Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Basic Res Cardiol ; 111(2): 20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26891724

ABSTRACT

Macrophages in the arterial intima sustain chronic inflammation during atherogenesis. Under hypercholesterolemic conditions murine Ly6C(high) monocytes surge in the blood and spleen, infiltrate nascent atherosclerotic plaques, and differentiate into macrophages that proliferate locally as disease progresses. Spleen tyrosine kinase (SYK) may participate in downstream signaling of various receptors that mediate these processes. We tested the effect of the SYK inhibitor fostamatinib on hypercholesterolemia-associated myelopoiesis and plaque formation in Apoe(-/-) mice during early and established atherosclerosis. Mice consuming a high cholesterol diet supplemented with fostamatinib for 8 weeks developed less atherosclerosis. Histologic and flow cytometric analysis of aortic tissue showed that fostamatinib reduced the content of Ly6C(high) monocytes and macrophages. SYK inhibition limited Ly6C(high) monocytosis through interference with GM-CSF/IL-3 stimulated myelopoiesis, attenuated cell adhesion to the intimal surface, and blocked M-CSF stimulated monocyte to macrophage differentiation. In Apoe(-/-) mice with established atherosclerosis, however, fostamatinib treatment did not limit macrophage accumulation or lesion progression despite a significant reduction in blood monocyte counts, as lesional macrophages continued to proliferate. Thus, inhibition of hypercholesterolemia-associated monocytosis, monocyte infiltration, and differentiation by SYK antagonism attenuates early atherogenesis but not established disease when local macrophage proliferation dominates lesion progression.


Subject(s)
Atherosclerosis/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Monocytes/drug effects , Myelopoiesis/drug effects , Oxazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/therapeutic use , Aminopyridines , Animals , Atherosclerosis/immunology , Atherosclerosis/prevention & control , Cell Adhesion/drug effects , Cells, Cultured , Disease Progression , Drug Evaluation, Preclinical , Female , Macrophages/drug effects , Mice , Morpholines , Oxazines/pharmacology , Pyridines/pharmacology , Pyrimidines , Random Allocation , Syk Kinase
2.
Cytotherapy ; 18(2): 186-97, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26596504

ABSTRACT

The prevalence of end-stage renal disease is emerging as a serious worldwide public health problem because of the shortage of donor organs and the need to take lifelong immunosuppressive medication in patients who receive a transplanted kidney. Recently, tissue bioengineering of decellularization and recellularization scaffolds has emerged as a novel strategy for organ regeneration, and we review the critical technologies supporting these methods. We present a summary of factors associated with experimental protocols that may shed light on the future development of kidney bioengineering and we discuss the cell sources and bioreactor techniques applied to the recellularization process. Finally, we review some artificial renal engineering technologies and their future prospects, such as kidney on a chip and the application of three-dimensional and four-dimensional printing in kidney tissue engineering.


Subject(s)
Guided Tissue Regeneration/methods , Kidney Failure, Chronic/therapy , Regeneration , Regenerative Medicine/trends , Tissue Engineering/methods , Animals , Bioreactors , Humans , Induced Pluripotent Stem Cells/cytology , Kidney/cytology , Kidney/pathology , Organogenesis , Rats , Tissue Scaffolds
3.
Rheumatology (Oxford) ; 54(1): 169-77, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25065010

ABSTRACT

OBJECTIVES: Spleen tyrosine kinase (SYK) is a core signalling protein that drives inflammatory responses and is fundamental to the propagation of signals via numerous immune receptors, including the B cell receptor and Fc receptors (FcRs). Fostamatinib, a small molecule SYK inhibitor, has shown evidence of ameliorating inflammation in RA patients. We sought to understand how the active metabolite of fostamatinib, R406, affects the inflammatory response at the cellular level. METHODS: Antigen-specific in vivo systems and in vitro fluorescence microscopy were combined to investigate the effects of fostamatinib on antigen-specific interactions between dendritic cells (DCs) and CD4(+) T cells. RESULTS: Although it has previously been shown that R406 reduces the response of DCs to immune complexes (ICs), we found that fostamatinib failed to reduce specific CD4(+) T cell proliferation in mice after immunization with ICs. However, we observed in vitro that R406 reduces both the area and duration of cellular interactions between IC-activated DCs and specific CD4(+) T cells during the initial phase of cellular crosstalk. This led to diminished proliferation of antigen-specific CD4(+) T cells after R406 treatment compared with vehicle controls. This decreased proliferative capacity of CD4(+) T cells was accompanied by reduced expression of the co-stimulatory molecules, inducible T cell co-stimulator (ICOS) and PD-1, and abrogation of the production of inflammatory cytokines such as IFN-γ and IL-17. CONCLUSION: Our findings indicate a potential mechanism by which this compound may be effective in inhibiting FcR-driven CD4(+) T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Oxazines/metabolism , Oxazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/metabolism , Pyridines/pharmacology , Administration, Oral , Aminopyridines , Animals , Antigen-Antibody Complex/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Animal , Morpholines , Oxazines/administration & dosage , Pyridines/administration & dosage , Pyrimidines , Syk Kinase
4.
Cytotherapy ; 17(8): 1015-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25981396

ABSTRACT

Allogeneic transplantation is the definitive treatment for patients with end-stage liver disease but is limited by donor shortage and very high cost. Through de-cellularization and re-cellularization methods, re-engineered liver may provide a promising alternative for treating patients with end-stage liver disease. To achieve this, the prevention of the native extracellular matrix ultrastructure plays a central role in de-cellularization protocol; the re-seeding cell types, as well as re-seeding strategies, need more explorations in re-cellularization protocol. Some success of this approach has been published in a rat model; however, the re-engineered liver remains functional in vivo for only several hours, which suggests that the recent protocol may be far from the ideal target. This Review highlights the challenges still to be overcome and presents an overview and summary of methods of de-cellularization and re-cellularization strategies, together with a view on future directions that may lead to the regeneration of a functional liver.


Subject(s)
End Stage Liver Disease/surgery , Hepatocytes/transplantation , Liver Regeneration/physiology , Liver Transplantation/methods , Liver/cytology , Tissue Engineering/trends , Animals , Extracellular Matrix/metabolism , Humans , Rats , Tissue Donors , Transplantation, Homologous
5.
Clin Exp Rheumatol ; 32(6): 878-82, 2014.
Article in English | MEDLINE | ID: mdl-25288220

ABSTRACT

OBJECTIVES: This paper aims to explore the functional significance of the P2X7 receptor in preclinical models of rheumatoid arthritis. METHODS: Preclinical studies in vivo were performed using the rat streptococcal cell wall (SCW) arthritis model. Ex vivo cultures of lipopolysaccharide (LPS)/benzoylbenzoyl adenosine triphosphate (BzATP)-stimulated human monocytes were generated to test the activities of a novel, highly specific inhibitor of human P2X7, AZD9056, on interleukin (IL)-1 and IL-18 release. RESULTS: P2X7 receptor expression was detected in inflamed synovial tissue after onset of SCW-induced arthritis in rats. Inhibition of P2X7 therein led to reduced articular inflammation and erosive progression. No effect was noted on acute-phase responses. Ex vivo, AZD9056 inhibited IL-1 and IL-18 release to BzATP in LPS-primed human monocytes. CONCLUSIONS: P2X7 receptor inhibition could represent a novel approach to the treatment of inflammatory arthritis. However, confirmatory clinical studies are warranted to further explore this possibility.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Molecular Targeted Therapy , Monocytes/drug effects , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Synovial Membrane/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Arthritis, Rheumatoid/metabolism , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Inflammation Mediators/metabolism , Interleukin-1/metabolism , Interleukin-18/metabolism , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Rats , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects , Synovial Membrane/immunology , Synovial Membrane/metabolism , Time Factors
6.
J Immunol ; 185(4): 2611-9, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20639492

ABSTRACT

The ATP-gated P2X(7) receptor (P2X(7)R) is a promising therapeutic target in chronic inflammatory diseases with highly specific antagonists currently under clinical trials for rheumatoid arthritis. Anti-inflammatory actions of P2X(7)R antagonists are considered to result from inhibition of P2X(7)R-induced release of proinflammatory cytokines from activated macrophages. However, P2X(7)Rs are also expressed in resting macrophages, suggesting that P2X(7)R may also signal via cytokine-independent mechanisms involved in joint disease. In this study, we examined P2X(7)R function in resting human lung macrophages and mouse bone marrow-derived macrophages and found that ATP induced rapid release of the lysosomal cysteine proteases cathepsin B, K, L, and S and that was independent of the presence of the proinflammatory cytokines IL-1beta and IL-18. Cathepsins released into the medium were effective to degrade collagen extracellular matrix. ATP-induced cathepsin release was abolished by P2X(7)R antagonists, absent from P2X(7)R(-/-) mouse macrophages, and not associated with cell death. Our results suggest P2X(7)R activation may play a novel and direct role in tissue damage through release of cathepsins independently of its proinflammatory actions via IL-1 cytokines.


Subject(s)
Cathepsins/metabolism , Cytokines/metabolism , Joint Diseases/metabolism , Macrophages/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/pharmacology , Animals , Blotting, Western , Cathepsin B/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Etanercept , Humans , Immunoglobulin G/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-1beta/metabolism , Joint Diseases/genetics , L Cells , Macrophages/cytology , Macrophages/drug effects , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Receptors, Tumor Necrosis Factor , Thiazoles/chemistry , Thiazoles/pharmacology
8.
Arthritis Rheum ; 62(11): 3154-60, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20662070

ABSTRACT

OBJECTIVE: To investigate both the preclinical effects of blocking the chemokine receptor CCR5 and the clinical effects of this approach on the signs and symptoms of rheumatoid arthritis (RA) in patients with active disease. METHODS: Preclinical evaluations of AZD5672, a small-molecule antagonist of CCR5, were performed, including studies of ligand binding and chemotaxis. The pharmacokinetics of AZD5672 were assessed in both single- and multiple-dose studies in healthy volunteers. A randomized, placebo-controlled, phase IIb study was conducted in patients with active RA receiving methotrexate. Treatment arms were AZD5672 (20, 50, 100, or 150 mg orally, once daily), matched placebo, or open-label etanercept (50 mg subcutaneously, once weekly). The primary end point was the proportion of patients achieving a 20% improvement response on the American College of Rheumatology improvement criteria (ACR20) at week 12. Secondary end points included the ACR20 over time, as well as 50% (ACR50) and 70% (ACR70) improvement responses, changes in individual components of the ACR improvement criteria, and disease activity measured with the Disease Activity Score based on the 28-joint count. RESULTS: AZD5672 was a highly potent and selective antagonist of CCR5, displaying nonproportional steady-state pharmacokinetics while inhibiting internalization of CCR5 in an ex vivo macrophage inflammatory protein 1ß stimulation assay in which AZD5672 was evaluated over the 20-150-mg dose range. In the phase IIb study testing this dose range in patients with RA (n = 371 patients randomized to received treatment), AZD5672 was generally well tolerated, with no unexpected adverse events. There was no statistically significant difference in the proportion of patients achieving an ACR20 response at week 12 between those receiving any dose of AZD5672 and those receiving placebo; etanercept was significantly more efficacious than AZD5672 and placebo. CONCLUSION: Despite a clear rationale for targeting CCR5, this clinical study showed that AZD5672, administered orally, did not have any clinical benefit, suggesting that CCR5 antagonism alone is unlikely to be a viable therapeutic strategy in RA.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Benzeneacetamides/therapeutic use , CCR5 Receptor Antagonists , Methotrexate/therapeutic use , Sulfonamides/therapeutic use , Antirheumatic Agents/therapeutic use , Chi-Square Distribution , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Treatment Outcome
9.
Cytotherapy ; 12(3): 349-60, 2010 May.
Article in English | MEDLINE | ID: mdl-20053145

ABSTRACT

Today, many patients suffer from acute liver failure and hepatoma. This is an area of high unmet clinical need as these conditions are associated with very high mortality. There is an urgent need to develop techniques that will enable liver tissue engineering or generate a bioartificial liver, which will maintain or improve liver function or offer the possibility of liver replacement. Liver tissue engineering is an innovative way of constructing an implantable liver and has the potential to alleviate the shortage of organ donors for orthotopic liver transplantation. In this review we describe, from an engineering perspective, progress in the field of liver tissue engineering, including three main aspects involving cell sources, scaffolds and vascularization.


Subject(s)
Liver , Tissue Engineering , Adipose Tissue/cytology , Animals , Cell Line , Fetal Blood/cytology , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Liver/cytology , Liver/physiology , Liver Failure/therapy , Liver Neoplasms/therapy , Stem Cells/cytology , Stem Cells/physiology , Telomerase/metabolism
10.
Curr Drug Discov Technol ; 17(1): 45-56, 2020.
Article in English | MEDLINE | ID: mdl-30648510

ABSTRACT

The unique nature of microgravity encountered in space provides an opportunity for drug discovery and development that cannot be replicated on Earth. From the production of superior protein crystals to the identification and validation of new drug targets to microarray analyses of transcripts attenuated by microgravity, there are numerous examples which demonstrate the benefit of exploiting the space environment. Moreover, studies conducted on Space Shuttle missions, the International Space Station and other craft have had a direct benefit for drug development programmes such as those directed against reducing bone and muscle loss or increasing bone formation. This review will highlight advances made in both drug discovery and development and offer some future insight into how drug discovery and associated technologies may be further advanced using the microgravity assist.


Subject(s)
Drug Development/methods , Drug Discovery/methods , Space Flight , Weightlessness , Animals , Bone Density/drug effects , Bone Density/physiology , Crystallography, X-Ray/methods , Disease Models, Animal , Drug Development/trends , Drug Discovery/trends , Gene Expression Profiling/methods , Humans , Mice , Muscular Atrophy/drug therapy , Muscular Atrophy/physiopathology , Oligonucleotide Array Sequence Analysis/methods , Osteoporosis/drug therapy , Osteoporosis/physiopathology
11.
New Bioeth ; 25(4): 295-317, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31558118

ABSTRACT

While humans have made enormous progress in the exploration and exploitation of Earth, exploration of outer space remains beyond current human capabilities. The principal challenges lie in current space technology and engineering which includes the protection of astronauts from the hazards of working and living in the space environment. These challenges may lead to a paradoxical situation where progress in space technology and the ability to ensure acceptable risk/benefit for human space exploration becomes dissociated and the rate of scientific discovery declines. In this paper, we discuss the predominant challenges of the space environment for human health and argue that development and deployment of a human enhancement policy, initially confined to astronauts - for the purpose of future human space programmes is a rational solution to these challenges.


Subject(s)
Astronauts , Bioethics , Biomedical Enhancement , Extraterrestrial Environment , Space Flight , Biomedical Enhancement/ethics , Dissent and Disputes , Humans
12.
Drug Saf ; 42(6): 769-784, 2019 06.
Article in English | MEDLINE | ID: mdl-30649752

ABSTRACT

INTRODUCTION: Tralokinumab is a monoclonal antibody (mAb) that neutralizes interleukin (IL)-13, a cytokine involved in the pathogenesis of asthma. OBJECTIVE: The objectives of this study were to characterize the potential immunogenic properties of tralokinumab and report data for anti-drug antibodies (ADAs) and hypersensitivity reactions from two phase III clinical trials. METHODS: The oligosaccharide structure of tralokinumab, Fab-arm exchange, and ADAs were characterized by standard techniques. Hypersensitivity adverse events (AEs) were evaluated in two pivotal clinical trials of tralokinumab in severe, uncontrolled asthma: STRATOS 1 and 2 (NCT02161757 and NCT02194699). RESULTS: No galactose-α-1,3-galactose (α-Gal) epitopes were found in the Fab region of tralokinumab and only 4.5% of glycoforms contained α-Gal in the Fc region. Under non-reducing conditions, Fab-arm exchange did not take place with another immunoglobulin (Ig) G4 mAb (mavrilimumab). However, following glutathione reduction, a hybrid antibody with monovalent bioactivity was detected. ADA incidences (titers) were as follows: STRATOS 1-every 2 weeks (Q2 W) 0.8% (26.0), every 4 weeks (Q4 W) 0.5% (26.0), placebo 0.8% (52.0); STRATOS 2-Q2 W 1.2% (39.0), placebo 0.8% (13.0). Participant-reported hypersensitivity AE rates were as follows: STRATOS 1-Q2 W 25.9%, Q4 W 25.0%, placebo 25.5%; STRATOS 2-Q2 W 13.2%, placebo 9.0%. External evaluation for anaphylaxis by Sampson criteria found no tralokinumab-related severe hypersensitivity or anaphylaxis reactions. CONCLUSION: Preclinical assessments suggested a low likelihood of immunogenicity for tralokinumab. In STRATOS 1 and 2, ADA incidence was low, no differences were found between tralokinumab-treated and placebo groups in reporting of hypersensitivity reactions, and there were no Sampson criteria-evaluated anaphylaxis events with tralokinumab treatment. Together, the results suggest that tralokinumab treatment would not increase the risk for severe hypersensitivity or anaphylactic reactions.


Subject(s)
Anaphylaxis/chemically induced , Antibodies, Monoclonal/adverse effects , Antibody Formation/immunology , Asthma/drug therapy , Drug Hypersensitivity/immunology , Adolescent , Adult , Aged , Anaphylaxis/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/immunology , Child , Double-Blind Method , Female , Humans , Male , Middle Aged , Young Adult , alpha-Galactosidase/immunology
13.
Immunotherapy ; 10(6): 473-490, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29536781

ABSTRACT

Tralokinumab, a fully human IgG4 monoclonal antibody, specifically neutralizes IL-13. The ATMOSPHERE clinical development program comprised four randomized, placebo-controlled clinical trials and an open-label study that aimed to assess the efficacy and safety of tralokinumab for the treatment of severe, uncontrolled asthma. The two pivotal trials (STRATOS 1 and STRATOS 2; NCT02161757 and NCT02194699) evaluated the efficacy and safety of tralokinumab, with STRATOS 1 identifying a subgroup most likely to demonstrate enhanced response to treatment. Further trials have assessed the ability of tralokinumab to reduce oral corticosteroid use (TROPOS; NCT02281357) and determined its mechanistic effects (MESOS; NCT02449473). An open-label study in Japanese individuals (NCT02902809) assessed the long-term safety and tolerability of tralokinumab in this population.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Animals , Biomarkers/blood , Disease Progression , Humans , Interleukin-13/blood , Interleukin-13/immunology , Randomized Controlled Trials as Topic
14.
Drug Saf ; 41(5): 489-509, 2018 05.
Article in English | MEDLINE | ID: mdl-29411337

ABSTRACT

INTRODUCTION: Interleukin-13 and interleukin-4 are type-II cytokines signalling through the shared type II interleukin-4 receptor. As a result of their structural similarity, interleukin-13 and interleukin-4 have overlapping functions in the mediation of type-II-driven diseases and are, therefore, promising targets of biologic drugs currently in development for the treatment of such diseases, including asthma and atopic dermatitis. OBJECTIVE: This systematic review was conducted to assess preclinical evidence of potential safety concerns related to blockade of interleukin-13 alone or interleukin-13 and interleukin-4 in combination. METHODS: We specifically examined risks related to infection, malignancy and the cardiovascular system. We systematically searched the BIOSIS, MEDLINE and EMBASE databases to identify preclinical studies published between January 2006 and October 2016 that addressed the effects of interleukin-13/interleukin-4 blockade and modulation on the risk of infection, malignancy and cardiovascular events. To provide a clinical context, we also performed a search for clinical trials targeting the interleukin-13/interleukin-4 pathways. Relevant data from preclinical and clinical trials were abstracted and presented descriptively. RESULTS: Aside from expected evidence that inhibition of interleukin-13 and interleukin-4 impaired host responses to helminth infections, we did not identify other preclinical evidence suggesting safety risks relating to infection, malignancy or cardiovascular events. We found no evidence in clinical trials suggesting serious safety concerns, i.e. increased risk for infections, malignancy or cardiovascular events from therapeutic modulation of the interleukin-13 pathway alone or the combined interleukin-13/interleukin-4 pathways. CONCLUSIONS: Although our findings are reassuring, long-term safety assessments of biologics that target the interleukin-13/interleukin-4 pathways currently in clinical development are needed.


Subject(s)
Interleukin-13/metabolism , Interleukin-4/metabolism , Signal Transduction/physiology , Animals , Asthma/metabolism , Humans , Risk
15.
Arthritis Rheumatol ; 70(9): 1419-1428, 2018 09.
Article in English | MEDLINE | ID: mdl-29669391

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is a chronic and degenerative autoimmune joint disease that leads to disability, reduced quality of life, and increased mortality. Although several synthetic and biologic disease-modifying antirheumatic drugs are available, there is still a medical need for novel drugs that control disease progression. As only 10% of experimental drug candidates for treatment of RA that enter phase I trials are eventually registered by the Food and Drug Administration, there is an immediate need for translational tools to facilitate early decision-making in drug development. In this study, we aimed to determine if the inability of fostamatinib (a small molecule inhibitor of Syk) to demonstrate sufficient efficacy in phase III of a previous clinical study could have been predicted earlier in the development process. METHODS: Biomarkers of bone, cartilage, and interstitial matrix turnover (C-telopeptide of type I collagen [CTX-I], matrix metalloproteinase-derived types I, II, and III collagen neoepitopes [C1M, C2M, and C3M]) were measured in 450 serum samples from the Oral Syk Inhibition in Rheumatoid Arthritis 1 study (OSKIRA-1, a phase III clinical study of the efficacy of fostamatinib in RA) at baseline and follow-up. Additionally, the same biomarkers were subsequently measured in conditioned media from osteoclast, cartilage, and synovial membrane cultured with the active metabolite of fostamatinib, R406, to assess the level of suppression induced by the drug. RESULTS: In OSKIRA-1 serum samples and osteoclast and cartilage cultures, fostamatinib suppressed the levels of CTX-I and C2M. In OSKIRA-1 serum samples and synovial membrane cultures, fostamatinib did not mediate any clinical or preclinical effect on either C1M or C3M, which have previously been associated with disease response and efficacy. CONCLUSION: These data demonstrate that translational biomarkers are a potential tool for early assessment and decision-making in drug development for RA treatment.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Drug Discovery/methods , Translational Research, Biomedical/methods , Aminopyridines , Biomarkers/analysis , Cartilage/drug effects , Cartilage/metabolism , Collagen/drug effects , Collagen/metabolism , Drug Development/methods , Humans , Morpholines , Oxazines/pharmacology , Pyridines/pharmacology , Pyrimidines , Synovial Membrane/drug effects , Synovial Membrane/metabolism
16.
Lancet Respir Med ; 6(7): 499-510, 2018 07.
Article in English | MEDLINE | ID: mdl-29793857

ABSTRACT

BACKGROUND: The role of interleukin 13 in airway inflammation and remodelling in asthma is unclear. Tralokinumab is a human monoclonal antibody that neutralises interleukin 13. We aimed to evaluate whether tralokinumab would have an effect on airway eosinophilic infiltration, blood and sputum eosinophil concentrations, eosinophil activation, and airway remodelling. METHODS: We did a multicentre, double-blind, randomised, placebo-controlled phase 2 trial at 15 centres across the UK, Denmark, and Canada. We enrolled participants of either sex aged 18-75 years with inadequately controlled moderate-to-severe asthma for 12 months or more, requiring treatment with inhaled corticosteroids at a stable dose. We randomly assigned participants (1:1) to receive tralokinumab (300 mg) or placebo by an interactive web-based system or voice response system. Participants and study personnel were masked to treatment allocation. Both tralokinumab and placebo were administered subcutaneously every 2 weeks. The primary outcome measure was change from baseline to week 12 in bronchial biopsy eosinophil count. Secondary outcome measures included change in blood and sputum eosinophil counts. Exploratory outcomes included fractional exhaled nitric oxide (FENO) and blood IgE concentrations. Safety analyses were carried out in all participants who received study drug. This trial is registered with ClinicalTrials.gov, number NCT02449473, and with the European Clinical Trials Database, EudraCT 2015-000857-19. FINDINGS: Between Sept 25, 2015, and June 21, 2017, 224 participants were enrolled and screened. Of these participants, 79 were randomly assigned to receive tralokinumab (n=39) or placebo (n=40). Tralokinumab did not significantly affect bronchial eosinophil count compared with placebo at week 12 (treatment effect ratio 1·43, 95% CI 0·63-3·27; p=0·39). Compared with placebo, tralokinumab did not significantly affect blood eosinophil count (treatment effect ratio 1·21, 95% CI 1·00-1·48; p=0·055) or sputum eosinophil count (0·57, 0·06-6·00; p=0·63), but FENO concentration (0·78, 0·63-0·96; p=0·023) and total blood IgE concentration (0·86, 0·77-0·97; p=0·014) were significantly reduced. 33 (85%) of 39 patients receiving tralokinumab and 32 (80%) of 40 receiving placebo reported at least one adverse event during the treatment period. No deaths in either treatment group were observed. Treatment-related adverse events occurred more frequently in the tralokinumab group than in the placebo group (11 [28%] of 39 vs seven [18%] of 40). INTERPRETATION: Tralokinumab did not significantly affect eosinophilic inflammation in bronchial submucosa, blood, or sputum compared with placebo, but did reduce FENO and IgE concentrations. These results suggest interleukin 13 is not crucial for eosinophilic airway inflammation control in moderate-to-severe asthma. FUNDING: AstraZeneca.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Inflammation/drug therapy , Adolescent , Adult , Aged , Asthma/physiopathology , Bronchi/drug effects , Bronchi/physiopathology , Canada , Denmark , Double-Blind Method , Eosinophilia/physiopathology , Female , Humans , Inflammation/physiopathology , Male , Middle Aged , Severity of Illness Index , Treatment Outcome , United Kingdom , Young Adult
18.
Expert Rev Gastroenterol Hepatol ; 11(11): 1009-1018, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28708431

ABSTRACT

INTRODUCTION: The outcome of a comparative efficacy and safety of vasoconstrictor therapies for treatment of patients with type 1 hepatorenal syndrome (HRS-1) remain inconclusive. Areas covered: We searched literature databases for randomized controlled trials (RCTs) until 31 January 2016, and included ten eligible RCTs. In conclusion, terlipressin was the most efficacious vasoconstrictor drug for HRS-1, but had a higher probability of causing AEs. Norepinephrine was an attractive alternative to terlipressin and associated with less AEs. Expert commentary: To date, most previous traditional meta-analyses included trials with a limited population and compared terlipressin alone or with albumin against no intervention or albumin. Since different HRS types have different diagnoses and show different responses to vasoconstrictors, it may be questionable to combine data from patients with type 1 and type 2 HRS, which has been reported for most previous meta-analyses. Thus, performing a high-quality network meta-analysis of the existing literature is a valuable way to interrogate published data and to draw conclusions which may inform on the best interventional strategy.


Subject(s)
Hepatorenal Syndrome/drug therapy , Lypressin/analogs & derivatives , Norepinephrine/therapeutic use , Vasoconstriction/drug effects , Vasoconstrictor Agents/therapeutic use , Albumins/therapeutic use , Comparative Effectiveness Research , Female , Hepatorenal Syndrome/classification , Hepatorenal Syndrome/diagnosis , Hepatorenal Syndrome/physiopathology , Humans , Lypressin/adverse effects , Lypressin/therapeutic use , Male , Middle Aged , Norepinephrine/adverse effects , Odds Ratio , Plasma Substitutes/therapeutic use , Randomized Controlled Trials as Topic , Risk Factors , Terlipressin , Treatment Outcome , Vasoconstrictor Agents/adverse effects
19.
Eur J Gastroenterol Hepatol ; 29(6): 698-705, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28240612

ABSTRACT

BACKGROUND AND AIM: Critically ill cirrhosis patients have an increased risk of morbidity and mortality, even after admission to the ICU. Our objectives were to compare the predictive accuracy of model for end-stage liver disease (MELD), MELD-Na, UK model for end-stage liver disease, and chronic liver failure-sequential organ failure assessment (CLIF-SOFA) by the development and validation of an easy-to-use prognostic model [named quick CLIF-SOFA (qCLIF-SOFA)] for early risk prediction in critically ill patients with cirrhosis. PATIENTS AND METHODS: Overall, 1460 patients were extracted from the MIMIC-III database and enrolled in this study at 30-day and 90-day follow-up. qCLIF-SOFA was developed in the established cohort (n=730) and a performance analysis was completed in the validation cohort (n=730) using area under the receiver operating characteristic curve. Results were compared with CLIF-SOFA. RESULTS: The performance of CLIF-SOFA was significantly better than that of MELD, MELD-Na, and UK model for end-stage liver disease for predicting both 30-day and 90-day mortality (all P<0.05). qCLIF-SOFA consisted of five independent factors (bilirubin, creatinine, international normalized ratio, mean arterial pressure, and vasopressin) associated with mortality. In the established cohort, CLIF-SOFA and qCLIF-SOFA predicted mortality with area under the receiver operating characteristic curve values of 0.768 versus 0.743 at 30-day, 0.747 versus 0.744 at 90-day, and 0.699 versus 0.706 at 1 year, respectively (all P>0.05). A similar result was observed in the validation cohort (0.735 vs. 0.734 at 30 days, 0.723 vs. 0.737 at 90 days, and 0.682 vs. 0.700 at 1 year, respectively, all P>0.05). CONCLUSION: The utility of CLIF-SOFA was further shown to predict mortality for critically ill cirrhosis patients. The novel and simpler qCLIF-SOFA model showed comparable accuracy compared with existing CLIF-SOFA for prognostic prediction.


Subject(s)
Decision Support Techniques , Liver Cirrhosis/diagnosis , Liver Cirrhosis/mortality , Liver Failure/diagnosis , Liver Failure/mortality , Models, Biological , Multiple Organ Failure/diagnosis , Multiple Organ Failure/mortality , Organ Dysfunction Scores , Aged , Area Under Curve , Arterial Pressure , Bilirubin/blood , Biomarkers/blood , Creatinine/blood , Critical Illness , Databases, Factual , Female , Humans , International Normalized Ratio , Kaplan-Meier Estimate , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Failure/blood , Liver Failure/etiology , Male , Middle Aged , Multiple Organ Failure/blood , Multiple Organ Failure/etiology , Predictive Value of Tests , Prognosis , ROC Curve , Reproducibility of Results , Risk Assessment , Risk Factors , Time Factors
20.
Eur J Gastroenterol Hepatol ; 29(10): 1166-1173, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28746121

ABSTRACT

BACKGROUND AND AIM: Upper gastrointestinal bleeding (UGIB) is a complication with a high mortality rate in critically ill patients presenting with cirrhosis. Today, there exist few accurate scoring models specifically designed for mortality risk assessment in critically ill cirrhotic patients with upper gastrointestinal bleeding (CICGIB). Our aim was to develop and evaluate a novel nomogram-based model specific for CICGIB. PATIENTS AND METHODS: Overall, 540 consecutive CICGIB patients were enrolled. On the basis of Cox regression analyses, the nomogram was constructed to estimate the probability of 30-day, 90-day, 270-day, and 1-year survival. An upper gastrointestinal bleeding-chronic liver failure-sequential organ failure assessment (UGIB-CLIF-SOFA) score was derived from the nomogram. Performance assessment and internal validation of the model were performed using Harrell's concordance index (C-index), calibration plot, and bootstrap sample procedures. UGIB-CLIF-SOFA was also compared with other prognostic models, such as CLIF-SOFA and model for end-stage liver disease, using C-indices. RESULTS: Eight independent factors derived from Cox analysis (including bilirubin, creatinine, international normalized ratio, sodium, albumin, mean artery pressure, vasopressin used, and hematocrit decrease>10%) were assembled into the nomogram and the UGIB-CLIF-SOFA score. The calibration plots showed optimal agreement between nomogram prediction and actual observation. The C-index of the nomogram using bootstrap (0.729; 95% confidence interval: 0.689-0.766) was higher than that of the other models for predicting survival of CICGIB. CONCLUSION: We have developed and internally validated a novel nomogram and an easy-to-use scoring system that accurately predicts the mortality probability of CICGIB on the basis of eight easy-to-obtain parameters. External validation is now warranted in future clinical studies.


Subject(s)
Decision Support Techniques , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/mortality , Liver Cirrhosis/complications , Liver Cirrhosis/mortality , Nomograms , Aged , Arterial Pressure , Biomarkers/blood , Chi-Square Distribution , Databases, Factual , Female , Gastrointestinal Hemorrhage/diagnosis , Humans , International Normalized Ratio , Kaplan-Meier Estimate , Liver Cirrhosis/blood , Liver Cirrhosis/diagnosis , Male , Middle Aged , Multivariate Analysis , Organ Dysfunction Scores , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Reproducibility of Results , Risk Assessment , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL