Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
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.
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
3.
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
4.
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
5.
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
6.
ACR Open Rheumatol ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010674

ABSTRACT

OBJECTIVE: Signal regulatory protein α (SIRPα) is found primarily on myeloid cells, including macrophages and neutrophils; binds to CD47; and regulates phagocytosis, antigen presentation, cellular fusion, cell proliferation, and migration. Therefore, SIRPα may be involved in the pathogenesis of autoimmune diseases, including systemic vasculitis. This study aimed to assess SIRPα expression in tissue samples from patients with vasculitis. METHODS: Immunohistochemical staining for SIRPα was performed on temporal artery (TA), kidney, and lung biopsy samples from patients with giant cell arteritis (GCA), patients with microscopic polyangiitis (MPA), patients with granulomatosis with polyangiitis (GPA), and patients without vasculitis. A score of SIRPα+ expression was calculated, derived from the percentages of monocytes, macrophages, and dendritic cells and neutrophils with different staining intensities in affected tissues. RESULTS: A total of 46 samples from patients with different vasculitides (GCA, MPA, and GPA) were included in the study. Tissue samples included TA samples from 15 patients with GCA; kidney samples from 11 and 9 patients with GPA and MPA, respectively; and lung samples from 11 patients with GPA. Most tissue samples from patients with active vasculitis (15 of 15 TA samples, 17 of 20 kidney samples, and 9 of 11 lung samples) showed SIRPα staining. SIRPα staining intensity was less in kidney samples compared to TA and lung samples. CONCLUSION: This study demonstrates high-level expression of SIRPα in macrophages and monocytes in affected tissue in systemic vasculitis. These findings provide a foundation for further studies exploring the role of the SIRPα-CD47 pathway in the pathogenesis of systemic vasculitis and the potential for the blockade of SIRPα and/or the depletion of SIRPα+ cells as treatment of systemic vasculitis.

7.
Blood Adv ; 7(16): 4258-4268, 2023 08 22.
Article in English | MEDLINE | ID: mdl-36897252

ABSTRACT

Dysregulated activation of the complement system is implicated in the onset or progression of several diseases. Most clinical-stage complement inhibitors target the inactive complement proteins present at high concentrations in plasma, which increases target-mediated drug disposition and necessitates high drug levels to sustain therapeutic inhibition. Furthermore, many efforts are aimed at inhibiting only terminal pathway activity, which leaves opsonin-mediated effector functions intact. We describe the discovery of SAR443809, a specific inhibitor of the alternative pathway C3/C5 convertase (C3bBb). SAR443809 selectively binds to the activated form of factor B (factor Bb) and inhibits alternative pathway activity by blocking the cleavage of C3, leaving the initiation of classical and lectin complement pathways unaffected. Ex vivo experiments with patient-derived paroxysmal nocturnal hemoglobinuria erythrocytes show that, although terminal pathway inhibition via C5 blockade can effectively inhibit hemolysis, proximal complement inhibition with SAR443809 inhibits both hemolysis and C3b deposition, abrogating the propensity for extravascular hemolysis. Finally, intravenous and subcutaneous administration of the antibody in nonhuman primates demonstrated sustained inhibition of complement activity for several weeks after injection. Overall, SAR443809 shows strong potential for treatment of alternative pathway-mediated disorders.


Subject(s)
Complement Factor B , Complement Pathway, Alternative , Animals , Complement Factor B/antagonists & inhibitors , Erythrocytes/drug effects , Hemolysis/drug effects , Complement C3-C5 Convertases/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Immune System Diseases/drug therapy , Immune System Diseases/enzymology , Humans , Macaca fascicularis , Antibodies/administration & dosage , Proteolysis/drug effects
8.
Adv Ther (Weinh) ; 5(6)2022 Jun.
Article in English | MEDLINE | ID: mdl-36211621

ABSTRACT

Chronic autoimmune demyelinating neuropathies are a group of rare neuromuscular disorders with complex, poorly characterized etiology. Here we describe a phenotypic, human-on-a-chip (HoaC) electrical conduction model of two rare autoimmune demyelinating neuropathies, chronic inflammatory demyelinating polyneuropathy (CIDP) and multifocal motor neuropathy (MMN), and explore the efficacy of TNT005, a monoclonal antibody inhibitor of the classical complement pathway. Patient sera was shown to contain anti-GM1 IgM and IgG antibodies capable of binding to human primary Schwann cells and induced pluripotent stem cell derived motoneurons. Patient autoantibody binding was sufficient to activate the classical complement pathway resulting in detection of C3b and C5b-9 deposits. A HoaC model, using a microelectrode array with directed axonal outgrowth over the electrodes treated with patient sera, exhibited reductions in motoneuron action potential frequency and conduction velocity. TNT005 rescued the serum-induced complement deposition and functional deficits while treatment with an isotype control antibody had no rescue effect. These data indicate that complement activation by CIDP and MMN patient serum is sufficient to mimic neurophysiological features of each disease and that complement inhibition with TNT005 was sufficient to rescue these pathological effects and provide efficacy data included in an investigational new drug application, demonstrating the model's translational potential.

10.
Proc Natl Acad Sci U S A ; 105(3): 1032-7, 2008 Jan 22.
Article in English | MEDLINE | ID: mdl-18195349

ABSTRACT

Glutamate is the primary excitatory neurotransmitter in the brain, and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) type glutamate receptors mediate most fast synaptic transmission. AMPA receptors are tetrameric assemblies composed from four possible subunits (GluR1-4). In hippocampal pyramidal cells, AMPA receptors are heteromeric receptors containing the GluR2 subunit and either GluR1 or GluR3. It is generally accepted that the trafficking of GluR1/GluR2 receptors to synapses requires activity, whereas GluR2/GluR3 receptors traffic constitutively. It has been suggested that the trafficking is governed by the cytoplasmic C termini of the subunits. Because the basis for this theory relied on the introduction of unnatural, homomeric, calcium-permeable AMPA receptors, we have used the GluR2(-/-) knock out mouse to determine whether the expression of mutated forms of GluR2 can rescue WT synaptic responses. We find that GluR2, lacking its entire C terminus, or a GluR2 chimera containing the C terminus of GluR1, is capable of trafficking to the synapse in the absence of activity. These findings suggest that the GluR2 C terminus is not required for GluR2 synaptic insertion.


Subject(s)
Receptors, AMPA/metabolism , Animals , Electrophysiology , Estriol/analogs & derivatives , Estriol/metabolism , Mice , Mice, Knockout , Patch-Clamp Techniques , Protein Subunits/metabolism , Protein Transport , Receptors, AMPA/deficiency , Receptors, AMPA/genetics
11.
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
12.
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
13.
J Neurosci ; 27(41): 11112-21, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17928453

ABSTRACT

We directly resolved discrete exocytic fusion events mediating insertion of AMPA-type glutamate receptors (AMPARs) to the somatodendritic surface of rat hippocampal pyramidal neurons, in slice and dissociated cultures, using protein tagging with a pH-sensitive GFP (green fluorescent protein) variant and rapid (10 frames/s) fluorescence microscopy. AMPAR-containing exocytic events occurred under basal culture conditions in both the cell body and dendrites; potentiating chemical stimuli produced an NMDA receptor-dependent increase in the frequency of individual exocytic events. The number of AMPARs inserted per exocytic event, estimated using single-molecule analysis, was quite uniform but individual events differed significantly in kinetic properties affecting the subsequent surface distribution of receptors. "Transient" events, from which AMPARs dispersed laterally immediately after surface insertion, generated a pronounced but short-lived (dissipating within approximately 1 s) increase in surface AMPAR fluorescence extending locally (2-5 microm) from the site of exocytosis. "Persistent" events, from which inserted AMPARs dispersed slowly (typically over 5-10 s), affected local surface receptor concentration to a much smaller degree. Both modes of exocytic insertion occurred throughout the dendritic shaft, but remarkably, neither mode of insertion was observed directly into synaptic spines. AMPARs entered spines preferentially from transient events occurring in the adjoining dendritic shaft, driven apparently by mass action and short-range lateral diffusion, and locally delivered AMPARs remained mostly in the mobile fraction. These results suggest a highly dynamic mechanism for both constitutive and activity-dependent surface delivery of AMPARs, mediated by kinetically distinct exocytic modes that differ in propensity to drive lateral entry of receptors to nearby synapses.


Subject(s)
Cell Membrane/physiology , Computer Systems , Excitatory Postsynaptic Potentials/physiology , Exocytosis/physiology , Receptors, AMPA/physiology , Animals , Animals, Newborn , Hippocampus/physiology , Organ Culture Techniques , Rats , Receptors, Cell Surface/physiology
14.
Clin Pharmacol Ther ; 104(4): 655-663, 2018 10.
Article in English | MEDLINE | ID: mdl-29737533

ABSTRACT

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody-mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement-mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first-in-human, double-blind, randomized, placebo-controlled, dose-escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration-effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 µg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Complement C1s/antagonists & inhibitors , Complement Inactivating Agents/administration & dosage , Complement Pathway, Classical/drug effects , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Austria , Complement C1s/immunology , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Middle Aged , Treatment Outcome , Young Adult
15.
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
16.
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
17.
J Neurosci ; 22(5): 1629-39, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11880493

ABSTRACT

Serotonin-gated ion channels (5-HT3) are members of the ligand-gated channel family, which includes channels that are opened directly by the neurotransmitter acetylcholine, GABA, glycine, or glutamate. Although there is general agreement that the second transmembrane domain (M2) lines the pore, the position of the gate in the M2 is less certain. Here, we used substituted cysteine accessibility method (SCAM) to provide new evidence for a centrally located gate that moves during channel activation. In the closed state, three cysteine substitutions, located on the extracellular side of M2, were modified by methanethiosulfonate (MTS) reagents. In contrast, 13 cysteine substitutions were modified in the open state with MTS reagents. The pattern of inhibition (every three to four substitutions) was consistent with an alpha helical structure for the middle and cytoplasmic segments of the M2 transmembrane domain. Unexpectedly, open-state modification of two amino acids in the center of M2 with three different MTS reagents prevented channels from fully closing in the absence of neurotransmitter. Our results are consistent with a model in which the central region of the M2 transmembrane domain is inaccessible in the closed state and moves during channel activation.


Subject(s)
Ethyl Methanesulfonate/analogs & derivatives , Ion Channel Gating/physiology , Receptors, Serotonin/metabolism , Serotonin/metabolism , Amino Acid Substitution , Animals , Cysteine/genetics , DNA, Complementary/genetics , DNA, Complementary/metabolism , Ethyl Methanesulfonate/pharmacology , Ion Channel Gating/drug effects , Ligands , Mesylates/pharmacology , Mice , Microinjections , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/metabolism , Patch-Clamp Techniques , Protein Conformation , Protein Structure, Tertiary/physiology , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT3 , Sequence Analysis, DNA , Structure-Activity Relationship , Xenopus
19.
Mol Immunol ; 54(3-4): 355-67, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23357789

ABSTRACT

Store operated calcium entry (SOCE) downstream of T cell receptor (TCR) activation in T lymphocytes has been shown to be mediated mainly through the Calcium Release Activated Calcium (CRAC) channel. Here, we compared the effects of a novel, potent and selective CRAC current inhibitor, 2,6-Difluoro-N-{5-[4-methyl-1-(5-methyl-thiazol-2-yl)-1,2,5,6-tetrahydro-pyridin-3-yl]-pyrazin-2-yl}-benzamide (RO2959), on T cell effector functions with that of a previously reported CRAC channel inhibitor, YM-58483, and a calcineurin inhibitor Cyclosporin A (CsA). Using both electrophysiological and calcium-based fluorescence measurements, we showed that RO2959 is a potent SOCE inhibitor that blocked an IP3-dependent current in CRAC-expressing RBL-2H3 cells and CHO cells stably expressing human Orai1 and Stim1, as well as SOCE in human primary CD4(+) T cells triggered by either TCR stimulation or thapsigargin treatment. Furthermore, we demonstrated that RO2959 completely inhibited cytokine production as well as T cell proliferation mediated by TCR stimulation or MLR (mixed lymphocyte reaction). Lastly, we showed by gene expression array analysis that RO2959 potently blocked TCR triggered gene expression and T cell functional pathways similar to CsA and another calcineurin inhibitor FK506. Thus, both from a functional and transcriptional level, our data provide evidence that RO2959 is a novel and selective CRAC current inhibitor that potently inhibits human T cell functions.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Anilides/pharmacology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CHO Cells , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Cell Proliferation/drug effects , Cricetinae , Cyclosporine/pharmacology , Cytokines/genetics , Cytokines/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Humans , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed/methods , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , Rats , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Stromal Interaction Molecule 1 , Tacrolimus/pharmacology , Thiadiazoles/pharmacology
20.
Br J Pharmacol ; 160(6): 1387-98, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20590629

ABSTRACT

BACKGROUND AND PURPOSE: Purinoceptors containing the P2X3 subunit (P2X3 homotrimeric and P2X2/3 heterotrimeric) are members of the P2X family of ion channels gated by ATP and may participate in primary afferent sensitization in a variety of pain-related diseases. The current work describes the in vitro pharmacological characteristics of AF-353, a novel, orally bioavailable, highly potent and selective P2X3/P2X2/3 receptor antagonist. EXPERIMENTAL APPROACH: The antagonistic potencies (pIC(50)) of AF-353 for rat and human P2X3 and human P2X2/3 receptors were determined using methods of radioligand binding, intracellular calcium flux and whole cell voltage-clamp electrophysiology. KEY RESULTS: The pIC(50) estimates for these receptors ranged from 7.3 to 8.5, while concentrations 300-fold higher had little or no effect on other P2X channels or on an assortment of receptors, enzymes and transporter proteins. In contrast to A-317491 and TNP-ATP, competition binding and intracellular calcium flux experiments suggested that AF-353 inhibits activation by ATP in a non-competitive fashion. Favourable pharmacokinetic parameters were observed in rat, with good oral bioavailability (%F = 32.9), reasonable half-life (t(1/2) = 1.63 h) and plasma-free fraction (98.2% protein bound). CONCLUSIONS AND IMPLICATIONS: The combination of a favourable pharmacokinetic profile with the antagonist potency and selectivity for P2X3 and P2X2/3 receptors suggests that AF-353 is an excellent in vivo tool compound for study of these channels in animal models and demonstrates the feasibility of identifying and optimizing molecules into potential clinical candidates, and, ultimately, into a novel class of therapeutics for the treatment of pain-related disorders.


Subject(s)
Adenosine Triphosphate/metabolism , Phenyl Ethers/pharmacology , Purinergic P2 Receptor Antagonists , Pyrimidines/pharmacology , Administration, Oral , Animals , Biological Availability , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Half-Life , Humans , Inhibitory Concentration 50 , Male , Patch-Clamp Techniques , Phenyl Ethers/administration & dosage , Phenyl Ethers/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Rats , Rats, Wistar , Receptors, Purinergic P2X2 , Receptors, Purinergic P2X3
SELECTION OF CITATIONS
SEARCH DETAIL