Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Open Microbiol J ; 11: 303-315, 2017.
Article in English | MEDLINE | ID: mdl-29290843

ABSTRACT

OBJECTIVE: Host derived markers on virally infected cells or virions may provide targets for the generation of antiviral agents. Recently, we identified phosphatidylserine (PS) as a host marker of virions and virally-infected cells. METHODS AND MATERIALS: Under normal physiological conditions, PS is maintained on the inner leaflet of the plasma membrane facing the cytosol. Following viral infection, activation or pre-apoptotic changes cause PS to become externalized. We have previously shown that bavituximab, a chimeric human-mouse antibody that binds PS complexed with ß2-glycoprotein I (ß2GP1), protected rodents against lethal Pichinde virus and cytomegalovirus infections. RESULTS: Here, we determined the antiviral activity of a fully human monoclonal antibody, PGN632, that directly binds to PS. Treatment with PGN632 protected 20% of guinea pigs with advanced infections of the hemorrhagic arenavirus, Pichinde, from death. Combining PGN632 with ribavirin improved the antiviral activity of both agents, such that the combination rescued 50% of animals from death. CONCLUSION: The major mechanisms of action of PGN632 appear to be opsonization of virus and antibody-dependent cellular cytotoxicity of virally-infected cells. PS-targeting agents may have utility in the treatment of viral diseases.

2.
Adv Clin Exp Med ; 25(3): 505-11, 2016.
Article in English | MEDLINE | ID: mdl-27629739

ABSTRACT

BACKGROUND: Doxorubicin and docetaxel-based chemotherapy regimens used in breast cancer patients are associated with high risk of febrile neutropenia (FN). Granulocyte colony-stimulating factors (G-CSF) are recommended for both treating and preventing chemotherapy-induced neutropenia. Increased thrombosis incidence in G-CSF treated patients was reported; however, the underlying mechanisms remain unclear. The principal activator of blood coagulation in cancer is tissue factor (TF). It additionally contributes to cancer progression and stimulates angiogenesis. The main proangiogenic factor is vascular endothelial growth factor (VEGF). OBJECTIVES: The aim of the study was to evaluate granulocyte-colony stimulating factor receptor (G-CSFR), tissue factor (TF) expression and vascular endothelial growth factor receptor (VEGF-R) bound VEGF in human breast cancer in loco. MATERIAL AND METHODS: G-CSFR, TF and VEGFR bound VEGF (VEGF: VEGFR) were assessed in 28 breast cancer tissue samples. Immunohistochemical (IHC) methodologies according to ABC technique and double staining IHC procedure were employed utilizing antibodies against G-CSFR, TF and VEGF associated with VEGFR (VEGF: VEGFR). RESULTS: Expression of G-CSFR was demonstrated in 20 breast cancer tissue specimens (71%). In 6 cases (21%) the expression was strong (IRS 9-12). Strong expression of TF was observed in all investigated cases (100%). Moreover, expression of VEGF: VEGFR was visualized in cancer cells (IRS 5-8). No presence of G-CSFR, TF or VEGF: VEGFR was detected on healthy breast cells. Double staining IHC studies revealed co-localization of G-CSFR and TF, G-CSFR and VEGF: VEGFR, as well as TF and VEGF: VEGFR on breast cancer cells and ECs. CONCLUSIONS: The results of the study indicate that GCSFR, TF and VEGF: VEGFR expression as well as their co-expression might influence breast cancer biology, and may increase thromboembolic adverse events incidence.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Thromboplastin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , Protein Binding
3.
PLoS One ; 11(7): e0158757, 2016.
Article in English | MEDLINE | ID: mdl-27463336

ABSTRACT

In the antiphospholipid syndrome (APS), patients produce antiphospholipid antibodies (aPL) that promote thrombosis and adverse pregnancy outcomes. Current therapy with anticoagulation is only partially effective and associated with multiple complications. We previously discovered that aPL recognition of cell surface ß2-glycoprotein I (ß2-GPI) initiates apolipoprotein E receptor 2 (apoER2)-dependent signaling in endothelial cells and in placental trophoblasts that ultimately promotes thrombosis and fetal loss, respectively. Here we sought to identify a monoclonal antibody (mAb) to ß2-GPI that negates aPL-induced processes in cell culture and APS disease endpoints in mice. In a screen measuring endothelial NO synthase (eNOS) activity in cultured endothelial cells, we found that whereas aPL inhibit eNOS, the mAb 1N11 does not, and instead 1N11 prevents aPL action. Coimmunoprecipitation studies revealed that 1N11 decreases pathogenic antibody binding to ß2-GPI, and it blocks aPL-induced complex formation between ß2-GPI and apoER2. 1N11 also prevents aPL antagonism of endothelial cell migration, and in mice it reverses the impairment in reendothelialization caused by aPL, which underlies the non-thrombotic vascular occlusion provoked by disease-causing antibodies. In addition, aPL inhibition of trophoblast proliferation and migration is negated by 1N11, and the more than 6-fold increase in fetal resorption caused by aPL in pregnant mice is prevented by 1N11. Furthermore, the promotion of thrombosis by aPL is negated by 1N11. Thus, 1N11 has been identified as an mAb that attenuates APS-related pregnancy complications and thrombosis in mice. 1N11 may provide an efficacious, mechanism-based therapy to combat the often devastating conditions suffered by APS patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antiphospholipid Syndrome/complications , Pregnancy Complications/prevention & control , Thrombosis/complications , Antiphospholipid Syndrome/prevention & control , Cells, Cultured , Endothelium, Vascular/pathology , Female , Fetal Resorption , Humans , Nitric Oxide Synthase Type III/metabolism , Pregnancy , Thrombosis/prevention & control , Trophoblasts/pathology
4.
Ann Surg Oncol ; 23(Suppl 5): 583-591, 2016 12.
Article in English | MEDLINE | ID: mdl-26847681

ABSTRACT

BACKGROUND: Currently, the only FDA-approved systemic therapy for hepatocellular carcinoma (HCC) is the multi-receptor tyrosine kinase inhibitor, sorafenib, which provides only modest clinical benefit. We recently showed that treatment with a phosphatidylserine (PS) targeting agent suppresses tumor growth by targeting tumor vasculature and reactivating antitumor immunity. METHODS: We tested the hypothesis that sorafenib increases PS exposure on tumor vasculature, thereby enhancing the antitumor efficacy of PS targeting. We evaluated the efficacy of combining a PS targeting agent (2aG4) with sorafenib in murine xenograft models of human HCC. RESULTS: Our results demonstrate that combination of 2aG4 and sorafenib had a superior therapeutic effect over single agent therapy. Mechanistic studies showed that sorafenib significantly increased PS exposure on tumor vasculature; the percentage of PS-positive vessels increased from 19 to 52, 23 to 68, and 30 to 55 % in PLC/PRF/5, C3A, and Huh7 tumors, respectively. Combination therapy significantly decreased tumor microvessel density and the level of M2 macrophages, while increasing the apoptotic index of tumor endothelial cells and the frequency of M1 macrophages. Furthermore, we report the findings of a Phase I clinical study of bavituximab, a chimeric version of 2aG4, combined with sorafenib in HCC patients. The Phase I results demonstrate the appropriate dose of bavituximab to be given with sorafenib in future clinical trials. CONCLUSIONS: Overall, these results strongly support the combination of bavituximab with sorafenib as a promising systemic therapeutic strategy for the treatment for advanced HCC patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phosphatidylserines/antagonists & inhibitors , Adult , Aged , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Carcinoma, Hepatocellular/blood supply , Endothelium/drug effects , Endothelium/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/blood supply , Male , Maximum Tolerated Dose , Mice , Middle Aged , Neoplasm Transplantation , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenotype , Phenylurea Compounds/administration & dosage , Phosphatidylserines/metabolism , Rituximab/pharmacology , Sorafenib , Xenograft Model Antitumor Assays
5.
Am J Nucl Med Mol Imaging ; 5(5): 493-503, 2015.
Article in English | MEDLINE | ID: mdl-26550540

ABSTRACT

Bavituximab is a chimeric monoclonal antibody with immune modulating and tumor-associated vascular disrupting properties demonstrated in models of non-small cell lung cancer (NSCLC). The molecular target of Bavituximab, phosphatidylserine (PS), is exposed on the outer leaflet of the membrane bi-layer of malignant vascular endothelial cells and tumor cells to a greater extent than on normal tissues. We evaluated the tumor-targeting properties of Bavituximab for imaging of NSCLC xenografts when radiolabeled with (111)In through conjugation with a bifunctional chelating agent, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). In vitro binding of (111)In-DOTA-Bavituximab to PS was determined by enzyme-linked immunosorbent assay (ELISA). Biodistribution of (111)In-DOTA-Bavituximab was conducted in normal rats, which provided data for dosimetry calculation. Single-photon emission computed tomography/computed tomography (SPECT/CT) imaging was performed in athymic nude rats bearing A549 NSCLC xenografts. At the molar conjugation ratio of 0.54 DOTA per Bavituximab, the PS binding affinity of (111)In-DOTA-Bavituximab was comparable to that of unmodified Bavituximab. Based on the quantitative SPECT/CT imaging data analysis, (111)In-DOTA-Bavituximab demonstrated tumor-specific uptake as measured by the tumor-tomuscle ratio, which peaked at 5.2 at 72 hr post-injection. In contrast, the control antibody only presented a contrast of 1.2 at the same time point.These findings may underlie the diagnostic efficacy and relative low rates of systemic vascular and immune-related toxicities of this immunoconjugate. Future applications of (111)In-DOTA-bavituximab may include prediction of efficacy, indication of tumor immunologic status, or characterization of radiographic findings.

6.
J Biomed Nanotechnol ; 10(5): 846-55, 2014 May.
Article in English | MEDLINE | ID: mdl-24734537

ABSTRACT

Phosphatidylserine (PS), normally restricted to the inner leaflet of the plasma membrane, becomes exposed on the outer surface of viable endothelial cells in tumor vasculature, but not in normal blood vessels. In the present study, we report the use of PGN635, a novel human monoclonal antibody that specifically targets PS, for in vivo molecular MRI of tumor vasculature. The F(ab')2 fragments of PGN635 were conjugated to polyethylene glycol (PEG) coated iron oxide nanoparticles (IO). Targeting specificity of the PS-targeted Nanoprobe, IO-PGN635F(ab')2 was first confirmed by in vitro MRI and histological staining. In vivo longitudinal MRI was then performed before and after i.v. injection of IO-PGN635F(ab')2 into mice bearing 4T1 breast tumors. T2-weighted MR images at 9.4 T revealed inhomogeneous signal loss in tumor as early as 2 h post injection. Furthermore, ionizing radiation induced a significant increase in PS exposure on tumor vascular endothelial cells, resulting in significantly enhanced and sustained tumor contrast (p < 0.05). Spatially heterogeneous MRI contrast correlated well with histological staining of tumor vascular endothelium. Our studies suggest that PS exposed within the lumen of tumor vasculature is a highly specific and useful biomarker for targeted MRI contrast agents.


Subject(s)
Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Molecular Imaging/methods , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphatidylserines/pharmacokinetics , Animals , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/pathology , Neoplasms, Experimental/radiotherapy , Neovascularization, Pathologic/radiotherapy , Radiotherapy, Conformal , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
7.
J Immunol Methods ; 407: 120-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24735771

ABSTRACT

The last decade has seen an exponential growth in the number of exosome-related publications. Although many of these studies have used exosomes from biological fluids (blood, and ascites or pleural effusions) the vast majority employed vesicles isolated from large volumes of tissue culture supernatants. While several techniques are available for their isolation, all require a significant reduction in volume to obtain sufficient concentrations for study. One approach is to concentrate the medium before proceeding with their isolation, however, these procedures are very time consuming and require specialized laboratory equipment. Here we provide a new and effective method for the isolation of tumor-derived exosomes based on "charge neutralization" with acetate. We show that titration of tissue culture supernatants with 0.1M acetate to pH4.75 results in immediate precipitation of virtually all the exosomes. The precipitated exosomes can be washed to remove residual media and are readily "resolubilized" upon resuspension in acetate-free buffer at neutral pH. This simple cost effective method significantly increases the yield of exosomes from an unlimited quantity of culture supernatants. Exosomes isolated by this technique are indistinguishable from exosomes recovered by direct ultracentrifugation.


Subject(s)
Exosomes/chemistry , Neoplasms/chemistry , Acetates/chemistry , Animals , Cell Line, Tumor , Chemical Precipitation , Culture Media, Conditioned/chemistry , Humans , Hydrogen-Ion Concentration , Mice , Salts/chemistry , Solubility , Ultracentrifugation
8.
PLoS One ; 8(12): e84864, 2013.
Article in English | MEDLINE | ID: mdl-24367699

ABSTRACT

Phosphatidylserine (PS) is an attractive target for imaging agents that identify tumors and assess their response to therapy. PS is absent from the surface of most cell types, but becomes exposed on tumor cells and tumor vasculature in response to oxidative stresses in the tumor microenvironment and increases in response to therapy. To image exposed PS, we used a fully human PS-targeting antibody fragment, PGN635 F(ab')2, that binds to complexes of PS and ß2-glycoprotein I. PGN635 F(ab')2 was labeled with the positron-emitting isotope iodine-124 ((124)I) and the resulting probe was injected into nude mice bearing subcutaneous or orthotopic human PC3 prostate tumors. Biodistribution studies showed that (124)I-PGN635 F(ab')2 localized with remarkable specificity to the tumors with little uptake in other organs, including the liver and kidneys. Clear delineation of the tumors was achieved by PET 48 hours after injection. Radiation of the tumors with 15 Gy or systemic treatment of the mice with 10 mg/kg docetaxel increased localization in the tumors. Tumor-to-normal (T/N) ratios were inversely correlated with tumor growth measured over 28 days. These data indicate that (124)I-PGN635 F(ab')2 is a promising new imaging agent for predicting tumor response to therapy.


Subject(s)
Immunoglobulin Fab Fragments , Iodine Radioisotopes , Phosphatidylserines/metabolism , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnosis , Animals , Docetaxel , Immunoglobulin Fab Fragments/metabolism , Iodine Radioisotopes/metabolism , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology , Taxoids , beta 2-Glycoprotein I/metabolism
9.
Cancer Immunol Res ; 1(4): 256-68, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24777853

ABSTRACT

Multiple tumor-derived factors are responsible for the accumulation and expansion of immune-suppressing myeloid-derived suppressor cells (MDSC) and M2-like tumor-associated macrophages (TAM) in tumors. Here, we show that treatment of tumor-bearing mice with docetaxel in combination with the phosphatidylserine-targeting antibody 2aG4 potently suppressed the growth and progression of prostate tumors, depleted M2-like TAMs, and MDSCs, and increased the presence of M1-like TAMs and mature dendritic cells in the tumors. In addition, the antibody markedly altered the cytokine balance in the tumor microenvironment from immunosuppressive to immunostimulatory. In vitro studies confirmed that 2aG4 repolarized TAMs from an M2- to an M1-like phenotype and drove the differentiation of MDSCs into M1-like TAMs and functional dendritic cells. These data suggest that phosphatidylserine is responsible for the expansion of MDSCs and M2-like TAMs in tumors, and that bavituximab, a phosphatidylserine-targeting antibody currently in clinical trials for cancer, could reverse this process and reactivate antitumor immunity.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Cell Differentiation , Macrophages/immunology , Phosphatidylserines/immunology , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytokines/immunology , Dendritic Cells/immunology , Docetaxel , Humans , Lymphocyte Activation , Male , Mice , Mice, SCID , Neoplasm Transplantation , Signal Transduction , Taxoids/therapeutic use , Tumor Microenvironment
10.
Biochem Pharmacol ; 84(11): 1534-40, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23022224

ABSTRACT

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.


Subject(s)
Vascular Endothelial Growth Factor A/pharmacokinetics , Alanine Transaminase/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Aspartate Aminotransferases/metabolism , Mice , Mice, Inbred BALB C , Tissue Distribution , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/therapeutic use
11.
MAbs ; 4(6): 648-52, 2012.
Article in English | MEDLINE | ID: mdl-23007482

ABSTRACT

Now in its 23rd and 10th years, respectively, the Antibody Engineering and Antibody Therapeutics conferences are the Annual Meeting of The Antibody Society. The scientific program covers the full spectrum of challenges in antibody research and development from basic science through clinical development. In this preview of the conferences, the chairs provide their thoughts on sessions that will allow participants to track emerging trends in (1) the development of next-generation immunomodulatory antibodies; (2) the complexity of the environment in which antibodies must function; (3) antibody-targeted central nervous system (CNS) therapies that cross the blood brain barrier; (4) the extension of antibody half-life for improved efficacy and pharmacokinetics (PK)/pharmacodynamics (PD); and (5) the application of next generation DNA sequencing to accelerate antibody research. A pre-conference workshop on Sunday, December 2, 2012 will update participants on recent intellectual property (IP) law changes that affect antibody research, including biosimilar legislation, the America Invents Act and recent court cases. Keynote presentations will be given by Andreas Plückthun (University of Zürich), who will speak on engineering receptor ligands with powerful cellular responses; Gregory Friberg (Amgen Inc.), who will provide clinical updates of bispecific antibodies; James D. Marks (University of California, San Francisco), who will discuss a systems approach to generating tumor targeting antibodies; Dario Neri (Swiss Federal Institute of Technology Zürich), who will speak about delivering immune modulators at the sites of disease; William M. Pardridge (University of California, Los Angeles), who will discuss delivery across the blood-brain barrier; and Peter Senter (Seattle Genetics, Inc.), who will present his vision for the future of antibody-drug conjugates. For more information on these meetings or to register to attend, please visit www.IBCLifeSciences.com/AntibodyEng or call 800-390-4078. Members of The Antibody Society and mAbs journal subscribers receive a 20% discount for meeting registration. To obtain this discount, email kdostie@ibcusa.com. mAbs is the official therapeutics journal of The Antibody Society and offers a discounted subscription to Society members for $49.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Immunotherapy/methods , Antibodies, Monoclonal/genetics , Antibodies, Neoplasm/genetics , California , Humans , Immunotherapy/trends , Immunotoxins , Protein Engineering , Societies, Scientific , Translational Research, Biomedical
12.
Neoplasia ; 14(4): 352-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22577350

ABSTRACT

Surgery is the most effective therapy for cancer in the United States, but disease still recurs in more than 40% of patients within 5 years after resection. Chemotherapy is given postoperatively to prevent relapses; however, this approach has had marginal success. After surgery, recurrent tumors depend on rapid neovascular proliferation to deliver nutrients and oxygen. Phosphatidylserine (PS) is exposed on the vascular endothelial cells in the tumor microenvironment but is notably absent on blood vessels in normal tissues. Thus, PS is an attractive target for cancer therapy after surgery. Syngeneic mice bearing TC1 lung cancer tumors were treated with mch1N11 (a novel mouse chimeric monoclonal antibody that targets PS), cisplatin (cis), or combination after surgery. Tumor relapses and disease progression were decreased 90% by combination therapy compared with a 50% response rate for cis alone (P = .02). Mice receiving postoperative mch1N11 had no wound-related complications or added systemic toxicity in comparison to control animals. Mechanistic studies demonstrated that the effects of mch1N11 were associated with a dense infiltration of inflammatory cells, particularly granulocytes. This strategy was independent of the adaptive immune system. Together, these data suggest that vascular-targeted strategies directed against exposed PS may be a powerful adjunct to postoperative chemotherapy in preventing relapses after cancer surgery.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endothelium, Vascular/drug effects , Neoplasm Recurrence, Local/prevention & control , Neoplasms, Experimental/drug therapy , Phosphatidylserines/antagonists & inhibitors , Animals , Cisplatin/administration & dosage , Combined Modality Therapy , Endothelium, Vascular/metabolism , Female , Flow Cytometry , Immunohistochemistry , Inflammation/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/surgery , Neovascularization, Pathologic/drug therapy
13.
J Cell Biochem ; 113(3): 956-66, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22034063

ABSTRACT

The mechanisms of nonclassical export of signal peptide-less proteins remain insufficiently understood. Here, we demonstrate that stress-induced unconventional export of FGF1, a potent and ubiquitously expressed mitogenic and proangiogenic protein, is associated with and dependent on the formation of membrane blebs and localized cell surface exposure of phosphatidylserine (PS). In addition, we found that the differentiation of promonocytic cells results in massive FGF1 release, which also correlates with membrane blebbing and exposure of PS. These findings indicate that the externalization of acidic phospholipids could be used as a pharmacological target to regulate the availability of FGF1 in the organism.


Subject(s)
Cell Surface Extensions/metabolism , Fibroblast Growth Factor 1/metabolism , Phosphatidylserines/analysis , Animals , Calcium/physiology , Cell Differentiation , Cell Membrane/metabolism , Cell Surface Extensions/chemistry , Cell Surface Extensions/ultrastructure , Cytoskeleton/metabolism , Humans , Mice , NIH 3T3 Cells , Phospholipid Transfer Proteins/physiology , Protein Transport/drug effects , Stress, Physiological , U937 Cells
14.
Transl Oncol ; 4(6): 355-64, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22191000

ABSTRACT

Phosphatidylserine (PS) is normally intracellular but becomes exposed on the luminal surface of vascular endothelial cells in tumors. It also becomes exposed on tumors cells responding to therapy. In the present study, we optically imaged exposed PS in vivo using PGN635, a novel monoclonal antibody that binds PS. The F(ab')(2) fragment of PGN635 was labeled with the near-infrared (NIR) dye, IRDye800CW. In vivo dynamic NIR imaging was performed after injection of 800CW-PGN635 into mice bearing radiation-treated or untreated U87 glioma xenografts growing subcutaneously or orthotopically. NIR optical imaging revealed a clear tumor contrast in nonirradiated subcutaneous U87 gliomas after injection of 800CW-PGN635. The tumor contrast was visible as early as 4 hours later and was maximal 24 hours later (tumor-to-normal tissue ratio [TNR] = 2.8 ± 0.7). Irradiation enhanced the tumor contrast at 24 hours (TNR = 4.0 ± 0.3). Similar results were observed for orthotopic gliomas. Localization of 800CW-PGN635 to tumors was antigen specific because 800CW-Aurexis, a control probe of irrelevant specificity, did not localize to the tumors, and preadministration of unlabeled PGN635 blocked the uptake of 800CW-PGN635. Fluorescence microscopy confirmed that 800CW-PGN635 was binding to PS-positive vascular endothelial cells in nonirradiated gliomas. Irradiation of the gliomas increased PS exposure on both tumor vascular endothelial cells and tumor cells and gave rise to an increase in tumor contrast with 800CW-PGN635 that was predictive of the reduction in tumor growth. 800CW-PGN635 may be a useful new imaging probe for detection of exposed PS in tumors responding to therapy.

15.
Clin Cancer Res ; 17(21): 6888-96, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21989064

ABSTRACT

PURPOSE: Bavituximab is a chimeric immunoglobulin G1 phosphatidylserine-targeting monoclonal antibody that triggers vascular disruption and enhances antitumor immune response. This phase I study assessed the safety and pharmacokinetics of bavituximab in patients with advanced solid tumors. EXPERIMENTAL DESIGN: Patients with refractory advanced solid tumors were enrolled into four sequential dose-escalation cohorts (0.1, 0.3, 1, or 3 mg/kg bavituximab weekly) with two dosing schedules. Patients in the 0.1 and 0.3 mg/kg cohorts received bavituximab on days 0, 28, 35, and 42. Patients in the 1 and 3 mg/kg cohorts were administered bavituximab on days 0, 7, 14, and 21. Safety, pharmacokinetics, and tumor response were assessed. RESULTS: Twenty-six patients were accrued. No maximum tolerated dose was reached. Six serious adverse events occurred in five patients, including one pulmonary embolism at 3 mg/kg, which was the only dose-limiting toxicity (DLT) in the study. Bavituximab half-life ranged from 37 to 47 hours, with no accumulation seen following administration of multiple doses. Activated partial thromboplastin time was modestly prolonged in vitro at the highest dose tested. As assessed on day 56, a total of 18 patients were evaluable for efficacy, of whom 10 had disease progression and none had an objective response. CONCLUSIONS: Bavituximab was well tolerated at doses ranging up to 3 mg/kg weekly. Pharmacokinetic studies support a weekly dosing regimen. Additional phase I and II clinical trials are in progress to investigate bavituximab in combination with chemotherapy and other molecularly targeted agents.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Blood Coagulation/drug effects , Blood Platelets/drug effects , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/blood , Neoplasms/drug therapy , Neoplasms/immunology , Phosphatidylserines/immunology
16.
BMC Cancer ; 11: 358, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849059

ABSTRACT

BACKGROUND: The fusion protein VEGF(121)/rGel composed of the growth factor VEGF(121) and the plant toxin gelonin targets the tumor neovasculature and exerts impressive anti-vascular effects. We have previously shown that VEGF(121)/rGel is cytotoxic to endothelial cells overexpressing VEGFR-2 but not to endothelial cells overexpressing VEGFR-1. In this study, we examined the basis for the specific toxicity of this construct and assessed its intracellular effects in vitro and in vivo. METHODS: We investigated the binding, cytotoxicity and internalization profile of VEGF(121)/rGel on endothelial cells expressing VEGFR-1 or VEGFR-2, identified its effects on angiogenesis models in vitro and ex vivo, and explored its intracellular effects on a number of molecular pathways using microarray analysis. RESULTS: Incubation of PAE/VEGFR-2 and PAE/VEGFR-1 cells with (125)I-VEGF(121)/rGel demonstrated binding specificity that was competed with unlabeled VEGF(121)/rGel but not with unlabeled gelonin. Assessment of the effect of VEGF(121)/rGel on blocking tube formation in vitro revealed a 100-fold difference in IC(50) levels between PAE/VEGFR-2 (1 nM) and PAE/VEGFR-1 (100 nM) cells. VEGF(121)/rGel entered PAE/VEGFR-2 cells within one hour of treatment but was not detected in PAE/VEGFR-1 cells up to 24 hours after treatment. In vascularization studies using chicken chorioallantoic membranes, 1 nM VEGF(121)/rGel completely inhibited bFGF-stimulated neovascular growth. The cytotoxic effects of VEGF(121)/rGel were not apoptotic since treated cells were TUNEL-negative with no evidence of PARP cleavage or alteration in the protein levels of select apoptotic markers. Microarray analysis of VEGF(121)/rGel-treated HUVECs revealed the upregulation of a unique "fingerprint" profile of 22 genes that control cell adhesion, apoptosis, transcription regulation, chemotaxis, and inflammatory response. CONCLUSIONS: Taken together, these data confirm the selectivity of VEGF(121)/rGel for VEGFR-2-overexpressing endothelial cells and represent the first analysis of genes governing intoxication of mammalian endothelial cells by a gelonin-based targeted therapeutic agent.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Aorta/cytology , Chick Embryo , Chickens , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Drug Delivery Systems , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Physiologic/drug effects , Recombinant Fusion Proteins/genetics , Ribosome Inactivating Proteins, Type 1/genetics , Swine , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
17.
Immunotherapy ; 3(8): 933-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21843081

ABSTRACT

Bavituximab is a chimeric monoclonal antibody directed against the membrane phospholipid phosphatidylserine. Phosphatidylserine exposure is increased on endothelial cells and apoptotic cancer cells in solid tumors, allowing tumor-specific targeting of bavituximab. Bavituximab binding results in tumor vessel occlusion and enhanced antitumor immunity. Preclinical investigations have demonstrated efficacy as monotherapy and in combination with other modalities against multiple cancer types. Phase I clinical trials of bavituximab monotherapy and in combination with chemotherapy in adults with refractory solid tumors have been completed. Phase II trials of bavituximab in combination with chemotherapy for the first- and second-line treatment of advanced non-small-cell lung cancer are currently ongoing. This article summarizes the preclinical development and clinical experience with bavituximab in non-small-cell lung cancer.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Phosphatidylserines/immunology , Animals , Antibodies, Monoclonal/adverse effects , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Trials, Phase II as Topic , Drug Evaluation, Preclinical , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology
18.
Vaccine ; 29(29-30): 4785-93, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21557977

ABSTRACT

Phosphatidylserine (PS), an anionic phospholipid normally restricted to the inner leaflet of the plasma membrane, is immunosuppressive when externalized on the outside of cell membranes. Exposed PS inhibits the maturation and function of dendritic cells (DCs), and induces the production of multiple immunosuppressive mediators. In the present study, we determined whether blocking these effects of PS while simultaneously introducing interleukin-2 (IL-2) could improve the immunogenicity of a whole-cell cancer vaccine. An immunocytokine (2aG4-IL2) was made by genetically linking IL-2 with a PS targeting antibody, 2aG4, that can block the immunosuppressive effects of PS. The 2aG4-IL2/4T1 vaccine was generated by coating the PS exposed on irradiated 4T1 cells with 2aG4-IL2. Tumor growth, spontaneous metastasis, and survival of vaccinated mice challenged with live 4T1 tumor cells were assessed. Eighty percent of mice inoculated with 2aG4-IL2/4T1 vaccine survived free of tumor, as compared with 20% in the 2aG4/4T1 group, 20% in the C44-IL2/4T1 group, and none in the C44/4T1 control group (P=0.001 for 2aG4-IL2/4T1 versus all others groups). The incidence, number of spontaneous lung metastases was significantly lower in the 2aG4-IL2/4T1 vaccinated group than in the other groups. Splenocytes from 2aG4-IL2/4T1 vaccinated mice had significantly higher 4T1 specific cytotoxicity and ability to secrete interferon-gamma (IFNγ) than did splenocytes from mice in the other groups. These results demonstrate that a potent whole-cell vaccine can be created by coating irradiated tumor cells with 2aG4-IL2. Such vaccine could potentially be an effective treatment modality for patients with residual disease or at "high-risk" for recurrence.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Breast Neoplasms/prevention & control , Cancer Vaccines/immunology , Interleukin-2/administration & dosage , Phosphatidylserines/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Cancer Vaccines/administration & dosage , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , Leukocytes, Mononuclear/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Protein Binding , Recombinant Proteins/administration & dosage , Spleen/immunology , Survival Analysis
19.
Anticancer Res ; 31(3): 843-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21498704

ABSTRACT

BACKGROUND: Colon cancer (CC) is frequently complicated by thromboembolic episodes. Thrombin plays a role in angiogenesis and among others induces the synthesis of vascular endothelial growth factor (VEGF) and its receptors (VEGFR-1 and VEGFR-2). The aim of this study was to assess the expression of prothrombin fragment F1+2 (F1+2), a byproduct in thrombin generation (indicating the presence of thrombin), in relation to the presence of VEGFR-2-bound VEGF (VEGF:VEGFR-2), as an indicator of VEGFR-2 activation in human CC tissue. MATERIALS AND METHODS: Immunohistochemical ABC and double staining studies were performed using antibodies against F1+2 and VEGF:VEGFR-2 in 59 specimens obtained from CC patients. RESULTS: Medium and high expression of both F1+2 and VEGF:VEGF2 in association with CC cells and endothelial cells was demonstrated. Moreover, coexpression of F1+2 and VEGF:VEGFR-2 was observed in the cells. CONCLUSION: The results may suggest a possible functional interaction between thrombin and VEGF-R2 stimulation in human CC in vivo.


Subject(s)
Colonic Neoplasms/metabolism , Peptide Fragments/metabolism , Prothrombin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adult , Colonic Neoplasms/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Protein Transport
20.
Neoplasia ; 13(4): 299-308, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21472134

ABSTRACT

We have previously shown that oxidative stress within the tumor microenvironment causes phosphatidylserine (PS) to redistribute from the inner to the outer membrane leaflet of the endothelial cells (EC) creating a highly specific marker for the tumor vasculature. Because the distribution of phosphatidylethanolamine (PE) and PS within the membrane is coregulated, we reasoned that PE would also be localized in the outer membrane leaflet of tumor EC. To demonstrate this, the PE-binding peptide duramycin was biotinylated and used to determine the distribution of PE on EC in vitro and in vivo. Exposure of cultured EC to hypoxia, acidity, reactive oxygen species, or irradiation resulted in the formation of membrane blebs that were intensely PE-positive. When biotinylated duramycin was intravenously injected into tumor-bearing mice, it preferentially localized to the luminal surface of the vascular endothelium. Depending on tumor type, 13% to 56% of the tumor vessels stained positive for PE. PE-positive vessels were observed in and around hypoxic regions of the tumor. With the exception of intertubular vessels of the kidney, normal vessels remained unstained. To test the potential of PE as a biomarker for imaging, duramycin was conjugated to the near-infrared fluorophore 800CW and used for optical imaging of RM-9 prostate carcinomas. The near-infrared probe was easily detected within tumors in live animals. These results show that PE, like PS, becomes exposed on tumor vascular endothelium of multiple types of tumors and holds promise as a biomarker for noninvasive imaging and drug targeting.


Subject(s)
Endothelium, Vascular/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Phosphatidylethanolamines/metabolism , Amino Acid Sequence , Animals , Antigens, Surface/metabolism , Bacteriocins/pharmacology , Cattle , Cell Line, Tumor , Drug Evaluation, Preclinical , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Models, Biological , Molecular Sequence Data , Neovascularization, Pathologic/pathology , Peptides/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL