Network


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

Hotspot


Dive into the research topics where Timothy F. Cloughesy is active.

Publication


Featured researches published by Timothy F. Cloughesy.


Annual Review of Pathology-mechanisms of Disease | 2014

Glioblastoma: From Molecular Pathology to Targeted Treatment

Timothy F. Cloughesy; Webster K. Cavenee; Paul S. Mischel

Glioblastoma (GBM) is one of the most lethal human cancers. Genomic analyses are defining the molecular architecture of GBM, uncovering relevant subsets of patients whose disease may require different treatments. Many pharmacological targets have been revealed, promising to transform patient care through targeted therapies. However, for most patients, clinical responses to targeted inhibitors are either not apparent or not durable. In this review, we address the challenge of developing more effective, molecularly guided approaches for the treatment of GBM patients. We summarize the current state of knowledge regarding molecular classifiers and examine their benefit for stratifying patients for treatment. We survey the molecular landscape of the disease, discussing the challenges raised by acquired drug resistance. Furthermore, we analyze the biochemical features of GBM, suggesting a next generation of drug targets, and we examine the contribution of tumor heterogeneity and its implications. We conclude with an analysis of the experimental approaches and their potential benefit to patients.


Journal of Neuro-oncology | 2012

Cilengitide in patients with recurrent glioblastoma: the results of NABTC 03-02, a phase II trial with measures of treatment delivery

Mark R. Gilbert; John G. Kuhn; Kathleen R. Lamborn; Frank S. Lieberman; Patrick Y. Wen; Minesh P. Mehta; Timothy F. Cloughesy; Andrew B. Lassman; Lisa M. DeAngelis; Susan M. Chang; Michael D. Prados

Cilengitide is a cyclic pentapeptide that is a specific inhibitor of the αvβ3 and αvβ5 integrins. Preclinical studies demonstrate antiangiogenic activity and anti-invasive activity in a number of glioma models. This study was designed to evaluate the efficacy and tumor delivery of cilengitide in patients with recurrent glioblastoma. Patients with recurrent glioblastoma who require a surgical resection for optimal clinical care received 3 intravenous doses of cilengitide at either 500 or 2000xa0mg (day -8, -4, -1) prior to undergoing tumor resection with corresponding blood samples for plasma to tumor comparisons. After recovery from surgery, patients were treated with cilengitide (2000xa0mg i.v. twice weekly, maximum of 2xa0years of treatment). The study accrued 30 patients with recurrent glioblastoma, 26 were evaluable for efficacy. The 6-month progression free survival rate was 12%. Cilengitide was detected in all tumor specimens with higher levels in the group receiving 2000xa0mg dosing while corresponding plasma concentrations were low, often below the lower limit of detection. These results confirm drug delivery and possibly retention in tumor. This study provides evidence that with established dosing, cilengitide is adequately delivered to the tumor, although as a single agent, efficacy in recurrent glioblastoma is modest. However, these results demonstrating drug delivery to tumor do support continued investigation of this agent as preliminary results from recent studies combining cilengitide with cytotoxic therapies are promising.


European Journal of Nuclear Medicine and Molecular Imaging | 2007

18F-fluorothymidine kinetics of malignant brain tumors

Christiaan Schiepers; Wei Chen; Magnus Dahlbom; Timothy F. Cloughesy; Carl K. Hoh; Sung-Cheng Huang

Purpose18F-labeled deoxy-fluorothymidine (FLT), a marker of cellular proliferation, has been used in PET tumor imaging. Here, the FLT kinetics of malignant brain tumors were investigated.MethodsSeven patients with high-grade tumors and two patients with metastases had 12 studies. After 1.5xa0MBq/kg 18F-FLT had been administered intravenously, dynamic PET studies were acquired for 75xa0min. Images were reconstructed with iterative algorithms, and corrections applied for attenuation and scatter. Parametric images were generated with factor analysis, and vascular input and tumor output functions were derived. Compartmental models were used to estimate the rate constants.ResultsThe standard three-compartment model appeared appropriate to describe 18F-FLT uptake. Corrections for blood volume, metabolites, and partial volume were necessary. Kinetic parameters were correlated with tumor pathology and clinical follow-up data. Two groups could be distinguished: lesions that were tumor predominant (TumP) and lesions that were treatment change predominant (TrcP). Both groups had a widely varying k1 (transport across the damaged BBB, range 0.02–0.2). Group TrcP had a relatively low k3 (phosphorylation rate, range 0.017–0.027), whereas k3 varied sevenfold in group TumP (range 0.015–0.11); the k3 differences were significant (pu2009<u20090.01). The fraction of transported FLT that is phosphorylated [k3/(k2+k3)] was able to separate the two groups (pu2009<u20090.001).ConclusionA three-compartment model with blood volume, metabolite, and partial volume corrections could adequately describe 18F-FLT kinetics in malignant brain tumors. Patients could be distinguished as having: (1) tumor-predominant or (2) treatment change-predominant lesions, with significantly different phosphorylation rates.


Journal of Neuro-oncology | 2007

A North American brain tumor consortium (NABTC 99-04) phase II trial of temozolomide plus thalidomide for recurrent glioblastoma multiforme

Morris D. Groves; Vinay K. Puduvalli; Susan M. Chang; Charles A. Conrad; Mark R. Gilbert; Ivo W. Tremont-Lukats; Ta Jen Liu; Pamela Peterson; David Schiff; Timothy F. Cloughesy; Patrick Y. Wen; Harry S. Greenberg; Lauren E. Abrey; Lisa M. DeAngelis; Kenneth R. Hess; Kathleen R. Lamborn; Michael D. Prados; Alfred Yung

BackgroundLaboratory and clinical data suggest that the anti-angiogenic agent, thalidomide, if combined with cytotoxic agents, may be effective against recurrent glioblastoma multiforme (GBM).ObjectivesTo determine 6-month progression-free survival (6PFS) and toxicity of temozolomide plus thalidomide in adults with recurrent GBM.Patients and methodsEligible patients had recurrent GBM after surgery, radiotherapy, and/or adjuvant chemotherapy. Temozolomide was given at 150–200xa0mg/m2/day on days 1–5 of each 28-day cycle. Thalidomide was given orally at 400xa0mg at bedtime (days 1–28) and increased to 1,200xa0mg as tolerated. Patients were evaluated with magnetic resonance imaging scans every 56xa0days. The study was designed to detect an increase of the historical 6PFS for GBM from 10 to 30%.ResultsForty-four patients were enrolled, 43 were evaluable for efficacy and safety. The study population included 15 women, 29 men; median age was 53xa0years (range 32–84); median Karnofsky performance status was 80% (range 60–100%). Thirty-six (82%) patients were chemotherapy-naïve. There were 57 reports of toxicity of grade 3 or greater. Non-fatal grade 3–4 granulocytopenia occurred in 15 patients (34%). The objective response rate was 7%. The estimated probability of being progression-free at 6xa0months with this therapy is 24% [95% confidence interval (C.I.) 12–38%]. The median time to progression is 15xa0weeks (95% C.I. 10–20xa0weeks). There was no observed correlation between serum levels of vascular endothelial growth factor, basic fibroblast growth factor, and IL-8 and the 6PFS outcome.ConclusionThis drug combination was reasonably safe, but with little indication of improvement compared to temozolomide alone.


Cancer Discovery | 2013

De-Repression of PDGFRβ Transcription Promotes Acquired Resistance to EGFR Tyrosine Kinase Inhibitors in Glioblastoma Patients

David Akhavan; Alexandra L. Pourzia; Alex A. Nourian; Kevin J. Williams; David Nathanson; Ivan Babic; Genaro R. Villa; Kazuhiro Tanaka; Ali Nael; Huijun Yang; Julie Dang; Harry V. Vinters; William H. Yong; Mitchell Flagg; Fuyuhiko Tamanoi; Takashi Sasayama; C. David James; Harley I. Kornblum; Timothy F. Cloughesy; Webster K. Cavenee; Steven J. Bensinger; Paul S. Mischel

UNLABELLEDnAcquired resistance to tyrosine kinase inhibitors (TKI) represents a major challenge for personalized cancer therapy. Multiple genetic mechanisms of acquired TKI resistance have been identified in several types of human cancer. However, the possibility that cancer cells may also evade treatment by co-opting physiologically regulated receptors has not been addressed. Here, we show the first example of this alternate mechanism in brain tumors by showing that EGF receptor (EGFR)-mutant glioblastomas (GBMs) evade EGFR TKIs by transcriptionally de-repressing platelet-derived growth factor receptor β (PDGFRβ). Mechanistic studies show that EGFRvIII signaling actively suppresses PDGFRβ transcription in an mTORC1- and extracellular signal-regulated kinase-dependent manner. Genetic or pharmacologic inhibition of oncogenic EGFR renders GBMs dependent on the consequently de-repressed PDGFRβ signaling for growth and survival. Importantly, combined inhibition of EGFR and PDGFRβ signaling potently suppresses tumor growth in vivo. These data identify a novel, nongenetic TKI resistance mechanism in brain tumors and provide compelling rationale for combination therapy.nnnSIGNIFICANCEnThese results provide the fi rst clinical and biologic evidence for receptor tyrosinekinase (RTK) switching as a mechanism of resistance to EGFR inhibitors in GBM and provide a molecular explanation of how tumors can become addicted to a non amplified, nonmutated, physiologically regulated RTK to evade targeted treatment.


Neuro-oncology | 2014

Report of the Jumpstarting Brain Tumor Drug Development Coalition and FDA clinical trials neuroimaging endpoint workshop (January 30, 2014, Bethesda MD)

Patrick Y. Wen; Timothy F. Cloughesy; Benjamin M. Ellingson; David A. Reardon; Howard A. Fine; Lauren E. Abrey; Karla V. Ballman; Martin Bendszuz; Jan C. Buckner; Susan M. Chang; Michael D. Prados; W.B. Pope; Alma Gregory Sorensen; Martin van den Bent; Wai Kwan A.lfred Yung

On January 30, 2014, a workshop was held on neuroimaging endpoints in high-grade glioma. This workshop was sponsored by the Jumpstarting Brain Tumor Drug Development Coalition, consisting of the National Brain Tumor Society, the Society for Neuro-Oncology, Accelerate Brain Cancer Cure, and the Musella Foundation for Research and Information, and conducted in collaboration with the Food and Drug Administration. The workshop included neuro-oncologists, neuroradiologists, radiation oncologists, neurosurgeons, biostatisticians, patient advocates, and representatives from industry, clinical research organizations, and the National Cancer Institute. This report summarizes the presentations and discussions of that workshop and the proposals that emerged to improve the Response Assessment in Neuro-Oncology (RANO) criteria and standardize neuroimaging parameters.


Clinical Cancer Research | 2013

The mTOR Kinase Inhibitors, CC214-1 and CC214-2, Preferentially Block the Growth of EGFRvIII-Activated Glioblastomas

Beatrice Gini; Ciro Zanca; Deliang Guo; Tomoo Matsutani; Kenta Masui; Shiro Ikegami; Huijun Yang; David Nathanson; Genaro R. Villa; David Shackelford; Shaojun Zhu; Kazuhiro Tanaka; Ivan Babic; David Akhavan; Kelly Y. Lin; Alvaro Assuncao; Yuchao Gu; Bruno Bonetti; Deborah Mortensen; Shuichan Xu; Heather Raymon; Webster K. Cavenee; Frank Furnari; C. David James; Guido Kroemer; James R. Heath; Kristen Hege; Rajesh Chopra; Timothy F. Cloughesy; Paul S. Mischel

Purpose: mTOR pathway hyperactivation occurs in approximately 90% of glioblastomas, but the allosteric mTOR inhibitor rapamycin has failed in the clinic. Here, we examine the efficacy of the newly discovered ATP-competitive mTOR kinase inhibitors CC214-1 and CC214-2 in glioblastoma, identifying molecular determinants of response and mechanisms of resistance, and develop a pharmacologic strategy to overcome it. Experimental Design: We conducted in vitro and in vivo studies in glioblastoma cell lines and an intracranial model to: determine the potential efficacy of the recently reported mTOR kinase inhibitors CC214-1 (in vitro use) and CC214-2 (in vivo use) at inhibiting rapamycin-resistant signaling and blocking glioblastoma growth and a novel single-cell technology—DNA Encoded Antibody Libraries—was used to identify mechanisms of resistance. Results: Here, we show that CC214-1 and CC214-2 suppress rapamycin-resistant mTORC1 signaling, block mTORC2 signaling, and significantly inhibit the growth of glioblastomas in vitro and in vivo. EGFRvIII expression and PTEN loss enhance sensitivity to CC214 compounds, consistent with enhanced efficacy in strongly mTOR-activated tumors. Importantly, CC214 compounds potently induce autophagy, preventing tumor cell death. Genetic or pharmacologic inhibition of autophagy greatly sensitizes glioblastoma cells and orthotopic xenografts to CC214-1- and CC214-2–induced cell death. Conclusions: These results identify CC214-1 and CC214-2 as potentially efficacious mTOR kinase inhibitors in glioblastoma, and suggest a strategy for identifying patients most likely to benefit from mTOR inhibition. In addition, this study also shows a central role for autophagy in preventing mTOR-kinase inhibitor-mediated tumor cell death, and suggests a pharmacologic strategy for overcoming it. Clin Cancer Res; 19(20); 5722–32. ©2013 AACR.


Neuro-oncology | 2002

A phase II trial of thymidine and carboplatin for recurrent malignant glioma: A north American brain tumor consortium study

H. Ian Robins; Susan M. Chang; Michael D. Prados; W. K. Alfred Yung; Kenneth R. Hess; David Schiff; Harry S. Greenberg; Karen Fink; Kelly Nicolas; John G. Kuhn; Timothy F. Cloughesy; Larry Junck; Minesh P. Mehta

A phase II study of irinotecan (CPT-11) was conducted at Duke University Medical Center, Durham, NC, to evaluate the activity of this agent in children with high-risk malignant brain tumors. A total of 22 children were enrolled in this study, including 13 with histologically verified recurrent malignant brain tumors (glioblastoma multiforme [GBM] 4, anaplastic astrocytoma 1, ependymoma 5, and medulloblastoma/primitive neuroectodermal tumor 3), 5 with recurrent diffuse pontine glioma, and 4 with newly diagnosed GBM. All patients with recurrent tumor had prior chemotherapy and/or irradiation. Each course of CPT-11 consisted of 125 mg/m ( 2 ) per week given i.v. for 4 weeks followed by a 2-week rest period. Patients with recurrent tumors received therapy until disease progression or unacceptable toxicity. Patients with newly diagnosed tumors initially received 3 cycles of treatment to assess tumor response and then were allowed radiotherapy at physicians choice; patients who demonstrated a response to CPT-11 prior to radiotherapy were allowed to continue the drug after radiation until disease progression or unacceptable toxicity. A 25% to 50% dose reduction was made for grade III-IV toxicity. Responses were assessed after every course by gadolinium-enhanced MRI of the brain and spine. Twenty-two patients received a median of 2 courses of CPT-11 (range, 1-16). Responses were seen in 4 of 9 patients with GBM or anaplastic astrocytoma (44%; 95% confidence interval, 11%-82%) (complete response in 2 patients with recurrent GBM lasting 9 months and 48+ months; partial response in one patient with a newly diagnosed midbrain GBM lasting 18 months prior to radiotherapy; and partial response lasting 11 months in 1 patient with recurrent anaplastic astrocytoma), 1 of 5 patients with recurrent ependymoma (partial response initially followed by stable disease lasting 11 months), and none of 5 patients with recurrent diffuse pontine glioma. Two of 3 patients with medulloblastoma/primitive neuroectodermal tumor had stable disease for 9 and 13 months. Toxicity was mainly myelosuppression, with 12 of 22 patients (50%) suffering grade II-IV neutropenia. Seven patients required dose reduction secondary to neutropenia. CPT-11, given in this schedule, appears to be active in children with malignant glioma, medulloblastoma, and ependymoma with acceptable toxicity. Ongoing studies will demonstrate if activity of CPT-11 can be enhanced when combined with alkylating agents, including carmustine and temozolomide.


Journal of Neuro-oncology | 2012

A dose escalation trial for the combination of erlotinib and sirolimus for recurrent malignant gliomas

Phioanh L. Nghiemphu; Albert Lai; Richard M. Green; David A. Reardon; Timothy F. Cloughesy

In order to achieve higher dosages than previously used in clinical trials, we conducted a phase I trial to determine the maximum tolerated dose (MTD) for the combination of erlotinib and sirolimus for the treatments of recurrent malignant gliomas. Patients with pathologically proven World Health Organization (WHO) grade III glioma and grade IV glioblastoma and radiographically proven tumor recurrence were eligible for this study. Treatments included once daily erlotinib, which was given alone for the first 7xa0days of treatments, then in combination with once daily sirolimus. Sirolimus was given with a loading dose on dayxa08 followed by a maintenance dose starting on dayxa09. Dose-limiting toxicity (DLT) was determined over the first 28xa0days of treatments, and the MTD was determined in a 3xa0+xa03 classic study design. 19 patients were enrolled, and 13 patients were eligible for MTD determination. The MTD was determined to be 150xa0mg daily for erlotinib and 5xa0mg daily (after a 15xa0mg loading dose) for sirolimus. The DLTs included rash and mucositis (despite maximal medical managements), hypophosphatemia, altered mental status, and neutropenia. The combination of erlotinib and sirolimus is difficult to tolerate at dosages higher than previously reported in phase II trials.


Clinical Cancer Research | 2017

Adaptive Global Innovative Learning Environment for Glioblastoma: GBM AGILE

Brian M. Alexander; Sujuan Ba; Mitchel S. Berger; Donald A. Berry; Webster K. Cavenee; Susan M. Chang; Timothy F. Cloughesy; Tao Jiang; Mustafa Khasraw; Wenbin Li; Robert Mittman; George Poste; Patrick Y. Wen; W. K. Alfred Yung; Anna Barker

Glioblastoma (GBM) is a deadly disease with few effective therapies. Although much has been learned about the molecular characteristics of the disease, this knowledge has not been translated into clinical improvements for patients. At the same time, many new therapies are being developed. Many of these therapies have potential biomarkers to identify responders. The result is an enormous amount of testable clinical questions that must be answered efficiently. The GBM Adaptive Global Innovative Learning Environment (GBM AGILE) is a novel, multi-arm, platform trial designed to address these challenges. It is the result of the collective work of over 130 oncologists, statisticians, pathologists, neurosurgeons, imagers, and translational and basic scientists from around the world. GBM AGILE is composed of two stages. The first stage is a Bayesian adaptively randomized screening stage to identify effective therapies based on impact on overall survival compared with a common control. This stage also finds the population in which the therapy shows the most promise based on clinical indication and biomarker status. Highly effective therapies transition in an inferentially seamless manner in the identified population to a second confirmatory stage. The second stage uses fixed randomization to confirm the findings from the first stage to support registration. Therapeutic arms with biomarkers may be added to the trial over time, while others complete testing. The design of GBM AGILE enables rapid clinical testing of new therapies and biomarkers to speed highly effective therapies to clinical practice. Clin Cancer Res; 24(4); 737–43. ©2017 AACR.

Collaboration


Dive into the Timothy F. Cloughesy's collaboration.

Top Co-Authors

Avatar

Susan M. Chang

University of California

View shared research outputs
Top Co-Authors

Avatar

Patrick Y. Wen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wei Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheri Geist

University of California

View shared research outputs
Top Co-Authors

Avatar

Webster K. Cavenee

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Brian M. Alexander

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge