Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancer Chemother Pharmacol ; 75(3): 579-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25577133

ABSTRACT

PURPOSE: To define the dose-limiting toxicity (DLT), maximum tolerated dose (MTD) and pharmacokinetics (PK) of PEP02, a novel liposome-encapsulated irinotecan, in patients with advanced refractory solid tumors. METHODS: Patients were enrolled in cohorts of one to three to receive escalating dose of PEP02 in a phase I trial. PEP02, from 60 to 180 mg/m(2), was given as a 90-min intravenous infusion, every 3 weeks. RESULTS: A total of 11 patients were enrolled into three dose levels: 60 (one patient), 120 (six patients) and 180 mg/m(2) (four patients). DLT was observed in three patients, one at 120 mg/m(2) (grade 3 catheter-related infection) and two at 180 mg/m(2) (grade 4 neutropenia lasting for >3 days in one, grade 4 hematological toxicities and grade 4 diarrhea in the other). MTD was determined as 120 mg/m(2). Comparing with those after free-form irinotecan in the literature, the dose-normalized PK of SN-38 (the active metabolite) after PEP02 was characterized by lower C max, prolonged terminal half-life and higher AUC but with significant inter-individual variation. One patient who died of treatment-related toxicity had significantly higher C max and AUC levels of SN-38 than those of the other three patients at 180 mg/m(2). Post hoc pharmacogenetic study showed that the patient had a combined heterozygosity genotype of UGT1A1*6/*28. Two patients had objective tumor response. CONCLUSIONS: PEP02 apparently modified the PK parameters of irinotecan and SN-38 by liposome encapsulation. The MTD of PEP02 monotherapy at 3-week interval is 120 mg/m(2), which will be the recommended dose for future studies.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/analogs & derivatives , Glucuronosyltransferase/genetics , Nanoparticles , Neoplasms/drug therapy , Adult , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Dose-Response Relationship, Drug , Female , Genotype , Half-Life , Humans , Infusions, Intravenous , Irinotecan , Liposomes , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Pharmacogenetics , Treatment Outcome
2.
Br J Cancer ; 109(4): 920-5, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23880820

ABSTRACT

BACKGROUND: PEP02, also known as MM-398, is a novel nanoliposomal irinotecan that has improved pharmacokinetics and tumour bio-distribution of the free drug. This phase 2 study evaluated PEP02 monotherapy as second-line treatment for pancreatic cancer. METHODS: Patients who had metastatic pancreatic adenocarcinoma, Karnofsky performance status ≥70, and had progressed following gemcitabine-based therapy were eligible. Intravenous injection of PEP02 120 mg m(-2) was given every 3 weeks. Simon 2-stage design was used. The primary objective was 3-month survival rate (OS(3-month)). RESULTS: A total of 40 patients were enrolled. The most common severe adverse events included neutropenia, abdominal pain, asthenia, and diarrhoea. Three patients (7.5%) achieved an objective response, with an additional 17 (42.5%) demonstrating stable disease for a minimum of two cycles. Ten (31.3%) of 32 patients with an elevated baseline CA19-9 had a >50% biomarker decline. The study met its primary end point with an OS(3-month) of 75%, with median progression-free survival and overall survival of 2.4 and 5.2 months, respectively. CONCLUSION: PEP02 demonstrates moderate antitumour activity with a manageable side effect profile for metastatic, gemcitabine-refractory pancreatic cancer patients. Given the limited treatment options available to this patient population, a phase 3 trial of PEP02 (MM-398), referred to as NAPOLI-1, is currently underway.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Sucrose/analogs & derivatives , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Camptothecin/therapeutic use , Disease-Free Survival , Drug Combinations , Female , Humans , Irinotecan , Liposomes , Male , Middle Aged , Pancreatic Neoplasms/pathology , Salvage Therapy , Sucrose/therapeutic use , Treatment Outcome
3.
Ann Oncol ; 24(6): 1567-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23406728

ABSTRACT

BACKGROUND: PEP02 is a novel highly stable liposomal nanocarrier formulation of irinotecan. This randomized phase II study evaluated the efficacy and safety of single agent PEP02 compared with irinotecan or docetaxel in the second-line treatment of advanced oesophago-gastric (OG) cancer. PATIENTS AND METHODS: Patients with locally advanced/metastatic disease who had failed one prior chemotherapy regimen were randomly assigned to PEP02 120 mg/m(2), irinotecan 300 mg/m(2) or docetaxel (Taxotere) 75 mg/m(2) every 3 weeks. The primary end point was objective response rate (ORR). Simon's two-stage design was used and the ORR of interest was 20% (α = 0.05, type II error ß = 0.10, null hypothesis of ORR was 5%). RESULTS: Forty-four patients per arm received treatment, and 124 were assessable for response. The ORR statistical threshold for the first stage was reached in all arms. In the intent-to-treat (ITT) population, ORRs were 13.6% (6/44), 6.8% (3/44) and 15.9% (7/44) in the PEP02, irinotecan and docetaxel arms, respectively. The median progression-free survival (PFS) and overall survival were similar between the trial arms. Commonest grade 3-4 adverse event reported was diarrhoea in the PEP02 and irinotecan groups (27.3% versus 18.2%). CONCLUSION: The ORR associated with PEP02 was comparable with docetaxel and numerically greater than that of irinotecan. PEP02 warrants further evaluation in the advanced gastric cancer setting.


Subject(s)
Adenocarcinoma/drug therapy , Camptothecin/analogs & derivatives , Drug Carriers/administration & dosage , Esophageal Neoplasms/drug therapy , Esophagogastric Junction/pathology , Stomach Neoplasms/drug therapy , Taxoids/administration & dosage , Adenocarcinoma/diagnosis , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Camptothecin/administration & dosage , Docetaxel , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/mortality , Esophagogastric Junction/drug effects , Female , Humans , Irinotecan , Male , Middle Aged , Nanocapsules/administration & dosage , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Survival Rate/trends
4.
Pharmacol Res ; 44(3): 213-20, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11529688

ABSTRACT

Leukotrienes (LTs) and platelet-activating factor (PAF) are important mediators of inflammation and allergy. LDP-392, a novel dual PAF receptor antagonist and 5-lipoxygenase (5-LO) inhibitor, has been identified. LDP-392 is 17.9-fold more potent than zileuton (5-LO inhibitor) in the RBL cytosolic 5-LO assay, and equally potent as MK 287 (PAF receptor antagonist) in the human platelet PAF receptor binding assay. The in vivo dual activities of LDP-392 were confirmed by measuring the inhibition of ex vivo LTB(4)production in rats and PAF-induced hemoconcentration in mice. Intravenous administration of LDP-392 demonstrated greater inhibition than zileuton, BN 50739 or MK 287 on arachidonic acid-induced ear edema and protected mice from LPS-induced lethality. Topical administration of LDP-392, in a dose-dependent manner, inhibited TPA-induced ear edema in mice and UVB-induced erythema in guinea-pigs. These data suggest that LDP-392, as a dual PAF receptor antagonist and 5-LO inhibitor, may be of greater clinical effectiveness.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Furans/pharmacology , Lipoxygenase Inhibitors , Lipoxygenase Inhibitors/pharmacology , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Urea/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arachidonate 5-Lipoxygenase/metabolism , Dose-Response Relationship, Drug , Edema/drug therapy , Erythema/drug therapy , Female , Guinea Pigs , Humans , Lipoxygenase Inhibitors/therapeutic use , Mice , Platelet Membrane Glycoproteins/metabolism , Rats , Urea/analogs & derivatives
5.
Transplantation ; 69(11): 2282-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10868627

ABSTRACT

BACKGROUND: Inasmuch as complement plays a critical role in many pathological processes and in xenograft rejection, efficient complement inhibitors are of great interest. Because the membrane-associated complement inhibitors are very effective, recombinant soluble molecules have been generated. METHODS: We tested the efficacy of complement activation blocker-2 (CAB-2), a recombinant soluble chimeric protein derived from human decay accelerating factor (DAF, CD55) and membrane cofactor protein (MCP, CD46), in two models of pig-to-human xenotransplantation in which tissue injury is complement mediated. The in vitro model consisted of porcine aortic endothelial cells and human serum, and the ex vivo model consisted of a porcine heart perfused with human blood. RESULTS: In vitro, addition of CAB-2 to serum inhibited cytotoxicity and the deposition of C4b and iC3b on the endothelial cells. Ex vivo, addition of CAB-2 to human blood prolonged organ survival from 17.3 +/- 6.4 min in controls to 108 +/- 55.6 min with 910 nM (100 microg/ml) CAB-2 and 219.8 +/- 62.7 min with 1820 nM (200 microg/ml) CAB-2. CAB-2 also retarded the onset of increased coronary vascular resistance. The complement activity of the perfusate was reduced by CAB-2, as was the generation of C3a and SC5b-9. The myocardial tissues had similar deposition of IgG, IgM, and Clq; however, CAB-2 reduced the deposition of C3, C4, and C9. Hearts surviving >240 min demonstrated trace to no deposition of C9 and normal histologic architecture. CONCLUSION: These results indicate that CAB-2 can function as an inhibitor of complement activation and markedly reduce tissue injury in models of pig-to-human xenotransplantation and thus may represent a useful therapeutic agent for xenotransplantation and other complement-mediated conditions.


Subject(s)
Antigens, CD/pharmacology , Complement Inactivator Proteins/pharmacology , Heart Transplantation , Myocardium/pathology , Recombinant Fusion Proteins/pharmacology , Transplantation, Heterologous , Animals , Antigens, CD/genetics , Blood/drug effects , CD55 Antigens/genetics , Chimera/genetics , Complement Inactivator Proteins/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Graft Survival/drug effects , Heart/physiopathology , Humans , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Myocardial Reperfusion Injury/prevention & control , Recombinant Fusion Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Solubility , Swine
6.
Circulation ; 100(5): 553-8, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430771

ABSTRACT

BACKGROUND: We previously demonstrated that inhibiting formation of terminal complement components (C5a and C5b-9) prevents platelet and neutrophil (PMN) but not monocyte activation during simulated extracorporeal circulation (SECC). This study examined whether earlier complement inhibition during SECC, blocking C3a formation, would additionally prevent monocyte activation. METHODS AND RESULTS: SECC was established by recirculating heparinized whole blood from human volunteers on a membrane oxygenator. CAB-2, a chimeric protein constructed from genes encoding the complement regulatory proteins CD46 and CD55, inactivates the C3/C5 convertases and blocks in vitro generation of C3a, C5a, and C5b-9. CAB-2 was used in 4 experiments at a final concentration of 300 micrograms/mL and 4 experiments at 30 micrograms/mL; 4 control runs used vehicle alone. Samples were assayed for C3a and C5b-9, monocyte activation (CD11b upregulation), PMN activation (CD11b upregulation and elastase release), and platelet activation (P-selectin expression and monocyte-platelet conjugate formation). CAB-2 at both doses significantly inhibited formation of C3a and C5b-9 during SECC. High-dose CAB-2 significantly blocked monocyte and PMN CD11b upregulation and PMN elastase release. CAB-2 also inhibited formation of platelet activation-dependent monocyte-platelet conjugates. CONCLUSIONS: Blockade of complement activation early in the common pathway inhibited monocyte CD11b upregulation during SECC, suggesting that early complement components contribute most to monocyte activation during SECC. As expected, PMN and platelet activation were blocked by terminal complement inhibition. This investigation further elucidates the relation between complement and blood cell activation during simulated cardiopulmonary bypass.


Subject(s)
Complement C3a/antagonists & inhibitors , Complement C3a/metabolism , Complement C5a/antagonists & inhibitors , Complement C5a/metabolism , Extracorporeal Circulation , Monocytes/metabolism , Recombinant Fusion Proteins/pharmacology , Blood Platelets/metabolism , CD11 Antigens/drug effects , CD11 Antigens/metabolism , Complement Activation/drug effects , Humans , Monocytes/drug effects , Neutrophils/metabolism , Platelet Activation/drug effects , Up-Regulation/drug effects
7.
Clin Immunol ; 91(1): 99-105, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10219260

ABSTRACT

Primate platelets are different from rodent and rabbit platelets in that they do not express receptors for C3a or C5a or immune adherence receptors. This study assessed the effects of immune complex (IC)-induced complement activation on primate platelets in the circulation. Cynomolgus monkeys (CYN, N = 4) immunized to bovine gamma globulin (BGG) were infused with BGG over 5 min to induce acute intravascular IC formation and complement activation. The studies were carried out under normal complement conditions (N = 12), partial complement inhibition (CAB-2 treated, N = 3), or total complement inhibition (CVF treated, N = 1). Under normal complement conditions, BGG infusion increased C3a levels from undetectable to an average of 11.9 +/- 2.6 micrograms/ml. At this time, decreases occurring in both circulating neutrophils (85 +/- 6%) and monocytes (78 +/- 6%) were significantly greater than decreases in circulating platelets (13 +/- 3%, p < 0.001). Partial complement inhibition had an equivocal effect on the BGG-induced changes in circulating leukocytes, while total complement inhibition abrogated these changes. In contrast, platelet changes were unaffected by complement inhibition. We conclude that, compared to circulating leukocytes, circulating platelets are insensitive to intravascular complement activation induced by IC in the nonhuman primate. These results contrast with previous studies in rodents which demonstrate strong effects of IC-induced intravascular complement activation on both circulating neutrophils and platelets.


Subject(s)
Antigen-Antibody Complex/biosynthesis , Blood Platelets/immunology , Complement Activation , Macaca fascicularis/blood , Macaca fascicularis/immunology , Animals , Antigen-Antibody Complex/blood , Blood Cell Count , Cattle , Complement C3a/metabolism , Immunization , Mice , Platelet Count , Rabbits , Rats , Species Specificity , gamma-Globulins/administration & dosage , gamma-Globulins/immunology
8.
J Med Chem ; 41(11): 1970-9, 1998 May 21.
Article in English | MEDLINE | ID: mdl-9599246

ABSTRACT

By incorporating an N-hydroxyurea functionality onto diaryltetrahydrofurans, a novel series of compounds was investigated as dual 5-lipoxygenese (5-LO) inhibitor and platelet-activating factor (PAF) receptor antagonist. These dual functional compounds were evaluated in vitro for 5-LO inhibition in RBL cell extracts and human whole blood, and PAF receptor antagonism in a receptor binding assay. PAF-induced hemoconcentration and arachidonic acid- and TPA-induced ear edema in mice were used to determine in vivo activities. The structure-activity relationship analysis to define a preclinical lead is presented. (+/-)-trans-2-[3-methoxy-4-(4-chlorophenylthioethoxy)-5-(N-methyl- N-h ydroxyureidyl)methylphenyl]-5-(3,4, 5-trimethoxyphenyl)tetrahydrofuran (40, CMI-392) was selected for further study. In the arachidonic acid-induced mouse ear edema model, 40 was more potent than either zileuton (a 5-LO inhibitor) or BN 50739 (a PAF receptor antagonist), and it demonstrated the same inhibitory effect as a physical combination of the latter two agents. These results suggest that a single compound which both inhibits leukotriene synthesis and blocks PAF receptor binding may provide therapeutic advantages over single-acting agents. The clinical development of compound 40 is in progress.


Subject(s)
Enzyme Inhibitors/pharmacology , Furans/pharmacology , Lipoxygenase Inhibitors , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Urea/analogs & derivatives , Animals , Arachidonic Acid/toxicity , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/ultrastructure , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Drug Evaluation, Preclinical , Edema/chemically induced , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Furans/chemical synthesis , Furans/chemistry , Hematocrit , Humans , Leukotriene B4/biosynthesis , Leukotriene B4/blood , Mice , Rats , Structure-Activity Relationship , Tetradecanoylphorbol Acetate/toxicity , Tumor Cells, Cultured , Urea/chemical synthesis , Urea/chemistry , Urea/pharmacology
9.
J Immunol ; 158(6): 2872-81, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058824

ABSTRACT

A chimeric gene was constructed from the genes coding for the human complement regulatory proteins, membrane cofactor protein (CD46) and decay-accelerating factor (CD55). The recombinant chimeric gene was transfected into Chinese hamster ovary cells. The gene product is a soluble, glycosylated, 110-kDa protein named complement activation blocker-2 (CAB-2). This protein possesses both factor I cofactor activity and decay-accelerating activity, and inactivates classical and alternative C3/C5 convertases in vitro. The specific activity of CAB-2 against cell-associated convertases is greater than that of soluble forms of either membrane cofactor protein or decay-accelerating factor or of both factors combined. CAB-2 also blocks the activation of complement in vivo, inhibiting both the Arthus reaction and Forssman shock in guinea pigs. Studies in rats demonstrate CAB-2 to exhibit favorable biphasic pharmacokinetics with a t1/2 alpha of 10 min and a t1/2 beta of 8 h; the beta phase accounts for 93% of the administered dose. CAB-2 may be an effective therapeutic treatment of acute human diseases in which excessive complement activation causes damage to normal tissues.


Subject(s)
Antigens, CD/genetics , CD55 Antigens/genetics , Complement Inactivator Proteins/genetics , Membrane Glycoproteins/genetics , Recombinant Fusion Proteins/pharmacology , Anaphylaxis/prevention & control , Animals , Antigens, CD/physiology , CD55 Antigens/physiology , Complement Activation/drug effects , Complement Inactivator Proteins/biosynthesis , Complement Inactivator Proteins/isolation & purification , Complement Inactivator Proteins/pharmacokinetics , Female , Guinea Pigs , Membrane Cofactor Protein , Membrane Glycoproteins/physiology , Protein Engineering , Rats , Rats, Inbred Strains , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacokinetics , Solubility
10.
J Am Soc Nephrol ; 5(11): 1888-94, 1995 May.
Article in English | MEDLINE | ID: mdl-7620086

ABSTRACT

Complement is a major mediator of tissue injury in several types of glomerulonephritis. However, no therapeutic agents that inhibit complement activation are available for human use. sCR1 (TP10, BRL 55736) is a recombinant, soluble human complement receptor 1 (CR1) molecule lacking transmembrane and cytoplasmic domains that inhibits C3 and C5 convertase activity by preferentially binding C4b and C3b. To test the efficacy of sCR1 on complement-mediated glomerulonephritis, rats were pretreated with sCR1 (60 mg/kg per day) before and during the induction of three models of complement-dependent glomerulonephritis (concanavalin A and antithymocyte serum models of proliferative glomerulonephritis, passive Heyman nephritis). Daily sCR1 and complement hemolytic activity levels were measured, and renal histology and urine protein excretion were examined. Mean serum sCR1 levels of 100 to 200 micrograms/mL were maintained with a reduction in complement hemolytic activity to less than 15% in most animals. In the antithymocyte serum model, sCR1-treated animals had significant reductions in mesangiolysis, glomerular platelet and macrophage infiltrates, and proteinuria at 48 h. In the concanavalin A model, sCR1 significantly reduced glomerular C3 and fibrin deposits, platelet infiltrates, and proteinuria at 48 h. In passive Heymann nephritis, proteinuria was also significantly reduced (199 +/- 8.5 versus 125 +/- 16 mg/day, P < 0.002) at 5 days. It was concluded that sCR1 significantly reduces both morphologic and functional consequences of several different types of complement-mediated glomerulonephritis and deserves evaluation as a potential therapeutic agent in complement-mediated immune glomerular disease in humans.


Subject(s)
Complement System Proteins/immunology , Glomerulonephritis/drug therapy , Receptors, Complement , Animals , Antilymphocyte Serum , Complement System Proteins/analysis , Concanavalin A/analysis , Concanavalin A/immunology , Disease Models, Animal , Glomerulonephritis/chemically induced , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Immunoglobulin G/analysis , Kidney Glomerulus/pathology , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Complement/analysis , Recombinant Proteins/analysis , Recombinant Proteins/blood , Recombinant Proteins/therapeutic use
11.
Ann Thorac Surg ; 55(3): 619-24, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8383955

ABSTRACT

Although complement activation during cardiopulmonary bypass (CPB) is well documented, its pathogenic role in postperfusion organ injury is unproven. In this study, soluble human complement receptor type 1 (sCR1), a potent inhibitor of complement activation, was used to determine the contribution of complement activation to pulmonary injury in a porcine model of CPB. In vitro experiments demonstrated that sCR1 inhibits both classic and alternative complement pathways in the pig. Seven control piglets and 6 piglets treated with sCR1 (12 mg/kg intravenously) underwent 2 hours of hypothermic (28 degrees C) CPB followed by 2 hours of observation. In control piglets, total hemolytic complement activity and functional activities of C3 and C5 declined to 61.3%, 67.8%, and 61.4% of prebypass values, respectively, after 2 hours of CPB. Plasma from animals treated with sCR1 had virtually no hemolytic activity (total hemolytic complement activity < 5% of baseline), demonstrating effective complement inhibition. Similar degrees of neutropenia developed in the two groups during CPB, and there was no difference in post-CPB lung tissue myeloperoxidase level. Two hours after CPB, pulmonary vascular resistance increased 338% in control piglets but only 147% in piglets pretreated with sCR1 (p < 0.05); the alveolar-arterial gradient was not significantly different between controls (331 +/- 52 mm Hg) and piglets receiving sCR1 (290 +/- 85 mm Hg). Histologic examination revealed similar degrees of pulmonary edema in both groups. These data constitute direct evidence that complement activation plays a pathogenic role in lung injury after CPB.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cardiopulmonary Bypass , Complement Activation , Receptors, Complement/metabolism , Animals , Cardiopulmonary Bypass/adverse effects , Complement C3/analysis , Complement C5/analysis , Complement Hemolytic Activity Assay , In Vitro Techniques , Leukocyte Count , Lung/enzymology , Lung/pathology , Neutrophils/pathology , Peroxidase/analysis , Pulmonary Circulation , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Swine , Vascular Resistance
12.
J Immunol ; 149(5): 1744-50, 1992 Sep 01.
Article in English | MEDLINE | ID: mdl-1324280

ABSTRACT

C receptor-1 is a protein involved in the regulation of C3 and C5-convertases. Recombinant human soluble C receptor-1 has recently been produced and shown to reduce infarct size in a rat model of myocardial ischemia/reperfusion injury. The present study aimed to investigate whether recombinant human soluble C receptor-1 exerts any protective effect on pulmonary injury produced in a rodent model of adult respiratory distress syndrome. In this model, Escherichia coli endotoxin (LPS, 0.1 microgram/kg) combined with platelet-activating factor (1 pmol/kg/min over 60 min, n = 10) caused microvascular lung injury characterized by elevation of myeloperoxidase activity, deposition of C3 and C5b-9 on the endothelium of pulmonary vessels, and pulmonary edema. Furthermore, bronchoalveolar lavage revealed increased neutrophil count and elevated protein concentration. These pulmonary responses were associated with elevated serum TNF-alpha. Pretreatment (10 min, i.v.) with recombinant human soluble C receptor-1 at 10 mg/kg (n = 13), but not at 1 mg/kg, prevented the LPS/platelet-activating factor-induced pulmonary edema (p less than 0.01) and changes in the bronchoalveolar lavage fluid cell count (p less than 0.01) and protein concentration (p less than 0.05), and attenuated the deposition of C3 and C5b-9 to lung vessels. There was no effect on lung myeloperoxidase activity and serum TNF-alpha. Also, C depletion by cobra venom factor (500 U/kg, i.v.) eliminated the pulmonary edema and elevated leukocyte count in bronchoalveolar lavage fluid, but had no effect on lung myeloperoxidase activity and serum TNF-alpha. These data suggest that C factors may play an important role in the pathophysiology of adult respiratory distress syndrome.


Subject(s)
Complement System Proteins/physiology , Endotoxins/toxicity , Lipopolysaccharides/toxicity , Lung/drug effects , Platelet Activating Factor/toxicity , Respiratory Distress Syndrome/etiology , Animals , Complement C3/analysis , Complement Membrane Attack Complex/analysis , Elapid Venoms/pharmacology , Leukocyte Count/drug effects , Male , Peroxidase/analysis , Rats , Rats, Inbred Strains , Receptors, Complement/physiology , Receptors, Complement 3b , Tumor Necrosis Factor-alpha/analysis
13.
J Immunol ; 149(5): 1723-8, 1992 Sep 01.
Article in English | MEDLINE | ID: mdl-1387151

ABSTRACT

We examined the role of C activation in ischemia reperfusion injury by inhibiting C activation in a rat model of mesenteric arterial occlusion. In anesthetized rats, 60 min of mesenteric arterial occlusion was followed by 3 h of reperfusion. PBS alone or containing soluble C receptor 1 (3 or 6 mg) was administered i.v. Controls underwent laparotomy without ischemia. Relative serum C activities were assessed by hemolytic assay, neutrophil (polymorphonuclear leukocyte) sequestration by tissue content of myeloperoxidase (MPO) activity, intestinal mucosal injury by histologic grading, lung vascular permeability by the ratio of bronchoalveolar lavage to blood concentration of radiolabeled BSA, and endothelial cell injury was quantified by measurement of plasma factor VIII-related Ag. After reperfusion, PBS-treated animals had increased intestinal MPO (0.048 +/- 0.007 U/g) compared to sham (0.022 +/- 0.005 U/g (p less than 0.05)) and intestinal mucosal injury score (2.490 +/- 0.221) compared to sham (0.331 +/- 0.045 (p less than 0.05)). Treatment with 6 mg soluble C receptor 1 15 min before reperfusion reduced intestinal MPO (0.017 +/- 0.003 U/g (p less than 0.05)) and mucosal injury (1.733 +/- 0.168 (p less than 0.05)) compared to PBS control. PBS-treated animals also demonstrated increased lung MPO (0.314 +/- 0.025 U/g vs 0.085 +/- 0.018 in sham (p less than 0.05)) and increased lung permeability (bronchoalveolar lavage/blood cpm 11.32 +/- 1.35 x 10(-3) vs sham 2.22 +/- 0.19 x 10(-3) (p less than 0.05)). Treatment with 6 mg soluble C receptor 1 15 min before reperfusion or at reperfusion reduced the lung permeability (bronchoalveolar lavage/blood cpm 3.90 +/- 0.79 x 10(-3) and 5.08 +/- 0.75, respectively (both p less than 0.05)) compared to PBS control, but did not reduce lung MPO (0.342 +/- 0.031 U/g and 0.246 +/- 0.025), respectively. Treatment with sCR1 also reduced the release of factor VIII-related Ag, 5-day mortality, and C hemolytic activity. In this model, C is a major mediator of intestinal injury and extraintestinal injury.


Subject(s)
Intestines/blood supply , Lung/blood supply , Receptors, Complement/physiology , Reperfusion Injury/prevention & control , Animals , Complement Activation , Intestines/pathology , Lung/metabolism , Lung/pathology , Male , Neutrophils/immunology , Permeability , Rats , Rats, Inbred Strains , Receptors, Complement 3b
14.
J Immunol ; 148(10): 3086-92, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1374449

ABSTRACT

Acute lung injury produced by deposition of IgA immune complexes is complement-dependent, neutrophil-independent, oxygen radical-mediated, and may be a result of the formation of the hydroxyl radical (HO) generated directly or indirectly from activated lung macrophages. The current studies were designed to evaluate further the pathophysiologic events that occur after intrapulmonary deposition of IgA immune complexes. Pretreatment of rats with the human recombinant soluble complement receptor-1 resulted in marked attenuation of IgA immune complex-induced lung injury. Intravenous administration of antibody to CD18, but not antibody to CD11b, was highly protective against lung injury. Treatment of animals with either anti-endothelial leukocyte-adhesion molecule-1 or anti-TNF-alpha, both of which were highly protective against IgG immune complex-induced lung injury, had no protective effects in the model of IgA immune complex-induced lung injury. Immunohistochemical analysis revealed up-regulation of the endothelial leukocyte adhesion molecule-1 in the pulmonary vasculature after deposition of IgA immune complexes. This up-regulation was TNF-alpha-dependent. The arginine analog, NG-monomethyl-L-arginine, was highly protective against IgA immune complex-induced lung injury. This protective effect was reversed by the co-presence of L-arginine (but not D-arginine). Protective interventions against IgA immune complex-induced lung injury were inversely correlated with the numbers of macrophages that could be retrieved by lung lavage. These data suggest fundamental differences in the pathogenesis of lung injury after intrapulmonary deposition of IgA immune complexes, as compared with injury caused by deposition of IgG immune complexes. In the latter, neutrophils, intrapulmonary generation of TNF-alpha, and up-regulation of pulmonary vascular endothelial leukocyte-adhesion molecule-1 are required for the full development of lung injury, whereas no such requirements appear in the case of IgA immune complex-induced lung injury. Full expression of IgA immune complex-induced lung injury also appears to require L-arginine, suggesting a possible role for nitric oxide or its derivatives in events ultimately leading to injury.


Subject(s)
Antigen-Antibody Complex/immunology , Antigens, CD/physiology , Arginine/physiology , Immunoglobulin A/immunology , Lung Diseases/etiology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Bronchoalveolar Lavage Fluid/cytology , CD18 Antigens , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/physiology , E-Selectin , Macrophage-1 Antigen/physiology , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Rats , Tumor Necrosis Factor-alpha/physiology , omega-N-Methylarginine
16.
J Immunol ; 148(5): 1479-85, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1311349

ABSTRACT

Complement activation is an important step for triggering of acute inflammatory reactions. Soluble human recombinant complement receptor type 1 (sCR1) blocks complement activation by both classical and alternative pathways. In addition to glycogen-induced peritonitis, three models of complement-dependent acute inflammatory injury have been used to assess the protective effects of sCR1: lung and dermal injury after intraalveolar or intradermal deposition of IgG immune complexes; acute lung injury resulting from intravascular activation of complement after the i.v. injection of cobra venom factor; and acute skin and lung injury (at 4 h) after thermal trauma involving 25 to 30% total body surface area. Vascular injury was quantified by increases in vascular permeability, hemorrhage, neutrophil infiltration, and, as indicated, tissue water content. Intravenous infusion of sCR1 reduced lung and dermal vascular injury in all models studied. In glycogen-induced peritoneal exudates sCR1-reduced neutrophil accumulation by 79%. In animals undergoing IgG immune complex-induced alveolitis, sCR1 treatment reduced vascular permeability and hemorrhage by 72 and 71%, respectively, and tissue accumulation of neutrophils was reduced by 68%. After cobra venom factor injection, sCR1 reduced increases in lung vascular permeability by 67%, hemorrhage by 73%, and lung myeloperoxidase content by 55%. Four hours after thermal injury of skin, sCR1-treated animals demonstrated significant protection against lung injury; increases in vascular permeability and hemorrhage were reduced by 45 and 46%, respectively, and myeloperoxidase content was lowered by 39%. In thermal injury of the skin, sCR1 injection reduced dermal vascular permeability by 25% at 1 h (p = NS) and 44% at 4 h. Water content in skin biopsies was also decreased. There was a dose-response relationship between the amount of sCR1 infused and the extent of protection in each of the injury models. These data demonstrate that sCR1 offers significant protection against complement-dependent tissue injury in the animal models studied and that the protective effects are related to reduced neutrophil content.


Subject(s)
Complement System Proteins/physiology , Neutrophils/physiology , Receptors, Complement , Animals , Antigen-Antibody Complex/physiology , Burns/physiopathology , Elapid Venoms/toxicity , Glycogen/pharmacology , Lung/drug effects , Lung/enzymology , Male , Peroxidase/analysis , Rabbits , Rats , Rats, Inbred Strains , Receptors, Complement/analysis , Receptors, Complement 3b , Recombinant Proteins/pharmacology
17.
Transplantation ; 52(5): 868-73, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1949173

ABSTRACT

In the guinea pig-to-rat model of hyperacute xenograft (Xg) rejection, the effect of complement inhibition using systemically administered soluble complement receptor type 1 (sCR1) on discordant cardiac Xg survival was investigated. In PBS-treated control Xg recipients (n = 13), hyperacute rejection was rapid, with a mean Xg survival of 17 +/- 4 min. Therapy with sCR1 prolonged survival of cardiac Xgs in a dose-dependent manner. A 3 mg/kg bolus of sCR1 (n = 4) prolonged Xg survival to 64 +/- 29 min (not significant). Increasing the sCR1 dose to 5.9 mg/kg (n = 4) significantly delayed Xg rejection to 71 +/- 17 min (P-0.026, log-rank test vs. control). In 10 recipients treated with 15 mg/kg sCR1, mean Xg survival was further prolonged to 189 +/- 36 min (P-0.0004) with no adverse effects. While 2 of 8 recipients receiving 60 mg/kg sCR1 died with functioning Xgs at 30 and 300 min due to anastomotic bleeding, Xg survival averaged over 12 hr (747 +/- 100 min, P-0.0004) in the remaining 6 recipients. sCR1 administration significantly inhibited serum complement activity in a parallel dose-dependent fashion, with the 60 mg/kg dose reducing complement activity by 95 +/- 1 and 96 +/- 1% five and 30 min following Xg reperfusion, respectively. Immunofluorescence microscopy revealed rat IgM bound to all cardiac Xgs in control as well as sCR1-treated recipients. In addition, serial histologic examination of cardiac Xgs harvested within 21 min of graft reperfusion revealed occlusive platelet aggregates within the coronary vessels as well as interstitial hemorrhage and myocardial necrosis in Xgs from control recipients, all of which were only minimally present in Xgs from recipients treated with sCR1. These studies show that complement inhibition with sCR1 significantly delays hyperacute cardiac Xg rejection in this discordant model and may be an important component in a therapeutic protocol for xenotransplantation.


Subject(s)
Graft Rejection/drug effects , Receptors, Complement , Transplantation, Heterologous/immunology , Animals , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , Graft Rejection/immunology , Guinea Pigs , Heart Transplantation/immunology , Hemolysis/drug effects , Male , Microscopy, Fluorescence , Myocardium/cytology , Rats , Rats, Inbred Lew
18.
J Immunol ; 146(1): 250-6, 1991 Jan 01.
Article in English | MEDLINE | ID: mdl-1824590

ABSTRACT

The human CR1 was genetically engineered by site directed mutagenesis into a truncated form which was secreted from transfected Chinese hamster ovary cells. This soluble recombinant CR1 (sCR1) was purified from the supernatants of the Chinese hamster ovary cells cultured in a hollow fiber bioreactor. sCR1 inhibits the C3 and C5 convertases of the classical and the alternative pathways in vitro. The ability of sCR1 to inhibit the immune complex-mediated inflammation in vivo was tested in a rat reversed passive Arthus reaction model. Administration of sCR1 at the dermal sites reduced the Arthus vasculitis in a dose-dependent manner as judged by both gross and microscopic examination, as well as by immunohistologic localization of C3 and C5b-9 neoantigen deposits. These data suggest that sCR1 inhibits the Arthus reaction by interrupting the activation of the C cascade, hence limiting the detrimental immune complex-induced tissue damage in vivo.


Subject(s)
Arthus Reaction/immunology , Complement System Proteins/metabolism , Receptors, Complement/physiology , Animals , Arthus Reaction/pathology , Complement Activation , Fluorescent Antibody Technique , Humans , Immunization, Passive , Molecular Weight , Rats , Rats, Inbred Strains , Receptors, Complement/chemistry , Receptors, Complement/genetics , Receptors, Complement 3b , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Skin/immunology , Skin/pathology , Solubility , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...