60 results on '"Nakada MT"'
Search Results
2. CNTO 95, a fully human anti alphav integrin antibody, inhibits cell signaling, migration, invasion, and spontaneous metastasis of human breast cancer cells.
- Author
-
Chen Q, Manning CD, Millar H, McCabe FL, Ferrante C, Sharp C, Shahied-Arruda L, Doshi P, Nakada MT, and Anderson GM
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Breast Neoplasms pathology, Cell Adhesion drug effects, Cell Line, Tumor, Female, Focal Adhesion Kinase 1 metabolism, Humans, Lung Neoplasms metabolism, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Mice, Mice, SCID, Neoplasm Invasiveness, Paxillin metabolism, Phosphorylation, Signal Transduction drug effects, Transplantation, Heterologous, Vitronectin metabolism, Antibodies, Monoclonal pharmacology, Breast Neoplasms metabolism, Cell Movement drug effects, Integrin alphaV immunology
- Abstract
CNTO 95 is a fully human monoclonal antibody that recognizes alphav integrins. Previous studies have shown that CNTO 95 exhibits both anti-tumor and anti-angiogenic activities (Trikha M et al., Int J Cancer 110:326-335, 2004). In this study we investigated the biological activities of CNTO 95 on breast tumor cells both in vitro and in vivo. In vitro treatment with CNTO 95 decreased the viability of breast tumor cells adhering to vitronectin. CNTO 95 inhibited tumor cell adhesion, migration, and invasion in vitro. CNTO 95 treatment also induced tyrosine dephosphorylation of focal adhesion kinase (FAK), and the docking protein paxillin that recruits both structural and signaling molecules to focal adhesions (Turner CE, Int J Biochem Cell Biol 30:955-959, 1998; O'Neil GM et al., Trends Cell Biol 10:111-119, 2000). These results suggest that CNTO 95 inhibits breast tumor cell growth, migration and invasion by interruption of alphav integrin mediated focal adhesions and cell motility signals. In vivo studies of CNTO 95 were conducted in an orthotopic breast tumor xenograft model. Treatment with CNTO 95 resulted in significant inhibition of both tumor growth and spontaneous metastasis of MDA-MB-231 cells to the lungs. CNTO 95 also inhibited lung metastasis in a separate experimental (tail vein injection) model of metastasis. The results presented here demonstrate the anti-tumor and anti-metastatic activities of CNTO 95 in breast cancer models and provide insight into the cellular and molecular mechanisms mediating its inhibitory effects on metastasis.
- Published
- 2008
- Full Text
- View/download PDF
3. Endothelial targeting of semi-permeable polymer nanocarriers for enzyme therapies.
- Author
-
Dziubla TD, Shuvaev VV, Hong NK, Hawkins BJ, Madesh M, Takano H, Simone E, Nakada MT, Fisher A, Albelda SM, and Muzykantov VR
- Subjects
- Animals, Antioxidants metabolism, Catalase chemistry, Catalase therapeutic use, Cell Adhesion Molecules metabolism, Cells, Cultured, Humans, Male, Mice, Mice, Inbred C57BL, Catalase metabolism, Cell Membrane Permeability, Drug Delivery Systems, Endothelial Cells metabolism, Nanostructures chemistry, Polymers chemistry, Polymers metabolism
- Abstract
The medical utility of proteins, e.g. therapeutic enzymes, is greatly restricted by their labile nature and inadequate delivery. Most therapeutic enzymes do not accumulate in their targets and are inactivated by proteases. Targeting of enzymes encapsulated into substrate-permeable polymer nano-carriers (PNC) impermeable for proteases might overcome these limitations. To test this hypothesis, we designed endothelial targeted PNC loaded with catalase, an H(2)O(2)-detoxifying enzyme, and tested if this approach protects against vascular oxidative stress, a pathological process implicated in ischemia-reperfusion and other disease conditions. Encapsulation of catalase (MW 247 kD), peroxidase (MW 42 kD) and xanthine oxidase (XO, MW 300 kD) into approximately 300 nm diameter PNC composed of co-polymers of polyethylene glycol and poly-lactic/poly-glycolic acid (PEG-PLGA) was in the range approximately 10% for all enzymes. PNC/catalase and PNC/peroxidase were protected from external proteolysis and exerted enzymatic activity on their PNC diffusible substrates, H(2)O(2) and ortho-phenylendiamine, whereas activity of encapsulated XO was negligible due to polymer impermeability to the substrate. PNC targeted to platelet-endothelial cell (EC) adhesion molecule-1 delivered active encapsulated catalase to ECs and protected the endothelium against oxidative stress in cell culture and animal studies. Vascular targeting of PNC-loaded detoxifying enzymes may find wide medical applications including management of oxidative stress and other toxicities.
- Published
- 2008
- Full Text
- View/download PDF
4. Alpha-v integrins as therapeutic targets in oncology.
- Author
-
Nemeth JA, Nakada MT, Trikha M, Lang Z, Gordon MS, Jayson GC, Corringham R, Prabhakar U, Davis HM, and Beckman RA
- Subjects
- Angiogenesis Inhibitors pharmacology, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Remodeling drug effects, Clinical Trials, Phase II as Topic, Humans, Integrin alphaV chemistry, Neoplasm Metastasis, Neoplasms etiology, Signal Transduction, Integrin alphaV physiology, Neoplasms drug therapy
- Abstract
Integrins are heterodimeric cell adhesion receptors that mediate intercellular communication through cell-extracellular matrix interactions and cell-cell interactions. Integrins have been demonstrated to play a direct role in cancer progression, specifically in tumor cell survival, tumor angiogenesis, and metastasis. Therefore, agents targeted against integrin function have potential as effective anticancer therapies. Numerous anti-integrin agents, including monoclonal antibodies and small-molecule inhibitors, are in clinical development for the treatment of solid and hematologic tumors. This review focuses on the role of alpha(v) integrins in cancer progression, the current status of integrin-targeted agents in development, and strategies for the clinical development of anti-integrin therapies.
- Published
- 2007
- Full Text
- View/download PDF
5. Alphav integrin-targeted immunoconjugates regress established human tumors in xenograft models.
- Author
-
Chen Q, Millar HJ, McCabe FL, Manning CD, Steeves R, Lai K, Kellogg B, Lutz RJ, Trikha M, Nakada MT, and Anderson GM
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Humanized, Antibody Specificity, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Line, Tumor, Drug Delivery Systems, Female, Flow Cytometry, Humans, Immunoconjugates chemistry, Immunotherapy, Mice, Rats, Xenograft Model Antitumor Assays, Antibodies, Monoclonal administration & dosage, Antineoplastic Agents administration & dosage, Immunoconjugates administration & dosage, Integrin alpha5 immunology, Neoplasms, Experimental drug therapy
- Abstract
Purpose: Targeted delivery of cytotoxic agents to solid tumors through cell surface antigens can potentially reduce systemic toxicity and increase the efficacy of the targeted compounds. The purpose of this study was to show the feasibility of treating solid tumors by targeting alpha(v) integrins with antibody-maytansinoid conjugates and to test the relative in vivo activities of several linker-maytansinoid chemistries., Experimental Design: CNTO 364, CNTO 365, and CNTO 366 are targeted cytotoxic agents created by conjugating the CNTO 95 anti-alpha(v) integrin antibody with three distinct maytansinoid-linker structures. These structures were designed to have varying degrees of chemical substitution surrounding the disulfide bond linking the cytotoxic agent to the antibody. A model conjugate was shown to be specifically cytotoxic in vitro and highly active against established human tumor xenografts in immunocompromised rats. The in vivo antitumor activities of CNTO 364, CNTO 365, and CNTO 366 were compared in rat xenograft models., Results: CNTO 365, with a linker chemistry of expected intermediate stability, was shown to be substantially more active than the other two conjugates with lesser or greater substitution around the disulfide linkage., Conclusion: CNTO 95-maytansinoid immunoconjugates are potent antitumor agents against alpha(v) integrin-expressing human carcinomas. These studies show for the first time the feasibility of targeting alpha(v) integrins on solid tumors with tumor-activated prodrugs. The DM4 linker-maytansinoid configuration of CNTO 365 was substantially more active in the models tested here when compared with alternative configurations with greater or lesser chemical substitution surrounding the linker.
- Published
- 2007
- Full Text
- View/download PDF
6. Phase I evaluation of a fully human anti-alphav integrin monoclonal antibody (CNTO 95) in patients with advanced solid tumors.
- Author
-
Mullamitha SA, Ton NC, Parker GJ, Jackson A, Julyan PJ, Roberts C, Buonaccorsi GA, Watson Y, Davies K, Cheung S, Hope L, Valle JW, Radford JA, Lawrance J, Saunders MP, Munteanu MC, Nakada MT, Nemeth JA, Davis HM, Jiao Q, Prabhakar U, Lang Z, Corringham RE, Beckman RA, and Jayson GC
- Subjects
- Adult, Aged, Dose-Response Relationship, Drug, Female, Humans, Immunohistochemistry, Integrin alphaV immunology, Magnetic Resonance Imaging, Male, Maximum Tolerated Dose, Middle Aged, Positron-Emission Tomography, Treatment Outcome, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Integrin alphaV metabolism, Neoplasms drug therapy
- Abstract
Purpose: A fully human monoclonal antibody to anti-alpha(v) integrins (CNTO 95) has been shown to inhibit angiogenesis and tumor growth in preclinical studies. We assessed the safety and pharmacokinetics of CNTO 95 in patients with advanced refractory solid tumors., Experimental Design: In this phase I trial, CNTO 95 (0.1, 0.3, 1.0, 3.0, and 10.0 mg/kg) was infused on days 0, 28, 35, and 42, and clinical assessments, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), and [(18)F]-2-fluorodeoxyglucose positron emission tomography (FDG-PET) were done. Patients achieving stable disease or better were eligible for extended dosing every 3 weeks for up to 12 months., Results: Among the 24 enrolled patients, CNTO 95 was associated with one episode of grade III and four episodes of grade II infusion-related fever (all responded to acetaminophen). Of the six patients who received extended dosing, one patient (10.0 mg/kg), with cutaneous angiosarcoma, had a 9-month partial response. Pre- and post-treatment lesion biopsies confirmed tumor cell alpha(v) integrin expression, as well as CNTO 95 penetration of the tumor and localization to tumor cells in association with reduced bcl-2 expression. A lesion in one patient (10.0 mg/kg) with stable ovarian carcinosarcoma was no longer detectable by FDG-PET by day 49. Exposure to CNTO 95 seemed to increase in a greater-than-dose-proportional manner; dose-dependent mean half-life ranged from 0.26 to 6.7 days., Conclusions: CNTO 95 was generally well tolerated. Six patients received extended therapy, including one patient with a prolonged response. Biopsy data confirmed tumor localization and pharmacodynamic activity.
- Published
- 2007
- Full Text
- View/download PDF
7. CNTO 859, a humanized anti-tissue factor monoclonal antibody, is a potent inhibitor of breast cancer metastasis and tumor growth in xenograft models.
- Author
-
Ngo CV, Picha K, McCabe F, Millar H, Tawadros R, Tam SH, Nakada MT, and Anderson GM
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacology, Breast Neoplasms pathology, Carcinoma prevention & control, Carcinoma secondary, Cell Proliferation drug effects, Female, Humans, Immunoglobulin G immunology, Immunoglobulin G pharmacology, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Mice, Mice, Inbred Strains, Xenograft Model Antitumor Assays, Antibodies, Monoclonal therapeutic use, Breast Neoplasms drug therapy, Carcinoma drug therapy, Immunoglobulin G therapeutic use, Lung Neoplasms drug therapy, Thromboplastin immunology
- Abstract
Thromboembolic complications are frequently associated with advanced cancer. Interestingly, one of the major initiators of blood coagulation, tissue factor (TF), is reported to be overexpressed in several tumor types and can be found on both tumor cells and tumor vasculature. Although the exact mechanisms have yet to be elucidated, TF expressed on tumor cells can trigger intracellular signaling events through various pathways that can lead to tumor angiogenesis, proliferation, and metastasis. There exists preclinical evidence that disruption of TF dependent signaling can effectively inhibit tumor cell migration, metastasis, and angiogenesis. Here, we report for the first time that an antibody to tissue factor can also prevent tumor growth in vivo. Prophylactic administration of CNTO 859, a humanized anti-human TF antibody, was shown to inhibit experimental lung metastasis of MDA-MB-231 human breast carcinoma cells by over 99% compared to a control antibody. Furthermore, therapeutic doses of CNTO 859 were shown to reduce tumor incidence and growth of orthotopically implanted MDA-MB-231 cells., ((c) 2006 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
8. A review of antibody therapeutics and antibody-related technologies for oncology.
- Author
-
Scallon BJ, Snyder LA, Anderson GM, Chen Q, Yan L, Weiner LM, and Nakada MT
- Subjects
- Humans, Antibodies therapeutic use, Immunotherapy methods, Neoplasms therapy
- Published
- 2006
- Full Text
- View/download PDF
9. Regulation of vascular endothelial growth factor expression by EMMPRIN via the PI3K-Akt signaling pathway.
- Author
-
Tang Y, Nakada MT, Rafferty P, Laraio J, McCabe FL, Millar H, Cunningham M, Snyder LA, Bugelski P, and Yan L
- Subjects
- Animals, Basigin pharmacology, Breast Neoplasms metabolism, Cell Line, Tumor, Fibroblasts metabolism, Humans, Matrix Metalloproteinases, Mice, Mitogen-Activated Protein Kinases metabolism, Neoplasm Transplantation, Neoplasms metabolism, Neovascularization, Pathologic, Phosphorylation, Proto-Oncogene Proteins c-jun metabolism, Recombinant Proteins pharmacology, Transplantation, Heterologous, p38 Mitogen-Activated Protein Kinases metabolism, Basigin metabolism, Oncogene Protein v-akt metabolism, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction, Vascular Endothelial Growth Factor A metabolism
- Abstract
Extracellular matrix metalloproteinase (MMP) inducer (EMMPRIN) is a cell surface glycoprotein overexpressed in many solid tumors. In addition to its ability to stimulate stromal MMP expression, tumor-associated EMMPRIN also induces vascular endothelial growth factor (VEGF) expression. To explore the underlying signaling pathways used by EMMPRIN, we studied the involvement of phosphoinositide 3-kinase (PI3K)-Akt, mitogen-activated protein kinase (MAPK), JUN, and p38 kinases in EMMPRIN-mediated VEGF regulation. Overexpression of EMMPRIN in MDA-MB-231 breast cancer cells stimulated the phosphorylation of only Akt and MAPKs but not that of JUN and p38 kinases. Conversely, inhibition of EMMPRIN expression resulted in suppressed Akt and MAPK phosphorylation. Furthermore, the PI3K-specific inhibitor LY294002 inhibited VEGF production by EMMPRIN-overexpressing cells in a dose- and time-dependent manner. On the other hand, the MAPK inhibitor U0126 did not affect VEGF production. In vivo, EMMPRIN-overexpressing tumors with elevated VEGF expression had a high level of phosphorylation of Akt and MAPK. Finally, when fibroblast cells were treated with recombinant EMMPRIN, Akt kinase but not MAPK was phosphorylated concomitant with an increase in VEGF production. Both the activation of Akt kinase and the induction of VEGF were specifically inhibited with a neutralizing antibody to EMMPRIN. Our results show that in both tumor and fibroblast cells EMMPRIN regulates VEGF production via the PI3K-Akt pathway but not via the MAPK, JUN, or p38 kinase pathways.
- Published
- 2006
- Full Text
- View/download PDF
10. Therapeutic potential of cytokine and chemokine antagonists in cancer therapy.
- Author
-
Yan L, Anderson GM, DeWitte M, and Nakada MT
- Subjects
- Disease Progression, Humans, Inflammation physiopathology, Risk Factors, Chemokines antagonists & inhibitors, Cytokines antagonists & inhibitors, Neoplasms therapy
- Abstract
A new paradigm is becoming widely accepted, that chronic inflammation, driven in part by chemokines and cytokines at the site of a tumour, may facilitate tumour progression instead of promoting anti-tumour immunity. Tumours and activated stromal cells secrete pro-inflammatory chemokines and cytokines that act either directly or indirectly through stimulation of the vascular endothelium to recruit leukocytes to the tumour. After activation, these tumour-associated leukocytes release angiogenic factors, mitogens, proteolytic enzymes, and chemotactic factors, recruiting more inflammatory cells and stimulating angiogenesis to sustain tumour growth and facilitate tumour metastasis. Breaking this cycle by inhibiting targets such as cytokines, chemokines and other inflammatory mediators, either alone, or more realistically, in combination with other therapies, such as anti-angiogenic or cytotoxic agents, may provide highly efficacious therapeutic regimens for the treatment of malignancies. This article reviews anti-cytokine and anti-chemokine therapies being pursued in cancer, and discusses in more detail anti-tumour necrosis factor-alpha (TNF) approaches.
- Published
- 2006
- Full Text
- View/download PDF
11. c7E3 Fab inhibits human tumor angiogenesis in a SCID mouse human skin xenograft model.
- Author
-
Nakada MT, Cao G, Sassoli PM, and DeLisser HM
- Subjects
- Abciximab, Animals, Cell Line, Tumor, Humans, Mice, Mice, SCID, Neovascularization, Pathologic prevention & control, Transplantation, Heterologous, Angiogenesis Inhibitors pharmacology, Antibodies, Monoclonal pharmacology, Immunoglobulin Fab Fragments pharmacology, Neovascularization, Pathologic drug therapy, Skin Transplantation
- Abstract
The alphavbeta3 integrin plays an important role in tumor growth and angiogenesis. Inhibition of this receptor by intact bivalent antibodies has been shown to inhibit angiogenesis and tumor growth. In this study we tested the chimeric Fab of 7E3 (c7E3 Fab), an antibody reactive with human platelet GPIIb/IIIa and alphavbeta3 to determine if it would inhibit in vivo angiogenesis and tumor growth in a SCID mouse/human skin tumor growth and angiogenesis model. c7E3 Fab inhibited human tumor angiogenesis and tumor growth. These data suggest monovalent antibody fragments devoid of antibody effector function can have efficacy in preclinical models of angiogenesis.
- Published
- 2006
- Full Text
- View/download PDF
12. Therapeutic effect of anti-TNF-alpha antibodies in an experimental model of anti-neutrophil cytoplasm antibody-associated systemic vasculitis.
- Author
-
Little MA, Bhangal G, Smyth CL, Nakada MT, Cook HT, Nourshargh S, and Pusey CD
- Subjects
- Animals, Antibodies, Antineutrophil Cytoplasmic biosynthesis, Autoimmune Diseases blood, Cell Movement, Disease Models, Animal, Leukocytes physiology, Nephritis therapy, Rats, Rats, Inbred WKY, Vasculitis blood, Antibodies, Antineutrophil Cytoplasmic immunology, Antibodies, Monoclonal therapeutic use, Autoimmune Diseases therapy, Tumor Necrosis Factor-alpha antagonists & inhibitors, Vasculitis therapy
- Abstract
The therapeutic options for anti-neutrophil cytoplasm antibody (ANCA)-associated systemic vasculitis (AASV) remain limited and hampered by adverse effects. One potential novel therapeutic avenue involves inhibition of TNF-alpha, with encouraging uncontrolled data in humans with one agent (infliximab) but disappointing controlled data from another (etanercept). For investigating the potential role of TNF-alpha as a therapeutic target in AASV, the effect of an anti-rat TNF-alpha mAb (CNTO 1081) in a rat model of AASV was investigated. For testing the effect of TNF-alpha blockade in this model, starting on day 28 after immunization (a point when glomerulonephritis is established), animals were randomized to treatment with CNTO 1081 or control mouse IgG. Treatment with CNTO 1081 significantly reduced albuminuria (mean 1.1 +/- 0.3 mg/24 h CNTO 1081 versus 8.0 +/- 1.9 controls; P < 0.05) and crescent formation (0% CNTO 1081 versus 60% controls; P < 0.05). Lung hemorrhage was also reduced (CNTO 1081: median score 0, range 0 to 2; controls: 2, range 1 to 3; P < 0.05). When analyzed by intravital microscopy, there was a 43% inhibition of leukocyte transmigration in mesenteric venules in response to topical CXCL1 (a neutrophil chemoattractant) in the CNTO 1081 group compared with controls (P < 0.001). Anti-myeloperoxidase antibody titers were similar in both groups throughout the study. In conclusion, these findings indicate that TNF-alpha plays an important role in the pathogenesis of experimental autoimmune vasculitis and suggest that blockade of this cytokine with an mAb is effective in treating established vasculitis. The therapeutic action of anti-TNF-alpha reagents may be mediated, in part, by suppression of the enhanced leukocyte-endothelial interactions in this disorder.
- Published
- 2006
- Full Text
- View/download PDF
13. Intraarterial reteplase and intravenous abciximab for treatment of acute ischemic stroke. A preliminary feasibility and safety study in a non-human primate model.
- Author
-
Qureshi AI, Suri MF, Ali Z, Ringer AJ, Boulos AS, Nakada MT, Alberico RA, Martin LB, Guterman LR, and Hopkins LN
- Subjects
- Abciximab, Acute Disease, Animals, Antibodies, Monoclonal administration & dosage, Disease Models, Animal, Feasibility Studies, Fibrinolytic Agents administration & dosage, Immunoglobulin Fab Fragments administration & dosage, Injections, Intralesional, Macaca fascicularis, Macaca mulatta, Platelet Aggregation Inhibitors administration & dosage, Random Allocation, Recombinant Proteins administration & dosage, Recombinant Proteins pharmacology, Tissue Plasminogen Activator administration & dosage, Antibodies, Monoclonal pharmacology, Fibrinolytic Agents pharmacology, Immunoglobulin Fab Fragments pharmacology, Intracranial Thrombosis drug therapy, Magnetic Resonance Imaging, Platelet Aggregation Inhibitors pharmacology, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Stroke drug therapy, Tissue Plasminogen Activator pharmacology
- Abstract
We performed a preliminary feasibility and safety study using intravenous (IV) administration of a platelet glycoprotein IIb/IIIa inhibitor (abciximab) in conjunction with intraarterial (IA) administration of a thrombolytic agent (reteplase) in a primate model of intracranial thrombosis. We introduced thrombus through superselective catheterization of the intracranial segment of the internal carotid artery in 16 primates. The animals were randomly assigned to receive IA reteplase and IV abciximab ( n =4), IA reteplase and IV placebo ( n =4), IA placebo and IV abciximab ( n =4) or IA and IV placebo ( n =4). Recanalization was assessed by serial angiography during the 6-h period after initiation of treatment. Postmortem magnetic resonance (MR) imaging was performed to determine the presence of cerebral infarction or intracranial hemorrhage. Partial or complete recanalization at 6 h after initiation of treatment (decrease of two or more points in pre-treatment angiographic occlusion grade) was observed in two animals treated with IA reteplase and IV abciximab, three animals treated with IA reteplase alone and one animal treated with IV abciximab alone. No improvement in perfusion was observed in animals that received IV and IA placebo. Cerebral infarction was demonstrated on postmortem MR imaging in three animals that received IA and IV placebo and in one animal each from the groups that received IA reteplase and IV abciximab or IV abciximab alone. One animal that received IV abciximab alone had a small intracerebral hemorrhage on MR imaging. IA reteplase with or without abciximab appeared to be the most effective regimen for achieving recanalization in our model of intracranial thrombosis. Further studies are required in experimental models to determine the optimal dose, method of administration and efficacy of these medications in acute ischemic stroke.
- Published
- 2005
- Full Text
- View/download PDF
14. Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases.
- Author
-
Tang Y, Nakada MT, Kesavan P, McCabe F, Millar H, Rafferty P, Bugelski P, and Yan L
- Subjects
- Animals, Antigens, CD biosynthesis, Antigens, CD genetics, Basigin, Breast Neoplasms enzymology, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Growth Processes physiology, Cell Line, Tumor, Cell Movement physiology, Coculture Techniques, Endothelial Cells cytology, Female, Fibroblasts cytology, Humans, Mice, Mice, Nude, Neovascularization, Pathologic enzymology, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Transplantation, Heterologous, Vascular Endothelial Growth Factor A antagonists & inhibitors, Antigens, CD physiology, Breast Neoplasms blood supply, Matrix Metalloproteinase 2 biosynthesis, Matrix Metalloproteinase 9 biosynthesis, Vascular Endothelial Growth Factor A biosynthesis
- Abstract
Matrix metalloproteinases (MMPs) are endopeptidases that play pivotal roles in promoting tumor disease progression, including tumor angiogenesis. In many solid tumors, MMP expression could be attributed to tumor stromal cells and is partially regulated by tumor-stroma interactions via tumor cell-associated extracellular matrix metalloproteinase inducer (EMMPRIN). The role of EMMPRIN during tumor angiogenesis and growth was explored by modulating EMMPRIN expression and activity using recombinant DNA engineering and neutralizing antibodies. In human breast cancer cells, changes in EMMPRIN expression influenced vascular endothelial growth factor (VEGF) production at both RNA and protein levels. In coculture of tumor cells and fibroblasts mimicking tumor-stroma interactions, VEGF expression was induced in an EMMPRIN- and MMP-dependent fashion, and was further enhanced by overexpressing EMMPRIN. Conversely, VEGF expression was inhibited by suppressing EMMPRIN expression in tumor cells, by neutralizing EMMPRIN activity, or by inhibiting MMPs. In vivo, EMMPRIN overexpression stimulated tumor angiogenesis and growth; both were significantly inhibited by antisense suppression of EMMPRIN. Expression of both human and mouse VEGF and MMP, derived from tumor and host cells, respectively, was regulated by EMMPRIN. These results suggest a novel tumor angiogenesis mechanism in which tumor-associated EMMPRIN functionally mediates tumor-stroma interactions and directly contributes to tumor angiogenesis and growth by stimulating VEGF and MMP expression.
- Published
- 2005
- Full Text
- View/download PDF
15. Tumor necrosis factor-alpha in the pathogenesis and treatment of cancer.
- Author
-
Anderson GM, Nakada MT, and DeWitte M
- Subjects
- Animals, Humans, Neoplasms therapy, Signal Transduction, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha therapeutic use, Neoplasms etiology, Tumor Necrosis Factor-alpha physiology
- Abstract
The critical pathogenic role of tumor necrosis factor (TNF)alpha in inflammatory disorders such as rheumatoid arthritis and inflammatory bowel disease is well established. The role played by TNFalpha in both the treatment and pathogenesis of cancer remains less understood. Recent advances help to create a framework for understanding seemingly paradoxical effects of TNFalpha as both an anti-tumour agent and a mediator of tumour growth. High pharmacological doses of TNFalpha combined with chemotherapy can regress otherwise intractable tumours, and efforts continue to optimize delivery to avoid severe toxicities. Mounting evidence demonstrates that pathophysiological concentrations of endogenous TNFalpha act to promote tumour genesis and growth. The cellular and molecular pathways mediating these phenomena are starting to be clarified. Current data support the continued development of both TNFalpha and anti-TNFalpha therapy for clinical treatment of cancers in distinct settings.
- Published
- 2004
- Full Text
- View/download PDF
16. CNTO 95, a fully human monoclonal antibody that inhibits alphav integrins, has antitumor and antiangiogenic activity in vivo.
- Author
-
Trikha M, Zhou Z, Nemeth JA, Chen Q, Sharp C, Emmell E, Giles-Komar J, and Nakada MT
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Antibody Affinity, Antibody Specificity, Aorta metabolism, Aorta pathology, Blotting, Western, Cattle, Cell Adhesion, Cell Division, Cell Line, Cell Line, Tumor, Cell Movement, Cells, Cultured, Collagen pharmacology, Dose-Response Relationship, Drug, Drug Combinations, Endothelium, Vascular cytology, Fibroblast Growth Factor 2 metabolism, Haplorhini, Humans, Integrin alphaVbeta3 metabolism, Integrins metabolism, Kinetics, Laminin pharmacology, Macaca fascicularis, Melanoma immunology, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Transplantation, Neovascularization, Pathologic, Placenta metabolism, Placenta pathology, Protein Binding, Proteoglycans pharmacology, Rats, Rats, Nude, Receptors, Vitronectin metabolism, Time Factors, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents pharmacology, Integrin alphaV chemistry, Melanoma therapy
- Abstract
Integrins of the alphav family, such as alphavbeta3 and alphavbeta5, are implicated in tumor-induced angiogenesis; but their role in tumor growth has not been fully explored. CNTO 95 is a fully human antibody that recognizes the alphav family of integrins and is likely to be less immunogenic in humans compared to chimeric or humanized antibodies. CNTO 95 bound to purified alphavbeta3 and alphavbeta5 with a Kd of approximately 200 pM and to alphav integrin-expressing human cells with a Kd of 1-24 nM. In vitro, CNTO 95 inhibited human melanoma cell adhesion, migration and invasion at doses ranging 7-20 nM. In a rat aortic ring sprouting assay, CNTO 95 (approx. 70 nM) completely inhibited sprouting. Using a human melanoma xenograft model in nude mice wherein CNTO 95 recognized alphavbeta3 and alphavbeta5 on human tumor cells but not mouse angiogenic integrins, CNTO 95 (10 mg/kg, 3 times/week) inhibited growth of human melanoma tumors in nude mice by approximately 80% (p = 0.0005), suggesting that CNTO 95 inhibited human tumor growth independently of its antiangiogenic activity. In a nude rat human xenograft model where CNTO 95 binds and blocks both tumor and host integrins, this antibody (10 mg/kg once/week) reduced final tumor weight by >99% (p < 0.0001). Based on these preclinical data, a dose-escalating phase I clinical trial in cancer patients has been initiated. To our knowledge, CNTO 95 is the first fully human MAb to alphav integrins that has potent antitumor and antiangiogenic properties in in vivo preclinical models., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
17. Tumor-stroma interaction: positive feedback regulation of extracellular matrix metalloproteinase inducer (EMMPRIN) expression and matrix metalloproteinase-dependent generation of soluble EMMPRIN.
- Author
-
Tang Y, Kesavan P, Nakada MT, and Yan L
- Subjects
- Antigens, CD biosynthesis, Antigens, CD genetics, Antigens, CD pharmacology, Antigens, Neoplasm biosynthesis, Antigens, Neoplasm genetics, Antigens, Neoplasm pharmacology, Basigin, Breast Neoplasms enzymology, Breast Neoplasms genetics, Cell Line, Tumor, Cells, Cultured, Coculture Techniques, Feedback, Physiological, Fibroblasts, Humans, Matrix Metalloproteinase 1 biosynthesis, Matrix Metalloproteinase 1 genetics, Matrix Metalloproteinase 1 metabolism, Matrix Metalloproteinases biosynthesis, Matrix Metalloproteinases genetics, Recombinant Proteins pharmacology, Solubility, Transfection, Antigens, CD metabolism, Antigens, Neoplasm metabolism, Breast Neoplasms metabolism, Gene Expression Regulation, Neoplastic drug effects, Matrix Metalloproteinases metabolism, Stromal Cells cytology, Stromal Cells metabolism
- Abstract
Matrix metalloproteinases (MMPs) are metal-dependent endopeptidases that play pivotal roles in tumor disease progression. In many solid tumors, MMPs are indeed produced by tumor stromal cells, rather than by tumor cells. This expression pattern is, at least in part, regulated by tumor-stroma interaction via tumor cell-associated extracellular matrix metalloproteinase inducer (EMMPRIN). In vitro, recombinant EMMPRIN dose-dependently stimulated MMP-1 production by primary human fibroblast cells. Interestingly, in addition to stimulating MMP expression, EMMPRIN also induced its own gene expression. To further explore this potential positive feedback regulatory mechanism, we generated human breast cancer cells expressing different levels of EMMPRIN. Coculture of EMMPRIN-positive tumor cells with fibroblast cells resulted in a concomitant stimulation of MMP-2, MMP-9, and EMMPRIN production. This induction was EMMPRIN dependent, was further enhanced by overexpression, and was reduced by antisense suppression of EMMPRIN expression in tumor cells. Increased expression of membrane-associated EMMPRIN was accompanied by an MMP-dependent generation of a soluble form of EMMPRIN representing a proteolytic cleavage product lacking the carboxyl terminus. On the basis of these findings, we propose a model in which tumor cell-associated EMMPRIN stimulates MMPs, as well as EMMPRIN expression in tumor stroma. Increased MMP activity in tumor local environment results in proteolytic cleavage of membrane-associated EMMPRIN, releasing soluble EMMPRIN. Soluble EMMPRIN in turn acts in a paracrine fashion on stroma cells that are both adjacent and distant to tumor sites to further stimulate the production of MMPs and additional EMMPRIN, which consequently contributes to tumor angiogenesis, tumor growth, and metastasis.
- Published
- 2004
18. Clot lysis in a primate model of peripheral arterial occlusive disease with use of systemic or intraarterial reteplase: addition of abciximab results in improved vessel reperfusion.
- Author
-
Nakada MT, Montgomery MO, Nedelman MA, Guerrero JL, Cohen SA, Barnathan ES, and Jordan RE
- Subjects
- Abciximab, Animals, Antibodies, Monoclonal administration & dosage, Drug Therapy, Combination, Femoral Artery, Fibrinolytic Agents administration & dosage, Immunoglobulin Fab Fragments administration & dosage, Infusions, Intra-Arterial, Injections, Intravenous, Macaca fascicularis, Platelet Aggregation drug effects, Recombinant Proteins administration & dosage, Tissue Plasminogen Activator administration & dosage, Vascular Patency, Antibodies, Monoclonal therapeutic use, Arterial Occlusive Diseases drug therapy, Fibrinolytic Agents therapeutic use, Immunoglobulin Fab Fragments therapeutic use, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Recombinant Proteins therapeutic use, Thrombosis drug therapy, Tissue Plasminogen Activator therapeutic use
- Abstract
Purpose: This study was designed to compare the ability of reteplase (a fibrinolytic agent) alone or in combination with abciximab (a monoclonal antibody antagonist of platelet glycoprotein IIb/IIIa) to achieve and sustain vessel patency in an acute model of peripheral arterial occlusive disease in cynomolgus monkeys., Materials and Methods: Total arterial occlusion was induced in the femoral arteries of 32 cynomolgus monkeys (eight groups of four) by endothelial injury and injection of thrombin-treated autologous blood. Reteplase was administered by intravenous bolus dose or by intraarterial infusion at the site of the clot. Abciximab was administered as a single weight-adjusted intravenous bolus dose. Platelet activity was measured by ex vivo platelet aggregation before and after abciximab treatment. Different groups of animals received sequential partial doses of reteplase with or without increasing doses of abciximab until either the weight-adjusted human dose equivalent of reteplase was reached or vessel recanalization was achieved., Results: Animals receiving reteplase-only regimens demonstrated variability in the times required for reperfusion and the permanence of the effect. The coadministration of abciximab at doses of the antibody that achieved near or full inhibition of platelet function generally decreased the time to reperfusion and resulted in more consistent and sustained vessel patency. In the case of systemic intravenous reteplase, the coadministration of abciximab resulted in effective reperfusion of thrombosed vessels at decreased doses of the lytic agent., Conclusions: Reteplase administered systemically or at the site of thrombotic occlusion restored blood flow for periods of varying duration in monkeys with acute femoral artery thrombosis. The coadministration of systemic intravenous abciximab to intravenous or intraarterial reteplase allowed the use of lower doses of fibrinolytic agent with more accelerated and sustained reperfusion.
- Published
- 2004
- Full Text
- View/download PDF
19. PECAM-directed immunotargeting of catalase: specific, rapid and transient protection against hydrogen peroxide.
- Author
-
Sweitzer TD, Thomas AP, Wiewrodt R, Nakada MT, Branco F, and Muzykantov VR
- Subjects
- Adenoviridae genetics, Antioxidants metabolism, Biotinylation, Cell Line, Tumor, Drug Delivery Systems, Endothelium, Vascular metabolism, Free Radicals, Genetic Therapy methods, Humans, Hydrogen Peroxide metabolism, Hydrogen Peroxide pharmacology, Immunoblotting, Kinetics, Liposomes metabolism, Microscopy, Confocal, Microscopy, Fluorescence, Oxidative Stress, Time Factors, Transfection, Catalase metabolism, Hydrogen Peroxide chemistry, Platelet Endothelial Cell Adhesion Molecule-1 metabolism
- Abstract
Vascular immunotargeting to Platelet-Endothelial Cell Adhesion Molecule-1 (PECAM) facilitates drug delivery to endothelium. We used human PECAM-transfected REN cells (REN/PECAM) as a model to compare targeting of antioxidant enzyme catalase conjugated with PECAM antibody (anti-PECAM/catalase) with adenoviral catalase delivery. Anti-PECAM/(125)I-catalase bound to REN/PECAM, but not to REN cells (70 vs. 1 ng/well vs. < 2 ng/well of unmodified catalase). At a virus-to-cell ratio of 1, elevated levels of catalase protein were detected by immunoblotting after adenoviral transfection of REN/PECAM and REN cells alike; H(2)O(2)-degrading activity of cell lysates was elevated at ratios of 10 and higher. REN/PECAM cells internalize 66% of cell-bound anti-PECAM/(125)I-catalase. Confocal microscopy localized anti-PECAM/catalase to intracellular vesicles, while catalase expressed by adenovirus was distributed in vesicles and throughout the cytosol. Within 15 min of delivery, anti-PECAM/catalase augmented H(2)O(2)-degrading activity and survival of H(2)O(2)-exposed REN/PECAM cells. The effects of conjugate delivery reached a plateau within 1 h and declined to the basal level within 12 h. In contrast, adenoviral delivery required several hours for transduction and development of the effects, but permitted much longer duration of protection (at least 48 h). Simultaneous exposure of REN/PECAM cells to anti-PECAM/catalase and catalase-encoding adenovirus afforded protection against H(2)O(2) with a rapid onset and a prolonged duration. Therefore, PECAM-directed immunotargeting provides a specific, antigen-directed intracellular delivery of catalase that affords a rapid but transient protection against H(2)O(2) and may complement gene delivery strategies for antioxidant protection.
- Published
- 2003
- Full Text
- View/download PDF
20. Monoclonal antibodies as therapeutics in oncology.
- Author
-
Trikha M, Yan L, and Nakada MT
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antigens, Neoplasm immunology, Clinical Trials, Phase III as Topic, Combined Modality Therapy methods, Drug Approval, Humans, Mice, Neoplasms immunology, Quality Control, Radioimmunotherapy methods, Antibodies, Monoclonal classification, Antibodies, Monoclonal therapeutic use, Immunotherapy methods, Neoplasms therapy
- Abstract
The specificity of antibodies has been harnessed to target cancer cells and the first therapeutic antibodies for use in oncology are now finding application in the clinic. Studies are currently under way to develop new and improved antibodies. Recent developments have been made in the identification of novel targets, including the use of genomic and proteomic technologies. Several methods are also being developed to enhance antibody efficacy.
- Published
- 2002
- Full Text
- View/download PDF
21. Abciximab pharmacodynamics are unaffected by antecedent therapy with other GPIIb/IIIa antagonists in non-human primates.
- Author
-
Nakada MT, Sassoli PM, Tam SH, Nedelman MA, Jordan RE, and Kereiakes DJ
- Subjects
- Abciximab, Animals, Binding Sites, Antibody drug effects, Dose-Response Relationship, Drug, Drug Therapy, Combination, Eptifibatide, Female, Macaca fascicularis, Male, Peptides pharmacology, Platelet Aggregation drug effects, Platelet Aggregation immunology, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Protein Binding drug effects, Protein Binding immunology, Tirofiban, Tyrosine analogs & derivatives, Tyrosine pharmacology, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal pharmacology, Immunoglobulin Fab Fragments metabolism, Immunoglobulin Fab Fragments pharmacology, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors
- Abstract
Background: Tirofiban and eptifibatide are currently approved for the medical stabilization of non-ST segment elevation acute coronary syndromes. In patients undergoing percutaneous coronary intervention (PCI) during infusion of these drugs, conversion to abciximab, which has long term proven clinical efficacy and cost-effectiveness, following PCI may be desirable. The purpose of this study was to determine if the binding or pharmacodynamics of abciximab is affected by a prior infusion of either tirofiban or eptifibatide., Methods: In vitro binding experiments were performed to determine if prior exposure to tirofiban or eptifibatide altered the affinity and extent of binding of abciximab to GPIIb/IIIa. For in vivo experiments, cynomolgus monkeys were pretreated with a bolus and 18 hour infusion of saline, tirofiban, or eptifibatide. At the end of the initial treatment, a bolus and 12 hr infusion of abciximab was started without delay. Inhibition of platelet aggregation, GPIIb/IIIa receptor blockade and abciximab pharmacokinetics were measured during and after both infusions., Results: Equilibrium binding of abciximab in vitro was unaffected by tirofiban or eptifibatide. The extent and duration of abciximab inhibition of ex vivo platelet aggregation, receptor blockade, and abciximab pharmacokinetics in monkeys during and after the abciximab infusion were not affected by prior infusion of the animals with tirofiban or eptifibatide., Conclusions: In vitro and in vivo studies revealed that the molecular interaction of abciximab with the platelet GPIIb/IIIa receptor is not altered by immediate prior exposure of platelets to small molecule GPIIb/IIIa antagonists. These preclinical studies suggest that the efficacy of abciximab should not be impaired if it is initiated following termination of therapy with small molecule GPIIb/IIIa antagonists.
- Published
- 2002
- Full Text
- View/download PDF
22. Effects of abciximab on the acute pathology of blood vessels after arterial stenting in nonhuman primates.
- Author
-
Palmerini T, Nedelman MA, Scudder LE, Nakada MT, Jordan RE, Smyth S, Gordon RE, Fallon JT, and Coller BS
- Subjects
- Abciximab, Animals, Aorta, Abdominal pathology, Arteries drug effects, Arteries injuries, Iliac Artery pathology, Immunohistochemistry, Macaca fascicularis, Microscopy, Electron, Thrombosis etiology, Time Factors, Antibodies, Monoclonal pharmacology, Anticoagulants pharmacology, Arteries pathology, Blood Platelets drug effects, Immunoglobulin Fab Fragments pharmacology, Leukocytes drug effects, Platelet Aggregation Inhibitors pharmacology, Stents adverse effects, Thrombosis prevention & control
- Abstract
Objectives: This study was designed to assess the effect of abciximab on platelet and leukocyte deposition 60 min after stent insertion in nonhuman primates., Background: Although it is well established that abciximab improves both short- and long-term clinical outcomes after stent placement, there have been no studies assessing its effect on early platelet and leukocyte deposition., Methods: Cynomolgus monkeys were pretreated with aspirin and either saline or a 0.4 mg/kg bolus of abciximab, and then subjected to angioplasty and Palmaz-Schatz stent placement in the common iliac artery or abdominal aorta. After 60 min, animals were euthanized and the stented artery was evaluated by immunohistochemistry and morphometry., Results: Complete occlusion of the stented vessel with a thin fibrin(ogen) meshwork and trapped blood occurred in two saline-treated and two abciximab-treated animals. In the four remaining saline-treated animals, a layer of erythrocytes trapped in a network of fibrin(ogen) was noted close to the vessel wall, and this was covered by a layer of large, irregular platelet thrombi. Leukocytes formed a monolayer on top of the platelets and near stent struts. In the four remaining abciximab-treated animals, the mean erythrocyte area was 65% smaller (p = 0.070), the platelet aggregate area was 89% smaller (p = 0.049) and the luminal area was 59% larger (p = 0.004). A monolayer of leukocytes also formed on top of the platelets and near stent struts., Conclusions: In control stented blood vessels in this study, platelet thrombi formed not at the vessel wall, but on top of an erythrocyte-rich layer, and platelets recruited leukocytes. Abciximab decreased the size of platelet thrombi, but did not prevent leukocyte recruitment.
- Published
- 2002
- Full Text
- View/download PDF
23. Multiple roles for platelet GPIIb/IIIa and alphavbeta3 integrins in tumor growth, angiogenesis, and metastasis.
- Author
-
Trikha M, Zhou Z, Timar J, Raso E, Kennel M, Emmell E, and Nakada MT
- Subjects
- Abciximab, Animals, Antibodies, Monoclonal pharmacology, Apoptosis drug effects, Apoptosis physiology, Blood Platelets physiology, Cattle, Cell Adhesion drug effects, Cell Adhesion physiology, Cell Division drug effects, Cell Division physiology, Cell Movement physiology, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Extracellular Matrix Proteins metabolism, Female, Fibrin metabolism, Fibrinolytic Agents pharmacology, Fibroblast Growth Factor 2 antagonists & inhibitors, Fibroblast Growth Factor 2 physiology, HT29 Cells, Humans, Immunoglobulin Fab Fragments pharmacology, Melanoma drug therapy, Melanoma secondary, Mice, Mice, Nude, Neovascularization, Pathologic blood, Neovascularization, Pathologic drug therapy, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Rats, Receptors, Vitronectin antagonists & inhibitors, Xenograft Model Antitumor Assays, Melanoma blood supply, Melanoma pathology, Neovascularization, Pathologic pathology, Platelet Glycoprotein GPIIb-IIIa Complex physiology, Receptors, Vitronectin physiology
- Abstract
In vivo tumor cells interact with a variety of host cells such as endothelial cells and platelets, and these interactions are mediated by integrins GPIIb/IIIa and alphavbeta3. We used chimeric (c) 7E3 Fab (ReoPro) and murine (m) 7E3 F(ab')(2) to elucidate the role of these integrins in angiogenesis, tumor growth, and metastasis. These antibodies are potent inhibitors of GPIIb/IIIa and alphavbeta3. c7E3 Fab inhibited alphavbeta3-mediated human umbilical vein endothelial (HUVEC) and melanoma cell adhesion, migration, invasion, and basic fibroblast growth factor stimulated proliferation of HUVECs (IC(50) values range from 0.15 to 5 microg/ml for different assays). In an in vitro angiogenesis assay, c7E3 Fab inhibited basic fibroblast growth factor and platelet-stimulated capillary formation of HUVECs (IC(50) = 10 microg/ml and 15 microg/ml, respectively), demonstrating that endothelial alphavbeta3 is important for sprouting, and platelet-stimulated sprouting is mediated by GPIIb/IIIa. In an experimental metastasis assay, a single pretreatment of human melanoma cells with c7E3 Fab (2.5 microg/ml) inhibited lung colonization of the tumor cells in severe combined immunodeficient mice. In vivo, m7E3 F(ab')(2) partially inhibited growth of human melanoma tumors in nude mice compared with control-treated animals. These data suggest that tumor cell-expressed integrins are important but not the only component involved in tumor growth. Because c7E3 Fab and m7E3 F(ab')(2) do not cross-react with murine integrins, this inhibition of metastasis and tumor growth is attributable to direct blockade of human tumor alphavbeta3 integrins. m7E3 F(ab')(2) completely blocked tumor formation and growth of human melanoma tumors growing in nude rats. In this xenograft model, m7E3 F(ab')(2) simultaneously binds to both human tumor and host platelet GPIIb/IIIa and endothelial alphavbeta3 integrins, thus participating as an antiangiogenic and an antitumor agent. Collectively, these results indicate that combined blockade of GPIIb/IIIa and alphavbeta3 affords significant antiangiogenic and antitumor benefit.
- Published
- 2002
24. Effect of anti-tumor necrosis factor-alpha polyclonal antibody on restenosis after balloon angioplasty in a rabbit atherosclerotic model.
- Author
-
Zhou Z, Lauer MA, Wang K, Forudi F, Zhou X, Song Xy, Solowski N, Kapadia SR, Nakada MT, Topol EJ, and Lincoff AM
- Subjects
- Animals, Goats, Hyperplasia pathology, Inflammation etiology, Inflammation pathology, Macrophages immunology, Mice, Rabbits, Tumor Necrosis Factor-alpha analysis, Tunica Intima pathology, Angioplasty, Balloon adverse effects, Antibodies pharmacology, Arteriosclerosis pathology, Graft Occlusion, Vascular pathology, Tumor Necrosis Factor-alpha immunology
- Abstract
Inflammation has been postulated to contribute to restenosis after balloon angioplasty. Tumor necrosis factor (TNF)-alpha is a pleiotropic proinflammatory cytokine involved in many features of inflammation. We examined the tissue expression pattern of TNF-alpha and the inflammatory response to arterial injury, and the effects of a goat anti-rabbit-TNF-alpha polyclonal antibody on tissue TNF-alpha expression, inflammation and restenosis in a rabbit atherosclerotic model. At different time points following air dessication and subsequent balloon injury, fresh rabbit femoral artery tissues were homogenized and analyzed for TNF-alpha levels by quantitative TNF-alpha bioassay. Rabbits were treated with a goat anti-rabbit-TNF-alpha polyclonal antibody, Serum and tissue TNF-alpha neutralization, macrophage infiltration (as an indicator of inflammation), and neointimal areas were determined. Balloon angioplasty increased tissue TNF-alpha expression 100000-fold over baseline, and this increase persisted over 6 days after arterial injury, serum anti-TNF-alpha antibody levels were sufficient to neutralize tissue TNF-alpha activity by 60-75%, macrophage infiltration was suppressed, but did not decrease the neointimal formation. These data indicate that tissue TNF-alpha levels were markedly increased after balloon angioplasty. Anti-TNF-alpha treatment was sufficient to neutralize tissue TNF-alpha activity, reduce inflammation, but did not inhibit neointimal formation following balloon angioplasty in a rabbit atherosclerotic model.
- Published
- 2002
- Full Text
- View/download PDF
25. Platelets inhibit the lysis of pulmonary microemboli.
- Author
-
Murciano JC, Harshaw D, Neschis DG, Koniaris L, Bdeir K, Medinilla S, Fisher AB, Golden MA, Cines DB, Nakada MT, and Muzykantov VR
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Blood Physiological Phenomena, Fibrinolytic Agents pharmacology, Humans, In Vitro Techniques, Male, Mice, Perfusion, Platelet Glycoprotein GPIIb-IIIa Complex immunology, Rats, Rats, Sprague-Dawley, Tissue Plasminogen Activator pharmacology, Blood Platelets physiology, Fibrinolysis physiology, Pulmonary Embolism physiopathology
- Abstract
Using tracings of (125)I-labeled fibrin(ogen) in rodents, we examined the hypothesis that platelets impede the lysis of pulmonary emboli. (125)I-Microemboli (ME, 3-10 micron diameter) lodged homogeneously throughout the lungs after intravenous injection in both rats and mice (60% of injected dose), caused no lethality, and underwent spontaneous dissolution (50 and 100% within 1 and 5 h, respectively). Although lung homogenates displayed the most intense fibrinolytic activity of all the major organs, dissolution of ME was much slower in isolated perfused lungs (IPL) than was observed in vivo. Addition of rat plasma to the perfusate facilitated ME dissolution in IPL to a greater extent than did addition of tissue-type plasminogen activator alone, suggesting that permeation of the clot by plasminogen is the rate-limited step in lysis. Platelet-containing ME injected in rats lysed much more slowly than did ME formed from fibrin alone. (125)I-Thrombi, formed in the pulmonary vasculature of mice in response to intravascular activation of platelets by injection of collagen and epinephrine, were essentially resistant to spontaneous dissolution. Moreover, injection of the antiplatelet glycoprotein IIb/IIIa antibody 7E3 F(ab')(2) facilitated spontaneous dissolution of pulmonary ME and augmented fibrinolysis by a marginally effective dose of Retavase (10 microg/kg) in rats. These studies show that platelets suppress pulmonary fibrinolysis. The mechanism(s) by which platelets stabilize ME and utility of platelet inhibitors to facilitate their dissolution deserves further study.
- Published
- 2002
- Full Text
- View/download PDF
26. Platelets and cancer: implications for antiangiogenic therapy.
- Author
-
Trikha M and Nakada MT
- Subjects
- Angiogenesis Inhibitors pharmacology, Angiogenesis Inhibitors therapeutic use, Blood Platelets physiology, Humans, Neoplasm Metastasis drug therapy, Neoplasm Metastasis prevention & control, Neoplasms drug therapy, Neoplasms pathology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic etiology, Thrombophilia blood, Thrombophilia etiology, Blood Platelets pathology, Neoplasms blood
- Abstract
Thromboembolism is one of the most common causes of death in cancer patients. Among the most frequent thrombotic complications in patients with cancer are disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, and thrombocytosis. Clearly, these complications arise as tumor cells interact with almost all components of the hemostatic system including platelets. Platelets participate in tumor progression by contributing to the metastatic cascade, protecting tumor cells from immune surveillance, regulating tumor cell invasion, and angiogenesis. Platelets contain one of the largest stores of angiogenic and mitogenic factors and the tumor vasculature is leaky, which allows platelets to come in contact with the tumor and deposit multiple angiogenic factors including vascular endothelial growth factor (VEGF) and thrombin to tumor cells, which in turn contributes to tumor progression. This article reviews the recent literature on how platelets contribute to tumor growth, angiogenesis, and metastasis.
- Published
- 2002
- Full Text
- View/download PDF
27. Glycoprotein IIb/IIIa antagonist, murine 7E3 F(ab') 2, and tissue plasminogen activator in focal ischemia: evaluation of efficacy and risk of hemorrhage with combination therapy.
- Author
-
Shuaib A, Yang Y, Nakada MT, Li Q, and Yang T
- Subjects
- Animals, Bleeding Time, Brain pathology, Brain Ischemia diagnostic imaging, Brain Ischemia pathology, Brain Ischemia physiopathology, Cerebral Angiography, Cerebral Hemorrhage chemically induced, Cerebral Hemorrhage epidemiology, Drug Therapy, Combination, Immunoglobulin Fab Fragments administration & dosage, Incidence, Male, Mice, Nervous System physiopathology, Plasminogen Activators adverse effects, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Rats, Rats, Wistar, Risk Factors, Tail blood supply, Tissue Plasminogen Activator adverse effects, Brain Ischemia drug therapy, Immunoglobulin Fab Fragments therapeutic use, Plasminogen Activators therapeutic use, Tissue Plasminogen Activator therapeutic use
- Abstract
Tissue hypoperfusion during cerebral ischemia results from occlusion of large and small vessels. Combination treatment strategies using fibrinolytics to thrombolyse an embolic clot and antiplatelet agents to prevent reocclusion and the formation of new platelet thrombi in the microcirculation may offer advantages over single-agent therapy. The authors report on the effects of tissue plasminogen activator (rt-PA), a glycoprotein (GP) IIb/IIIa receptor antagonist, 7E3 F(ab') 2, or a combination of the two agents in a focal embolic model of cerebral ischemia in Wistar rats. Focal ischemia was produced by introducing an autologous thrombus into the right side middle cerebral artery. Forty-six male Wistar rats were randomly divided into 6 groups: control (n = 8), 7E3 F(ab') 2 (n = 9, 6 mg/kg), rt-PA (n = 9, 10 mg/kg), rt-PA (n = 6, 20 mg/kg), and 7E3 F(ab') 2 with either 10 mg/kg (n = 10) (low-dose combination) or 20 mg/kg (n = 6) (high-dose combination) rt-PA. Evaluation of neurobehavioral scores, cerebral angiography, bleeding time, and measurement of brain infarction volume were used to determine efficacy. All actively treated groups showed a significant reduction in the infarct volume. Animals treated with 7E3 F(ab') 2 showed reduced infarction volumes (24.0 +/- 5.1%) compared with controls (42.43 +/- 5.6%, P < 0.02). Treatment with rt-PA significantly reduced infarction volume (20.7 +/- 3.3, = 0.01) at 10 mg/kg and at 20 mg/kg (19.5 +/- 8.2%, P < 0.05). Compared with vehicle-treated animals, the low-dose combination (16.4 +/- 5.5, P < 0.003) and high-dose combination (23.7 +/- 6.2%, P < 0.05) showed significant reduction in infarction volume. Cerebral angiography revealed significantly better recanalization in the combination group (5/6 animals in the high dose and 4/6 in low dose) compared with animals treated with 7E3 F(ab') 2 (3/10) or rt-PA alone (2/6). Bleeding time significantly increased from 11.25 +/- 1.9 minutes in the control group to 17 +/- 3.1 minutes in the rt-PA group, 24.5 +/- 2.6 minutes in the 7E3 F(ab') 2 group, 25.7 +/- 3.1 minutes in the low-dose combination group, and 32.5 +/- 4.7 minutes in the high-dose combination group. The incidence of intercerebral hemorrhage was highest in the high-dose combination group (6 of 6 animals) and lowest in the single treatment with 7E3 F(ab') 2 alone (1 of 10 animals) ( P < 0.05). Our data show that murine 7E3 F(ab') 2 alone has therapeutic effects when used after cerebral ischemia. Although this study suggests that higher doses of thrombolytic combined with anti-GPIIb/IIIa therapy may increases the risk of intracranial hemorrhage, the data also support the notion that anti-GPIIb/IIIa agents can safely be combined with low doses of thrombolytic agent to produce significant attenuation of neuronal damage with no increase in the incidence of cerebral hemorrhage.
- Published
- 2002
- Full Text
- View/download PDF
28. The beta3 integrin antagonist m7E3 reduces matrix metalloproteinase activity and smooth muscle cell migration.
- Author
-
Bendeck MP and Nakada MT
- Subjects
- Abciximab, Animals, Antigens, CD, Blood Platelets drug effects, Blood Platelets physiology, Carotid Arteries cytology, Carotid Arteries drug effects, Carotid Arteries physiology, Carotid Artery Injuries pathology, Cell Division drug effects, Cell Movement drug effects, Integrin beta3, Male, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular pathology, Rats, Rats, Sprague-Dawley, Tunica Intima anatomy & histology, Tunica Intima drug effects, Antibodies, Monoclonal pharmacology, Carotid Artery Injuries physiopathology, Immunoglobulin Fab Fragments pharmacology, Matrix Metalloproteinase Inhibitors, Muscle, Smooth, Vascular physiopathology, Platelet Membrane Glycoproteins antagonists & inhibitors
- Abstract
Treatment with c7E3 (abciximab, ReoPro) has been associated with a reduction in coronary events and the need for revascularization. Some of these beneficial effects may be due to blockade of the alphavbeta3 integrin receptor on smooth muscle cells (SMCs), however very little is known about the mechanisms involved. The current studies were designed to test the hypothesis that beta3 integrin antagonists inhibit the arterial response to injury by reducing matrix metalloproteinase (MMP) activity in the vessel wall. Male Sprague-Dawley rats were treated with daily intraperitoneal injections of the monoclonal antibody m7E3 at a dose of 6 mg/kg/day. MMP-9 activity was reduced by 73%, and MMP-2 activity by 75%, in the injured carotids of the m7E3-treated rats compared to saline-treated controls. By contrast, tissue inhibitor metalloproteinase (TIMP) activity was not changed. SMC migration assayed at 4 days after injury was reduced from 56.7 +/- 14 cells/mm(2) intimal surface area in controls to 17.5 +/- 5 cells/mm(2) in m7E3-treated rats (p = 0.02). Medial cell replication measured at 4 days and intimal cell replication measured at 7 days were not affected. Intimal cross-sectional area, measured 14 days after injury was reduced by 28% after m7E3 treatment (p = 0.05). Intimal smooth muscle cell number and the ratio of intima/media cross-section area were also reduced. By contrast, intimal SMC density was not affected by m7E3 treatment, indicating no effect on matrix accumulation. We conclude that treatment with m7E3 reduced SMC migration following vascular injury, possibly via an inhibitory effect on MMP activity, and this resulted in a decrease in intimal size at 14 days after injury., (Copyright 2001 S. Karger AG, Basel)
- Published
- 2001
- Full Text
- View/download PDF
29. Abciximab suppresses the rise in levels of circulating inflammatory markers after percutaneous coronary revascularization.
- Author
-
Lincoff AM, Kereiakes DJ, Mascelli MA, Deckelbaum LI, Barnathan ES, Patel KK, Frederick B, Nakada MT, and Topol EJ
- Subjects
- Abciximab, Biomarkers blood, C-Reactive Protein metabolism, Female, Humans, Inflammation blood, Inflammation etiology, Infusions, Intravenous, Interleukin-6 blood, Male, Middle Aged, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Treatment Outcome, Tumor Necrosis Factor-alpha metabolism, United States, Angioplasty, Balloon, Coronary adverse effects, Antibodies, Monoclonal administration & dosage, Immunoglobulin Fab Fragments administration & dosage, Inflammation prevention & control, Platelet Aggregation Inhibitors administration & dosage
- Abstract
Background: Previous investigators have shown that systemic markers of inflammation may be increased in patients with acute ischemic syndromes or after percutaneous coronary revascularization and that persistent elevation in these markers is predictive of excess risk of subsequent adverse cardiac events. By virtue of its cross-reactivity with the glycoprotein IIb/IIIa, avbeta3, and alphaMbeta2 receptors, abciximab may reduce inflammatory processes. Methods and Results-- Assays for the inflammatory markers C-reactive protein, interleukin-6, and tumor necrosis factor-alpha were performed on serum samples obtained from 160 patients in a placebo-controlled, randomized trial of abciximab during angioplasty. Eighty patients each had received a placebo or abciximab bolus plus a 12-hour infusion. Serum samples were drawn at baseline (before revascularization), 24 to 48 hours after study drug administration, and 4 weeks after study drug administration. Between baseline and 24 to 48 hours, the increase in C-reactive protein was 32% less in patients receiving abciximab than placebo (P=0.025); the rise in interleukin-6 levels was 76% less in the abciximab group (P<0.001); and the rise in tumor necrosis factor-alpha levels was 100% less with abciximab therapy (P=0.112). By 4 weeks, most marker levels had returned to baseline, with no significant differences between placebo and abciximab groups., Conclusions: Systemic markers of inflammation increase in the first 24 to 48 hours after angioplasty, but the magnitude of that rise is diminished by periprocedural abciximab. Some of the long-term clinical benefit derived from this agent may be related to an anti-inflammatory effect.
- Published
- 2001
- Full Text
- View/download PDF
30. Regulation of PECAM-1 in endothelial cells during cell growth and migration.
- Author
-
RayChaudhury A, Elkins M, Kozien D, and Nakada MT
- Subjects
- Blotting, Northern, Cell Communication, Cells, Cultured, Humans, Platelet Endothelial Cell Adhesion Molecule-1 analysis, RNA, Messenger analysis, Umbilical Veins, Cell Division, Cell Movement, Endothelium, Vascular chemistry, Endothelium, Vascular cytology, Gene Expression Regulation, Platelet Endothelial Cell Adhesion Molecule-1 genetics
- Abstract
Endothelial cells (EC) that form the inner lining of blood vessels remain quiescent in the normal adult vasculature except during angiogenesis and reendothelialization, which result in EC proliferation and migration. EC placed in culture at subconfluent density also undergo cell multiplication and movement. This report demonstrates that whereas in confluent EC in a compact monolayer, the EC-EC adhesion molecule platelet-endothelial cell adhesion molecule-1 (PECAM-1) is strongly expressed at cell borders, little or no PECAM-1 immunostaining is detected in sparse or migrating cultured EC. Consistent with this observation, steady-state PECAM-1 mRNA expression was much lower in subconfluent EC than in confluent EC. The absence of PECAM-1 expression in sparse EC appeared not to be linked to ability to proliferate, since PECAM-1 expression remained low even in the presence of nitric oxide (NO) or mitomycin C, agents that inhibit EC growth. However, another growth-inhibitory agent, TGF-beta 1, did not alter PECAM-1 staining. Based on these observations, it is hypothesized that cell-associated mechanical forces underlying cell tensegrity regulate PECAM-1 expression.
- Published
- 2001
- Full Text
- View/download PDF
31. 7E3 F(ab')2, an effective antagonist of rat alphaIIbbeta3 and alphavbeta3, blocks in vivo thrombus formation and in vitro angiogenesis.
- Author
-
Sassoli PM, Emmell EL, Tam SH, Trikha M, Zhou Z, Jordan RE, and Nakada MT
- Subjects
- Abciximab, Animals, Antibodies, Monoclonal chemistry, Aorta injuries, Aorta pathology, Disease Models, Animal, Dose-Response Relationship, Drug, Immunoglobulin Fab Fragments chemistry, Kinetics, Male, Mice, Microcirculation, Platelet Aggregation drug effects, Platelet Glycoprotein GPIIb-IIIa Complex immunology, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Rats, Rats, Inbred Strains, Receptors, Vitronectin immunology, Receptors, Vitronectin metabolism, Thrombosis prevention & control, Antibodies, Monoclonal pharmacology, Immunoglobulin Fab Fragments pharmacology, Neovascularization, Physiologic drug effects, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Receptors, Vitronectin antagonists & inhibitors, Thrombosis drug therapy
- Abstract
Abciximab (c7E3 Fab, ReoPro) blocks GPIIb/IIIa and alphavbeta3 and inhibits thrombotic and proliferative events only in humans and non-human primates. The bivalent F(ab')2 fragment is an effective anti-thrombotic agent in canine models. In the present study, 7E3 F(ab')2 was also found to bind to rat GPIIb/IIIa (KD = 27 +/- 4 microg/mL) and alphavbeta3 (KD = 9 +/- 8 microg/mL), to block in vitro rat platelet aggregation (IC50 = 16 +/- 6 microg/mL), and to inhibit alphavbeta3-mediated microvessel sprout formation in a rat aortic ring assay. Following administration of 7E3 F(ab')2 (4 mg/kg) to rats, platelet aggregation was completely blocked for up to 6 h and thrombus formation in response to a rat abdominal aorta double crush injury was prevented. Effective chronic dosing was achieved with 6 mg/kg daily I.P. injections. In vitro mixing experiments indicated that 7E3 F(ab')2 redistributed to unlabeled platelets in 2 h. Ex vivo, 7E3 F(ab')2 was detected on platelets for up to 4 days after a single 4-mg/kg injection. These data suggest that 7E3 F(ab')2 may be a useful agent to study the effects of GPIIb/IIIa and alphavbeta3 blockade in rat models of thrombosis and vascular disease.
- Published
- 2001
32. Angiographic evaluation of middle cerebral artery reperfusion caused by platelet glycoprotein IIb/IIIa receptor complex antagonist murine 7E3 F(ab')2 in a model of focal cerebral ischemia in rats.
- Author
-
Yang Y, Li Q, Nakada MT, Yang T, and Shuaib A
- Subjects
- Animals, Antibodies, Monoclonal adverse effects, Bleeding Time, Brain Ischemia complications, Cerebral Hemorrhage chemically induced, Cerebral Hemorrhage epidemiology, Cerebral Infarction pathology, Incidence, Male, Nervous System Diseases etiology, Nervous System Diseases physiopathology, Rats, Rats, Wistar, Tail blood supply, Antibodies, Monoclonal therapeutic use, Brain Ischemia drug therapy, Brain Ischemia physiopathology, Cerebral Angiography, Middle Cerebral Artery physiopathology, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Reperfusion
- Abstract
Object: Antagonists of the glycoprotein IIb/IIIa (GPIIb/IIIa) receptor complex are currently used for the treatment of acute coronary syndromes. The platelet GPIIb/IIIa mediates platelet aggregation, and blocking this receptor complex can reduce or prevent arterial thrombosis. To study the recanalization efficacy of a GPIIb/IIIa antagonist in treating cerebral ischemia, we investigated the therapeutic effects of murine 7E3 F(ab'), in a focal embolic cerebral ischemia model in rats., Methods: Focal cerebral ischemia was produced by introducing an autologous thrombus into the right side of the middle cerebral artery (MCA). Thirty male Wistar rats were randomly divided into three groups of 10 rats each: control, 7E3 F(ab')2 administered 1 hour postischemia, and 7E3 F(ab')2 administered 3 hours postischemia. Animals in the therapeutic groups received intravenous infusion of 6 mg/kg 7E3 F(ab')2 at 1 or 3 hours following cerebral embolization. Brain infarct volume, neurobehavioral scores, duration of bleeding, and findings on angiograms of the MCA (before and after infusion) were assessed in all animals. Angiographic evaluation revealed full MCA recanalization in three of 10 animals in each 7E3 F(ab')2 treatment group. Animals in these groups exhibited a significant reduction in infarct volume when compared with animals in the control group: 1) infarct volume 1 hour postischemia, 22 +/- 13.9% (p = 0.005); 2) infarct volume 3 hours postischemia, 22.1 +/- 14.8% (p = 0.008); and 3) infarct volume in control animals, 42.4 +/- 16%. Postischemia treatment with 7E3 F(ab')2 also improved the animal's neurobehavioral performance. The duration of bleeding significantly increased by more than two times, but there was no associated increase in intracerebral hemorrhage in any group., Conclusions: On the basis of their findings, the authors conclude that murine 7E3 F(ab'), is a potent and safe antiplatelet agent in this experimental focal embolic cerebral ischemia model. Neuronal lesions were significantly reduced when the treatment was delayed up to 3 hours.
- Published
- 2001
- Full Text
- View/download PDF
33. Separate pathways for cellular uptake of ferric and ferrous iron.
- Author
-
Conrad ME, Umbreit JN, Moore EG, Hainsworth LN, Porubcin M, Simovich MJ, Nakada MT, Dolan K, and Garrick MD
- Subjects
- Amino Acid Substitution, Animals, Antigens, CD metabolism, Biological Transport, Cations metabolism, Cations, Divalent metabolism, Cell Line, Chlorides pharmacokinetics, Humans, Integrin beta3, K562 Cells, Kidney, Manganese Compounds pharmacokinetics, Mice, Platelet Membrane Glycoproteins metabolism, Rats, Recombinant Proteins metabolism, Transfection, Zinc Compounds pharmacokinetics, Carrier Proteins metabolism, Cation Transport Proteins, Ferric Compounds pharmacokinetics, Ferrous Compounds pharmacokinetics, Iron-Binding Proteins
- Abstract
Separate pathways for transport of nontransferrin ferric and ferrous iron into tissue cultured cells were demonstrated. Neither the ferric nor ferrous pathway was shared with either zinc or copper. Manganese shared the ferrous pathway but had no effect on cellular uptake of ferric iron. We postulate that ferric iron was transported into cells via beta(3)-integrin and mobilferrin (IMP), whereas ferrous iron uptake was facilitated by divalent metal transporter-1 (DMT-1; Nramp-2). These conclusions were documented by competitive inhibition studies, utilization of a beta(3)-integrin antibody that blocked uptake of ferric but not ferrous iron, development of an anti-DMT-1 antibody that blocked ferrous iron and manganese uptake but not ferric iron, transfection of DMT-1 DNA into tissue culture cells that showed enhanced uptake of ferrous iron and manganese but neither ferric iron nor zinc, hepatic metal concentrations in mk mice showing decreased iron and manganese but not zinc or copper, and data showing that the addition of reducing agents to tissue culture media altered iron binding to proteins of the IMP and DMT-1 pathways. Although these experiments show ferric and ferrous iron can enter cells via different pathways, they do not indicate which pathway is dominant in humans.
- Published
- 2000
- Full Text
- View/download PDF
34. GPIIb-IIIa antagonist-induced reduction in platelet surface factor V/Va binding and phosphatidylserine expression in whole blood.
- Author
-
Furman MI, Krueger LA, Frelinger AL 3rd, Barnard MR, Mascelli MA, Nakada MT, and Michelson AD
- Subjects
- Abciximab, Antibodies, Monoclonal pharmacology, Blood Platelets metabolism, Collagen pharmacology, Dose-Response Relationship, Drug, Eptifibatide, Flow Cytometry, Humans, Immunoglobulin Fab Fragments pharmacology, Infant, Newborn, Membrane Proteins metabolism, Peptides pharmacology, Protein Binding, Thrombasthenia blood, Thrombin antagonists & inhibitors, Thrombin pharmacology, Tirofiban, Tyrosine pharmacology, Factor V metabolism, Phosphatidylserines blood, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Tyrosine analogs & derivatives
- Abstract
In addition to inhibition of platelet aggregation, GPIIb-IIIa antagonists may reduce thrombotic events via other mechanisms. In a novel whole blood flow cytometric system, we investigated the effects of GPIIb-IIIa antagonists, in the presence or absence of thrombin inhibitors, on platelet surface-bound factor V/Va and platelet surface phospholipids. Diluted venous blood was incubated with either buffer or a GPIIb-IIIa antagonist (abciximab, tirofiban, or eptifibatide). Some samples were pre-incubated with clinically relevant concentrations of unfractionated heparin (UFH), a low molecular weight heparin, a direct thrombin inhibitor, or buffer only. Platelets were then activated and labeled with mAb V237 (factor V/Va-specific) or annexin V (binds phosphatidylserine), fixed, and analyzed by flow cytometry. In the absence of thrombin inhibitors, GPIIb-IIIa antagonists (especially abciximab) significantly reduced agonist-induced platelet procoagulant activity, as determined by reduced binding of V237 and annexin V. At high pharmacologic concentrations, unfractionated heparin and enoxaparin, but not hirudin, further reduced factor V/Va binding to the surface of activated platelets in the presence of GPIIb-IIa antagonists. Agonist-induced platelet procoagulant activity was reduced in a patient with Glanzmann's thrombasthenia. We conclude that GPIIb-IIIa antagonists reduce platelet procoagulant activity in whole blood and heparin and enoxaparin augment this reduction. Fibrinogen binding to GPIIb-IIIa is important in the generation of platelet procoagulant activity.
- Published
- 2000
35. Divergent effects of platelet-endothelial cell adhesion molecule-1 and beta 3 integrin blockade on leukocyte transmigration in vivo.
- Author
-
Thompson RD, Wakelin MW, Larbi KY, Dewar A, Asimakopoulos G, Horton MA, Nakada MT, and Nourshargh S
- Subjects
- Abciximab, Animals, Antibody Specificity, Cell Migration Inhibition, Cell Movement immunology, Endothelium, Vascular immunology, Endothelium, Vascular ultrastructure, Immunoglobulin Fab Fragments pharmacology, Immunosuppressive Agents pharmacology, Integrin beta3, Interleukin-1 pharmacology, Leukocytes cytology, Leukocytes immunology, Male, Mesentery blood supply, N-Formylmethionine Leucyl-Phenylalanine pharmacology, Neutrophils immunology, Protein Structure, Tertiary, Rats, Rats, Sprague-Dawley, Venules immunology, Venules ultrastructure, Antibodies, Blocking pharmacology, Antibodies, Monoclonal pharmacology, Antigens, CD immunology, Platelet Endothelial Cell Adhesion Molecule-1 immunology, Platelet Membrane Glycoproteins antagonists & inhibitors, Platelet Membrane Glycoproteins immunology
- Abstract
The final stage in the migration of leukocytes to sites of inflammation involves movement of leukocytes through the endothelial cell layer and the perivascular basement membrane. Both platelet-endothelial cell adhesion molecule-1 (PECAM-1/CD31) and the integrin alphavbeta3 have been implicated in this process, and in vitro studies have identified alphavbeta3 as a heterotypic ligand for PECAM-1. In the present study we have addressed the roles of these molecules by investigating and comparing the effects of PECAM-1 and alphavbeta3 blockade on leukocyte migration in vivo. For this purpose we have examined the effects of neutralizing Abs directed against PECAM-1 (domain 1-specific, mAb 37) and beta3 integrins (mAbs 7E3 and F11) on leukocyte responses in the mesenteric microcirculation of anesthetized rats using intravital microscopy. The anti-PECAM-1 mAb suppressed leukocyte extravasation, but not leukocyte rolling or firm adhesion, elicited by IL-1beta in a dose-dependent manner (e.g., 67% inhibition at 10 mg/kg 37 Fab), but had no effect on FMLP-induced leukocyte responses. Analysis by electron microscopy suggested that this suppression was due to an inhibition of neutrophil migration through the endothelial cell barrier. By contrast, both anti-beta3 integrin mAbs, 7E3 F(ab')2 (5 mg/kg) and F11 F(ab')2 (5 mg/kg), selectively reduced leukocyte extravasation induced by FMLP (38 and 46%, respectively), but neither mAb had an effect on IL-1beta-induced leukocyte responses. These findings indicate roles for both PECAM-1 and beta3 integrins in leukocyte extravasation, but do not support the concept that these molecules act as counter-receptors in mediating leukocyte transmigration.
- Published
- 2000
- Full Text
- View/download PDF
36. Monoclonal antibodies to alphaVbeta3 (7E3 and LM609) inhibit sickle red blood cell-endothelium interactions induced by platelet-activating factor.
- Author
-
Kaul DK, Tsai HM, Liu XD, Nakada MT, Nagel RL, and Coller BS
- Subjects
- Abciximab, Adult, Animals, Antibodies, Monoclonal, Humanized, Cecum blood supply, Cell Adhesion drug effects, Endothelium, Vascular drug effects, Erythrocytes drug effects, Erythrocytes immunology, Humans, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Rats, Receptors, Vitronectin immunology, Reference Values, Venules physiology, Anemia, Sickle Cell blood, Antibodies, Monoclonal pharmacology, Cell Adhesion physiology, Endothelium, Vascular physiology, Erythrocytes physiology, Immunoglobulin Fab Fragments pharmacology, Platelet Activating Factor pharmacology, Receptors, Vitronectin physiology
- Abstract
Abnormal interaction of sickle red blood cells (SS RBC) with the vascular endothelium has been implicated as a factor in the initiation of vasoocclusion in sickle cell anemia. Both von Willebrand factor (vWf) and thrombospondin (TSP) play important roles in mediating SS RBC-endothelium interaction and can bind to the endothelium via alphaVbeta3 receptors. We have used monoclonal antibodies (MoAb) directed against alphaVbeta3 and alphaIIbbeta3 (GPIIb/IIIa) integrins to dissect the role of these integrins in SS RBC adhesion. The murine MoAb 7E3 inhibits both alphaVbeta3 and alphaIIbbeta3 (GPIIb/IIIa), whereas MoAb LM609 selectively inhibits alphaVbeta3, and MoAb 10E5 binds only to alphaIIbbeta3. In this study, we have tested the capacity of these MoAbs to block platelet-activating factor (PAF)-induced SS RBC adhesion in the ex vivo mesocecum vasculature of the rat. Infusion of washed SS RBC in preparations treated with PAF (200 pg/mL), with or without a control antibody, resulted in extensive adhesion of these cells in venules, accompanied by frequent postcapillary blockage and increased peripheral resistance units (PRU). PAF also caused increased endothelial surface and interendothelial expression of endothelial vWf. Importantly, pretreatment ofthe vasculature with either MoAb 7E3 F(ab')(2) or LM609, but not 10E5 F(ab')(2), after PAF almost completely inhibited SS RBC adhesion in postcapillary venules, the sites of maximal adhesion and frequent blockage. The inhibition of adhesion with 7E3 or LM609 was accompanied by smaller increases in PRU and shorter pressure-flow recovery times. Thus, blockade of alphaVbeta3 may constitute a potential therapeutic approach to prevent SS RBC-endothelium interactions under flow conditions. (Blood. 2000;95:368-374)
- Published
- 2000
37. Contortrostatin, a homodimeric disintegrin, binds to integrin alphavbeta5.
- Author
-
Zhou Q, Nakada MT, Brooks PC, Swenson SD, Ritter MR, Argounova S, Arnold C, and Markland FS
- Subjects
- Agkistrodon, Animals, Binding Sites, Cell Adhesion drug effects, Dimerization, Disintegrins chemistry, Disintegrins pharmacokinetics, Humans, Integrins chemistry, Kinetics, Neoplasm Invasiveness, Radioligand Assay, Receptors, Vitronectin chemistry, Receptors, Vitronectin metabolism, Tumor Cells, Cultured, Urinary Bladder Neoplasms, Viper Venoms, Disintegrins pharmacology, Integrins metabolism
- Abstract
Contortrostatin is a homodimeric disintegrin from snake venom. We have shown that contortrostatin binds to integrins alphaIIbbeta3, alpha5beta1, and alphavbeta3. We now use several criteria to demonstrate the binding of contortrostatin to alphavbeta5. First, incubation of T24 cells, which express alphavbeta3 and alphavbeta5, with antibody against alphavbeta3 failed to completely inhibit adhesion of cells to vitronectin. However, pretreatment of the cells with contortrostatin or the combination of antibodies against alphavbeta3 and alphavbeta5 completely blocked adhesion to vitronectin. By contrast, either anti-alphavbeta5 alone or contortrostatin blocked adhesion of an alphavbeta3-negative T24 subline. Second, contortrostatin as well as anti-alphavbeta5 inhibits invasion of OVCAR-5, which express only alphavbeta5. Third, contortrostatin binds to purified alphavbeta5 in a saturable manner. Finally, radioligand binding assays yielded a K(d) value of 24 nM for [(125)I]contortrostatin binding to alphavbeta5. This investigation identifies alphavbeta5 as a binding site for contortrostatin. Blockage of alphavbeta5 by contortrostatin inhibits alphavbeta5-mediated adhesion and invasion., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
38. Antibodies against the first Ig-like domain of human platelet endothelial cell adhesion molecule-1 (PECAM-1) that inhibit PECAM-1-dependent homophilic adhesion block in vivo neutrophil recruitment.
- Author
-
Nakada MT, Amin K, Christofidou-Solomidou M, O'Brien CD, Sun J, Gurubhagavatula I, Heavner GA, Taylor AH, Paddock C, Sun QH, Zehnder JL, Newman PJ, Albelda SM, and DeLisser HM
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal biosynthesis, Antibodies, Monoclonal metabolism, Binding Sites, Antibody, Binding, Competitive, Cattle, Cell Adhesion immunology, Cell Aggregation immunology, Cell Movement immunology, Endothelium, Vascular immunology, Endothelium, Vascular metabolism, Epitope Mapping, Humans, L Cells, Mice, Mice, SCID, Molecular Sequence Data, Neutrophils immunology, Neutrophils pathology, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Platelet Endothelial Cell Adhesion Molecule-1 physiology, Protein Structure, Tertiary, Rats, Recombinant Fusion Proteins biosynthesis, Skin Transplantation pathology, Antibodies, Monoclonal pharmacology, Immunoglobulins immunology, Neutrophil Activation immunology, Peptide Fragments immunology, Platelet Endothelial Cell Adhesion Molecule-1 immunology
- Abstract
Platelet endothelial cell adhesion molecule (PECAM-1), a member of the Ig superfamily, is found on endothelial cells and neutrophils and has been shown to be involved in the migration of leukocytes across the endothelium. Adhesion is mediated, at least in part, through binding interactions involving its first N-terminal Ig-like domain, but it is still unclear which sequences in this domain are required for in vivo function. Therefore, to identify functionally important regions of the first Ig-like domain of PECAM-1 that are required for the participation of PECAM-1 in in vivo neutrophil recruitment, a panel of mAbs against this region of PECAM-1 was generated and characterized in in vitro adhesion assays and in an in vivo model of cutaneous inflammation. It was observed that mAbs that disrupted PECAM-1-dependent homophilic adhesion in an L cell aggregation assay also blocked TNF-alpha-induced intradermal accumulation of neutrophils in a transmigration model using human skin transplanted onto SCID mice. Localization of the epitopes of these Abs indicated that these function-blocking Abs mapped to specific regions on either face of domain 1. This suggests that these regions of the first Ig-like domain may contain or be close to binding sites involved in PECAM-1-dependent homophilic adhesion, and thus may represent potential targets for the development of antiinflammatory reagents.
- Published
- 2000
- Full Text
- View/download PDF
39. Loss of endothelial surface expression of E-selectin in a patient with recurrent infections.
- Author
-
DeLisser HM, Christofidou-Solomidou M, Sun J, Nakada MT, and Sullivan KE
- Subjects
- Bacterial Infections pathology, Cell Adhesion, Cell Movement immunology, Child, E-Selectin genetics, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Female, Humans, Immunity, Innate genetics, Inflammation genetics, Inflammation immunology, Leukocytes immunology, Leukocytes pathology, Bacterial Infections immunology, E-Selectin biosynthesis, Endothelium, Vascular immunology
- Abstract
Neutrophil accumulation at sites of inflammation is mediated by specific groups of cell adhesion molecules including the beta2 (CD18) integrins on leukocytes and the selectins (P- and E-selectin on the endothelium and L-selectin on the leukocyte). This is supported by studies of patients with leukocyte adhesion deficiency syndromes whose leukocytes are genetically deficient in the expression of beta2 integrins or selectin carbohydrate ligands (eg, sialyl-Lewis(x)). However, inherited deficiency or dysfunction of endothelial cell adhesion molecules involved in leukocyte recruitment has not been previously described. In this report we describe a child with recurrent infections and clinical evidence of impaired pus formation reminiscent of a leukocyte adhesion deficiency syndrome, but whose neutrophils were functionally normal and expressed normal levels of CD18, L-selectin, and sialyl-Lewis(x). In contrast, immunohistochemical staining of inflamed tissue from the patient showed the absence of E-selectin from the endothelium, although E-selectin mRNA was present. However, E-selectin protein was expressed as significantly elevated levels of circulating soluble E-selectin were detected, the molecular size of which was consistent with a proteolytically cleaved form of E-selectin. Gene sequencing failed to show evidence of a secreted mutant variant. These data represent, to our knowledge, the first description of a potentially inherited dysfunction of an endothelial cell adhesion molecule involved in leukocyte recruitment and provide additional human evidence of the importance of endothelial selectins in the inflammatory response.
- Published
- 1999
40. Contortrostatin, a dimeric disintegrin from Agkistrodon contortrix contortrix, inhibits angiogenesis.
- Author
-
Zhou Q, Nakada MT, Arnold C, Shieh KY, and Markland FS Jr
- Abstract
Contortrostatin, a 13.5 kDa disulfide-linked homodimeric polypeptide possessing an Arg-Gly-Asp sequence, was isolated from venom of the southern copperhead snake. Daily injection of contortrostatin into the primary tumor of human breast cancer MDA-MB-435 carried in nude mice significantly inhibited tumor growth and neovascularization of the tumor tissue. On the chick embryo chorioallantoic membrane, contortrostatin inhibited angiogenesis induced by MDA-MB-435 cells, basic fibroblast growth factor, and vascular endothelial growth factor. In addition, contortrostatin effectively blocked adhesion of human umbilical vein endothelial cells (HUVEC) to immobilized vitronectin and significantly inhibited invasion of HUVEC through a Matrigel barrier. Competitive binding assays and adhesion assays with different integrin antibodies suggested that integrin alpha(v)beta3 is a binding site for contortrostatin on vascular endothelial cells. Detachment of HUVEC from vitronectin by contortrostatin induced apoptosis. HUVEC adhered and spread well on immobilized contortrostatin without undergoing apoptosis, suggesting that it is the inhibition of adhesion and spreading of HUVEC on extracellular matrix proteins, rather than binding of contortrostatin to integrins per se, that triggers apoptosis. We conclude that contortrostatin binds to alpha(v)beta3, and interferes with the anchorage-dependent survival mechanism of the vascular endothelial cells, and the mobility of the cells. The consequent suppression of angiogenesis is an important component of the antineoplastic activity of contortrostatin.
- Published
- 1999
- Full Text
- View/download PDF
41. Inhibition of angiogenesis and tumor growth by murine 7E3, the parent antibody of c7E3 Fab (abciximab; ReoPro).
- Author
-
Varner JA, Nakada MT, Jordan RE, and Coller BS
- Abstract
Angiogenesis plays an essential role in the growth and dissemination of solid tumor cancers. The expression of endothelial cell integrin alpha(v)beta3 has been shown to increase during vascular proliferation associated with human tumors. Selective antagonists of alpha(v)beta3 can block angiogenesis and tumor growth by inducing programmed cell death in proliferating endothelial cells. Monoclonal antibody 7E3, an antagonist of the human, but not murine, integrins alpha(v)beta3 and alphaIIbbeta3 (GPIIb/IIIa), inhibits platelet aggregation. It is the parent antibody of a mouse/human chimeric antibody fragment approved for adjunctive therapy of patients undergoing percutaneous coronary interventions to prevent ischemic complications (c7E3Fab; abciximab; ReoPro). To evaluate the potential of 7E3 to inhibit human angiogenesis and tumor growth independent of its antiplatelet effects, we established integrin alpha(v)beta3-negative human melanoma tumors in full-thickness human skin grafted onto SCID mice. The resulting tumors induce a human angiogenic response as assessed by the immunoreactivity of vascular cells with monoclonal antibodies specific for human CD31. Administration of 7E3 prevented or significantly inhibited the growth of tumors, and this effect correlated with a significant reduction in the number of blood vessels supplying the tumors. These results support the previous findings that blockade of integrin alpha(v)beta3 inhibits angiogenesis and tumor growth and indicates that dual inhibitors of alpha(v)beta3 and alphaIIbbeta3 are effective in blocking tumor growth and angiogenesis.
- Published
- 1999
- Full Text
- View/download PDF
42. Effects of beta3-integrin blockade (c7E3) on the response to angioplasty and intra-arterial stenting in atherosclerotic nonhuman primates.
- Author
-
Deitch JS, Williams JK, Adams MR, Fly CA, Herrington DM, Jordan RE, Nakada MT, Jakubowski JA, and Geary RL
- Subjects
- Abciximab, Animals, Arteries, Blood Coagulation drug effects, Combined Modality Therapy, Drug Evaluation, Preclinical, Hematologic Tests, Hyperplasia drug therapy, Lipids blood, Macaca fascicularis, Male, Stents, Treatment Outcome, Angioplasty, Balloon, Coronary, Antibodies, Monoclonal therapeutic use, Arteriosclerosis therapy, Immunoglobulin Fab Fragments therapeutic use, Integrins antagonists & inhibitors, Platelet Aggregation Inhibitors therapeutic use
- Abstract
Because the beta3-antagonist abciximab (c7E3 Fab) has significantly improved late outcomes after coronary angioplasty, the beta3 integrins have been implicated in the arterial response to injury. However, the mechanisms underlying this benefit are unknown. The observation that c7E3 binds beta3 integrins on vascular cells (alphavbeta3) with affinity equal to that for the platelet glycoprotein IIb/IIIa integrin has led to the hypothesis that c7E3 may act directly on the artery wall to prevent restenosis after angioplasty. To test this hypothesis, we studied the effects of c7E3 on structural changes within the artery wall after angioplasty or stent angioplasty in 23 male cynomolgus monkeys with established atherosclerosis. Animals were randomly assigned to receive either a bolus of c7E3 (0.4 mg/kg IV, n=11) followed by a 48-hour infusion (0. 2 microg. kg-1. min-1) or an equal volume of vehicle (n=12). Animals received weight-adjusted aspirin and heparin and then underwent unilateral iliac artery experimental angioplasty and subclavian artery stent angioplasty (Palmaz). Iliac artery lumen diameter (LD) was determined by angiography at baseline (LDPre), after angioplasty (LDPost), and 35 days later (LDDay35). Arteries were then fixed by perfusion and removed for analysis. Lumen, intima, media, and external elastic lamina (EEL) areas were measured in iliac artery cross sections. Values from each injured iliac artery were normalized to the contralateral uninjured iliac artery to control for interanimal variability in baseline artery size and atherosclerosis extent. Intimal area was also measured in subclavian stent cross sections. c7E3 blocked platelet aggregation and prolonged the bleeding time from 2.8+/-1.1 to 19.8+/-2.5 minutes, P<0.001. Experimental angioplasty increased LDPost an average of 28%, and the initial gain was similar in both groups (P=NS). Despite an anti-platelet effect, c7E3 did not inhibit iliac lumen narrowing (LDDay35-LDPost: c7E3, -0.69+/-0.17 versus vehicle, -0.99+/-.17 mm, P=0.35); intimal hyperplasia (neointima area: c7E3, 1.12+/-.28 versus vehicle, 1.22+/-.20 mm2, P=0.77); or decrease in artery wall size (EEL area [percent of uninjured control]: c7E3, 101+/-7% versus vehicle, 121+/-7%). Stent intimal hyperplasia was also unaltered by c7E3 treatment (neointimal area: c7E3, 1.09+/-0.16 versus vehicle, 1. 28+/-0.11 mm2, P=0.36). These results suggest that the benefits of c7E3 treatment in coronary angioplasty were not from inhibition of intimal hyperplasia or improved artery wall remodeling. Alternative mechanisms should be explored to explain improved late outcomes after angioplasty in patients treated with c7E3.
- Published
- 1998
- Full Text
- View/download PDF
43. Abciximab (ReoPro, chimeric 7E3 Fab) demonstrates equivalent affinity and functional blockade of glycoprotein IIb/IIIa and alpha(v)beta3 integrins.
- Author
-
Tam SH, Sassoli PM, Jordan RE, and Nakada MT
- Subjects
- Abciximab, Binding, Competitive immunology, Cells, Cultured, Coronary Vessels cytology, Cross Reactions, Endothelium, Vascular drug effects, Fluorescein-5-isothiocyanate, Fluorescent Dyes, Humans, Iodine Radioisotopes, Keratinocytes cytology, Muscle, Skeletal cytology, Muscle, Smooth, Vascular drug effects, Platelet Glycoprotein GPIIb-IIIa Complex immunology, Receptors, Vitronectin immunology, Recombinant Fusion Proteins pharmacology, Species Specificity, Umbilical Veins cytology, Antibodies, Monoclonal pharmacology, Immunoglobulin Fab Fragments pharmacology, Platelet Aggregation Inhibitors pharmacology, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Receptors, Vitronectin antagonists & inhibitors
- Abstract
Background: Large, randomized, and blinded clinical trials (EPIC, EPILOG, and CAPTURE) have demonstrated that abciximab (ReoPro, chimeric 7E3 Fab) markedly reduces thrombotic events associated with percutaneous transluminal coronary interventions. The marked early benefits at 30 days were sustained at 6 months and 3 years. Initially developed because of its efficacy in blocking GP IIb/IIIa (alphaIIb/beta3) receptors on platelets, abciximab also binds with equivalent affinity to alpha(v)beta3., Methods and Results: This study presents a detailed characterization of the alphavss3 interaction, including the ability of abciximab to (1) bind with comparable affinity to alpha(v)beta3 and GP IIb/IIIa, (2) inhibit alpha(v)beta3 and GP IIb/IIIa-mediated cell adhesion in vitro with IC50 values approximating binding KD values, and (3) redistribute between GP IIb/IIIa and alpha(v)beta3 integrins in vitro., Conclusions: As an antagonist of not only GP IIb/IIIa but also alpha(v)beta3, abciximab may provide additional clinical benefit in preventing alpha(v)beta3-mediated effects such as thrombin generation, clot retraction, or smooth muscle cell migration and proliferation. Abciximab binds with equivalent affinity to both GP IIb/IIIa and alphavss3 and redistributes between the 2 integrin receptors in vitro. Abciximab has been previously shown to circulate on platelets for up to 2 weeks. Taken together, these findings suggest that abciximab may have the ability to inhibit both GP IIb/IIIa and alpha(v)beta3 for extended periods.
- Published
- 1998
- Full Text
- View/download PDF
44. Neutralization of TNF by the antibody cA2 reveals differential regulation of adhesion molecule expression on TNF-activated endothelial cells.
- Author
-
Nakada MT, Tam SH, Woulfe DS, Casper KA, Swerlick RA, and Ghrayeb J
- Subjects
- Actin Cytoskeleton metabolism, Antineoplastic Agents pharmacology, Blotting, Northern, Cell Adhesion Molecules genetics, Cell Line, Cell Survival drug effects, Colchicine pharmacology, Cytochalasin B pharmacology, Down-Regulation, E-Selectin genetics, E-Selectin metabolism, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Humans, Infliximab, Intercellular Adhesion Molecule-1 genetics, Intercellular Adhesion Molecule-1 metabolism, Microtubules metabolism, Nucleic Acid Synthesis Inhibitors pharmacology, Protein Synthesis Inhibitors pharmacology, Tumor Necrosis Factor-alpha immunology, Tumor Necrosis Factor-alpha pharmacology, Umbilical Veins cytology, Up-Regulation, Vascular Cell Adhesion Molecule-1 genetics, Vascular Cell Adhesion Molecule-1 metabolism, Antibodies, Monoclonal pharmacology, Cell Adhesion Molecules metabolism, Endothelium, Vascular metabolism, Gene Expression Regulation drug effects, Tumor Necrosis Factor-alpha metabolism
- Abstract
Upregulation of adhesion proteins plays an important role in mediating inflammation. The induction of adhesive molecules has been well studied, but the reversibility of their expression has not been well characterized. A neutralizing anti-TNF monoclonal antibody (cA2) was used to study the down regulation of TNF-induced E-selectin, vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) on cultured human umbilical vein endothelial cells (HUVECs). Addition of cA2 following TNF stimulation of HUVECs enhanced the rate of E-selectin and VCAM-1 down-regulation from the cell surface and also reduced steady state E-selectin and VCAM-1 mRNA levels. The cA2-mediated disappearance of E-selectin, but not VCAM-1 protein was microtubule and not microfilament dependent. Neutralization of TNF only slightly reduced ICAM-1 cell surface levels following initial TNF stimulation, suggesting a slower turnover of ICAM-1 compared to E-selectin and VCAM-1. Microtubule inhibition during TNF stimulation partially inhibited E-selectin, VCAM-1 and ICAM-1 mRNA upregulation. VCAM-1 and ICAM-1 cell surface expression were similarly partially inhibited, however, E-selectin levels were unaffected, presumably due to the dual, opposing effect of inhibiting protein expression and inhibiting internalization. Microfilament inhibition during protein induction specifically inhibited the maximal expression of VCAM-1 protein and mRNA, without affecting E-selectin or ICAM-1. These data support the notion that E-selectin, VCAM-1, and ICAM-1 expression are differentially regulated on HUVECs and suggest that TNF neutralizing therapies may be effective because of their ability to reduce the levels of pre-existing adhesion proteins.
- Published
- 1998
- Full Text
- View/download PDF
45. Beta3 integrins are upregulated after vascular injury and modulate thrombospondin- and thrombin-induced proliferation of cultured smooth muscle cells.
- Author
-
Stouffer GA, Hu Z, Sajid M, Li H, Jin G, Nakada MT, Hanson SR, and Runge MS
- Subjects
- Abciximab, Angioplasty, Balloon adverse effects, Animals, Antibodies, Monoclonal pharmacology, Brachial Artery injuries, Brachial Artery metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Division drug effects, Cells, Cultured, Humans, Immunoglobulin Fab Fragments pharmacology, Integrin beta3, Integrins metabolism, Male, Muscle, Smooth, Vascular drug effects, Papio, Platelet Aggregation Inhibitors pharmacology, Signal Transduction, Thrombin pharmacology, Thrombospondins pharmacology, Up-Regulation, Antibodies, Monoclonal metabolism, Antigens, CD metabolism, Immunoglobulin Fab Fragments metabolism, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Platelet Aggregation Inhibitors metabolism, Platelet Membrane Glycoproteins metabolism
- Abstract
Background: Treatment with an antibody that binds beta3 integrins (abciximab; c7E3 Fab) at the time of coronary angioplasty decreases the need for repeat revascularization. Two potential mechanisms have been proposed to explain this effect: (1) inhibition of platelet aggregation or (2) interruption of ligand binding to beta3 integrins on the smooth muscle cell (SMC) surface. We examined the latter hypothesis by determining (1) if beta3 integrin expression is upregulated after vascular injury in the baboon, (2) if 7E3 binds beta3 integrins on cultured SMC, and (3) if beta3 integrin activation plays a role in proliferation of cultured SMC., Methods and Results: Results demonstrated that immunostaining for beta3 integrins was present in the neointima 1 week after balloon withdrawal injury of baboon brachial arteries and that beta3 integrin expression colocalized with alpha-actin-positive cells. In contrast, staining for beta3 integrins was undetectable in contralateral uninjured brachial arteries. 7E3 bound to cultured human aortic SMC with an affinity (KD=3.3 nmol/L) similar to 7E3 binding to endothelial cells or platelets. Cotreatment with 7E3 partially inhibited thrombospondin-induced or alpha-thrombin-induced proliferation but not PDGF-induced or serum-induced proliferation., Conclusions: In summary, these studies demonstrate that vascular cell beta3 integrin expression is increased after injury, that 7E3 binds to cultured SMC with high affinity, and that beta3 activation is important for thrombospondin-induced or alpha-thrombin-induced proliferation. These results support the hypothesis that beta3 integrins play a role in SMC growth responses after balloon injury.
- Published
- 1998
- Full Text
- View/download PDF
46. Neutrophil platelet endothelial cell adhesion molecule-1 participates in neutrophil recruitment at inflammatory sites and is down-regulated after leukocyte extravasation.
- Author
-
Christofidou-Solomidou M, Nakada MT, Williams J, Muller WA, and DeLisser HM
- Subjects
- Animals, Antibodies, Monoclonal immunology, Down-Regulation, Endothelium, Vascular physiology, Humans, Mice, Mice, Inbred BALB C, Mice, SCID, Tumor Necrosis Factor-alpha pharmacology, Chemotaxis, Leukocyte, Inflammation physiopathology, Neutrophils physiology, Platelet Endothelial Cell Adhesion Molecule-1 physiology
- Abstract
Platelet endothelial cell adhesion molecule-1 (PECAM-1), a member of the Ig superfamily, is found on endothelial cells and neutrophils, and has been shown to be involved in the migration of leukocytes across the endothelium. Although studies have supported a role for endothelial PECAM-1 in this process, the participation of neutrophil PECAM-1 in vivo has not been unambiguously demonstrated. Therefore, to examine the involvement of neutrophil PECAM-1 in leukocyte recruitment, we studied the effect of a blocking Ab against murine PECAM-1 on neutrophil recruitment in an established model of murine peritonitis and in a murine model for studying leukocyte-endothelial interactions involving the human vasculature. These studies not only confirmed that neutrophil PECAM-1 is important in the accumulation of neutrophils at inflammatory sites, but that extravasated neutrophils displayed decreased surface expression of PECAM-1. In vitro, the surface expression of murine neutrophil PECAM-1 was not decreased significantly by inflammatory mediators, but was reduced after transendothelial migration. These studies, consistent with previous in vitro observations, confirm that neutrophil PECAM-1, as well as endothelial PECAM-1, is involved in the recruitment of neutrophils into inflammatory sites in vivo, and suggest that the expression of neutrophil PECAM-1 is down-regulated after extravasation into inflamed tissues possibly as a result of engagement of its ligand.
- Published
- 1997
47. Structure-function studies on synthetic peptides derived from the 109-118 lectin domain of selectins.
- Author
-
Tam SH, Nakada MT, Kruszynski M, Fieles WE, Taylor AH, Mervic M, and Heavner GA
- Subjects
- Amino Acid Sequence, Cell Adhesion drug effects, E-Selectin drug effects, Humans, Inflammation drug therapy, Molecular Sequence Data, Neutrophils cytology, Neutrophils drug effects, Peptide Fragments pharmacology, Peptide Fragments therapeutic use, Structure-Activity Relationship, E-Selectin chemistry, Peptide Fragments chemistry
- Abstract
Previously, we identified several peptides corresponding to amino acid sequences within the lectin domains of selectins that inhibit neutrophil (PMN) adhesion to P-selectin. Here we focused on one of the active regions, 109-118, which contains residues that have been identified as critical for E-selectin binding to the sialyl Lexis X (sLex) counter receptor. Analogues were synthesized and examined for their inhibitory effect on PMN binding to P-selectin and E-selectin immunoglobulin fusion proteins (P-IgG, E-IgG) and also on P-IgG and E-IgG binding to sLex coated surfaces. Peptide sequences which inhibited PMN binding to the fusion proteins were not necessarily those that inhibited fusion protein binding to sLex. In addition, various amino acid substitutions could be tolerated at the 111 and 113 positions without altering inhibitory activity. Modeling suggests that structural conformations of peptide analogues could explain the differences in biological activity of peptide analogues compared to mutants of the native protein.
- Published
- 1996
- Full Text
- View/download PDF
48. Determination of the core sequence of an antagonist of selectin-dependent leukocyte adhesion and correlation of its structure with molecular modeling studies.
- Author
-
Kruszynski M, Nakada MT, Tam SH, Taylor AH, Fieles WE, and Heavner GA
- Subjects
- Amino Acid Sequence, Humans, L-Selectin chemistry, Molecular Sequence Data, P-Selectin pharmacology, Peptide Fragments pharmacology, Repetitive Sequences, Nucleic Acid, Cell Adhesion drug effects, E-Selectin chemistry, Models, Molecular, Neutrophils physiology, P-Selectin chemistry, Peptide Fragments chemistry
- Abstract
The sequence 36-50 from the lectin domain of human P-selectin has been previously identified as a weak inhibitor of selectin-dependent leukocyte adhesion. A series of C- and N-terminally truncated peptides was synthesized to determine the limits of the active core region within the parent sequence. Deletions from both the N- and C-termini gave significant increases in inhibitory activity and identified 41-50 or 36-49 as minimum active sequences, but surprisingly not the common 41-49 peptide. All peptides tested showed parallel inhibition of both P- and E-selectin-dependent adhesion. A molecular model of the lectin domain was constructed using homology modeling. Examination of this model suggests one hypothesis to explain the increase in activity on deletion of Asp36.
- Published
- 1996
- Full Text
- View/download PDF
49. The mouse/human chimeric monoclonal antibody cA2 neutralizes TNF in vitro and protects transgenic mice from cachexia and TNF lethality in vivo.
- Author
-
Siegel SA, Shealy DJ, Nakada MT, Le J, Woulfe DS, Probert L, Kollias G, Ghrayeb J, Vilcek J, and Daddona PE
- Subjects
- Animals, Blood Coagulation Factors biosynthesis, Blood Coagulation Factors genetics, Cachexia physiopathology, Cell Adhesion drug effects, Cell Adhesion Molecules biosynthesis, Cell Adhesion Molecules genetics, Cell Division drug effects, Cells, Cultured, E-Selectin, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Fibroblasts drug effects, Fibroblasts metabolism, Gene Expression Regulation drug effects, Humans, Infliximab, Intercellular Adhesion Molecule-1 biosynthesis, Intercellular Adhesion Molecule-1 genetics, Interleukin-6 metabolism, Mice, Mice, Transgenic, Neutralization Tests, Neutrophils drug effects, Recombinant Proteins pharmacology, Respiratory Burst drug effects, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha toxicity, Umbilical Veins, Antibodies, Monoclonal immunology, Cachexia prevention & control, Recombinant Fusion Proteins immunology, Thromboplastin, Tumor Necrosis Factor-alpha immunology
- Abstract
The pleiotropic cytokine tumour necrosis factor-alpha (TNF) is thought to play a central role in infectious, inflammatory and autoimmune diseases. Critical to the understanding and management of TNF-associated pathology is the development of highly specific agents capable of modifying TNF activity. We evaluated the ability of a high affinity mouse/human chimeric anti-TNF monoclonal antibody (cA2) to neutralize the in vitro and in vivo biological effects of TNF. cA2 inhibited TNF-induced mitogenesis and IL-6 secretion by human fibroblasts, TNF-priming of human neutrophils, and the stimulation of human umbilical vein endothelial cells by TNF as measured by the expression of E-selectin, ICAM-1 and procoagulant activity. cA2 also specifically blocked TNF-induced adherence of human neutrophils to an endothelial cell monolayer. Receptor binding studies suggested that neutralization resulted from cA2 blocking of TNF binding to both p55 and p75 TNF receptors on the cells. In vivo, repeated administration of cA2 to transgenic mice that constitutively express human TNF reversed the cachectic phenotype and prevented subsequent mortality. These results demonstrated that cA2 effectively neutralized a broad range of TNF biological activities both in vitro and in vivo.
- Published
- 1995
- Full Text
- View/download PDF
50. Impaired immune responsiveness is an essential component in persistent central nervous system infection with gross murine leukemia virus.
- Author
-
Korostoff JM, Nakada MT, Markman JF, and Gaulton GN
- Subjects
- Animals, Animals, Newborn, Antibodies, Viral analysis, Antibodies, Viral immunology, Antibody Formation, Antibody Specificity, Antigen-Antibody Complex analysis, Histocompatibility Antigens analysis, Immune Tolerance, Leukemia, Experimental blood, Mice, Mice, Inbred BALB C, Retroviridae immunology, AKR murine leukemia virus, Central Nervous System Diseases physiopathology, Immune System physiopathology, Leukemia, Experimental immunology
- Abstract
Exposure of newborn mice to Gross murine leukemia virus (GMuLV) results in persistent viral infection of the central nervous system (CNS) white matter. Animals exposed to virus as neonates showed a marked depression in GMuLV-specific B lymphocyte function as evidenced by significant decreases in adult and neonatal anti-GMuLV antibody levels. Immunohistochemical analyses showed that the sites of GMuLV infection in the CNS were also devoid of major histocompatibility complex (MHC) class I and II protein expression, although transplantation of GMuLV-infected brain tissue to the kidney capsules of immunocompetent mice induced a potent mononuclear cell graft infiltrate. These results indicate that persistent GMuLV infection of the CNS is linked to both impairment of anti-GMuLV peripheral immune responses and deficient antigen-presenting cell function within the CNS.
- Published
- 1991
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.