Searched for: in-biosketch:true
person:snudem01
STRATEGIES FOR THE SURGICAL MANAGEMENT OF PEDIATRIC OPTIC PATHWAY GLIOMAS - EXPERIENCE WITH 100 PATIENTS [Meeting Abstract]
Hidalgo, Eveline Teresa; Kvint, Svetlana; Thomas, Cheddhi; Orrilac, Cordelia; North, Emily; Dastagirzada, Yosef; Snuderl, Matija; Wisoff, Jeffrey H.
ISI:000379749000377
ISSN: 1522-8517
CID: 2964242
Strategies for the surgical management of pediatric optic pathway gliomas-experience with 100 patients [Meeting Abstract]
Teresa, Hidalgo E; Kvint, S; Thomas, C; Orrilac, C; North, E; Dastagirzada, Y; Snuderl, M; Wisoff, J H
Introduction: Pediatric optic pathway gliomas (OPGs) are often considered benign, but can have detrimental effects on the quality of life, impair vision and are a potentially lethal disease. The aim of this study is to report the characteristics and outcomes of surgically treated pediatric OPGs and to identify candidates for different treatment strategies. Methods: Retrospective chart review of consecutive pediatric patients with surgically treated OPGs by a single surgeon at our institution from 1985-2015. Three treatment pathways were defined: (1) surgery without planned adjuvant therapy; (2) surgery with planned adjuvant therapy; and, (3) patients with prior treatment.Results: 100 patients-55 male and 45 female-were included in analysis. 8 patients had NF1. Pathology revealed pilocytic astrocytoma (45%), pilomyxoid astrocytoma (9%), ganglioglioma (3%), and unknown (35%). Radiologic location of the tumor was: hypothalamic 86%, involvement of only chiasm and/or tract in 12%. Median age at diagnosis was 4 years, median age at surgery was 6 years, and median time from diagnosis to surgery was 1 year. Pathway 1: 39 patients; median PFS 73+17 months; OS rate 74%; median follow-up 117 months. Pathway 2: 10 patients; median PFS 29+15 months; OS rate 70%; median follow-up 59 months. Pathway 3: 51 patients; median PFS 33+8 months; OS rate 80%; median follow-up was 73 months. Conclusion: The role of surgery in the treatment of pediatric OPGs depends on patient characteristics and tumor biology. With the adequate therapeutic strategy, long-term PFS and OS can be achieved
EMBASE:611934154
ISSN: 1523-5866
CID: 2258952
Endothelium-independent primitive myxoid vascularization creates invertebrate-like channels to maintain blood supply in optic gliomas [Meeting Abstract]
Snuderl, M; Zhang, G; Wu, P; Jennings, T; Shroff, S; Ortenzi, V; Jain, R; Cohen, B; Reidy, J; Dushay, M; Wisoff, J; Harter, D; Karajannis, M; Fenyo, D; Neubert, T; Zagzag, D
INTRODUCTION: Optic gliomas are classified as pilocytic astrocytoma (PA) or pilomyxoid astrocytoma (PMXA). Abundant bluish chondroid myxoid matrix is characteristic of PMXA but not PA. We sought to investigate the molecular composition of myxoid matrix and its biologic role in angiogenesis of optic gliomas. We reviewed clinical and pathological data on a cohort of 120 patients with optic glioma diagnosed at NYU Langone Medical Center from 1996 to 2014. We analyzed microvascular density (MVD), perfusion, hypoxia and proliferation by immunohistochemistry and ultrastructural features by electron microscopy. To identify the composition of the myxoid matrix in PMXA we performed liquid chromatography-mass spectrometry (LC-MS) without sample fractionation quantified using peptide spectral counts. PMXA showed significantly lower MVD by CD34 (8.1 vs 14.5, p-value < 0.002) and Erg (7 vs. 13.6, p-value 0.003) than PA, however GLUT-1 showed equal perfusion. Electron microscopy showed that PMXA contain both regular blood vessels with endothelial lining and channels completely lacking endothelial and smooth muscle cells. LC-MS stratified optic gliomas into three distinct groups. We identified 5389 proteins of which 188 were differentially expressed in the three groups (p<0.05, Benjamini-Hochberg adjustment). Between PA and PMXA, we found that most of differentially expressed proteins (146/188) displayed a positive fold change (increasing in PMXA relative to PA), and a minority (42/188) showed a negative fold change. The most abundant extracellular matrix proteins were a chondroitin sulfate proteoglycan versican (VCAN 3.7-fold increase Q=0.000463) and its paralog vertebrate Hyaluronan And Proteoglycan Link Protein 1 (HAPLN1, 22-fold increase from the PA to the PMXA group Q=4.60x10-7). Optic gliomas can develop endothelium-independent channels reminiscent of those in invertebrates to maintain blood supply. The myxoid matrix is composed of VCAN and its linking paralog HAPLN1. Targeting the myxoid matrix may provide novel avenues for therapy of optic gliom
EMBASE:622711609
ISSN: 1554-6578
CID: 3188352
Global loss of histone H3K27 trimethylation in atypical and anaplastic meningiomas [Meeting Abstract]
Liechty, B; Katz, L; Fatterpekar, G; Sen, R; Silverman, J; Golfinos, J; Sen, C; Zagzag, D; Snuderl, M
H3K27 downregulates gene transcription. When H3K27 is trimethylated, it is tightly associated with inactive gene promoters. In malignant gliomas, the loss of histone H3K27 trimethylation is strongly associated with underlying K27M mutation; however the role of H3K27 in meningiomas has not been completely elucidated. Atypical and anaplastic meningiomas (WHO Grade II and III) are associated with higher risk of recurrence following gross total resection; however the molecular biology of anaplastic progression is not completely understood. We performed histological and molecular analysis of 14 WHO Grade II and III meningiomas and compared them with 6 locally invasive WHO Grade I meningiomas. Grade and atypical features were correlated with expression of histone H3K27 trimethylation by immunohistochemistry. Staining intensity (none, weak, moderate, strong) and extent of staining (0-100% of the tumor) were evaluated semi-quantitatively. We also tested the tumors for K27M mutation by mutation specific antibody. Out of 14 high grade meningiomas, 10 showed a complete loss of K27 trimethyl staining and 4 tumors showed small foci of preserved trimethyl staining, mostly in areas close to the dura; however staining intensity was weak. In contrary, all 6 (100%) WHO Grade I tumors showed preserved multifocal trimethyl mark expression in 25-50% of the tumor cells, with moderate (5) or strong (1) staining intensity. All tumors were negative for histone H3K27M mutation by immunohistochemistry. Atypical and anaplastic meningiomas show almost uniform loss of histone H3K27 trimethylation staining. However this loss of trimethylation is not caused by histone H3K27M mutation. Loss of histone H3K27 trimethylation leads to dysregulation of the PRC2 complex, which is involved in repression of non-cell-type specific promoters and may contribute to aggressive behavior. Clinically, loss of histone H3K27 trimethylation can be used as a diagnostic marker for a high grade meningiomaswhen histological features are inconclusive
EMBASE:622711546
ISSN: 1554-6578
CID: 3188362
Histopathologic correlates of newly defined brain tumor entities identified through molecular classification [Meeting Abstract]
Orr, B; Sturm, D; Toprak, U; Hovestadt, V; Jones, D; Capper, D; Northcott, P; Allen, S; Phillips, J; Perry, A; Mobley, B; Schniederjan, M; Santi, M; Buccoliero, A; Dahiya, S; Snuderl, M; Gajjar, A; Aldape, K; Von, Deimling A; Pfister, S; Ellison, D; Korshunov, A; Kool, M
High grade pediatric brain tumors, particularly embryonal tumors and small cell high-grade gliomas, are challenging to classify because they lack clear morphologic differentiation and our current repertoire of discriminating biomarkers is limited. Inappropriate classification has important consequences with respect to adjuvant therapy and increases the heterogeneity of clinical trial cohorts, which potentially obscures promising treatment leads. Recently, using molecular profiling of a large cohort of pediatric brain tumors, we identified four novel entities designated CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2), CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC), CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1), and CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR), each characterized by distinct recurrent molecular abnormalities and clinicopathologic features. While these tumors demonstrate significant histologic overlap with other brain tumor types, they are associated with recurrent morphologic features and immunophenotypes. We discuss the histopathologic correlates of these new brain tumor entities and emerging biomarkers that distinguish these tumors from other high-grade pediatric brain tumors. Incorporation of these tumors into the WHO classification will allow for improved design of future clinical trials and facilitate targeted approaches to treatment
EMBASE:622711534
ISSN: 1554-6578
CID: 3188372
Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival
Kloepper, Jonas; Riedemann, Lars; Amoozgar, Zohreh; Seano, Giorgio; Susek, Katharina; Yu, Veronica; Dalvie, Nisha; Amelung, Robin L; Datta, Meenal; Song, Jonathan W; Askoxylakis, Vasileios; Taylor, Jennie W; Lu-Emerson, Christine; Batista, Ana; Kirkpatrick, Nathaniel D; Jung, Keehoon; Snuderl, Matija; Muzikansky, Alona; Stubenrauch, Kay G; Krieter, Oliver; Wakimoto, Hiroaki; Xu, Lei; Munn, Lance L; Duda, Dan G; Fukumura, Dai; Batchelor, Tracy T; Jain, Rakesh K
Inhibition of the vascular endothelial growth factor (VEGF) pathway has failed to improve overall survival of patients with glioblastoma (GBM). We previously showed that angiopoietin-2 (Ang-2) overexpression compromised the benefit from anti-VEGF therapy in a preclinical GBM model. Here we investigated whether dual Ang-2/VEGF inhibition could overcome resistance to anti-VEGF treatment. We treated mice bearing orthotopic syngeneic (Gl261) GBMs or human (MGG8) GBM xenografts with antibodies inhibiting VEGF (B20), or Ang-2/VEGF (CrossMab, A2V). We examined the effects of treatment on the tumor vasculature, immune cell populations, tumor growth, and survival in both the Gl261 and MGG8 tumor models. We found that in the Gl261 model, which displays a highly abnormal tumor vasculature, A2V decreased vessel density, delayed tumor growth, and prolonged survival compared with B20. In the MGG8 model, which displays a low degree of vessel abnormality, A2V induced no significant changes in the tumor vasculature but still prolonged survival. In both the Gl261 and MGG8 models A2V reprogrammed protumor M2 macrophages toward the antitumor M1 phenotype. Our findings indicate that A2V may prolong survival in mice with GBM by reprogramming the tumor immune microenvironment and delaying tumor growth.
PMCID:4843473
PMID: 27044098
ISSN: 1091-6490
CID: 2120752
Dual inhibition of Ang-2 and VEGF receptors normalizes tumor vasculature and prolongs survival in glioblastoma by altering macrophages
Peterson, Teresa E; Kirkpatrick, Nathaniel D; Huang, Yuhui; Farrar, Christian T; Marijt, Koen A; Kloepper, Jonas; Datta, Meenal; Amoozgar, Zohreh; Seano, Giorgio; Jung, Keehoon; Kamoun, Walid S; Vardam, Trupti; Snuderl, Matija; Goveia, Jermaine; Chatterjee, Sampurna; Batista, Ana; Muzikansky, Alona; Leow, Ching Ching; Xu, Lei; Batchelor, Tracy T; Duda, Dan G; Fukumura, Dai; Jain, Rakesh K
Glioblastomas (GBMs) rapidly become refractory to anti-VEGF therapies. We previously demonstrated that ectopic overexpression of angiopoietin-2 (Ang-2) compromises the benefits of anti-VEGF receptor (VEGFR) treatment in murine GBM models and that circulating Ang-2 levels in GBM patients rebound after an initial decrease following cediranib (a pan-VEGFR tyrosine kinase inhibitor) administration. Here we tested whether dual inhibition of VEGFR/Ang-2 could improve survival in two orthotopic models of GBM, Gl261 and U87. Dual therapy using cediranib and MEDI3617 (an anti-Ang-2-neutralizing antibody) improved survival over each therapy alone by delaying Gl261 growth and increasing U87 necrosis, effectively reducing viable tumor burden. Consistent with their vascular-modulating function, the dual therapies enhanced morphological normalization of vessels. Dual therapy also led to changes in tumor-associated macrophages (TAMs). Inhibition of TAM recruitment using an anti-colony-stimulating factor-1 antibody compromised the survival benefit of dual therapy. Thus, dual inhibition of VEGFR/Ang-2 prolongs survival in preclinical GBM models by reducing tumor burden, improving normalization, and altering TAMs. This approach may represent a potential therapeutic strategy to overcome the limitations of anti-VEGFR monotherapy in GBM patients by integrating the complementary effects of anti-Ang2 treatment on vessels and immune cells.
PMCID:4843449
PMID: 27044097
ISSN: 1091-6490
CID: 2120762
New Brain Tumor Entities Emerge from Molecular Classification of CNS-PNETs
Sturm, Dominik; Orr, Brent A; Toprak, Umut H; Hovestadt, Volker; Jones, David T W; Capper, David; Sill, Martin; Buchhalter, Ivo; Northcott, Paul A; Leis, Irina; Ryzhova, Marina; Koelsche, Christian; Pfaff, Elke; Allen, Sariah J; Balasubramanian, Gnanaprakash; Worst, Barbara C; Pajtler, Kristian W; Brabetz, Sebastian; Johann, Pascal D; Sahm, Felix; Reimand, Juri; Mackay, Alan; Carvalho, Diana M; Remke, Marc; Phillips, Joanna J; Perry, Arie; Cowdrey, Cynthia; Drissi, Rachid; Fouladi, Maryam; Giangaspero, Felice; Lastowska, Maria; Grajkowska, Wieslawa; Scheurlen, Wolfram; Pietsch, Torsten; Hagel, Christian; Gojo, Johannes; Lotsch, Daniela; Berger, Walter; Slavc, Irene; Haberler, Christine; Jouvet, Anne; Holm, Stefan; Hofer, Silvia; Prinz, Marco; Keohane, Catherine; Fried, Iris; Mawrin, Christian; Scheie, David; Mobley, Bret C; Schniederjan, Matthew J; Santi, Mariarita; Buccoliero, Anna M; Dahiya, Sonika; Kramm, Christof M; von Bueren, Andre O; von Hoff, Katja; Rutkowski, Stefan; Herold-Mende, Christel; Fruhwald, Michael C; Milde, Till; Hasselblatt, Martin; Wesseling, Pieter; Rossler, Jochen; Schuller, Ulrich; Ebinger, Martin; Schittenhelm, Jens; Frank, Stephan; Grobholz, Rainer; Vajtai, Istvan; Hans, Volkmar; Schneppenheim, Reinhard; Zitterbart, Karel; Collins, V Peter; Aronica, Eleonora; Varlet, Pascale; Puget, Stephanie; Dufour, Christelle; Grill, Jacques; Figarella-Branger, Dominique; Wolter, Marietta; Schuhmann, Martin U; Shalaby, Tarek; Grotzer, Michael; van Meter, Timothy; Monoranu, Camelia-Maria; Felsberg, Jorg; Reifenberger, Guido; Snuderl, Matija; Forrester, Lynn Ann; Koster, Jan; Versteeg, Rogier; Volckmann, Richard; van Sluis, Peter; Wolf, Stephan; Mikkelsen, Tom; Gajjar, Amar; Aldape, Kenneth; Moore, Andrew S; Taylor, Michael D; Jones, Chris; Jabado, Nada; Karajannis, Matthias A; Eils, Roland; Schlesner, Matthias; Lichter, Peter; von Deimling, Andreas; Pfister, Stefan M; Ellison, David W; Korshunov, Andrey; Kool, Marcel
Primitive neuroectodermal tumors of the central nervous system (CNS-PNETs) are highly aggressive, poorly differentiated embryonal tumors occurring predominantly in young children but also affecting adolescents and adults. Herein, we demonstrate that a significant proportion of institutionally diagnosed CNS-PNETs display molecular profiles indistinguishable from those of various other well-defined CNS tumor entities, facilitating diagnosis and appropriate therapy for patients with these tumors. From the remaining fraction of CNS-PNETs, we identify four new CNS tumor entities, each associated with a recurrent genetic alteration and distinct histopathological and clinical features. These new molecular entities, designated "CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2)," "CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC)," "CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1)," and "CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR)," will enable meaningful clinical trials and the development of therapeutic strategies for patients affected by poorly differentiated CNS tumors.
PMCID:5139621
PMID: 26919435
ISSN: 1097-4172
CID: 1965622
A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer
Whitley, Melodi Javid; Cardona, Diana M; Lazarides, Alexander L; Spasojevic, Ivan; Ferrer, Jorge M; Cahill, Joan; Lee, Chang-Lung; Snuderl, Matija; Blazer, Dan G 3rd; Hwang, E Shelley; Greenup, Rachel A; Mosca, Paul J; Mito, Jeffrey K; Cuneo, Kyle C; Larrier, Nicole A; O'Reilly, Erin K; Riedel, Richard F; Eward, William C; Strasfeld, David B; Fukumura, Dai; Jain, Rakesh K; Lee, W David; Griffith, Linda G; Bawendi, Moungi G; Kirsch, David G; Brigman, Brian E
Local recurrence is a common cause of treatment failure for patients with solid tumors. Intraoperative detection of microscopic residual cancer in the tumor bed could be used to decrease the risk of a positive surgical margin, reduce rates of reexcision, and tailor adjuvant therapy. We used a protease-activated fluorescent imaging probe, LUM015, to detect cancer in vivo in a mouse model of soft tissue sarcoma (STS) and ex vivo in a first-in-human phase 1 clinical trial. In mice, intravenous injection of LUM015 labeled tumor cells, and residual fluorescence within the tumor bed predicted local recurrence. In 15 patients with STS or breast cancer, intravenous injection of LUM015 before surgery was well tolerated. Imaging of resected human tissues showed that fluorescence from tumor was significantly higher than fluorescence from normal tissues. LUM015 biodistribution, pharmacokinetic profiles, and metabolism were similar in mouse and human subjects. Tissue concentrations of LUM015 and its metabolites, including fluorescently labeled lysine, demonstrated that LUM015 is selectively distributed to tumors where it is activated by proteases. Experiments in mice with a constitutively active PEGylated fluorescent imaging probe support a model where tumor-selective probe distribution is a determinant of increased fluorescence in cancer. These co-clinical studies suggest that the tumor specificity of protease-activated imaging probes, such as LUM015, is dependent on both biodistribution and enzyme activity. Our first-in-human data support future clinical trials of LUM015 and other protease-sensitive probes.
PMCID:4794335
PMID: 26738797
ISSN: 1946-6242
CID: 2042672
Advancing methylation profiling in neuropathology: Diagnosis and clinical management [Meeting Abstract]
Kannan, Kasthuri S; Tsirigos, Aristotelis; Serrano, Jonathan; Forrester, Lynn Ann; Faustin, Arline; Thomas, Cheddhi; Capper, David; Hovestadt, Volker; Pfister, Stefan M; Jones, David TW; Sill, Martin; Schrimpf, Daniel; von Deimling, Andreas; Heguy, Adriana; Gardner, Sharon L; Allen, Jeffrey; Hedvat, Cyrus; Zagzag, David; Snuderl, Matija; Karajannis, Matthias A
ISI:000369082700032
ISSN: 1557-3265
CID: 2687512