Try a new search

Format these results:

Searched for:

person:chia01

Total Results:

121


PIK3CA activating mutations are associated with more disseminated disease at presentation and earlier recurrence in glioblastoma

Tanaka, Shota; Batchelor, Tracy T; Iafrate, A John; Dias-Santagata, Dora; Borger, Darrell R; Ellisen, Leif W; Yang, Daniel; Louis, David N; Cahill, Daniel P; Chi, Andrew S
Phosphatidylinositol 3-kinase signaling promotes cell growth and survival and is frequently activated in infiltrative gliomas. Activating mutations in PIK3CA gene are observed in 6-15% of glioblastomas, although their clinical significance is largely undescribed. The objective of this study was to examine whether PIK3CA mutations are associated with a specific clinical phenotype in glioblastoma. We retrospectively reviewed 157 consecutive newly diagnosed glioblastoma patients from December 2009 to June 2012 who underwent molecular profiling consisting of targeted hotspot genotyping, fluorescence in situ hybridization for gene amplification, and methylation-specific PCR for O6-methylguanine-DNA methyltransferase promoter methylation. Molecular alterations were correlated with clinical features, imaging and outcome. The Cancer Genome Atlas data was analyzed as a validation set. There were 91 males; median age was 58 years (range, 23-85). With a median follow-up of 20.9 months, median progression-free survival (PFS) and estimated overall survival (OS) were 11.9 and 24.0 months, respectively. Thirteen patients (8.3%) harbored PIK3CA mutation, which was associated with younger age (mean 49.4 vs. 58.1 years, p = 0.02). PIK3CA mutation correlated with shorter PFS (median 6.9 vs. 12.4 months, p = 0.01) and OS (median 21.2 vs. 24.2 months, p = 0.049) in multivariate analysis. A significant association between PIK3CA mutation and more disseminated disease at diagnosis, as defined by gliomatosis, multicentric lesions, or distant leptomeningeal lesions, was observed (46.2% vs. 11.1%, p = 0.004). In conclusion, despite the association with younger age, PIK3CA activating mutations are associated with earlier recurrence and shorter survival in adult glioblastoma. The aggressive course of these tumors may be related to their propensity for disseminated presentation.
PMID: 31036078
ISSN: 2051-5960
CID: 3854502

First clinical experience with DRD2/3 antagonist ONC201 in H3 K27M-mutant pediatric diffuse intrinsic pontine glioma: a case report [Case Report]

Hall, Matthew D; Odia, Yazmin; Allen, Joshua E; Tarapore, Rohinton; Khatib, Ziad; Niazi, Toba N; Daghistani, Doured; Schalop, Lee; Chi, Andrew S; Oster, Wolfgang; Mehta, Minesh P
Diffuse intrinsic pontine gliomas (DIPGs) frequently harbor the histone H3 K27M mutation. Gliomas with this mutation commonly overexpress dopamine receptor (DR) D2 and suppress DRD5, leading to enhanced sensitivity to DRD2 antagonism. This study reports the first clinical experience with the DRD2/3 antagonist ONC201 as a potential targeted therapy for H3 K27M–mutant DIPG. One pediatric patient (a 10-year-old girl) with H3 K27M–mutant DIPG was enrolled in an investigator-initiated, IRB-approved compassionate-use study and began single-agent ONC201 treatment 1 month after completing radiotherapy. The study endpoints were clinical and radiographic response (primary) and toxicities (secondary).
PMID: 30952114
ISSN: 1933-0715
CID: 3935732

Buparlisib in Patients With Recurrent Glioblastoma Harboring Phosphatidylinositol 3-Kinase Pathway Activation: An Open-Label, Multicenter, Multi-Arm, Phase II Trial

Wen, Patrick Y; Touat, Mehdi; Alexander, Brian M; Mellinghoff, Ingo K; Ramkissoon, Shakti; McCluskey, Christine S; Pelton, Kristine; Haidar, Sam; Basu, Sankha S; Gaffey, Sarah C; Brown, Loreal E; Martinez-Ledesma, Juan Emmanuel; Wu, Shaofang; Kim, Jungwoo; Wei, Wei; Park, Mi-Ae; Huse, Jason T; Kuhn, John G; Rinne, Mikael L; Colman, Howard; Agar, Nathalie Y R; Omuro, Antonio M; DeAngelis, Lisa M; Gilbert, Mark R; de Groot, John F; Cloughesy, Timothy F; Chi, Andrew S; Roberts, Thomas M; Zhao, Jean J; Lee, Eudocia Q; Nayak, Lakshmi; Heath, James R; Horky, Laura L; Batchelor, Tracy T; Beroukhim, Rameen; Chang, Susan M; Ligon, Azra H; Dunn, Ian F; Koul, Dimpy; Young, Geoffrey S; Prados, Michael D; Reardon, David A; Yung, W K Alfred; Ligon, Keith L
PURPOSE/OBJECTIVE:Phosphatidylinositol 3-kinase (PI3K) signaling is highly active in glioblastomas. We assessed pharmacokinetics, pharmacodynamics, and efficacy of the pan-PI3K inhibitor buparlisib in patients with recurrent glioblastoma with PI3K pathway activation. METHODS:This study was a multicenter, open-label, multi-arm, phase II trial in patients with PI3K pathway-activated glioblastoma at first or second recurrence. In cohort 1, patients scheduled for re-operation after progression received buparlisib for 7 to 13 days before surgery to evaluate brain penetration and modulation of the PI3K pathway in resected tumor tissue. In cohort 2, patients not eligible for re-operation received buparlisib until progression or unacceptable toxicity. Once daily oral buparlisib 100 mg was administered on a continuous 28-day schedule. Primary end points were PI3K pathway inhibition in tumor tissue and buparlisib pharmacokinetics in cohort 1 and 6-month progression-free survival (PFS6) in cohort 2. RESULTS:and proliferation were not significant. Tumor-to-plasma drug level was 1.0. In cohort 2, four (8%) of 50 patients reached 6-month PFS6, and the median PFS was 1.7 months (95% CI, 1.4 to 1.8 months). The most common grade 3 or greater adverse events related to treatment were lipase elevation (n = 7 [10.8%]), fatigue (n = 4 [6.2%]), hyperglycemia (n = 3 [4.6%]), and elevated ALT (n = 3 [4.6%]). CONCLUSION/CONCLUSIONS:Buparlisib had minimal single-agent efficacy in patients with PI3K-activated recurrent glioblastoma. Although buparlisib achieved significant brain penetration, the lack of clinical efficacy was explained by incomplete blockade of the PI3K pathway in tumor tissue. Integrative results suggest that additional study of PI3K inhibitors that achieve more-complete pathway inhibition may still be warranted.
PMID: 30715997
ISSN: 1527-7755
CID: 3794892

Cell surface Notch ligand DLL3 is a therapeutic target in isocitrate dehydrogenase mutant glioma

Spino, Marissa; Kurz, Sylvia C; Chiriboga, Luis; Serrano, Jonathan; Zeck, Briana; Sen, Namita; Patel, Seema; Shen, Guomiao; Vasudevaraja, Varshini; Tsirigos, Aristotelis; Suryadevara, Carter M; Frenster, Joshua D; Tateishi, Kensuke; Wakimoto, Hiroaki; Jain, Rajan; Riina, Howard A; Nicolaides, Theodore; Sulman, Erik P; Cahill, Daniel P; Golfinos, John G; Isse, Kumiko; Saunders, Laura R; Zagzag, David; Placantonakis, Dimitris G; Snuderl, Matija; Chi, Andrew S
PURPOSE/OBJECTIVE:Isocitrate dehydrogenase (IDH) mutant gliomas are a distinct glioma molecular subtype for which no effective molecularly-directed therapy exists. Low-grade gliomas, which are 80-90% IDH mutant, have high RNA levels of the cell surface Notch ligand DLL3. We sought to determine DLL3 expression by immunohistochemistry in glioma molecular subtypes and the potential efficacy of an anti-DLL3 antibody drug conjugate (ADC), rovalpituzumab tesirine (Rova-T), in IDH mutant glioma. EXPERIMENTAL DESIGN/METHODS:We evaluated DLL3 expression by RNA using TCGA data and by immunohistochemistry in a discovery set of 63 gliomas and 20 non-tumor brain tissues and a validation set of 62 known IDH wildtype and mutant gliomas using a monoclonal anti-DLL3 antibody. Genotype was determined using a DNA methylation array classifier or by sequencing. The effect of Rova-T on patient-derived endogenous IDH mutant glioma tumorspheres was determined by cell viability assay. RESULTS:Compared to IDH wildtype glioblastoma, IDH mutant gliomas have significantly higher DLL3 RNA (P<1x10-15) and protein by immunohistochemistry (P=0.0014 and P<4.3x10-6 in the discovery and validation set, respectively). DLL3 immunostaining was intense and homogeneous in IDH mutant gliomas, retained in all recurrent tumors, and detected in only 1 of 20 non-tumor brains. Patient-derived IDH mutant glioma tumorspheres overexpressed DLL3 and were potently sensitive to Rova-T in an antigen-dependent manner. CONCLUSIONS:DLL3 is selectively and homogeneously expressed in IDH mutant gliomas and can be targeted with Rova-T in patient-derived IDH mutant glioma tumorspheres. Our findings are potentially immediately translatable and have implications for therapeutic strategies that exploit cell surface tumor-associated antigens.
PMID: 30397180
ISSN: 1078-0432
CID: 3455762

There is an exception to every rule-T2-FLAIR mismatch sign in gliomas

Johnson, Derek R; Kaufmann, Timothy J; Patel, Sohil H; Chi, Andrew S; Snuderl, Matija; Jain, Rajan
The T2-FLAIR mismatch sign, in which a low-grade glioma is hyperintense on T2-weighted MR and centrally hypointense on T2-weighted FLAIR MR, has been reported as 100% specific for IDH-mutant astrocytomas in several series. We report several cases of "false positive" T2-FLAIR mismatch sign occurring outside the context of IDH-mutant astrocytomas, predominantly in children or young adults with pediatric-type gliomas. These results suggest caution in the interpretation of the T2-FLAIR mismatch sign in the pediatric glioma population.
PMID: 30565056
ISSN: 1432-1920
CID: 3557052

Genetically distinct glioma stem-like cell xenografts established from paired glioblastoma samples harvested before and after molecularly targeted therapy

Tanaka, Shota; Luk, Samantha; Kiyokawa, Juri; Onozato, Maristela L; Iafrate, A John; Shah, Khalid; Martuza, Robert L; Rabkin, Samuel D; Batchelor, Tracy T; Cahill, Daniel P; Chi, Andrew S; Wakimoto, Hiroaki
Intratumoural heterogeneity underlies tumour escape from molecularly targeted therapy in glioblastoma. A cell-based model preserving the evolving molecular profiles of a tumour during treatment is key to understanding the recurrence mechanisms and development of strategies to overcome resistance. In this study, we established a matched pair of glioblastoma stem-like cell (GSC) cultures from patient glioblastoma samples before and after epidermal growth factor receptor (EGFR)-targeted therapy. A patient with recurrent glioblastoma (MGG70R) harboring focal, high-level EGFR amplification received the irreversible EGFR tyrosine kinase inhibitor dacomitinib. The tumour that subsequently recurred (MGG70RR) showed diploid EGFR, suggesting inhibitor-mediated elimination of EGFR-amplified tumour cells and propagation of EGFR non-amplified cell subpopulations. The MGG70R-GSC line established from MGG70R formed xenografts retaining EGFR amplification and EGFR overexpression, while MGG70RR-GSC established from MGG70RR generated tumours that lacked EGFR amplification and EGFR overexpression. MGG70R-GSC-derived intracranial xenografts were more proliferative than MGG70RR-GSC xenografts, which had upregulated mesenchymal markers, mirroring the pathological observation in the corresponding patient tumours. In vitro MGG70R-GSC was more sensitive to EGFR inhibitors than MGG70RR-GSC. Thus, these molecularly distinct GSC lines recapitulated the subpopulation alteration that occurred during glioblastoma evasion of targeted therapy, and offer a valuable model facilitating therapeutic development for recurrent glioblastoma.
PMCID:6333836
PMID: 30644426
ISSN: 2045-2322
CID: 3594762

Phase II Study of Iniparib with Concurrent Chemoradiation in Patients with Newly Diagnosed Glioblastoma

Blakeley, Jaishri O; Grossman, Stuart A; Chi, Andrew S; Mikkelsen, Tom; Rosenfeld, Myrna R; Ahluwalia, Manmeet S; Nabors, Louis B; Eichler, April; Garcia-Ribas, Ignacio; Desideri, Serena; Ye, Xiaobu
PURPOSE/OBJECTIVE:Iniparib is a purported prodrug causing cell death through intracellular conversion to nitro radical ions. We assessed the efficacy and safety of iniparib with standard radiation therapy (RT) and temozolomide (TMZ) in patients with newly diagnosed glioblastoma (GBM). EXPERIMENTAL DESIGN/METHODS:Adults meeting eligibility criteria were enrolled in this prospective, single arm, open-label multi-institution phase II trial with median overall survival (mOS) compared to a historical control as the primary objective. A safety run-in component of RT+TMZ+iniparib (n=5) was followed by an efficacy study (n=76) with the recommended phase II doses of iniparib (8.0 mg/kg IV twice/week with RT + daily TMZ followed by 8.6 mg/kg IV twice/week with 5/28 day TMZ). RESULTS:The median age of the 81 evaluable participants was 58 years (63% male). Baseline KPS was ≥80% in 87% of participants. The mOS was 22 months (95%CI: 17-24) and the hazard rate was 0.44 (95%CI: 0.35-0.55) per-person year of follow-up. The 2 and 3 year survival rates were 38% and 25%, respectively. Treatment-related grade 3 adverse events (AE) occurred in 27% of patients; nine patients had AEs requiring drug discontinuation including: infusion-related reaction, rash, gastritis, increased liver enzymes, and thrombocytopenia. CONCLUSIONS:Iniparib is well tolerated with RT and TMZ in patients with newly diagnosed GBM at up to 17.2mg/kg weekly. The primary objective of improved mOS compared with historical a control was met, indicating potential antitumor activity of iniparib in this setting. Dosing optimization (frequency and sequence) is needed prior to additional efficacy studies.
PMID: 30131387
ISSN: 1078-0432
CID: 3245842

Plasma cell-free circulating tumor DNA (ctDNA) detection in longitudinally followed glioblastoma patients using TERT promoter mutation-specific droplet digital PCR assays

Cordova, Christine; Syeda, Mahrukh M; Corless, Broderick; Wiggins, Jennifer M; Patel, Amie; Kurz, Sylvia Christine; Delara, Malcolm; Sawaged, Zacharia; Utate, Minerva; Placantonakis, Dimitris; Golfinos, John; Schafrick, Jessica; Silverman, Joshua Seth; Jain, Rajan; Snuderl, Matija; Zagzag, David; Shao, Yongzhao; Karlin-Neumann, George Alan; Polsky, David; Chi, Andrew S
ORIGINAL:0014231
ISSN: 1527-7755
CID: 4032352

NONINVASIVE PERFUSION IMAGING BIOMARKER OF MALIGNANT GENOTYPE IN ISOCITRATE DEHYDROGENASE MUTANT GLIOMAS [Meeting Abstract]

Mureb, Monica; Jain, Rajan; Poisson, Laila; Littig, Ingrid Aguiar; Neto, Lucidio Nunes; Wu, Chih-Chin; Ng, Victor; Patel, Sohil; Patel, Seema; Serrano, Jonathan; Kurz, Sylvia; Cahill, Daniel; Bendszus, Martin; von Deimling, Andreas; Placantonakis, Dimitris; Golfinos, John; Kickingereder, Philipp; Snuderl, Matija; Chi, Andrew
ISI:000509478703153
ISSN: 1522-8517
CID: 4530372

Probing tumor microenvironment in patients with newly diagnosed glioblastoma during chemoradiation and adjuvant temozolomide with functional MRI

Ina Ly, K; Vakulenko-Lagun, Bella; Emblem, Kyrre E; Ou, Yangming; Da, Xiao; Betensky, Rebecca A; Kalpathy-Cramer, Jayashree; Duda, Dan G; Jain, Rakesh K; Chi, Andrew S; Plotkin, Scott R; Batchelor, Tracy T; Sorensen, Gregory; Rosen, Bruce R; Gerstner, Elizabeth R
Functional MRI may identify critical windows of opportunity for drug delivery and distinguish between early treatment responders and non-responders. Using diffusion-weighted, dynamic contrast-enhanced, and dynamic susceptibility contrast MRI, as well as pro-angiogenic and pro-inflammatory blood markers, we prospectively studied the physiologic tumor-related changes in fourteen newly diagnosed glioblastoma patients during standard therapy. 153 MRI scans and blood collection were performed before chemoradiation (baseline), weekly during chemoradiation (week 1-6), monthly before each cycle of adjuvant temozolomide (pre-C1-C6), and after cycle 6. The apparent diffusion coefficient, volume transfer coefficient (Ktrans), and relative cerebral blood volume (rCBV) and flow (rCBF) were calculated within the tumor and edema regions and compared to baseline. Cox regression analysis was used to assess the effect of clinical variables, imaging, and blood markers on progression-free (PFS) and overall survival (OS). After controlling for additional covariates, high baseline rCBV and rCBF within the edema region were associated with worse PFS (microvessel rCBF: HR = 7.849, p = 0.044; panvessel rCBV: HR = 3.763, p = 0.032; panvessel rCBF: HR = 3.984; p = 0.049). The same applied to high week 5 and pre-C1 Ktrans within the tumor region (week 5 Ktrans: HR = 1.038, p = 0.003; pre-C1 Ktrans: HR = 1.029, p = 0.004). Elevated week 6 VEGF levels were associated with worse OS (HR = 1.034; p = 0.004). Our findings suggest a role for rCBV and rCBF at baseline and Ktrans and VEGF levels during treatment as markers of response. Functional imaging changes can differ substantially between tumor and edema regions, highlighting the variable biologic and vascular state of tumor microenvironment during therapy.
PMID: 30459364
ISSN: 2045-2322
CID: 3480592