Network


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

Hotspot


Dive into the research topics where David M. Irvin is active.

Publication


Featured researches published by David M. Irvin.


Nature Communications | 2014

Vascular channels formed by subpopulations of PECAM1+ melanoma cells

James M. Dunleavey; Lin Xiao; Joshua Thompson; Mi Mi Kim; Janiel M. Shields; Sarah E. Shelton; David M. Irvin; Victoria E. Brings; David W. Ollila; Rolf A. Brekken; Paul A. Dayton; Juan M. Melero-Martin; Andrew C. Dudley

Targeting the vasculature remains a promising approach for treating solid tumors; however, the mechanisms of tumor neovascularization are diverse and complex. Here we uncover a new subpopulation of melanoma cells that express the vascular cell adhesion molecule PECAM1, but not VEGFR-2, and participate in a PECAM1-dependent form of vasculogenic mimicry (VM). Clonally-derived PECAM1+ tumor cells coalesce to form PECAM1-dependent networks in vitro and they generate well-perfused, VEGF-independent channels in mice. The neural crest specifier AP-2α is diminished in PECAM1+ melanoma cells and is a transcriptional repressor of PECAM1. Reintroduction of AP-2α into PECAM1+ tumor cells represses PECAM1 and abolishes tube-forming ability whereas AP-2α knockdown in PECAM1− tumor cells up-regulates PECAM1 expression and promotes tube formation. Thus, VM-competent subpopulations, rather than all cells within a tumor, may instigate VM, supplant host-derived endothelium, and form PECAM1-dependent conduits that are not diminished by neutralizing VEGF.


Brain Pathology | 2017

Intrinsic astrocyte heterogeneity influences tumor growth in glioma mouse models.

David M. Irvin; Robert S. McNeill; Ryan E. Bash; C. Ryan Miller

The influence of cellular origin on glioma pathogenesis remains elusive. We previously showed that mutations inactivating Rb and Pten and activating Kras transform astrocytes and induce tumorigenesis throughout the adult mouse brain. However, it remained unclear whether astrocyte subpopulations were susceptible to these mutations. We therefore used genetic lineage tracing and fate mapping in adult conditional, inducible genetically engineered mice to monitor transformation of glial fibrillary acidic protein (GFAP) and glutamate aspartate transporter (GLAST) astrocytes and immunofluorescence to monitor cellular composition of the tumor microenvironment over time. Because considerable regional heterogeneity exists among astrocytes, we also examined the influence of brain region on tumor growth. GFAP astrocyte transformation induced uniformly rapid, regionally independent tumor growth, but transformation of GLAST astrocytes induced slowly growing tumors with significant regional bias. Transformed GLAST astrocytes had reduced proliferative response in culture and in vivo and malignant progression was delayed in these tumors. Recruited glial cells, including proliferating astrocytes, oligodendrocyte progenitors and microglia, were the majority of GLAST, but not GFAP astrocyte‐derived tumors and their abundance dynamically changed over time. These results suggest that intrinsic astrocyte heterogeneity, and perhaps regional brain microenvironment, significantly contributes to glioma pathogenesis.


Neuro-oncology | 2017

Combination therapy with potent PI3K and MAPK inhibitors overcomes adaptive kinome resistance to single agents in preclinical models of glioblastoma

Robert S. McNeill; Demitra A. Canoutas; Timothy J. Stuhlmiller; Harshil Dhruv; David M. Irvin; Ryan E. Bash; Steven P. Angus; Laura E. Herring; Jeremy M. Simon; Kasey R. Skinner; Juanita C. Limas; Xin Chen; Ralf S. Schmid; Marni B. Siegel; Amanda E.D. Van Swearingen; Michael J. Hadler; Erik P. Sulman; Jann N. Sarkaria; Carey K. Anders; Lee M. Graves; Michael E. Berens; Gary L. Johnson; C. Ryan Miller

Background Glioblastoma (GBM) is the most common and aggressive primary brain tumor. Prognosis remains poor despite multimodal therapy. Developing alternative treatments is essential. Drugs targeting kinases within the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) effectors of receptor tyrosine kinase (RTK) signaling represent promising candidates. Methods We previously developed a non-germline genetically engineered mouse model of GBM in which PI3K and MAPK are activated via Pten deletion and KrasG12D in immortalized astrocytes. Using this model, we examined the influence of drug potency on target inhibition, alternate pathway activation, efficacy, and synergism of single agent and combination therapy with inhibitors of these 2 pathways. Efficacy was then examined in GBM patient-derived xenografts (PDX) in vitro and in vivo. Results PI3K and mitogen-activated protein kinase kinase (MEK) inhibitor potency was directly associated with target inhibition, alternate RTK effector activation, and efficacy in mutant murine astrocytes in vitro. The kinomes of GBM PDX and tumor samples were heterogeneous, with a subset of the latter harboring MAPK hyperactivation. Dual PI3K/MEK inhibitor treatment overcame alternate effector activation, was synergistic in vitro, and was more effective than single agent therapy in subcutaneous murine allografts. However, efficacy in orthotopic allografts was minimal. This was likely due to dose-limiting toxicity and incomplete target inhibition. Conclusion Drug potency influences PI3K/MEK inhibitor-induced target inhibition, adaptive kinome reprogramming, efficacy, and synergy. Our findings suggest that combination therapies with highly potent, brain-penetrant kinase inhibitors will be required to improve patient outcomes.


Neuro-oncology | 2018

MerTK as a therapeutic target in glioblastoma.

Jing Wu; Lauren N. Frady; Ryan E. Bash; Stephanie M. Cohen; Allison N. Schorzman; Yu Ting Su; David M. Irvin; William C. Zamboni; Xiaodong Wang; Stephen V. Frye; Matthew G. Ewend; Erik P. Sulman; Mark R. Gilbert; H. Shelton Earp; C. Ryan Miller

Background Glioma-associated macrophages and microglia (GAMs) are components of the glioblastoma (GBM) microenvironment that express MerTK, a receptor tyrosine kinase that triggers efferocytosis and can suppress innate immune responses. The aim of the study was to define MerTK as a therapeutic target using an orally bioavailable inhibitor, UNC2025. Methods We examined MerTK expression in tumor cells and macrophages in matched patient GBM samples by double-label immunohistochemistry. UNC2025-induced MerTK inhibition was studied in vitro and in vivo. Results MerTK/CD68+ macrophages increased in recurrent tumors while MerTK/glial fibrillary acidic protein-positive tumor cells did not. Pharmacokinetic studies showed high tumor exposures of UNC2025 in a syngeneic orthotopic allograft mouse GBM model. The same model mice were randomized to receive vehicle, daily UNC2025, fractionated external beam radiotherapy (XRT), or UNC2025/XRT. Although median survival (21, 22, 35, and 35 days, respectively) was equivalent with or without UNC2025, bioluminescence imaging (BLI) showed significant growth delay with XRT/UNC2025 treatment and complete responses in 19%. The responders remained alive for 60 days and showed regression to 1%-10% of pretreatment BLI tumor burden; 5 of 6 were tumor free by histology. In contrast, only 2% of 98 GBM mice of the same model treated with XRT survived 50 days and none survived 60 days. UNC2025 also reduced CD206+ macrophages in mouse tumor samples. Conclusions These results suggest that MerTK inhibition combined with XRT has a therapeutic effect in a subset of GBM. Further mechanistic studies are warranted.


Neuro-oncology | 2017

Genomic profiles of low-grade murine gliomas evolve during progression to glioblastoma

Mark Vitucci; David M. Irvin; Robert S. McNeill; Ralf S. Schmid; Jeremy M. Simon; Harshil Dhruv; Marni B. Siegel; Andrea M. Werneke; Ryan E. Bash; Seungchan Kim; Michael E. Berens; C. R. Miller

Background Gliomas are diverse neoplasms with multiple molecular subtypes. How tumor-initiating mutations relate to molecular subtypes as these tumors evolve during malignant progression remains unclear. Methods We used genetically engineered mouse models, histopathology, genetic lineage tracing, expression profiling, and copy number analyses to examine how genomic tumor diversity evolves during the course of malignant progression from low- to high-grade disease. Results Knockout of all 3 retinoblastoma (Rb) family proteins was required to initiate low-grade tumors in adult mouse astrocytes. Mutations activating mitogen-activated protein kinase signaling, specifically KrasG12D, potentiated Rb-mediated tumorigenesis. Low-grade tumors showed mutant Kras-specific transcriptome profiles but lacked copy number mutations. These tumors stochastically progressed to high-grade, in part through acquisition of copy number mutations. High-grade tumor transcriptomes were heterogeneous and consisted of 3 subtypes that mimicked human mesenchymal, proneural, and neural glioblastomas. Subtypes were confirmed in validation sets of high-grade mouse tumors initiated by different driver mutations as well as human patient-derived xenograft models and glioblastoma tumors. Conclusion These results suggest that oncogenic driver mutations influence the genomic profiles of low-grade tumors and that these, as well as progression-acquired mutations, contribute strongly to the genomic heterogeneity across high-grade tumors.


PLOS ONE | 2018

Intra-cavity stem cell therapy inhibits tumor progression in a novel murine model of medulloblastoma surgical resection

Onyinyechukwu Okolie; David M. Irvin; Juli R. Bagó; Kevin T. Sheets; Andrew Satterlee; Abigail G. Carey-Ewend; Vivien Lettry; Raluca Dumitru; Scott W. Elton; Matthew G. Ewend; C. Ryan Miller; Shawn D. Hingtgen

Background Cytotoxic neural stem cells (NSCs) have emerged as a promising treatment for Medulloblastoma (MB), the most common malignant primary pediatric brain tumor. The lack of accurate pre-clinical models incorporating surgical resection and tumor recurrence limits advancement in post-surgical MB treatments. Using cell lines from two of the 5 distinct MB molecular sub-groups, in this study, we developed an image-guided mouse model of MB surgical resection and investigate intra-cavity NSC therapy for post-operative MB. Methods Using D283 and Daoy human MB cells engineered to express multi-modality optical reporters, we created the first image-guided resection model of orthotopic MB. Brain-derived NSCs and novel induced NSCs (iNSCs) generated from pediatric skin were engineered to express the pro-drug/enzyme therapy thymidine kinase/ganciclovir, seeded into the post-operative cavity, and used to investigate intra-cavity therapy for post-surgical MB. Results We found that surgery reduced MB volumes by 92%, and the rate of post-operative MB regrowth increased 3-fold compared to pre-resection growth. Real-time imaging showed NSCs rapidly homed to MB, migrating 1.6-fold faster and 2-fold farther in the presence of tumors, and co-localized with MB present in the contra-lateral hemisphere. Seeding of cytotoxic NSCs into the post-operative surgical cavity decreased MB volumes 15-fold and extended median survival 133%. As an initial step towards novel autologous therapy in human MB patients, we found skin-derived iNSCs homed to MB cells, while intra-cavity iNSC therapy suppressed post-surgical tumor growth and prolonged survival of MB-bearing mice by 123%. Conclusions We report a novel image-guided model of MB resection/recurrence and provide new evidence of cytotoxic NSCs/iNSCs delivered into the surgical cavity effectively target residual MB foci.


Neuro-oncology | 2016

Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model

Onyinyechukwu Okolie; Juli R. Bagó; Ralf S. Schmid; David M. Irvin; Ryan E. Bash; C. Ryan Miller; Shawn D. Hingtgen


Neuro-oncology | 2017

PDTM-18. INVESTIGATING INTRA-CAVITY NEURAL STEM CELL THERAPY FOR POST-SURGICAL DISEASE IN A NOVEL SURGICAL RESECTION/RECURRENCE MODEL OF MEDULLOBLASTOMA

Onyinyechukwu Okolie; Elizabeth Finch; David M. Irvin; Juli R. Bagó; Kevin T. Sheets; Andrew Satterlee; Raluca Dumitru; Simon Khagi; Scott W. Elton; Matthew G. Ewend; Shawn D. Hingtgen


Neuro-oncology | 2016

TMOD-05. REGIONAL ASTROCYTE HETEROGENEITY INFLUENCES EVOLUTION OF LOW-GRADE GLIOMA DURING MALIGNANT PROGRESSION

David M. Irvin; Mark Vitucci; Robert S. McNeill; Ryan E. Bash; C. Ryan Miller


American Journal of Pathology | 2016

BRAF Mutations Open Doors for N-Ethyl-N-Nitrosourea–Induced Gliomagenesis

Robert S. McNeill; David M. Irvin; C. Ryan Miller

Collaboration


Dive into the David M. Irvin's collaboration.

Top Co-Authors

Avatar

C. Ryan Miller

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ryan E. Bash

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Robert S. McNeill

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ralf S. Schmid

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Juli R. Bagó

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Matthew G. Ewend

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Onyinyechukwu Okolie

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Shawn D. Hingtgen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Andrew Satterlee

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Erik P. Sulman

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge