Network


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

Hotspot


Dive into the research topics where Terry Van Dyke is active.

Publication


Featured researches published by Terry Van Dyke.


Genome Biology | 2007

Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors

Jason I. Herschkowitz; Karl Simin; Victor J. Weigman; Igor Mikaelian; Jerry Usary; Zhiyuan Hu; Karen Rasmussen; Laundette P Jones; Shahin Assefnia; Subhashini Chandrasekharan; Michael G. Backlund; Yuzhi Yin; Andrey Khramtsov; Roy Bastein; John Quackenbush; Robert I. Glazer; Powel H. Brown; Jeffrey Green; Levy Kopelovich; Priscilla A. Furth; Juan P. Palazzo; Olufunmilayo I. Olopade; Philip S. Bernard; Gary A. Churchill; Terry Van Dyke; Charles M. Perou

BackgroundAlthough numerous mouse models of breast carcinomas have been developed, we do not know the extent to which any faithfully represent clinically significant human phenotypes. To address this need, we characterized mammary tumor gene expression profiles from 13 different murine models using DNA microarrays and compared the resulting data to those from human breast tumors.ResultsUnsupervised hierarchical clustering analysis showed that six models (TgWAP-Myc, TgMMTV-Neu, TgMMTV-PyMT, TgWAP-Int3, TgWAP-Tag, and TgC3(1)-Tag) yielded tumors with distinctive and homogeneous expression patterns within each strain. However, in each of four other models (TgWAP-T121, TgMMTV-Wnt1, Brca1Co/Co;TgMMTV-Cre;p53+/- and DMBA-induced), tumors with a variety of histologies and expression profiles developed. In many models, similarities to human breast tumors were recognized, including proliferation and human breast tumor subtype signatures. Significantly, tumors of several models displayed characteristics of human basal-like breast tumors, including two models with induced Brca1 deficiencies. Tumors of other murine models shared features and trended towards significance of gene enrichment with human luminal tumors; however, these murine tumors lacked expression of estrogen receptor (ER) and ER-regulated genes. TgMMTV-Neu tumors did not have a significant gene overlap with the human HER2+/ER- subtype and were more similar to human luminal tumors.ConclusionMany of the defining characteristics of human subtypes were conserved among the mouse models. Although no single mouse model recapitulated all the expression features of a given human subtype, these shared expression features provide a common framework for an improved integration of murine mammary tumor models with human breast tumors.


Cell | 1994

p53-Dependent apoptosis suppresses tumor growth and progression in vivo

Holly Symonds; Leonard Krall; Lee Remington; Mayte Saenz-Robles; Scott W. Lowe; Tyler Jacks; Terry Van Dyke

To determine the contribution of p53 loss to tumor progression, we have induced abnormal proliferation in the brain choroid plexus epithelium of transgenic mice using a SV40 T antigen fragment that perturbs pRB family function but does not affect p53 function. Tumors induced by this mutant develop slowly compared with those induced by wild-type T antigen. Suppressed tumor growth is directly attributable to p53 function, since rapid tumor development occurs when the T antigen fragment is expressed in p53-null mice. In p53-heterozygous mice, stochastic loss of the wild-type p53 allele results in the focal emergence of aggressive tumor nodules characteristic of tumor progression. In each case, aggressive tumor development in the absence of p53 function corresponds to a decrease in the level of apoptosis. These results provide in vivo evidence that p53-dependent apoptosis, occurring in response to oncogenic events, is a critical regulator of tumorigenesis.


PLOS Biology | 2003

Pten Dose Dictates Cancer Progression in the Prostate

Lloyd C. Trotman; Masaru Niki; Zohar A. Dotan; Jason A. Koutcher; Antonio Di Cristofano; Andrew Xiao; Alan S Khoo; Pradip Roy-Burman; Norman M. Greenberg; Terry Van Dyke; Carlos Cordon-Cardo; Pier Paolo Pandolfi

Complete inactivation of the PTEN tumor suppressor gene is extremely common in advanced cancer, including prostate cancer (CaP). However, one PTEN allele is already lost in the vast majority of CaPs at presentation. To determine the consequence of PTEN dose variations on cancer progression, we have generated by homologous recombination a hypomorphic Pten mouse mutant series with decreasing Pten activity: Ptenhy/+ > Pten+/− > Ptenhy/− (mutants in which we have rescued the embryonic lethality due to complete Pten inactivation) > Pten prostate conditional knockout (Ptenpc) mutants. In addition, we have generated and comparatively analyzed two distinct Ptenpc mutants in which Pten is inactivated focally or throughout the entire prostatic epithelium. We find that the extent of Pten inactivation dictate in an exquisite dose-dependent fashion CaP progression, its incidence, latency, and biology. The dose of Pten affects key downstream targets such as Akt, p27Kip1, mTOR, and FOXO3. Our results provide conclusive genetic support for the notion that PTEN is haploinsufficient in tumor suppression and that its dose is a key determinant in cancer progression.


Cell | 1984

Transgenic mice harboring SV40 t-antigen genes develop characteristic brain tumors

Ralph L. Brinster; Howard Y. Chen; Albee Messing; Terry Van Dyke; Arnold J. Levine; Richard D. Palmiter

A high percentage of transgenic mice developing from eggs microinjected with plasmids containing the SV40 early region genes and a metallothionein fusion gene develop tumors within the choroid plexus. A line of mice has been established in which nearly every affected animal succumbs to this brain tumor. Thymic hypertrophy and kidney pathology are also observed in some mice. SV40 T-antigen mRNA and protein are readily detected in affected tissues; however, SV40 T-antigen gene expression is barely detectable in unaffected tissues or in susceptible tissues prior to overt pathology, suggesting that tumorigenesis depends upon activation of the SV40 genes. Comparison of DNA from tumor tissue (or cell lines derived from tumors) with DNA from unaffected tissues reveals structural rearrangements as well as changes in DNA methylation of the foreign DNA. The SV40 genes are frequently amplified in tumor tissue, which further indicates that their expression is intimately involved in tumorigenesis in transgenic mice.


Cancer Discovery | 2013

Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790M-Mediated Resistance in NSCLC

Annette O Walter; Robert Tjin Tham Sjin; Henry J Haringsma; Kadoaki Ohashi; Jing Sun; Kwangho Lee; Aleksander Dubrovskiy; Matthew T. Labenski; Zhendong Zhu; Zhigang Wang; Michael Sheets; Thia St Martin; Russell Karp; Dan van Kalken; Prasoon Chaturvedi; Deqiang Niu; M. Nacht; Russell C. Petter; William F. Westlin; Kevin Lin; Sarah S. Jaw-Tsai; Mitch Raponi; Terry Van Dyke; Jeff Etter; Zoe Weaver; William Pao; Juswinder Singh; Andrew Simmons; Thomas Harding; Andrew E. Allen

UNLABELLED Patients with non-small cell lung cancer (NSCLC) with activating EGF receptor (EGFR) mutations initially respond to first-generation reversible EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by acquired resistance, frequently driven by the EGFR(T790M) mutation. CO-1686 is a novel, irreversible, and orally delivered kinase inhibitor that specifically targets the mutant forms of EGFR, including T790M, while exhibiting minimal activity toward the wild-type (WT) receptor. Oral administration of CO-1686 as single agent induces tumor regression in EGFR-mutated NSCLC tumor xenograft and transgenic models. Minimal activity of CO-1686 against the WT EGFR receptor was observed. In NSCLC cells with acquired resistance to CO-1686 in vitro, there was no evidence of additional mutations or amplification of the EGFR gene, but resistant cells exhibited signs of epithelial-mesenchymal transition and demonstrated increased sensitivity to AKT inhibitors. These results suggest that CO-1686 may offer a novel therapeutic option for patients with mutant EGFR NSCLC. SIGNIFICANCE We report the preclinical development of a novel covalent inhibitor, CO-1686, that irreversibly and selectively inhibits mutant EGFR, in particular the T790M drug-resistance mutation, in NSCLC models. CO-1686 is the fi rst drug of its class in clinical development for the treatment of T790M-positive NSCLC, potentially offering potent inhibition of mutant EGFR while avoiding the on-target toxicity observed with inhibition of the WT EGFR.


Cell | 2002

Cancer Modeling in the Modern Era: Progress and Challenges

Terry Van Dyke; Tyler Jacks

Genetically engineered mouse models have contributed extensively to the field of cancer research. The ability to manipulate the mouse germline affords numerous approaches toward understanding the complexities of this disease, possibly providing accurate preclinical models for therapeutic and diagnostic advances. This review highlights some of the current strategies for modeling cancer in the mouse, recent accomplishments, and key remaining challenges.


Cell | 2005

Selective Evolution of Stromal Mesenchyme with p53 Loss in Response to Epithelial Tumorigenesis

Reginald Hill; Yurong Song; Robert D. Cardiff; Terry Van Dyke

Our understanding of cancer has largely come from the analysis of aberrations within the tumor cell population. Yet it is increasingly clear that the tumor microenvironment can significantly influence tumorigenesis. For example, the mesenchyme can support the growth of tumorigenic epithelium. However, whether fibroblasts are subject to genetic/epigenetic changes as a result of selective pressures conferred by oncogenic stress in the epithelium has not been experimentally assessed. Recent analyses of some human carcinomas have shown tumor-suppressor gene mutations within the stroma, suggesting that the interplay among multiple cell types can select for aberrations nonautonomously during tumor progression. We demonstrate that this indeed occurs in a mouse model of prostate cancer where epithelial cell cycle disruption via cell-specific inhibition of pRb function induces a paracrine p53 response that suppresses fibroblast proliferation in associated stroma. This interaction imposes strong selective pressure yielding a highly proliferative mesenchyme that has undergone p53 loss.


Cell | 2014

Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy.

Mara H. Sherman; Ruth T. Yu; Dannielle D. Engle; Ning Ding; Annette R. Atkins; Hervé Tiriac; Eric A. Collisson; Frances Connor; Terry Van Dyke; Serguei Kozlov; Philip Martin; Tiffany W. Tseng; David W. Dawson; Timothy R. Donahue; Atsushi Masamune; Tooru Shimosegawa; Minoti V. Apte; Jeremy S. Wilson; Beverly Ng; Sue Lynn Lau; Jenny E. Gunton; Geoffrey M. Wahl; Tony Hunter; Jeffrey A. Drebin; Peter J. O’Dwyer; Christopher Liddle; David A. Tuveson; Michael Downes; Ronald M. Evans

The poor clinical outcome in pancreatic ductal adenocarcinoma (PDA) is attributed to intrinsic chemoresistance and a growth-permissive tumor microenvironment. Conversion of quiescent to activated pancreatic stellate cells (PSCs) drives the severe stromal reaction that characterizes PDA. Here, we reveal that the vitamin D receptor (VDR) is expressed in stroma from human pancreatic tumors and that treatment with the VDR ligand calcipotriol markedly reduced markers of inflammation and fibrosis in pancreatitis and human tumor stroma. We show that VDR acts as a master transcriptional regulator of PSCs to reprise the quiescent state, resulting in induced stromal remodeling, increased intratumoral gemcitabine, reduced tumor volume, and a 57% increase in survival compared to chemotherapy alone. This work describes a molecular strategy through which transcriptional reprogramming of tumor stroma enables chemotherapeutic response and suggests vitamin D priming as an adjunct in PDA therapy. PAPERFLICK:


Molecular Therapy | 2008

Engineering and selection of shuffled AAV genomes: a new strategy for producing targeted biological nanoparticles.

Wuping Li; Aravind Asokan; Zhijian Wu; Terry Van Dyke; Nina DiPrimio; Jarrod S. Johnson; Lakshmanan Govindaswamy; Mavis Agbandje-McKenna; Stefan Leichtle; D. Eugene Redmond; Thomas J. McCown; Kimberly B. Petermann; Norman E. Sharpless; Richard Jude Samulski

We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.


Molecular Cell | 1998

Key Roles for E2F1 in Signaling p53-Dependent Apoptosis and in Cell Division within Developing Tumors

Huichin Pan; Chaoying Yin; Nicholas J. Dyson; Ed Harlow; Lili Yamasaki; Terry Van Dyke

Apoptosis induced by the p53 tumor suppressor can attenuate cancer growth in preclinical animal models. Inactivation of the pRb proteins in mouse brain epithelium by the T121 oncogene induces aberrant proliferation and p53-dependent apoptosis. p53 inactivation causes aggressive tumor growth due to an 85% reduction in apoptosis. Here, we show that E2F1 signals p53-dependent apoptosis since E2F1 deficiency causes an 80% apoptosis reduction. E2F1 acts upstream of p53 since transcriptional activation of p53 target genes is also impaired. Yet, E2F1 deficiency does not accelerate tumor growth. Unlike normal cells, tumor cell proliferation is impaired without E2F1, counterbalancing the effect of apoptosis reduction. These studies may explain the apparent paradox that E2F1 can act as both an oncogene and a tumor suppressor in experimental systems.

Collaboration


Dive into the Terry Van Dyke's collaboration.

Top Co-Authors

Avatar

Yurong Song

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chaoying Yin

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chi-Ping Day

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Glenn Merlino

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lucy Lu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Zoe Weaver Ohler

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Rajaa El Meskini

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Dale O. Cowley

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chunyu Yang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Cari Graff-Cherry

Science Applications International Corporation

View shared research outputs
Researchain Logo
Decentralizing Knowledge