Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Clin Immunol ; 251: 109629, 2023 06.
Article in English | MEDLINE | ID: mdl-37149117

ABSTRACT

The objective of this study was to characterize the complement-inhibiting activity of SAR445088, a novel monoclonal antibody specific for the active form of C1s. Wieslab® and hemolytic assays were used to demonstrate that SAR445088 is a potent, selective inhibitor of the classical pathway of complement. Specificity for the active form of C1s was confirmed in a ligand binding assay. Finally, TNT010 (a precursor to SAR445088) was assessed in vitro for its ability to inhibit complement activation associated with cold agglutinin disease (CAD). TNT010 inhibited C3b/iC3b deposition on human red blood cells incubated with CAD patient serum and decreased their subsequent phagocytosis by THP-1 cells. In summary, this study identifies SAR445088 as a potential therapeutic for the treatment of classical pathway-driven diseases and supports its continued assessment in clinical trials.


Subject(s)
Anemia, Hemolytic, Autoimmune , Complement C1s , Humans , Complement C1s/metabolism , Complement Activation , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Complement Inactivating Agents/therapeutic use , Complement Pathway, Classical
2.
Immunohorizons ; 3(11): 519-530, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690560

ABSTRACT

Terminal complement pathway inhibition at the level of C5 alleviates symptoms of several diseases associated with complement overactivation. However, C5 blockade is associated with an increased risk of invasive meningococcal disease despite immunization. Targeting specific complement pathways proximal to C5 provides the theoretical advantage of leaving the other pathways (including the terminal pathway) intact for immune surveillance. We aimed to address the risk of Neisseria meningitidis and Streptococcus pneumoniae infections when inhibiting the classical pathway (CP) using a specific C1s inhibitor (TNT005). Addition of TNT005 to 20% normal human serum that contained anti-meningococcal capsular Ab decreased C4 deposition 8-fold and abrogated killing of N. meningitidis, despite leaving C3 deposition intact. TNT005 impaired killing of N. meningitidis in 78% nonimmune human plasma and 78% whole blood but permitted killing in both when specific anti-capsular Ab was added. Simultaneously inhibiting both the CP and alternative pathway (AP) blocked killing of Ab-coated N. meningitidis in whole blood. Blocking the AP alone abrogated C3 deposition, whereas TNT005 only partially inhibited (∼40% decrease) C3 deposition on S. pneumoniae coated with anti-capsular Ab. Blocking either the CP or AP alone did not impair killing of pneumococci in whole blood containing specific Ab (<10% survival at 3 h); however, blocking both pathways resulted in ∼35% bacterial survival. These data suggest that killing of N. meningitidis or S. pneumoniae in whole blood containing specific anti-capsular Abs is unimpeded by TNT005. Meningococcal and pneumococcal capsular conjugate vaccines may mitigate risk of these infections in patients receiving C1s inhibitors.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Capsules/immunology , Complement C1/antagonists & inhibitors , Neisseria meningitidis/immunology , Streptococcus pneumoniae/immunology , Antibodies, Monoclonal/immunology , Bacterial Proteins/immunology , Complement C4/immunology , Complement Pathway, Classical/drug effects , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology
3.
J Invest Dermatol ; 139(12): 2417-2424.e2, 2019 12.
Article in English | MEDLINE | ID: mdl-31229501

ABSTRACT

Deposition of autoantibodies (α-BP180 and BP230) and complement along the dermal-epidermal-junction is a hallmark of bullous pemphigoid and was shown to be important for pathogenesis. Given the adverse effects of standard treatment (glucocorticoids, immunosuppressants), there is an unmet need for safe and effective therapies. In this phase 1 trial, we evaluated the safety and activity of BIVV009 (sutimlimab, previously TNT009), a targeted C1s inhibitor, in 10 subjects with active or past bullous pemphigoid (NCT02502903). Four weekly 60 mg/kg infusions of BIVV009 proved sufficient for inhibition of the classical complement pathway in all patients, as measured by CH50. C3c deposition along the dermal-epidermal junction was partially or completely abrogated in 4 of 5 patients, where it was present at baseline. BIVV009 was found to be safe and tolerable in this elderly population, with only mild to moderate adverse events reported (e.g., headache, fatigue). One serious adverse event (i.e., fatal cardiac decompensation) occurred at the end of the post-treatment observation period in an 84-year-old patient with a history of diabetes and heart failure, but was deemed unlikely to be related to the study drug. This trial provides the first results with a complement-targeting therapy in bullous pemphigoid, to our knowledge, and supports further studies on BIVV009's efficacy and safety in this population.


Subject(s)
Autoantigens/administration & dosage , Complement C3/metabolism , Complement Pathway, Classical/drug effects , Dermis/pathology , Dystonin/administration & dosage , Epidermis/pathology , Non-Fibrillar Collagens/administration & dosage , Pemphigoid, Bullous/drug therapy , Aged , Aged, 80 and over , Dermis/metabolism , Epidermis/metabolism , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Pemphigoid, Bullous/metabolism , Pemphigoid, Bullous/pathology , Collagen Type XVII
4.
J Immunol ; 202(4): 1200-1209, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30635392

ABSTRACT

The classical pathway of complement (CP) can mediate C3 opsonization of Ags responsible for the costimulation and activation of cognate B lymphocytes. In this manner, the complement system acts as a bridge between the innate and adaptive immune systems critical for establishing a humoral response. However, aberrant complement activation is often observed in autoimmune diseases in which C3 deposition on self-antigens may serve to activate self-reactive B cell clones. In this study, we use BIVV009 (Sutimlimab), a clinical stage, humanized mAb that specifically inhibits the CP-specific serine protease C1s to evaluate the impact of upstream CP antagonism on activation and proliferation of normal and autoimmune human B cells. We report that BIVV009 significantly inhibited complement-mediated activation and proliferation of primary human B cells. Strikingly, CP antagonism suppressed human Ig-induced activation of B cells derived from patients with rheumatoid arthritis. These results suggest that clinical use of CP inhibitors in autoimmune patients may not only block complement-mediated tissue damage, but may also prevent the long-term activation of autoimmune B cells and the production of autoantibodies that contribute to the underlying pathologic condition of these diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Arthritis, Rheumatoid/drug therapy , B-Lymphocytes/drug effects , Complement C1s/antagonists & inhibitors , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Proliferation/drug effects , Complement Activation/drug effects , Complement C1s/immunology , Humans
5.
Blood ; 133(9): 893-901, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30559259

ABSTRACT

Cold agglutinin disease is a difficult-to-treat autoimmune hemolytic anemia in which immunoglobulin M antibodies bind to erythrocytes and fix complement, resulting in predominantly extravascular hemolysis. This trial tested the hypothesis that the anti-C1s antibody sutimlimab would ameliorate hemolytic anemia. Ten patients with cold agglutinin disease participated in the phase 1b component of a first-in-human trial. Patients received a test dose of 10-mg/kg sutimlimab followed by a full dose of 60 mg/kg 1 to 4 days later and 3 additional weekly doses of 60 mg/kg. All infusions were well tolerated without premedication. No drug-related serious adverse events were observed. Seven of 10 patients with cold agglutinin disease responded with a hemoglobin increase >2 g/dL. Sutimlimab rapidly increased hemoglobin levels by a median of 1.6 g/dL within the first week, and by a median of 3.9 g/dL (interquartile range, 1.3-4.5 g/dL; 95% confidence interval, 2.1-4.5) within 6 weeks (P = .005). Sutimlimab rapidly abrogated extravascular hemolysis, normalizing bilirubin levels within 24 hours in most patients and normalizing haptoglobin levels in 4 patients within 1 week. Hemolytic anemia recurred when drug levels were cleared from the circulation 3 to 4 weeks after the last dose of sutimlimab. Reexposure to sutimlimab in a named patient program recapitulated the control of hemolytic anemia. All 6 previously transfused patients became transfusion-free during treatment. Sutimlimab was safe, well tolerated, and rapidly stopped C1s complement-mediated hemolysis in patients with cold agglutinin disease, significantly increasing hemoglobin levels and precluding the need for transfusions. This trial was registered at www.clinicaltrials.gov as #NCT02502903.


Subject(s)
Anemia, Hemolytic, Autoimmune/drug therapy , Anemia, Hemolytic/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Complement C1s/antagonists & inhibitors , Hemolysis/drug effects , Severity of Illness Index , Aged , Anemia, Hemolytic/etiology , Anemia, Hemolytic, Autoimmune/complications , Complement C1s/immunology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Prospective Studies
6.
Transplantation ; 101(10): 2410-2418, 2017 10.
Article in English | MEDLINE | ID: mdl-28926521

ABSTRACT

BACKGROUND: Complement may play a key role in antibody-mediated rejection. A promising therapeutic approach may be classical pathway (CP) inhibition at the level of early component C1. METHODS: In this first-in-human, double-blind, randomized placebo-controlled phase 1 trial, we evaluated the safety and complement inhibitory effect of TNT009, a humanized monoclonal anti-C1s antibody. Sixty-four adult healthy volunteers received either single (n = 48; 7 consecutive cohorts, 0.3-100 mg/kg) or 4 weekly infusions (n = 16; 2 consecutive cohorts, 30 and 60 mg/kg per infusion) of TNT009 or placebo. To assess the effect of treatment on complement activity, sera from dosed subjects were analyzed in a CP activation assay evaluating C3d deposition on HLA-coated microbeads spiked with alloantibodies. RESULTS: Single doses of TNT009 at 3 to 100 mg/kg uniformly and profoundly inhibited HLA antibody-mediated C3d deposition (≥86% after 60 minutes), whereby the duration of CP inhibition (2-14 days) was dose-dependent. Four weekly doses persistently blocked complement for 5 to 6 weeks. Ex vivo serum CP activity was profoundly inhibited when TNT009 concentrations exceeded 20 µg/mL. Infusions were well tolerated without serious or severe adverse events. CONCLUSIONS: Treatment with TNT009 was safe and potently inhibited CP activity. Future studies in patients are required to assess the potential of TNT009 for preventing or treating antibody-mediated rejection.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Complement Activation/drug effects , Complement C1s/antagonists & inhibitors , Complement Inactivating Agents/administration & dosage , HLA Antigens/immunology , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/therapeutic use , Complement C1s/immunology , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/blood , Double-Blind Method , Drug Administration Schedule , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Time Factors , Treatment Outcome
7.
Transplantation ; 101(7): 1559-1572, 2017 07.
Article in English | MEDLINE | ID: mdl-28640789

ABSTRACT

BACKGROUND: Antibody-mediated rejection (AMR) of most solid organs is characterized by evidence of complement activation and/or intragraft macrophages (C4d + and CD68+ biopsies). We previously demonstrated that crosslinking of HLA I by antibodies triggered endothelial activation and monocyte adhesion. We hypothesized that activation of the classical complement pathway at the endothelial cell surface by HLA antibodies would enhance monocyte adhesion through soluble split product generation, in parallel with direct endothelial activation downstream of HLA signaling. METHODS: Primary human aortic endothelial cells (HAEC) were stimulated with HLA class I antibodies in the presence of intact human serum complement. C3a and C5a generation, endothelial P-selectin expression, and adhesion of human primary and immortalized monocytes (Mono Mac 6) were measured. Alternatively, HAEC or monocytes were directly stimulated with purified C3a or C5a. Classical complement activation was inhibited by pretreatment of complement with an anti-C1s antibody (TNT003). RESULTS: Treatment of HAEC with HLA antibody and human complement increased the formation of C3a and C5a. Monocyte recruitment by human HLA antibodies was enhanced in the presence of intact human serum complement or purified C3a or C5a. Specific inhibition of the classical complement pathway using TNT003 or C1q-depleted serum significantly reduced adhesion of monocytes in the presence of human complement. CONCLUSIONS: Despite persistent endothelial viability in the presence of HLA antibodies and complement, upstream complement anaphylatoxin production exacerbates endothelial exocytosis and leukocyte recruitment. Upstream inhibition of classical complement may be therapeutic to dampen mononuclear cell recruitment and endothelial activation characteristic of microvascular inflammation during AMR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Complement Inactivating Agents/pharmacology , Complement Pathway, Classical/drug effects , Endothelial Cells/drug effects , HLA-A Antigens/immunology , Immunosuppressive Agents/pharmacology , Monocytes/drug effects , Cells, Cultured , Coculture Techniques , Complement C3a/pharmacology , Complement C5a/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Exocytosis/drug effects , HLA-A Antigens/metabolism , Humans , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Monocytes/immunology , Monocytes/metabolism , P-Selectin/immunology , P-Selectin/metabolism
8.
Clin Immunol ; 163: 84-90, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26769276

ABSTRACT

Complement activation contributes to inflammation in many diseases, yet it also supports physiologic apoptotic cells (AC) clearance and its downstream immunosuppressive effects. The roles of individual complement components in AC phagocytosis have been difficult to dissect with artificially depleted sera. Using human in vitro systems and the novel antibody complement C1s inhibitor TNT003, we uncoupled the role of the enzymatic activation of the classical pathway from the opsonizing role of C1q in mediating a) the phagocytosis of early and late AC, and b) the immunosuppressive capacity of early AC. We found that C1s inhibition had a small impact on the physiologic clearance of early AC, leaving their immunosuppressive properties entirely unaffected, while mainly inhibiting the phagocytosis of late apoptotic/secondary necrotic cells. Our data suggest that C1s inhibition may represent a valuable therapeutic strategy to control classical pathway activation without causing significant AC accumulation in diseases without defects in AC phagocytosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Complement Activation/drug effects , Complement C1q/drug effects , Complement C1s/antagonists & inhibitors , Cytokines/drug effects , Cytophagocytosis/drug effects , Immune Tolerance/drug effects , Macrophages/drug effects , Apoptosis/immunology , Complement Activation/immunology , Complement C1q/immunology , Complement C1q/metabolism , Complement C1s/metabolism , Complement C3b/drug effects , Complement C3b/immunology , Complement C3b/metabolism , Cytokines/immunology , Cytophagocytosis/immunology , Humans , In Vitro Techniques , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Interleukin-6/immunology , Jurkat Cells , Macrophages/immunology , Phagocytosis/drug effects , Phagocytosis/immunology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology
9.
Blood ; 123(26): 4015-22, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24695853

ABSTRACT

Activation of the classical pathway (CP) of complement is often associated with autoimmune disorders in which disease pathology is linked to the presence of an autoantibody. One such disorder is cold agglutinin disease (CAD), an autoimmune hemolytic anemia in which autoantibodies (cold agglutinins) bind to red blood cells (RBCs) at low temperatures. Anemia occurs as a result of autoantibody-mediated CP activation on the surface of the erythrocyte, leading to the deposition of complement opsonins that drive extravascular hemolysis in the liver. Here we test the effects of TNT003, a mouse monoclonal antibody targeting the CP-specific serine protease C1s, on CP activity induced by cold agglutinins on human RBCs. We collected 40 individual CAD patient samples and showed that TNT003 prevented cold agglutinin-mediated deposition of complement opsonins that promote phagocytosis of RBCs. Furthermore, we show that by preventing CP activation, TNT003 also prevents cold agglutinin-driven generation of anaphylatoxins. Finally, we provide evidence that CP activity in CAD patients terminates prior to activation of the terminal cascade, supporting the hypothesis that the primary route of RBC destruction in these patients occurs via extravascular hemolysis. Our results support the development of a CP inhibitor for the treatment of CAD.


Subject(s)
Anemia, Hemolytic, Autoimmune/drug therapy , Antibodies, Monoclonal, Murine-Derived/pharmacology , Complement Activation/drug effects , Complement C1s/antagonists & inhibitors , Serine Proteases/blood , Serine Proteinase Inhibitors/pharmacology , Aged , Anaphylatoxins/metabolism , Anemia, Hemolytic, Autoimmune/enzymology , Anemia, Hemolytic, Autoimmune/pathology , Animals , Cell Line, Tumor , Complement C1s/metabolism , Female , Hemolysis/drug effects , Humans , Male , Mice , Middle Aged
10.
Cancer Res ; 73(11): 3316-25, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23667176

ABSTRACT

LD22-4, an 86-amino acid fragment of the basic fibroblast growth factor, is an inhibitor of cell migration. LD22-4 inhibits the migration of various tumor cells, endothelial cells, and fibroblasts in vitro and suppresses tumor growth and angiogenesis in vivo. LD22-4 is effective in the presence of multiple growth factors, either alone or in combination, as well as haptotactic factors. LD22-4 inhibits the rate of malignant gliomas prepared from U87MG cells in an orthotopic mouse model by 90% compared with untreated mice. Using U87MG cells, we identified the LD22-4 membrane receptor as neuropilin 1 (NRP1). The identification of NRP1 as the LD22-4 receptor was based upon mass spectrometric analysis of proteins that bind to LD22-4, immunoprecipitation of an NRP1-LD22-4 complex formed during incubation of LD22-4 with U87MG cells, LD22-4-NRP1 coimmunoprecipitation studies, and binding of LD22-4 to HEK293 cells expressing NRP1. In contrast, NRP1 binding of an inactive mutant of LD22-4 was substantially reduced. As is typical of NRP1-binding proteins, LD22-4 itself binds to heparin and requires heparan sulfate for binding to cells. The addition of heparin to migration assays increased the inhibitory activity of LD22-4. In addition to a heparin-binding region, LD22-4 contains a 5-amino acid C-terminus that matches an NRP1 consensus binding sequence. Thus, direct binding experiments, dependence on heparan sulfate, and the presence of a NRP1 consensus binding sequence indicate that NRP1 is the binding site of LD22-4 and mediates inhibition of cell migration.


Subject(s)
Cell Movement/drug effects , Fibroblast Growth Factor 2/pharmacology , Glioma/drug therapy , Neuropilin-1/metabolism , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Cell Growth Processes/drug effects , Female , Fibroblast Growth Factor 2/chemistry , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Heparin/chemistry , Heparin/metabolism , Humans , Mice , Mice, Nude , Neuropilin-1/antagonists & inhibitors , Peptide Fragments/chemistry , Protein Binding , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 14(7): 2137-44, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381955

ABSTRACT

PURPOSE: ATN-161 (Ac-PHSCN-NH(2)) is an integrin-binding peptide that is currently in phase II trials in cancer patients. This peptide has been shown to have antitumor activity in a number of different preclinical models. EXPERIMENTAL DESIGN: In this study, we examined the binding, biodistribution, and dose and biomarker response of ATN-161 in several animal models. RESULTS: ATN-161 bound to the beta subunit of a number of different integrins implicated in tumor growth and progression, which depended on its cysteine thiol. The peptide had antiangiogenic activity in the Matrigel plug model, and this activity could be reversed by inhibitors of protein kinase A, an effector of alpha(5)beta(1)-dependent angiogenesis. A labeled analogue of ATN-161, ATN-453, localized to neovessels but not to preexisting vasculature in vivo. The half-life of the peptide when localized to a tumor was much longer than in plasma. Dose-response studies in the Matrigel plug model of angiogenesis or a Lewis lung carcinoma model of tumor growth showed a U-shaped dose-response curve with 1 to 10 mg/kg given thrice a week, being the optimal dose range of ATN-161. Two additional pharmacodynamic models of angiogenesis (dynamic contrast-enhanced magnetic resonance imaging and measurement of endothelial cell progenitors) also revealed U-shaped dose-response curves. CONCLUSIONS: The presence of a U-shaped dose-response curve presents a significant challenge to identifying a biologically active dose of ATN-161. However, the identification of biomarkers of angiogenesis that also exhibit this same U-shaped response should allow the translation of those biomarkers to the clinic, allowing them to be used to identify the active dose of ATN-161 in phase II studies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms, Experimental/drug therapy , Oligopeptides/pharmacology , Animals , Biomarkers, Tumor/analysis , Dose-Response Relationship, Drug , Humans , Mice , Neovascularization, Pathologic/drug therapy
12.
Nat Struct Mol Biol ; 15(4): 422-3, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18376415

ABSTRACT

The urokinase receptor (uPAR) can recognize several ligands. The structural basis for this multiple ligand recognition by uPAR is unknown. This study reports the crystal structures of uPAR in complex with both urokinase (uPA) and vitronectin and reveal that uPA occupies the central cavity of the receptor, whereas vitronectin binds at the outer side of the receptor. These results provide a structural understanding of one receptor binding to two ligands.


Subject(s)
Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Vitronectin/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Cell Surface/chemistry , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/chemistry , Vitronectin/chemistry
13.
Mol Cancer Ther ; 5(9): 2271-80, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985061

ABSTRACT

PURPOSE: Integrins are expressed by numerous tumor types including breast cancer, in which they play a crucial role in tumor growth and metastasis. In this study, we evaluated the ability of ATN-161 (Ac-PHSCN-NH2), a 5-mer capped peptide derived from the synergy region of fibronectin that binds to alpha5beta1 and alphavbeta3 in vitro, to block breast cancer growth and metastasis. EXPERIMENTAL DESIGN: MDA-MB-231 human breast cancer cells were inoculated s.c. in the right flank, or cells transfected with green fluorescent protein (MDA-MB-231-GFP) were inoculated into the left ventricle of female BALB/c nu/nu mice, resulting in the development of skeletal metastasis. Animals were treated with vehicle alone or by i.v. infusion with ATN-161 (0.05-1 mg/kg thrice a week) for 10 weeks. Tumor volume was determined at weekly intervals and tumor metastasis was evaluated by X-ray, microcomputed tomography, and histology. Tumors were harvested for histologic evaluation. RESULT: Treatment with ATN-161 caused a significant dose-dependent decrease in tumor volume and either completely blocked or caused a marked decrease in the incidence and number of skeletal as well as soft tissue metastases. This was confirmed histologically as well as radiographically using X-ray and microcomputed tomography. Treatment with ATN-161 resulted in a significant decrease in the expression of phosphorylated mitogen-activated protein kinase, microvessel density, and cell proliferation in tumors grown in vivo. CONCLUSION: These studies show that ATN-161 can block breast cancer growth and metastasis, and provides a rationale for the clinical development of ATN-161 for the treatment of breast cancer.


Subject(s)
Adenocarcinoma/drug therapy , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Oligopeptides/pharmacology , Adenocarcinoma/pathology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Radiography/instrumentation , Soft Tissue Neoplasms/prevention & control , Soft Tissue Neoplasms/secondary , Transfection , Xenograft Model Antitumor Assays
14.
Science ; 311(5761): 656-9, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16456079

ABSTRACT

The urokinase plasminogen activator binds to its cellular receptor with high affinity and initiates signaling cascades that are implicated in pathological processes including tumor growth, metastasis, and inflammation. We report the crystal structure at 1.9 angstroms of the urokinase receptor complexed with the urokinase amino-terminal fragment and an antibody against the receptor. The three domains of urokinase receptor form a concave shape with a central cone-shaped cavity where the urokinase fragment inserts. The structure provides insight into the flexibility of the urokinase receptor that enables its interaction with a wide variety of ligands and a basis for the design of urokinase-urokinase receptor antagonists.


Subject(s)
Receptors, Cell Surface/chemistry , Urokinase-Type Plasminogen Activator/chemistry , Antibodies/chemistry , Antibodies/metabolism , Crystallography, X-Ray , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/metabolism
15.
Cancer Res ; 65(17): 7775-81, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140945

ABSTRACT

Pancreatic carcinomas express high levels of urokinase-type plasminogen activator (uPA) and its receptor (uPAR), both of which mediate cell migration and invasion. We investigated the hypotheses that (a) insulin-like growth factor-I (IGF-I)- and hepatocyte growth factor (HGF)-mediated migration and invasion of human pancreatic carcinoma cells require uPA and uPAR function and (b) inhibition of uPAR inhibits tumor growth, retroperitoneal invasion, and hepatic metastasis of human pancreatic carcinomas in mice. Using transwell assays, we investigated the effect of IGF-I and HGF on L3.6pl migration and invasion. We measured the induction of uPA and uPAR following treatment of cells with IGF-I and HGF using immunoprecipitation and Western blot analysis. The importance of uPA and uPAR on L3.6pl cell migration and invasion was studied by inhibiting their activities with amiloride and antibodies before cytokine treatment. In an orthotopic mouse model of human pancreatic carcinoma, we evaluated the effect of anti-uPAR monoclonal antibodies with and without gemcitabine on primary tumor growth, retroperitoneal invasion, and hepatic metastasis. IGF-I and HGF mediated cell migration and invasion in L3.6pl cells. In addition, IGF-I and HGF induced uPA and uPAR expression in L3.6pl cells. In vitro, blockade of uPA and uPAR activity inhibited IGF-I- and HGF-mediated cell migration and invasion. Treatment of mice with anti-uPAR monoclonal antibody significantly decreased pancreatic tumor growth and hepatic metastasis and completely inhibited retroperitoneal invasion. Our study shows the importance of the uPA/uPAR system in pancreatic carcinoma cell migration and invasion. These findings suggest that uPAR is a potential target for therapy in patients with pancreatic cancer.


Subject(s)
Cell Movement/physiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Amiloride/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/drug effects , Hepatocyte Growth Factor/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Liver Neoplasms, Experimental/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Proto-Oncogene Proteins c-met/physiology , Receptor, IGF Type 1/physiology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/biosynthesis
16.
Ann Surg ; 241(5): 748-56; discussion 756-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15849510

ABSTRACT

OBJECTIVE: To determine whether insulinlike growth factor-I (IGF-I) and hepatocyte growth factor (HGF) cooperate to induce migration and invasion of human colorectal carcinoma (CRC) cells and whether the effects of IGF-I and/or HGF are mediated through activation of the urokinase plasminogen activator (uPA)/uPA receptor (uPAR) system, a central mediator of tumor-cell migration and invasion. SUMMARY BACKGROUND DATA: CRC cells must invade through the basement membrane of the colon and migrate to form metastases. CRC cells are known to overexpress IGF-I receptor (IGF-IR), c-Met, and uPAR, 3 cell-surface receptors known to mediate cell migration and invasion. We hypothesized that IGF-IR and c-Met cooperate to induce migration and invasion in CRC cells and that this signaling is dependent on uPAR. METHODS: KM12L4 human CRC cells were treated with IGF-I, HGF, or IGF-I + HGF in transwell migration and invasion chambers; cells that had migrated or invaded were counted. To determine the role of c-Met in IGF-I-induced migration and invasion, c-Met was inhibited by infection of cells with an adenovirus containing a c-Met ribozyme; transwell assays were then repeated. To determine the role of the uPA/uPAR system in IGF-I-induced CRC cell migration and invasion, transwell assays were repeated after pretreating cells with the uPA inhibitor amiloride or with neutralizing antibodies to uPA and uPAR. RESULTS: IGF-I and HGF, alone or in combination, increased cell migration and invasion. The c-Met ribozyme inhibited IGF-I- and HGF-mediated migration and invasion, indicating that c-Met is essential for these processes. uPA and uPAR inhibition blocked IGF-I- and HGF-mediated migration and invasion, suggesting that uPAR is downstream of IGF/IGF-IR and HGF/c-Met in the signaling pathways that mediate cell migration and invasion. CONCLUSIONS: IGF-I and HGF cooperate to induce migration and invasion of CRC cells, and c-Met and uPA/uPAR are required for IGF-I-mediated migration and invasion. In our in vitro model of CRC migration and invasion, uPA and uPAR appear to be downstream of IGF-IR and c-Met and are required for migration and invasion. Elucidation of the pathways that contribute to tumor progression and metastasis should provide a foundation for the rational development and use of targeted therapies for CRC.


Subject(s)
Cell Movement/physiology , Colorectal Neoplasms/pathology , Insulin-Like Growth Factor I/physiology , Neoplasm Invasiveness , Proto-Oncogene Proteins c-met/physiology , Receptors, Cell Surface/physiology , Cell Line, Tumor , Hepatocyte Growth Factor/physiology , Humans , Receptors, Urokinase Plasminogen Activator , Signal Transduction/physiology , Urokinase-Type Plasminogen Activator/physiology
17.
J Biol Chem ; 278(32): 29863-72, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12754207

ABSTRACT

Urokinase-type plasminogen activator (uPA) induces cell adhesion and chemotactic movement. uPA signaling requires its binding to uPA receptor (uPAR/CD87), but how glycosylphosphatidylinositol-anchored uPAR mediates signaling is unclear. uPAR is a ligand for several integrins (e.g. alpha 5 beta 1) and supports cell-cell interaction by binding to integrins on apposing cells (in trans). We studied whether binding of uPAR to alpha 5 beta 1 in cis is involved in adhesion and migration of Chinese hamster ovary cells in response to immobilized uPA. This process was temperature-sensitive and required mitogen-activated protein kinase activation. Anti-uPAR antibody or depletion of uPAR blocked, whereas overexpression of uPAR enhanced, cell adhesion to uPA. Adhesion to uPA was also blocked by deletion of the growth factor domain (GFD) of uPA and by anti-GFD antibody, whereas neither the isolated uPA kringle nor serine protease domain supported adhesion directly. Interestingly, anti-alpha 5 antibody, RGD peptide, and function-blocking mutations in alpha 5 beta 1 blocked adhesion to uPA. uPA-induced cell migration also required GFD, uPAR, and alpha 5 beta 1, but alpha 5 beta 1 alone did not support uPA-induced adhesion and migration. Thus, binding of uPA causes uPAR to act as a ligand for alpha 5 beta 1 to induce cell adhesion, intracellular signaling, and cell migration. We demonstrated that uPA induced RGD-dependent binding of uPAR to alpha 5 beta 1 in solution. These results suggest that uPA-induced adhesion and migration of Chinese hamster ovary cells occurs as a consequence of (a) uPA binding to uPAR through GFD, (b) the subsequent binding of a uPA.uPAR complex to alpha 5 beta 1 via uPAR, and (c) signal transduction through alpha 5 beta 1.


Subject(s)
Integrin alpha5beta1/physiology , Receptors, Cell Surface/metabolism , Signal Transduction , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cell Adhesion , Cell Movement , Cricetinae , DNA, Complementary/metabolism , Dimerization , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibronectins/metabolism , Glycosylphosphatidylinositols/metabolism , Ligands , MAP Kinase Signaling System , Models, Biological , Mutation , Oligopeptides/pharmacology , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptors, Urokinase Plasminogen Activator , Temperature
18.
Int J Cancer ; 104(4): 496-503, 2003 Apr 20.
Article in English | MEDLINE | ID: mdl-12584749

ABSTRACT

Integrin alpha(5)beta(1) is expressed on activated endothelial cells and plays a critical role in tumor angiogenesis. We hypothesized that a novel integrin alpha(5)beta(1) antagonist, ATN-161, would inhibit angiogenesis and growth of liver metastases in a murine model. We further hypothesized that combining ATN-161 with 5-fluorouracil (5-FU) chemotherapy would enhance the antineoplastic effect. Murine colon cancer cells (CT26) were injected into spleens of BALB/c mice to produce liver metastases. Four days thereafter, mice were given either ATN-161 (100 mg/kg, every 3rd day) or saline by intraperitoneal injection, with or without combination of continuous-infusion 5-FU (100 mg/kg/2 weeks), which was started on day 7. On day 20 after tumor cell inoculation, mice were killed and liver weights and number of liver metastases were determined. A follow-up study on survival was also conducted in which mice were randomized to receive ATN-161, 5-FU or ATN-161+5-FU. Combination therapy with ATN-161+5-FU significantly reduced tumor burden (liver weight) and number of liver metastases (p<0.02). Liver tumors in the ATN-161 and ATN-161+5-FU groups had significantly fewer microvessels (p<0.05) than tumors in the control or 5-FU-treated groups. Unlike treatment with either agent alone, ATN-161+5-FU significantly increased tumor cell apoptosis and decreased tumor cell proliferation (p<0.03) and improved overall survival (p<0.03, log-rank test). Targeting integrin alpha(5)beta(1) in combination with 5-FU infusion reduced liver metastases formation and improved survival in this colon cancer model. The enhancement of antineoplastic activity from the combination of anti-angiogenic therapy and chemotherapy may be a promising approach for treating metastatic colorectal cancer.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Colorectal Neoplasms/drug therapy , Fluorouracil/administration & dosage , Integrin alpha5beta1/antagonists & inhibitors , Liver Neoplasms/secondary , Oligopeptides/administration & dosage , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Integrin alpha5beta1/physiology , Liver Neoplasms/blood supply , Liver Neoplasms/prevention & control , Male , Mice , Mice, Inbred BALB C
19.
Cancer Res ; 62(18): 5344-50, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12235005

ABSTRACT

Histidine-proline-rich glycoprotein (HPRG) is an abundant multidomain plasma protein evolutionarily related to high-molecular-weight kininogen. The cleaved form of high-molecular-weight kininogen has recently been demonstrated to exhibit antiangiogenic activities in vitro (J. C. Zhang et al., FASEB J., 14: 2589-2600, 2000), mediated primarily through domain 5. HPRG contains a histidine-proline-rich (H/P) domain with sequence and functional similarities to HKa-D5. We hypothesized that HPRG may also have antiangiogenic properties, localized within its H/P domain. The H/P domain is highly conserved among species, and because rabbit H/P domain is more resistant to internal proteolytic cleavage than the human domain, the rabbit HPRG (rbHPRG) was primarily used to assess the antiangiogenic activity of HPRG. Rabbit HPRG inhibited human umbilical vein endothelial cell (HUVEC) tube formation stimulated by fibroblast growth factor-2 (FGF-2) or vascular endothelial growth factor on a Matrigel surface as well as cell proliferation of FGF-2 stimulated HUVECs. The antiangiogenic activity of rbHPRG was localized to the H/P domain by use of proteolytic fragments of rbHPRG and was further confirmed and characterized in two in vivo models of angiogenesis: the chorioallantoic membrane of the chick assay and the mouse Matrigel plug assay. Caspase-3 activation was observed in HUVECs stimulated with FGF-2 in the presence of rbHPRG, suggesting that apoptosis of activated endothelial cells may be one of the mechanisms underlying its antiangiogenic activity. Finally, the H/P domain of rbHPRG reduced tumor cell number when tumor cells were co-inoculated in the Matrigel plug assay. In conclusion, the H/P domain within HPRG induces the apoptosis of activated endothelial cells leading to potent antiangiogenic effects.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Proteins/pharmacology , Angiogenesis Inhibitors/physiology , Animals , Cell Division/drug effects , Chick Embryo , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Physiologic/drug effects , Plasminogen/pharmacology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Structure, Tertiary , Proteins/physiology , Rabbits , Repetitive Sequences, Amino Acid , Sequence Homology, Amino Acid
20.
J Biol Chem ; 277(34): 31056-61, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12063256

ABSTRACT

The 24-kDa form of basic fibroblast growth factor inhibits the migration of endothelial cells and mammary carcinoma cells while continuing to promote cell proliferation. This molecule consists of the 18-kDa fibroblast growth factor sequence plus an additional 55 amino acids at the amino-terminal end. Antibody neutralization studies suggested that the inhibition of migration is associated with these 55 amino acids, whereas the promotion of proliferation localizes to the 18-kDa domain. To determine whether 24kD basic fibroblast growth factor could be modified to eliminate its effect on cell proliferation but retain its inhibition of migration, portions of the carboxyl-terminal end of 24kD fibroblast growth factor were deleted, and the products were tested on MCF-7 and endothelial cells. A protein consisting of the 55 amino acids of the amino-terminal end and the first 31 amino acids of 18kD basic fibroblast growth factor (ATE+31) inhibited migration by 80% but did not promote cell growth. Arginine to alanine substitutions within the first 21 amino acids of the carboxyl-terminal end substantially reduced the efficacy of ATE+31, whereas substitutions in the remaining part of the molecule had no effect. Competition binding experiments showed that ATE+31 does not compete with 24kD basic fibroblast growth factor for binding to fibroblast growth factor receptor 1. In an in vivo matrigel plug assay, 150 nm ATE+31 peptide reduced angiogenesis by 80%. These studies demonstrate that the amino-terminal end of 24kD basic fibroblast growth factor is responsible for an activity that inhibits the migration rates of cultured cells as well as the angiogenic response in vivo.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Peptide Fragments/pharmacology , Cell Movement/drug effects , Female , Humans , Molecular Weight , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...