Try a new search

Format these results:

Searched for:

person:eisels01

in-biosketch:true

Total Results:

48


Evaluation of the SSTR2-targeted radiopharmaceutical 177Lu-DOTATATE and SSTR2-specific 68Ga-DOTATATE PET as imaging biomarker in patients with intracranial meningioma

Kurz, Sylvia C; Zan, Elcin; Cordova, Christine; Troxel, Andrea B; Barbaro, Marissa; Silverman, Joshua S; Snuderl, Matija; Zagzag, David; Kondziolka, Douglas; Golfinos, John G; Chi, Andrew S; Sulman, Erik P
BACKGROUND:There are no effective medical therapies for patients with meningioma who progress beyond surgical and radiotherapeutic interventions. Somatostatin receptor Type 2 (SSTR2) represents a promising treatment target in meningiomas. In this multicenter, single-arm phase II clinical study (NCT03971461), the SSTR2-targeting radiopharmaceutical 177Lu-DOTATATE is evaluated for its feasibility, safety, and therapeutic efficacy in these patients. PATIENTS AND METHODS/METHODS:Adult patients with progressive intracranial meningiomas received 177Lu-DOTATATE at a dose of 7.4 GBq (200 mCi) every eight weeks for four cycles. 68Ga-DOTATATE PET-MRI was performed before and six months after begin of treatment. The primary endpoint was progression-free survival (PFS) at 6 months (PFS-6). Secondary endpoints were safety and tolerability, overall survival (OS) at 12 months (OS-12), median PFS, and median OS. RESULTS:Fourteen patients (F=11, M=3) with progressive meningiomas (WHO 1=3, 2=10, 3=1) were enrolled. Median age was 63.1 (range 49.7-78) years. All patients previously underwent tumor resection and at least one course of radiation. Treatment with 177Lu-DOTATATE was well tolerated. Seven patients (50%) achieved PFS-6. Best radiographic response by modified Macdonald criteria was stable disease (SD) in all seven patients. A >25% reduction in 68Ga-DOTATATE (PET) was observed in five meningiomas and two patients. In one lesion, this corresponded to >50% reduction in bidirectional tumor measurements (MRI). CONCLUSIONS:Treatment with 177Lu-DOTATATE was well tolerated. The predefined PFS-6 threshold was met in this interim analysis, thereby allowing this multicenter clinical trial to continue enrollment. 68Ga-DOTATATE PET may be a useful imaging biomarker to assess therapeutic outcome in patients with meningioma.
PMID: 38048045
ISSN: 1557-3265
CID: 5595302

ONC201 (Dordaviprone) in Recurrent H3 K27M-Mutant Diffuse Midline Glioma

Arrillaga-Romany, Isabel; Gardner, Sharon L; Odia, Yazmin; Aguilera, Dolly; Allen, Joshua E; Batchelor, Tracy; Butowski, Nicholas; Chen, Clark; Cloughesy, Timothy; Cluster, Andrew; de Groot, John; Dixit, Karan S; Graber, Jerome J; Haggiagi, Aya M; Harrison, Rebecca A; Kheradpour, Albert; Kilburn, Lindsay; Kurz, Sylvia C; Lu, Guangrong; MacDonald, Tobey J; Mehta, Minesh; Melemed, Allen S; Nghiemphu, Phioanh Leia; Ramage, Samuel C; Shonka, Nicole; Sumrall, Ashley; Tarapore, Rohinton; Taylor, Lynne; Umemura, Yoshie; Wen, Patrick Y
PURPOSE/OBJECTIVE:Histone 3 (H3) K27M-mutant diffuse midline glioma (DMG) has a dismal prognosis with no established effective therapy beyond radiation. This integrated analysis evaluated single-agent ONC201 (dordaviprone), a first-in-class imipridone, in recurrent H3 K27M-mutant DMG. METHODS:Fifty patients (pediatric, n = 4; adult, n = 46) with recurrent H3 K27M-mutant DMG who received oral ONC201 monotherapy in four clinical trials or one expanded access protocol were included. Eligible patients had measurable disease by Response Assessment in Neuro-Oncology (RANO) high-grade glioma (HGG) criteria and performance score (PS) ≥60 and were ≥90 days from radiation; pontine and spinal tumors were ineligible. The primary end point was overall response rate (ORR) by RANO-HGG criteria. Secondary end points included duration of response (DOR), time to response (TTR), corticosteroid response, PS response, and ORR by RANO low-grade glioma (LGG) criteria. Radiographic end points were assessed by dual-reader, blinded independent central review. RESULTS:The ORR (RANO-HGG) was 20.0% (95% CI, 10.0 to 33.7). The median TTR was 8.3 months (range, 1.9-15.9); the median DOR was 11.2 months (95% CI, 3.8 to not reached). The ORR by combined RANO-HGG/LGG criteria was 30.0% (95% CI, 17.9 to 44.6). A ≥50% corticosteroid dose reduction occurred in 7 of 15 evaluable patients (46.7% [95% CI, 21.3 to 73.4]); PS improvement occurred in 6 of 34 evaluable patients (20.6% [95% CI, 8.7 to 37.9]). Grade 3 treatment-related treatment-emergent adverse events (TR-TEAEs) occurred in 20.0% of patients; the most common was fatigue (n = 5; 10%); no grade 4 TR-TEAEs, deaths, or discontinuations occurred. CONCLUSION/CONCLUSIONS:ONC201 monotherapy was well tolerated and exhibited durable and clinically meaningful efficacy in recurrent H3 K27M-mutant DMG.
PMID: 38335473
ISSN: 1527-7755
CID: 5632032

Clinical utility of whole-genome DNA methylation profiling as a primary molecular diagnostic assay for central nervous system tumors-A prospective study and guidelines for clinical testing

Galbraith, Kristyn; Vasudevaraja, Varshini; Serrano, Jonathan; Shen, Guomiao; Tran, Ivy; Abdallat, Nancy; Wen, Mandisa; Patel, Seema; Movahed-Ezazi, Misha; Faustin, Arline; Spino-Keeton, Marissa; Roberts, Leah Geiser; Maloku, Ekrem; Drexler, Steven A; Liechty, Benjamin L; Pisapia, David; Krasnozhen-Ratush, Olga; Rosenblum, Marc; Shroff, Seema; Boué, Daniel R; Davidson, Christian; Mao, Qinwen; Suchi, Mariko; North, Paula; Hopp, Amanda; Segura, Annette; Jarzembowski, Jason A; Parsons, Lauren; Johnson, Mahlon D; Mobley, Bret; Samore, Wesley; McGuone, Declan; Gopal, Pallavi P; Canoll, Peter D; Horbinski, Craig; Fullmer, Joseph M; Farooqui, Midhat S; Gokden, Murat; Wadhwani, Nitin R; Richardson, Timothy E; Umphlett, Melissa; Tsankova, Nadejda M; DeWitt, John C; Sen, Chandra; Placantonakis, Dimitris G; Pacione, Donato; Wisoff, Jeffrey H; Teresa Hidalgo, Eveline; Harter, David; William, Christopher M; Cordova, Christine; Kurz, Sylvia C; Barbaro, Marissa; Orringer, Daniel A; Karajannis, Matthias A; Sulman, Erik P; Gardner, Sharon L; Zagzag, David; Tsirigos, Aristotelis; Allen, Jeffrey C; Golfinos, John G; Snuderl, Matija
BACKGROUND/UNASSIGNED:Central nervous system (CNS) cancer is the 10th leading cause of cancer-associated deaths for adults, but the leading cause in pediatric patients and young adults. The variety and complexity of histologic subtypes can lead to diagnostic errors. DNA methylation is an epigenetic modification that provides a tumor type-specific signature that can be used for diagnosis. METHODS/UNASSIGNED:We performed a prospective study using DNA methylation analysis as a primary diagnostic method for 1921 brain tumors. All tumors received a pathology diagnosis and profiling by whole genome DNA methylation, followed by next-generation DNA and RNA sequencing. Results were stratified by concordance between DNA methylation and histopathology, establishing diagnostic utility. RESULTS/UNASSIGNED:Of the 1602 cases with a World Health Organization histologic diagnosis, DNA methylation identified a diagnostic mismatch in 225 cases (14%), 78 cases (5%) did not classify with any class, and in an additional 110 (7%) cases DNA methylation confirmed the diagnosis and provided prognostic information. Of 319 cases carrying 195 different descriptive histologic diagnoses, DNA methylation provided a definitive diagnosis in 273 (86%) cases, separated them into 55 methylation classes, and changed the grading in 58 (18%) cases. CONCLUSIONS/UNASSIGNED:DNA methylation analysis is a robust method to diagnose primary CNS tumors, improving diagnostic accuracy, decreasing diagnostic errors and inconclusive diagnoses, and providing prognostic subclassification. This study provides a framework for inclusion of DNA methylation profiling as a primary molecular diagnostic test into professional guidelines for CNS tumors. The benefits include increased diagnostic accuracy, improved patient management, and refinements in clinical trial design.
PMCID:10355794
PMID: 37476329
ISSN: 2632-2498
CID: 5536102

Activity of Adagrasib (MRTX849) in Brain Metastases: Preclinical Models and Clinical Data From Patients With KRASG12C-Mutant Non-Small Cell Lung Cancer

Sabari, Joshua K; Velcheti, Vamsidhar; Shimizu, Kazuhide; Strickland, Matthew R; Heist, Rebecca S; Singh, Mohini; Nayyar, Naema; Giobbie-Hurder, Anita; Digumarthy, Subba R; Gainor, Justin F; Rajan, Anant P; Nieblas-Bedolla, Edwin; Burns, Aaron C; Hallin, Jill; Olson, Peter; Christensen, James G; Kurz, Sylvia C; Brastianos, Priscilla K; Wakimoto, Hiroaki
PURPOSE/OBJECTIVE:Patients with KRAS-mutant non-small cell lung cancer (NSCLC) with brain metastases (BM) have a poor prognosis. Adagrasib (MRTX849), a potent oral small molecule KRASG12C inhibitor, irreversibly and selectively binds KRASG12C, locking it in its inactive state. Adagrasib has been optimized for favorable pharmacokinetic (PK) properties, including long half-life (~24 hours), extensive tissue distribution, dose-dependent PK, and central nervous system penetration, however BM-specific anti-tumor activity of KRASG12C inhibitors remains to be fully characterized. EXPERIMENTAL DESIGN/METHODS:A retrospective database query identified patients with KRAS-mutant NSCLC to understand their propensity to develop BM. Preclinical studies assessed physiochemical and PK properties of adagrasib. Mice bearing intracranial KRASG12C-mutant NSCLC xenografts (LU99-Luc/H23-Luc/LU65-Luc) were treated with clinically relevant adagrasib doses and levels of adagrasib in plasma, cerebrospinal fluid (CSF), and brain were determined along with anti-tumor activity. Preliminary clinical data were collected from 2 patients with NSCLC with untreated BM who had received adagrasib 600 mg BID in the Phase 1b cohort of the KRYSTAL-1 trial; CSF was collected, adagrasib concentrations measured, and anti-tumor activity in BM evaluated. RESULTS:Patients with KRAS-mutant NSCLC demonstrated high propensity to develop BM ({greater than or equal to}40%). Adagrasib penetrated into CSF and demonstrated tumor regression and extended survival in multiple preclinical BM models. In 2 patients with NSCLC and untreated BM, CSF concentrations of adagrasib measured above the target cellular IC50. Both patients demonstrated corresponding BM regression, supporting potential clinical activity of adagrasib in the brain. CONCLUSIONS:These data support further development of adagrasib in patients with KRASG12C-mutant NSCLC with untreated BM.
PMID: 35404402
ISSN: 1557-3265
CID: 5204272

Practical guidance for telemedicine use in neuro-oncology

Strowd, Roy E; Dunbar, Erin M; Gan, Hui K; Kurz, Sylvia; Jordan, Justin T; Mandel, Jacob J; Mohile, Nimish A; Nevel, Kathryn S; Taylor, Jennie W; Ullrich, Nicole J; Welch, Mary R; Wasilewski, Andrea; Mrugala, Maciej M
While the COVID-19 pandemic has catalyzed the expansion of telemedicine into nearly every specialty of medicine, few articles have summarized current practices and recommendations for integrating virtual care in the practice of neuro-oncology. This article identifies current telemedicine practice, provides practical guidance for conducting telemedicine visits, and generates recommendations for integrating virtual care into neuro-oncology practice. Practical aspects of telemedicine are summarized including when to use and not use telemedicine, how to conduct a virtual visit, who to include in the virtual encounter, unique aspects of telehealth in neuro-oncology, and emerging innovations.
PMCID:8965064
PMID: 35371525
ISSN: 2054-2577
CID: 5219512

Vascular disorders

Chapter by: Kurz, Sylvia C.; Rogers, Lisa R.
in: Handbook of Neuro-Oncology Neuroimaging by
[S.l.] : Elsevier, 2022
pp. 81-86
ISBN: 9780128229958
CID: 5501052

Efficacy of laser interstitial thermal therapy (LITT) for newly diagnosed and recurrent IDH wild-type glioblastoma

de Groot, John F; Kim, Albert H; Prabhu, Sujit; Rao, Ganesh; Laxton, Adrian W; Fecci, Peter E; O'Brien, Barbara J; Sloan, Andrew; Chiang, Veronica; Tatter, Stephen B; Mohammadi, Alireza M; Placantonakis, Dimitris G; Strowd, Roy E; Chen, Clark; Hadjipanayis, Constantinos; Khasraw, Mustafa; Sun, David; Piccioni, David; Sinicrope, Kaylyn D; Campian, Jian L; Kurz, Sylvia C; Williams, Brian; Smith, Kris; Tovar-Spinoza, Zulma; Leuthardt, Eric C
Background/UNASSIGNED:wild-type glioblastoma. Methods/UNASSIGNED:wild-type newly diagnosed and recurrent glioblastoma patients who were treated with laser ablation at 14 US centers between January 2016 and May 2019. Data were monitored for accuracy. Statistical analysis included individual variable summaries, multivariable differences in survival, and median survival numbers. Results/UNASSIGNED:promoter methylation, adjuvant chemotherapy within 12 weeks, and tumor volume <3 cc. Conclusions/UNASSIGNED:wild-type newly diagnosed glioblastoma is comparable to outcomes observed in other tumor resection studies when those patients undergo radiation and chemotherapy following LITT.
PMCID:9122789
PMID: 35611270
ISSN: 2632-2498
CID: 5283892

Association of hyperglycemia and molecular subclass on survival in IDH-wildtype glioblastoma

Liu, Elisa K; Vasudevaraja, Varshini; Sviderskiy, Vladislav O; Feng, Yang; Tran, Ivy; Serrano, Jonathan; Cordova, Christine; Kurz, Sylvia C; Golfinos, John G; Sulman, Erik P; Orringer, Daniel A; Placantonakis, Dimitris; Possemato, Richard; Snuderl, Matija
BACKGROUND/UNASSIGNED:Hyperglycemia has been associated with worse survival in glioblastoma. Attempts to lower glucose yielded mixed responses which could be due to molecularly distinct GBM subclasses. METHODS/UNASSIGNED:Clinical, laboratory, and molecular data on 89 IDH-wt GBMs profiled by clinical next-generation sequencing and treated with Stupp protocol were reviewed. IDH-wt GBMs were sub-classified into RTK I (Proneural), RTK II (Classical) and Mesenchymal subtypes using whole-genome DNA methylation. Average glucose was calculated by time-weighting glucose measurements between diagnosis and last follow-up. RESULTS/UNASSIGNED:= .02). Methylation clustering did not identify unique signatures associated with high or low glucose levels. Metabolomic analysis of 23 tumors showed minimal variation across metabolites without differences between molecular subclasses. CONCLUSION/UNASSIGNED:Higher average glucose values were associated with poorer OS in RTKI and Mesenchymal IDH-wt GBM, but not RTKII. There were no discernible epigenetic or metabolomic differences between tumors in different glucose environments, suggesting a potential survival benefit to lowering systemic glucose in selected molecular subtypes.
PMCID:9653172
PMID: 36382106
ISSN: 2632-2498
CID: 5384812

Functional connectivity of the default mode, dorsal attention and fronto-parietal executive control networks in glial tumor patients

Tordjman, Mickael; Madelin, Guillaume; Gupta, Pradeep Kumar; Cordova, Christine; Kurz, Sylvia C; Orringer, Daniel; Golfinos, John; Kondziolka, Douglas; Ge, Yulin; Wang, Ruoyu Luie; Lazar, Mariana; Jain, Rajan
PURPOSE/OBJECTIVE:Resting state functional magnetic resonance imaging (rsfMRI) is an emerging tool to explore the functional connectivity of different brain regions. We aimed to assess the disruption of functional connectivity of the Default Mode Network (DMN), Dorsal Attention Network(DAN) and Fronto-Parietal Network (FPN) in patients with glial tumors. METHODS:rsfMRI data acquired on 3T-MR of treatment-naive glioma patients prospectively recruited (2015-2019) and matched controls from the 1000 functional-connectomes-project were analyzed using the CONN functional toolbox. Seed-Based Connectivity Analysis (SBCA) and Independent Component Analysis (ICA, with 10 to 100 components) were performed to study reliably the three networks of interest. RESULTS:). For the FPN, increased connectivity was noted in the precuneus, posterior cingulate gyrus, and frontal cortex. No difference in the connectivity of the networks of interest was demonstrated between low- and high-grade gliomas, as well as when stratified by their IDH1-R132H (isocitrate dehydrogenase) mutation status. CONCLUSION/CONCLUSIONS:Altered functional connectivity is reliably found with SBCA and ICA in the DMN, DAN, and FPN in glioma patients, possibly explained by decreased connectivity between the cerebral hemispheres across the corpus callosum due to disruption of the connections.
PMID: 33528739
ISSN: 1573-7373
CID: 4789692

Dissecting the immunosuppressive tumor microenvironments in Glioblastoma-on-a-Chip for optimized PD-1 immunotherapy

Cui, Xin; Ma, Chao; Vasudevaraja, Varshini; Serrano, Jonathan; Tong, Jie; Peng, Yansong; Delorenzo, Michael; Shen, Guomiao; Frenster, Joshua; Morales, Renee-Tyler Tan; Qian, Weiyi; Tsirigos, Aristotelis; Chi, Andrew S; Jain, Rajan; Kurz, Sylvia C; Sulman, Erik P; Placantonakis, Dimitris G; Snuderl, Matija; Chen, Weiqiang
Programmed cell death protein-1 (PD-1) checkpoint immunotherapy efficacy remains unpredictable in glioblastoma (GBM) patients due to the genetic heterogeneity and immunosuppressive tumor microenvironments. Here, we report a microfluidics-based, patient-specific 'GBM-on-a-Chip' microphysiological system to dissect the heterogeneity of immunosuppressive tumor microenvironments and optimize anti-PD-1 immunotherapy for different GBM subtypes. Our clinical and experimental analyses demonstrated that molecularly distinct GBM subtypes have distinct epigenetic and immune signatures that may lead to different immunosuppressive mechanisms. The real-time analysis in GBM-on-a-Chip showed that mesenchymal GBM niche attracted low number of allogeneic CD154+CD8+ T-cells but abundant CD163+ tumor-associated macrophages (TAMs), and expressed elevated PD-1/PD-L1 immune checkpoints and TGF-β1, IL-10, and CSF-1 cytokines compared to proneural GBM. To enhance PD-1 inhibitor nivolumab efficacy, we co-administered a CSF-1R inhibitor BLZ945 to ablate CD163+ M2-TAMs and strengthened CD154+CD8+ T-cell functionality and GBM apoptosis on-chip. Our ex vivo patient-specific GBM-on-a-Chip provides an avenue for a personalized screening of immunotherapies for GBM patients.
PMID: 32909947
ISSN: 2050-084x
CID: 4589392