receptor monoclonal antibody
Recently Published Documents


TOTAL DOCUMENTS

269
(FIVE YEARS 26)

H-INDEX

52
(FIVE YEARS 1)

2021 ◽  
Vol 23 (5) ◽  
pp. 739-748
Author(s):  
V. A. Vizir ◽  
A. S. Sadomov ◽  
O. V. Demidenko

The aim is to familiarize practitioners with the clinical case of tocilizumab use in the combination treatment of the coronavirus disease (COVID-19) patient with concomitant rheumatoid arthritis. Materials and methods. The clinical case shows our own follow-up of COVID-19 clinical course in the patient with concomitant rheumatoid arthritis during combination treatment with the use of a recombinant humanized anti-interleukin-6 receptor monoclonal antibody tocilizumab. Results. The patient with a severe COVID-19 course, whose examination and treatment results are given in the article, was comorbid for rheumatoid arthritis. The cytokine storm development at the hospital stage was confirmed by an increase in markers of systemic inflammation: C-reactive protein, D-dimer, fibrinogen, an almost 50-fold increase in serum interleukin-6 level, as well as absolute and relative lymphocytopenia. Despite the anti-inflammatory therapy administered with systemic corticosteroids, the patient’s condition progressively worsened. After assessing the indications and contraindications, it was decided to use the interleukin-6 receptor inhibitor tocilizumab, followed by rapid clinical, laboratory and X-ray positive response to the treatment. The understanding of tocilizumab use in patients with COVID-19 at the current stage was formed based on the comparative analysis of our own clinical case data and the results of relevant clinical trials, world recommendations and guidelines. Conclusions. The use of recombinant humanized anti-interleukin-6 receptor monoclonal antibody tocilizumab in the combination treatment of severe COVID-19 with concomitant rheumatoid arthritis is pathogenetically based and decreases the main clinical and laboratory signs of cytokine storm, respiratory failure, improves chest x-ray findings and reduces the length of hospital stay. Further large randomized placebo-controlled trials including the population of patients with various comorbid conditions are needed to clarify conclusively the place and role of anti-cytokine drugs in the treatment of COVID-19 patients.


2021 ◽  
Vol 80 (Suppl 1) ◽  
pp. 600.1-600
Author(s):  
A. Abdul Razzack ◽  
S. Abdul Razzack ◽  
P. Shenasan ◽  
N. Shenasan ◽  
S. Mishra ◽  
...  

Background:Type I interferons such as Anifrolumab have been implicated in Systemic lupus erythematosus (SLE) pathogenesis on the basis of increased interferon-stimulated gene expression and genetic susceptibility. Little is known regarding its efficacy and safety profile.Objectives:To assess the efficacy and safety of Anifrolumab in patients with SLE.Methods:Electronic databases (PubMed, Embase, Scopus, Cochrane) were searched from inception until December 15th, 2020. Unadjusted odds ratios (OR) were calculated from dichotomous data using Mantel Haenszel (M-H) random-effects with statistical significance to be considered if the confidence interval excludes 1 and p<0.05. The primary outcome of interest was British Isles Lupus Assessment Group (BILAG)-based Composite Lupus Assessment (BICLA). Secondary outcomes included the proportion of patients who achieved an SLE responder index of 4 (SRI-4) reduction of 50% or more in the Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI), reductions in the glucocorticoid dose and adverse effects.Results:A total of three studies1,2,3 with 839 participants (Anifrolumab=372, Placebo=467) were included in our analysis. Follow-up duration was at week 52. A statistically significant different was observed in the Anifrolumab arm in terms of BICLA response (OR 0.44 95%CI 0.34-0.59;p < 0.00001, I2=4), ≥50% reduction in CLASI activity score (OR 0.36 95%CI 0.21-0.60;p=0.0001, I2=0), glucocorticoid reduction (OR 0.41 95%CI 0.28-0.59;p<0.00001; I2=0) and SRI-4 response (OR 0.52 95% CI 0.30-0.90; p=0.02, I2=75). However, Adverse events were less likely in the placebo arm as compared to Anifrolumab (OR 1.54 95%CI 1.05-2.25; p=0.03; I2=0).Conclusion:Anifrolumab was found to be more effective than placebo for the management of SLE, but may also cause more severe adverse effects.References:[1]Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez C, Bae SC, Brohawn PZ, Pineda L, Berglind A, Tummala R; TULIP-2 Trial Investigators. Trial of Anifrolumab in Active Systemic Lupus Erythematosus. N Engl J Med. 2020 Jan 16;382(3):211-221. doi: 10.1056/NEJMoa1912196. Epub 2019 Dec 18. PMID: 31851795.[2]Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, Illei GG, Drappa J, Wang L, Yoo S; CD1013 Study Investigators. Anifrolumab, an Anti-Interferon-α Receptor Monoclonal Antibody, in Moderate-to-Severe Systemic Lupus Erythematosus. Arthritis Rheumatol. 2017 Feb;69(2):376-386. doi: 10.1002/art.39962. PMID: 28130918; PMCID: PMC5299497.[3]Furie RA, Morand EF, Bruce IN, et al. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): a randomised, controlled, phase 3 trial. Lancet Rheumatol 2019; 1(4):e208-e219.Disclosure of Interests:None declared


2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
Ileana Peride ◽  
Andrei Niculae ◽  
Corina Adriana Balas ◽  
Ana-Maria Nechita ◽  
Ionel Alexandru Checherita

Abstract Background and Aims Since the beginning of 2019, once COVID-19 pandemic was declared, there is a keen interest in understanding the impact of SARS-CoV-2 infection on chronic kidney disease (CKD) patients, regarding the influence on CKD progression and the suitable therapy options, as most of the indicated medications are contraindicated for a glomerular filtration rate (GFR) below 30 ml/min, and, in addition, there is a little experience in dialyzed patients. The aim of our single-center pilot study is to determine the influence of SARS-CoV-2 infection on CKD patients’ (dialyzed or not) outcome and hospitalization rate. Method We evaluated the patients diagnosed with COVID-19, admitted in our Department between October and December 2020. The inclusion criteria were: age &gt; 18 years old, diagnosis of CKD – predialysis and hemodialyzed patients. The exclusion criteria were: patients without pre-existing CKD. All included subjects signed the patients’ consent. To all included patients we performed the following tests: total blood count, erythrocyte sedimentation rate, C-reactive protein, fibrinogen, ferritin, serum free iron, serum creatinine, urea, uric acid, calcemia, total proteins, electrolytes and acid-base balance, urinary exams (including urine culture), coagulation and lipid profile, quantitative D-dimer, IL-6, procalcitonin, and imaging tests (CT, pulmonary Rx, abdominal ultrasonography). The patients were monitored by the infectious disease medical team that adjusted the therapy according to the patients’ lab and imagistic results. The specific treatment for SARS-CoV-2 infection included primary anti-interleukin receptor monoclonal antibody drugs (such as Anakinra, Tocilizumab), corticotherapy (dexamethasone), anti-retroviral therapy (remdesivir, favipiravir) only in hemodialyzed patients or in those presenting an eGFR &gt; 30 mL/min, antibiotics, antifungal drugs, and oxygen-therapy. Usually, anti-interleukin receptor monoclonal antibody consisted in 7 doses, administrated every 48 hours. The dose of all other recommended drugs was adapted according to the patients’ eGFR. Results A total of 63 patients were admitted in our Department and were under our care, presenting medium or severe forms of SARS-CoV-2 infection. After applying the inclusion and exclusion criteria, only 38 patients were considered eligible: 21 male patients (mean age 63.52 ± 13.82 years), and 17 female patients (mean age 67.24 ± 12.83 years). 31.57% represented the percentage of death during the hospitalization (due to the severity of the disease, 4 patients died within 24 hours) in patients presenting heterogenous comorbidities, such as diabetes mellitus, hypertension, pre-existing glomerulonephritis and/or oncological pathologies; we also noticed that female gender represented 58.33% of the deceased patients. The mean hospitalization period in the deceased patients was 6.42 ± 5.38 days – 4 ± 3.21 days in female gender, and 9.8 ± 6.30 days in male gender. Conclusion Most of our patients, although diagnosed with medium and severe forms of SARS-CoV-2 infection, presented a favorable evolution, and an adequate response to the specific medication. We observed that most of the deceased cases were female patients, and compared to the male deceased subject, female deceased patients presented a lower period of hospitalization. Therefore, probably female CKD patients with comorbidities and diagnosed with COVID-19 are more predisposed to an unfavorable prognosis. Further and larger clinical trials are necessary to validate the impact of SARS-CoV-2 infection on mortality in CKD patients.


PLoS ONE ◽  
2021 ◽  
Vol 16 (3) ◽  
pp. e0249049
Author(s):  
Karen Liao ◽  
Keguan Chen ◽  
Sara Brett ◽  
Andrew Gehman ◽  
Ann M. Schwartz ◽  
...  

Interleukin-7 (IL-7) signaling modulates T cell activity and is implicated in numerous autoimmune diseases. An anti-IL-7 receptor monoclonal antibody (GSK2618960) biotherapeutic was evaluated in healthy subjects for safety, pharmacokinetics (PK), pharmacodynamics (PD) and immunogenicity in a single-dose escalation phase I study. We found that antibodies against GSK2618960 (i.e., anti-drug antibodies or ADA) developed in 83% and 100% of GSK2618960-treated subjects in the 0.6 and 2.0 mg/kg dose cohorts, respectively. Of the ADA positive subjects, 64% (7 of 11) had detectable neutralizing activity. Further investigation revealed the presence of GSK2618960-specific memory B cells, indicating the development of immunological memory for the ADAs. Ex vivo stimulation of peripheral blood mononuclear cell (PBMC) samples demonstrated a relatively strong CD4+ T cell proliferation response to GSK2618960 as compared to the control anti-RSV antibody (which is known to have only low immunogenic potential), confirming the high immunogenic potential of GSK2618960. Furthermore, GSK2618960 was found to bind in vitro monocyte-derived dendritic cells (DCs). GSK2618960 treatment of PBMCs increased the proportion of DC cells showing an increase in expression of CD83, CD86 and CD209, which indicated enhanced DC differentiation and activation relative to the isotype control anti-β amyloid antibody. Collectively, the evidence supports that the high incidence of observed clinical immunogenicity was likely related to the receptor-mediated activity by GSK2618960.


2021 ◽  
Vol 12 ◽  
Author(s):  
Marika Bogdani ◽  
Linda Faxius ◽  
Malin Fex ◽  
Anita Ramelius ◽  
Anya Wernersson ◽  
...  

The depleting Vβ13a T cell receptor monoclonal antibody (mAb) 17D5 prevents both induced and spontaneous autoimmune diabetes in BB rats. Here it was tested in congenic DRLyp/Lyp rats, all of which spontaneously developed diabetes. Starting at 40 days of age, rats were injected once weekly with either saline, His42 Vβ16 mAb, or 17D5 mAb and monitored for hyperglycemia. Diabetes occurred in 100% (n = 5/5) of saline-treated rats (median age, 66 days; range 55–73), and in 100% (n = 6/6) of His42-treated rats (median age, 69 days; range 59–69). Diabetes occurred in fewer (n = 8/11, 73%) 17D5-treated rats at a later age (median 76 days, range 60–92). Three (27%) of the 17D5-treated rats were killed at 101–103 days of age without diabetes (17D5 no-diabetes rats). Survival analysis demonstrated that 17D5 mAb delayed diabetes onset. Saline- and His42-treated rats had severely distorted islets with substantial loss of insulin-positive cells. These rats exhibited prominent hyaluronan (HA) staining, with the intra-islet HA+ accumulations measuring 5,000 ± 2,400 µm2 and occupying 36 ± 12% of islet area, and severe (grade 4) insulitis with abundant infiltration by CD68+, CD3+, and CD8+ cells. The 17D5 mAb-treated rats with delayed diabetes onset exhibited less severe insulitis (predominantly grade 3). In contrast, the 17D5 no-diabetes rats had mostly normal islets, with insulin+ cells representing 76 ± 3% of islet cells. In these rats, the islet HA deposits were significantly smaller than in the diabetic rats; the intra-islet HA+ areas were 1,200 ± 300 µm2 and accounted for 8 ± 1% of islet area. Also, islet-associated CD68+ and CD3+ cells occurred less frequently (on average in 60 and 3% of the islets, respectively) than in the diabetes rats (present in &gt;95% of the islets). No CD8+ cells were detected in islets in all 17D5 no-diabetes rats. We conclude that mAb 17D5 delayed diabetes in DRLyp/Lyp rats and markedly reduced expression of HA and concomitant infiltration of CD68+, CD3+, and CD8+ cells. Our findings underscore the importance of refining immune suppression in prevention or intervention clinical trials to use mAb reagents that are directed against specific T cell receptors.


2021 ◽  
Vol 24 ◽  
pp. 1-15
Author(s):  
Mitsutoshi Kimura ◽  
Kazuki Kurihara ◽  
Hajime Moteki ◽  
Masahiko Ogihara

Background: We investigated the signal transduction pathway associated with growth hormone (GH)-stimulated DNA synthesis and proliferation in primary cultured hepatocytes. Methods: Adult rat hepatocytes were isolated from normal livers by two-step in situ collagenase perfusion to facilitate disaggregation of the adult rat liver. Then hepatocytes were cultured in serum-free Williams’ medium E supplemented with GH (1-100 ng/ml) in the presence or absence of test reagents. GH-induced hepatocyte DNA synthesis and proliferation were determined, and the phosphorylation activities of Janus kinase (JAK) 2 (JAK2) (p125 kDa), p95-kDa RTK, and ERK1/2 were measured by western blotting. Results: Hepatocytes grown in serum-free defined medium proliferated within 5 h of culture in the presence of GH (100 ng/ml) in a concentration- and time-dependent manner (EC50 75 ng/ml). These proliferative effects of GH were almost completely blocked by an anti-GH receptor monoclonal antibody (85 ng/ml) and an anti-insulin-like growth factor (IGF)-I receptor monoclonal antibody. In addition, the proliferative effects of GH were significantly blocked by a JAK2 inhibitor (TG101209, 10−6 M), as well as specific signal-transducing inhibitors of phospholipase C (PLC; U-73122, 10−6 M), RTK (AG538, 10−6 M), phosphoinositide 3-kinase (PI3K; LY294002, 10−6 M), mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK; PD98059, 10−6 M), and mammalian target of rapamycin (mTOR; rapamycin, 10 ng/ml). GH significantly induced the phosphorylations of JAK2 (p125 kDa), p95-kDa IGF-I receptor tyrosine kinase (RTK), and ERK2 in this order according to western blotting analysis. Conclusions: The proliferative action of GH is mediated by two main signaling pathways. One includes activation of the GH receptor/JAK2/PLC/Ca2+ pathway, and the other involves activation of the p95-kDa IGF-I RTK/PI3K/ERK2/mTOR pathway in primary cultures of adult rat hepatocytes.


Doctor Ru ◽  
2021 ◽  
Vol 20 (3) ◽  
pp. 70-72
Author(s):  
A.V. Aksenov ◽  
◽  
E.A. Ivanovskaya ◽  
◽  

Objective of the Paper: To demonstrate a case of successful use of tocilizumab in a child with systemic juvenile idiopathic arthritis (JIA). Key Points. A clinical variant of JIA is systemic arthritis (systemic JIA). Currently, management of systemic JIA includes tocilizumab, a recombinant humanised anti–interleukin-6 receptor monoclonal antibody. The article describes a case of successful use of tocilizumab in a child with systemic JIA after inefficient therapy with glucocorticosteroids and methotrexate. Conclusion. Systemic JIA is the severest clinical JIA variant. Tocilizumab allows achieving the quiescent disease stage where the standard therapy with glucocorticosteroids and methotrexate is ineffective. Keywords: system arthritis, juvenile idiopathic arthritis, tocilizumab, children.


Sign in / Sign up

Export Citation Format

Share Document