Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by James Sheridan.
Lancet Neurology | 2010
Daniel Wynn; Michael Kaufman; Xavier Montalban; Timothy Vollmer; Jack Simon; Jacob Elkins; Gilmore O'neill; Lauri Neyer; James Sheridan; Chungchi Wang; Alice Fong; John Rose
BACKGROUND Daclizumab, a humanised monoclonal antibody, reduced multiple sclerosis disease activity in previous non-randomised studies. We aimed to assess whether daclizumab reduces disease activity in patients with active relapsing multiple sclerosis who are receiving interferon beta treatment. METHODS We did a phase 2, randomised, double-blind, placebo-controlled study at 51 centres in the USA, Canada, Germany, Italy, and Spain. Patients with active relapsing multiple sclerosis who were taking interferon beta were randomly assigned to receive add-on subcutaneous daclizumab 2 mg/kg every 2 weeks (interferon beta and high-dose daclizumab group), daclizumab 1 mg/kg every 4 weeks (interferon beta and low-dose daclizumab group), or interferon beta and placebo for 24 weeks. The randomisation scheme was generated by Facet Biotech. All patients and assessors were masked to treatment with the exception of Facet Biotech bioanalysts who prepared data for the data safety monitoring board or generated pharmacokinetic or pharmacodynamic data, a drug accountability auditor, and the site pharmacist. The primary endpoint was total number of new or enlarged gadolinium contrast-enhancing lesions measured on brain MRI scans every 4 weeks between weeks 8 and 24. Effects of daclizumab on prespecified subsets of lymphocytes and quantitative T-cell proliferative response were assessed in an exploratory pharmacodynamic substudy. Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00109161. FINDINGS From May, 2005, to March, 2006, 288 patients were assessed for eligibility, and 230 were randomly assigned to receive interferon beta and high-dose daclizumab (n=75), interferon beta and low-dose daclizumab (n=78), or interferon beta and placebo (n=77). The adjusted mean number of new or enlarged gadolinium contrast-enhancing lesions was 4.75 in the interferon beta and placebo group compared with 1.32 in the interferon beta and high-dose daclizumab group (difference 72%, 95% CI 34% to 88%; p=0.004) and 3.58 in the interferon beta and low-dose daclizumab group (25%, -76% to 68%; p=0.51). In the pharmacodynamic substudy, daclizumab was not associated with significant changes in absolute numbers of T cells, B cells, or natural killer cells, or T-cell proliferative response compared with interferon beta alone. The number of CD56(bright) natural killer cells was seven to eight times higher in both daclizumab groups than in the interferon beta and placebo group (interferon beta and low-dose daclizumab group p=0.002; interferon beta and high-dose daclizumab group p<0.0001). Common adverse events were equally distributed across groups. INTERPRETATION Add-on daclizumab treatment reduced the number of new or enlarged gadolinium contrast-enhancing lesions compared with interferon beta alone and might reduce multiple sclerosis disease activity to a greater extent than interferon beta alone. FUNDING Facet Biotech and Biogen Idec.
Multiple Sclerosis Journal | 2011
James Sheridan; Ying Zhang; Katherine Riester; Meina T Tang; Lyubov Efros; Jia Shi; Jeffrey Harris; Vladimir Vexler; Jacob Elkins
Objective: The objective of this study was to evaluate whether interleukin-2 (IL-2) receptor expression on CD56bright natural killer (NK) cells predicted CD56bright NK cell expansion and therapeutic response to daclizumab (DAC) in multiple sclerosis (MS). Methods: DAC exposure, CD56bright NK cell counts, IL-2 receptor alpha (CD25) and beta (CD122) subunits, and new or enlarged lesions on brain MRI were measured in 64 subjects in a pharmacokinetic/pharmacodynamic substudy of the phase 2 CHOICE trial at multiple time points. Peripheral blood mononuclear cell (PBMC) samples were obtained from healthy subjects to assess the relationship among DAC treatment, intermediate affinity IL-2 signaling, and CD56bright NK cell expansion. Results: Increased CD56bright NK cell counts in DAC/interferon beta (IFNβ)-treated subjects were observed by day 14, the first post-dosing time point (mean [SD] ln{CD56bright NK cell count}: DAC high/IFNβ, 2.01 [1.25]; DAC low/IFNβ, 2.29 [1.06]; placebo/IFNβ, 1.01 [1.03]; adjusted p = 0.003), and persisted throughout the treatment period. Higher DAC dose predicted a faster rate of CD56bright NK cell expansion (p < 0.001), but individual subjects’ increases in CD56bright NK cells from baseline levels were only weakly correlated with DAC exposure (r2 = 0.167). Higher expression of the intermediate-affinity IL-2 receptor subunit (CD122) on CD56bright NK cells at baseline predicted fewer new gadolinium-enhanced (Gd+) lesions during the treatment period (1.77 vs. 0.62 adjusted mean new Gd+ lesions during weeks 8–24, lowest vs. highest quartile of percentage CD122+ CD56bright NK cells; p = 0.033) and a greater increase in CD56bright NK cell counts at the end of DAC dosing (p = 0.029). Conclusion: CD56bright NK cell expansion after DAC treatment appears to reflect individual differences in the capacity for intermediate-affinity IL-2 signaling and could provide a basis for predicting clinical response to DAC in MS.
Journal of Immunology | 2015
David J. Huss; Devangi Mehta; Akanksha Sharma; Xiaojun You; Katherine Riester; James Sheridan; Lakshmi Amaravadi; Jacob Elkins; Jason D. Fontenot
Regulatory T cells (Tregs) mediate immune tolerance to self and depend on IL-2 for homeostasis. Treg deficiency, dysfunction, and instability are implicated in the pathogenesis of numerous autoimmune diseases. There is considerable interest in therapeutic modulation of the IL-2 pathway to treat autoimmunity, facilitate transplantation tolerance, or potentiate tumor immunotherapy. Daclizumab is a humanized mAb that binds the IL-2 receptor α subunit (IL-2Rα or CD25) and prevents IL-2 binding. In this study, we investigated the effect of daclizumab-mediated CD25 blockade on Treg homeostasis in patients with relapsing-remitting multiple sclerosis. We report that daclizumab therapy caused an ∼50% decrease in Tregs over a 52-wk period. Remaining FOXP3+ cells retained a demethylated Treg-specific demethylated region in the FOXP3 promoter, maintained active cell cycling, and had minimal production of IL-2, IFN-γ, and IL-17. In the presence of daclizumab, IL-2 serum concentrations increased and IL-2Rβγ signaling induced STAT5 phosphorylation and sustained FOXP3 expression. Treg declines were not associated with daclizumab-related clinical benefit or cutaneous adverse events. These results demonstrate that Treg phenotype and lineage stability can be maintained in the face of CD25 blockade.
Multiple Sclerosis Journal | 2014
Ying Zhang; M McClellan; Lyubov Efros; D Shi; Bibiana Bielekova; Mt Tang; Vladimir Vexler; James Sheridan
Daclizumab is a humanized monoclonal antibody that prevents interleukin-2 (IL-2) binding to CD25, blocking IL-2 signaling by cells that require high-affinity IL-2 receptors to mediate IL-2 signaling. The phase 2a CHOICE study evaluating daclizumab as a treatment for multiple sclerosis (MS) included longitudinal analysis of activated T cell counts. Whereas an exposure-dependent relationship was observed between daclizumab and reductions in HLA-DR+-activated T cells, a similar relationship was not observed for reductions in CD25 levels. The objective of this report is to determine the mechanism by which daclizumab reduces CD25 levels on peripheral blood mononuclear cells (PBMCs) using cytometric techniques. Daclizumab reduced T cell CD25 levels through a mechanism that required the daclizumab-Fc domain interaction with Fc receptors (FcR) on monocytes, but not on natural killer (NK) cells, and was unrelated to internalization or cell killing. Activated CD4+ T cells and FoxP3+ Treg cells showed evidence of trogocytosis of the CD25 antigen in the presence of monocytes. A daclizumab variant that retained affinity for CD25 but lacked FcR binding did not induce trogocytosis and was significantly less potent as an inhibitor of IL-2-induced proliferation of PBMCs. In conclusion, Daclizumab-induced monocyte-mediated trogocytosis of CD25 from T cells appears to be an additional mechanism contributing to daclizumab inhibition of IL-2 signaling.
Expert Review of Clinical Pharmacology | 2014
James Sheridan; Randy R. Robinson; John Rose
Daclizumab is a monoclonal antibody specific for the IL-2R α chain (CD25). Daclizumab has been observed to have multiple mechanisms of action, which may contribute to beneficial effects in immune-related disease and particularly in relapsing and remitting multiple sclerosis (RRMS). These include inhibition of activated immune cells, increase of regulatory natural killer cells, effects on dendritic cells, inhibition of innate lymphoid tissue inducer cells and altered responses involving IL-2 transpresentation. The antibody has shown considerable promise in open-label and early Phase II clinical trials when used as a monotherapy, or in combination with IFN-β. In recently completed randomized trials in RRMS, treatment with daclizumab monotherapy compared with placebo resulted in clinically meaningful and statistically significant reductions in relapses, active lesions on brain MRI and slowing of disability progression. A large Phase III trial in RRMS is ongoing.
mAbs | 2016
Bishu Ganguly; Balaji Balasa; Lyubov Efros; Paul R. Hinton; Stephen Hartman; Archana B. Thakur; Joanna M. Xiong; Brian Schmidt; Randy R. Robinson; Thierry Sornasse; Vladimir Vexler; James Sheridan
ABSTRACT The CD25-binding antibody daclizumab high-yield process (DAC HYP) is an interleukin (IL)-2 signal modulating antibody that shares primary amino acid sequence and CD25 binding affinity with Zenapax®, a distinct form of daclizumab, which was approved for the prevention of acute organ rejection in patients receiving renal transplants as part of an immunosuppressive regimen that includes cyclosporine and corticosteroids. Comparison of the physicochemical properties of the two antibody forms revealed the glycosylation profile of DAC HYP differs from Zenapax in both glycan distribution and the types of oligosaccharides, most notably high-mannose, galactosylated and galactose-α-1,3-galactose (α-Gal) oligosaccharides, resulting in a DAC HYP antibody material that is structurally distinct from Zenapax. Although neither antibody elicited complement-dependent cytotoxicity in vitro, DAC HYP antibody had significantly reduced levels of antibody-dependent cell-mediated cytotoxicity (ADCC). The ADCC activity required natural killer (NK) cells, but not monocytes, suggesting the effects were mediated through binding to Fc-gamma RIII (CD16). Incubation of each antibody with peripheral blood mononuclear cells also caused the down-modulation of CD16 expression on NK cells and the CD16 down-modulation was greater for Zenapax in comparison to that observed for DAC HYP. The substantive glycosylation differences between the two antibody forms and corresponding greater Fc-mediated effector activities by Zenapax, including cell killing activity, manifest as a difference in the biological function and pharmacology between DAC HYP and Zenapax.
Journal of Neuroimmunology | 2014
David J. Huss; Devangi Mehta; Akanksha Sharma; Xiaojun You; Katherine Riester; James Sheridan; Lakshmi Amaravadi; Jacob Elkins; Jason D. Fontenot
Regulatory T cells (Tregs) mediate immune tolerance to self and depend on IL-2 for homeostasis. Treg deficiency, dysfunction, and instability are implicated in the pathogenesis of numerous autoimmune diseases. There is considerable interest in therapeutic modulation of the IL-2 pathway to treat autoimmunity, facilitate transplantation tolerance, or potentiate tumor immunotherapy. Daclizumab is a humanized mAb that binds the IL-2 receptor a subunit (IL-2R a or CD25) and prevents IL-2 binding. In this study, we investigated the effect of daclizumab-mediated CD25 blockade on Treg homeostasis in patients with relapsing-remitting multiple sclerosis. We report that daclizumab therapy caused an ~50% decrease in Tregs over a 52-wk period. Remaining FOXP3+ cells retained a demethylated Treg-specific demethylated region in the FOXP3 promoter, maintained active cell cycling, and had minimal production of IL-2, IFN- g, and IL-17. In the presence of daclizumab, IL-2 serum concentrations increased and IL-2R bg signaling induced STAT5 phosphorylation and sustained FOXP3 expression. Treg declines were not associated with daclizumab-related clinical benefit or cutaneous adverse events. These results demonstrate that Treg phenotype and lineage stability can be maintained in the face of CD25 blockade.
Neurology | 2012
Jacob S. Elkins; James Sheridan; Lakshmi Amaravadi; Katherine Riester; Gilmore O'Neill
Archive | 2017
Lakshmi Amaravadi; Jacob S. Elkins; Katherine Riester; James Sheridan; David J. Huss; Devangi Mehta; Akanksha Sharma
Neurology | 2016
Sami Fam; Oksana Mokliatchouk; Devangi Mehta; Katherine Riester; James Sheridan; Peter McCroskery; Jacob Elkins; Lakshmi Amaravadi