. Smith; Vida Tafoya; Rosina T. Lis; Edward C. Stack; Myrna R. Rosenfeld; Patrick Y. Wen

BACKGROUND Among patients with glioblastoma (GBM) who progress on standard temozolomide, the optimal therapy is unknown. Resistance to temozolomide is partially mediated by O(6)-methylguanine-DNA methyltransferase (MGMT). Because MGMT may be depleted by prolonged temozolomide administration, dose-intense schedules may overcome resistance. METHODS This was a multicenter, phase 2, single-arm study of temozolomide (75-100 mg/m(2)/day) for 21 days of each 28-day cycle. Patients had GBM in first recurrence after standard therapy. The primary end point was 6-month progression-free survival (PFS6). RESULTS Fifty-eight participants were accrued, 3 of whom were ineligible for analysis; one withdrew before response assessment. There were 33 men (61%), with a median age of 57 years (range, 25-79 years) and a median Karnofsky performance score of 90 (range, 60-100). Of 47 patients with MGMT methylation results, 36 (65%) had methylated tumors. There were 7 (13%) partial responses, and PFS6 was only 11%. Response and PFS did not depend on MGMT status; MSH2, MLH1, or ERCC1 expression; the number of prior temozolomide cycles; or the time off temozolomide. Treatment was well tolerated, with limited grade 3 neutropenia (n = 2) or thrombocytopenia (n = 2). CONCLUSIONS Dose-intense temozolomide on this schedule is safe in recurrent GBM. However, efficacy is marginal and predictive biomarkers are needed.


Neuro-oncology | 2015

Medical management of brain tumors and the sequelae of treatment

David Schiff; Eudocia Q. Lee; Lakshmi Nayak; Andrew D. Norden; David A. Reardon; Patrick Y. Wen

Patients with malignant brain tumors are prone to complications that negatively impact their quality of life and sometimes their overall survival as well. Tumors may directly provoke seizures, hypercoagulable states with resultant venous thromboembolism, and mood and cognitive disorders. Antitumor treatments and supportive therapies also produce side effects. In this review, we discuss major aspects of supportive care for patients with malignant brain tumors, with particular attention to management of seizures, venous thromboembolism, corticosteroids and their complications, chemotherapy including bevacizumab, and fatigue, mood, and cognitive dysfunction.


Journal of Clinical Oncology | 2012

Bayesian Adaptive Randomized Trial Design for Patients With Recurrent Glioblastoma

Lorenzo Trippa; Eudocia Q. Lee; Patrick Y. Wen; Tracy T. Batchelor; Timothy F. Cloughesy; Giovanni Parmigiani; Brian M. Alexander

PURPOSE To evaluate whether the use of Bayesian adaptive randomized (AR) designs in clinical trials for glioblastoma is feasible and would allow for more efficient trials. PATIENTS AND METHODS We generated an adaptive randomization procedure that was retrospectively applied to primary patient data from four separate phase II clinical trials in patients with recurrent glioblastoma. We then compared AR designs with more conventional trial designs by using realistic hypothetical scenarios consistent with survival data reported in the literature. Our primary end point was the number of patients needed to achieve a desired statistical power. RESULTS If our phase II trials had been a single, multiarm trial using AR design, 30 fewer patients would have been needed compared with a multiarm balanced randomized (BR) design to attain the same power level. More generally, Bayesian AR trial design for patients with glioblastoma would result in trials with fewer overall patients with no loss in statistical power and in more patients being randomly assigned to effective treatment arms. For a 140-patient trial with a control arm, two ineffective arms, and one effective arm with a hazard ratio of 0.6, a median of 47 patients would be randomly assigned to the effective arm compared with 35 in a BR trial design. CONCLUSION Given the desire for control arms in phase II trials, an increasing number of experimental therapeutics, and a relatively short time for events, Bayesian AR designs are attractive for clinical trials in glioblastoma.

Collaboration


Dive into the Eudocia Q. Lee's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian M. Alexander

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge