103 results on '"Yung, W. K."'
Search Results
2. EGFR suppresses p53 function by promoting p53 binding to DNA-PKcs: a noncanonical regulatory axis between EGFR and wild-type p53 in glioblastoma.
- Author
-
Ding, Jie, Li, Xiaolong, Khan, Sabbir, Zhang, Chen, Gao, Feng, Sen, Shayak, Wasylishen, Amanda R, Zhao, Yang, Lozano, Guillermina, Koul, Dimpy, and Yung, W K Alfred
- Published
- 2022
- Full Text
- View/download PDF
3. Report of National Brain Tumor Society roundtable workshop on innovating brain tumor clinical trials: building on lessons learned from COVID-19 experience.
- Author
-
Lee, Eudocia Q, Selig, Wendy, Meehan, Clair, Bacha, Jeffrey, Barone, Amy, Bloomquist, Erik, Chang, Susan M, Groot, John F de, Galanis, Evanthia, Hassan, Islam, Kalidas, Chitkala, Khasraw, Mustafa, Kvedar, Joseph C, Lassman, Andrew B, Puduvalli, Vinay, Sahebjam, Solmaz, Schwamm, Lee H, Tamir, Sharon, Welch, Mary, and Yung, W K Alfred
- Published
- 2021
- Full Text
- View/download PDF
4. PARP-mediated PARylation of MGMT is critical to promote repair of temozolomide-induced O6-methylguanine DNA damage in glioblastoma.
- Author
-
Wu, Shaofang, Li, Xiaolong, Gao, Feng, Groot, John F de, Koul, Dimpy, and Yung, W K Alfred
- Published
- 2021
- Full Text
- View/download PDF
5. Radiographic read paradigms and the roles of the central imaging laboratory in neuro-oncology clinical trials.
- Author
-
Ellingson, Benjamin M, Brown, Matthew S, Boxerman, Jerrold L, Gerstner, Elizabeth R, Kaufmann, Timothy J, Cole, Patricia E, Bacha, Jeffrey A, Leung, David, Barone, Amy, Colman, Howard, Bent, Martin J van den, Wen, Patrick Y, Yung, W K Alfred, Cloughesy, Timothy F, and Goldin, Jonathan G
- Published
- 2021
- Full Text
- View/download PDF
6. Integrated analysis of telomerase enzymatic activity unravels an association with cancer stemness and proliferation.
- Author
-
Noureen, Nighat, Wu, Shaofang, Lv, Yingli, Yang, Juechen, Alfred Yung, W. K., Gelfond, Jonathan, Wang, Xiaojing, Koul, Dimpy, Ludlow, Andrew, and Zheng, Siyuan
- Subjects
ENZYMATIC analysis ,CANCER cell proliferation ,TELOMERASE ,TELOMERES - Abstract
Active telomerase is essential for stem cells and most cancers to maintain telomeres. The enzymatic activity of telomerase is related but not equivalent to the expression of TERT, the catalytic subunit of the complex. Here we show that telomerase enzymatic activity can be robustly estimated from the expression of a 13-gene signature. We demonstrate the validity of the expression-based approach, named EXTEND, using cell lines, cancer samples, and non-neoplastic samples. When applied to over 9,000 tumors and single cells, we find a strong correlation between telomerase activity and cancer stemness. This correlation is largely driven by a small population of proliferating cancer cells that exhibits both high telomerase activity and cancer stemness. This study establishes a computational framework for quantifying telomerase enzymatic activity and provides new insights into the relationships among telomerase, cancer proliferation, and stemness. Telomerase activity correlates with distinct cell states, but can be challenging to quantify. Here the authors quantify telomerase activity across a range of biological samples using the expression of 13 genes, and show it correlates with cancer cell proliferation and stemness. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
7. Consensus recommendations for a dynamic susceptibility contrast MRI protocol for use in high-grade gliomas.
- Author
-
Boxerman, Jerrold L, Quarles, Chad C, Hu, Leland S, Erickson, Bradley J, Gerstner, Elizabeth R, Smits, Marion, Kaufmann, Timothy J, Barboriak, Daniel P, Huang, Raymond H, Wick, Wolfgang, Weller, Michael, Galanis, Evanthia, Kalpathy-Cramer, Jayashree, Shankar, Lalitha, Jacobs, Paula, Chung, Caroline, Bent, Martin J van den, Chang, Susan, Yung, W K Al, and Cloughesy, Timothy F
- Published
- 2020
- Full Text
- View/download PDF
8. A Phase Ib/II, open-label, multicenter study of INC280 (capmatinib) alone and in combination with buparlisib (BKM120) in adult patients with recurrent glioblastoma.
- Author
-
van den Bent, Martin, Azaro, Analia, De Vos, Filip, Sepulveda, Juan, Yung, W. K. Alfred, Wen, Patrick Y., Lassman, Andrew B., Joerger, Markus, Tabatabai, Ghazaleh, Rodon, Jordi, Tiedt, Ralph, Zhao, Sylvia, Kirsilae, Tiina, Cheng, Yi, Vicente, Sergio, Balbin, O. Alejandro, Zhang, Hefei, and Wick, Wolfgang
- Abstract
Purpose: To estimate the maximum tolerated dose (MTD) and/or identify the recommended Phase II dose (RP2D) for combined INC280 and buparlisib in patients with recurrent glioblastoma with homozygous phosphatase and tensin homolog (PTEN) deletion, mutation or protein loss. Methods: This multicenter, open-label, Phase Ib/II study included adult patients with glioblastoma with mesenchymal-epithelial transcription factor (c-Met) amplification. In Phase Ib, patients received INC280 as capsules or tablets in combination with buparlisib. In Phase II, patients received INC280 only. Response was assessed centrally using Response Assessment in Neuro-Oncology response criteria for high-grade gliomas. All adverse events (AEs) were recorded and graded. Results: 33 patients entered Phase Ib, 32 with altered PTEN. RP2D was not declared due to potential drug–drug interactions, which may have resulted in lack of efficacy; thus, Phase II, including 10 patients, was continued with INC280 monotherapy only. Best response was stable disease in 30% of patients. In the selected patient population, enrollment was halted due to limited activity with INC280 monotherapy. In Phase Ib, the most common treatment-related AEs were fatigue (36.4%), nausea (30.3%) and increased alanine aminotransferase (30.3%). MTD was identified at INC280 Tab 300 mg twice daily + buparlisib 80 mg once daily. In Phase II, the most common AEs were headache (40.0%), constipation (30.0%), fatigue (30.0%) and increased lipase (30.0%). Conclusion: The combination of INC280/buparlisib resulted in no clear activity in patients with recurrent PTEN-deficient glioblastoma. More stringent molecular selection strategies might produce better outcomes. Trial registration: NCT01870726. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
9. NRG oncology RTOG 9006: a phase III randomized trial of hyperfractionated radiotherapy (RT) and BCNU versus standard RT and BCNU for malignant glioma patients.
- Author
-
Ali, Arif N., Zhang, Peixin, Yung, W. K. Alfred, Chen, Yuhchyau, Movsas, Benjamin, Urtasun, Raul C., Jones, Christopher U., Choi, Kwang N., Michalski, Jeff M., Fischbach, A. Jennifer, Markoe, Arnold M., Schultz, Christopher J., Penas-Prado, Marta, Garg, Madhur K., Hartford, Alan C., Kim, Harold E., Won, Minhee, and Curran, Walter J.
- Abstract
From 1990 to 1994, patients with newly diagnosed malignant gliomas were enrolled and randomized between hyperfractionated radiation (HFX) of 72.0 Gy in 60 fractions given twice daily and 60.0 Gy in 30 fractions given once daily. All patients received 80 mg/m
2 of 1,3 bis(2 chloroethyl)-1 nitrosourea on days 1–3 q8 weeks for 1 year. Patients were stratified by age, KPS, and histology. The primary endpoint was overall survival (OS), with secondary endpoints including progression-free survival (PFS) and toxicity. Out of the 712 patients accrued, 694 (97.5%) were analyzable cases (350 HFX, 344 standard arm). There was no significant difference between the arms on overall acute or late treatment-related toxicity. No statistically significant effect for HFX, as compared to standard therapy, was found on either OS, with a median survival time (MST) of 11.3 versus 13.1 months (p = 0.20) or PFS, with a median PFS time of 5.7 versus 6.9 months (p = 0.18). The treatment effect on OS remained insignificant based on the multivariate analysis (hazard ratio 1.16; p = 0.0682). When OS was analyzed by histology subgroup there was also no significant difference between the two arms for patients with glioblastoma multiforme (MST: 10.3 vs. 11.2 months; p = 0.34), anaplastic astrocytoma (MST: 69.8 vs. 50.0 months; p = 0.91) or anaplastic oligodendroglioma (MST: 92.1 vs. 66.5 months; p = 0.33). Though this trial provided many invaluable secondary analyses, there was no trend or indication of a benefit to HFX radiation to 72.0 Gy in any subset of malignant glioma patients. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
10. High-pitch Dual-source Computed tomography Coupled with Sinogram-affirmed Iterative Reconstruction: Image Quality and Radiation Dose in Children.
- Author
-
Lee, J. C. Y., Tse, K. S., Lau, H. Y., Fok, W. S., Chan, K. K., Chat, W. H., Yung, W. K., and Mui, T. M.
- Published
- 2018
- Full Text
- View/download PDF
11. Phase I study of sorafenib and tipifarnib for recurrent glioblastoma: NABTC 05-02.
- Author
-
Nghiemphu, Phioanh Leia, Ebiana, Victoria Asuquo, Wen, Patrick, Gilbert, Mark, Abrey, Lauren E., Lieberman, F., DeAngelis, Lisa M., Robins, H. Ian, Yung, W. K. Alfred, Chang, Susan, Drappatz, Jan, Mehta, Minesh P., Levin, Victor A., Aldape, Kenneth, Dancey, Janet E., Wright, J. J., Prados, Michael, Kuhn, John, and Cloughesy, Timothy F.
- Abstract
Recurrent glioblastoma (GBM) has a very low 6-month progression free survival (PFS) with currently available treatments. Combination chemotherapy to target multiple cell signaling pathways is currently being investigated in order to improve prognosis for recurrent disease. The purpose of this phase I study was to determine the maximum tolerated dose (MTD) for the combination of tipifarnib and sorafenib for the treatment of recurrent GBM. Patients with pathologically proven WHO grade IV GBM and radiographically proven tumor recurrence were eligible for this study. Treatments included sorafenib at twice daily and escalating dosages of tipifarnib. Dose-limiting toxicity (DLT) was determined over the first 28-days of treatments, and the MTD was determined in a 3 + 3 study design. We enrolled 24 patients, and 21 patients completed the MTD period. The study was stopped early with no MTD determination for excessive toxicities. The last dose level reached was sorafenib at 200 mg twice a day and tipifarnib 100 mg twice a day on an alternating week schedule. The DLTs included diarrhea, lipase elevation, hypophosphatemia, and arthralgia. The combination of sorafenib and tipifarnib has excessive toxicities and full single agent dosages could not be achieved in combination. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
12. Activation of WEE1 confers resistance to PI3K inhibition in glioblastoma.
- Author
-
Shaofang Wu, Shuzhen Wang, Feng Gao, Luyuan Li, Siyuan Zheng, Alfred Yung, W. K., and Koul, Dimpy
- Published
- 2018
- Full Text
- View/download PDF
13. A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse.
- Author
-
Yung, W K A, Albright, R E, Olson, J, Fredericks, R, Fink, K, Prados, M D, Brada, M, Spence, A, Hohl, R J, Shapiro, W, Glantz, M, Greenberg, H, Selker, R G, Vick, N A, Rampling, R, Friedman, H, Phillips, P, Bruner, J, Yue, N, and Yung, W K
- Subjects
GLIOBLASTOMA multiforme ,ALKYLATING agents ,ANTINEOPLASTIC agents ,BENZAMIDE ,BRAIN tumors ,COMPARATIVE studies ,GLIOMAS ,RESEARCH methodology ,MEDICAL cooperation ,PROGNOSIS ,QUALITY of life ,QUESTIONNAIRES ,RESEARCH ,STATISTICAL sampling ,TIME ,DISEASE relapse ,EVALUATION research ,RANDOMIZED controlled trials ,DACARBAZINE ,THERAPEUTICS - Abstract
A randomized, multicentre, open-label, phase II study compared temozolomide (TMZ), an oral second-generation alkylating agent, and procarbazine (PCB) in 225 patients with glioblastoma multiforme at first relapse. Primary objectives were to determine progression-free survival (PFS) at 6 months and safety for TMZ and PCB in adult patients who failed conventional treatment. Secondary objectives were to assess overall survival and health-related quality of life (HRQL). TMZ was given orally at 200 mg/m(2)/day or 150 mg/m(2)/day (prior chemotherapy) for 5 days, repeated every 28 days. PCB was given orally at 150 mg/m(2)/day or 125 mg/m(2)/day (prior chemotherapy) for 28 days, repeated every 56 days. HRQL was assessed using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30 [+3]) and the Brain Cancer Module 20 (BCM20). The 6-month PFS rate for patients who received TMZ was 21%, which met the protocol objective. The 6-month PFS rate for those who received PCB was 8% (P = 0.008, for the comparison). Overall PFS significantly improved with TMZ, with a median PFS of 12.4 weeks in the TMZ group and 8.32 weeks in the PCB group (P = 0.0063). The 6-month overall survival rate for TMZ patients was 60% vs. 44% for PCB patients (P = 0.019). Freedom from disease progression was associated with maintenance of HRQL, regardless of treatment received. TMZ had an acceptable safety profile; most adverse events were mild or moderate in severity. [ABSTRACT FROM AUTHOR]
- Published
- 2000
- Full Text
- View/download PDF
14. Brain Malignancy Steering Committee clinical trials planning workshop: Report from the Targeted Therapies Working Group.
- Author
-
Alexander, Brian M., Galanis, Evanthia, Alfred Yung, W. K., Ballman, Karla V., Boyett, James M., Cloughesy, Timothy F., Degroot, John F., Huse, Jason T., Mann, Bhupinder, Mason, Warren, Mellinghoff, Ingo K., Mikkelsen, Tom, Mischel, Paul S., O’Neill, Brian P., Prados, Michael D., Sarkaria, Jann N., Tawab-Amiri, Abdul, Trippa, Lorenzo, Ye, Xiaobu, and Ligon, Keith L.
- Published
- 2015
- Full Text
- View/download PDF
15. Regional nerve injury after intra-arterial chemotherapy.
- Author
-
Castellanos, A. M., Glass, J. Peter, Yung, W. K. Alfred, Glass, J P, and Yung, W K
- Published
- 1987
- Full Text
- View/download PDF
16. Use of [18F]fluorodeoxyglucose positron emission tomography in patients with primary malignant brain tumors.
- Author
-
Janus, Todd J., Kim, E. Edmund, Tilbury, Roy, Bruner, Janet M., Yung, W. K. Alfred, Janus, T J, Kim, E E, Tilbury, R, Bruner, J M, and Yung, W K
- Published
- 1993
- Full Text
- View/download PDF
17. Suppression of human glioma growth by adenovirus-mediated Rb gene transfer.
- Author
-
Fueyo, J., Gomez-Manzano, C., Yung, W. K. A., Liu, T.-J., Alemany, R., Bruner, J. M., Chintala, S. K., Rao, J. S., Levin, V. A., Kyritsis, A. P., and Yung, W K
- Published
- 1998
- Full Text
- View/download PDF
18. Brain Metastasis.
- Author
-
Tonn, Jörg-Christian, Grossman, Stuart A., Rutka, James T., Westphal, Manfred, Dorai, Zeena, Sawaya, Raymond, and Alfred Yung, W. K.
- Published
- 2006
- Full Text
- View/download PDF
19. Planned conservative management of placenta accreta - experience of a regional general hospital.
- Author
-
Lo, Tsz Kin, Yung, W. K., Lau, W. L., Law, Bassanio, Lau, Samuel, and Leung, W. C.
- Subjects
PLACENTA ,HOSPITALS ,WOMEN'S health ,THERAPEUTIC embolization ,UTERUS - Abstract
Objective: There are only a few series treating ≥10 cases of accreta conservatively, all from university teaching hospitals, with reported success rate of 60-85%. We reported the first series of accreta managed by planned uterine conservation in the setting of non-university district general hospital. Methods: Women with placenta previa overlying previous cesarean scar who desired uterine conservation were included. For cases with accreta confirmed during cesarean delivery, placenta was purposefully left behind, followed immediately by uterine artery embolization. Cases were followed in our special postnatal clinic. Charts were reviewed to retrieve clinical details. Results: Among 15 cases of placenta previa overlying cesarean scar opting for conservative management, 12 (80%) were confirmed to be accreta intra-operatively. They had 20-100% of the adherent placentae retained (median 90%) and their uterus preserved. Postpartum, abnormal vaginal bleeding and/or infection led to unscheduled readmission in 67% (8/12), all managed conservatively. Sonographic resolution of placenta took 2-13 months (median 6.6), and was later than menstrual return in 11 cases. Conclusions: Successful planned conservative management of placenta accreta is feasible in the setting of district general hospital with facilities for interventional radiology. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. A High Notch Pathway Activation Predicts Response to γ Secretase Inhibitors in Proneural Subtype of Glioma Tumor-Initiating Cells.
- Author
-
Saito, Norihiko, Fu, Jun, Zheng, Siyuan, Yao, Jun, Wang, Shuzhen, Liu, Diane D., Yuan, Ying, Sulman, Erik P., Lang, Frederick F., Colman, Howard, Verhaak, Roel G., Yung, W. K. Alfred, and Koul, Dimpy
- Subjects
SECRETASE inhibitors ,GLIOMAS ,PROTEOMICS ,GENETIC transcription ,BRAIN tumors ,CELLULAR signal transduction - Abstract
Genomic, transcriptional, and proteomic analyses of brain tumors reveal subtypes that differ in pathway activity, progression, and response to therapy. However, a number of small molecule inhibitors under development vary in strength of subset and pathway-specificity, with molecularly targeted experimental agents tending toward stronger specificity. The Notch signaling pathway is an evolutionarily conserved pathway that plays an important role in multiple cellular and developmental processes. We investigated the effects of Notch pathway inhibition in glioma tumor-initiating cell (GIC, hereafter GIC) populations using γ secretase inhibitors. Drug cytotoxicity testing of 16 GICs showed differential growth responses to the inhibitors, stratifying GICs into responders and nonresponders. Responder GICs had an enriched proneural gene signature in comparison to nonresponders. Also gene set enrichment analysis revealed 17 genes set representing active Notch signaling components NOTCH1, NOTCH3, HES1, MAML1, DLL-3, JAG2, and so on, enriched in responder group. Analysis of The Cancer Genome Atlas expression dataset identified a group (43.9%) of tumors with proneural signature showing high Notch pathway activation suggesting γ secretase inhibitors might be of potential value to treat that particular group of proneural glioblastoma (GBM). Inhibition of Notch pathway by γ secretase inhibitor treatment attenuated proliferation and self-renewal of responder GICs and induces both neuronal and astrocytic differentiation. In vivo evaluation demonstrated prolongation of median survival in an intracranial mouse model. Our results suggest that proneural GBM characterized by high Notch pathway activation may exhibit greater sensitivity to γ secretase inhibitor treatment, holding a promise to improve the efficiency of current glioma therapy. S tem C ells 2014;32:301-312 [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
21. IDH1/2 mutations target a key hallmark of cancer by deregulating cellular metabolism in glioma.
- Author
-
Zhang, Chunzhi, Moore, Lynette M., Li, Xia, Yung, W. K. Alfred, and Zhang, Wei
- Published
- 2013
- Full Text
- View/download PDF
22. Phase 1/1b study of lonafarnib and temozolomide in patients with recurrent or temozolomide refractory glioblastoma.
- Author
-
Yust‐Katz, Shlomit, Liu, Diane, Yuan, Ying, Liu, Vivien, Kang, Sanghee, Groves, Morris, Puduvalli, Vinay, Levin, Victor, Conrad, Charles, Colman, Howard, Hsu, Sigmonid, Yung, W. K. Alfred, and Gilbert, Mark R.
- Subjects
PHARMACEUTICAL research ,FARNESYLTRANSFERASE ,GLIOBLASTOMA multiforme treatment ,CANCER chemotherapy ,CANCER relapse - Abstract
BACKGROUND Lonafarnib is an oral selective farnesyltransferase inhibitor, a class of drugs which have shown activity in preclinical glioma models. Temozolomide (TMZ) is an alkylating agent that is the first-line chemotherapy for glioblastoma. METHODS The current study combined the cytotoxic agent TMZ with the cytostatic agent lonafarnib for patients with recurrent glioblastoma to establish a maximum tolerated dose (MTD) of the combination and its preliminary efficacy. Three dose cohorts of lonafarnib were studied in the phase 1 component of the trial (100 mg twice daily [bid], 150 mg bid, and 200 bid) with dose-dense schedule of TMZ (150 mg/m
2 daily) administered in an alternating weekly schedule. After establishing the MTD of lonafarnib, a subsequent expansion phase 1b was undertaken to evaluate efficacy, primarily measured by 6-month progression-free survival (PFS-6). RESULTS Fifteen patients were enrolled into the phase 1 component and 20 patients into the phase 1b component. The MTD of lonafarnib in combination with TMZ was 200 mg bid. Among the patients enrolled into the study, 34 were eligible for 6-month progression evaluation and 35 patients were evaluable for time-to-progression analysis. The PFS-6 rate was 38% (95% confidence interval [CI] = 22%, 56%) and the median PFS was 3.9 months (95% CI = 2.5, 8.4). The median disease-specific survival was 13.7 months (95% CI = 8.9, 22.1). Hematologic toxicities, particularly lymphopenia, were the most common grade 3 and 4 adverse events. There were no treatment-related deaths. CONCLUSIONS These results demonstrate that TMZ can be safely combined with a farnesyltransferase inhibitor and that this regimen is active, although the current study cannot determine the relative contributions of the 2 agents or the contribution of the novel administration schedule. Cancer 2013;119:2747-2753. © 2013 American Cancer Society. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
23. Phase I/II study of sorafenib in combination with temsirolimus for recurrent glioblastoma or gliosarcoma: North American Brain Tumor Consortium study 05-02.
- Author
-
Lee, Eudocia Q., Kuhn, John, Lamborn, Kathleen R., Abrey, Lauren, DeAngelis, Lisa M., Lieberman, Frank, Robins, H. Ian, Chang, Susan M., Yung, W. K. Alfred, Drappatz, Jan, Mehta, Minesh P., Levin, Victor A., Aldape, Kenneth, Dancey, Janet E., Wright, John J., Prados, Michael D., Cloughesy, Timothy F., Gilbert, Mark R., and Wen, Patrick Y.
- Published
- 2012
- Full Text
- View/download PDF
24. Moving toward molecular classification of diffuse gliomas in adults.
- Author
-
Theeler, Brett J., Alfred Yung, W. K., Fuller, Gregory N., and De Groot, John F.
- Published
- 2012
- Full Text
- View/download PDF
25. Identification of Common Prognostic Gene Expression Signatures with Biological Meanings from Microarray Gene Expression Datasets.
- Author
-
Jun Yao, Qi Zhao, Ying Yuan, Xiaoming Liu, Yung, W. K. Alfred, Weinstein, John N., Li Zhang, and Hoshida, Yujin
- Subjects
GENE expression ,ALGORITHM research ,PROPORTIONAL hazards models ,BREAST cancer research ,SEX chromosomes ,LUNG tumors - Abstract
Numerous prognostic gene expression signatures for breast cancer were generated previously with few overlap and limited insight into the biology of the disease. Here we introduce a novel algorithm named SCoR (Survival analysis using Cox proportional hazard regression and Random resampling) to apply random resampling and clustering methods in identifying gene features correlated with time to event data. This is shown to reduce overfitting noises involved in microarray data analysis and discover functional gene sets linked to patient survival. SCoR independently identified a common poor prognostic signature composed of cell proliferation genes from six out of eight breast cancer datasets. Furthermore, a sequential SCoR analysis on highly proliferative breast cancers repeatedly identified T/B cell markers as favorable prognosis factors. In glioblastoma, SCoR identified a common good prognostic signature of chromosome 10 genes from two gene expression datasets (TCGA and REMBRANDT), recapitulating the fact that loss of one copy of chromosome 10 (which harbors the tumor suppressor PTEN) is linked to poor survival in glioblastoma patients. SCoR also identified prognostic genes on sex chromosomes in lung adenocarcinomas, suggesting patient gender might be used to predict outcome in this disease. These results demonstrate the power of SCoR to identify common and biologically meaningful prognostic gene expression signatures. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
26. Phase 2 trial of irinotecan and thalidomide in adults with recurrent anaplastic glioma.
- Author
-
Giglio, Pierre, Dhamne, Megha, Hess, Kenneth R., Gilbert, Mark R., Groves, Morris D., Levin, Victor A., Kang, Sanghee L., Ictech, Sandra E., Liu, Vivien, Colman, Howard, Conrad, Charles A., Loghin, Monica, de Groot, John, Yung, W. K. Alfred, and Puduvalli, Vinay K.
- Subjects
CLINICAL trials ,IRINOTECAN ,THALIDOMIDE ,CANCER relapse ,GLIOMA treatment ,COMPARATIVE studies ,CANCER invasiveness - Abstract
BACKGROUND: Therapeutic options for patients with anaplastic gliomas (AGs) are limited despite better insights into glioma biology. The authors previously reported improved outcome in patients with recurrent glioblastoma treated with thalidomide and irinotecan compared with historical controls. Here, results of the AG arm of the study are reported, using this drug combination. METHODS: Adults with recurrent AG previously treated with radiation therapy, with Karnofsky performance score ≥70, adequate organ function and not on enzyme-inducing anticonvulsants were enrolled. Treatment was in 6-week cycles with irinotecan at 125 mg/m
2 weekly for 4 weeks followed by 2 weeks off, and thalidomide at 100 mg daily increased to 400 mg/day as tolerated. The primary endpoint was progression-free survival rate at 6 months (PFS-6), and the secondary endpoints were overall survival (OS) and response rate (RR). RESULTS: In 39 eligible patients, PFS-6 for the intent-to-treat population was 36% (95% confidence interval [CI] = 21%, 53%), median PFS was 13 weeks (95% CI = 6%, 28%) and RR was 10%(95% CI = 3%, 24%). Radiological findings included 2 complete and 2 partial responses and 17 stable disease. Median OS from study registration was 62 weeks, (95% CI = 51, 144). Treatment-related toxicities (grade 3 or higher) included neutropenia, diarrhea, nausea, and fatigue; 6 patients experienced venous thromboembolism. Four deaths were attributable to treatment-related toxicities: 1 from pulmonary embolism, 2 from colitis, and 1 from urosepsis. CONCLUSIONS: The combination of thalidomide and irinotecan did not achieve sufficient efficacy to warrant further investigation against AG, although a subset of patients experienced prolonged PFS/OS. A trial of the more potent thalidomide analogue, lenalidomide, in combination with irinotecan against AG is currently ongoing. Cancer 2012;3599-3606. © 2011 American Cancer Society. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
27. Is surgery at progression a prognostic marker for improved 6-month progression-free survival or overall survival for patients with recurrent glioblastoma?
- Author
-
Clarke†, Jennifer L., Ennis†, Michele M., Yung, W. K. Alfred, Chang, Susan M., Wen, Patrick Y., Cloughesy, Timothy F., DeAngelis, Lisa M., Robins, H. Ian, Lieberman, Frank S., Fine, Howard A., Abrey, Lauren, Gilbert, Mark R., Mehta, Minesh, Kuhn, John G., Aldape, Kenneth D., Lamborn, Kathleen R., and Prados, Michael D.
- Published
- 2011
28. Identification of novel synergistic targets for rational drug combinations with PI3 kinase inhibitors using siRNA synthetic lethality screening against GBM.
- Author
-
Kim, Yong-Wan, Liu, Ta Jen, Koul, Dimpy, Tiao, Ningyi, Feroze, Abdullah H., Wang, Jing, Powis, Garth, and Yung, W. K. Alfred
- Published
- 2011
- Full Text
- View/download PDF
29. Advances in Translational Research in Neuro-oncology.
- Author
-
Juan Fueyo, Gomez-Manzano, Candelaria, and Yung, W. K. Alfred
- Abstract
During the last decade, we have witnessed several key advances in the field of neurooncology. First, there were conceptual advances in the molecular and cell biology of malignant gliomas including the discovery in 2004 of brain tumor stem cells. Second, the Cancer Genome Atlas project has been extremely useful in the discovery of new molecular markers, including mutations in the IDH1 gene, and has led to a new classification of gliomas based on the differentiation status and mesenchymal transformation. In addition, use of the 1p/19q marker and O
6 -methylguanine-DNA methyltransferase methylation status have been identified as guides for patient selection for therapies and represent the first steps toward personalized medicine for treating gliomas. Finally, progress has been made in treatment strategies including the establishment of temozolomide as the criterion standard for treating gliomas, the adoption of bevacizumab in the clinical setting, and developments in experimental biological therapies including cancer vaccines and oncolytic adenoviruses. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
30. Combined action of the dinuclear platinum compound BBR3610 with the PI3-K inhibitor PX-866 in glioblastoma.
- Author
-
Gwak, Ho-Shin, Shingu, Takashi, Chumbalkar, Vaibhav, Hwang, Yeo-Hyeon, DeJournett, Robert, Latha, Khatri, Koul, Dimpy, Alfred Yung, W. K., Powis, Garth, Farrell, Nicholas P., and Bögler, Oliver
- Abstract
Polynuclear platinum compounds are more effective at killing glioblastoma cells than cisplatin, work by a different mechanism, and typically do not induce high levels of apoptosis at early time points after exposure. Here, we tested the hypothesis that combining BBR3610, the most potent polynuclear platinum, with a phosphoinositide-3-kinase (PI3K) inhibitor would promote apoptosis and enhance the impact on glioblastoma cells. The PI3K pathway is commonly activated in glioblastoma and promotes tumor cell survival, suggesting that its inhibition would make cells more sensitive to cytotoxic agents. We chose PX-866 as a PI3K inhibitor as it is a clinically promising agent being evaluated for brain tumor therapy. Combining BBR3610 and PX-866 resulted in synergistic killing of cultured glioma cells and an extension of survival in an orthotopic xenograft animal model. Both agents alone induced autophagy, and this appeared to be saturated, because when they were combined no additional autophagy was observed. However, the combination of PX-866 and BBR3610 did induce statistically significant increases in the level of apoptosis, associated with a reduction in pAkt and pBad, as well as inhibition of transwell migration. We conclude that combining polynuclear platinums with PI3K inhibitors has translational potential and alters the cellular response to include early apoptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
31. AMPK/TSC2/mTOR-signaling intermediates are not necessary for LKB1-mediated nuclear retention of PTEN tumor suppressor.
- Author
-
Liu, Juinn-Lin, Mao, Zhenyu, Gallick, Gary E., and Yung, W. K. Alfred
- Published
- 2011
- Full Text
- View/download PDF
32. Corticosteroid Use in Patients with Glioblastoma at First or Second Relapse Treated with Bevacizumab in the BRAIN Study.
- Author
-
VREDENBURGH, JAMES J., CLOUGHESY, TIMOTHY, SAMANT, MEGHNA, PRADOS, MICHAEL, WEN, PATRICK Y., MIKKELSEN, TOM, SCHIFF, DAVID, ABREY, LAUREN E., YUNG, W. K. ALFRED, PALEOLOGOS, NINA, NICHOLAS, MARTIN K., JENSEN, RANDY, DAS, ASHA, and FRIEDMAN, HENRY S.
- Subjects
CORTICOSTEROIDS ,HORMONE therapy ,ANTINEOPLASTIC agents ,ENZYME inhibitors ,BRAIN tumors ,CANCER relapse ,EPIDERMAL growth factor ,MONOCLONAL antibodies ,HEALTH outcome assessment ,RANDOMIZED controlled trials ,TREATMENT effectiveness - Abstract
Background. Vascular endothelial growth factor inhibitors have corticosteroid-sparing effects in patients with high-grade gliomas. We assessed corticosteroid use in patients with recurrent glioblastoma treated with bevacizumab (BEV) in the BRAIN study (J Clin Oncol 2009;27:4733- 4740). Methods. BRAIN was a phase II, multicenter, randomized, noncomparative trial of BEV alone (n=85) or in combination with irinotecan (CPT-11) (n =82) in adults with recurrent glioblastoma. Median corticosteroid dose for patients who used corticosteroids at baseline was summarized by treatment arm; the percentage of patients who had sustained (≥50% corticosteroid dose reduction for≥50% of time on study drug) or complete (discontinuation of corticosteroid for ≥25% of time on study drug) reduction in corticosteroid dose overall and by objective response and progression-free survival was calculated. The incidence of corticosteroid-related adverse events was summarized. Results. In each treatment group, 50% of patients were using systemic corticosteroids at baseline. The majority of those experienced a reduction in dose while receiving BEV-based therapy. Thirteen (30.2%) BEV and 20 (46.5%) BEV + CPT-11 patients had a sustained reduction of corticosteroid dose; 7 (16.3%) BEV and 9 (20.9%) BEV + CPT-11 patients had a complete reduction of corticosteroid dose. The majority of patients who had an objective response or progression-free survival >6 months experienced corticosteroid dose reduction. Approximately 64% of patients who used corticosteroids while receiving BEV-based therapy experienced infection. Conclusion. BEV may have corticosteroid-sparing effects in patients with recurrent glioblastoma. Corticosteroid reduction may positively affect patient health-related quality of life. Given the exploratory nature of the analyses in a noncomparative study, these results should be interpreted cautiously. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
33. Cellular and in vivo activity of a novel PI3K inhibitor, PX-866, against human glioblastoma.
- Author
-
Koul, Dimpy, Shen, Ruijun, Kim, Yong-Wan, Kondo, Yasuko, Lu, Yiling, Bankson, Jim, Ronen, Sabrina M., Kirkpatrick, D. Lynn, Powis, Garth, and Yung, W. K. Alfred
- Published
- 2010
- Full Text
- View/download PDF
34. Biomarkers of disease: cerebrospinal fluid vascular endothelial growth factor (VEGF) and stromal cell derived factor (SDF)-1 levels in patients with neoplastic meningitis (NM) due to breast cancer, lung cancer and melanoma.
- Author
-
Groves, Morris D., Hess, Kenneth R., Puduvalli, Vinay K., Colman, Howard, Conrad, Charles A., Gilbert, Mark R., Weinberg, Jeffrey, Cristofanilli, Massimo, Yung, W. K. Alfred, and Ta-Jen Liu
- Abstract
Background Breast cancer, lung cancer and melanoma metastasize to the meninges in 5–15% of patients. The identification of specific biomarkers of disease may allow for earlier diagnosis and treatment. Preclinical evidence suggests the possible relevance of SDF-1 and VEGF in the homing and neoangiogenesis of metastases. We chose to measure these molecules in the cerebrospinal fluid (CSF) of melanoma, breast, and lung cancer patients being evaluated for neoplastic meningitis (NM). Materials and Methods We collected CSF from patients with these cancers who were being evaluated for possible NM. CSF was assayed for SDF-1 and VEGF levels using Enzyme-linked Immunosorbent Assay (ELISA) assays. Results CSF samples from 89 patients met criteria for analysis, including 41 with breast cancer, 35 with lung cancer and 13 with melanoma. Twenty-five percent (22/89) of all samples were positive for malignant cells; 8/41 (20%) from breast cancer, 10/35 (29%) from lung cancer and 4/13 (31%) from melanoma. CSF VEGF levels were available from 83 patients, and were elevated (>20 pg/ml) in 15/22 (68%) of patients with positive CSF cytology and normal (<20 pg/ml) in 59/61 (97%) of patients with negative CSF cytology. The two patients with negative CSF cytology who also had elevated CSF VEGF levels had MRI evidence of NM. CSF SDF-1 levels were available from 81 patients, and were elevated (>950 pg/ml) in 11/18 (61%) of patients with positive CSF cytology and normal (<950 pg/ml) in 57/63 (90%) of patients with negative CSF cytology. Conclusions Elevated CSF levels of VEGF are sensitive and highly specific for the diagnosis of NM from breast cancer, lung cancer and melanoma, and may serve as a useful biomarker of NM in high risk patients. CSF SDF-1 levels add little to the diagnostic information provided by CSF VEGF. Evaluation of CSF VEGF levels as a trigger for early treatment in high risk breast cancer, lung cancer and melanoma patients at risk for NM, is warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
35. Two phase II trials of temozolomide with interferon-alpha2b (pegylated and non-pegylated) in patients with recurrent glioblastoma multiforme.
- Author
-
Groves, M. D., Puduvalli, V. K., Gilbert, M. R., Levin, V. A., Conrad, C. A., Liu, V. H., Hunter, K., Meyers, C., Hess, K. R., and Alfred Yung, W. K.
- Subjects
GLIOBLASTOMA multiforme ,THERAPEUTIC use of interferons ,CLINICAL drug trials ,DRUG efficacy ,THROMBOCYTOPENIA ,LEUCOPENIA ,PATIENTS ,ANTINEOPLASTIC agents ,BRAIN tumors ,CANCER relapse ,CLINICAL trials ,COMPARATIVE studies ,GLIOMAS ,INTERFERONS ,RESEARCH methodology ,MEDICAL cooperation ,POLYETHYLENE glycol ,PROGNOSIS ,PROTEINS ,RECOMBINANT proteins ,RESEARCH ,EVALUATION research ,TREATMENT effectiveness ,KAPLAN-Meier estimator ,KARNOFSKY Performance Status ,DACARBAZINE - Abstract
Background:Because of the poor outcomes for patients with recurrent glioblastoma multiforme (GBM), and some laboratory and clinical evidence of efficacy using interferon in GBM, we assessed the toxicity and efficacy of temozolomide (TMZ) combined with either short-acting (IFN) or long-acting (pegylated) interferon α2b (PEG) in two single-arm phase II studies, and compared the results to 6-month progression-free survival (PFS-6) data from historical controls.Methods:Two single-arm phase II studies were carried out in adults with GBM. Patients were treated with the standard regimen of TMZ (150–200 mg m
−2 per day × 5 days every month) combined with either 4 million units per m2 subcutaneously (SQ) three times weekly of IFN or 0.5 μg kg−1 SQ weekly of PEG. Physical exams and imaging evaluations were carried out every 8 weeks.Results:On the IFN study, 34 adults (74% men) were enrolled, and 29 adults (55% men) on the PEG study; median Karnofsky performance status was 80 and 90 for the IFN and PEG studies, respectively. Grade 3 or 4 toxicities were common, leucopoenia and thrombocytopoenia occurring in 35–38% and 18–21% of patients, respectively. Grade 3 or 4 fatigue occurred in 18% of patients on both studies. Lymphopoenia was infrequent. PFS-6 was 31% for 29 evaluable patients in the IFN study and 38% for 26 evaluable patients in the PEG study.Conclusion:In recurrent GBM patients, both studies of standard dose TMZ with either IFN or PEG showed improved efficacy when compared to historical controls, or reports using TMZ alone. Even though the TMZ+PEG study met criteria for further study, the results of both of these studies must be considered in light of the standard of care (TMZ plus radiotherapy) for newly diagnosed GBM, which has evolved since the inception of these studies. Despite the results of the current studies being eclipsed by the new GBM standard of care, these results can still inform the development of newer approaches for GBM, either in an earlier, upfront setting, or by extrapolation of the results and consideration of the use of PEG or IFN in conjunction with other antiglioma strategies.British Journal of Cancer (2009) 101, 615–620. doi:10.1038/sj.bjc.6605189 www.bjcancer.com [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
36. Exploratory Analysis of the Copy Number Alterations in Glioblastoma Multiforme.
- Author
-
Freire, Pablo, Vilela, Marco, Deus, Helena, Yong-Wan Kim, Koul, Dimpy, Colman, Howard, Aldape, Kenneth D., Bogler, Oliver, Alfred Yung, W. K., Coombes, Kevin, Mills, Gordon B., Vasconcelos, Ana T., and Almeida, Jonas S.
- Subjects
GLIOBLASTOMA multiforme ,GLIOMAS ,NERVOUS system tumors ,CANCER genetics ,CANCER genes ,TUMOR suppressor genes ,GENOMES ,GENETICS - Abstract
Background: The Cancer Genome Atlas project (TCGA) has initiated the analysis of multiple samples of a variety of tumor types, starting with glioblastoma multiforme. The analytical methods encompass genomic and transcriptomic information, as well as demographic and clinical data about the sample donors. The data create the opportunity for a systematic screening of the components of the molecular machinery for features that may be associated with tumor formation. The wealth of existing mechanistic information about cancer cell biology provides a natural reference for the exploratory exercise. Methodology/Principal Findings: Glioblastoma multiforme DNA copy number data was generated by The Cancer Genome Atlas project for 167 patients using 227 aCGH experiments, and was analyzed to build a catalog of aberrant regions. Genome screening was performed using an information theory approach in order to quantify aberration as a deviation from a centrality without the bias of untested assumptions about its parametric nature. A novel Cancer Genome Browser software application was developed and is made public to provide a user-friendly graphical interface in which the reported results can be reproduced. The application source code and stand alone executable are available at http://code.google. com/p/cancergenome and http://bioinformaticstation.org, respectively. Conclusions/Significance: The most important known copy number alterations for glioblastoma were correctly recovered using entropy as a measure of aberration. Additional alterations were identified in different pathways, such as cell proliferation, cell junctions and neural development. Moreover, novel candidates for oncogenes and tumor suppressors were also detected. A detailed map of aberrant regions is provided. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
37. Comprehensive genomic characterization defines human glioblastoma genes and core pathways.
- Author
-
McLendon, Roger, Friedman, Allan, Bigner, Darrell, Van Meir, Erwin G., Brat, Daniel J., Mastrogianakis, Gena M., Olson, Jeffrey J., Mikkelsen, Tom, Lehman, Norman, Aldape, Ken, Yung, W. K. Alfred, Bogler, Oliver, Weinstein, John N., Berg, Vanden, Berger, Mitchel, Prados, Michael, Muzny, Donna, Morgan, Margaret, Scherer, Steve, and Sabo, Aniko
- Subjects
GLIOBLASTOMA multiforme ,CANCER cells ,GENOMES ,METHYLATION ,GENE expression ,DNA - Abstract
Human cancer cells typically harbour multiple chromosomal aberrations, nucleotide substitutions and epigenetic modifications that drive malignant transformation. The Cancer Genome Atlas (TCGA) pilot project aims to assess the value of large-scale multi-dimensional analysis of these molecular characteristics in human cancer and to provide the data rapidly to the research community. Here we report the interim integrative analysis of DNA copy number, gene expression and DNA methylation aberrations in 206 glioblastomas—the most common type of adult brain cancer—and nucleotide sequence aberrations in 91 of the 206 glioblastomas. This analysis provides new insights into the roles of ERBB2, NF1 and TP53, uncovers frequent mutations of the phosphatidylinositol-3-OH kinase regulatory subunit gene PIK3R1, and provides a network view of the pathways altered in the development of glioblastoma. Furthermore, integration of mutation, DNA methylation and clinical treatment data reveals a link between MGMT promoter methylation and a hypermutator phenotype consequent to mismatch repair deficiency in treated glioblastomas, an observation with potential clinical implications. Together, these findings establish the feasibility and power of TCGA, demonstrating that it can rapidly expand knowledge of the molecular basis of cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
38. Phase-1 trial of gefitinib and temozolomide in patients with malignant glioma: a North American brain tumor consortium study.
- Author
-
Prados, Michael D., Yung, W. K. A., Wen, Patrick Y., Junck, Larry, Cloughesy, Timothy, Fink, Karen, Chang, Susan, Robins, H. Ian, Dancey, Janet, and Kuhn, John
- Subjects
GLIOMAS ,BRAIN tumors ,PHARMACOKINETICS ,GLIOBLASTOMA multiforme ,RADIOTHERAPY ,DRUG therapy ,PATIENTS - Abstract
This is a phase-I study of gefitinib in combination with temozolomide in patients with gliomas. The goal of the study was to define the maximum tolerated dose (MTD) and to characterize the pharmacokinetics of gefitinib when combined with temozolomide. Patients were stratified according to co-administration of enzyme-inducing anti-epileptic drugs (EIAEDs). There were 26 evaluable patients enrolled (16 on EIAEDs, 10 not on EIAEDs). All but seven patients had Glioblastoma Multiforme (GBM), and only six cases had a Karnosfsky Performance Status (KPS) of less than 80; median age was 51 years. All had received prior radiotherapy and 14 patients had no prior chemotherapy. The starting dose of temozolomide was 150 mg/m
2 /day for 5 days every 28 days and could be escalated to a maximum dose of 200 mg/m2 /day in subsequent cycles. The starting dose of gefitinib was 500 mg/day given by mouth on a continuous basis. Dose-limiting toxicity was assessed in cycle one only. For patients on EIAEDs, the MTD of gefitinib was 1,000 mg/day in combination with temozolomide. Dose-limiting toxicity (DLT) was due to diarrhea, nausea and vomiting. For patients not on EIAEDs, the MTD was 250 mg/day in combination with temozolomide. The DLT was due to increases in liver transaminases. Rash was not a significant toxicity at these dose levels. The peak concentration and AUC0-24hr at the 500 mg dose level was 1.8 and 2.5-fold lower, respectively, in the EIAED group compared to the non-EIAED group; trough levels of gefitinib increased in both groups consistent with the reported terminal half-life ranging from 27 to 51 h. The recommended phase-2 dose of gefitinib when used in combination with temozolomide is 1,000 and 250 mg/day, respectively, for patients on or not on EIAEDs. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
39. A Carbon Nanomattress: A New Nanosystem with Intrinsic, Tunable, Damping Properties.
- Author
-
Teo, E. H. T., Yung, W. K. P., Chua, D. H. C., and Tay, B. K.
- Published
- 2007
- Full Text
- View/download PDF
40. Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo.
- Author
-
Alonso, M. M., Gomez-Manzano, C., Jiang, H., Bekele, N. B., Piao, Y., Yung, W. K. A., Alemany, R., and Fueyo, J.
- Subjects
GLIOMAS ,DRUG therapy ,ADENOVIRUSES ,VIRAL proteins ,CELL lines ,CANCER treatment ,GENE therapy ,ANTINEOPLASTIC agents ,CANCER genes - Abstract
Novel therapies are clearly needed for gliomas, and the combination of oncolytic vectors with chemotherapy possesses a significant hope for the treatment of this malignancy. In addition, combination with chemotherapy allows for lower virus doses to achieve anticancer effect, thus resulting in lower undesirable toxicities due to viral proteins. In this work, we sought to determine whether combination of an oncolytic adenovirus ICOVIR-5, with RAD001 or temozolomide (TMZ) could result in enhanced anti-glioma effect in vivo. We assessed the in vitro cytotoxic effect and replication properties of ICOVIR-5 in combination with RAD001 or TMZ in U87 MG glioma cell line by MTT and TCID
50 , respectively. Our data showed that in vitro treatment with RAD001 or TMZ not only interfered with adenovirus replication but, in addition, enhanced its oncolytic properties. To evaluate the in vivo anticancer effect, athymic mice bearing glioma xenografts (5 × 105 U87 MG cells/animal) received a single intratumoral injection of ICOVIR-5 (107 PFU/animal). RAD001 was given as a regimen of 5 mg/kg 5 days per week until the end of the experiment and TMZ was administered for 5 days at 7.5 mg/kg/mice. Of significance, combination of ICOVIR-5 with RAD001 or TMZ showed a potent anti-glioma effect in vivo, resulting in a dramatic extension of the median animal survival and in 20–40% animals becoming free of disease beyond 90 days.Cancer Gene Therapy (2007) 14, 756–761; doi:10.1038/sj.cgt.7701067; published online 8 June 2007 [ABSTRACT FROM AUTHOR]- Published
- 2007
- Full Text
- View/download PDF
41. E2F1 and Telomerase.
- Author
-
Alonso, Marta M., Fueyo, Juan, Yung, W. K. Alfred, and Gomez-Manzano, Candelaria
- Published
- 2006
- Full Text
- View/download PDF
42. Expression of Transcription Factor E2F1 and Telomerase in Gliobtastomas: Mechanistic Linkage and Prognostic Significance.
- Author
-
Alonso, Marta M., Fueyo, Juan, Shay, Jerry W., Aldape, Kenneth D., Hong Jiang, Ok-Hee Lee, Johnson, David G., Jing Xu, Kondo, Yasuko, Kanzawa, Takao, Kyo, Satoru, Bekele, B. Nebiyou, Xian Zhou, Nigro, Janice, McDonald, J. Matthew, Yung, W. K. Alfred, and Gomez-Manzano, Candelaria
- Subjects
RETINOBLASTOMA ,TELOMERASE ,GLIOMAS ,TUMOR suppressor genes ,TRANSCRIPTION factors ,PROGNOSIS ,ADENOVIRUSES ,CANCER cells - Abstract
Background: Several tumor suppressor pathways have been identified as modulators of telomerase function. We examined the functional role of the retinoblastoma-E2F1 pathway in regulating telomerase activity in malignant gliomas. Methods: Adenovirus vectors were used to transfer cDNAs into human glioblastoma and sarcoma cells. Telomerase activity was assessed with a telomere repeat amplification protocol. Promoter activity in cancer cells was assessed with promoter-luciferase reporter constructs. Promoter binding was assessed with the chromatin immunoprecipitation (ChIP) assay. We isolated astrocytes from E2F1 transgenic mice and normal mice for in vivo studies. We evaluated the expression of E2F1 and hTERT (the catalytic subunit of human telomerase) mRNAs by reverse transcriptase-polymerase chain reaction and proteins in human glioblastoma samples by immunoblot analysis. Associations between survival among 61 glioblastoma multiforme patients and expression of E2F1 and hTERT mRNA and protein were examined with Kaplan-Meier analysis, the log-rank test, and Cox proportional hazards regression models. All statistical tests were two-sided. Results: Ectopic E2F1 expression increased hTERT promoter activity in cancer cells. We detected an interaction between E2F1 protein and the hTERT promoter. Transgenic E2F1 astrocytes contained functional telomerase protein. E2F1 mRNA expression and hTERT mRNA expression were statistically significantly correlated in human glioblastoma specimens (R = .81 P«.001). Longer median survival was statistically significantly associated with lower E2F1 mRNA expression in tumors (103.6 weeks) rather than with higher expression (46.1 weeks) (difference = 57.5 weeks; 95% confidence interval [CI] = 14.7 to 159.7; log-rank P = .002). E2F1 mRNA was the only factor that was statistically significantly associated with overall survival in a multivariable model (P = .04). Among 27 patients with glioblastoma multiforme samples, the expression of E2F1 protein was statistically significantly associated with survival (log-rank P«.001). Conclusions: E2F1 may participate in telomerase activity regulation in malignant glioma cells. Its expression appears to be strongly associated with the survival of patients with malignant brain tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
43. Modeling prognosis for patients with malignant astrocytic gliomas: Quantifying the expression of multiple genetic markers and clinical variables.
- Author
-
Yi-Hong Zhou, Hess, Kenneth R., Longjian Liu, Linskey, Mark. E., and Yung, W. K. Alfred
- Subjects
ASTROCYTOMAS ,GLIOBLASTOMA multiforme ,GENETICS ,POLYMERASE chain reaction ,GENE expression ,NEOVASCULARIZATION - Abstract
The disparate lengths of survival among patients with malignant astrocytic gliomas (anaplastic astrocytomas [AAs] and glioblastoma multiforme [GBM]) cannot be adequately accounted for by clinical variables (patient age, histology, and recurrent status). Using real-time quantitative reverse transcription-polymerase chain reaction, we quantified the expression of four genes that were putative prognostic markers (CDK4, IGFBP2, MMP2, and RPS9) in a set of 43 AAs, 41 GBMs, and seven adjacent normal brain tissues. We previously explicated the expression and prognostic value of PAX6, PTEN, VEGF, and EGFR in these glioma tissues and established a comprehensive prognostic model (Zhou et al., 2003). This study attempts to improve that model by including four additional genetic markers, which exhibited a differential expression (P < 0.001) among tumor grades and between tumor and normal tissues. By including eight log-scaled gene expression variables, three clinical variables, and interaction terms among the eight genes, we established a prognostic model that accounted for two thirds of the variation (R²) in survival for this set of patients. To improve the R² of the model without compromising its clinical utility, our data demonstrated that incorporating genes from different pathways markedly strengthens the model. Spearman rank correlation analysis of gene expression demonstrated a statistically significant positive correlation (P < 0.01) between the expression of IGFBP2-MMP2 and IGFBP2-VEGF in GBMs, but not in AAs. This finding suggests that the expression of IGFBP2 is associated with pathways activated specifically in GBMs that result in enhancing invasiveness and angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
44. Nuclear PTEN-Mediated Growth Suppression Is Independent of Akt Down-Regulation.
- Author
-
Juinn-Lin Liu, Xiaoyang Sheng, Hortobagyi, Zsuzsanna K., Zhenyu Mao, Gallick, Gary E., and Yung, W. K. Alfred
- Subjects
TUMOR suppressor proteins ,TUMOR suppressor genes ,TUMORS ,CELL membranes ,GENETIC mutation ,PROTEIN kinases - Abstract
The tumor suppressor gene PTEN is a phosphoinositide phosphatase that is inactivated by deletion and/or mutation in diverse human tumors. Wild-type PTEN is expressed both in the cytoplasm and nucleus in normal cells, with a preferential nuclear localization in differentiated or resting cells. To elucidate the relationship between PTEN's subcellular localization and its biologic activities, we constructed different PTEN mutants that targeted PTEN protein into different subcellular compartments. Our data show that the subcellular localization patterns of a PTEN (ΔPDZB) mutant versus a G129R phosphatase mutant were indistinguishable from those of wild-type PTEN. In contrast, the Myr-PTEN mutant demonstrated an enhanced association with the cell membrane. We found that nuclear PTEN alone is capable of suppressing anchorage-independent growth and facilitating G
1 arrest in U251MG cells without inhibiting Akt activity. Nuclear compartment-specific PTEN-induced growth suppression is dependent on possessing a functional lipid phosphatase domain. In addition, the down-regulation of p70S6K could be mediated, at least in part, through activation of AMP-activated protein kinase in an Akt-independent fashion. Introduction of a constitutively active mutant of Akt, Akt-DD, only partially rescues nuclear PTEN-mediated growth suppression. Our collective results provide the first direct evidence that PTEN can contribute to G1 growth arrest through an Akt-independent signaling pathway. [ABSTRACT FROM AUTHOR]- Published
- 2005
- Full Text
- View/download PDF
45. ?24-hyCD adenovirus suppresses glioma growth in vivo by combining oncolysis and chemosensitization.
- Author
-
Conrad, Charles, Miller, C. Ryan, Yongjie Ji, Gomez-Manzano, Candeleria, Bharara, Suman, McMurray, John S., Lang, Frederick F., Wong, Franklin, Sawaya, Raymond, Yung, W. K. Alfred, and Fueyo, Juan
- Subjects
ADENOVIRUSES ,DNA viruses ,GLIOMAS ,NERVOUS system tumors ,CANCER genetics ,CYTOKINES ,CELLULAR immunity ,CANCER cells - Abstract
Replication-competent adenoviruses could provide an efficient method for delivering therapeutic genes to tumors. The most promising strategies among adenovirus-based oncolytic systems are designed to exploit free E2F-1 activity in cancer cells, which in the absence of pRb activates transcription and regulates the expression of genes involved in differentiation, proliferation, and apoptosis. We previously developed?24, an E1A-mutant, conditionally replicative oncolytic adenovirus. Here, we examine the ability of a second-generation?24 (?24-hyCD) engineered to express a humanized form of the Saccharomyces cerevisiae cytosine deaminase gene (hyCD). Real-time quantitative PCR, Western blotting, thin-layer chromatography, and radioisotope quantitative enzymatic assays confirmed the production of a catalytically active hyCD enzyme in the setting of an oncolytic infection in vitro; other experiments assessing local production of 5-fluorouracil and a concomitant bystander effect showed improved cytotoxicity. The IC
50 dose of 5-fluorocytosine (5-FC) required for a complete cytopathic effect by the?24-hyCD virus was fivefold lower than with?24 alone in U251MG and U87MG malignant glioma (MG) cell lines. Intratumoral treatment of mice bearing intracranial U87MG xenografts with?24-hyCD+5-FC significantly improved survival, confirming that?24-hyCD with 5-FC is a more efficient anticancer tool than?24 alone. Histopathologically,?24-hyCD replication was accompanied by progressively augmented oncolysis and drug-induced necrosis. These findings demonstrate that?24-hyCD with concomitant systemic 5-FC is a significant improvement over the earlier?24 oncolytic tumor-selective strategy for therapy of experimental gliomas.Cancer Gene Therapy (2005) 12, 284-294. doi:10.1038/sj.cgt.7700750 Published online 14 January 2005 [ABSTRACT FROM AUTHOR]- Published
- 2005
- Full Text
- View/download PDF
46. Genetically modified adenoviruses against gliomas: from bench to bedside.
- Author
-
Gomez-Manzano C, Yung WKA, Alemany R, Fueyo J, Gomez-Manzano, Candelaria, Yung, W K Alfred, Alemany, Ramon, and Fueyo, Juan
- Published
- 2004
- Full Text
- View/download PDF
47. 13-cis -Retinoic acid in the treatment of recurrent glioblastoma multiforme.
- Author
-
Siew-Ju See, Levin, Victor A., Yung, W.-K. Alfred, Hess, Kenneth R., and Groves, Morris D.
- Subjects
ISOTRETINOIN ,GLIOBLASTOMA multiforme ,CANCER treatment ,CANCER patients ,MEDICAL records ,RADIOTHERAPY ,DRUG therapy ,THERAPEUTICS - Abstract
Basic science and clinical investigations have demonstrated that 13-cis-retinoic acid (cRA) has activity against alignant gliomas. To assess its effectiveness in the setting of recurrent glioblastoma multiforme (GBM), we performed a retrospective analysis of the medical records and neuroimaging results of patients with recurrent GBM who were treated with cRA. The toxicity profile of cRA, response, and effect on progression-free survival from initiation of treatment were end points of our analysis. Eighty-two of 85 patients with a median age of 51 years received at least 1 full cycle of cRA. At the initiation of cRA treatment, the median Karnofsky performance score was 80. All patients had failed conventional radiotherapy. Seven patients were chemonaive, whereas 75 patients had received some form of chemotherapy. Radiographic partial responses, minor responses, and stable disease were seen in 4%, 8%, and 34% of patients, respectively. Two patients were not assessable. Progression-free survival and overall survival after initiation of cRA were 10.0 and 24.6 weeks, respectively. Sixmonth progression-free survival was 19% for the entire group. Grade 3 or 4 toxicity developed in 14 patients (16%), one of whom developed pancreatitis and died. The results of this study demonstrate only modest efficacy for cRA therapy in this cohort of heavily pretreated patients with recurrent GBM. This data supports the use of cRA in such patients, but its further evaluation in larger, prospective, controlled studies with or without other noncytotoxic and cytotoxic agents may be warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
48. Phase II study of neoadjuvant 1, 3-bis (2-chloroethyl)-1-nitrosourea and temozolomide for newly diagnosed anaplastic glioma: a North American Brain Tumor Consortium Trial.
- Author
-
Chang, Susan M, Prados, Michael D, Yung, W K Alfred, Fine, Howard, Junck, Larry, Greenberg, Harry, Robins, H Ian, Mehta, Minesh, Fink, Karen L, Jaeckle, Kurt A, Kuhn, John, Hess, Kenneth, and Schold, Clifford
- Published
- 2004
49. A novel E1A-E1B mutant adenovirus induces glioma regression in vivo.
- Author
-
Gomez-Manzano, Candelaria, Alemany, Ramon, Lemoine, Michael G, Mitlianga, Paraskevi, Khan, Asadullah, Alonso, Marta, Ta-Jen Liu, Fueyo, Juan, Balague, Cristina, Hong Jiang, Lang, Frederick F., Conrad, Charles A., Bekele, B. Nebiyou, and Yung, W. K. Alfred
- Subjects
GLIOMAS ,BRAIN tumors ,SURGICAL therapeutics ,ADENOVIRUSES ,CLINICAL trials - Abstract
Malignant gliomas are the most frequently occurring primary brain tumors and are resistant to conventional therapy. Conditionally replicating adenoviruses are a novel strategy in glioma treatment. Clinical trials using E1B mutant adenoviruses have been reported recently and E1A mutant replication-competent adenoviruses are in advanced preclinical testing. Here we constructed a novel replication-selective adenovirus (CB1) incorporating a double deletion of a 24?bp Rb-binding region in the E1a gene, and a 903?bp deleted region in the E1b gene that abrogates the expression of a p53-binding E1B-55?kDa protein. CB1 exerted a potent anticancer effect in vitro in U-251 MG, U-373 MG, and D-54 MG human glioma cell lines, as assessed by qualitative and quantitative viability assays. Replication analyses demonstrated that CB1 replicates in vitro in human glioma cells. Importantly, CB1 acquired a highly attenuated replicative phenotype in both serum-starved and proliferating normal human astrocytes. In vivo experiments using intracranially implanted D-54 MG glioma xenografts in nude mice showed that a single dose of CB1 (1.5 × 10
8 ?PFU/tumor) significantly improved survival. Immunohistochemical analyses of expressed adenoviral proteins confirmed adenoviral replication within the tumors. The CB1 oncolytic adenovirus induces a potent antiglioma effect and could ultimately demonstrate clinical relevance and therapeutic utility.Oncogene (2004) 23, 1821-1828. doi:10.1038/sj.onc.1207321 [ABSTRACT FROM AUTHOR]- Published
- 2004
- Full Text
- View/download PDF
50. Phase 1 trial of irinotecan (CPT-11) in patients with recurrent malignant glioma: A North American Brain Tumor Consortium study.
- Author
-
Prados, Michael D., Yung, W. K. A., Jaeckle, Kurt A., Robins, H. Ian, Mehta, Minesh P., Fine, Howard A., Wen, Patrick Y., Cloughesy, Timothy F., Chang, Susan M., Nicholas, M. Kelly, Schiff, David, Greenberg, Harry S., Junck, Larry, Fink, Karen L., Hess, Kenneth R., and Kuhn, John
- Subjects
THERAPEUTICS ,PHARMACOKINETICS ,DRUGS ,GLIOMAS ,ONCOLOGY ,MEDICAL research - Abstract
This study was conducted to determine the maximum tolerated dose and dose-limiting toxicity of irinotecan (CPT-11) administered every 3 weeks to adults with progressive malignant glioma who were treated with enzymeinducing antiepileptic drug (EIAED) therapy, and to compare the pharmacokinetics with those in patients not on EIAED therapy treated at the recommended phase 2 dose for other cancers. The CPT-11 dose was 350 mg/m² i.v. every 3 weeks and remained fixed in patients not on EIAED therapy, but the dose was escalated by 50-mg/m2 increments in patients on EIAED therapy. CPT-11 and its metabolites SN-38, SN-38 glucuronide (SN-38G), and APC (7-ethyl-10[4-N-(5 aminopentanoic acid)-1-piperidine]- carbonyloxycamptothecin) were characterized in both groups. Patients on EIAEDs received 350 to 800 mg/m² of CPT-11. Dose-limiting toxicity was due to grade 3 diarrhea despite maximal doses of loperamide. The systemic levels of CPT-11, APC, SN-38G, and SN-38 were all lower in the EIAED group. There was a moderate-to- fair relationship between CPT-11 dose and the area under the curve (AUC) for CPT-11 and APC over the dosage range of 350 to 800 mg/m², but no relationship between CPT-11 dose and the AUC for SN-38 or SN-38G. At the 750-mg/m² dose, the AUC for CPT-11 (21.6 μg ⊗ h/ml) matched the AUC (21.6 μg ⊗ h/ml) in the non- EIAED group treated with 350 mg/m² of CPT-11. We conclude that the recommended phase 2 dose of CPT-11 for patients on EIAEDs is 750 mg/m² when given every 3 weeks. A phase 2 study of patients with recurrent malignant glioma is ongoing to assess the efficacy of CPT-11 when the dose is stratified according to the use of EIAEDs. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.