Network


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

Hotspot


Dive into the research topics where Shane E. Holloway is active.

Publication


Featured researches published by Shane E. Holloway.


Molecular Cancer Research | 2005

Molecular Consequences of Silencing Mutant K-ras in Pancreatic Cancer Cells: Justification for K-ras-Directed Therapy

Jason B. Fleming; Guo Liang Shen; Shane E. Holloway; Mishel Davis; Rolf A. Brekken

Mutation of the K-ras gene is an early event in the development of pancreatic adenocarcinoma and, therefore, RNA interference (RNAi) directed toward mutant K-ras could represent a novel therapy. In this study, we examine the phenotypic and molecular consequences of exposure of pancreatic tumor cells to mutant-specific K-ras small interfering RNA. Specific reduction of activated K-ras via RNAi in Panc-1 and MiaPaca-2 cells resulted in cellular changes consistent with a reduced capacity to form malignant tumors. These changes occur through distinct mechanisms but likely reflect an addiction of each cell line to oncogene stimulation. Both cell lines show reduced proliferation after K-ras RNAi, but only MiaPaca-2 cells showed increased apoptosis. Both cell lines showed reduced migration after K-ras knockdown, but changes in integrin levels were not consistent between the cell lines. Both cell lines showed alteration of the level of GLUT-1, a metabolism-associated gene that is downstream of c-myc, with Panc-1 cells demonstrating decreased GLUT-1 levels, whereas MiaPaca-2 cells showed increased levels of expression after K-ras knockdown. Furthermore, after K-ras RNAi, there was a reduction in angiogenic potential of both Panc-1 and MiaPaca-2 cells. Panc-1 cells increased the level of expression of thrombospondin-1, an endogenous inhibitor of angiogenesis, whereas MiaPaca-2 cells decreased the production of vascular endothelial growth factor, a primary stimulant of angiogenesis in pancreatic tumors. We have found that silencing mutant K-ras through RNAi results in alteration of tumor cell behavior in vitro and suggests that targeting mutant K-ras specifically might be effective against pancreatic cancer in vivo.


Cancer Research | 2008

Vascular Endothelial Growth Factor Receptor 2 Mediates Macrophage Infiltration into Orthotopic Pancreatic Tumors in Mice

Sean P. Dineen; Kristi D. Lynn; Shane E. Holloway; Andrew F. Miller; James P. Sullivan; David S. Shames; Adam W. Beck; Carlton C. Barnett; Jason B. Fleming; Rolf A. Brekken

Macrophages are an abundant inflammatory cell type in the tumor microenvironment that can contribute to tumor growth and metastasis. Macrophage recruitment into tumors is mediated by multiple cytokines, including vascular endothelial growth factor (VEGF), which is thought to function primarily through VEGF receptor (VEGFR) 1 expressed on macrophages. Macrophage infiltration is affected by VEGF inhibition. We show that selective inhibition of VEGFR2 reduced macrophage infiltration into orthotopic pancreatic tumors. Our studies show that tumor-associated macrophages express VEGFR2. Furthermore, peritoneal macrophages from tumor-bearing animals express VEGFR2, whereas peritoneal macrophages from non-tumor-bearing animals do not. To our knowledge, this is the first time that tumor-associated macrophages have been shown to express VEGFR2. Additionally, we found that the cytokine pleiotrophin is sufficient to induce VEGFR2 expression on macrophages. Pleiotrophin has previously been shown to induce expression of endothelial cell markers on macrophages and was present in the microenvironment of orthotopic pancreatic tumors. Finally, we show that VEGFR2, when expressed by macrophages, is essential for VEGF-stimulated migration of tumor-associated macrophages. In summary, tumor-associated macrophages express VEGFR2, and selective inhibition of VEGFR2 reduces recruitment of macrophages into orthotopic pancreatic tumors. Our results show an underappreciated mechanism of action that may directly contribute to the antitumor activity of angiogenesis inhibitors that block the VEGFR2 pathway.


International Journal of Cancer | 2006

Combination of a monoclonal anti‐phosphatidylserine antibody with gemcitabine strongly inhibits the growth and metastasis of orthotopic pancreatic tumors in mice

Adam W. Beck; Troy A. Luster; Andrew F. Miller; Shane E. Holloway; Chris R. Conner; Carlton C. Barnett; Philip E. Thorpe; Jason B. Fleming; Rolf A. Brekken

Pancreatic cancer continues to have a dismal prognosis and novel therapy is needed. In this study, we evaluate a promising new target for therapy, phosphatidylserine (PS). PS is an anionic phospholipid located normally on the inner leaflet of the plasma membrane in mammalian cells. In the tumor microenvironment, PS becomes externalized on vascular endothelium. The monoclonal antibody 3G4 binds PS and promotes an inflammatory response against tumor blood vessels, resulting in reduction of tumor growth. Mice with orthotopic pancreatic tumors were treated with 3G4, gemcitabine or a combination of both drugs. Tumor burden including pancreas weight and metastatic lesions (liver, lymph node and peritoneal) were reduced 3‐ to 5‐fold by the combination therapy as compared with 1.5‐ to 2‐fold with 3G4 and gemcitabine alone, respectively. Treatment of tumor‐bearing animals with the combination therapy increased macrophage infiltration into the tumor mass 10‐fold and reduced microvessel density in the tumor by 2.5‐fold compared with tumors from untreated animals. Gemcitabine alone and 3G4 alone were less effective than the combination of the 2 agents together. The additive therapeutic effect of both agents appears to be because chemotherapy increases PS exposure on tumor vascular endothelium and amplifies the target for attack by 3G4. In conclusion, 3G4 enhanced the anti‐tumor and anti‐metastatic activity of gemcitabine without contributing to toxicity.


Experimental Biology and Medicine | 2008

Forced expression of MMP9 rescues the loss of angiogenesis and abrogates metastasis of pancreatic tumors triggered by the absence of host SPARC

Shanna A. Arnold; Emilia Mira; Sabeeha Muneer; Grzegorz Korpanty; Adam W. Beck; Shane E. Holloway; Santos Mañes; Rolf A. Brekken

Pancreatic adenocarcinoma is characterized by desmoplasia, local invasion, and metastasis. These features are regulated in part by MMP9 and SPARC. To explore the interaction of SPARC and MMP9 in cancer, we first established orthotopic pancreatic tumors in SPARC-null and wild-type mice with the murine pancreatic adenocarcinoma cell line, PAN02. MMP9 expression was higher in tumors from wild-type compared to SPARC-null mice. Coincident with lower MMP9 expression, tumors grown in SPARC-null mice were significantly larger, had decreased ECM deposition and reduced microvessel density compared to wild-type controls. In addition, metastasis was enhanced in the absence of host SPARC. Therefore, we next analyzed the orthotopic tumor growth of PAN02 cells transduced with MMP9 or a control empty vector. Forced expression of MMP9 by the PAN02 cells resulted in larger tumors in both wild-type and SPARC-null animals compared to empty vector controls and further diminished ECM deposition. Importantly, forced expression of MMP9 within the tumor reversed the decrease in angiogenesis and abrogated the metastatic potential displayed by control tumors grown in SPARC-null mice. Finally, contrary to the in vivo results, MMP9 increased cell migration in vitro, which was blocked by the addition of SPARC. These results suggest that SPARC and MMP9 interact to regulate many stages of tumor progression including ECM deposition, angiogenesis and metastasis.


Annals of Surgical Oncology | 2006

Selective Blockade of Vascular Endothelial Growth Factor Receptor 2 With an Antibody Against Tumor-Derived Vascular Endothelial Growth Factor Controls the Growth of Human Pancreatic Adenocarcinoma Xenografts

Shane E. Holloway; Adam W. Beck; Latha Shivakumar; Jessica Shih; Jason B. Fleming; Rolf A. Brekken

BackgroundVascular endothelial growth factor (VEGF), a key regulator of angiogenesis, is critical for growth of human pancreatic adenocarcinoma. Preclinical studies demonstrate that blockade of VEGF activity can control the growth of pancreatic tumors in mice. In this study, we evaluated the efficacy of 2C3, an antibody that inhibits VEGF receptor 2 activation by human VEGF, to inhibit the growth of human pancreatic adenocarcinoma in mice.MethodsHuman pancreatic cancer cell lines (MiaPaca-2, Panc-1, and Capan-1) were used to establish xenografts in nu/nu mice. The expression of VEGF and its receptors was determined in each cell line. Proliferation of tumor cells in vitro and tumor growth in vivo in the presence of 2C3 or a control antibody was evaluated. The effect of 2C3 on tumor weight, total vessel density, number of pericyte-associated vessels, and tumor perfusion was determined, and the level of 2C3 in the serum of animals was measured by enzyme-linked immunosorbent assay.Results2C3 did not affect the proliferation of cells in culture. 2C3 was present and active in the serum of tumor-bearing animals treated with 2C3, and these animals showed a decrease in tumor burden compared with control-treated mice. Therapy with 2C3 resulted in reduced vascular function, measured by a decrease in vessel density and in the percentage of vessels associated with pericytes. Furthermore, tumors derived from Capan-1 cells demonstrated decreased perfusion after treatment with 2C3.ConclusionsBlockade of VEGF receptor 2 activation by tumor-derived VEGF decreases tumor vessel function and growth of some human pancreatic adenocarcinoma cell lines in mice.


Cancer | 2010

Intravenous delivery of the plasma fraction of stored packed erythrocytes promotes pancreatic cancer growth in immunocompetent mice

Carlton C. Barnett; Adam W. Beck; Shane E. Holloway; Marguerite Kehler; Marie K. Schluterman; Rolf A. Brekken; Jason B. Fleming; Christopher C. Silliman

Perioperative blood transfusion in pancreatic cancer patients has been linked to decreased survival; however, a causal mechanism has not been determined. During the processing and storage of packed erythrocytes, biologically active molecules accumulated in the acellular plasma fraction; therefore, the authors hypothesized that the plasma fraction of stored packed erythrocytes promoted tumor progression.


International Journal of Gastrointestinal Cancer | 2003

A clinically relevant model of human pancreatic adenocarcinoma identifies patterns of metastasis associated with alterations of the TGF-β/Smad4 signaling pathway

Shane E. Holloway; Mishel Davis; Raffat Jaber; Jason B. Fleming

AbstractGenetic alterations impacting the TGF-β/Smad4 pathway are found in nearly all pancreatic adenocarcinomas, and recent reports have identified a relationship between DPC4/Smad4 expression and patient survival. In this study we use a clinically relevant animal model of pancreatic cancer to examine the impact of these genetic changes on the biology of pancreatic cancer. Methods: Using high-density oligonucleotide DNA microarray technology, a comprehensive examination of the components of the TGF-β/Smad4 pathway was performed on three human pancreatic adenocarcinoma cell lines. The in vitro and in vivo growth characteristics of these cell lines was then compared. Finally, using a clinically relevant orthotopic xenograft model of pancreatic cancer, primary tumor growth and metastases were measured for pancreatic tumors derived from each cell line. Results: Examination of the TGF-β/Smad4 pathway components identified that these three cell lines possess molecular profiles consistent with ∼90% of pancreatic adenocarcinoma tumors in patients. A significant discrepancy between in vitro and in vivo growth characteristics of each cell line was identified. When tumors from each cell line were established in nu/nu mice, each cell line exhibited distinct metastatic profiles. Data from these studies is consistent, with clinical observations concerning DPC4/Smad4 and patient outcome. Conclusion: Using an orthotopic model of tumor growth and metastasis identifies distinct metastatic profiles associated with molecular alterations of the TGF-β/Smad4 pathway and provides insight with regard to the biologic consequences of these changes.


Journal of The American College of Surgeons | 2005

Increased expression of Cyr61 (CCN1) identified in peritoneal metastases from human pancreatic cancer

Shane E. Holloway; Adam W. Beck; Luc Girard; M. Raffat Jaber; Carlton C. Barnett; Rolf A. Brekken; Jason B. Fleming


Journal of Surgical Research | 2002

Health-Related Quality of Life and Postoperative Length of Stay for Patients with Colorectal Cancer

Shane E. Holloway; George A. Sarosi; Lawrence T. Kim; Fiemu E. Nwariaku; Grant E. O'Keefe; Linda S. Hynan; Charlene Jones; Thomas Anthony


Journal of Surgical Research | 2004

Ultrasound-guided placement of bioresorbable fibers into pancreatic adenocarcinoma tumors achieves prolonged adenoviral gene transfer

Adam W. Beck; Shane E. Holloway; J.M. Ganter; R.C. Eberhart; K.D. Nelson; Jason B. Fleming

Collaboration


Dive into the Shane E. Holloway's collaboration.

Top Co-Authors

Avatar

Jason B. Fleming

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rolf A. Brekken

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adam W. Beck

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Carlton C. Barnett

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Andrew F. Miller

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mishel Davis

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sean P. Dineen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Charlene Jones

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chris R. Conner

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge