GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 81, No. Suppl 1 ( 2022-06), p. 1344-1345
    Abstract: A major limitation of biologic therapy is formation of anti-drug antibodies (ADA). We previously found ADA to rituximab (RTX) are more prevalent in patients undergoing treatment for systemic lupus erythematosus (SLE) than rheumatoid arthritis and vasculitis (1). In addition, we demonstrated that ADA to RTX predict subsequent infusion related reactions (2). However, little is known regarding the long-term dynamics of ADA to RTX in patients undergoing treatment for SLE. Objectives In this study we evaluated the longitudinal impact of ADA positivity with particular focus on; 1) Risk factors for development of ADA. 2) Impact of ADA on treatment response. 3) Influence of ADA on RTX drug kinetics over time. 4) The capacity of ADA to neutralise RTX. Methods Patients with SLE undergoing treatment with RTX were recruited to this study (n=35). Serum samples were collected at the following intervals post-treatment; 1-3 months (defined as ‘early’ post-treatment), 6 months, 12 months, 36 months (n=114). Clinical and laboratory data was collected pre-treatment and at each follow-up time point. Response to treatment was assessed by improvement in SLEDAI-2K score from baseline and also according to BILAG as previously described (3). ADA were detected using an electrochemiluminescent immunoassay. Serum RTX levels were measured by ELISA. ADA status was defined according to the following patterns over time; persistently negative, persistently positive (0-15 AU/ml) and persistently high positive (≥16 AU/ml, upper quartile). A complement dependent cytotoxic assay was used to determine neutralising capability of ADA in a subgroup of positive samples (n=38). Results ADA to RTX were found to be persistently positive in 64.3% of patients over the 36-month follow-up period and there was no significant difference in baseline disease activity (BILAG / SLEDAI-2K) between those who were subsequently ADA positive vs negative. ADA positive patients had a younger age at diagnosis of SLE when compared with ADA negative (mean 22.50 ± 9.10 vs 37.29 ± 11.31 years, p=0.002, Figure 1 A). Multivariate logistic regression found a 22% decrease in risk of ADA positivity for each addition year after diagnosis (p=0.03). Figure 1. ADA positive patients had a significantly lower C3 level at baseline (mean 0.61 ± 0.23 g/L vs 0.87 ± 0.30 g/L, p=0.026), which remained lower at each subsequent time point post-treatment up to 12 months post-treatment (Figure 1B). At 1-3 months post-RTX, patients who were ADA positive had a significantly lower circulating drug level than ADA negative (p 〈 0.001, Figure 1 C). In terms of clinical response, ADA positive patients had an initial significant improvement in disease activity (SLEDAI-2K) by 3 months (p 〈 0.001). However, response was not maintained at 12 months (Figure 1 D). In comparison, ADA negative patients showed a significant improvement in SLEDAI-2K at 6 months and this was maintained across the 36-month follow-up period (Figure 1 E). BILAG defined relapse was more common at six months post-treatment in ADA positive patients (22%) and ADA highly positive patients (33%) than those who were ADA negative (in which there were no cases of relapse within the first six months, Figure 1 F). At 12-months post-RTX, a higher rate of BILAG defined Major Response was seen in those who were ADA negative (80%) when compared with ADA positive (44%) and high positive (36%) as shown in Figure 1 G. Finally, antibodies derived from all ADA positive samples (38/38) were found to neutralise RTX in vitro . Conclusion ADA to RTX were common and persisted over the 36-month period of this study. ADA associated with earlier serum drug elimination, increased relapse rates and demonstrated neutralising capacity suggesting that ADA could be a significant limitation to sustained response to treatment in clinical practice. References [1]Faustini F et al. Arthritis research & therapy. 2021;23(1):211 [2]Wincup C et al. Annals of the rheumatic diseases. 2019;78(8):1140-2 [3]Md Yusof MY et al Annals of the rheumatic diseases. 2017;76(11):1829-36 Disclosure of Interests None declared
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 79, No. Suppl 1 ( 2020-06), p. 179.2-179
    Abstract: Juvenile-onset systemic lupus erythematosus (JSLE) is a complex and heterogeneous disease characterised by diagnosis and treatment delays. An unmet need exists to better characterise the immunological profile of JSLE patients and investigate its links with the disease trajectory over time. Objectives: A machine learning (ML) approach was applied to explore new diagnostic signatures for JSLE based on immune-phenotyping data and stratify patients by specific immune characteristics to investigate longitudinal clinical outcome. Methods: Immune-phenotyping of 28 T-cell, B-cell and myeloid-cell subsets in 67 age and sex-matched JSLE patients and 39 healthy controls (HCs) was performed by flow cytometry. A balanced random forest (BRF) ML predictive model was developed (10,000 decision trees). 10-fold cross validation, Sparse Partial Least Squares-Discriminant Analysis (sPLS-DA) and logistic regression was used to validate the model. Longitudinal clinical data were related to the immunological features identified by ML analysis. Results: The BRF-model discriminated JSLE patients from healthy controls with 91% prediction accuracy suggesting that JSLE patients could be distinguished from HCs with high confidence using immunological parameters. The top-ranked immunological features from the BRF-model were confirmed using sPLS-DA and logistic regression and included CD19+ unswitched memory B-cells, naïve B-cells, CD14 + monocytes and total CD4 + , CD8 + and memory T-cell subsets. K-mean clustering was applied to stratify patients using the validated signature. Four groups were identified, each with a distinct immune and clinical profile. Notably, CD8 + T-cell subsets were important in driving patient stratification while B-cell markers were similarly expressed across the JSLE cohort. JSLE patients with elevated effector memory CD8 + T-cell frequencies had more persistently active disease over time, and this was associated with increased treatment burden and prevalence of lupus nephritis. Finally, network analysis identified specific clinical features associated with each of the top JSLE immune-signature variables. Conclusion: Using a combined ML approach, a distinct immune signature was identified that discriminated between JSLE patients and HCs and further stratified patients. This signature could have diagnostic and therapeutic implications. Further immunological association studies are warranted to develop data-driven personalised medicine approaches for JSLE. Acknowledgments: Lupus UK, Rosetrees Trust, Versus Arthritis Disclosure of Interests: George Robinson: None declared, Junjie Peng: None declared, Pierre Dönnes: None declared, Leda Coelewij: None declared, Meena Naja: None declared, Anna Radziszewska: None declared, Chris Wincup: None declared, Hannah Peckham: None declared, David Isenberg Consultant of: Study Investigator and Consultant to Genentech, Yiannis Ioannou: None declared, Ines Pineda Torra: None declared, Coziana Ciurtin Grant/research support from: Pfizer, Consultant of: Roche, Modern Biosciences, Elizabeth Jury: None declared
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2020
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 80, No. Suppl 1 ( 2021-06), p. 8.1-8
    Abstract: Males and females have altered immune responses resulting in variation in autoimmune and cardiovascular disease risk (CVR). Recently, these differences have played a role in the inflammatory response to COVID-19. Sex differences exist in the frequency and activity of immune-cell subsets but mechanisms underlying sexual dimorphism remain unknown. Juvenile-onset systemic lupus erythematosus (JSLE) is an autoimmune disorder that commonly emerges during puberty, has a strong female prevalence (female:male ratio, 4.5:1) and results in an increased CVR. JSLE is characterised by chronic inflammation and dyslipidaemia, where cardiovascular disease is a leading cause of mortality for patients. Our previous work identified a link between immune cell function and lipid metabolism in adult-onset SLE. We hypothesised that sex hormones could influence both lipid metabolism and immune cell function and this could determine sex-specific susceptibility to JSLE and associated CVR. Objectives: We investigated the role of sex hormones in modifying systemic lipid metabolism and inflammation. Methods: Nuclear magnetic resonance spectroscopy based serum metabolomics measuring over 130 lipoproteins (14-subsets with lipid compositions), flow cytometry measuring immune-cells, and RNA-sequencing were used to assess the metabolic and immune profile in young, pre/post-pubertal males (n=10/17) and females (n=10/23) and in individuals with gender-dysphoria (GD) under cross-hormone treatment (trans-male/female, n=26/25). This analysis was also performed on a cohort of post-pubertal male (n=12) and female (n=23) JSLE patients. Data was analysed by logistic regression, balanced random forest machine learning (BRF-ML), differential gene expression (DEG) and pathway analysis. Results: Post-pubertal males had significantly reduced cardio-protective high-density lipoprotein (HDL) subsets (p 〈 0.0001) and increased cardio-pathogenic very-low-density lipoprotein subsets (p 〈 0.0001) compared to females. These differences were not observed pre-puberty and were reversed significantly by cross-hormone treatment in GD individuals, suggesting that sex hormones regulate lipid metabolism in-vivo . BRF-ML (28 immune-cell subsets) identified an increased frequency of anti-inflammatory regulatory T-cells (Tregs) in post-pubertal males compared to females (p=0.0097). These Tregs were also more suppressive in males compared to females. Differences in Treg frequency were seen pre-puberty and were not altered by sex hormone treatment in GD individuals. However, Treg DEGs and functional transcriptomic pathways altered between post-pubertal males and females, including those involved in inflammatory signalling, overlapped with those altered by hormones in GD, suggesting hormones may also drive Treg functional changes. In addition, HDL metabolites modified by hormones showed differential associations with Treg phenotypes between post-pubertal males and females. Strikingly, sex differences in lipoproteins and Tregs were lost in JSLE, suggesting hormone signalling could be dysregulated in the pathogenesis of autoimmunity and could increase CVR for patients. Conclusion: Sex hormones drive altered lipoprotein metabolism and functional transcriptomic pathways in Tregs. Males have a lipoprotein profile associated with increased CVR, but a more anti-inflammatory immune profile compared to females. Together, this could explain sex differences in inflammatory disease susceptibilities and inform future sex-specific therapeutic strategies for the management of both JSLE and CVR. Acknowledgements: Lupus UK Rosetrees Trust Versus Arthritis NIHR UCLH Biomedical Research Centre Disclosure of Interests: None declared
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 81, No. Suppl 1 ( 2022-06), p. 483.3-484
    Abstract: Cardiovascular disease (CVD) is a leading cause of mortality in patients with systemic lupus erythematosus (SLE, female:male ratio of 9:1) through accelerated atherosclerosis, the build-up of lipids and inflammation in the major artery walls, compared to age and sex matched healthy individuals. This is due to chronic inflammation, dyslipidaemia and other cardiometabolic defects that exacerbate with age (1). SLE in women aged of 35-44 increases the risk of coronary artery disease by 50 times and there is a 100-300-fold increased CVD-related mortality risk in young patients that develop SLE before the age of 18. Investigating metabolic defects in young patients and how they progress with age could help us understand the progressive mechanisms of atherosclerosis in SLE. Objectives This study investigated detailed changes in the metabolomic profile of female patients with SLE and matched healthy controls across age. Methods Serum NMR metabolomics ( 〉 250 metabolites, Nightingale) covering glycolysis metabolites, amino acids and 130 lipid measures was performed on serum from a cohort of female SLE patients (n=164, 13-72 years of age, median 35) and matched healthy controls (HCs, n=120, 15-76 years of age, median 36) and analysed by linear regression and Venn analysis. Multiple t-tests (corrected for multiple comparisons by false discovery rate) were used to assess unique metabolic changes by age group between SLE patients and HCs (≤25, n=62/43; 26-49, n=50/46; ≥50, n=52/31) and dysregulated metabolic pathways were assessed using metaboanalyst software. The metabolic impact of disease activity measures and treatments was assessed by Spearman correlations and unpaired t-tests respectively. Results Twenty-five metabolites were significantly altered in all SLE age groups compared to HCs, dominated by atheroprotective high density lipoprotein (HDL) subsets and their surface-bound peptide, apolipoprotein(Apo)A1, all of which were significantly decreased in SLE compared to HCs (p 〈 0.0001 in ages combined). In addition, the concentration of glycoprotein acetyls (GlycA, inflammatory biomarker) was increased in SLE in all age groups compared to HCs (p 〈 0.0001 in ages combined). Importantly, ApoA1 correlated negatively with disease activity measures (SLEDAI, p=0.005; BILAG, p=0.0009; dsDNA, p=0.003; ESR, p=0.0006) and positively with lymphocyte count (p=0.0005), whilst GlycA correlated positively with ESR (p 〈 0.0001) and CRP (p 〈 0.0001). Alternatively, metabolites unique to specific age groups in SLE compared to HCs included reduced amino acid subsets in the ≤25 age group, increased atherogenic very low density lipoproteins and reduced polyunsaturated fatty acids in the 26-49 age group, and increased atherogenic low density lipoproteins in the ≥50 age group. Separately, metabolites associated with the glycolysis pathway (p=0.004, metaboanalyist), including acetone, citrate, creatinine, glycerol, lactate and pyruvate, had significant positive correlations with age in SLE patients, but not in HCs. These metabolites were not significantly associated with disease activity measures. However, pyruvate (p=0.01) and lactate (p=0.009) were significantly upregulated in prednisolone treated patients, whilst citrate (p=0.002) and creatinine (p= 0.005) were downregulated in hydroxychloroquine treated patients. Conclusion Increasing HDL (ApoA1) levels whilst maintaining low disease activity in patients with SLE from a young age could improve cardiometabolic risk outcomes. This could be achieved through improved nutrition, lipid targeted therapies and better treatment strategies. Focusing on understanding and monitoring biomarkers of the glycolytic pathway could aid treatment decisions and help avoid adverse metabolic effects of current anti-inflammatory therapies in SLE (1). References [1]Robinson G.A, Pineda-Torra I, Ciurtin C, Jury E.C. Lipid metabolism in autoimmune rheumatic disease: implications for modern and conventional therapies . J Clin Invest. 2022;132(2):e148552. https://doi.org/10.1172/JCI148552 . Acknowledgements The authors would like to thank all of the patients and healthy blood donors, as well as Prof. Arne Akbar and Dr. Chris Wincup for additional patient and healthy donor samples. Disclosure of Interests None declared.
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Clinical Pathology, BMJ, Vol. 52, No. 9 ( 1999-09-01), p. 697-700
    Type of Medium: Online Resource
    ISSN: 0021-9746
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 1999
    detail.hit.zdb_id: 2028928-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 75, No. Suppl 2 ( 2016-06), p. 180.4-181
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2016
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 81, No. Suppl 1 ( 2022-06), p. 480.2-481
    Abstract: Sjögren’s syndrome (SS) is an autoimmune rheumatic disease characterised by dryness resulting from chronic lymphocytic infiltration of the exocrine glands. Patients also present with other extraglandular manifestations such as arthritis, anemia and fatigue or various organ and systems involvement. The disease is more frequent in women aged 30-50. However, in rare cases, the disease starts in childhood and is known as juvenile SS (JSS) or childhood SS. Children have different clinical manifestations compared to adults, with dryness being less common, making the diagnosis very challenging 1 . Objectives To investigate in depth the immune cell profile of patients with JSS for better understanding of disease pathogenesis. Methods Peripheral blood was collected from a cohort of patients with JSS while attending appointments at UCLH clinics. None had received B-cell depletion therapy. Immune-phenotyping of 29 immune-cell subsets, including B and T cells, in peripheral blood from patients with JSS (n=10) and age and sex-matched healthy controls (n=10) was performed using flow cytometry as we have performed previously for patients with adult onset SS 2 . Data were analysed using multiple t-tests and compared with the adult SS immune phenotype. Results Patients with JSS had an average age of 18 years (range 16-21) with an average age of disease onset at 14 years (range 12-18). Up to 60% of patients presented Anti-Ro autoantibodies while 50% presented Anti-La autoantibodies. Patients with JSS had an altered immune profile compared to age matched healthy controls (average of 18 years, range 15-25). In the B cell compartment, JSS patients had higher frequencies of Total CD19+ B cells (p=0.0044), Naïve B cells (CD19+IgD+CD27-) (p=0.0183) and bm2 (CD19+IgD+CD38+) (p=0.0490) whereas memory B cell subsets such as early bm5 (CD19+IgD-CD38+) and late bm5 (CD19+IgD-CD38-) were significantly reduced (p=0.0249, and p=0.0117 respectively), similar to the profile seen in patients with adult-SS. Interestingly, in the CD4+ T cell compartment, central memory (CD4+CD27+CD45RA-) T cells were significantly reduced (p= 〈 0,0001) but effector memory (CD4+CD27-CD45-) and effector memory-re-expressing-CD45RA (EMRA, CD4+CD27-CD45RA+) T-cell subsets were significantly elevated (p=0.0171 and p=0.0002 respectively). These changes were not identified in adult-SS patients. Finally, unlike our observations in patients with adult-onset SS there was no widespread deregulation of CD8+ T cell subsets in JSS patients; only a significant increase in CD8+CD25-CD127+ responders T cells (p=0.0392) was observed in JSS patients versus healthy. Conclusion This is the first pilot study investigating the immunophenotype profile of patients with JSS. Our preliminary findings suggest altered immune phenotypes in both B-cell and T cell compartments and for B cells are in concordance with previous immunophenotyping studies in adult SS (predominance of naïve and lower frequencies of memory B cells), suggesting an immunological rationale for the use of similar therapies. Further studies, comparing the adult with the juvenile phenotype could help stratify patients for targetted therapies and improve treatment in this rare disease in children for which no evidence-based recommnedations exist. References [1]Ciurtin C et al. Barriers to translational research in Sjögren’s syndrome with childhood onset: challenges of recognising and diagnosing an orphan rheumatic disease. Lancet Rheumatology. 2021; https://doi.org/10.1016/S2665-9913 (20)30393-3. [2]Martin-Gutierrez L, Peng J, et al. Stratification of Patients with Sjogren’s Syndrome and Patients With Systemic Lupus Erythematosus According to Two Shared Immune Cell Signatures, With Potential Therapeutic Implications. Arthritis Rheumatol. 2021;73(9):1626-37. Disclosure of Interests None declared.
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Gut, BMJ, Vol. 55, No. 11 ( 2006-11-01), p. 1553-1560
    Type of Medium: Online Resource
    ISSN: 0017-5749
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2006
    detail.hit.zdb_id: 1492637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 81, No. Suppl 1 ( 2022-06), p. 234.2-235
    Abstract: Systemic lupus erythematosus (SLE), a chronic, inflammatory autoimmune disease, predominantly affects women with a 9:1 female:male incidence. Cardiovascular disease (CVD) is a leading cause of mortality in SLE via accelerated atherosclerosis: the build-up of cells and lipids in the vascular wall and the main pathology underlying CVD. Objectives To define molecular profiles of SLE with subclinical atherosclerosis using multi-omics data analysis and clinical data in a well-characterised cohort of CVD-free SLE women. Methods Multi-omics analyses were conducted to explore the molecular signatures of SLE patients with (SLE-P) and without (SLE-NP) subclinical atherosclerosis defined by non-invasive ultrasound scanning of the carotid and femoral arteries. SLE blood CD14+ monocyte transcriptomes were investigated by bulk RNA-sequencing (SLE-P N=13, SLE-NP N=8), and targeted serum cardiometabolic and cardiovascular proteomics (OLINK) were used to explore matched protein expression (SLE-P N=17, SLE-NP N=20) (no difference in disease activity between groups). Bioinformatics approaches, including pathway and disease module enrichment analyses and extended protein-protein interaction networks, further defined molecular profiles of SLE patients with atherosclerosis from patients that remained plaque free. Gene signature-derived interferon (IFN) scores were applied to investigate heterogeneous subgroups within the cohort as a measure of inflammation. Results Distinct monocyte gene and protein expression profiles were identified in SLE and enriched in biological pathways relating to extracellular mechanisms, including purinergic and cytokine signalling. Lipid regulatory mechanisms were enriched in SLE-P, whereas SLE-NP patient’s transcriptome and proteome profiles were defined by pathways relating to inflammation. Specifically, the type-I IFN pathway was exclusively reduced in SLE-P compared to SLE-NP. IFN scores derived from published IFN-responsive gene expression signatures stratified patients into significantly distinct subgroups (high versus low IFN-response, p=0.0001) with 66% (N=14) of patients showing high IFN expression across multiple signatures not associated with age, ethnicity, or disease activity. However, IFN scores did not predict the presence of sub-clinical atherosclerosis and further heterogeneity was revealed with 46% of SLE-P patients showing a low IFN response (N=6). Further, a measure of plaque lipid content (echogenicity) was inversely correlated with IFN score (grey scale median, p=0.03, r=-0.8) which may reflect distinct plaque phenotypes between these subgroups relating to clinical presentation and risk of cardiovascular events. Conclusion Lipid dysregulation is a key mechanism that drives atherosclerosis pathology and genes and proteins relating to lipid metabolism distinguished SLE patients with and without subclinical atherosclerosis. Differences in levels of interferons and other inflammatory molecules may contribute to unique patterns of gene expression between SLE patients. A distinct subset of SLE-P patients showed low interferon expression, which may be suggestive of a dampened immune response in early subclinical CVD. Further elucidating the complexity of lipid dysregulation, inflammation and immune function in atherosclerosis in SLE will help improve patient stratification towards investigating the efficacy of anti-atherosclerotic therapies. Disclosure of Interests None declared
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Annals of the Rheumatic Diseases, BMJ, Vol. 75, No. Suppl 2 ( 2016-06), p. 947.2-947
    Type of Medium: Online Resource
    ISSN: 0003-4967 , 1468-2060
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2016
    detail.hit.zdb_id: 1481557-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...