Searched for: in-biosketch:true
person:sulmae01
PRMT6 methylation of RCC1 regulates mitosis, tumorigenicity, and radiation response of glioblastoma stem cells
Huang, Tianzhi; Yang, Yongyong; Song, Xiao; Wan, Xuechao; Wu, Bingli; Sastry, Namratha; Horbinski, Craig M; Zeng, Chang; Tiek, Deanna; Goenka, Anshika; Liu, Fabao; Brennan, Cameron W; Kessler, John A; Stupp, Roger; Nakano, Ichiro; Sulman, Erik P; Nishikawa, Ryo; James, Charles David; Zhang, Wei; Xu, Wei; Hu, Bo; Cheng, Shi-Yuan
Aberrant cell proliferation is a hallmark of cancer, including glioblastoma (GBM). Here we report that protein arginine methyltransferase (PRMT) 6 activity is required for the proliferation, stem-like properties, and tumorigenicity of glioblastoma stem cells (GSCs), a subpopulation in GBM critical for malignancy. We identified a casein kinase 2 (CK2)-PRMT6-regulator of chromatin condensation 1 (RCC1) signaling axis whose activity is an important contributor to the stem-like properties and tumor biology of GSCs. CK2 phosphorylates and stabilizes PRMT6 through deubiquitylation, which promotes PRMT6 methylation of RCC1, which in turn is required for RCC1 association with chromatin and activation of RAN. Disruption of this pathway results in defects in mitosis. EPZ020411, a specific small-molecule inhibitor for PRMT6, suppresses RCC1 arginine methylation and improves the cytotoxic activity of radiotherapy against GSC brain tumor xenografts. This study identifies a CK2α-PRMT6-RCC1 signaling axis that can be therapeutically targeted in the treatment of GBM.
PMID: 33539787
ISSN: 1097-4164
CID: 4776592
Proton Therapy Reduces the Likelihood of High-Grade Radiation-Induced Lymphopenia in Glioblastoma Patients: Phase II Randomized Study of Protons vs. Photons
Mohan, Radhe; Liu, Amy Y; Brown, Paul D; Mahajan, Anita; Dinh, Jeffrey; Chung, Caroline; McAvoy, Sarah; McAleer, Mary Frances; Lin, Steven H; Li, Jing; Ghia, Amol J; Zhu, Cong; Sulman, Erik P; de Groot, John F; Heimberger, Amy B; McGovern, Susan L; Grassberger, Clemens; Shih, Helen; Ellsworth, Susannah; Grosshans, David R
BACKGROUND:We investigated differences in radiation-induced grade 3+ lymphopenia (G3+L), defined as an absolute lymphocyte count (ALC) nadir of <500 cells/µL, after proton therapy (PT) or X-ray (photon) therapy (XRT) for patients with glioblastoma (GBM). METHODS:Patients enrolled in a randomized phase II trial received PT (n=28) or XRT (n=56), concomitantly with temozolomide. ALC was measured before, weekly during, and within 1 month after radiotherapy. Whole-brain mean dose (WBMD) and brain dose-volume indices were extracted from planned dose distributions. Univariate and multivariate logistic regression analyses were used to identify independent predictive variables. The resulting model was evaluated using receiver operating characteristic (ROC) curve analysis. RESULTS:Rates of G3+L were lower in men (7/47 [15%] vs. women (19/37 [51%], P<0.001)), and for PT (4/28 [14%] vs. XRT (22/56 [39%], P=0.024)). G3+L was significantly associated with baseline ALC, WBMD, and brain volumes receiving 5‒40 Gy(RBE) or higher (i.e., V5 through V40). Stepwise multivariate logistic regression analysis identified being female (odds ratio [OR] 6.2, 05% confidence interval [CI] 1.95‒22.4, P=0.003), baseline ALC (OR 0.18, 95% CI 0.05‒0.51, P=0.003) and whole brain V20 (OR 1.07, 95% CI 1.03‒1.13, P=0.002) as the strongest predictors. ROC analysis yielded an area under the curve of 0.86 (95% CI 0.79-0.94) for the final G3+L prediction model. CONCLUSIONS:Sex, baseline ALC, and whole brain V20 were the strongest predictors of G3+L for patients with GBM treated with radiation and temozolomide. PT reduced brain volumes receiving low and intermediate doses and, consequently, reduced G3+L.
PMID: 32750703
ISSN: 1523-5866
CID: 4553972
The state of neuro-oncology during the COVID-19 pandemic: a worldwide assessment
Mrugala, Maciej M; Ostrom, Quinn T; Pressley, Shelley M; Taylor, Jennie W; Thomas, Alissa A; Wefel, Jeffrey S; Coven, Scott L; Acquaye, Alvina A; Haynes, Chas; Agnihotri, Sameer; Lim, Michael; Peters, Katherine B; Sulman, Erik P; Salcido, Joanne T; Butowski, Nicholas A; Hervey-Jumper, Shawn; Mansouri, Alireza; Oliver, Kathy R; Porter, Alyx B; Nassiri, Farshad; Schiff, David; Dunbar, Erin M; Hegi, Monika E; Armstrong, Terri S; van den Bent, Martin J; Chang, Susan M; Zadeh, Gelareh; Chheda, Milan G
Background/UNASSIGNED:It remains unknown how the COVID-19 pandemic has changed neuro-oncology clinical practice, training, and research efforts. Methods/UNASSIGNED:We performed an international survey of practitioners, scientists, and trainees from 21 neuro-oncology organizations across 6 continents, April 24-May 17, 2020. We assessed clinical practice and research environments, institutional preparedness and support, and perceived impact on patients. Results/UNASSIGNED:Of 582 respondents, 258 (45%) were US-based and 314 (55%) international. Ninety-four percent of participants reported changes in their clinical practice. Ninety-five percent of respondents converted at least some practice to telemedicine. Ten percent of practitioners felt the need to see patients in person, specifically because of billing concerns and pressure from their institutions. Sixty-seven percent of practitioners suspended enrollment for at least one clinical trial, including 62% suspending phase III trial enrollments. More than 50% believed neuro-oncology patients were at increased risk for COVID-19. Seventy-one percent of clinicians feared for their own personal safety or that of their families, specifically because of their clinical duties; 20% had inadequate personal protective equipment. While 69% reported increased stress, 44% received no psychosocial support from their institutions. Thirty-seven percent had salary reductions and 63% of researchers temporarily closed their laboratories. However, the pandemic created positive changes in perceived patient satisfaction, communication quality, and technology use to deliver care and mediate interactions with other practitioners. Conclusions/UNASSIGNED:The pandemic has changed treatment schedules and limited investigational treatment options. Institutional lack of support created clinician and researcher anxiety. Communication with patients was satisfactory. We make recommendations to guide clinical and scientific infrastructure moving forward and address the personal challenges of providers and researchers.
PMCID:7928618
PMID: 34007966
ISSN: 2632-2498
CID: 4877152
Phase II trial of proton therapy versus photon IMRT for GBM: secondary analysis comparison of progression-free survival between RANO versus clinical assessment
Al Feghali, Karine A; Randall, James W; Liu, Diane D; Wefel, Jeffrey S; Brown, Paul D; Grosshans, David R; McAvoy, Sarah A; Farhat, Maguy A; Li, Jing; McGovern, Susan L; McAleer, Mary F; Ghia, Amol J; Paulino, Arnold C; Sulman, Erik P; Penas-Prado, Marta; Wang, Jihong; de Groot, John; Heimberger, Amy B; Armstrong, Terri S; Gilbert, Mark R; Mahajan, Anita; Guha-Thakurta, Nandita; Chung, Caroline
Background/UNASSIGNED:This secondary image analysis of a randomized trial of proton radiotherapy (PT) versus photon intensity-modulated radiotherapy (IMRT) compares tumor progression based on clinical radiological assessment versus Response Assessment in Neuro-Oncology (RANO). Methods/UNASSIGNED:Eligible patients were enrolled in the randomized trial and had MR imaging at baseline and follow-up beyond 12 weeks from completion of radiotherapy. "Clinical progression" was based on a clinical radiology report of progression and/or change in treatment for progression. Results/UNASSIGNED:= .04) groups. There were 31 (46.3%) discrepant cases of which 17 had RANO progression more than a month prior to clinical progression, and 14 had progression by RANO but not clinical criteria. Conclusions/UNASSIGNED:Based on this secondary analysis of a trial of PT versus IMRT for glioblastoma, while no difference in PFS was noted relative to treatment technique, RANO criteria identified progression more often and earlier than clinical assessment. This highlights the disconnect between measures of tumor response in clinical trials versus clinical practice. With growing efforts to utilize real-world data and personalized treatment with timely adaptation, there is a growing need to improve the consistency of determining tumor progression within clinical trials and clinical practice.
PMCID:8320688
PMID: 34337411
ISSN: 2632-2498
CID: 5191122
World Cancer Day 2021 - Perspectives in Pediatric and Adult Neuro-Oncology
Sulman, Erik P; Eisenstat, David D
Significant advances in our understanding of the molecular genetics of pediatric and adult brain tumors and the resulting rapid expansion of clinical molecular neuropathology have led to improvements in diagnostic accuracy and identified new targets for therapy. Moreover, there have been major improvements in all facets of clinical care, including imaging, surgery, radiation and supportive care. In selected cohorts of patients, targeted and immunotherapies have resulted in improved patient outcomes. Furthermore, adaptations to clinical trial design have facilitated our study of new agents and other therapeutic innovations. However, considerable work remains to be done towards extending survival for all patients with primary brain tumors, especially children and adults with diffuse midline gliomas harboring Histone H3 K27 mutations and adults with isocitrate dehydrogenase (IDH) wild-type, O6 guanine DNA-methyltransferase gene (MGMT) promoter unmethylated high grade gliomas. In addition to improvements in therapy and care, access to the advances in technology, such as particle radiation or biologic therapy, neuroimaging and molecular diagnostics in both developing and developed countries is needed to improve the outcome of patients with brain tumors.
PMCID:8142853
PMID: 34041027
ISSN: 2234-943x
CID: 4888132
Multiomics profiling of primary lung cancers and distant metastases reveals immunosuppression as a common characteristic of tumor cells with metastatic plasticity
Lee, Won-Chul; Reuben, Alexandre; Hu, Xin; McGranahan, Nicholas; Chen, Runzhe; Jalali, Ali; Negrao, Marcelo V; Hubert, Shawna M; Tang, Chad; Wu, Chia-Chin; Lucas, Anthony San; Roh, Whijae; Suda, Kenichi; Kim, Jihye; Tan, Aik-Choon; Peng, David H; Lu, Wei; Tang, Ximing; Chow, Chi-Wan; Fujimoto, Junya; Behrens, Carmen; Kalhor, Neda; Fukumura, Kazutaka; Coyle, Marcus; Thornton, Rebecca; Gumbs, Curtis; Li, Jun; Wu, Chang-Jiun; Little, Latasha; Roarty, Emily; Song, Xingzhi; Lee, J Jack; Sulman, Erik P; Rao, Ganesh; Swisher, Stephen; Diao, Lixia; Wang, Jing; Heymach, John V; Huse, Jason T; Scheet, Paul; Wistuba, Ignacio I; Gibbons, Don L; Futreal, P Andrew; Zhang, Jianhua; Gomez, Daniel; Zhang, Jianjun
BACKGROUND:Metastasis is the primary cause of cancer mortality accounting for 90% of cancer deaths. Our understanding of the molecular mechanisms driving metastasis is rudimentary. RESULTS:We perform whole exome sequencing (WES), RNA sequencing, methylation microarray, and immunohistochemistry (IHC) on 8 pairs of non-small cell lung cancer (NSCLC) primary tumors and matched distant metastases. Furthermore, we analyze published WES data from 35 primary NSCLC and metastasis pairs, and transcriptomic data from 4 autopsy cases with metastatic NSCLC and one metastatic lung cancer mouse model. The majority of somatic mutations are shared between primary tumors and paired distant metastases although mutational signatures suggest different mutagenesis processes in play before and after metastatic spread. Subclonal analysis reveals evidence of monoclonal seeding in 41 of 42 patients. Pathway analysis of transcriptomic data reveals that downregulated pathways in metastases are mainly immune-related. Further deconvolution analysis reveals significantly lower infiltration of various immune cell types in metastases with the exception of CD4+ T cells and M2 macrophages. These results are in line with lower densities of immune cells and higher CD4/CD8 ratios in metastases shown by IHC. Analysis of transcriptomic data from autopsy cases and animal models confirms that immunosuppression is also present in extracranial metastases. Significantly higher somatic copy number aberration and allelic imbalance burdens are identified in metastases. CONCLUSIONS:Metastasis is a molecularly late event, and immunosuppression driven by different molecular events, including somatic copy number aberration, may be a common characteristic of tumors with metastatic plasticity.
PMCID:7640699
PMID: 33148332
ISSN: 1474-760x
CID: 4664222
Full automation of spinal stereotactic radiosurgery and stereotactic body radiation therapy treatment planning using Varian Eclipse scripting
Teruel, Jose R; Malin, Martha; Liu, Elisa K; McCarthy, Allison; Hu, Kenneth; Cooper, Bejamin T; Sulman, Erik P; Silverman, Joshua S; Barbee, David
The purpose of this feasibility study is to develop a fully automated procedure capable of generating treatment plans with multiple fractionation schemes to improve speed, robustness, and standardization of plan quality. A fully automated script was implemented for spinal stereotactic radiosurgery/stereotactic body radiation therapy (SRS/SBRT) plan generation using Eclipse v15.6 API. The script interface allows multiple dose/fractionation plan requests, planning target volume (PTV) expansions, as well as information regarding distance/overlap between spinal cord and targets to drive decision-making. For each requested plan, the script creates the course, plans, field arrangements, and automatically optimizes and calculates dose. The script was retrospectively applied to ten computed tomography (CT) scans of previous cervical, thoracic, and lumbar spine SBRT patients. Three plans were generated for each patient - simultaneous integrated boost (SIB) 1800/1600 cGy to gross tumor volume (GTV)/PTV in one fraction; SIB 2700/2100 cGy to GTV/PTV in three fractions; and 3000 cGy to PTV in five fractions. Plan complexity and deliverability patient-specific quality assurance (QA) was performed using ArcCHECK with an Exradin A16 chamber inserted. Dose objectives were met for all organs at risk (OARs) for each treatment plan. Median target coverage was GTV V100% = 87.3%, clinical target volume (CTV) V100% = 95.7% and PTV V100% = 88.0% for single fraction plans; GTV V100% = 95.6, CTV V100% = 99.6% and PTV V100% = 97.2% for three fraction plans; and GTV V100% = 99.6%, CTV V100% = 99.1% and PTV V100% = 97.2% for five fraction plans. All plans (n = 30) passed patient-specific QA (>90%) at 2%/2 mm global gamma. A16 chamber dose measured at isocenter agreed with planned dose within 3% for all cases. Automatic planning for spine SRS/SBRT through scripting increases efficiency, standardizes plan quality and approach, and provides a tool for target coverage comparison of different fractionation schemes without the need for additional resources.
PMID: 32965754
ISSN: 1526-9914
CID: 4605772
Genetic driver mutations introduced in identical cell-of-origin in murine glioblastoma reveal distinct immune landscapes but similar response to checkpoint blockade
Chen, Zhihong; Herting, Cameron J; Ross, James L; Gabanic, Ben; Puigdelloses Vallcorba, Montse; Szulzewsky, Frank; Wojciechowicz, Megan L; Cimino, Patrick J; Ezhilarasan, Ravesanker; Sulman, Erik P; Ying, Mingyao; Ma'ayan, Avi; Read, Renee D; Hambardzumyan, Dolores
Glioblastoma (GBM) is the most aggressive primary brain tumor. In addition to being genetically heterogeneous, GBMs are also immunologically heterogeneous. However, whether the differences in immune microenvironment are driven by genetic driver mutation is unexplored. By leveraging the versatile RCAS/tv-a somatic gene transfer system, we establish a mouse model for Classical GBM by introducing EGFRvIII expression in Nestin-positive neural stem/progenitor cells in adult mice. Along with our previously published Nf1-silenced and PDGFB-overexpressing models, we investigate the immune microenvironments of the three models of human GBM subtypes by unbiased multiplex profiling. We demonstrate that both the quantity and composition of the microenvironmental myeloid cells are dictated by the genetic driver mutations, closely mimicking what was observed in human GBM subtypes. These myeloid cells express high levels of the immune checkpoint protein PD-L1; however, PD-L1 targeted therapies alone or in combination with irradiation are unable to increase the survival time of tumor-bearing mice regardless of the driver mutations, reflecting the outcomes of recent human trials. Together, these results highlight the critical utility of immunocompetent mouse models for preclinical studies of GBM, making these models indispensable tools for understanding the resistance mechanisms of immune checkpoint blockade in GBM and immune cell-targeting drug discovery.
PMID: 32639068
ISSN: 1098-1136
CID: 4526912
Radiation with STAT3 blockade triggers dendritic cell-T cell interactions in the glioma microenvironment and therapeutic efficacy
Ott, Martina; Kassab, Cynthia; Marisetty, Anantha; Hashimoto, Yuuri; Wei, Jun; Zamler, Daniel B; Leu, Jia-Shiun; Tomaszowski, Karl-Heinz; Sabbagh, Aria; Fang, Dexing; Gupta, Pravesh; Priebe, Waldemar; Zielinski, Rafal; Burks, Jared K; Long, James P; Kong, Ling-Yuan; Fuller, Gregory N; de Groot, John F; Sulman, Erik P; Heimberger, Amy B
BACKGROUND:Patients with central nervous system (CNS) tumors are typically treated with radiation therapy, but this is not curative and results in the upregulation of phosphorylated signal transducer and activator of transcription 3 (p-STAT3), which drives invasion, angiogenesis, and immune suppression. Therefore, we investigated the combined effect of an inhibitor of STAT3 and whole-brain radiation therapy (WBRT) in a murine model of glioma. METHODS:C57BL/6 mice underwent intracerebral implantation of GL261 glioma cells, WBRT, and treatment with WP1066, a blood-brain barrier (BBB)-penetrant inhibitor of the STAT3 pathway, or the two in combination. The role of the immune system was evaluated using tumor rechallenge strategies, immune incompetent backgrounds, immunofluorescence, immune phenotyping of tumor-infiltrating immune cells (via flow cytometry), and nanostring gene expression analysis of 770 immune-related genes from immune cells, including those directly isolated from the tumor microenvironment. RESULTS:The combination of WP1066 and WBRT resulted in long-term survivors and enhanced median survival time relative to monotherapy in the GL261 glioma model (combination vs. control p<0.0001). Immunological memory appeared to be induced, because mice were protected during subsequent tumor rechallenge. The therapeutic effect of the combination was completely lost in immune incompetent animals. Nanostring analysis and immunofluorescence revealed immunological reprograming in the CNS tumor microenvironment specifically affecting dendritic-cell antigen presentation and T cell effector functions. CONCLUSION/CONCLUSIONS:This study indicates that the combination of STAT3 inhibition and WBRT enhances the therapeutic effect against gliomas in the CNS by inducing dendritic cell and T cell interactions in the CNS tumor.
PMID: 32605912
ISSN: 1078-0432
CID: 4504182
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