880 results on '"Mark R. Gilbert"'
Search Results
52. Data from Protein Kinase B (PKB/AKT) Protects IDH-Mutated Glioma from Ferroptosis via Nrf2
- Author
-
Chunzhang Yang, Terri S. Armstrong, Mark R. Gilbert, Kareem A. Zaghloul, Patrick J. Cimino, Yang Zhang, Nicole J. Briceno, Dionne L. Davis, Wei Zhang, Hua Song, Meili Zhang, Fengchao Lang, Fu-Ju Chou, and Yang Liu
- Abstract
Purpose:Mutations of the isocitrate dehydrogenase (IDH) gene are common genetic mutations in human malignancies. Increasing evidence indicates that IDH mutations play critical roles in malignant transformation and progression. However, the therapeutic options for IDH-mutated cancers remain limited. In this study, the investigation of patient cohorts revealed that the PI3K/protein kinase B (AKT) signaling pathways were enhanced in IDH-mutated cancer cells.Experimental Design:In this study, we investigated the gene expression profile in IDH-mutated cells using RNA sequencing after the depletion of AKT. Gene set enrichment analysis (GSEA) and pathway enrichment analysis were used to discover altered molecular pathways due to AKT depletion. We further investigated the therapeutic effect of the AKT inhibitor, ipatasertib (Ipa), combined with temozolomide (TMZ) in cell lines and preclinical animal models.Results:GSEA and pathway enrichment analysis indicated that the PI3K/AKT pathway significantly correlated with Nrf2-guided gene expression and ferroptosis-related pathways. Mechanistically, AKT suppresses the activity of GSK3β and stabilizes Nrf2. Moreover, inhibition of AKT activity with Ipa synergizes with the genotoxic agent TMZ, leading to overwhelming ferroptotic cell death in IDH-mutated cancer cells. The preclinical animal model confirmed that combining Ipa and TMZ treatment prolonged survival.Conclusions:Our findings highlighted AKT/Nrf2 pathways as a potential synthetic lethality target for IDH-mutated cancers.
- Published
- 2023
- Full Text
- View/download PDF
53. Supplementary Figure S4 from Protein Kinase B (PKB/AKT) Protects IDH-Mutated Glioma from Ferroptosis via Nrf2
- Author
-
Chunzhang Yang, Terri S. Armstrong, Mark R. Gilbert, Kareem A. Zaghloul, Patrick J. Cimino, Yang Zhang, Nicole J. Briceno, Dionne L. Davis, Wei Zhang, Hua Song, Meili Zhang, Fengchao Lang, Fu-Ju Chou, and Yang Liu
- Abstract
(A) Heatmap of gene expression profile for glioma specimen. The leading edge of the KEGG_MTOR_SIGNALING_PATHWAY geneset was shown. (B) Dose-response curve measured the cell viability in IDH1WT NHA cells in response to TMZ/Ipa combination treatment. (C) isobologram analysis showed the synergistic effect of TMZ and Ipa in IDH1WT NHA cells. (D) The combination index (C.I.) was calculated to show the additive effect of TMZ and Ipa.
- Published
- 2023
- Full Text
- View/download PDF
54. Interview with Dr. Mellinghoff from Differential Sensitivity of Glioma- versus Lung Cancer–Specific EGFR Mutations to EGFR Kinase Inhibitors
- Author
-
Ingo K. Mellinghoff, Timothy F. Cloughesy, Minesh P. Mehta, Paul S. Mischel, John G. Kuhn, William H. Yong, Lisa M. DeAngelis, Andrew B. Lassman, Nian Wu, Steve Horvath, Howard A. Fine, Michael D. Prados, Susan M. Chang, Kathleen R. Lamborn, Patrick Y. Wen, Jan Drappatz, David A. Reardon, Mark R. Gilbert, W. K. Alfred Yung, Frank Lieberman, Linda M. Liau, Adriana Heguy, Cameron W. Brennan, Alicia Pedraza, Julie Dang, Daisuke Kuga, Akio Iwanami, Shaojun Zhu, Hui Tao, Nicolas Yannuzzi, Milan G. Chheda, Barbara Oldrini, Sara Kubek, Phioanh Leia Nghiemphu, Christian Grommes, Carl Campos, Daniel Rohle, H. Ian Robins, and Igor Vivanco
- Abstract
mp3 file (11 MB). In the May edition of the Cancer Discovery podcast, Science Writer Elizabeth McKenna talks with Ingo K. Mellinghoff about his paper, which suggests that the disappointing clinical activity of first-generation EGFR inhibitors in glioblastoma versus lung cancer may be attributed to the different conformational requirements of mutant EGFR in these two cancer types.
- Published
- 2023
- Full Text
- View/download PDF
55. Supplementary Figures 1-10 from Differential Sensitivity of Glioma- versus Lung Cancer–Specific EGFR Mutations to EGFR Kinase Inhibitors
- Author
-
Ingo K. Mellinghoff, Timothy F. Cloughesy, Minesh P. Mehta, Paul S. Mischel, John G. Kuhn, William H. Yong, Lisa M. DeAngelis, Andrew B. Lassman, Nian Wu, Steve Horvath, Howard A. Fine, Michael D. Prados, Susan M. Chang, Kathleen R. Lamborn, Patrick Y. Wen, Jan Drappatz, David A. Reardon, Mark R. Gilbert, W. K. Alfred Yung, Frank Lieberman, Linda M. Liau, Adriana Heguy, Cameron W. Brennan, Alicia Pedraza, Julie Dang, Daisuke Kuga, Akio Iwanami, Shaojun Zhu, Hui Tao, Nicolas Yannuzzi, Milan G. Chheda, Barbara Oldrini, Sara Kubek, Phioanh Leia Nghiemphu, Christian Grommes, Carl Campos, Daniel Rohle, H. Ian Robins, and Igor Vivanco
- Abstract
PDF file - 689K
- Published
- 2023
- Full Text
- View/download PDF
56. Supplementary Tables 1-8 from Differential Sensitivity of Glioma- versus Lung Cancer–Specific EGFR Mutations to EGFR Kinase Inhibitors
- Author
-
Ingo K. Mellinghoff, Timothy F. Cloughesy, Minesh P. Mehta, Paul S. Mischel, John G. Kuhn, William H. Yong, Lisa M. DeAngelis, Andrew B. Lassman, Nian Wu, Steve Horvath, Howard A. Fine, Michael D. Prados, Susan M. Chang, Kathleen R. Lamborn, Patrick Y. Wen, Jan Drappatz, David A. Reardon, Mark R. Gilbert, W. K. Alfred Yung, Frank Lieberman, Linda M. Liau, Adriana Heguy, Cameron W. Brennan, Alicia Pedraza, Julie Dang, Daisuke Kuga, Akio Iwanami, Shaojun Zhu, Hui Tao, Nicolas Yannuzzi, Milan G. Chheda, Barbara Oldrini, Sara Kubek, Phioanh Leia Nghiemphu, Christian Grommes, Carl Campos, Daniel Rohle, H. Ian Robins, and Igor Vivanco
- Abstract
PDF file - 104K
- Published
- 2023
- Full Text
- View/download PDF
57. Supplementary Figure and Table Legends, Methods from Differential Sensitivity of Glioma- versus Lung Cancer–Specific EGFR Mutations to EGFR Kinase Inhibitors
- Author
-
Ingo K. Mellinghoff, Timothy F. Cloughesy, Minesh P. Mehta, Paul S. Mischel, John G. Kuhn, William H. Yong, Lisa M. DeAngelis, Andrew B. Lassman, Nian Wu, Steve Horvath, Howard A. Fine, Michael D. Prados, Susan M. Chang, Kathleen R. Lamborn, Patrick Y. Wen, Jan Drappatz, David A. Reardon, Mark R. Gilbert, W. K. Alfred Yung, Frank Lieberman, Linda M. Liau, Adriana Heguy, Cameron W. Brennan, Alicia Pedraza, Julie Dang, Daisuke Kuga, Akio Iwanami, Shaojun Zhu, Hui Tao, Nicolas Yannuzzi, Milan G. Chheda, Barbara Oldrini, Sara Kubek, Phioanh Leia Nghiemphu, Christian Grommes, Carl Campos, Daniel Rohle, H. Ian Robins, and Igor Vivanco
- Abstract
PDF file - 133K
- Published
- 2023
- Full Text
- View/download PDF
58. Supplementary Table S1 from Protein Kinase B (PKB/AKT) Protects IDH-Mutated Glioma from Ferroptosis via Nrf2
- Author
-
Chunzhang Yang, Terri S. Armstrong, Mark R. Gilbert, Kareem A. Zaghloul, Patrick J. Cimino, Yang Zhang, Nicole J. Briceno, Dionne L. Davis, Wei Zhang, Hua Song, Meili Zhang, Fengchao Lang, Fu-Ju Chou, and Yang Liu
- Abstract
Patient sample information
- Published
- 2023
- Full Text
- View/download PDF
59. Figure S3 from Dopamine Receptor D5 is a Modulator of Tumor Response to Dopamine Receptor D2 Antagonism
- Author
-
Joshua E. Allen, Wolfgang Oster, Patrick Y. Wen, Tracy T. Batchelor, Isabel Arrillaga-Romany, Robert Wechsler-Reya, Jessica Rusert, Mark R. Gilbert, Deric M. Park, Jinkyu Jung, Neil Charter, Sean Deacon, Rohinton S. Tarapore, Jessica Durrant, Alexander VanEngelenburg, Faye Doherty, Olivier Elemento, Wafik S. El-Deiry, Sophie Oster, C. Leah B. Kline, Coryandar Gilvary, Neel S. Madhukar, and Varun V. Prabhu
- Abstract
Figure S3
- Published
- 2023
- Full Text
- View/download PDF
60. Supplementary Data, Supplementary Tables S1-S4, Supplementary Figure S1 from Bayesian Optimal Interval Design: A Simple and Well-Performing Design for Phase I Oncology Trials
- Author
-
Mark R. Gilbert, Susan G. Hilsenbeck, Kenneth R. Hess, and Ying Yuan
- Abstract
Supplementary Data. Supplementary Table S1. Dose escalation and de-escalation thresholds for different target toxicity rates. Supplementary Table S2. Sixteen true toxicity scenarios used in the simulation study with the target DLT rate of 0.15 (left panel) and 0.3 (right panel). Supplementary Table S3. BOIN design with tighter de-escalation thresholds. Supplementary Table S4. Dose escalation and de-escalation rule for 3+3 and mTPI. Supplementary Figure S1. Sample size distribution of the 3+3 design when the true toxicity rates of 5 dose levels are 0.12, 0.2, 0.3, 0.4 and 0.5, respectively.
- Published
- 2023
- Full Text
- View/download PDF
61. Supplemental Table 1 from Genetic Modulation of Neurocognitive Function in Glioma Patients
- Author
-
Jeffrey S. Wefel, Melissa L. Bondy, Terri S. Armstrong, Mark R. Gilbert, Charles A. Conrad, Carol J. Etzel, Fu-Wen Liang, Spiridon Tsavachidis, Georgina N. Armstrong, Nicholas Boehling, Michael E. Scheurer, Erik P. Sulman, Renke Zhou, and Yanhong Liu
- Abstract
Demographic and Clinical Characteristics of Glioma Patients in the Parent Epidemiological Study (N=1247).
- Published
- 2023
- Full Text
- View/download PDF
62. Supplementary materials-Neutrophil chemotaxis videos from Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas
- Author
-
Mark R. Gilbert, John I. Gallin, Douglas B. Kuhns, William D. Figg, Katherine R. Calvo, Terri S. Armstrong, Javier Gonzales, Heather Leeper, Marta Penas-Prado, Eric M. Burton, Brett J. Theeler, Orwa Aboud, Christine Cordova, Nicole Lollo, Christine Siegel, Nancy Garren, Ramya Antony, Lisa Boris, Kelly Mentges, Ewa Grajkowska, Matthew Lindsley, Christine Bryla, Salman Ahmad, Elizabeth Vera, Tito R. Mendoza, Madison K. Butler, Guangyang Yu, Ying Pang, Yu-Ting Su, Tristan M. Sissung, Cody J. Peer, Danielle Fink, Debra A. Long Priel, Ying Yuan, and Jing Wu
- Abstract
Neutrophils chemotaxis video
- Published
- 2023
- Full Text
- View/download PDF
63. Supplementary Data from MGMT Promoter Methylation Cutoff with Safety Margin for Selecting Glioblastoma Patients into Trials Omitting Temozolomide: A Pooled Analysis of Four Clinical Trials
- Author
-
Michael Weller, Josef Straub, Wim Van Criekinge, Greg Jones, L. Burt Nabors, Olivier L. Chinot, Mark R. Gilbert, Roger Stupp, Thierry Gorlia, Els Genbrugge, and Monika E. Hegi
- Abstract
Supplementary Tables and Figures
- Published
- 2023
- Full Text
- View/download PDF
64. Data from MGMT Promoter Methylation Cutoff with Safety Margin for Selecting Glioblastoma Patients into Trials Omitting Temozolomide: A Pooled Analysis of Four Clinical Trials
- Author
-
Michael Weller, Josef Straub, Wim Van Criekinge, Greg Jones, L. Burt Nabors, Olivier L. Chinot, Mark R. Gilbert, Roger Stupp, Thierry Gorlia, Els Genbrugge, and Monika E. Hegi
- Abstract
Purpose:The methylation status of the O6-methylguanine DNA methyltransferase (MGMT) gene promoter is predictive for benefit from temozolomide in glioblastoma (GBM). A clinically optimized cutoff was sought allowing patient selection for therapy without temozolomide, while avoiding to withhold it from patients who may potentially benefit.Experimental Design: Quantitative MGMT methylation-specific PCR data were obtained for newly diagnosed patients with GBM screened or treated with standard radiotherapy and temozolomide in four randomized trials. The pooled dataset was randomly split into a training and test dataset. The unsupervised cutoff was obtained at a 50% probability to be (un)methylated. ROC analysis identified an optimal cutoff supervised by overall survival (OS).Results:For 4,041 patients valid MGMT results were obtained, whereof 1,725 were randomized. The unsupervised cutoff in the training dataset was 1.27 (log2[1,000 × (MGMT+1)/ACTB]), separating unmethylated and methylated patients. The optimal supervised cutoff for unmethylated patients was −0.28 (AUC = 0.61), classifying “truly unmethylated” (≤−0.28) and “gray zone” patients (>−0.28, ≤1.27), the latter comprising approximately 10% of cases. In contrast, for patients with MGMT methylation (>1.27) more methylation was not related to better outcome. Both methylated and gray zone patients performed significantly better for OS than truly unmethylated patients [HR = 0.35, 95% confidence interval (CI), 0.27–0.45, P < 0.0001; HR = 0.58, 95% CI, 0.43–0.78, P < 0.001], validated in the test dataset. The MGMT assay was highly reproducible upon retesting of 218 paired samples (R2 = 0.94).Conclusions:Low MGMT methylation (gray zone) may confer some sensitivity to temozolomide treatment, hence the lower safety margin should be considered for selecting patients with unmethylated GBM into trials omitting temozolomide.
- Published
- 2023
- Full Text
- View/download PDF
65. Supplementary materials-Clinical trial protocol from Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas
- Author
-
Mark R. Gilbert, John I. Gallin, Douglas B. Kuhns, William D. Figg, Katherine R. Calvo, Terri S. Armstrong, Javier Gonzales, Heather Leeper, Marta Penas-Prado, Eric M. Burton, Brett J. Theeler, Orwa Aboud, Christine Cordova, Nicole Lollo, Christine Siegel, Nancy Garren, Ramya Antony, Lisa Boris, Kelly Mentges, Ewa Grajkowska, Matthew Lindsley, Christine Bryla, Salman Ahmad, Elizabeth Vera, Tito R. Mendoza, Madison K. Butler, Guangyang Yu, Ying Pang, Yu-Ting Su, Tristan M. Sissung, Cody J. Peer, Danielle Fink, Debra A. Long Priel, Ying Yuan, and Jing Wu
- Abstract
Clinical trial protocol
- Published
- 2023
- Full Text
- View/download PDF
66. Data from Molecular Study of Malignant Gliomas Treated with Epidermal Growth Factor Receptor Inhibitors: Tissue Analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01
- Author
-
Eric C. Holland, Michael Prados, W.A. Yung, Susan Chang, Mark R. Gilbert, Patrick Wen, Lisa M. DeAngelis, John K. Cowell, Norma J. Nowak, Richard Wilson, John G. Kuhn, Alan H. Shih, William Pao, Kathleen Lamborn, Chelsea N. Grefe, Frank S. Lieberman, Lauren E. Abrey, Jeffrey R. Razier, Michael R. Rossi, and Andrew B. Lassman
- Abstract
Purpose: We investigated the molecular effect of the epidermal growth factor receptor (EGFR) inhibitors erlotinib and gefitinib in vivo on all available tumors from patients treated on North American Brain Tumor Consortium trials 01-03 and 00-01 for recurrent or progressive malignant glioma.Experimental Design: EGFR expression and signaling during treatment with erlotinib or gefitinib were analyzed by Western blot and compared with pre–erlotinib/gefitinib–exposed tissue or unexposed controls. Tumors were also analyzed for EGFR mutations and for other genomic abnormalities by array-based comparative genomic hybridization. Clinical data were used to associate molecular features with tumor sensitivity to erlotinib or gefitinib.Results: Erlotinib and gefitinib did not markedly affect EGFR activity in vivo. No lung signature mutations of EGFR exons 18 to 21 were observed. There was no clear association between erlotinib/gefitinib sensitivity and deletion or amplification events on array-based comparative genomic hybridization analysis, although novel genomic changes were identified.Conclusions: As erlotinib and gefitinib were generally ineffective at markedly inhibiting EGFR phosphorylation in these tumors, other assays may be needed to detect molecular effects. Additionally, the mechanism of erlotinib/gefitinib sensitivity likely differs between brain and lung tumors. Finally, novel genomic changes, including deletions of chromosomes 6, 21, and 22, represent new targets for further research.
- Published
- 2023
- Full Text
- View/download PDF
67. Figure S5 from Mitochondrial NIX Promotes Tumor Survival in the Hypoxic Niche of Glioblastoma
- Author
-
Deric M. Park, Mark R. Gilbert, Roger Abounader, Jeremy N. Rich, Amber J. Giles, Brian J. Williams, Hua Song, Wei Zhang, Orieta Celiku, Ying Zhang, and Jinkyu Jung
- Abstract
NIX-mediated mitophagy is related with hypoxia-induced macroautophagy.
- Published
- 2023
- Full Text
- View/download PDF
68. Data from Mitochondrial NIX Promotes Tumor Survival in the Hypoxic Niche of Glioblastoma
- Author
-
Deric M. Park, Mark R. Gilbert, Roger Abounader, Jeremy N. Rich, Amber J. Giles, Brian J. Williams, Hua Song, Wei Zhang, Orieta Celiku, Ying Zhang, and Jinkyu Jung
- Abstract
Cancer cells rely on mitochondrial functions to regulate key survival and death signals. How cancer cells regulate mitochondrial autophagy (mitophagy) in the tumor microenvironment as well as utilize mitophagy as a survival signal is still not well understood. Here, we elucidate a key survival mechanism of mitochondrial NIX-mediated mitophagy within the hypoxic region of glioblastoma, the most malignant brain tumor. NIX was overexpressed in the pseudopalisading cells that envelop the hypoxic–necrotic regions, and mitochondrial NIX expression was robust in patient-derived glioblastoma tumor tissues and glioblastoma stem cells. NIX was required for hypoxia and oxidative stress–induced mitophagy through NFE2L2/NRF2 transactivation. Silencing NIX impaired mitochondrial reactive oxygen species clearance, cancer stem cell maintenance, and HIF/mTOR/RHEB signaling pathways under hypoxia, resulting in suppression of glioblastoma survival in vitro and in vivo. Clinical significance of these findings was validated by the compelling association between NIX expression and poor outcome for patients with glioblastoma. Taken together, our findings indicate that the NIX-mediated mitophagic pathway may represent a key therapeutic target for solid tumors, including glioblastoma.Significance:NIX-mediated mitophagy regulates tumor survival in the hypoxic niche of glioblastoma microenvironment, providing a potential therapeutic target for glioblastoma.
- Published
- 2023
- Full Text
- View/download PDF
69. Figure S7-S10 from Mitochondrial NIX Promotes Tumor Survival in the Hypoxic Niche of Glioblastoma
- Author
-
Deric M. Park, Mark R. Gilbert, Roger Abounader, Jeremy N. Rich, Amber J. Giles, Brian J. Williams, Hua Song, Wei Zhang, Orieta Celiku, Ying Zhang, and Jinkyu Jung
- Abstract
NIX is functional for responding oxidative stress signal.
- Published
- 2023
- Full Text
- View/download PDF
70. Data from Dopamine Receptor D5 is a Modulator of Tumor Response to Dopamine Receptor D2 Antagonism
- Author
-
Joshua E. Allen, Wolfgang Oster, Patrick Y. Wen, Tracy T. Batchelor, Isabel Arrillaga-Romany, Robert Wechsler-Reya, Jessica Rusert, Mark R. Gilbert, Deric M. Park, Jinkyu Jung, Neil Charter, Sean Deacon, Rohinton S. Tarapore, Jessica Durrant, Alexander VanEngelenburg, Faye Doherty, Olivier Elemento, Wafik S. El-Deiry, Sophie Oster, C. Leah B. Kline, Coryandar Gilvary, Neel S. Madhukar, and Varun V. Prabhu
- Abstract
Purpose:Dopamine receptor D2 (DRD2) is a G protein–coupled receptor antagonized by ONC201, an anticancer small molecule in clinical trials for high-grade gliomas and other malignancies. DRD5 is a dopamine receptor family member that opposes DRD2 signaling. We investigated the expression of these dopamine receptors in cancer and their influence on tumor cell sensitivity to ONC201.Experimental Design:The Cancer Genome Atlas was used to determine DRD2/DRD5 expression broadly across human cancers. Cell viability assays were performed with ONC201 in >1,000 Genomic of Drug Sensitivity in Cancer and NCI60 cell lines. IHC staining of DRD2/DRD5 was performed on tissue microarrays and archival tumor tissues of glioblastoma patients treated with ONC201. Whole exome sequencing was performed in RKO cells with and without acquired ONC201 resistance. Wild-type and mutant DRD5 constructs were generated for overexpression studies.Results:DRD2 overexpression broadly occurs across tumor types and is associated with a poor prognosis. Whole exome sequencing of cancer cells with acquired resistance to ONC201 revealed a de novo Q366R mutation in the DRD5 gene. Expression of Q366R DRD5 was sufficient to induce tumor cell apoptosis, consistent with a gain-of-function. DRD5 overexpression in glioblastoma cells enhanced DRD2/DRD5 heterodimers and DRD5 expression was inversely correlated with innate tumor cell sensitivity to ONC201. Investigation of archival tumor samples from patients with recurrent glioblastoma treated with ONC201 revealed that low DRD5 expression was associated with relatively superior clinical outcomes.Conclusions:These results implicate DRD5 as a negative regulator of DRD2 signaling and tumor sensitivity to ONC201 DRD2 antagonism.
- Published
- 2023
- Full Text
- View/download PDF
71. Table S1 from Dopamine Receptor D5 is a Modulator of Tumor Response to Dopamine Receptor D2 Antagonism
- Author
-
Joshua E. Allen, Wolfgang Oster, Patrick Y. Wen, Tracy T. Batchelor, Isabel Arrillaga-Romany, Robert Wechsler-Reya, Jessica Rusert, Mark R. Gilbert, Deric M. Park, Jinkyu Jung, Neil Charter, Sean Deacon, Rohinton S. Tarapore, Jessica Durrant, Alexander VanEngelenburg, Faye Doherty, Olivier Elemento, Wafik S. El-Deiry, Sophie Oster, C. Leah B. Kline, Coryandar Gilvary, Neel S. Madhukar, and Varun V. Prabhu
- Abstract
Table S1
- Published
- 2023
- Full Text
- View/download PDF
72. Supplementary materials from Mitochondrial NIX Promotes Tumor Survival in the Hypoxic Niche of Glioblastoma
- Author
-
Deric M. Park, Mark R. Gilbert, Roger Abounader, Jeremy N. Rich, Amber J. Giles, Brian J. Williams, Hua Song, Wei Zhang, Orieta Celiku, Ying Zhang, and Jinkyu Jung
- Abstract
Methods of supplementary figures and additional information of materials.
- Published
- 2023
- Full Text
- View/download PDF
73. Data from Chemosensitivity of IDH1-Mutated Gliomas Due to an Impairment in PARP1-Mediated DNA Repair
- Author
-
Jing Wu, Chunzhang Yang, Mark R. Gilbert, Matthew G. Ewend, Jeffrey MacDonald, Seog In Moon, Wendy Bautista, Yu-Ting Su, Lauren N. Frady, Katherine Tech, Yang Liu, Jakub Kwintkiewicz, and Yanxin Lu
- Abstract
Mutations in isocitrate dehydrogenase (IDH) are the most prevalent genetic abnormalities in lower grade gliomas. The presence of these mutations in glioma is prognostic for better clinical outcomes with longer patient survival. In the present study, we found that defects in oxidative metabolism and 2-HG production confer chemosensitization in IDH1-mutated glioma cells. In addition, temozolomide (TMZ) treatment induced greater DNA damage and apoptotic changes in mutant glioma cells. The PARP1-associated DNA repair pathway was extensively compromised in mutant cells due to decreased NAD+ availability. Targeting the PARP DNA repair pathway extensively sensitized IDH1-mutated glioma cells to TMZ. Our findings demonstrate a novel molecular mechanism that defines chemosensitivity in IDH-mutated gliomas. Targeting PARP-associated DNA repair may represent a novel therapeutic strategy for gliomas. Cancer Res; 77(7); 1709–18. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
74. Data from Bayesian Optimal Interval Design: A Simple and Well-Performing Design for Phase I Oncology Trials
- Author
-
Mark R. Gilbert, Susan G. Hilsenbeck, Kenneth R. Hess, and Ying Yuan
- Abstract
Despite more than two decades of publications that offer more innovative model-based designs, the classical 3 + 3 design remains the most dominant phase I trial design in practice. In this article, we introduce a new trial design, the Bayesian optimal interval (BOIN) design. The BOIN design is easy to implement in a way similar to the 3 + 3 design, but is more flexible for choosing the target toxicity rate and cohort size and yields a substantially better performance that is comparable with that of more complex model-based designs. The BOIN design contains the 3 + 3 design and the accelerated titration design as special cases, thus linking it to established phase I approaches. A numerical study shows that the BOIN design generally outperforms the 3 + 3 design and the modified toxicity probability interval (mTPI) design. The BOIN design is more likely than the 3 + 3 design to correctly select the MTD and allocate more patients to the MTD. Compared with the mTPI design, the BOIN design has a substantially lower risk of overdosing patients and generally a higher probability of correctly selecting the MTD. User-friendly software is freely available to facilitate the application of the BOIN design. Clin Cancer Res; 22(17); 4291–301. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
75. Data from Genetic Modulation of Neurocognitive Function in Glioma Patients
- Author
-
Jeffrey S. Wefel, Melissa L. Bondy, Terri S. Armstrong, Mark R. Gilbert, Charles A. Conrad, Carol J. Etzel, Fu-Wen Liang, Spiridon Tsavachidis, Georgina N. Armstrong, Nicholas Boehling, Michael E. Scheurer, Erik P. Sulman, Renke Zhou, and Yanhong Liu
- Abstract
Purpose: Accumulating evidence supports the contention that genetic variation is associated with neurocognitive function in healthy individuals and increased risk for neurocognitive decline in a variety of patient populations, including cancer patients. However, this has rarely been studied in glioma patients.Experimental Design: To identify the effect of genetic variants on neurocognitive function, we examined the relationship between the genotype frequencies of 10,967 single-nucleotide polymorphisms in 580 genes related to five pathways (inflammation, DNA repair, metabolism, cognitive, and telomerase) and neurocognitive function in 233 newly diagnosed glioma patients before surgical resection. Four neuropsychologic tests that measured memory (Hopkins Verbal Learning Test—Revised), processing speed (Trail Making Test A), and executive function (Trail Making Test B, Controlled Oral Word Association) were examined.Results: Eighteen polymorphisms were associated with processing speed and 12 polymorphisms with executive function. For processing speed, the strongest signals were in IRS1 rs6725330 in the inflammation pathway (P = 2.5 × 10−10), ERCC4 rs1573638 in the DNA repair pathway (P = 3.4 × 10−7), and ABCC1 rs8187858 in metabolism pathway (P = 6.6 × 10−7). For executive function, the strongest associations were in NOS1 rs11611788 (P = 1.8 × 10−8) and IL16 rs1912124 (P = 6.0 × 10−7) in the inflammation pathway, and POLE rs5744761 (P = 6.0 × 10−7) in the DNA repair pathway. Joint effect analysis found significant gene polymorphism-dosage effects for processing speed (Ptrend = 9.4 × 10−16) and executive function (Ptrend = 6.6 × 10−15).Conclusions: Polymorphisms in inflammation, DNA repair, and metabolism pathways are associated with neurocognitive function in glioma patients and may affect clinical outcomes. Clin Cancer Res; 21(14); 3340–6. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
76. Supplementary Table Legend from Molecular Study of Malignant Gliomas Treated with Epidermal Growth Factor Receptor Inhibitors: Tissue Analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01
- Author
-
Eric C. Holland, Michael Prados, W.A. Yung, Susan Chang, Mark R. Gilbert, Patrick Wen, Lisa M. DeAngelis, John K. Cowell, Norma J. Nowak, Richard Wilson, John G. Kuhn, Alan H. Shih, William Pao, Kathleen Lamborn, Chelsea N. Grefe, Frank S. Lieberman, Lauren E. Abrey, Jeffrey R. Razier, Michael R. Rossi, and Andrew B. Lassman
- Abstract
Supplementary Table Legend from Molecular Study of Malignant Gliomas Treated with Epidermal Growth Factor Receptor Inhibitors: Tissue Analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01
- Published
- 2023
- Full Text
- View/download PDF
77. Data from Novel Targeting of Transcription and Metabolism in Glioblastoma
- Author
-
Jing Wu, Mark R. Gilbert, Chunzhang Yang, Zhengping Zhuang, Mones Abu-Asab, Mioara Larion, Thomas Estok, Kristan Meetze, Wei Zhang, Orieta Celiku, Aiguo Li, Dragan Maric, Adrian Lita, Gabriel Vasconcelos, Hallie Lappin, Li-Yuan Chen, Qi Zhang, Hua Song, Herui Wang, Robert Chen, and Yu-Ting Su
- Abstract
Purpose: Glioblastoma (GBM) is highly resistant to treatment, largely due to disease heterogeneity and resistance mechanisms. We sought to investigate a promising drug that can inhibit multiple aspects of cancer cell survival mechanisms and become an effective therapeutic for GBM patients.Experimental Design: To investigate TG02, an agent with known penetration of the blood–brain barrier, we examined the effects as single agent and in combination with temozolomide, a commonly used chemotherapy in GBM. We used human GBM cells and a syngeneic mouse orthotopic GBM model, evaluating survival and the pharmacodynamics of TG02. Mechanistic studies included TG02-induced transcriptional regulation, apoptosis, and RNA sequencing in treated GBM cells as well as the investigation of mitochondrial and glycolytic function assays.Results: We demonstrated that TG02 inhibited cell proliferation, induced cell death, and synergized with temozolomide in GBM cells with different genetic background but not in astrocytes. TG02-induced cytotoxicity was blocked by the overexpression of phosphorylated CDK9, suggesting a CDK9-dependent cell killing. TG02 suppressed transcriptional progression of antiapoptotic proteins and induced apoptosis in GBM cells. We further demonstrated that TG02 caused mitochondrial dysfunction and glycolytic suppression and ultimately ATP depletion in GBM. A prolonged survival was observed in GBM mice receiving combined treatment of TG02 and temozolomide. The TG02-induced decrease of CDK9 phosphorylation was confirmed in the brain tumor tissue.Conclusions: TG02 inhibits multiple survival mechanisms and synergistically decreases energy production with temozolomide, representing a promising therapeutic strategy in GBM, currently under investigation in an ongoing clinical trial. Clin Cancer Res; 24(5); 1124–37. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
78. Supplementary Figure and Data from Chemosensitivity of IDH1-Mutated Gliomas Due to an Impairment in PARP1-Mediated DNA Repair
- Author
-
Jing Wu, Chunzhang Yang, Mark R. Gilbert, Matthew G. Ewend, Jeffrey MacDonald, Seog In Moon, Wendy Bautista, Yu-Ting Su, Lauren N. Frady, Katherine Tech, Yang Liu, Jakub Kwintkiewicz, and Yanxin Lu
- Abstract
Supplementary figures and tables
- Published
- 2023
- Full Text
- View/download PDF
79. Supplementary Data from Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas
- Author
-
Mark R. Gilbert, John I. Gallin, Douglas B. Kuhns, William D. Figg, Katherine R. Calvo, Terri S. Armstrong, Javier Gonzales, Heather Leeper, Marta Penas-Prado, Eric M. Burton, Brett J. Theeler, Orwa Aboud, Christine Cordova, Nicole Lollo, Christine Siegel, Nancy Garren, Ramya Antony, Lisa Boris, Kelly Mentges, Ewa Grajkowska, Matthew Lindsley, Christine Bryla, Salman Ahmad, Elizabeth Vera, Tito R. Mendoza, Madison K. Butler, Guangyang Yu, Ying Pang, Yu-Ting Su, Tristan M. Sissung, Cody J. Peer, Danielle Fink, Debra A. Long Priel, Ying Yuan, and Jing Wu
- Abstract
Supplementary Figures and Tables
- Published
- 2023
- Full Text
- View/download PDF
80. Supplementary Table 1 from Phase I Study of Vorinostat in Combination with Temozolomide in Patients with High-Grade Gliomas: North American Brain Tumor Consortium Study 04-03
- Author
-
Patrick Y. Wen, Michael D. Prados, Igor Espinoza-Delgado, Matthew M. Ames, Xiabu Ye, Serena Desideri, Jihong Xu, Renee M. McGovern, Andrew B. Lassman, Frank S. Lieberman, H. Ian Robins, Mark R. Gilbert, W. K. Alfred Yung, Jan Drappatz, Susan M. Chang, Timothy F. Cloughesy, Kathleen R. Lamborn, John G. Kuhn, Joel M. Reid, Vinay K. Puduvalli, and Eudocia Q. Lee
- Abstract
PDF file, 80K, Supplementary Table 1: Temozolomide 150 mg/m2 Pharmacokinetic Parameters (first cycle).
- Published
- 2023
- Full Text
- View/download PDF
81. Data from Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas
- Author
-
Mark R. Gilbert, John I. Gallin, Douglas B. Kuhns, William D. Figg, Katherine R. Calvo, Terri S. Armstrong, Javier Gonzales, Heather Leeper, Marta Penas-Prado, Eric M. Burton, Brett J. Theeler, Orwa Aboud, Christine Cordova, Nicole Lollo, Christine Siegel, Nancy Garren, Ramya Antony, Lisa Boris, Kelly Mentges, Ewa Grajkowska, Matthew Lindsley, Christine Bryla, Salman Ahmad, Elizabeth Vera, Tito R. Mendoza, Madison K. Butler, Guangyang Yu, Ying Pang, Yu-Ting Su, Tristan M. Sissung, Cody J. Peer, Danielle Fink, Debra A. Long Priel, Ying Yuan, and Jing Wu
- Abstract
Purpose:To investigate the toxicity profile and establish an optimal dosing schedule of zotiraciclib with temozolomide in patients with recurrent high-grade astrocytoma.Patients and Methods:This two-stage phase I trial determined the MTD of zotiraciclib combined with either dose-dense (Arm1) or metronomic (Arm2) temozolomide using a Bayesian Optimal Interval design; then a randomized cohort expansion compared the progression-free survival rate at 4 months (PFS4) of the two arms for an efficient determination of a temozolomide schedule to combine with zotiraciclib at MTD. Pharmacokinetic and pharmacogenomic profiling were included. Patient-reported outcome was evaluated by longitudinal symptom burden.Results:Fifty-three patients were enrolled. Dose-limiting toxicities were neutropenia, diarrhea, elevated liver enzymes, and fatigue. MTD of zotiraciclib was 250 mg in both arms and thus selected for the cohort expansion. Dose-dense temozolomide plus zotiraciclib (PSF4 40%) compared favorably with metronomic temozolomide (PFS4 25%). Symptom burden worsened at cycle 2 but stabilized by cycle 4 in both arms. A significant decrease in absolute neutrophil count and neutrophil reactive oxygen species production occurred 12–24 hours after an oral dose of zotiraciclib but both recovered by 72 hours. Pharmacokinetic/pharmacogenomic analyses revealed that the CYP1A2_5347T>C (rs2470890) polymorphism was associated with higher AUCinf value.Conclusions:Zotiraciclib combined with temozolomide is safe in patients with recurrent high-grade astrocytomas. Zotiraciclib-induced neutropenia can be profound but mostly transient, warranting close monitoring rather than treatment discontinuation. Once validated, polymorphisms predicting drug metabolism may allow personalized dosing of zotiraciclib.
- Published
- 2023
- Full Text
- View/download PDF
82. Supplementary Legends from Dopamine Receptor D5 is a Modulator of Tumor Response to Dopamine Receptor D2 Antagonism
- Author
-
Joshua E. Allen, Wolfgang Oster, Patrick Y. Wen, Tracy T. Batchelor, Isabel Arrillaga-Romany, Robert Wechsler-Reya, Jessica Rusert, Mark R. Gilbert, Deric M. Park, Jinkyu Jung, Neil Charter, Sean Deacon, Rohinton S. Tarapore, Jessica Durrant, Alexander VanEngelenburg, Faye Doherty, Olivier Elemento, Wafik S. El-Deiry, Sophie Oster, C. Leah B. Kline, Coryandar Gilvary, Neel S. Madhukar, and Varun V. Prabhu
- Abstract
Supplementary legends for Figures S1-S5 and Tables S1-S2.
- Published
- 2023
- Full Text
- View/download PDF
83. Supplementary Table 2 from Phase I Study of Vorinostat in Combination with Temozolomide in Patients with High-Grade Gliomas: North American Brain Tumor Consortium Study 04-03
- Author
-
Patrick Y. Wen, Michael D. Prados, Igor Espinoza-Delgado, Matthew M. Ames, Xiabu Ye, Serena Desideri, Jihong Xu, Renee M. McGovern, Andrew B. Lassman, Frank S. Lieberman, H. Ian Robins, Mark R. Gilbert, W. K. Alfred Yung, Jan Drappatz, Susan M. Chang, Timothy F. Cloughesy, Kathleen R. Lamborn, John G. Kuhn, Joel M. Reid, Vinay K. Puduvalli, and Eudocia Q. Lee
- Abstract
PDF file, 62K, Supplementary Table 2: Grade 3 or 4 events with relationship unrelated to unlikely to combination therapy (Vorinostat + TMZ).
- Published
- 2023
- Full Text
- View/download PDF
84. Supplementary Table 1 from Molecular Study of Malignant Gliomas Treated with Epidermal Growth Factor Receptor Inhibitors: Tissue Analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01
- Author
-
Eric C. Holland, Michael Prados, W.A. Yung, Susan Chang, Mark R. Gilbert, Patrick Wen, Lisa M. DeAngelis, John K. Cowell, Norma J. Nowak, Richard Wilson, John G. Kuhn, Alan H. Shih, William Pao, Kathleen Lamborn, Chelsea N. Grefe, Frank S. Lieberman, Lauren E. Abrey, Jeffrey R. Razier, Michael R. Rossi, and Andrew B. Lassman
- Abstract
Supplementary Table 1 from Molecular Study of Malignant Gliomas Treated with Epidermal Growth Factor Receptor Inhibitors: Tissue Analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01
- Published
- 2023
- Full Text
- View/download PDF
85. Data from Phase I Study of Vorinostat in Combination with Temozolomide in Patients with High-Grade Gliomas: North American Brain Tumor Consortium Study 04-03
- Author
-
Patrick Y. Wen, Michael D. Prados, Igor Espinoza-Delgado, Matthew M. Ames, Xiabu Ye, Serena Desideri, Jihong Xu, Renee M. McGovern, Andrew B. Lassman, Frank S. Lieberman, H. Ian Robins, Mark R. Gilbert, W. K. Alfred Yung, Jan Drappatz, Susan M. Chang, Timothy F. Cloughesy, Kathleen R. Lamborn, John G. Kuhn, Joel M. Reid, Vinay K. Puduvalli, and Eudocia Q. Lee
- Abstract
Purpose: A phase I, dose-finding study of vorinostat in combination with temozolomide (TMZ) was conducted to determine the maximum tolerated dose (MTD), safety, and pharmacokinetics in patients with high-grade glioma (HGG).Experimental Design: This phase I, dose-finding, investigational study was conducted in two parts. Part 1 was a dose-escalation study of vorinostat in combination with TMZ 150 mg/m2/day for 5 days every 28 days. Part 2 was a dose-escalation study of vorinostat in combination with TMZ 150 mg/m2/day for 5 days of the first cycle and 200 mg/m2/day for 5 days of the subsequent 28-day cycles.Results: In part 1, the MTD of vorinostat administered on days 1 to 7 and 15 to 21 of every 28-day cycle, in combination with TMZ, was 500 mg daily. Dose-limiting toxicities (DLT) included grade 3 anorexia, grade 3 ALT, and grade 5 hemorrhage in the setting of grade 4 thrombocytopenia. In part 2, the MTD of vorinostat on days 1 to 7 and 15 to 21 of every 28-day cycle, combined with TMZ, was 400 mg daily. No DLTs were encountered, but vorinostat dosing could not be escalated further due to thrombocytopenia. The most common serious adverse events were fatigue, lymphopenia, thrombocytopenia, and thromboembolic events. There were no apparent pharmacokinetic interactions between vorinostat and TMZ. Vorinostat treatment resulted in hyperacetylation of histones H3 and H4 in peripheral mononuclear cells.Conclusion: Vorinostat in combination with temozolomide is well tolerated in patients with HGG. A phase I/II trial of vorinostat with radiotherapy and concomitant TMZ in newly diagnosed glioblastoma is underway. Clin Cancer Res; 18(21); 6032–9. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
86. Recurrent ACVR1 mutations in posterior fossa ependymoma
- Author
-
Drew Pratt, Calixto-Hope G. Lucas, Pavalan Panneer Selvam, Zied Abdullaev, Courtney Ketchum, Martha Quezado, Terri S. Armstrong, Mark R. Gilbert, Antonios Papanicolau-Sengos, Mark Raffeld, Hyoyoung Choo-Wosoba, Priya Chan, Nicholas Whipple, MacLean Nasrallah, Mariarita Santi, Vijay Ramaswamy, Caterina Giannini, Timothy A. Ritzmann, Richard G. Grundy, Anna Burford, Chris Jones, Cynthia Hawkins, Sriram Venneti, David A. Solomon, and Kenneth Aldape
- Subjects
Cellular and Molecular Neuroscience ,Ependymoma ,Mutation ,Humans ,Infratentorial Neoplasms ,Neurology (clinical) ,Activin Receptors, Type I ,Pathology and Forensic Medicine - Published
- 2022
- Full Text
- View/download PDF
87. Diagnosis and Management of Stroke in Adults with Primary Brain Tumor
- Author
-
Edina Komlodi-Pasztor, Mark R. Gilbert, and Terri S. Armstrong
- Subjects
Adult ,Stroke ,Oncology ,Brain Neoplasms ,Risk Factors ,Humans - Abstract
Purpose of Review This article reviews the risk factors, clinical presentations, differential diagnosis, and the types of strokes frequently seen in patients with primary brain neoplasms. This includes a discussion of approaches with a review of the available literature and provides recommendations for primary and secondary prevention specific to this patient population. Recent Findings Strokes in patients with brain tumors are often multifactorial. However, tailored approaches to stroke care are necessary to achieve optimal patient outcomes, AHA/ASA stroke guidelines provide little information on the management of stroke in cancer patients. A comprehensive algorithm for diagnosis for stroke in primary CNS tumor patients is proposed. Summary Understanding the potential complex etiology of stroke in patients with brain tumors is essential to provide appropriate treatment and initiate optimal prevention measures early in the cancer treatment program. Optimal care therefore requires a comprehensive approach including a variety of specialists and healthcare providers.
- Published
- 2022
- Full Text
- View/download PDF
88. A critical analysis of neuro-oncology clinical trials
- Author
-
Yeonju Kim, Terri S Armstrong, Mark R Gilbert, and Orieta Celiku
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BackgroundLimitations in trial design, accrual, and data reporting impact efficient and reliable drug evaluation in cancer clinical trials. These concerns have been recognized in neuro-oncology but have not been comprehensively evaluated. We conducted a semi-automated survey of adult interventional neuro-oncology trials, examining design, interventions, outcomes, and data availability trends.MethodsTrials were selected programmatically from ClinicalTrials.gov using primary malignant central nervous system tumor classification terms. Regression analyses assessed design and accrual trends; effect size analysis utilized survival rates among trials investigating survival.ResultsOf 3038 reviewed trials, most trials reporting relevant information were nonblinded (92%), single group (65%), nonrandomized (51%), and studied glioblastomas (47%) or other gliomas. Basic design elements were reported by most trials, with reporting increasing over time (OR = 1.24, P < .00001). Trials assessing survival outcomes were estimated to assume large effect sizes of interventions when powering their designs. Forty-two percent of trials were completed; of these, 38% failed to meet their enrollment target, with worse accrual over time (R = −0.94, P < .00001) and for US versus non-US based trials (OR = 0.5, P < .00001). Twenty-eight percent of completed trials reported partial results, with greater reporting for US (34.6%) versus non-US based trials (9.3%, P < .00001). Efficacy signals were detected by 15%–23% of completed trials reporting survival outcomes.ConclusionLow randomization rates, underutilization of controls, and overestimation of effect size, particularly pronounced in early-phase trials, impede generalizability of results. Suboptimal designs may be driven by accrual challenges, underscoring the need for cooperative efforts and novel designs. The limited results reporting highlights the need to incentivize data reporting and harmonization.
- Published
- 2023
- Full Text
- View/download PDF
89. Feasibility of a virtual reality intervention targeting distress and anxiety symptoms in patients with primary brain tumors: Interim analysis of a phase 2 clinical trial
- Author
-
Amanda L. King, Kayla N. Roche, Heather E. Leeper, Elizabeth Vera, Tito Mendoza, Kelly Mentges, Alvina A. Acquaye-Mallory, Kendra A. Adegbesan, Lisa Boris, Eric Burton, Anna Choi, Ewa Grajkowska, Tricia Kunst, Jason Levine, Nicole Lollo, Hope Miller, Marissa Panzer, Marta Penas-Prado, Valentina Pillai, Lily Polskin, Jennifer Reyes, Solmaz Sahebjam, Macy L. Stockdill, Brett J. Theeler, Jing Wu, Mark R. Gilbert, and Terri S. Armstrong
- Subjects
Cancer Research ,Neurology ,Oncology ,Neurology (clinical) - Abstract
Purpose Cancer patients experience distress and anxiety when undergoing imaging studies to monitor disease status, yet these symptoms are not always appropriately identified or well-managed. This interim analysis of a phase 2 clinical trial explored feasibility and acceptability of a virtual reality relaxation (VR) intervention for primary brain tumor (PBT) patients at the time of clinical evaluation. Methods English speaking, adult PBT patients with previous reports of distress and upcoming neuroimaging were recruited between March of 2021 and March 2022. A brief VR session was done within 2 weeks prior to neuroimaging with patient-reported outcomes (PROs) collected before and immediately post-intervention. Self-directed VR use over the next 1 month was encouraged with additional PROs assessments at 1 and 4 weeks. Feasibility metrics included enrollment, eligibility, attrition, and device-related adverse effects with satisfaction measured with qualitative phone interviews. Results Fifty-five patients were approached via email, 40 (73%) responded and 20 (50%) enrolled (9 declines, 11 screen fails). 65% of participants were ≤ 50 years, 50% were male, 90% were White/non-Hispanic, 85% had good KPS (≥ 90), and most were on active treatment. All patients completed the VR intervention, PROs questionnaires, weekly check-ins, and qualitative interview. Most (90%) reported frequent VR use and high satisfaction and only 7 mild AEs were recorded (headache, dizziness, nausea, neck pain). Conclusion This interim analysis supports feasibility and acceptability of a novel VR intervention to target psychological symptoms for PBT patients. Trial enrollment will continue to assess for intervention efficacy. Trial Registration NCT04301089 registered on 3/9/2020.
- Published
- 2023
- Full Text
- View/download PDF
90. 'Tumor Treating Fields' delivered via electromagnetic induction have varied effects across glioma cell lines and electric field amplitudes
- Author
-
Rea Ravin, Teddy X. Cai, Aiguo Li, Nicole Briceno, Randall H. Pursley, Marcial Garmendia-Cedillos, Tom Pohida, Herui Wang, Zhengping Zhuang, Jing Cui, Nicole Y. Morgan, Nathan H. Williamson, Mark R. Gilbert, and Peter J. Basser
- Subjects
Article - Abstract
Previous studies reported that alternating electric fields (EFs) in the intermediate frequency (100 – 300 kHz) and low intensity (1 – 3 V/cm) regime — termed “Tumor Treating Fields” (TTFields) — have a specific, anti-proliferative effect on glioblastoma multiforme (GBM) cells. However, the mechanism(s) of action remain(s) incompletely understood, hindering the clinical adoption of treatments based on TTFields. To advance the study of such treatmentin vitro, we developed an inductive device to deliver EFs to cell cultures which improves thermal and osmolar regulation compared to prior devices. Using this inductive device, we applied continuous, 200 kHz electromagnetic fields (EMFs) with a radial EF amplitude profile spanning 0 – 6.5 V/cm to cultures of primary rat astrocytes and several human GBM cell lines — U87, U118, GSC827, and GSC923 — for a duration of 72 hours. Cell density was assessed via segmented pixel densities from GFP expression (U87, U118) or from staining (astrocytes, GSC827, GSC923). Further RNA-Seq analyses were performed on GSC827 and GSC923 cells. Treated cultures of all cell lines exhibited little to no change in proliferation at lower EF amplitudes (0 – 3 V/cm). At higher amplitudes (> 4 V/cm), different effects were observed. Apparent cell densities increased (U87), decreased (GSC827, GSC923), or showed little change (U118, astrocytes). RNA-Seq analyses on treated and untreated GSC827 and GSC923 cells revealed differentially expressed gene sets of interest, such as those related to cell cycle control. Up- and down-regulation, however, was not consistent across cell lines nor EF amplitudes. Our results indicate no consistent, anti-proliferative effect of 200 kHz EMFs across GBM cell lines and thus contradict previousin vitrofindings. Rather, effects varied across different cell lines and EF amplitude regimes, highlighting the need to assess the effect(s) of TTFields and similar treatments on a per cell line basis.
- Published
- 2023
- Full Text
- View/download PDF
91. Objective response rate targets for recurrent glioblastoma clinical trials based on the historic association between objective response rate and median overall survival
- Author
-
Benjamin M Ellingson, Patrick Y Wen, Susan M Chang, Martin van den Bent, Michael A Vogelbaum, Gang Li, Shanpeng Li, Jiyoon Kim, Gilbert Youssef, Wolfgang Wick, Andrew B Lassman, Mark R Gilbert, John F de Groot, Michael Weller, Evanthia Galanis, Timothy F Cloughesy, and Neurology
- Subjects
Cancer Research ,SDG 3 - Good Health and Well-being ,Oncology ,Neurology (clinical) - Abstract
Durable objective response rate (ORR) remains a meaningful endpoint in recurrent cancer; however, the target ORR for single-arm recurrent glioblastoma trials has not been based on historic information or tied to patient outcomes. The current study reviewed 68 treatment arms comprising 4793 patients in past trials in recurrent glioblastoma in order to judiciously define target ORRs for use in recurrent glioblastoma trials. ORR was estimated at 6.1% [95% CI 4.23; 8.76%] for cytotoxic chemothera + pies (ORR = 7.59% for lomustine, 7.57% for temozolomide, 0.64% for irinotecan, and 5.32% for other agents), 3.37% for biologic agents, 7.97% for (select) immunotherapies, and 26.8% for anti-angiogenic agents. ORRs were significantly correlated with median overall survival (mOS) across chemotherapy (R2 = 0.4078, P < .0001), biologics (R2 = 0.4003, P = .0003), and immunotherapy trials (R2 = 0.8994, P < .0001), but not anti-angiogenic agents (R2 = 0, P = .8937). Pooling data from chemotherapy, biologics, and immunotherapy trials, a meta-analysis indicated a strong correlation between ORR and mOS (R2 = 0.3900, P < .0001; mOS [weeks] = 1.4xORR + 24.8). Assuming an ineffective cytotoxic (control) therapy has ORR = 7.6%, the average ORR for lomustine and temozolomide trials, a sample size of ≥40 patients with target ORR>25% is needed to demonstrate statistical significance compared to control with a high level of confidence (P < .01) and adequate power (>80%). Given this historic data and potential biases in patient selection, we recommend that well-controlled, single-arm phase II studies in recurrent glioblastoma should have a target ORR >25% (which translates to a median OS of approximately 15 months) and a sample size of ≥40 patients, in order to convincingly demonstrate antitumor activity. Crucially, this response needs to have sufficient durability, which was not addressed in the current study.
- Published
- 2023
- Full Text
- View/download PDF
92. Effects of Cognitive Reserve on Cognition in Individuals With Central Nervous System Disease
- Author
-
Varna Jammula, Diane Cooper, Terri Armstrong, Heather Leeper, and Mark R. Gilbert
- Subjects
cognition ,Cognitive Neuroscience ,central nervous system disease ,Standardized test ,CINAHL ,Review ,Neuropsychological Tests ,Central nervous system disease ,Cognitive Reserve ,Central Nervous System Diseases ,Medicine ,Humans ,Association (psychology) ,Cognitive reserve ,business.industry ,Neuropsychology ,Cognition ,General Medicine ,medicine.disease ,brain injury ,Psychiatry and Mental health ,Neuropsychology and Physiological Psychology ,Systematic review ,Educational Status ,business ,Clinical psychology - Abstract
Cognitive reserve (CR) has been proposed to account for functional outcome differences in brain pathology and its clinical manifestations. The purpose of our paper is to systematically review the effects of CR on cognitive outcomes in individuals with neurodegenerative and structural CNS diseases. We performed a systematic search of PubMed, CINAHL (Cumulative Index to Nursing and Allied Health Literature), and PsychInfo using the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Seventeen studies met the predetermined inclusion criteria and were selected for review. Education level was the most commonly used measure for CR, and various neuropsychological tests were used to measure cognitive outcomes. Regardless of the CNS disease of the individuals, almost all of the studies reported a positive association between CR and cognitive outcomes when they were evaluated cross-sectionally. However, when evaluated longitudinally, CR had either no effect on, or a negative association with, cognitive outcomes. Based on studies across a broad spectrum of CNS diseases, our findings suggest that CR may serve as a predictor of cognitive outcomes in individuals with CNS diseases. However, studies to date are limited by a lack of imaging analyses and standardized assessment strategies. The ability to use a standardized measure to assess the longitudinal effects of CR may allow for the development of more targeted treatment methods, resulting in improved disease outcomes for individuals.
- Published
- 2021
93. Multilayer interface tracking model of pure tungsten oxidation
- Author
-
Shu Huang, Ryan Kerr, Samuel Murphy, Mark R Gilbert, and Jaime Marian
- Subjects
Mechanics of Materials ,Modeling and Simulation ,General Materials Science ,Condensed Matter Physics ,Computer Science Applications - Abstract
We present a numerical model to predict oxide scale growth on tungsten surfaces under exposure to oxygen at high temperatures. The model captures the formation of four thermodynamically-compatible oxide sublayers, WO2, WO2.72, WO2.9, and WO3, on top of the metal substrate. Oxide layer growth is simulated by tracking the oxide/oxide and oxide/metal interfaces using a sharp-interface Stefan model coupled to diffusion kinetics. The model is parameterized using selected experimental measurements and electronic structure calculations of the diffusivities of all the oxide subphases involved. We simulate oxide growth at temperatures of 600∘C and above, extracting the power law growth exponents in each case, which we find to deviate from classical parabolic growth in several cases. We conduct a comparison of the model predictions with an extensive experimental data set, with reasonable agreement at most temperatures. While many gaps in our understanding still exist, this work is a first attempt at embedding the thermodynamic and kinetic complexity of tungsten oxide growth into a comprehensive mesoscale kinetic model that attempts to capture the essential features of tungsten oxidation to fill existing knowledge gaps and guide and enhance future tungsten oxidation models.
- Published
- 2022
- Full Text
- View/download PDF
94. 379 Human Endogenous Retrovirus-K (HML-2) Contributes to a Unique Stem-Cell Niche in Glioblastoma
- Author
-
Ashish Harish Shah, Sarah Rivas, Tara Doucet-Ohare, Vaidya Govindarajan, Catherine DeMarino, Leonel Ampie, Yeshavanath Banasavadi-Siddegowda, Dragan Maric, Rob Suter, Myoung-hwa Lee, Kareem A. Zaghloul, Stuart Walbridge, Marta Garcia-Montojo, Joe Steiner, Kory Johnson, Mark R. Gilbert, John D. Heiss, and Avindra Nath
- Subjects
Surgery ,Neurology (clinical) - Published
- 2023
- Full Text
- View/download PDF
95. Recurrent fusions in PLAGL1 define a distinct subset of pediatric-type supratentorial neuroepithelial tumors
- Author
-
Konstantin Okonechnikov, Christina Blume, Mariëtte E.G. Kranendonk, Stephanie Bunkowski, Dominik Sturm, Matija Snuderl, David R Ghasemi, Damian Stichel, Philipp Sievers, David T.W. Jones, Richard Grundy, Christian Mawrin, Ofelia Cruz, Andreas von Deimling, David W. Ellison, Tuyu Zheng, Daniel Schrimpf, Mark R. Gilbert, Lenka Krskova, Pascale Varlet, Hildegard Dohmen, Till Acker, Henning B. Boldt, Sophie C Henneken, Kenneth Aldape, Stefan Rutkowski, Mariona Suñol, Andrey Korshunov, Stefan M. Pfister, Julia Benzel, David Capper, Wolf Mueller, Ulrich Schüller, Sebastian Brandner, Patrick N. Harter, Zied Abdullaev, Celso Pouget, Rudi Beschorner, Kendra K Maaß, Viktoria Ruf, Pieter Wesseling, Mélanie Pagès, Nada Jabado, Terri S. Armstrong, Patricia Kohlhof-Meinecke, Martin Sill, Marcel Kool, Koichi Ichimura, Felix Sahm, Guido Reifenberger, Kristian W. Pajtler, Wolfgang Wick, David E. Reuss, Leonille Schweizer, Christine Stadelmann, Cinzia Lavarino, Ales Vicha, Michal Zapotocky, Noreen Akhtar, Pathology, CCA - Cancer biology and immunology, and CCA - Imaging and biomarkers
- Subjects
Ependymoma ,Male ,Pathology ,medicine.medical_specialty ,SOX10 ,Cell Cycle Proteins ,Biology ,Supratentorial ,Pathology and Forensic Medicine ,Neuroepithelial tumor ,Fusion gene ,Pleomorphic adenoma ,OLIG2 ,Cellular and Molecular Neuroscience ,FOXO1 ,medicine ,Humans ,Oncogene Fusion ,ddc:610 ,EP300 ,Child ,PLAGL1 ,Original Paper ,Tumor Suppressor Proteins ,EWSR1 ,Supratentorial Neoplasms ,medicine.disease ,DNA methylation ,Gene fusion ,Female ,Neurology (clinical) ,Genomic imprinting ,Transcription Factors - Abstract
Ependymomas encompass a heterogeneous group of central nervous system (CNS) neoplasms that occur along the entire neuroaxis. In recent years, extensive (epi-)genomic profiling efforts have identified several molecular groups of ependymoma that are characterized by distinct molecular alterations and/or patterns. Based on unsupervised visualization of a large cohort of genome-wide DNA methylation data, we identified a highly distinct group of pediatric-type tumors (n = 40) forming a cluster separate from all established CNS tumor types, of which a high proportion were histopathologically diagnosed as ependymoma. RNA sequencing revealed recurrent fusions involving the pleomorphic adenoma gene-like 1 (PLAGL1) gene in 19 of 20 of the samples analyzed, with the most common fusion being EWSR1:PLAGL1 (n = 13). Five tumors showed a PLAGL1:FOXO1 fusion and one a PLAGL1:EP300 fusion. High transcript levels of PLAGL1 were noted in these tumors, with concurrent overexpression of the imprinted genes H19 and IGF2, which are regulated by PLAGL1. Histopathological review of cases with sufficient material (n = 16) demonstrated a broad morphological spectrum of tumors with predominant ependymoma-like features. Immunohistochemically, tumors were GFAP positive and OLIG2- and SOX10 negative. In 3/16 of the cases, a dot-like positivity for EMA was detected. All tumors in our series were located in the supratentorial compartment. Median age of the patients at the time of diagnosis was 6.2 years. Median progression-free survival was 35 months (for 11 patients with data available). In summary, our findings suggest the existence of a novel group of supratentorial neuroepithelial tumors that are characterized by recurrent PLAGL1 fusions and enriched for pediatric patients.
- Published
- 2021
- Full Text
- View/download PDF
96. Independently validated sex-specific nomograms for predicting survival in patients with newly diagnosed glioblastoma: NRG Oncology RTOG 0525 and 0825
- Author
-
Deborah T. Blumenthal, Robin Buerki, Mitchell Machtay, Valerie Panet-Raymond, Stephanie L. Pugh, Nirav Patil, Eashwar Somasundaram, James R. Connor, Andrew E. Sloan, H. Ian Robins, Grant K. Hunter, Marta Penas-Prado, Justin D. Lathia, Serah Choi, Kristin A Waite, John C. Flickinger, Lynn S. Ashby, Maria Werner-Wasik, Joshua B. Rubin, Michael E. Berens, Merideth M Wendland, Mark R. Gilbert, Jill S. Barnholtz-Sloan, and Minesh P. Mehta
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Survival ,Malignant brain tumor ,Newly diagnosed ,Extent of resection ,Nomogram ,Internal medicine ,Sex differences ,medicine ,Humans ,In patient ,Promoter Regions, Genetic ,Proportional Hazards Models ,business.industry ,Brain Neoplasms ,medicine.disease ,Prognosis ,Sex specific ,Clinical trial ,Nomograms ,Neurology ,Clinical Study ,Female ,Neurology (clinical) ,business ,Glioblastoma - Abstract
Background/purpose Glioblastoma (GBM) is the most common primary malignant brain tumor. Sex has been shown to be an important prognostic factor for GBM. The purpose of this study was to develop and independently validate sex-specific nomograms for estimation of individualized GBM survival probabilities using data from 2 independent NRG Oncology clinical trials. Methods This analysis included information on 752 (NRG/RTOG 0525) and 599 (NRG/RTOG 0825) patients with newly diagnosed GBM. The Cox proportional hazard models by sex were developed using NRG/RTOG 0525 and significant variables were identified using a backward selection procedure. The final selected models by sex were then independently validated using NRG/RTOG 0825. Results Final nomograms were built by sex. Age at diagnosis, KPS, MGMT promoter methylation and location of tumor were common significant predictors of survival for both sexes. For both sexes, tumors in the frontal lobes had significantly better survival than tumors of multiple sites. Extent of resection, and use of corticosteroids were significant predictors of survival for males. Conclusions A sex specific nomogram that assesses individualized survival probabilities (6-, 12- and 24-months) for patients with GBM could be more useful than estimation of overall survival as there are factors that differ between males and females. A user friendly online application can be found here—https://npatilshinyappcalculator.shinyapps.io/SexDifferencesInGBM/.
- Published
- 2021
97. Report of Canonical BCR-ABL1 Fusion in Glioblastoma
- Author
-
Rosandra N. Kaplan, Olga Kim, Alice Ranjan, Martha Quezado, Jing Wu, Mythili Merchant, Liqiang Xi, Guangyang Yu, Megan Mackey, Kevin Camphausen, Matthias Holdhoff, Young K. Song, Zied Abdullaev, Kenneth Aldape, Kareem A. Zaghloul, David O. Kamson, Jun Wei, Terri S. Armstrong, Javed Khan, Sivasish Sindiri, Xinyu Wen, Ying Pang, Mark R. Gilbert, Svetlana Pack, Lisa Boris, Madison Butler, Ramya Antony, Orwa Aboud, and Hsien-Chao Chou
- Subjects
Cancer Research ,Bcr abl1 ,Fusion ,Oncology ,business.industry ,medicine ,Cancer research ,Case Reports ,medicine.disease ,business ,Glioblastoma - Published
- 2021
- Full Text
- View/download PDF
98. Impact of the methylation classifier and ancillary methods on CNS tumor diagnostics
- Author
-
Martha Quezado, Hye-Jung Chung, Lola Saidkhodjaeva, Manoj Tyagi, Zhichao Wu, Sushma Nagaraj, Mark R. Gilbert, Andreas von Deimling, Rust Turakulov, Shannon Skarshaug, Michael Newford, Antonios Papanicolau-Sengos, Drew Pratt, Kenneth Aldape, Kayla O’Donnell, Abigail K. Suwala, Candice Perry, Svetlana Pack, Vineela Gangalapudi, Shirin Karimi, Farshad Nassiri, Yasin Mamatjan, Zied Abdullaev, Felix Sahm, Valerie Zgonc, Liqiang Xi, Gelareh Zadeh, Mark Raffeld, Kristin Valdez, and Eytan Ruppin
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Confounding ,Brain tissue ,Neuropathology ,Methylation ,New diagnosis ,Dna methylation profiling ,Internal medicine ,Basic and Translational Investigations ,Medicine ,Neurology (clinical) ,CNS TUMORS ,business ,Classifier (UML) - Abstract
Background Accurate CNS tumor diagnosis can be challenging, and methylation profiling can serve as an adjunct to classify diagnostically difficult cases. Methods An integrated diagnostic approach was employed for a consecutive series of 1258 surgical neuropathology samples obtained primarily in a consultation practice over 2-year period. DNA methylation profiling and classification using the DKFZ/Heidelberg CNS tumor classifier was performed, as well as unsupervised analyses of methylation data. Ancillary testing, where relevant, was performed. Results Among the received cases in consultation, a high-confidence methylation classifier score (>0.84) was reached in 66.4% of cases. The classifier impacted the diagnosis in 46.7% of these high-confidence classifier score cases, including a substantially new diagnosis in 26.9% cases. Among the 289 cases received with only a descriptive diagnosis, methylation was able to resolve approximately half (144, 49.8%) with high-confidence scores. Additional methods were able to resolve diagnostic uncertainty in 41.6% of the low-score cases. Tumor purity was significantly associated with classifier score (P = 1.15e−11). Deconvolution demonstrated that suspected glioblastomas (GBMs) matching as control/inflammatory brain tissue could be resolved into GBM methylation profiles, which provided a proof-of-concept approach to resolve tumor classification in the setting of low tumor purity. Conclusions This work assesses the impact of a methylation classifier and additional methods in a consultative practice by defining the proportions with concordant vs change in diagnosis in a set of diagnostically challenging CNS tumors. We address approaches to low-confidence scores and confounding issues of low tumor purity.
- Published
- 2021
- Full Text
- View/download PDF
99. Adenosine A2A Receptor Activation Enhances Blood–Tumor Barrier Permeability in a Rodent Glioma Model
- Author
-
Jessica Bills, Meili Zhang, Dionne Davis, Monica Manglani, Kunio Nagashima, Stuart Walbridge, Brandon Foster, Dorian B. McGavern, Priya Shankarappa, Amélie Vézina, DreeAnna Morris, Matthew McCord, Hua Song, Jessica Kindrick, William D. Figg, Wei Zhang, Leslie L. Muldoon, Sadhana Jackson, Mark R. Gilbert, and Cody J. Peer
- Subjects
Cancer Research ,Receptor, Adenosine A2A ,Adenosine A2A receptor ,Mice, SCID ,Transfection ,Article ,Mice ,Rats, Nude ,Downregulation and upregulation ,Glioma ,Animals ,Humans ,Medicine ,Molecular Biology ,Temozolomide ,Brain Neoplasms ,business.industry ,medicine.disease ,Survival Analysis ,Rats ,Regadenoson ,Endothelial stem cell ,Disease Models, Animal ,Oncology ,Blood-Brain Barrier ,Paracellular transport ,Cancer research ,Female ,Stem cell ,business ,medicine.drug - Abstract
The blood–tumor barrier (BTB) limits the entry of effective chemotherapeutic agents into the brain for treatment of malignant tumors like glioblastoma. Poor drug entry across the BTB allows infiltrative glioma stem cells to evade therapy and develop treatment resistance. Regadenoson, an FDA-approved adenosine A2A receptor (A2AR) agonist, has been shown to increase drug delivery across the blood–brain barrier in non–tumor-bearing rodents without a defined mechanism of enhancing BTB permeability. Here, we characterize the time-dependent impact of regadenoson on brain endothelial cell interactions and paracellular transport, using mouse and rat brain endothelial cells and tumor models. In vitro, A2AR activation leads to disorganization of cytoskeletal actin filaments by 30 minutes, downregulation of junctional protein expression by 4 hours, and reestablishment of endothelial cell integrity by 8 hours. In rats bearing intracranial gliomas, regadenoson treatment results in increase of intratumoral temozolomide concentrations, yet no increased survival noted with combined temozolomide therapy. These findings demonstrate regadenoson's ability to induce brain endothelial structural changes among glioma to increase BTB permeability. The use of vasoactive mediators, like regadenoson, which transiently influences paracellular transport, should further be explored to evaluate their potential to enhance central nervous system treatment delivery to aggressive brain tumors. Implications: This study provides insight on the use of a vasoactive agent to increase exposure of the BTB to chemotherapy with intention to improve glioma treatment efficacy.
- Published
- 2021
- Full Text
- View/download PDF
100. Germline polymorphisms in
- Author
-
Michael E, Scheurer, Renke, Zhou, Mark R, Gilbert, Melissa L, Bondy, Erik P, Sulman, Ying, Yuan, Yanhong, Liu, Elizabeth, Vera, Merideth M, Wendland, Emad F, Youssef, Volker W, Stieber, Ritsuko R, Komaki, John C, Flickinger, Lawrence C, Kenyon, H Ian, Robins, Grant K, Hunter, Ian R, Crocker, Samuel T, Chao, Stephanie L, Pugh, and Terri S, Armstrong
- Abstract
We sought to identify clinical and genetic predictors of temozolomide-related myelotoxicity among patients receiving therapy for glioblastoma.Patients (23% of patients developed myelotoxicity (Myelotoxicity during concurrent chemoradiation with temozolomide is an uncommon but serious event, often leading to treatment cessation. Successful prediction of toxicity may lead to more cost-effective individualized monitoring of at-risk subjects. The addition of genetic factors greatly enhanced our ability to predict toxicity among a group of similarly treated glioblastoma patients.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.