Searched for: in-biosketch:true
person:sulmae01
NEUROCOGNITIVE FUNCTION (NCF) OUTCOMES OF RTOG FOUNDATION 3508: A PHASE 3 TRIAL OF ABT-414 WITH CONCURRENT CHEMORADIATION AND ADJUVANT TEMOZOLOMIDE IN PATIENTS WITH EGFR-AMPLIFIED NEWLY DIAGNOSED GBM [Meeting Abstract]
Wefel, Jeffrey S.; Won, Minhee; Lassman, Andrew; Stern, Yaakov; Wang, Tony; Aldape, Kenneth; Armstrong, Terri; Vogelbaum, Michael; Sulman, Erik; Moazami, Golnaz; Macsai, Marian; Gilbert, Mark; Bain, Earle; Blot, Vincent; Gan, Hui; Preusser, Matthias; Ansell, Peter; Samanta, Suvajit; Kundu, Madan; Seidel, Clemens; de Vos, Filip; Hsu, Sigmund; Cardona, Andres; Lombardi, Giuseppe; Bentsion, Dmitry; Peterson, Richard; Gedye, Craig; Lebrun-Frenay, Christine; Wick, Antje; Pugh, Stephanie; Curran, Walter; Mehta, Minesh
ISI:000590061300218
ISSN: 1522-8517
CID: 4688112
Aberrant DNA Methylation Predicts Melanoma-Specific Survival in Patients with Acral Melanoma
Pradhan, Dinesh; Jour, George; Milton, Denái; Vasudevaraja, Varshini; Tetzlaff, Michael T; Nagarajan, Priyadharsini; Curry, Jonathan L; Ivan, Doina; Long, Lihong; Ding, Yingwen; Ezhilarasan, Ravesanker; Sulman, Erik P; Diab, Adi; Hwu, Wen-Jen; Prieto, Victor G; Torres-Cabala, Carlos Antonio; Aung, Phyu P
Acral melanoma (AM) is a rare, aggressive type of cutaneous melanoma (CM) with a distinct genetic profile. We aimed to identify a methylome signature distinguishing primary acral lentiginous melanoma (PALM) from primary non-lentiginous AM (NALM), metastatic ALM (MALM), primary non-acral CM (PCM), and acral nevus (AN). A total of 22 PALM, nine NALM, 10 MALM, nine PCM, and three AN were subjected to genome-wide methylation analysis using the Illumina Infinium Methylation EPIC array interrogating 866,562 CpG sites. A prominent finding was that the methylation profiles of PALM and NALM were distinct. Four of the genes most differentially methylated between PALM and NALM or MALM were HHEX, DIPK2A, NELFB, and TEF. However, when primary AMs (PALM + NALM) were compared with MALM, IFITM1 and SIK3 were the most differentially methylated, highlighting their pivotal role in the metastatic potential of AMs. Patients with NALM had significantly worse disease-specific survival (DSS) than patients with PALM. Aberrant methylation was significantly associated with aggressive clinicopathologic parameters and worse DSS. Our study emphasizes the importance of distinguishing the two epigenetically distinct subtypes of AM. We also identified novel epigenetic prognostic biomarkers that may serve to risk-stratify patients with AM and may be leveraged for the development of targeted therapies.
PMID: 31888295
ISSN: 2072-6694
CID: 4251252
Inhibition of Nuclear PTEN Tyrosine Phosphorylation Enhances Glioma Radiation Sensitivity through Attenuated DNA Repair
Ma, Jianhui; Benitez, Jorge A; Li, Jie; Miki, Shunichiro; Ponte de Albuquerque, Claudio; Galatro, Thais; Orellana, Laura; Zanca, Ciro; Reed, Rachel; Boyer, Antonia; Koga, Tomoyuki; Varki, Nissi M; Fenton, Tim R; Nagahashi Marie, Suely Kazue; Lindahl, Erik; Gahman, Timothy C; Shiau, Andrew K; Zhou, Huilin; DeGroot, John; Sulman, Erik P; Cavenee, Webster K; Kolodner, Richard D; Chen, Clark C; Furnari, Frank B
PMID: 31821785
ISSN: 1878-3686
CID: 4234392
Functional and topographic effects on DNA methylation in IDH1/2 mutant cancers
Bledea, Ramona; Vasudevaraja, Varshini; Patel, Seema; Stafford, James; Serrano, Jonathan; Esposito, Gianna; Tredwin, Lilian M; Goodman, Nina; Kloetgen, Andreas; Golfinos, John G; Zagzag, David; Weigelt, Britta; Iafrate, A John; Sulman, Erik P; Chi, Andrew S; Dogan, Snjezana; Reis-Filho, Jorge S; Chiang, Sarah; Placantonakis, Dimitris; Tsirigos, Aristotelis; Snuderl, Matija
IDH1/2 mutations are early drivers present in diverse human cancer types arising in various tissue sites. IDH1/2 mutation is known to induce a global hypermethylator phenotype. However, the effects on DNA methylation across IDH mutant cancers and functionally different genome regions, remain unknown. We analyzed DNA methylation data from IDH1/2 mutant acute myeloid leukemia, oligodendroglioma, astrocytoma, solid papillary breast carcinoma with reverse polarity, sinonasal undifferentiated carcinoma and cholangiocarcinoma, which clustered by their embryonal origin. Hypermethylated common probes affect predominantly gene bodies while promoters in IDH1/2 mutant cancers remain unmethylated. Enhancers showed global hypermethylation, however commonly hypomethylated enhancers were associated with tissue differentiation and cell fate determination. We demonstrate that some chromosomes, chromosomal arms and chromosomal regions are more affected by IDH1/2 mutations while others remain resistant to IDH1/2 mutation induced methylation changes. Therefore IDH1/2 mutations have different methylation effect on different parts of the genome, which may be regulated by different mechanisms.
PMID: 31727977
ISSN: 2045-2322
CID: 4185902
Molecular Profiling of Long-Term IDH-wildtype Glioblastoma Survivors
Burgenske, Danielle M; Yang, Jie; Decker, Paul A; Kollmeyer, Thomas M; Kosel, Matthew L; Mladek, Ann C; Caron, Alissa A; Vaubel, Rachael A; Gupta, Shiv K; Kitange, Gaspar J; Sicotte, Hugues; Youland, Ryan S; Remonde, Dioval; Voss, Jesse S; Fritcher, Emily G Barr; Kolsky, Kathryn L; Ida, Cristiane M; Meyer, Fredric B; Lachance, Daniel H; Parney, Ian J; Kipp, Benjamin R; Giannini, Caterina; Sulman, Erik P; Jenkins, Robert B; Eckel-Passow, Jeanette E; Sarkaria, Jann N
BACKGROUND:Glioblastoma (GBM) represents an aggressive cancer type with a median survival of only 14 months. With fewer than 5% of patients surviving five years, comprehensive profiling of these rare patients could elucidate prognostic biomarkers that may confer better patient outcomes. We utilized multiple molecular approaches to characterize the largest patient cohort of long-term IDH-wildtype GBM survivors (LTS) to date. METHODS:Retrospective analysis was performed on 49 archived formalin-fixed paraffin embedded tumor specimens from patients diagnosed with GBM at the Mayo Clinic between December 1995 and September 2013. These patient samples were subdivided into two groups based on survival (12 LTS, 37 short-term survivors (STS)) and subsequently examined by mutation sequencing, copy number analysis, methylation profiling, and gene expression. RESULTS:Of the 49 patients analyzed in this study, LTS were younger at diagnosis (p=0.016), more likely to be female (p=0.048), and MGMT promoter methylated (UniD, p=0.01). IDH-wildtype STS and LTS demonstrated classic GBM mutations and copy number changes. Pathway analysis of differentially expressed genes showed LTS enrichment for sphingomyelin metabolism, which has been linked to decreased GBM growth, invasion, and angiogenesis. STS enriched for DNA repair and cell cycle control networks. CONCLUSIONS:While our findings largely report remarkable similarity between these LTS and more typical STS, unique attributes were observed in regard to altered gene expression and pathway enrichment. These attributes may be valuable prognostic markers and are worth further examination. Importantly, this study also underscores the limitations of existing biomarkers and classification methods in predicting patient prognosis.
PMID: 31346613
ISSN: 1523-5866
CID: 3988222
Prospective trial of conventionally fractionated dose constraints for re-irradiation of primary brain tumors [Meeting Abstract]
McGovern, S; Luo, D; Johnson, M; Nguyen, K; Li, J; McAleer, M; Yeboa, D; Grosshans, D; Ghia, A; Chung, C; Thall, P; Sulman, E; Brown, P; Mahajan, A
PURPOSE/OBJECTIVE: Dose constraints for re-irradiation of recurrent primary brain tumors are not well-established, especially for treatment volumes too large for stereotactic radiotherapy. This prospective trial was performed to test dose constraints for conventionally-fractionated re-irradiation of recurrent primary brain tumors MATERIALS/METHODS: A singleinstitution, prospective trial of 21 adults with recurrent brain tumors was performed. Electronic dosimetry records from the first course of radiation (RT1) were obtained and deformed onto the simulation CT for the second course of radiation (RT2). Treatment plans for RT2 were developed that met protocol-assigned dose constraints for RT2 alone and the composite dose of RT1+RT2. Dose constraints were also based on histology and interval since RT1. The primary endpoint was the rate of symptomatic brain necrosis after RT2.
RESULT(S): Twenty one adults enrolled from March 2017 to May 2018. Twelve had glioblastoma, four had oligodendroglioma, two had anaplastic astrocytoma, and one each had choroid plexus papilloma, hemangiopericytoma, and pleomorphic xanthroastrocytoma (PXA). Twenty patients were treated with VMAT and one was treated with proton CSI. Median RT1-RT2 interval was 45 months (range, 9-141 months). Median RT2 dose was 42.8 Gy (range, 17.5-60 Gy). Median PTV volume was 208 cc (range, 7-1537 cc). Median imaging followup was 9 months (range, 1-20 months). Two months after RT2, the patient with PXA developed a trapped temporal horn adjacent to the RT2 treatment volume; pathology from emergent resection revealed necrotic brain tissue. The patient recovered fully and lived another 18 months until dying of disease progression. No other patient developed symptomatic radionecrosis. Median overall survival from RT2 for all patients was 11 months (range, 3-20 months).
CONCLUSION(S): Re-irradiation can be performed with conventionally fractionated schemes. Given the low rate of symptomatic radionecrosis, the dose constraints described here are a starting point for future studies of conventionally fractionated re-irradiation
EMBASE:631168219
ISSN: 1523-5866
CID: 4388202
A randomized, double-blind, placebocontrolled phase 3 trial of depatuxizumab mafodotin (ABT-414) in epidermal growth factor receptor (EGFR) amplified (AMP) newly diagnosed glioblastoma (NGBM) [Meeting Abstract]
Lassman, A; Pugh, S; Wang, T; Aldape, K; Gan, H; Preusser, M; Vogelbaum, M; Sulman, E; Won, M; Zhang, P; Moazami, G; Macsai, M; Gilbert, M; Bain, E; Blot, V; Ansell, P; Samanta, S; Kundu, M; Seidel, C; De, Vos F; Hsu, S; Cardona, A; Lombardi, G; Bentsion, D; Peterson, R; Gedye, C; Lebrun-Frenay, C; Wick, A; Curran, W; Mehta, M
BACKGROUND: Approximately 50% of nGBMs harbor EGFR-amp. Depatuxizumab mafodotin (depatux-m) is an antibody drug conjugate: a monoclonal antibody that binds activated EGFR (wild-type and EGFRvIII mutant) linked to a microtubule-inhibitor toxin. Pre-clinical and earlier clinical trials suggested efficacy.
METHOD(S): RTOGF 3508/AbbVie M13-813 (INTELLANCE-1, NCT02573324) was a phase 3 academic-industry collaboration (RTOG-Foundation, AbbVie). Eligible adults (KPS >= 70, EGFR-amp nGBM, centrally confirmed histology and biomarkers) were randomized 1:1 to radiotherapy (RT) and temozolomide and either depatux-m (2.0 mg/kg during RT, 1.25 mg/kg thereafter, q 14 days) or placebo, stratified by region of world, RPA class, MGMT methylation, and EGFRvIII mutation. Primary endpoint was overall survival (OS), with 640 patients planned for randomization; 441 events yielded 85% power to detect 25% reduction in hazard of death (HR 0.75), one-sided 2.5% level of significance by stratified weighted log-rank.
RESULT(S): 2229 patients were screened and 639 (median age 60, range 22-84; 394 men, 62%) randomized. Pre-specified interim analysis after 346 events (>= 75% required) found no OS improvement for depatux-m over placebo (median 18.9 vs. 18.7 months, HR 1.01, 95% CI 0.82-1.25, one-sided p= 0.63). Progression-free survival (PFS) trended toward depatux-m (median 8.0 vs. 6.3 months; HR 0.84, 95% CI 0.70-1.02), particularly among the ~50% with EGFRvIII mutation (median 8.3 vs. 5.9 months, HR 0.72, 95% CI 0.56-0.93) but without an OS improvement (median 19.8 vs. 18.2, HR=0.95, 95% CI 0.71-1.27). Ocular side effects (grade >= 1) occurred in 95% of depatux-m treated patients, 61% grade 3-4, causing 12% to discontinue, and were the most common treatment related adverse events.
CONCLUSION(S): Interim analysis demonstrated no OS benefit for treating EGFR-amp nGBM with depatux-m. PFS trended toward favoring depatux-m, particularly in the EGFRvIII harboring subgroup. No new important safety risks were identified. The trial was stopped for futility. Active patients are permitted to continue treatment
EMBASE:631169019
ISSN: 1523-5866
CID: 4388062
MEK inhibitors induces neuronal differentiation in EGFR amplified glioma stem like cells [Meeting Abstract]
Balasubramaniyan, V; Park, S; Piao, Y; Martinez, E; Dong, J; Mittal, S; Khan, S; Zhang, Z -Y; Sulman, E; DeGroot, J
The median survival for patients with glioblastoma (GBM) is 12-15 months highlighting the need for better therapeutic strategies for this deadly disease. Genomic and epigenomic sequencing analysis at the single cell level have identified multiple genomic aberrations as potential targets for therapeutic intervention in GBM. EGFR and PDGFR amplification are evident in nearly 40% and 12% of human GBM, respectively. Although the first and second-generation EGFR tyrosine kinase small molecule inhibitors failed to show long term therapeutic benefit in GBM patients, multiple factors such as incorrect patient selection, acquired resistance, and drug-target heterogeneity may all lead to clinical failure of targeted therapies. Although the multilevel genomic characterization of gliomas are increasing, the clinical translation of these findings is beginning to unravel. In this study, we attempted to correlate the genomic variations using an unbiased high throughput drug screen using primary glioma stem-like cell (GSCs) as our model system. An unbiased high-throughput screen utilizing our GSC models identified that glioblastoma cells harboring focal EGFR amplification are sensitive to mitogen-activated protein kinase (MEK) inhibitors. MEK inhibition induced apoptosis in EGFR amplified cells at low concentration. RNA sequence analysis of cells treated with MEK inhibitors revealed upregulation of genes related to neuronal differentiation and down regulation of MEK target genes in MEK sensitive glioma stem cells. Additionally, RNA sequencing of GSCs with acquired MEK inhibitor resistance demonstrated an upregulation of oncogenic transcription factor ETS Variant Gene 1 (ETV1) as a mediator of resistance. Overall our data suggest that the MEK inhibition in combination with ETV inhibitors could be a potential therapeutic target for a subset of GBM patients
EMBASE:631169154
ISSN: 1523-5866
CID: 4388012
Prospective phase II randomized trial comparing proton therapy vs. photon imrt for GBM: Secondary analysis comparison of progression free survival between rano vs. clinical and radiological assessment [Meeting Abstract]
Al, Feghali K; Randall, J; Wefel, J; Guha-Thakurta, N; Grosshans, D; Dibaj, S; McAvoy, S; Li, J; McGovern, S; McAleer, M; Ghia, A; Paulino, A; Sulman, E; Penas-Prado, M; Wang, J; De, Groot J; Heimberger, A; Armstrong, T; Gilbert, M; Mahajan, A; Brown, P; Chung, C
PURPOSE: To compare tumor progression based on clinical radiological assessment and on Response Assessment in Neuro-Oncology (RANO) criteria between GBM patients treated with proton radiotherapy (PT) vs. photon intensity modulated radiotherapy (IMRT).
METHOD(S): Eligible patients were enrolled on the described prospective phase II trial and had MR imaging at baseline and follow-up beyond 12 weeks from treatment completion. 'Clinical' progression was based on a radiology report of progression in combination with changes in treatment due to suspected disease progression. A single blinded observer applied RANO criteria to determine the RANO-based tumor progression.
RESULT(S): Of 90 enrolled patients, 66 were evaluable, with median follow-up of 19.8 (Range: 3.2-65.1) months; median of 22.6 months for PT (n=25) vs. 18.9 months for IMRT (n=41). Median time to progression (TTP) was 7.9 months based on clinical progression criteria (8.1 months IMRT, 6.3 months PT) and 7.2 months (7.3 months IMRT, 5.7 months PT) by RANO criteria (p=ns for all). Median 'clinical' progression-free survival (PFS) was 8.7 (Range: 6.4-11.1) months; 8.9 months IMRT vs. 8.7 months PT (p=0.065). Median RANO PFS was 8.3 (range, 5.8-11.6) months: 8.3 months IMRT vs. 6.9 months PT (p=0.226). There were 14 discrepant cases: 3 had progression based on 'clinical' but not RANO criteria, and 11 had progression based on RANO but not 'clinical' criteria.
CONCLUSION(S): Based on this secondary analysis of a randomized trial of PT vs. IMRT for GBM, there was no difference in tumor progression relative to treatment technique used. There was no statistical difference in PFS noted between clinical and RANO-based assessments, but RANO criteria identified progression more often than clinical assessment, and TTP was shortened with the use of RANO criteria alone. Further development of tumor assessment tools that improve consistency and accuracy of determining tumor progression are needed to guide therapeutic trials in GBM
EMBASE:631168483
ISSN: 1523-5866
CID: 4388142
Prospective phase II randomized trial comparing proton therapy vs. photon IMRT for newly diagnosed GBM: Secondary analysis comparison of gender and neutrophil-lymphocyte ratio (NLR) in GBM outcomes [Meeting Abstract]
Randall, J; Al, Feghali K; Wefel, J; Grosshans, D; Dibaj, S; Milton, D; McAvoy, S; Li, J; McGovern, S; McAleer, M; Ghia, A; Paulino, A; Sulman, E; Penas-Prado, M; Wang, J; DeGroot, J; Heimberger, A; Armstrong, T; Gilbert, M; Mahajan, A; Brown, P; Chung, C
BACKGROUND: While glioblastoma (GBM) is more prevalent in males, studies show that females with GBM tend to have longer overall survival (OS) than males. Pretreatment neutrophil to lymphocyte ratio (NLR) has also proven to be prognostic in GBM, with lower NLR having favorable outcomes. This secondary analysis of a prospective randomized trial of proton vs. photon intensity modulated radiotherapy aims to explore the interaction of gender and NLR on GBM outcomes.
METHOD(S): Analysis was performed on the full patient population. Kaplan-Meier methods estimated OS with censoring at last follow-up for those who were alive. Univariate (UVA) and multivariate (MVA)Cox proportional hazards models assessed predictors of OS.
RESULT(S): Of 90 patients, 77 were included (43 males; 34 females) with median age of 52 years (range: 26-82 years). Median OS was longer for females than males (30.7 vs 18.2 months, p=0.004). On UVA, patients with NLR below median value (NLR= 3.1) tended to have longer OS than those above median, though not meeting statistical significance (23.1 vs. 17.9 months, p=0.051). Difference in OS was statistically significant in females (OS 36.4 months NLR >median vs. 16.7 months NLR< median, p=0.002), but not in males (OS 17.8 months NLR >median vs. 19.1 NLR< median, p=0.95). MVA analysis was consistent, with female gender predicting reduced hazard ratio (HR) (0.28, p=0.034) and females with below median NLR showing a reduced HR over those with above median (0.28, p=0.005). Again, males did not benefit (HR 0.90, p=0.77).
CONCLUSION(S): Consistent with prior publications, females and all patients with lower pre-treatment NLR with newly diagnosed GBM had longer OS. However, combining these two factors revealed that the benefits from lower pre-treatment NLR were conferred only in females with no impact on males. This different impact of NLR between genders may suggest innate immune differences in gender during response to malignancy
EMBASE:631168178
ISSN: 1523-5866
CID: 4388212