Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Clin Pharmacol Ther ; 116(1): 247-256, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38676311

ABSTRACT

B- and T-lymphocyte attenuator (BTLA; CD272) is an immunoglobulin superfamily member and part of a family of checkpoint inhibitory receptors that negatively regulate immune cell activation. The natural ligand for BTLA is herpes virus entry mediator (HVEM; TNFRSF14), and binding of HVEM to BTLA leads to attenuation of lymphocyte activation. In this study, we evaluated the role of BTLA and HVEM expression in the pathogenesis of systemic lupus erythematosus (SLE), a multisystem autoimmune disease. Peripheral blood mononuclear cells from healthy volunteers (N = 7) were evaluated by mass cytometry by time-of-flight to establish baseline expression of BTLA and HVEM on human lymphocytes compared with patients with SLE during a self-reported flare (N = 5). High levels of BTLA protein were observed on B cells, CD4+, and CD8+ T cells, and plasmacytoid dendritic cells in healthy participants. HVEM protein levels were lower in patients with SLE compared with healthy participants, while BTLA levels were similar between SLE and healthy groups. Correlations of BTLA-HVEM hub genes' expression with patient and disease characteristics were also analyzed using whole blood gene expression data from patients with SLE (N = 1,760) and compared with healthy participants (N = 60). HVEM, being one of the SLE-associated genes, showed an exceptionally strong negative association with disease activity. Several other genes in the BTLA-HVEM signaling network were strongly (negative or positive) correlated, while BTLA had a low association with disease activity. Collectively, these data provide a clinical rationale for targeting BTLA with an agonist in SLE patients with low HVEM expression.


Subject(s)
Lupus Erythematosus, Systemic , Receptors, Immunologic , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/blood , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Female , Adult , Male , Middle Aged , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Case-Control Studies
2.
Arthritis Rheumatol ; 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38556917

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is an autoimmune disease in which the joint lining or synovium becomes highly inflamed and majorly contributes to disease progression. Understanding pathogenic processes in RA synovium is critical for identifying therapeutic targets. We performed laser capture microscopy (LCM) followed by RNA sequencing (LCM-RNAseq) to study regional transcriptomes throughout RA synovium. METHODS: Synovial lining, sublining, and vessel samples were captured by LCM from seven patients with RA and seven patients with osteoarthritis (OA). RNAseq was performed on RNA extracted from captured tissue. Principal component analysis was performed on the sample set by disease state. Differential expression analysis was performed between disease states based on log2 fold change and q value parameters. Pathway analysis was performed using the Reactome Pathway Database on differentially expressed genes among disease states. Significantly enriched pathways in each synovial region were selected based on the false discovery rate. RESULTS: RA and OA transcriptomes were distinguishable by principal component analysis. Pairwise comparisons of synovial lining, sublining, and vessel samples between RA and OA revealed substantial differences in transcriptional patterns throughout the synovium. Hierarchical clustering of pathways based on significance revealed a pattern of association between biologic function and synovial topology. Analysis of pathways uniquely enriched in each region revealed distinct phenotypic abnormalities. As examples, RA lining samples were marked by anomalous immune cell signaling, RA sublining samples were marked by aberrant cell cycle, and RA vessel samples were marked by alterations in heme scavenging. CONCLUSION: LCM-RNAseq confirms reported transcriptional differences between the RA synovium and the OA synovium and provides evidence supporting a relationship between synovial topology and molecular anomalies in RA.

3.
JCI Insight ; 8(13)2023 07 10.
Article in English | MEDLINE | ID: mdl-37427592

ABSTRACT

B cells contribute to multiple aspects of autoimmune disorders, and B cell-targeting therapies, including B cell depletion, have been proven to be efficacious in treatment of multiple autoimmune diseases. However, the development of novel therapies targeting B cells with higher efficacy and a nondepleting mechanism of action is highly desirable. Here we describe a nondepleting, high-affinity anti-human CD19 antibody LY3541860 that exhibits potent B cell inhibitory activities. LY3541860 inhibits B cell activation, proliferation, and differentiation of primary human B cells with high potency. LY3541860 also inhibits human B cell activities in vivo in humanized mice. Similarly, our potent anti-mCD19 antibody also demonstrates improved efficacy over CD20 B cell depletion therapy in multiple B cell-dependent autoimmune disease models. Our data indicate that anti-CD19 antibody is a highly potent B cell inhibitor that may have potential to demonstrate improved efficacy over currently available B cell-targeting therapies in treatment of autoimmune conditions without causing B cell depletion.


Subject(s)
Autoimmune Diseases , B-Lymphocytes , Mice , Animals , Antigens, CD19 , Autoimmune Diseases/drug therapy
4.
Sci Rep ; 11(1): 18671, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548595

ABSTRACT

The selective Bruton tyrosine kinase (BTK) inhibitor poseltinib has been shown to inhibit the BCR signal transduction pathway and cytokine production in B cells (Park et al. Arthritis Res. Ther. 18, 91, https://doi.org/10.1186/s13075-016-0988-z , 2016). This study describes the translation of nonclinical research studies to a phase I clinical trial in healthy volunteers in which pharmacokinetics (PKs) and pharmacodynamics (PDs) were evaluated for dose determination. The BTK protein kinase inhibitory effects of poseltinib in human peripheral blood mononuclear cells (PBMCs) and in rats with collagen-induced arthritis (CIA) were evaluated. High-dimensional phosphorylation analysis was conducted on human immune cells such as B cells, CD8 + memory cells, CD4 + memory cells, NK cells, neutrophils, and monocytes, to map the impact of poseltinib on BTK/PLC and AKT signaling pathways. PK and PD profiles were evaluated in a first-in-human study in healthy donors, and a PK/PD model was established based on BTK occupancy. Poseltinib bound to the BTK protein and modulated BTK phosphorylation in human PBMCs. High-dimensional phosphorylation analysis of 94 nodes showed that poseltinib had the highest impact on anti-IgM + CD40L stimulated B cells, however, lower impacts on anti-CD3/CD-28 stimulated T cells, IL-2 stimulated CD4 + T cells and NK cells, M-CSF stimulated monocytes, or LPS-induced granulocytes. In anti-IgM + CD40L stimulated B cells, poseltinib inhibited the phosphorylation of BTK, AKT, and PLCγ2. Moreover, poseltinib dose dependently improved arthritis disease severity in CIA rat model. In a clinical phase I trial for healthy volunteers, poseltinib exhibited dose-dependent and persistent BTK occupancy in PBMCs of all poseltinib-administrated patients in the study. More than 80% of BTK occupancy at 40 mg dosing was maintained for up to 48 h after the first dose. A first-in-human healthy volunteer study of poseltinib established target engagement with circulating BTK protein. Desirable PK and PD properties were observed, and a modeling approach was used for rational dose selection for subsequent trials. Poseltinib was confirmed as a potential BTK inhibitor for the treatment of autoimmune diseases.Trial registration: This article includes the results of a clinical intervention on human participants [NCT01765478].


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aniline Compounds/pharmacology , Models, Biological , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacokinetics , Animals , Clinical Trials, Phase II as Topic , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Molecular Docking Simulation , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Rats
5.
Rheumatology (Oxford) ; 60(11): 5390-5396, 2021 11 03.
Article in English | MEDLINE | ID: mdl-33580248

ABSTRACT

OBJECTIVES: Heterogeneity of SLE patients in clinical trials remains a challenge for developing new therapies. This study used a combinatorial analysis of four molecular biomarkers to define key sources of heterogeneity. METHODS: Combinations of IFN (high/low), anti-dsDNA (+/-) and C3 and C4 (low/normal) were used to subset n = 1747 patients from two randomized phase III trials. A dichotomous classification scheme defined SLE (+) as: IFN (high), anti-dsDNA (+), C3 (low) and/or C4 (low). SLE (-) required all of the following: IFN (low), anti-dsDNA (-), C3 (normal) and C4 (normal). Additional analyses subset the data further by IFN, anti-dsDNA and complement. RESULTS: The trials enrolled n = 2262 patients of which n = 1747 patients had data for IFN, anti-dsDNA, C3 and C4 at baseline. There were n = 247 patients in the SLE (-) population and n = 1500 patients in the SLE (+) population. The SLE (-) population had more mucocutaneous and musculoskeletal disease at baseline, while SLE (+) had more haematological, renal and vascular involvement. There was lower concomitant medication use in the SLE (-) population for corticosteroids and immunosuppressants, except for MTX. Time to severe flare was significantly longer in SLE (-) vs SLE (+) (P < 0.0001) and SRI-4 response rate was significantly lower in SLE (-) vs SLE (+) (P = 0.00016). The USA had more SLE (-) patients (22%) than Mexico/Central America/South America (10%), Europe (7%) and the rest of the world (5%). CONCLUSION: Combinatorial analysis of four molecular biomarkers revealed subsets of SLE patients that discriminated by disease manifestations, concomitant medication use, geography, time to severe flare and SRI-4 response. These data may be useful for designing clinical trials and identifying subsets of patients for analysis. Rheumatology key messages SLE patients from a P3 trial were categorized by IFN, anti-dsDNA, C3 and C4 status. Patients lacking molecular markers of SLE distinguished from biomarker positive patients on multiple clinical parameters. Biomarker negative patients have distinct disease characteristics that may impact clinical trial outcomes.


Subject(s)
Antibodies, Antinuclear/blood , Complement C3/metabolism , Complement C4/metabolism , Interferons/blood , Lupus Erythematosus, Systemic/diagnosis , Humans , Lupus Erythematosus, Systemic/blood , Phenotype
6.
Lancet ; 392(10143): 222-231, 2018 07 21.
Article in English | MEDLINE | ID: mdl-30043749

ABSTRACT

BACKGROUND: Patients with systemic lupus erythematosus have substantial unmet medical need. Baricitinib is an oral selective Janus kinase (JAK)1 and JAK2 inhibitor that we hypothesised might have therapeutic benefit in patients with systemic lupus erythematosus. METHODS: In this double-blind, multicentre, randomised, placebo-controlled, 24-week phase 2 study, patients were recruited from 78 centres in 11 countries. Eligible patients were aged 18 years or older, had a diagnosis of systemic lupus erythematosus, and had active disease involving skin or joints. We randomly assigned patients (1:1:1) to receive once-daily baricitinib 2 mg, baricitinib 4 mg, or placebo for 24 weeks. The primary endpoint was the proportion of patients achieving resolution of arthritis or rash at week 24, as defined by Systemic Lupus Erythematosus Disease Activity Index-2000 (SLEDAI-2K). Efficacy and safety analyses included all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT02708095. FINDINGS: Between March 24, 2016, and April 27, 2017, 314 patients were randomly assigned to receive placebo (n=105), baricitinib 2 mg (n=105), or baricitinib 4 mg (n=104). At week 24, resolution of SLEDAI-2K arthritis or rash was achieved by 70 (67%) of 104 patients receiving baricitinib 4 mg (odds ratio [OR] vs placebo 1·8, 95% CI 1·0-3·3; p=0·0414) and 61 (58%) of 105 patients receiving baricitinib 2 mg (OR 1·3, 0·7-2·3; p=0·39). Adverse events were reported in 68 (65%) patients in the placebo group, 75 (71%) patients in the baricitinib 2 mg group, and 76 (73%) patients in the baricitinib 4 mg group. Serious adverse events were reported in ten (10%) patients receiving baricitinib 4 mg, 11 (10%) receiving baricitinib 2 mg, and five (5%) receiving placebo; no deaths were reported. Serious infections were reported in six (6%) patients with baricitinib 4 mg, two (2%) with baricitinib 2 mg, and one (1%) with placebo. INTERPRETATION: The baricitinib 4 mg dose, but not the 2 mg dose, significantly improved the signs and symptoms of active systemic lupus erythematosus in patients who were not adequately controlled despite standard of care therapy, with a safety profile consistent with previous studies of baricitinib. This study provides the foundation for future phase 3 trials of JAK1/2 inhibition with baricitinib as a new potential oral therapy for systemic lupus erythematosus. FUNDING: Eli Lilly and Company.


Subject(s)
Arthritis/drug therapy , Azetidines/administration & dosage , Exanthema/drug therapy , Janus Kinase Inhibitors/administration & dosage , Lupus Erythematosus, Systemic/drug therapy , Sulfonamides/administration & dosage , Administration, Oral , Adult , Azetidines/adverse effects , Cholesterol/blood , Cholesterol, HDL/blood , Creatine Kinase/blood , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Infections/epidemiology , Janus Kinase Inhibitors/adverse effects , Male , Middle Aged , Platelet Count , Purines , Pyrazoles , Sulfonamides/adverse effects
7.
Rheumatology (Oxford) ; 57(1): 125-133, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29045736

ABSTRACT

Objective: The SLE Responder Index (SRI) is a composite endpoint used in SLE trials. This investigation examined the clinical trial elements that drive response measured by the SRI. Methods: Analyses are based on data from two phase 3 trials (n = 2262) that evaluated the impact of an anti-B-cell activating factor antibody on disease activity using SRI-5 as the primary endpoint (ClinicalTrials.gov NCT01196091 and NCT01205438). Results: The SRI-5 response rate at week 52 for all patients was 32.8%. Non-response due to a lack of SLEDAI improvement, concomitant medication non-compliance or dropout was 31, 16.5 and 19.1%, respectively. Non-response due to deterioration in BILAG or Physician's Global Assessment after SLEDAI improvement, concomitant medication compliance and trial completion was 0.5%. Disease activity in three SLEDAI organ systems was highly prevalent at baseline: mucocutaneous, 90.6%; musculoskeletal, 82.9%; and immunologic, 71.6%. Disease activity in each of the other organ systems was <11% of patients. Four clinical manifestations were highly prevalent at baseline: arthritis, 82.6%; rash, 69.2%; alopecia, 58.2%; and mucosal ulcer, 32.5%. The combined prevalence of renal, vascular and CNS disease at baseline was 17.6%; these patients had high SRI-5 response rates. Adjustments to corticosteroids were allowed during the first 24 weeks. Increases in corticosteroids above 2.5 mg/day were observed in 16.2% of placebo patients over the first 24 weeks after randomization. Conclusion: The primary drivers of SRI-5 response were SLEDAI improvement, concomitant medication adherence and trial completion. Arthritis, rash, alopecia and mucosal ulcer were the most prevalent clinical manifestations at baseline. Corticosteroid increases and rare, highly weighted disease manifestations in SLEDAI can confound the SRI signal.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Adrenal Cortex Hormones/therapeutic use , B-Cell Activating Factor/antagonists & inhibitors , Clinical Trials as Topic , Clinical Trials, Phase III as Topic , Drug Therapy, Combination , Humans , Lupus Erythematosus, Systemic/physiopathology , Medication Adherence , Randomized Controlled Trials as Topic , Treatment Outcome
8.
Arthritis Rheumatol ; 69(3): 643-654, 2017 03.
Article in English | MEDLINE | ID: mdl-27723281

ABSTRACT

OBJECTIVE: To characterize baseline gene expression and pharmacodynamically induced changes in whole blood gene expression in 1,760 systemic lupus erythematosus (SLE) patients from 2 phase III, 52-week, randomized, placebo-controlled, double-blind studies in which patients were treated with the BAFF-blocking IgG4 monoclonal antibody tabalumab. METHODS: Patient samples were obtained from SLE patients from the ILLUMINATE-1 and ILLUMINATE-2 studies, and control samples were obtained from healthy donors. Blood was collected in Tempus tubes at baseline, week 16, and week 52. RNA was analyzed using Affymetrix Human Transcriptome Array 2.0 and NanoString. RESULTS: At baseline, expression of the interferon (IFN) response gene was elevated in patients compared with controls, with 75% of patients being positive for this IFN response gene signature. There was, however, substantial heterogeneity of IFN response gene expression and complex relationships among gene networks. The IFN response gene signature was a predictor of time to disease flare, independent of anti-double-stranded DNA (anti-dsDNA) antibody and C3 and C4 levels, and overall disease activity. Pharmacodynamically induced changes in gene expression following tabalumab treatment were extensive, occurring predominantly in B cell-related and immunoglobulin genes, and were consistent with other pharmacodynamic changes including anti-dsDNA antibody, C3, and immunoglobulin levels. CONCLUSION: SLE patients demonstrated increased expression of an IFN response gene signature (75% of patients had an elevated IFN response gene signature) at baseline in ILLUMINATE-1 and ILLUMINATE-2. Substantial heterogeneity of gene expression was detected among individual patients and in gene networks. The IFN response gene signature was an independent risk factor for future disease flares. Pharmacodynamic changes in gene expression were consistent with the mechanism of BAFF blockade by tabalumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B-Cell Activating Factor/antagonists & inhibitors , Gene Expression/genetics , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Gene Expression/drug effects , Humans , Male , Middle Aged , Young Adult
9.
Arthritis Rheum ; 63(3): 622-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21360491

ABSTRACT

OBJECTIVE: To assess the safety of rituximab in combination with a tumor necrosis factor (TNF) inhibitor and methotrexate (MTX) in patients with rheumatoid arthritis (RA). METHODS: Adult patients with active RA (≥ 5 swollen and ≥ 5 tender joints) receiving a stable dose of MTX (10-25 mg/week) and stable dose of TNF inhibitor (etanercept or adalimumab) for ≥ 12 weeks were randomized 2:1 to receive one course of rituximab or placebo, given intravenously at a dose of 2 × 500 mg. The primary end point was the proportion of patients developing ≥ 1 serious infection through week 24. RESULTS: Fifty-one patients were treated with either rituximab or placebo in combination with background MTX and a TNF inhibitor. Baseline characteristics were generally balanced between groups, except for corticosteroid usage (36% in the rituximab arm versus 17% in the placebo arm). A serious infection (pneumonia) was observed in 1 patient (3%) in the rituximab group after 14.4 patient-years of exposure (6.95 events per 100 patient-years, 95% confidence interval 0.98-49.35), compared with none in the placebo group at week 24. Infections were reported in 18 patients (55%) and 11 patients (61%) in the rituximab and placebo groups, respectively. Grade 3 infections were reported in 3 patients (9%) receiving rituximab and in none of the patients receiving placebo. No grade 4 infections were observed, nor were there any opportunistic, fungal, or tuberculosis infections. Serious adverse events (SAEs) were reported in 2 rituximab-treated patients (pneumonia and coronary artery occlusion), whereas there were no SAEs reported in placebo-treated patients. At week 24, the percentage of patients achieving an American College of Rheumatology 20% (ACR20) improvement response was 30% in the rituximab group compared with 17% in the placebo group, and ACR50 responses were achieved by 12% and 6% of patients, respectively. CONCLUSION: The preliminary safety profile of rituximab in combination with a TNF inhibitor and MTX was consistent with the safety profile of rituximab in combination with MTX in other RA trials without a TNF inhibitor, with no new safety signals observed. SAEs were numerically more frequent in the rituximab group, and there was no clear evidence of an efficacy advantage in patients receiving rituximab in combination with a TNF inhibitor and MTX.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/adverse effects , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/drug therapy , Immunoglobulin G/adverse effects , Methotrexate/adverse effects , Acute Disease , Adalimumab , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antirheumatic Agents/administration & dosage , Double-Blind Method , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Etanercept , Female , Humans , Immunoglobulin G/administration & dosage , Infusions, Intravenous , Male , Methotrexate/administration & dosage , Middle Aged , Placebos , Receptors, Tumor Necrosis Factor/administration & dosage , Rituximab , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Young Adult
10.
Arthritis Rheum ; 58(8): 2470-80, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18668592

ABSTRACT

OBJECTIVE: To investigate whether treatment with abetimus delays renal flare in patients with lupus nephritis. Secondary objectives included evaluation of the effect of abetimus on C3 levels, anti-double-stranded DNA (anti-dsDNA) antibody levels, use of high-dose corticosteroids and/or cyclophosphamide, and major systemic lupus erythematosus (SLE) flare. METHODS: We conducted a randomized, placebo-controlled study of treatment with abetimus at 100 mg/week for up to 22 months in SLE patients. Three hundred seventeen patients with a history of renal flare and anti-dsDNA levels >15 IU/ml were randomized to a treatment group (158 abetimus, 159 placebo); 298 (94%) were enrolled in the intent-to-treat (ITT) population (145 abetimus, 153 placebo), based on the presence of high-affinity antibodies for the oligonucleotide epitope of abetimus at baseline screening. RESULTS: Abetimus did not significantly prolong time to renal flare, time to initiation of high-dose corticosteroid and/or cyclophosphamide treatment, or time to major SLE flare. However, there were 25% fewer renal flares in the abetimus group compared with the placebo group (17 of 145 abetimus-treated patients [12%] versus 24 of 153 placebo-treated patients [16%]). Abetimus treatment decreased anti-dsDNA antibody levels (P < 0.0001), and reductions in anti-dsDNA levels were associated with increases in C3 levels (P < 0.0001). More patients in the abetimus group experienced > or =50% reductions in proteinuria at 1 year, compared with the placebo group (nominal P = 0.047). Trends toward reduced rates of renal flare and major SLE flare were noted in patients treated with abetimus who had impaired renal function at baseline. Treatment with abetimus for up to 22 months was well tolerated. CONCLUSION: Abetimus at 100 mg/week significantly reduced anti-dsDNA antibody levels but did not significantly prolong time to renal flare when compared with placebo. Multiple positive trends in renal end points were observed in the abetimus treatment group.


Subject(s)
Lupus Erythematosus, Systemic/complications , Lupus Nephritis/etiology , Lupus Nephritis/prevention & control , Oligonucleotides/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Adult , Antibodies, Anti-Idiotypic/blood , Complement C3/metabolism , Cyclophosphamide/therapeutic use , DNA/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/drug therapy , Male , Middle Aged , Oligonucleotides/adverse effects , Quality of Life , Treatment Outcome
11.
J Clin Pharmacol ; 48(8): 909-18, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511650

ABSTRACT

Abetimus sodium is an oligonucleotide-based investigational drug designed to treat patients with lupus nephritis by specifically reducing anti-double-stranded DNA antibody levels. The safety and pharmacokinetics of abetimus were evaluated in 24 healthy volunteers at intravenous doses of 600 mg, 1200 mg, and 2400 mg. The mean half-life ranged from 0.8 to 1.5 hours. Maximum exposure assessed by maximum observed plasma concentration was dose proportional. Total exposure assessed by area under the plasma concentration-time curve was dose proportional between 1200-mg and 2400-mg doses and greater than proportionate between the 600-mg and 1200-mg doses. Abetimus was well tolerated in all dose groups, with adverse events observed in 33.3% (2/6) of placebo subjects and 16.7% (3/18) subjects receiving abetimus. No clinically significant effects were observed on laboratory values, vital signs, or electrocardiogram, with the exception of a transient, dose-dependent, prolongation in activated partial thromboplastin time. In vitro coagulation studies suggested that the effect on activated partial thromboplastin time was attributable to a nonspecific interaction rather than specific factor depletion. Exposure to abetimus at intravenous doses of 600 mg, 1200 mg, and 2400 mg was well tolerated. Total exposure assessed by area under the plasma concentration-time curve was greater than dose proportional across the dose range of 600 mg to 2400 mg.


Subject(s)
Oligonucleotides/adverse effects , Oligonucleotides/pharmacokinetics , Adolescent , Adult , Area Under Curve , Blood Coagulation/drug effects , Double-Blind Method , Female , Half-Life , Humans , Injections, Intravenous , Male , Oligonucleotides/administration & dosage , Partial Thromboplastin Time , Prothrombin Time
12.
J Pharmacol Exp Ther ; 324(2): 867-75, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17993604

ABSTRACT

Semicarbazide-sensitive amine oxidase (SSAO, amine oxidase, copper-containing 3, and vascular adhesion protein-1) is a copper-containing enzyme that catalyzes the oxidative deamination of primary amines to an aldehyde, ammonia, and hydrogen peroxide. SSAO is also involved in leukocyte migration to sites of inflammation, and the enzymatic activity of SSAO is essential to this role. Thus, inhibition of SSAO enzyme activity represents a target for the development of small molecule anti-inflammatory compounds. Here, we have characterized the novel SSAO inhibitor, Z-3-fluoro-2-(4-methoxybenzyl)allylamine hydrochloride (LJP 1586), and assessed its anti-inflammatory activity. LJP 1586 is a potent inhibitor of rodent and human SSAO activity, with IC(50) values between 4 and 43 nM. The selectivity of LJP 1586 was confirmed with a broad panel of receptors and enzymes that included the monoamine oxidases A and B. Oral administration of LJP 1586 resulted in complete inhibition of rat lung SSAO, with an ED(50) between 0.1 and 1 mg/kg, and a pharmacodynamic half-life of greater than 24 h. In a mouse model of inflammatory leukocyte trafficking oral dosing with LJP 1586 resulted in significant dose-dependent inhibition of neutrophil accumulation, with an effect comparable to that of anti-leukocyte function-associated antigen-1 antibody. In a rat model of LPS-induced lung inflammation, administration of 10 mg/kg LJP 1586 resulted in a 55% significant reduction in transmigrated cells recovered by bronchoalveolar lavage. The results demonstrate that a selective, orally active small molecule inhibitor of SSAO is an effective anti-inflammatory compound in vivo and provide further support for SSAO as a therapeutic anti-inflammatory target.


Subject(s)
Allylamine/analogs & derivatives , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Amines/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Allylamine/chemistry , Allylamine/pharmacology , Allylamine/therapeutic use , Amine Oxidase (Copper-Containing)/metabolism , Amines/chemistry , Amines/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , CHO Cells , Cricetinae , Cricetulus , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Inflammation/drug therapy , Inflammation/enzymology , Lung/drug effects , Lung/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
13.
Arthritis Rheum ; 54(8): 2558-67, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16868978

ABSTRACT

OBJECTIVE: Results of previous studies suggest that anti-beta2-glycoprotein I (anti-beta2GPI) antibodies in complex with beta2GPI activate platelets in a dysregulated manner, potentially contributing to the prothrombotic tendency associated with the antiphospholipid syndrome (APS). We undertook this study to investigate the possible contribution of the GPIb-IX-V receptor to platelet activation mediated by the anti-beta2GPI antibody-beta2GPI complex. METHODS: In vitro methods were used in the present study. The interaction between beta2GPI and the GPIbalpha subunit of the GPIb-IX-V receptor was delineated using direct binding and competitive inhibition assays. The interaction between the anti-beta2GPI antibody-beta2GPI complex and platelets was studied using a novel method in which the Fc portion of the antibody was immobilized using protein A coated onto a microtiter plate. Platelet activation was assessed by two methods; one involved measuring thromboxane B2 production and the other involved assessment of the activation of the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta intracellular signaling pathway. The contribution of the GPIbalpha receptor to platelet activation induced by the anti-beta2GPI antibody-beta2GPI complex was assessed by observing the influence of 2 anti-GPIbalpha antibodies (AK2 and SZ2) directed against distinct epitopes. RESULTS: This study showed that beta(2)GPI could bind to the GPIbalpha receptor. The anti-beta2GPI antibody-beta2GPI complex was able to activate platelets, and this effect was inhibited by anti-GPIbalpha antibody directed against epitope Leu-36-Gln-59, but not by anti-GPIbalpha antibody directed against residues Tyr-276-Glu-282. CONCLUSION: Our findings show that inappropriate platelet activation by the anti-beta2GPI antibody-beta2GPI complex via the GPIbalpha receptor may contribute to the prothrombotic tendency associated with APS.


Subject(s)
Blood Platelets/immunology , Glycoproteins/immunology , Platelet Activation/immunology , Platelet Glycoprotein GPIb-IX Complex/immunology , Antibodies, Blocking/pharmacology , Autoantibodies/immunology , Binding Sites, Antibody/immunology , Blood Platelets/drug effects , Blood Platelets/metabolism , Glycoproteins/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thromboxane B2/metabolism , beta 2-Glycoprotein I
14.
J Med Chem ; 49(7): 2166-73, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16570912

ABSTRACT

In an attempt to examine the effect of inhibition of semicarbazide-sensitive amine oxidase (SSAO; EC 1.4.3.6, also known as VAP-1) as a novel anti-inflammatory target, the structure/mechanism based design and synthesis of a series of novel hydrazino-containing small molecules are described. The in vitro biological results show that compounds 4a,c are highly potent SSAO inhibitors with notable selectivity toward SSAO over monoamine oxidases A and B (MAO-A and MAO-B). SAR studies based on compound 4c were performed, and the results are discussed. The most potent and selective compound, 4a (IC(50) = 2 nM), is an orally active, competitive, and apparently irreversible inhibitor of SSAO that is effective at reducing disease incidence and severity in an in vivo animal disease model of multiple sclerosis.


Subject(s)
Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Hydrazines/chemical synthesis , Acute Disease , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Hydrazines/chemistry , Hydrazines/pharmacology , In Vitro Techniques , Lung/drug effects , Lung/enzymology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Rats , Structure-Activity Relationship
15.
J Pharmacol Exp Ther ; 317(1): 19-29, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16339390

ABSTRACT

Endothelial vascular adhesion protein-1 (VAP-1) facilitates leukocyte adhesion and infiltration. This relates partly to the function of VAP-1 as a semicarbazide-sensitive amine oxidase (SSAO). We examined the effects of VAP-1/SSAO inhibition [via LJP-1207 (N'-(2-phenyl-allyl)-hydrazine hydrochloride)] on pial venular leukocyte adhesion and infiltration (at 2-10 h of reperfusion) and neuropathology (at 72 h of reperfusion) after transient forebrain ischemia (TFI). A model associated with increased postischemic inflammation was used-i.e., diabetic ovariectomized (OVX) female rats given chronic estrogen replacement therapy (ERT). We compared rats treated, either at the onset or at 6 h of reperfusion, with saline or LJP-1207. Additional rats, rendered neutropenic 24 h before TFI, were studied. In saline-treated controls, intravascular accumulation of adherent leukocytes gradually increased, reaching 15 to 20% of the venular area, at which point neutrophil infiltration commenced (at approximately 6 h). In the rats given LJP-1207 at the onset of reperfusion, limited neutrophil adhesion ( approximately 5% maximum) and no infiltration were observed. These results generally paralleled those in neutropenic rats. In rats treated at 6 h of reperfusion, the pattern of neutrophil adhesion was similar to that of the saline-treated group up to 6 h, but further infiltration was essentially prevented. Neurologic outcomes and histopathology were similar to one another in the LJP-1207-treated and neutropenic groups and significantly improved over those in saline-treated controls. Thus, VAP-1-mediated post-TFI leukocyte adhesion/infiltration in diabetic OVX females given chronic ERT contributes substantially to neuropathology. One implication is that specifically preventing leukocyte infiltration provides a substantial measure of neuroprotection. This could explain the finding of LJP-1207 having at least a 6-h therapeutic window in this model.


Subject(s)
Amine Oxidase (Copper-Containing)/physiology , Brain Ischemia/pathology , Brain/pathology , Cell Adhesion Molecules/physiology , Diabetes Mellitus, Experimental/pathology , Estradiol/pharmacology , Inflammation/pathology , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Female , Hydrazines/pharmacology , Inflammation/complications , Inflammation/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Ovariectomy , Rats , Rats, Sprague-Dawley
16.
J Pharmacol Exp Ther ; 315(2): 553-62, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16081681

ABSTRACT

Human semicarbazide-sensitive amine oxidase (SSAO) or vascular adhesion protein-1 (VAP-1) is a copper-containing amine oxidase (AOC3, EC 1.4.3.6) that has both enzymatic and adhesive function. SSAO catalyzes the oxidative deamination of primary amines, resulting in the formation of the corresponding aldehyde and release of hydrogen peroxide and ammonia. Membrane-bound SSAO is an inflammation-inducible endothelial cell adhesion molecule that mediates the interaction between leukocytes and activated endothelial cells in inflamed vessels. Both the direct adhesive and enzymatic functions seem to be involved in the adhesion cascade. LJP 1207 [N'-(2-phenyl-allyl)-hydrazine hydrochloride] is a potent (human SSAO IC(50) = 17 nM), selective, and orally available SSAO inhibitor that blocks both the enzymatic and adhesion functions of SSAO/VAP-1. In a mouse model of ulcerative colitis, LJP 1207 significantly reduces mortality, loss of body weight, and colonic cytokine levels. Quantitative histopathological assessment of colitis activity in this model showed a highly significant suppression of inflammation, injury, and ulceration scores in the animals treated with the SSAO/VAP-1 inhibitor. LJP 1207 also reduced serum levels of tumor necrosis factor-alpha and interleukin 6 in lipopolysaccharide (LPS)-challenged mice and prolonged survival post-LPS-induced endotoxemia. Therapeutic and prophylactic administration of LJP 1207 in the rat carrageenan footpad model also markedly inhibited swelling and inflammation. Overall, the data suggest that small molecule SSAO/VAP-1 inhibitors may provide clinical benefit in the treatment of acute and chronic inflammatory diseases.


Subject(s)
Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Anti-Inflammatory Agents , Hydrazines/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Amine Oxidase (Copper-Containing)/genetics , Animals , Carrageenan , Cell Adhesion/drug effects , Cloning, Molecular , Colitis/chemically induced , Colitis/drug therapy , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Edema/chemically induced , Edema/drug therapy , Endothelial Cells/drug effects , Endotoxemia/chemically induced , Endotoxemia/drug therapy , Female , Inflammation/drug therapy , Inflammation/pathology , Male , Mice , Mice, Inbred Strains , Oxazolone , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
17.
Arthritis Rheum ; 52(4): 1129-37, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15818711

ABSTRACT

OBJECTIVE: To examine the relationship between changes in anti-double-stranded DNA (anti-dsDNA) antibody levels and the risk of renal flare in patients with systemic lupus erythematosus (SLE), using data from 2 randomized, controlled trials. METHODS: Analyses were based on 487 patients with SLE and a history of lupus nephritis who had an anti-dsDNA antibody titer >/=15 IU/ml at baseline, as measured by Farr assay. Results are presented for the combined population of patients, the placebo arms, and the drug treatment arms in which a dsDNA-based bioconjugate (abetimus sodium; LJP 394) was used. RESULTS: Changes in anti-dsDNA antibody levels were inversely correlated with changes in the C3 level (P < 0.0001 in both trials). Cox proportional hazards regression models showed that changes in anti-dsDNA antibody levels correlated with the risk of renal flare. The models predicted that a point estimate of a 50% reduction in anti-dsDNA antibody levels is associated with a 52% reduction (95% confidence interval [95% CI] 26-68%, nominal P = 0.0007) and a 53% reduction (95% CI 33-69%, nominal P < 0.0001) in the risk of renal flare in the 2 trials, respectively. In the 2 trials, the incidence of renal flare was lower in patients with sustained reductions in anti-dsDNA antibodies (3.0% and 4.1%, respectively) than in patients with stable or increasing antibody levels (21.3% and 20.3%, respectively). CONCLUSION: Changes in anti-dsDNA antibody levels were directly correlated with the risk of renal flare and inversely correlated with changes in the C3 level. Reducing anti-dsDNA antibody levels may represent a therapeutic objective in SLE patients with lupus nephritis, because it is associated with a reduced risk of renal flare.


Subject(s)
Antibodies, Antinuclear/analysis , DNA/immunology , Lupus Nephritis/immunology , Adjuvants, Immunologic/therapeutic use , Adolescent , Adult , Aged , Creatinine/blood , DNA/drug effects , Female , Humans , Lupus Nephritis/pathology , Lupus Nephritis/prevention & control , Male , Middle Aged , Oligonucleotides/therapeutic use , Proteinuria , Retrospective Studies , Secondary Prevention , Time Factors
18.
J Biol Chem ; 280(2): 907-12, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15522884

ABSTRACT

The fifth domain (DV) of beta2-glycoprotein I (beta2GPI) is important for binding a number of ligands including phospholipids and factor XI (FXI). Beta2GPI is proteolytically cleaved in DV by plasmin but not by thrombin, VIIa, tissue plasminogen activator, or uPA. Following proteolytic cleavage of DV by plasmin, beta2GPI retains binding to FXI but not to phospholipids. Native beta2GPI, but not cleaved beta2GPI, inhibits activation of FXI by thrombin and factor XIIa, attenuating a positive feedback mechanism for additional thrombin generation. In this report, we have defined the FXI/FXIa binding site on beta2GPI using site-directed mutagenesis. We show that the positively charged residues Lys284, Lys286, and Lys287 in DV are essential for the interaction of beta2GPI with FXI/FXIa. We also demonstrate that FXIa proteolytically cleaves beta2GPI at Lys317-Thr318 in DV. Thus, FXIa cleavage of beta2GPI in vivo during thrombus formation may accelerate FXI activation by decreasing the inhibitory effect of beta2GPI.


Subject(s)
Factor XI/metabolism , Factor XIa/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Lysine/metabolism , Protein Processing, Post-Translational , Threonine/metabolism , Binding Sites , Fibrinolysin/metabolism , Glycoproteins/genetics , Humans , Mutation/genetics , Peptide Hydrolases/metabolism , Protein Binding , Protein Structure, Tertiary , Thrombin/metabolism , Time Factors , Tissue Plasminogen Activator/metabolism , beta 2-Glycoprotein I
19.
BioDrugs ; 18(5): 297-305, 2004.
Article in English | MEDLINE | ID: mdl-15377172

ABSTRACT

Autoantibodies to beta2-glycoprotein I (beta2GPI) are believed to be the primary cause of coagulation abnormalities in patients with antiphospholipid syndrome (APS). Clinical features include a range of life-threatening thrombotic events and microangiopathies affecting multiple organ systems. Current standard of care relies on long-term, high-intensity anticoagulation and is associated with a high risk for serious bleeding events. The relation between autoantibodies and the pathophysiology of APS is not clearly understood, but numerous in vitro studies have characterized the effects of antiphospholipid autoantibodies on various components of the coagulation cascade, including tissue factor and the protein C pathway. The fine specificity of autoantibodies to beta2GPI is a subject of considerable debate; however, a body of evidence may offer resolution by integrating concepts of antibody affinity and assay sensitivity with carefully designed molecular studies. An investigational new therapy for APS is based on the approach that pathogenic antibodies may be reduced via depletion of circulating autoantibodies and induction of immune tolerance at the B-cell level. Preliminary results from a phase I/II clinical trial with LJP 1082, a B-cell toleragen, indicate the drug was well tolerated and may warrant further development for reduction of thrombotic events in patients with APS.


Subject(s)
Antiphospholipid Syndrome/drug therapy , Immunosuppressive Agents/therapeutic use , Antiphospholipid Syndrome/etiology , Autoantibodies/adverse effects , Autoantibodies/immunology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Glycoproteins/adverse effects , Glycoproteins/immunology , Glycoproteins/therapeutic use , Humans , Immunosuppressive Agents/immunology , Methods , Models, Biological , Organic Chemicals , Polyethylene Glycols/therapeutic use , beta 2-Glycoprotein I
20.
Clin Immunol ; 112(2): 129-35, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15240155

ABSTRACT

Autoantibodies in antiphospholipid syndrome react predominantly with the plasma protein beta2-glycoprotein I (beta2GPI). Work by a large number of investigators has led to considerable progress in detecting and understanding beta2GPI reactivity with autoantibodies. Characterization of B cell epitopes on beta2GPI has benefited from an appreciation of its interactions with anionic phospholipids and a variety of microplate surfaces. In particular, autoantibodies to beta2GPI are of sufficiently low affinity to require high concentrations of antigen for detectable reactivity. Moreover, some microplate surfaces do not support the proper orientation of beta2GPI to allow display of epitopes in a manner accessible to autoantibodies. These concepts have helped to explain previous notions that exposure of cryptic beta2GPI epitopes may require interactions with anionic surfaces. Finally, we review evidence identifying a dominant B cell epitope that is partially defined by residues Gly40 and Arg43 on the amino terminal domain of beta2GPI.


Subject(s)
Autoantibodies/immunology , Epitopes, B-Lymphocyte/immunology , Glycoproteins/immunology , Models, Immunological , Animals , Antibody Affinity , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/physiopathology , Humans , beta 2-Glycoprotein I
SELECTION OF CITATIONS
SEARCH DETAIL
...