Try a new search

Format these results:

Searched for:

person:sulmae01

in-biosketch:true

Total Results:

264


Glioblastoma Clinical Trials: Current Landscape and Opportunities for Improvement

Bagley, Stephen J; Kothari, Shawn; Rahman, Rifaquat; Lee, Eudocia Q; Dunn, Gavin P; Galanis, Evanthia; Chang, Susan M; Nabors, Louis Burt; Ahluwalia, Manmeet S; Stupp, Roger; Mehta, Minesh P; Reardon, David A; Grossman, Stuart A; Sulman, Erik P; Sampson, John H; Khagi, Simon; Weller, Michael; Cloughesy, Timothy F; Wen, Patrick Y; Khasraw, Mustafa
Therapeutic advances for glioblastoma have been minimal over the past 2 decades. In light of the multitude of recent phase III trials that have failed to meet their primary endpoints following promising preclinical and early-phase programs, a Society for Neuro-Oncology Think Tank was held in November 2020 to prioritize areas for improvement in the conduct of glioblastoma clinical trials. Here, we review the literature, identify challenges related to clinical trial eligibility criteria and trial design in glioblastoma, and provide recommendations from the Think Tank. In addition, we provide a data-driven context with which to frame this discussion by analyzing key study design features of adult glioblastoma clinical trials listed on ClinicalTrials.gov as "recruiting" or "not yet recruiting" as of February 2021.
PMID: 34561269
ISSN: 1557-3265
CID: 5178432

Update on Radiation Therapy for Central Nervous System Tumors

Rahman, Rifaquat; Sulman, Erik; Haas-Kogan, Daphne; Cagney, Daniel N
Radiation therapy has long been a critical modality of treatment of patients with central nervous system tumors, including primary brain tumors, brain metastases, and meningiomas. Advances in radiation technology and delivery have allowed for more precise treatment to optimize patient outcomes and minimize toxicities. Improved understanding of the molecular underpinnings of brain tumors and normal brain tissue response to radiation will allow for continued refinement of radiation treatment approaches to improve clinical outcomes for brain tumor patients. With continued advances in precision and delivery, radiation therapy will continue to be an important modality to achieve optimal outcomes of brain tumor patients.
PMID: 34711456
ISSN: 1558-1977
CID: 5042732

Author Correction: Atrx inactivation drives disease-defining phenotypes in glioma cells of origin through global epigenomic remodeling

Danussi, Carla; Bose, Promita; Parthasarathy, Prasanna T; Silberman, Pedro C; Van Arnam, John S; Vitucci, Mark; Tang, Oliver Y; Heguy, Adriana; Wang, Yuxiang; Chan, Timothy A; Riggins, Gregory J; Sulman, Erik P; Lang, Frederick F; Creighton, Chad J; Deneen, Benjamin; Miller, C Ryan; Picketts, David J; Kannan, Kasthuri; Huse, Jason T
PMID: 34987156
ISSN: 2041-1723
CID: 5107202

Germline polymorphisms in MGMT associated with temozolomide-related myelotoxicity risk in patients with glioblastoma treated on NRG Oncology/RTOG 0825

Scheurer, Michael E; Zhou, Renke; Gilbert, Mark R; Bondy, Melissa L; Sulman, Erik P; Yuan, Ying; Liu, Yanhong; Vera, Elizabeth; Wendland, Merideth M; Youssef, Emad F; Stieber, Volker W; Komaki, Ritsuko R; Flickinger, John C; Kenyon, Lawrence C; Robins, H Ian; Hunter, Grant K; Crocker, Ian R; Chao, Samuel T; Pugh, Stephanie L; Armstrong, Terri S
Background/UNASSIGNED:We sought to identify clinical and genetic predictors of temozolomide-related myelotoxicity among patients receiving therapy for glioblastoma. Methods/UNASSIGNED:= 591) receiving therapy on NRG Oncology/RTOG 0825 were included in the analysis. Cases were patients with severe myelotoxicity (grade 3 and higher leukopenia, neutropenia, and/or thrombocytopenia); controls were patients without such toxicity. A risk-prediction model was built and cross-validated by logistic regression using only clinical variables and extended using polymorphisms associated with myelotoxicity. Results/UNASSIGNED:had better prediction (AUC = 0.827). Receiving combination chemotherapy (OR, 1.82; 95% CI, 1.02-3.27) and being female (OR, 4.45; 95% CI, 2.45-8.08) significantly increased myelotoxicity risk. For each additional minor allele in the polymorphisms, the risk increased by 64% (OR, 1.64; 95% CI, 1.43-1.89). Conclusions/UNASSIGNED: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.
PMCID:9587696
PMID: 36299794
ISSN: 2632-2498
CID: 5359552

Clinical value of DNA methylation in practice: A prospective molecular neuropathology study [Meeting Abstract]

Galbraith, Kristyn; Shen, Guomiao; Serrano, Jonathan; Vasudevaraja, Varshini; Tran, Ivy; Movahed-Ezazi, Misha; Harter, David; Hidalgo, Eveline; Wisoff, Jeffrey; Orringer, Daniel; Placantonakis, Dimitris; Gardner, Sharon; William, Christopher; Zagzag, David; Allen, Jeffrey; Sulman, Erik; Golfinos, John; Snuderl, Matija
ISI:000798368400125
ISSN: 0022-3069
CID: 5244322

PDPN marks a subset of aggressive and radiation-resistant glioblastoma cells

Modrek, Aram S; Eskilsson, Eskil; Ezhilarasan, Ravesanker; Wang, Qianghu; Goodman, Lindsey D; Ding, Yingwen; Zhang, Ze-Yan; Bhat, Krishna P L; Le, Thanh-Thuy T; Barthel, Floris P; Tang, Ming; Yang, Jie; Long, Lihong; Gumin, Joy; Lang, Frederick F; Verhaak, Roel G W; Aldape, Kenneth D; Sulman, Erik P
Treatment-resistant glioma stem cells are thought to propagate and drive growth of malignant gliomas, but their markers and our ability to target them specifically are not well understood. We demonstrate that podoplanin (PDPN) expression is an independent prognostic marker in gliomas across multiple independent patient cohorts comprising both high- and low-grade gliomas. Knockdown of PDPN radiosensitized glioma cell lines and glioma-stem-like cells (GSCs). Clonogenic assays and xenograft experiments revealed that PDPN expression was associated with radiotherapy resistance and tumor aggressiveness. We further demonstrate that knockdown of PDPN in GSCs in vivo is sufficient to improve overall survival in an intracranial xenograft mouse model. PDPN therefore identifies a subset of aggressive, treatment-resistant glioma cells responsible for radiation resistance and may serve as a novel therapeutic target.
PMCID:9434399
PMID: 36059614
ISSN: 2234-943x
CID: 5336872

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

DNA damage drives DNA methylation and 3D chromatin organization alterations in glioblastoma [Meeting Abstract]

Modrek, Aram S.; Do, Catherine; Zhang, Zeyan; Deng, Yingwen; Karp, Jerome; Ezhilarasan, Ravesanker; Cova, Giulia; Snuderl, Matija; Tsirigos, Aristotelis; Skok, Jane; Sulman, Erik P.
ISI:000892509507561
ISSN: 0008-5472
CID: 5526672

ADAPTIVE RESPONSES TO GENOME-WIDE DNA DAMAGE RESULT IN TOPOLOGIC GENOME REORGANIZATION IN GLIOBLASTOMA [Meeting Abstract]

Modrek, Aram; Do, Catherine; Zhang, Zeyan; Deng, Yingwen; Karp, Jerome; Ezhilarasan, Ravesanker; Valor, Belen; Cova, Giulia; Jafari, Matiar; Snuderl, Matija; Tsirigos, Aristotelis; Skok, Jane; Sulman, Erik
ISI:000888571000458
ISSN: 1522-8517
CID: 5526662

Treatment for Brain Metastases: ASCO-SNO-ASTRO Guideline

Vogelbaum, Michael A; Brown, Paul D; Messersmith, Hans; Brastianos, Priscilla K; Burri, Stuart; Cahill, Dan; Dunn, Ian F; Gaspar, Laurie E; Gatson, Na Tosha N; Gondi, Vinai; Jordan, Justin T; Lassman, Andrew B; Maues, Julia; Mohile, Nimish; Redjal, Navid; Stevens, Glen; Sulman, Erik; van den Bent, Martin; Wallace, H James; Weinberg, Jeffrey S; Zadeh, Gelareh; Schiff, David
PURPOSE/OBJECTIVE:To provide guidance to clinicians regarding therapy for patients with brain metastases from solid tumors. METHODS:ASCO convened an Expert Panel and conducted a systematic review of the literature. RESULTS:Thirty-two randomized trials published in 2008 or later met eligibility criteria and form the primary evidentiary base. RECOMMENDATIONS/CONCLUSIONS:Surgery is a reasonable option for patients with brain metastases. Patients with large tumors with mass effect are more likely to benefit than those with multiple brain metastases and/or uncontrolled systemic disease. Patients with symptomatic brain metastases should receive local therapy regardless of the systemic therapy used. For patients with asymptomatic brain metastases, local therapy should not be deferred unless deferral is specifically recommended in this guideline. The decision to defer local therapy should be based on a multidisciplinary discussion of the potential benefits and harms that the patient may experience. Several regimens were recommended for non-small-cell lung cancer, breast cancer, and melanoma. For patients with asymptomatic brain metastases and no systemic therapy options, stereotactic radiosurgery (SRS) alone should be offered to patients with one to four unresected brain metastases, excluding small-cell lung carcinoma. SRS alone to the surgical cavity should be offered to patients with one to two resected brain metastases. SRS, whole brain radiation therapy, or their combination are reasonable options for other patients. Memantine and hippocampal avoidance should be offered to patients who receive whole brain radiation therapy and have no hippocampal lesions and 4 months or more expected survival. Patients with asymptomatic brain metastases with either Karnofsky Performance Status ≤ 50 or Karnofsky Performance Status < 70 with no systemic therapy options do not derive benefit from radiation therapy.Additional information is available at www.asco.org/neurooncology-guidelines.
PMID: 34932393
ISSN: 1527-7755
CID: 5108772