Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
2.
J Thromb Haemost ; 11(6): 1137-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528042

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is a multidomain protein that negatively regulates the coagulation cascade. TFPI inhibits the tissue factor (TF)-activated factor VII-activated FX (FXa) complex during TF-mediated coagulation initiation. The aptamer BAX 499 binds specifically to TFPI and inhibits its function, mediating a procoagulant effect in both in vitro and in vivo models of hemophilia. OBJECTIVES: This study sought to identify the regions of TFPI that are critical for BAX 499 binding, and to determine how binding mediates aptamer inhibition of TFPI. METHODS AND RESULTS: In vitro biochemical methods were used to evaluate the BAX 499 interaction with and inhibition of TFPI. Binding experiments indicated that the full-length TFPI protein is required for tight aptamer binding. Binding-competition experiments implicated the Kunitz 1, Kunitz 3 and C-terminal domains of TFPI in aptamer binding, a finding that is supported by hydrogen-deuterium exchange experiments, and indicated that aptamer and FXa can bind simultaneously to TFPI. In enzymatic assays, BAX 499 inhibited TFPI in a manner that is distinct from domain-specific antibodies, and aptamer inhibitory activity is reduced in the presence of the TFPI cofactor protein S. CONCLUSIONS: These studies demonstrate that BAX 499 binds to TFPI via multiple domains of the protein in a manner that is distinct from other TFPI inhibitors, mediating a mechanism of inhibition that does not involve direct competition with FXa. With this unique inhibitory mechanism, BAX 499 provides a useful tool for studying TFPI biology in health and disease.


Subject(s)
Aptamers, Nucleotide/chemistry , Lipoproteins/antagonists & inhibitors , Lipoproteins/chemistry , Thromboplastin/chemistry , Antibodies/chemistry , Blood Coagulation/drug effects , Coagulants/chemistry , Deuterium Exchange Measurement , Enzyme-Linked Immunosorbent Assay , Factor Xa/chemistry , Hemophilia A/drug therapy , Humans , Hydrogen/chemistry , Inhibitory Concentration 50 , Peptides/chemistry , Protein Binding , Protein S/chemistry , Protein Structure, Tertiary , Thromboplastin/antagonists & inhibitors
3.
J Thromb Haemost ; 10(8): 1581-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22632032

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is the major inhibitor of tissue factor-initiated coagulation, making it an interesting and novel therapeutic target in hemophilia treatment. The aptamer BAX499 (formerly ARC19499) is designed to improve hemostasis by specifically inhibiting TFPI. OBJECTIVES: The aim of the study was to examine the concentration-dependent augmentation of clotting by BAX499. METHODS: Whole blood clot formation was quantified by rotational thromboelastometry and thromboelastography, and thrombin generation in platelet-poor plasma was assessed with the calibrated automated thrombogram, in samples from patients with congenital hemophilia A (N=55) and B (N=11), patients with acquired hemophilia A (N=1), and healthy controls (N=37). RESULTS: BAX499 significantly improved clotting of samples from hemophilic patients in a concentration-dependent manner, resulting in clotting profiles in samples from patients with severe hemophilia that were similar to those of healthy controls. CONCLUSION: BAX499 improved ex vivo clotting parameters in blood and plasma from patients with hemophilia A and B with different severity of disease, and also in a patient with acquired hemophilia. These results further support the contention that anti TFPI strategies may be an effective treatment for hemophilic patients.


Subject(s)
Aptamers, Nucleotide/pharmacology , Blood Coagulation/drug effects , Hemophilia A/blood , Hemophilia B/blood , Hemostatics/pharmacology , Lipoproteins/antagonists & inhibitors , Signal Transduction/drug effects , Adolescent , Adult , Aged , Austria , Blood Coagulation/genetics , Case-Control Studies , Child , Child, Preschool , Dose-Response Relationship, Drug , Hemophilia A/diagnosis , Hemophilia A/genetics , Hemophilia B/diagnosis , Hemophilia B/genetics , Humans , India , Lipoproteins/blood , Lipoproteins/genetics , Middle Aged , Severity of Illness Index , Thrombelastography , Thrombin/metabolism , Whole Blood Coagulation Time , Young Adult
4.
J Thromb Haemost ; 9(9): 1825-34, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21696535

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is a major regulator of clotting initiation and a promising target for pro- and anticoagulation therapy. The aptamer BAX499 (formerly ARC19499) is a high-affinity specific TFPI antagonist designed to improve hemostasis. However, it is not clear how stimulation of coagulation onset by inactivating TFPI will affect spatial and temporal clot propagation. OBJECTIVE: To examine the BAX499 effect on clotting in a spatial, reaction-diffusion experimental system in comparison with that of recombinant activated factor VII (rVIIa). METHODS: Clotting in plasma activated by immobilized tissue factor (TF) was monitored by videomicroscopy. RESULTS: BAX499 dose-dependently improved coagulation in normal and hemophilia A plasma activated with TF at 2 pmole m(-2) by shortening lag time and increasing clot size by up to ~2-fold. The effect was TFPI specific as confirmed by experiments in TFPI-depleted plasma with or without TFPI supplementation. Clotting improvement was half-maximal at 0.7 nm of BAX499 and reached a plateau at 10 nm, remaining there at concentrations up to 1000 nm. The BAX499 effect decreased with TF surface density increase. RVIIa improved clotting in hemophilia A plasma activated with TF at 2 or 20 pmole m(-2) , both by shortening lag time and increasing spatial velocity of clot propagation; its effects were strongly concentration dependent. CONCLUSIONS: BAX499 significantly improves spatial coagulation by inhibiting TFPI in a spatially localized manner that is different to that observed with rVIIa.


Subject(s)
Aptamers, Nucleotide/pharmacology , Blood Coagulation/drug effects , Fibrin/biosynthesis , Lipoproteins/antagonists & inhibitors , Aptamers, Nucleotide/administration & dosage , Blood Coagulation/physiology , Computer Simulation , Factor VIIa/administration & dosage , Factor VIIa/pharmacology , Hemophilia A/blood , Hemophilia A/drug therapy , Hemostasis/drug effects , Hemostasis/physiology , Humans , In Vitro Techniques , Lipoproteins/deficiency , Lipoproteins/physiology , Male , Microscopy, Video , Models, Biological , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
5.
Thromb Haemost ; 87(5): 824-30, 2002 May.
Article in English | MEDLINE | ID: mdl-12038784

ABSTRACT

Hemophilia B therapy requires intravenous (IV) infusions of large volumes of factor IX due to the low concentration of factor IX in concentrates (approximately 100 IU/mL). High concentration recombinant factor IX (rFIX) could be a significant advance since it would reduce the large volumes necessary for IV dosing and allow for low-volume subcutaneous (SC) administration. To evaluate high concentration factor IX, we produced formulations with either 2,000 or 4,000 IU/mL and studied the SC bioavailability in beagle dogs, cynomolgus monkeys and hemophilia B dogs along with efficacy in hemophilia B dogs. Beagle dog SC bioavailability was 86.4% using a 2000 IU/mL formulation and 77.0% using a 4000 IU/mL formulation. Monkey bioavailability of a 4000 IU/mL formulation of rFIX was 34.8%. A single SC administration of 200 IU/kg (4000 IU/mL) of rFIX to hemophilia B dogs, produced factor IX clotting activity above 5% for 5 days with a bioavailability of 48.6%. High concentration SC rFIX has an acceptable pharmacokinetic profile in monkeys and dogs, and produces a sustained FIX activity in hemophilic dogs.


Subject(s)
Factor IX/pharmacokinetics , Animals , Biological Availability , Disease Models, Animal , Dog Diseases/drug therapy , Dogs , Enzyme-Linked Immunosorbent Assay , Factor IX/administration & dosage , Factor IX/therapeutic use , Hemophilia B/drug therapy , Hemophilia B/veterinary , Injections, Subcutaneous , Macaca fascicularis , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Species Specificity
6.
Thromb Haemost ; 85(3): 445-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11307812

ABSTRACT

The purpose of this paper was to establish proof of concept for administration of human recombinant F.IX (rF.IX) by inhalation for therapy of hemophilia B. The pharmacokinetics of intratracheal (IT) administration of rF.IX was studied in nine hemophilia B dogs randomized into 3 groups that received 200 IU/kg IT, 1,000 IU/kg IT, or 200 IU/kg intravenously (IV). IT rF.IX produced therapeutic levels of F.IX antigen and activity and the pharmacokinetic parameters were consistent with a slow release from a depot site within the lungs. Bioavailability compared to IV administration was 11% for 200 IU/kg IT and 4.9% for 1,000 IU/kg. The whole blood clotting time began to shorten at 2 h but F.IX bioactivity was not detected until 8 h post infusion in both IT groups. In all groups, F.IX activity was detected through 72 h post administration. These data demonstrate that biologically active rF.IX can reach the systemic circulation when given IT. Aerosolization of rF.IX may provide a needle-free therapeutic option for delivery of rF.IX to hemophilia B patients.


Subject(s)
Dog Diseases/drug therapy , Factor IX/administration & dosage , Factor IX/pharmacokinetics , Hemophilia B/veterinary , Administration, Inhalation , Animals , Antibodies, Heterophile/blood , Biological Availability , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Factor IX/immunology , Hemophilia B/drug therapy , Humans , Injections, Intravenous , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Therapeutic Equivalency
7.
Shock ; 15(4): 285-90, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11303727

ABSTRACT

P-selectin is a major component in the early interaction between platelets, endothelial cells, and inflammatory cells in the initial phases of the innate immune response. The major ligand for P-selectin is P-selectin glycoprotein ligand-1 (PSGL-1) and this ligand is expressed on the surface of monocyte, lymphocyte, and neutrophil membranes. A truncated form of recombinant human P-selectin glycoprotein ligand-1 has been covalently linked to immunoglobulin G (rPSGL-Ig) and this fusion peptide functions as a competitive inhibitor of PSGL-1. As an inhibitor of neutrophil-endothelial cell adherence, rPSGL-Ig is in early clinical development for the treatment of ischemia reperfusion injury. To determine the potential for deleterious effects from inhibition in P-selectin-mediated neutrophil attachment in the presence of bacterial infection, the effects of therapeutic doses of rPSGL-Ig were tested in three standard laboratory sepsis models. The experimental models included: the murine systemic Listeria monocytogenes infection model, the Pseudomonas aeruginosa bacteremia model in neutropenic rats, and the cecal ligation and puncture (CLP)-induced peritonitis model in rats. Recombinant human PSGL-Ig had no adverse effects on mortality or immune clearance in systemic bacterial infection in any of the three infection models. The PSGL-1 inhibitor did significantly decrease local neutrophil infiltration and bacterial clearance in the peritoneum following CLP, but this did not increase the systemic levels of proinflammatory cytokines, the quantitative levels of bacteremia, or the overall mortality rate following CLP. The results indicate that rPSGL-Ig did not exacerbate infection in these experimental sepsis models.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bacterial Infections/drug therapy , Chemotaxis, Leukocyte/drug effects , Immunoconjugates/therapeutic use , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Bacteremia/blood , Bacteremia/drug therapy , Bacteremia/immunology , Bacteremia/microbiology , Bacterial Infections/immunology , Cecum/injuries , Cell Adhesion/drug effects , Cytokines/blood , Depression, Chemical , Drug Evaluation, Preclinical , Female , Genes, Immunoglobulin , Humans , Immunoconjugates/pharmacology , Immunoconjugates/toxicity , Immunoglobulin Fc Fragments , Immunoglobulin G/genetics , Intestinal Perforation/complications , Listeriosis/drug therapy , Listeriosis/immunology , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/toxicity , Mice , Mice, Inbred C57BL , Neutropenia/chemically induced , Neutropenia/complications , Neutrophils/drug effects , Neutrophils/physiology , P-Selectin/physiology , Peritonitis/drug therapy , Peritonitis/immunology , Pseudomonas Infections/blood , Pseudomonas Infections/drug therapy , Pseudomonas Infections/immunology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/toxicity , Safety , Specific Pathogen-Free Organisms
8.
Microcirculation ; 8(1): 15-24, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11296849

ABSTRACT

OBJECTIVE: Leukocyte rolling is recognized as an important event in facilitating the extravasation of leukocytes from the vascular to the interstitial compartment, and is mediated by the selectin family of cell adhesion molecules. The aim of this study was to evaluate and characterize the rolling behavior of leukocytes in a model of acute inflammation using a novel soluble selectin ligand directed against P-selectin. METHODS: Feline mesenteric postcapillary venules were visualized using intravital microscopy prior to and following exposure to leukotriene C4 (LTC4) in animals pretreated with vehicle (saline) and the P-selectin antagonist rPSGL-Ig. RESULTS: A concentration of 500 pM LTC4 induced a threefold and sixfold elevation in leukocyte rolling flux and adhesion, respectively, compared to baseline values (p < 0.05). Administration of rPSGL-Ig had no effect on LTC4-induced leukocyte rolling flux but significantly attenuated the increase in the fraction of rolling leukocytes (p < 0.05). In addition, rPSGL-Ig inhibited the LTC4-induced reductions in leukocyte rolling velocity (p < 0.001). Finally, LTC4-induced leukocyte adhesion in animals pretreated with rPSGL-Ig was reduced by 60%, compared to vehicle-treated animals (p < 0.05). CONCLUSIONS: LTC4 induces leukocyte rolling and adhesion in feline mesenteric venules in a dose-dependent manner. Administration of rPSGL-Ig inhibits LTC4-induced reductions in leukocyte rolling velocity and attenuates the elevation in the fraction of rolling leukocytes produced by LTC4 stimulation. This suggests that rPSGL-Ig may be used to reduce leukocyte rolling and adhesion, and subsequently attenuate tissue injury during inflammation.


Subject(s)
Inflammation/drug therapy , Leukocytes/drug effects , Leukocytes/physiology , P-Selectin/drug effects , P-Selectin/physiology , Venules/drug effects , Amino Acid Sequence , Animals , Cats , Cell Adhesion/drug effects , Cell Movement/drug effects , Disease Models, Animal , Humans , Inflammation/pathology , Inflammation/physiopathology , Leukotriene C4/pharmacology , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Mesenteric Veins/drug effects , Mesenteric Veins/pathology , Mesenteric Veins/physiopathology , Molecular Sequence Data , Phlebitis/drug therapy , Phlebitis/pathology , Phlebitis/physiopathology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Solubility , Venules/pathology , Venules/physiopathology
9.
Circulation ; 103(8): 1128-34, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11222477

ABSTRACT

BACKGROUND: P-selectin mediates leukocyte recruitment to activated platelets and endothelium through its high-affinity receptor P-selectin glycoprotein ligand-1 (PSGL-1). Platelet and leukocyte activation and binding have been reported after coronary angioplasty and were correlated with restenosis. We investigated the effect of a recombinant soluble PSGL-1 (rPSGL-Ig) on the adhesion of platelets and neutrophils and the development of restenosis after double arterial injury. METHODS AND RESULTS: Four weeks after angioplasty of both carotid arteries in pigs, a second angioplasty was performed at the same sites, 15 minutes after a single administration of vehicle or rPSGL-1 (1 mg/kg IV). Animals were euthanized 1 hour, 4 hours, 1 week, or 4 weeks later. Adhesion of autologous (51)Cr-platelets and (111)In-neutrophils was quantified and histological/morphometric analyses were performed. Although rPSGL-Ig did not affect adherence of these cells 1 hour after injury, it significantly reduced the adhesion of platelets (50% at 4 hours and 85% at 1 week) and neutrophils (50% at 4 hours and 78% at 1 week) to deeply injured arteries. At 4 weeks, the residual lumen was 63% larger in rPSGL-Ig-treated arteries as compared with control arteries (6.1+/-0.6 versus 3.8+/-0.1 mm(2); P:<0.002). The neointimal area was slightly reduced (0.5 in rPSGL-Ig versus 0.7 mm(2) in control). The ratio of the external elastic lamina of injured to uninjured reference segments was >1 in treated arteries and <1 in control arteries. CONCLUSIONS: P-selectin antagonism with rPSGL-Ig inhibits early platelet/leukocyte adhesion on injured arteries and reduces restenosis through a positive impact on vascular remodeling. Hence, rPSGL-Ig may have potential in the prevention of restenosis.


Subject(s)
Angioplasty , Constriction, Pathologic/prevention & control , Membrane Glycoproteins/therapeutic use , Animals , Blood Platelets/drug effects , Blood Platelets/physiology , Cell Adhesion/drug effects , Cell Communication/drug effects , Constriction, Pathologic/pathology , Disease Models, Animal , Membrane Glycoproteins/genetics , Neutrophils/drug effects , Neutrophils/physiology , Recombinant Proteins/therapeutic use , Recurrence , Solubility , Swine
10.
Blood ; 97(2): 465-72, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11154224

ABSTRACT

Interleukin (IL)-11 is a cytokine with thrombopoietic activity that has been shown to increase plasma von Willebrand factor (vWf) in preliminary clinical studies. This led to further evaluation of the effect of recombinant human (rh)IL-11 on vWf and factor VIII (FVIII) secretion. In vitro, rhIL-11 did not increase vWf production by cultured endothelial cells, which suggests an indirect mechanism. Also, in vivo, plasma vWf was not elevated in mice shortly after a single intravenous (IV) bolus injection of 250 or 1000 microg/kg rhIL-11. The effect of continuous exposure to rhIL-11 was accessed by treating wild type mice for 7 consecutive days with subcutaneous 250 microg/kg/d rhIL-11. Platelet counts increased by 25% and 40% after 4 and 7 days, respectively. Plasma vWf and FVIII levels increased 2-fold after 4 and 7 days. Surprisingly, no effect of rhIL-11 on vWf or FVIII messenger RNA was observed, which suggests that the regulation by rhIL-11 occurs after transcription. No increase in soluble P-selectin was observed after rhIL-11 treatment, indicating that platelet activation is not the source of elevated vWf. Similarly to wild type mice, vWf heterozygous mice responded to rhIL-11 treatment by a significant increase in platelet counts and vWf and FVIII levels. Importantly, in vWf-deficient mice, rhIL-11 also induced a significant increase in FVIII independent of vWf and was able to reduce skin bleeding time. These results suggest that a clinical evaluation of the effects of rhIL-11-induced vWf/FVIII elevation in maintaining hemostasis in mild hemophilia A or von Willebrand disease would be worthwhile.


Subject(s)
Factor VIII/drug effects , Interleukin-11/pharmacology , von Willebrand Diseases/blood , von Willebrand Factor/drug effects , Animals , Bleeding Time , Cell Culture Techniques , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Factor VIII/metabolism , Female , Heterozygote , Humans , Interleukin-11/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , P-Selectin/blood , P-Selectin/drug effects , Platelet Count , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Time Factors , Umbilical Cord/cytology , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
11.
Arterioscler Thromb Vasc Biol ; 20(11): 2483-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073856

ABSTRACT

Leukocyte adhesion, transendothelial migration, and stasis are important components in the pathogenesis of deep vein thrombosis. Anesthetized cats were treated with saline, a recombinant soluble form of P-selectin glycoprotein ligand-1 (rPSGL-Ig), or an E- and L-selectin antibody (EL-246) before exposure and occlusion of a jugular vein. After 2 or 6 hours of occlusion, jugular veins were perfused with buffer, fixed, and prepared for scanning electron microscopy. In cats receiving saline, 2 and 6 hours of occlusion produced moderate levels of leukocyte and platelet adhesion and endothelial cell injury. Treatment of cats with rPSGL-Ig or EL-246 had no apparent effect on the magnitude of cell adhesion and endothelial cell injury compared with no treatment. After 6 hours of occlusion, the presence of a mural thrombus in untreated veins was observed and confirmed by scanning electron microscopy. Pretreatment of cats with rPSGL-Ig completely (4.0 mg/kg) or partially (1.0 mg/kg) prevented the occurrence of thrombi in the jugular veins. The reduction in thrombosis by rPSGL-Ig treatment after 6 hours of venous stasis, in the absence of any effect on leukocyte-mediated endothelial cell injury, suggests an antithrombotic mechanism of action for this protein.


Subject(s)
P-Selectin/physiology , Venous Thrombosis/prevention & control , Venous Thrombosis/physiopathology , Animals , Cats , Cell Adhesion , Cell Movement , Humans , Immunoglobulin G/pharmacology , Injections, Intravenous , Leukocyte Count , Ligands , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/pharmacology , Mice , Microscopy, Electron, Scanning , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology , Venous Thrombosis/pathology
12.
Blood ; 94(12): 4263-73, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10590071

ABSTRACT

Interleukin-12 (IL-12) is a heterodimeric cytokine mediating a dynamic interplay between T cells and antigen-presenting cells (APCs). Preclinical studies have demonstrated that recombinant murine IL-12 (rmIL-12) promotes specific antitumor immunity mediated by T cells in several types of tumors. However, the in vivo antitumor properties of IL-12 in acute myeloid leukemia (AML) have not been previously reported. We show here in a murine AML model that systemic administration of rmIL-12 significantly delays tumor growth but is incapable of rescuing mice from lethal leukemia. In contrast, AML cells genetically modified to express IL-12 (IL12-AML) using murine stem cell virus (MSCV) p40 + p35 elicit very potent antileukemic activity. Vaccines with lethally irradiated IL12-AML cells protect naive mice against challenge with wild-type AML cells and, more importantly, can cure mice bearing a considerable leukemic burden. Immunized mice show no signs of systemic IL-12 toxicity and their spleen histology is comparable with naive mice spleen. In vivo depletion of IL-12, interferon-gamma (IFN-gamma), or CD8(+) T cells after injections with live IL12-AML cells abrogates completely the antileukemia immune responses. Studies on the in vitro effects of IFN-gamma on AML cells demonstrate enhanced expression of major histocompatibility complex (MHC) and accessory molecules and induction of the costimulatory molecules B7.1 and B7.2, but no significant direct antiproliferative effect. (51)Cr release assays show that rejection of live IL12-AML cells supports the development of long-lasting leukemia-specific cytotoxic T lymphocyte (CTL) activity. In conclusion, our results demonstrate that IL12-AML vaccination is a safe and potent immunotherapeutic approach that has a great potential to eliminate minimal residual disease in patients with AML.


Subject(s)
Cancer Vaccines , Cytotoxicity, Immunologic , Interleukin-12/immunology , Leukemia, Experimental/immunology , Leukemia, Myeloid/immunology , Acute Disease , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cross Reactions , Epitopes/immunology , Gene Transfer Techniques , Hematopoietic Stem Cells/immunology , Interleukin-12/administration & dosage , Interleukin-12/genetics , Leukemia, Experimental/prevention & control , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology
13.
Circulation ; 99(10): 1363-9, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077522

ABSTRACT

BACKGROUND: We investigated whether administration of a soluble recombinant P-selectin glycoprotein ligand-1 chimera (rPSGL-Ig) in conjunction with thrombolytic therapy would enhance thrombolysis by preventing ongoing interactions of leukocytes with platelets and the injured arterial wall. METHODS AND RESULTS: An occlusive thrombus was formed in an internal iliac artery of Yorkshire pigs by placement of a copper coil in the artery under fluoroscopic guidance. Pigs then received heparin and, 15 minutes later, either vehicle or rPSGL-Ig followed by infusion with 25 mg tissue plasminogen activator according to the 90-minute regimen. Blood flow through the artery was monitored by angiography and scored on a scale of 0 to 3. Lysis of the thrombus was accelerated by 70% in pigs treated with rPSGL-Ig 250 microg/kg compared with control (13.3+/-5.0 versus 44. 4+/-13.3 minutes; n=9 each). Eight of 9 control pigs reoccluded in 13.8+/-16.9 minutes after the end of tissue plasminogen activator infusion, whereas no reocclusion was observed in 8 of 9 pigs in the rPSGL-Ig group. When the dose of rPSGL-Ig was increased to 500 microg/kg, time to lysis was shortened by 61% from control (18.0+/-8. 4 versus 46.0+/-8.9 minutes). Reocclusion occurred in 6.0+/-15.2 minutes in control but not in any rPSGL-Ig-treated pig (n=5 each). In addition, near-normal flow (score 2 or 3) after thrombolysis was achieved 59% and 58% faster in the 2 rPSGL-Ig groups than in their respective controls. CONCLUSIONS: Inhibition of leukocyte accumulation at the site of thrombosis with rPSGL-Ig may represent a safe therapeutic intervention that could be important in accelerating thrombolysis, achieving optimal reperfusion, and reducing incidence of acute reocclusion.


Subject(s)
Arterial Occlusive Diseases/drug therapy , Fibrinolytic Agents/therapeutic use , Iliac Artery , Immunoconjugates/therapeutic use , Membrane Glycoproteins/therapeutic use , P-Selectin/physiology , Thrombolytic Therapy , Thrombosis/drug therapy , Animals , Arterial Occlusive Diseases/prevention & control , Blood Proteins/analysis , Cell Adhesion/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Female , Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Immunoconjugates/pharmacology , Male , Membrane Glycoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Recurrence , Safety , Solubility , Swine , Thrombosis/prevention & control , Tissue Plasminogen Activator/therapeutic use
14.
Radiat Oncol Investig ; 6(2): 71-80, 1998.
Article in English | MEDLINE | ID: mdl-9572683

ABSTRACT

Interleukin-12 (IL-12), a naturally occurring cytokine, has demonstrated antitumor activity in several murine solid tumors. The Lewis lung carcinoma was used to study the most effective scheduling of recombinant murine interleukin-12 (rmIL-12) administration with fractionated radiation therapy. The effect of the schedule of rmIL-12 administration alone or along with a 1- or 2-week fractionated radiation therapy regimen was examined. Beginning rmIL-12 prior to or at the same time as radiation therapy and extending rmIL-12 through the radiation regimen and beyond produced the longest tumor growth delays. Those treatment regimens which were most effective against the primary tumor were also most effective in decreasing the number of lung metastases on day 20. To further assess the immunotherapeutic effects from rmIL-12 administration, the efficacy of rmIL-12 with fractionated radiation therapy delivered to a right hind-limb tumor was measured as tumor growth delay in an unirradiated left hind-limb tumor. There was some difference in the tumor growth delay between the unirradiated tumor in the animals bearing an irradiated tumor in the contralateral leg, and the tumors in animals receiving rmIL-12 only. Recombinant murine granulocyte-macrophage-colony stimulating factor (rmGM-CSF) was also an antitumor agent active against the Lewis lung carcinoma and produced an additive effect in combination with fractionated radiation therapy in this tumor. rmIL-12 was a radiation sensitizer in the Lewis lung carcinoma. When rmIL-12 (45-microg/kg) and rmGM-CSF (45 microg/kg) were administered together with fractionated radiation therapy, a marked increase in tumor growth delay resulted. This treatment combination also nearly ablated lung metastases on day 20 in these animals. These results may serve as a useful guide in developing clinical protocols, including rmIL-12 and fractionated radiation therapy.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/radiotherapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Interleukin-12/administration & dosage , Animals , Combined Modality Therapy , Drug Administration Schedule , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Radiation Dosage , Time Factors
15.
Mol Reprod Dev ; 46(1): 92-5, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8981369

ABSTRACT

Infiltration of monocytes into arteries is an early event in the pathogenesis of atherosclerosis. This recruitment is interpreted as enhancing lesion development, but it could also be a host response limiting lipid accumulation. The ability of macrophages to limit cholesterol uptake, however, can be reduced by the impaired mobility and metabolic activity associated with foam cell development. As lesions enlarge, foam cells die and become the nidus for the necrotic core. Treatments to improve viability might improve foam cell function and promote regression. Macrophage colony-stimulating factor (M-CSF) is vital to monocyte/macrophage differentiation, proliferation, and activation. We found that foam cells of Watanabe heritable hyperlipidemic (WHHL) rabbits had faint staining for M-CSF. Treatment of rabbits with recombinant human M-CSF (rhM-CSF) increased M-CSF staining, which correlated with reduced cholesterol content of these foam cells.


Subject(s)
Arteriosclerosis/metabolism , Hypercholesterolemia/genetics , Macrophage Colony-Stimulating Factor/biosynthesis , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Arteriosclerosis/etiology , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Cell Movement , Cholesterol/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Foam Cells/metabolism , Foam Cells/pathology , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/therapeutic use , Male , Rabbits , Recombinant Proteins/therapeutic use
16.
Crit Rev Immunol ; 17(5-6): 545-53, 1997.
Article in English | MEDLINE | ID: mdl-9419442

ABSTRACT

Interleukin 12 (IL-12), a novel heterodimeric protein produced primarily by antigen-presenting cells, serves as a key regulator of innate and adaptive immune responses. In addition to being a potent inducer of IFN-gamma, IL-12 is widely considered to be the principal cytokine that regulates the generation of Th1 type effector cells. As the successful induction of experimental autoimmune encephalomyelitis (EAE) is associated with a strong Th1 type cellular response, we have evaluated the role of IL-12 in regulating the pathogenesis of EAE in SJL/J mice and Lewis rats. In both settings, treatment with IL-12 was found to accelerate the onset and increase the severity and duration of clinical disease. More importantly, administration of IL-12 to Lewis rats that had recovered from primary disease was found to trigger clinical relapse. In all instances, IL-12-induced exacerbation was associated with a profound increase in iNOS positive macrophages within the perivascular lesions. Although IL-12-induced IFN-gamma does not appear to be required for exacerbation of disease, neutralizing antibodies against murine IL-12 delay the onset and reduce the severity of adoptively transferred EAE, indicating a role for endogenous IL-12 as regulator of disease. Based on the above findings, effective inhibition of IL-12 in vivo may have great therapeutic value in the treatment of MS and other Th1-associated inflammatory disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Interleukin-12/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Humans , Interleukin-12/pharmacology , Lymphocyte Activation , Mice , Multiple Sclerosis/prevention & control , Rats , Rats, Inbred Lew , T-Lymphocytes/cytology
17.
Clin Cancer Res ; 3(9): 1661-7, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9815857

ABSTRACT

The antitumor activity of interleukin (IL)-12, a naturally occurring cytokine, has been demonstrated in several murine solid tumors. Animals bearing established B16 melanoma or MB-49 bladder carcinoma were used to study the most effective scheduling of recombinant murine IL-12 (rmIL-12), along with systemic chemotherapy. rmIL-12 (0. 45, 4.5, or 45 microgram/kg) was more effective as a single agent when administered to mice bearing the MB-49 bladder carcinoma at the highest dose for 11 doses rather than for 5 doses. In combination with chemotherapy (Adriamycin, cyclophosphamide, or 5-fluorouracil), rmIL-12 administration did not increase the toxicity of the chemotherapy, and there was increased antitumor activity with each rmIL-12-drug combination. Administering rmIL-12 (45 microgram/kg) on days 4-14, along with Adriamycin, cyclophosphamide, or 5-fluorouracil on days 7-11, resulted in 2.2-2.7-fold increases in tumor growth delay, compared with the chemotherapy alone against the primary tumor, and a marked decrease in the number of lung metastases on day 20. Because the B16 melanoma grows more slowly than the MB-49 bladder carcinoma, allowing multiple courses of chemotherapy, cyclophosphamide could be administered. The rmIL-12 (45 microgram/kg)-cyclophosphamide combination regimen that was most effective overlapped 2 days with the terminal portion of the chemotherapy treatment. There was a parallel increase in the response of the primary tumor and metastatic disease to the lungs. Administration of rmIL-12 to animals bearing the MB-49 bladder carcinoma or the B16 melanoma was compatible with coadministration of chemotherapy at full dose without additional toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Transitional Cell/therapy , Immunologic Factors/administration & dosage , Interleukin-12/administration & dosage , Melanoma, Experimental/therapy , Urinary Bladder Neoplasms/therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/secondary , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/toxicity , Doxorubicin/administration & dosage , Doxorubicin/toxicity , Drug Administration Schedule , Fluorouracil/administration & dosage , Fluorouracil/toxicity , Immunologic Factors/therapeutic use , Interleukin-12/therapeutic use , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/immunology
18.
Eur J Immunol ; 27(12): 3438-46, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9464833

ABSTRACT

Administration of recombinant murine interleukin (rmIL)-12 to MB49.1 tumor-bearing mice results in dose-dependent regression of the primary tumor and the generation of protective antitumor immunity in the majority of animals. rmIL-12 administration is associated with a marked increase in lymph node cellularity that is predominantly due to the expansion of B220+ B cells as well as CD8+ T cells. Stimulation of lymph node cells from rmIL-12-treated, but not control tumor-bearing mice, with MB49.1 tumor cells in vitro was shown to enhance the secretion of interferon (IFN)-gamma. The magnitude of this in vitro response was dependent on the dose of rmIL-12 administered in vivo and mirrored the change in circulating serum IFN-gamma. Furthermore, at the height of the in vitro response to tumor stimulation, the addition of a neutralizing antibody to murine IL-12 suppressed IFN-gamma production, indicating a role for endogenous IL-12 in this antigen-specific cytokine response. Although studies in SCID mice confirmed that an appropriate T cell response was required for rmIL-12-mediated antitumor activity, in immunocompetent animals early tumor regression was not accompanied by cellular infiltration of the tumor. In contrast, a profound increase in tumor-associated inducible nitric oxide synthase (iNOS) was observed in mice receiving rmIL-12 which preceded T cell infiltration of the tumor which could be detected during the second week of IL-12 treatment. Direct tumor killing through the cytotoxic actions of NO via the iNOS pathway may serve as a way of generating tumor antigen which enables the host to mount a subsequent T cell response against the tumor.


Subject(s)
Immunity, Cellular , Interleukin-12/immunology , Neoplasms, Experimental/immunology , Animals , Interleukin-12/administration & dosage , Mice , Mice, Inbred C57BL , Mice, SCID , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology
19.
Blood ; 88(7): 2603-10, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8839853

ABSTRACT

Recombinant human factor IX (rFIX) has been expressed in transduced cultured cell systems since 1985. Because there has been limited in vivo testing of rFIX in hemophilia B subjects, this study was undertaken using the severe hemophilia B canines of the Chapel Hill strain. Three groups of hemophilic dogs received either 50, 100, or 200 IU/kg of rFIX. As a control, a fourth group of hemophilic dogs received 50 IU/kg of a high purity, plasma-derived human FIX (pdFIX). The coagulant and hemostatic effects of rFIX and pdFIX were similar with all comparative dosing regimens. Based on activity data, the elimination half-life of rFIX was 18.9 +/- 2.3 hours and pdFIX was 17.9 +/- 2.1 hours. A prophylactic regimen administering rFIX daily resulted in a continuous therapeutic level of plasma FIX and was accompanied by a two-fold increase in recovery levels by day 5, compared to that observed with administration of a single bolus. The mechanisms of the high to complete recovery of FIX with the prophylactic regimen could depend not only on the degree of saturation of the vascular endothelial binding sites but also on the altered dynamics of the balance of FIX distribution between the intravascular and extravascular compartments. The pharmacokinetic (PK) parameters for rFIX and pdFIX were similar. However, the relative PK values for V1 and V5s of both products on day 5 differed greatly from day 1 and may reflect the changing equilibrium of FIX between compartments with elevated levels of plasma FIX. Neutralizing antihuman FIX antibodies resulting from human FIX antigen being administered to FIX deficient dogs were observed beginning at 14 days. The antigenicity of rFIX and pdFIX appeared to be comparable. Despite the very different procedures used for production of rFIX and pdFIX products, in vivo testing in hemophilia B dogs showed the functional behavior of these products is similar; they are highly effective for replacement therapy and for prophylaxis.


Subject(s)
Dog Diseases/therapy , Factor IX/therapeutic use , Hemophilia B/veterinary , Recombinant Fusion Proteins/therapeutic use , Animals , Antibody Formation , Dog Diseases/blood , Dog Diseases/genetics , Dogs/blood , Dogs/immunology , Drug Evaluation/veterinary , Endothelium, Vascular/metabolism , Factor IX/immunology , Factor IX/pharmacokinetics , Hemophilia B/blood , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Immunization , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Species Specificity
20.
Stem Cells ; 14(5): 517-32, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8888493

ABSTRACT

We have extensively characterized the hematological response of normal and myelosuppressed nonhuman primates to treatment with recombinant human interleukin 11 (rHuIL-11) in vivo. In normal cynomolgus monkeys, rHuIL-11 significantly increased peripheral platelet counts when administered at doses of 10 micrograms/kg/day to 100 micrograms/kg/day either by constant i.v. infusion or s.c. injection. As few as four days of rHuIL-11 treatment were sufficient to increase peripheral platelet counts significantly. In addition, extending the treatment period enhanced both the magnitude and the duration of the response. Bone marrow megakaryocytes from animals treated with 100 micrograms/kg/day of rHuIL-11 were increased in size compared to controls and were ultrastructurally normal. A nonhuman primate myelosuppression model using carboplatin, which causes severe thrombocytopenia with platelet counts of < or = 20 x 10(3) platelets/microliters, was developed. This novel model was used to evaluate the effectiveness of rHuIL-11 in platelet restoration. rHuIL-11, administered s.c. at a dose of 125 micrograms/kg/day either concurrently or following chemotherapy, prevented severe thrombocytopenia in addition to accelerating platelet recovery compared to control animals receiving no rHuIL-11. These data demonstrate that rHuIL-11 has potent in vivo thrombopoietic effects when administered to normal and myelosuppressed nonhuman primates, and that rHuIL-11 can be an important therapy to reduce the severity and duration of thrombocytopenia following chemotherapy.


Subject(s)
Blood Platelets/metabolism , Hematopoietic Stem Cells/metabolism , Immunosuppression Therapy , Interleukin-11/pharmacology , Animals , Blood Platelets/cytology , Blood Proteins/drug effects , Cell Differentiation/drug effects , Cell Size/drug effects , Dose-Response Relationship, Drug , Female , Hematopoiesis/drug effects , Humans , Leukocyte Count/drug effects , Macaca fascicularis , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Platelet Count/drug effects , Recombinant Proteins/pharmacology , Reticulocytes/cytology , Reticulocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...