Network


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

Hotspot


Dive into the research topics where Troy A. Luster is active.

Publication


Featured researches published by Troy A. Luster.


Clinical Cancer Research | 2009

Antiphosphatidylserine Antibody Combined with Irradiation Damages Tumor Blood Vessels and Induces Tumor Immunity in a Rat Model of Glioblastoma

Jin He; Yi Yin; Troy A. Luster; Linda Watkins; Philip E. Thorpe

Purpose: The vascular targeting antibody bavituximab is being combined with chemotherapy in clinical trials in cancer patients. Bavituximab targets the membrane phospholipid, phosphatidylserine, complexed with 2-glycoprotein I. Phosphatidylserine is normally intracellular but becomes exposed on the luminal surface of vascular endothelium in tumors. Phosphatidylserine exposure on tumor vessels is increased by chemotherapy and irradiation. Here, we determined whether treatment with the murine equivalent of bavituximab, 2aG4, combined with irradiation can suppress tumor growth in a rat model of glioblastoma. Experimental Design: F98 glioma cells were injected into the brains of syngeneic rats where they grow initially as a solid tumor and then infiltrate throughout the brain. Rats with established tumors were treated with 10 Gy whole brain irradiation and 2aG4. Results: Combination treatment doubled the median survival time of the rats, and 13 of animals were rendered disease free. Neither treatment given individually was as effective. We identified two mechanisms. First, irradiation induced phosphatidylserine exposure on tumor blood vessels and enhanced antibody-mediated destruction of tumor vasculature by monocytes/macrophages. Second, the antibody treatment induced immunity to F98 tumor cells, which are normally weakly immunogenic. Surviving rats were immune to rechallenge with F98 tumor cells. In vitro, 2aG4 enhanced the ability of dendritic cells (DCs) to generate F98-specific cytotoxic T cells. Phosphatidylserine exposure, which is induced on tumor cells by irradiation, likely suppresses tumor antigen presentation, and 2aG4 blocks this tolerogenic effect. Conclusion: Bavituximab combined with radiotherapy holds promise as a vascular targeting and immune enhancement strategy for the treatment of human glioblastoma. (Clin Cancer Res 2009;15(22):687180)


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.


Clinical Cancer Research | 2007

Radiation-Enhanced Vascular Targeting of Human Lung Cancers in Mice with a Monoclonal Antibody That Binds Anionic Phospholipids

Jin He; Troy A. Luster; Philip E. Thorpe

Purpose: New treatment strategies aimed at damaging tumor vasculature could potentially improve tumor response to radiation therapy. We recently showed that anionic phospholipids, principally phosphatidylserine, are specifically exposed on the luminal surface of tumor blood vessels. Here we tested the hypothesis that radiation therapy can increase phosphatidylserine exposure on lung tumor vasculature, thereby enhancing the antitumor properties of the anti-phosphatidylserine antibody 2aG4. Experimental Design: The therapeutic efficacy of radiation therapy plus 2aG4 was tested in nude mice bearing radiation-resistant A549 human lung tumors. Radiation-induced phosphatidylserine exposure on endothelial cells and A549 tumor cells was analyzed by immunofluoresence staining. The mechanism of the enhanced antitumor effect was examined by histology and antibody-dependent cell-mediated cytotoxicity experiments. Results: Focal irradiation of A549 human lung cancer xenografts increased the percentage of tumor vessels with exposed phosphatidylserine from 4% to 26%. Treatment of mice bearing A549 tumors with 2aG4 plus focal radiation therapy inhibited tumor growth by 80% and was superior to radiation therapy or 2aG4 alone (P < 0.01). Combination therapy reduced blood vessel density and enhanced monocyte infiltration into the tumor mass beyond that observed with individual treatments. In vitro, 2aG4 enhanced the ability of macrophages to kill endothelial cells with exposed phosphatidylserine in an Fc′-dependent manner. Conclusion: These results suggest that 2aG4 enhances the antitumor effects of radiation therapy by increasing antibody-dependent cell-mediated cytotoxicity toward tumor vessels with externalized phosphatidylserine. Bavituximab, a chimeric version of 2aG4 in clinical trials, has the potential to enhance the therapeutic efficacy of radiation therapy in lung cancer patients.


Journal of Biological Chemistry | 2006

Plasma Protein β-2-Glycoprotein 1 Mediates Interaction between the Anti-tumor Monoclonal Antibody 3G4 and Anionic Phospholipids on Endothelial Cells

Troy A. Luster; Jin He; Xianming Huang; Sourindra Maiti; Alan J. Schroit; Philip G. de Groot; Philip E. Thorpe

A promising target on tumor vasculature is phosphatidylserine (PS), an anionic phospholipid that resides exclusively on the inner leaflet of the plasma membrane of resting mammalian cells. We have shown previously that PS becomes exposed on the surface of endothelial cells (EC) in solid tumors. To target PS on tumor vasculature, the murine monoclonal antibody 3G4 was developed. 3G4 localizes to tumor vasculature, inhibits tumor growth, and enhances anti-tumor chemotherapies without toxicity in mice. A chimeric version of 3G4 is in clinical trials. In this study, we investigated the basis for the interaction between 3G4 and EC with surface-exposed PS. We demonstrate that antibody binding to PS is dependent on plasma protein β-2-glycoprotein 1 (β2GP1). β2GP1 is a 50-kDa glycoprotein that binds weakly to anionic phospholipids under physiological conditions. We show that 3G4 enhances binding of β2GP1 to EC induced to expose PS. We also show that divalent 3G4-β2GP1 complexes are required for enhanced binding, since 3G4 Fab′ fragments do not bind EC with exposed PS. Finally, we demonstrate that an artificial dimeric β2GP1 construct binds to EC with exposed PS in the absence of 3G4, confirming that antibody binding is mediated by dimerization of β2GP1. Together, these data indicate that 3G4 targets tumor EC by increasing the avidity of β2GP1 for anionic phospholipids through formation of multivalent 3G4-β2GP1 complexes.


Biochemical Pharmacology | 2012

Pharmacodynamics, tissue distribution, toxicity studies and antitumor efficacy of the vascular targeting fusion toxin VEGF121/rGel.

Khalid A. Mohamedali; Gang Niu; Troy A. Luster; Philip E. Thorpe; Haokao Gao; Xiaoyuan Chen; Michael G. Rosenblum

As a part of an ongoing assessment of its mechanism of action, we evaluated the in vivo pharmacokinetics, tissue distribution, toxicity and antitumor efficacy of VEGF(121)/rGel, a novel fusion protein. Pharmacokinetic studies showed that VEGF(121)/rGel cleared from the circulation in a biphasic manner with calculated half-lives of 0.3 and 6h for the alpha and beta phases, respectively. Pharmacokinetic evaluation of (64)Cu-DOTA-VEGF(121)/rGel showed relatively high blood retention 30 min after injection (26.6 ± 1.73% ID/g), dropping to 11.8 ± 2.83% and 0.82 ± 0.11% ID/g at 60 and 240 min post injection, respectively. Tissue uptake studies showed that kidneys, liver and tumor had the highest drug concentrations 48 h after administration. The maximum tolerated dose (MTD), based on a QOD×5 i.v. administration schedule, was found to be 18 mg/kg with an LD(50) of 25mg/kg. Treatment of BALB/c mice with VEGF(121)/rGel at doses up to the MTD caused no alterations in hematologic parameters. However, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) parameters increased in a dose-related manner. The no-observable-adverse-effect-level (NOAEL) was determined to be 20% of the MTD (3.6 mg/kg). VEGF(121)/rGel treatment of mice bearing orthotopically-placed MDA-MB-231 breast tumors caused increased vascular permeability of tumor tissue by 53% compared to saline-treated controls. Immunohistochemical analysis showed significant tumor hypoxia and necrosis as a consequence of vascular damage. In summary, VEGF(121)/rGel appears to be an effective therapeutic agent causing focused damage to tumor vasculature with minimal toxic effects to normal organs. This agent appears to be an excellent candidate for further clinical development.


Neoplasia | 2005

The Vascular-Ablative Agent VEGF121/rGel Inhibits Pulmonary Metastases of MDA-MB-231 Breast Tumors

Sophia Ran; Khalid A. Mohamedali; Troy A. Luster; Philip E. Thorpe; Michael G. Rosenblum


Archive | 2006

Fc-fusion constructs binding to phosphatidylserine and their therapeutic use

Philip E Thorpe; Troy A. Luster; Steven W. King


Cancer Research | 2007

Fusion proteins composed of mouse IgG2a Fc and mouse beta-2-glycoprotein 1 bind to endothelial cells with exposed phosphatidylserine

Troy A. Luster; Philip E. Thorpe


Cancer Research | 2018

Abstract 4935: High-throughput immune-oncology screen identifies EGFR inhibitors as potent enhancers of CTL antigen-specific tumor cell killing

Patrick Lizotte; Troy A. Luster; Megan E. Cavanaugh; Luke J. Taus; Abha Dhaneshwar; Naomi Mayman; Aaron Yang; Mark A. Bittinger; Paul Kirschmeier; Nathanael S. Gray; David A. Barbie; Pasi A. Jänne


Archive | 2006

Constructions fixant la phosphatidylserine et leur utilisation pour le traitement de maladies

Philip E Thorpe; Troy A. Luster; Steven W. King

Collaboration


Dive into the Troy A. Luster's collaboration.

Top Co-Authors

Avatar

Philip E. Thorpe

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jin He

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Khalid A. Mohamedali

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael G. Rosenblum

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Philip E Thorpe

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Steven W. King

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xianming Huang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam W. Beck

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alan J. Schroit

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge