Network


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

Hotspot


Dive into the research topics where Adam W. Beck is active.

Publication


Featured researches published by Adam W. Beck.


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.


BMC Cancer | 2008

The Adnectin CT-322 is a novel VEGF receptor 2 inhibitor that decreases tumor burden in an orthotopic mouse model of pancreatic cancer

Sean P. Dineen; Laura A. Sullivan; Adam W. Beck; Andrew F. Miller; Juliet G. Carbon; Roni Mamluk; Henry Wong; Rolf A. Brekken

BackgroundPancreatic cancer continues to have a 5-year survival of less than 5%. Therefore, more effective therapies are necessary to improve prognosis in this disease. Angiogenesis is required for tumor growth, and subsequently, mediators of angiogenesis are attractive targets for therapy. Vascular endothelial growth factor (VEGF) is a well-characterized mediator of tumor angiogenesis that functions primarily by binding and activating VEGF receptor 2 (VEGFR2). In this study, we evaluate the use of CT-322, a novel biologic (Adnectin). This small protein is based on a human fibronectin domain and has beneficial properties in that it is fully human, stable, and is produced in bacteria. CT-322 binds to and inhibits activation of VEGFR2.MethodsThe efficacy of CT-322 was evaluated in vivo using two orthotopic pancreatic tumor models. The first model was a human tumor xenograft where MiaPaCa-2 cells were injected into the tail of the pancreas of nude mice. The second model was a syngeneic tumor using Pan02 cells injected into pancreas of C57BL/6J mice. In both models, therapy was initiated once primary tumors were established. Mice bearing MiaPaCa-2 tumors were treated with vehicle or CT-322 alone. Gemcitabine alone or in combination with CT-322 was added to the treatment regimen of mice bearing Pan02 tumors. Therapy was given twice a week for six weeks, after which the animals were sacrificed and evaluated (grossly and histologically) for primary and metastatic tumor burden. Primary tumors were also evaluated by immunohistochemistry for the level of apoptosis (TUNEL), microvessel density (MECA-32), and VEGF-activated blood vessels (Gv39M).ResultsTreatment with CT-322 was effective at preventing pancreatic tumor growth and metastasis in orthotopic xenograft and syngeneic models of pancreatic cancer. Additionally, CT-322 treatment increased apoptosis, reduced microvessel density and reduced the number of VEGF-activated blood vessels in tumors. Finally, CT-322, in combination with gemcitabine was safe and effective at controlling the growth of syngeneic pancreatic tumors in immunocompetent mice.ConclusionWe conclude that CT-322 is an effective anti-VEGFR2 agent and that further investigation of CT-322 for the treatment of pancreatic cancer is warranted.


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.


Journal of Gastrointestinal Surgery | 2011

Accumulation of PRO-Cancer Cytokines in the Plasma Fraction of Stored Packed Red Cells

Douglas Benson; Adam W. Beck; Marie Schluterman Burdine; Rolf A. Brekken; Christopher C. Silliman; Carlton C. Barnett

IntroductionPerioperative blood transfusion has been linked to decreased survival in pancreatic cancer; however, the exact causal mechanism has not been elucidated. Allogeneic transfusions are known to expose patients to foreign cells and lipid mediators. We hypothesize that stored packed red cells (pRBCs) contain pro-cancer cytokines that augment tumor progression. We analyzed the plasma fraction of stored pRBCs for pro-cancer cytokines and evaluated the affect of both storage time and leukocyte reduction on these mediators.MethodsChemiarray™ analysis for pro-cancer cytokines was performed on the acellular plasma fraction of stored leukocyte-reduced (LR) and non-leukocyte-reduced (NLR) pRBCs at dayxa01 (D.1–fresh) and dayxa042 (D.42–outdate) of storage. Elevated expression of monocyte chemotactic protein-1 (MCP-1), regulated on activation, normal T cell expressed and secreted (RANTES), angiogenin, tumor necrosis factor-alpha (TNF-α), epidermal growth factor (EGF), and platelet-derived growth factor (PDGF) was found. Specific enzyme-linked immunosorbent assay was performed for each of these factors in LR and NLR blood at D.1, dayxa028, and D.42. Data were analyzed by ANOVA. A p value ≤0.05 was considered significant; Nu2009≥u20094 per group. Migration assays were performed using inhibitors of EGF (gefitinib) and PDGF (imatinib) on murine pancreatic adenocarcinoma cells (Pan02) exposed to D.1 and D.42 LR and NLR plasma. Proliferation assays were performed on Pan02 cells to test the inhibition of PDGF.ResultsMCP-1 levels increased with storage time in LR blood, 86.3u2009±u20096.3xa0pg/ml at D.1 vs. 121.2u2009±u20096.1xa0pg/ml at D.42 (pu2009=u20090.007), and NLR blood, 78.2u2009±u20097.3xa0pg/ml at D.1 vs. 647.8u2009±u2009220.7xa0pg/ml at D.42 (pu2009=u20090.02). RANTES levels are lower in LR compared to NLR stored blood, 3.0u2009±u20091.9 vs. 15.8u2009±u20090.7xa0pg/ml at D.42 (pu2009<u20090.001), but similar in D.1 blood, 13.8u2009±u20091.8xa0pg/ml in LR vs. 12.0u2009±u20091.6xa0pg/ml in NLR. Angiogenin levels were different between LR and NLR blood, 0xa0pg/ml (undetectable) vs. 44.2u2009±u20093.7xa0pg/ml (pu2009<u20090.001). Storage time did not affect concentration. TNF-α levels were not different between LR and NLR blood, and there was no storage time effect on concentration. EGF and PDGF levels increased with storage time in NLR blood only, 216.4u2009±u20093.8xa0pg/ml at D.1 vs. 1,436.4u2009±u2009238.6xa0pg/ml at D.42 for EGF (pu2009=u20090.001), and 61.6u2009±u20096.0xa0pg/ml at D.1 vs. 76.5u2009±u20091.7xa0pg/ml at D.42 (pu2009=u20090.003) for PDGF. Inhibition of EGF reduced migration in Pan02 cells treated with D.42 NLR blood, 245.9u2009±u200911.2 vs. 164.6u2009±u200910.6 cells/hpf (pu2009<u20090.001). Inhibition of PDGF had no effect on Pan02 migration and reduced cell proliferation in cells treated with D.42 NLR, 181.1u2009±u20091.5% over control vs. 157.5u2009±u20092.1% (pu2009<u20090.001).ConclusionPro-cancer cytokines that can augment tumor progression were identified in pRBCs. Some of these factors are present in fresh blood. The soluble factors identified herein may represent possible therapeutic targets to offset negative effects of transfusion. These data stress the need for efforts in cancer patients to reduce transfusion requirements if needed.


Breast Cancer Research and Treatment | 2006

Telomerase immortalization of human mammary epithelial cells derived from a BRCA2 mutation carrier

Cheryl M. Lewis; Brittney Shea Herbert; Dawei Bu; Shane Halloway; Adam W. Beck; Ashleen Shadeo; Cindy Zhang; Raheela Ashfaq; Jerry W. Shay; David M. Euhus

SummaryA novel human mammary epithelial cell line, HME348, was established from benign breast tissue from a 44-year-old germ-line BRCA2 mutation carrier with a history of stage 1 breast cancer. Mutation analysis showed that the patient had a known 6872del4 BRCA2 heterozygous mutation. The human mammary epithelial cells passaged in culture exhibited cellular replicative aging as evidenced by telomere shortening, lack of telomerase activity, and senescence. Ectopic expression of telomerase (hTERT) reconstituted telomerase activity in these cells and led to the immortalization of the cells. When grown on glass, the majority of immortalized HME348 cells expressed ESA and p63 with a small population also expressing EMA. In three-dimensional Matrigel® culture, HME348 cells formed complex branching acini structures that expressed luminal (EMA, CK18) and myoepithelial (p63, CALLA, CK14) markers. Three clones derived from this culture were also p63+/ESA+/EMA+/− on glass but formed similar acinar structures with both luminal and myoepithelial cell differentiation in Matrigel® confirming the mammary progenitor nature of these cells. Additionally, the experimentally immortalized HME348 cells formed acini in cleared mammary fat pads in vivo. As this is the first report establishing and characterizing a benign human mammary epithelial cell line derived from a BRCA2 patient without the use of viral oncogenes, these cells may be useful for the study of BRCA2 function in breast morphogenesis and carcinogenesis.


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 Vascular Surgery | 2008

Aortic reconstruction with femoral-popliteal vein: Graft stenosis incidence, risk and reintervention

Adam W. Beck; Erin H. Murphy; Jennie A. Hocking; Carlos H. Timaran; Frank R. Arko; G. Patrick Clagett


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 Adam W. Beck's collaboration.

Top Co-Authors

Avatar

Rolf A. Brekken

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jason B. Fleming

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shane E. Holloway

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

Sean P. Dineen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlos H. Timaran

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cheryl M. Lewis

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
Researchain Logo
Decentralizing Knowledge