Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Donald M. O’Rourke is active.

Publication


Featured researches published by Donald M. O’Rourke.


Nature | 2012

IDH mutation impairs histone demethylation and results in a block to cell differentiation

Chao Lu; Patrick S. Ward; Gurpreet S. Kapoor; D. Rohle; Sevin Turcan; Omar Abdel-Wahab; Christopher R. Edwards; Raya Khanin; Maria E. Figueroa; Ari Melnick; Kathryn E. Wellen; Donald M. O’Rourke; Shelley L. Berger; Timothy A. Chan; Ross L. Levine; Ingo K. Mellinghoff; Craig B. Thompson

Recurrent mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 have been identified in gliomas, acute myeloid leukaemias (AML) and chondrosarcomas, and share a novel enzymatic property of producing 2-hydroxyglutarate (2HG) from α-ketoglutarate. Here we report that 2HG-producing IDH mutants can prevent the histone demethylation that is required for lineage-specific progenitor cells to differentiate into terminally differentiated cells. In tumour samples from glioma patients, IDH mutations were associated with a distinct gene expression profile enriched for genes expressed in neural progenitor cells, and this was associated with increased histone methylation. To test whether the ability of IDH mutants to promote histone methylation contributes to a block in cell differentiation in non-transformed cells, we tested the effect of neomorphic IDH mutants on adipocyte differentiation in vitro. Introduction of either mutant IDH or cell-permeable 2HG was associated with repression of the inducible expression of lineage-specific differentiation genes and a block to differentiation. This correlated with a significant increase in repressive histone methylation marks without observable changes in promoter DNA methylation. Gliomas were found to have elevated levels of similar histone repressive marks. Stable transfection of a 2HG-producing mutant IDH into immortalized astrocytes resulted in progressive accumulation of histone methylation. Of the marks examined, increased H3K9 methylation reproducibly preceded a rise in DNA methylation as cells were passaged in culture. Furthermore, we found that the 2HG-inhibitable H3K9 demethylase KDM4C was induced during adipocyte differentiation, and that RNA-interference suppression of KDM4C was sufficient to block differentiation. Together these data demonstrate that 2HG can inhibit histone demethylation and that inhibition of histone demethylation can be sufficient to block the differentiation of non-transformed cells.


Science Translational Medicine | 2017

A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma

Donald M. O’Rourke; MacLean P. Nasrallah; Arati Desai; J. Joseph Melenhorst; Keith Mansfield; Jennifer J.D. Morrissette; Maria Martinez-Lage; Steven Brem; Eileen Maloney; Angela Shen; Randi Isaacs; Suyash Mohan; Gabriela Plesa; Simon F. Lacey; Jean-Marc Navenot; Zhaohui Zheng; Bruce L. Levine; Hideho Okada; Carl H. June; Jennifer Brogdon; Marcela V. Maus

A trial of autologous T cells redirected to a specific mutation in glioblastoma patients illustrates mechanisms of resistance. Speeding toward CAR T cell therapy for glioblastoma Chimeric antigen receptor (CAR) T cells have been successfully implemented for treating leukemia and are now being investigated for solid tumors. O’Rourke et al. conducted a phase 1 safety study of autologous CAR T cells targeted to EGFR variant III in glioblastoma patients. Treatment seemed to be well tolerated, which is critical because other CAR T cell products have been implicated in devastating central nervous system complications. Of the 10 patients enrolled, 7 had surgical intervention, allowing for some analysis of the tumors and T cells in patients’ brains. The results of this trial indicate that CAR T cell therapy is a viable option for treating glioblastoma. We conducted a first-in-human study of intravenous delivery of a single dose of autologous T cells redirected to the epidermal growth factor receptor variant III (EGFRvIII) mutation by a chimeric antigen receptor (CAR). We report our findings on the first 10 recurrent glioblastoma (GBM) patients treated. We found that manufacturing and infusion of CAR-modified T cell (CART)–EGFRvIII cells are feasible and safe, without evidence of off-tumor toxicity or cytokine release syndrome. One patient has had residual stable disease for over 18 months of follow-up. All patients demonstrated detectable transient expansion of CART-EGFRvIII cells in peripheral blood. Seven patients had post–CART-EGFRvIII surgical intervention, which allowed for tissue-specific analysis of CART-EGFRvIII trafficking to the tumor, phenotyping of tumor-infiltrating T cells and the tumor microenvironment in situ, and analysis of post-therapy EGFRvIII target antigen expression. Imaging findings after CART immunotherapy were complex to interpret, further reinforcing the need for pathologic sampling in infused patients. We found trafficking of CART-EGFRvIII cells to regions of active GBM, with antigen decrease in five of these seven patients. In situ evaluation of the tumor environment demonstrated increased and robust expression of inhibitory molecules and infiltration by regulatory T cells after CART-EGFRvIII infusion, compared to pre–CART-EGFRvIII infusion tumor specimens. Our initial experience with CAR T cells in recurrent GBM suggests that although intravenous infusion results in on-target activity in the brain, overcoming the adaptive changes in the local tumor microenvironment and addressing the antigen heterogeneity may improve the efficacy of EGFRvIII-directed strategies in GBM.


Journal of Neuro-oncology | 2010

Activated EGFR signaling increases proliferation, survival, and migration and blocks neuronal differentiation in post-natal neural stem cells

Angel Ayuso-Sacido; Jennifer A. Moliterno; Sebila Kratovac; Gurpreet S. Kapoor; Donald M. O’Rourke; Eric C. Holland; Jose Manuel Garcia-Verdugo; Neeta S. Roy; John A. Boockvar

Recent evidence supports the notion that transformation of undifferentiated neural stem cell (NSC) precursors may contribute to the development of glioblastoma multiforme (GBM). The over-expression and mutation of the epidermal growth factor receptor (EGFR), along with other cellular pathway mutations, plays a significant role in GBM maintenance progression. Though EGFR signaling is important in determining neural cell fate and conferring astrocyte differentiation, there is a limited understanding of its role in NSC and tumor stem cell (TSC) biology. We hypothesized that EGFR expression and mutation in post-natal NSCs may contribute to cellular aggressiveness including enhanced cellular proliferation, survival and migration. Stable subclones of C17.2 murine NSCs were transfected to over-express either the wild-type EGFR (wtEGFR) or its most common mutated variant EGFRvIII. Activated EGFR signaling in these cells induced behaviors characteristic of GBM TSCs, including enhanced proliferation, survival and migration, even in the absence of EGF ligand. wtEGFR activation was also found to block neuronal differentiation and was associated with a dramatic increase in chemotaxis in the presence of EGF. EGFRvIII expression lead to an increase in NSC proliferation and survival, while it simultaneously blocked neuronal differentiation and promoted glial fate. Our findings suggest that activated EGFR signaling enhances the aggressiveness of NSCs. Understanding the regulatory mechanisms of NSCs may lend insight into deregulated mechanisms of GBM TSC invasion, proliferation, survival and resistance to current treatment modalities.


European Journal of Endocrinology | 2011

Clinically silent somatotroph adenomas are common

Alisha Wade; Jennifer Baccon; M. Sean Grady; Kevin Judy; Donald M. O’Rourke; Peter J. Snyder

Objective Somatotroph adenomas are typically recognized when they secrete GH excessively and cause acromegaly. Both ‘silent’ somatotroph adenomas (immunohistochemical evidence of GH excess without biochemical or clinical evidence) and ‘clinically silent’ somatotroph adenomas (immunohistochemical and biochemical evidence but no clinical evidence) have occasionally been reported. The relative frequency of each presentation is unknown. The goal of this study was, therefore, to determine the frequency of clinically silent somatotroph adenomas, a group that is potentially recognizable in vivo. Design We retrospectively identified 100 consecutive patients who had surgically excised and histologically confirmed pituitary adenomas. Methods Each pituitary adenoma was classified immunohistochemically by pituitary cell type. Somatotroph adenomas were further classified as ‘classic’ (obvious clinical features of acromegaly and elevated serum IGF1), ‘subtle’ (subtle clinical features of acromegaly and elevated IGF1), ‘clinically silent’ (no clinical features of acromegaly but elevated IGF1), and ‘silent’ (no clinical features of acromegaly and normal IGF1). Results Of the 100 consecutive pituitary adenomas, 29% were gonadotroph/glycoprotein, 24% somatotroph, 18% null cell, 15% corticotroph, 6% lactotroph, 2% thyrotroph, and 6% not classifiable. Of the 24 patients with somatotroph adenomas, classic accounted for 45.8%, subtle 16.7%, clinically silent 33.3%, and silent 4.2%. Conclusions Clinically silent somatotroph adenomas are more common than previously appreciated, representing one-third of all somatotroph adenomas. IGF1 should be measured in all patients with a sellar mass, because identification of a mass as a somatotroph adenoma expands the therapeutic options and provides a tumor marker to monitor treatment.


Journal of Clinical Investigation | 2016

c-Met–mediated endothelial plasticity drives aberrant vascularization and chemoresistance in glioblastoma

Menggui Huang; Tianrun Liu; Peihong Ma; R. Alan Mitteer; Zhenting Zhang; Hyun Jun Kim; Eujin Yeo; Duo Zhang; Peiqiang Cai; Chunsheng Li; Lin Zhang; Botao Zhao; Laura Roccograndi; Donald M. O’Rourke; Nadia Dahmane; Yanqing Gong; Constantinos Koumenis; Yi Fan

Aberrant vascularization is a hallmark of cancer progression and treatment resistance. Here, we have shown that endothelial cell (EC) plasticity drives aberrant vascularization and chemoresistance in glioblastoma multiforme (GBM). By utilizing human patient specimens, as well as allograft and genetic murine GBM models, we revealed that a robust endothelial plasticity in GBM allows acquisition of fibroblast transformation (also known as endothelial mesenchymal transition [Endo-MT]), which is characterized by EC expression of fibroblast markers, and determined that a prominent population of GBM-associated fibroblast-like cells have EC origin. Tumor ECs acquired the mesenchymal gene signature without the loss of EC functions, leading to enhanced cell proliferation and migration, as well as vessel permeability. Furthermore, we identified a c-Met/ETS-1/matrix metalloproteinase-14 (MMP-14) axis that controls VE-cadherin degradation, Endo-MT, and vascular abnormality. Pharmacological c-Met inhibition induced vessel normalization in patient tumor-derived ECs. Finally, EC-specific KO of Met inhibited vascular transformation, normalized blood vessels, and reduced intratumoral hypoxia, culminating in suppressed tumor growth and prolonged survival in GBM-bearing mice after temozolomide treatment. Together, these findings illustrate a mechanism that controls aberrant tumor vascularization and suggest that targeting Endo-MT may offer selective and efficient strategies for antivascular and vessel normalization therapies in GBM, and possibly other malignant tumors.


Academic Radiology | 2015

Automated Tumor Volumetry Using Computer-Aided Image Segmentation

Bilwaj Gaonkar; Luke Macyszyn; Michel Bilello; Mohammed Salehi Sadaghiani; Hamed Akbari; Mark A. Attiah; Zarina S. Ali; Xiao Da; Yiqang Zhan; Donald M. O’Rourke; Sean M. Grady; Christos Davatzikos

RATIONALE AND OBJECTIVES Accurate segmentation of brain tumors, and quantification of tumor volume, is important for diagnosis, monitoring, and planning therapeutic intervention. Manual segmentation is not widely used because of time constraints. Previous efforts have mainly produced methods that are tailored to a particular type of tumor or acquisition protocol and have mostly failed to produce a method that functions on different tumor types and is robust to changes in scanning parameters, resolution, and image quality, thereby limiting their clinical value. Herein, we present a semiautomatic method for tumor segmentation that is fast, accurate, and robust to a wide variation in image quality and resolution. MATERIALS AND METHODS A semiautomatic segmentation method based on the geodesic distance transform was developed and validated by using it to segment 54 brain tumors. Glioblastomas, meningiomas, and brain metastases were segmented. Qualitative validation was based on physician ratings provided by three clinical experts. Quantitative validation was based on comparing semiautomatic and manual segmentations. RESULTS Tumor segmentations obtained using manual and automatic methods were compared quantitatively using the Dice measure of overlap. Subjective evaluation was performed by having human experts rate the computerized segmentations on a 0-5 rating scale where 5 indicated perfect segmentation. CONCLUSIONS The proposed method addresses a significant, unmet need in the field of neuro-oncology. Specifically, this method enables clinicians to obtain accurate and reproducible tumor volumes without the need for manual segmentation.


Clinical Neurology and Neurosurgery | 2011

Treatment of steroid refractory, Gamma Knife related radiation necrosis with bevacizumab: Case report and review of the literature

Matthew R. Sanborn; Shabbar F. Danish; Myrna R. Rosenfeld; Donald M. O’Rourke; John Y. K. Lee

Radiation necrosis is the most significant complication associted with Gamma Knife radiosurgery. It typically becomes manifest s a necrotic white matter lesion 3 or more months following treatent [3]. Treatment volume and radiation dose are the two most mportant predictors of radiation necrosis. Once radiation necrois has become clinically apparent treatment has historically been imited to corticosteroids. Antiplatelet agents, anticoagulants and yperbaric oxygen have been studied but there is currently minimal igh quality evidence to support their use in routine clinical pracice [6]. If the radiation necrosis fails to respond to steroid treatment herapeutic options are limited. Recent trials have highlighted a potential role for bevacizumab, a onoclonal antibody directed against vascular endothelial growth actor (VEGF) in the treatment of radiation necrosis from convenional radiation therapy [14]. We present a case of symptomatic adiation necrosis following Gamma Knife radiosurgery for muliple atypical WHO grade II meningiomas initially unresponsive o steroid treatment and ultimately responsive to bevacizumab herapy.


Immunologic Research | 1998

Immunologic approaches to inhibiting cell-surface-residing oncoproteins in human tumors.

Donald M. O’Rourke; Mark I. Greene

The erbB family of receptor tyrosine kinases are growth factor receptors that are overexpressed or mutated in a large variety of human cancers. Studies of erbB-mediated signal transduction will lead to an understanding of the role played by this family of receptors in normal and transformed cells. In this article, we discuss the contemporary understanding of the structure and function of these receptors, and how these features might be exploited in immunologie strategies of receptor-based growth inhibition. The first part of this article details the structure of erbB receptors as it relates to the process of transformation of cells and the malignant phenotype in human tumors. In the second part of this article, we discuss immunologie approaches to therapy for cancers in which surface-residing erbB receptors are overexpressed or mutated, with an emphasis on studies targeting the pl85neu/c-erbB2 oncoprotein. The potential for antireceptor immunity and the evolution of small molecules for receptor-based immunotherapy are discussed. These studies provide a basis for the application of receptor-based strategies of growth inhibition in erbB-expressing human cancers.


Cancer Biology & Therapy | 2007

EGFR inhibition in glioblastoma cells induces G2/M arrest and is independent of p53

Gurpreet S. Kapoor; Antoire Christie; Donald M. O’Rourke

Mutations involving the TP53 gene are frequently identified in up to 50% of all human tumors, including glioblastomas. Analysis of expression patterns of TP53 in glioblastomas shows that it is mainly mutated in secondary glioblastomas and is less common in primary GBMs. However, the prognostic significance of TP53 loss of function in astrocytomas has always been controversial. In contrast, EGFR/erbB2 complexes have been implicated in the poor prognosis of several cancers, including glioblastomas. Our previous work showed that transforming phenotypes could be inhibited by interfering with active EGFR/erbB2 complex using mutant erbB2 proteins in wild-type p53 GBM cells.1, 2 To assess the dependence of EGFR inhibited phenotype on p53, we used three mutant p53 glioblastoma cell lines in the present study and showed that mutant erbB2 can be exploited to inhibit EGFR-mediated oncogenic transformation irrespective of p53 status. Ectopic expression of a mutant erbB2 receptor (T691S) in mutant p53 GBM cells resulted in slower growth rate than empty vector controls. T691S-expressing clones exhibited a more flattened and non-transformed morphology. Consistently, T691S inhibited transformation in soft agar assays and tumor formation in nude mice independent of p53 status. Biochemical analysis showed reduced Akt and GSK-3 α/β, but not p42/44MAPK phosphorylation, in T691S-expressing cells, when compared to parental controls, suggesting the PI3-K pathway may be more relevant than MAPK for glial cell transformation. Cell cycle analysis showed reduced cyclin D1 and CDK6 and increased phospho-Cdc-2 (Tyr15) and p15INK4B in erbB2-inhibited cells, suggesting that non-functional EGFR/erbB2 complexes exert their inhibitory effects at various stages of the cell cycle to block the progression of cells through G2/M via Akt/GSK-3/Cdc2 pathway. Collectively, these observations provide a basis for receptor-based therapies that disable erbB receptors and inhibit proliferative signals in erbB-expressing human cancers including glioblastomas, regardless of their TP53 status.


NeuroImage: Clinical | 2016

Population-based MRI atlases of spatial distribution are specific to patient and tumor characteristics in glioblastoma.

Michel Bilello; Hamed Akbari; Xiao Da; Jared M. Pisapia; Suyash Mohan; Ronald L. Wolf; Donald M. O’Rourke; Maria Martinez-Lage; Christos Davatzikos

Background and purpose In treating glioblastoma (GB), surgical and chemotherapeutic treatment guidelines are, for the most part, independent of tumor location. In this work, we compiled imaging data from a large cohort of GB patients to create statistical atlases illustrating the disease spatial frequency as a function of patient demographics as well as tumor characteristics. Materials and methods Two-hundred-six patients with pathology-proven glioblastoma were included. Of those, 65 had pathology-proven recurrence and 113 had molecular subtype and genetic information. We used validated software to segment the tumors in all patients and map them from patient space into a common template. We then created statistical maps that described the spatial location of tumors with respect to demographics and tumor characteristics. We applied a chi-square test to determine whether pattern differences were statistically significant. Results The most frequent location for glioblastoma in our patient population is the right temporal lobe. There are statistically significant differences when comparing patterns using demographic data such as gender (p = 0.0006) and age (p = 0.006). Small and large tumors tend to occur in separate locations (p = 0.0007). The tumors tend to occur in different locations according to their molecular subtypes (p < 10− 6). The classical subtype tends to spare the frontal lobes, the neural subtype tend to involve the inferior right frontal lobe. Although the sample size is limited, there was a difference in location according to EGFR VIII genotype (p < 10− 4), with a right temporal dominance for EFGR VIII negative tumors, and frontal lobe dominance in EGFR VIII positive tumors. Conclusions Spatial location of GB is an important factor that correlates with demographic factors and tumor characteristics, which should therefore be considered when evaluating a patient with GB and might assist in personalized treatment.

Collaboration


Dive into the Donald M. O’Rourke's collaboration.

Top Co-Authors

Avatar

Nadia Dahmane

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Steven Brem

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hamed Akbari

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Zev A. Binder

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arati Desai

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Rozycki

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge