National Institute of Neuroscience, National Center of Neurology and Psychiatry

Author Of 1 Presentation

Neuromyelitis Optica and Anti-MOG Disease Oral Presentation

FC01.03 - Effect of satralizumab on relapse severity in neuromyelitis optica spectrum disorder (NMOSD): results from the Phase III SAkura studies

Speakers
Presentation Number
FC01.03
Presentation Topic
Neuromyelitis Optica and Anti-MOG Disease
Lecture Time
13:24 - 13:36

Abstract

Background

NMOSD is an autoimmune disorder characterized by acute, unpredictable relapses that result in accumulating disability. Satralizumab, a humanized, monoclonal recycling antibody that targets the interleukin-6 receptor, reduced relapse frequency and had a favourable safety profile vs placebo in two randomized, phase 3 clinical trials: SAkuraSky (satralizumab in combination with baseline immunosuppressants; NCT02028884), and SAkuraStar (satralizumab monotherapy; NCT02073279).

Objectives

To assess the impact of satralizumab on relapse severity in patients with NMOSD.

Methods

Patients in the SAkura studies received satralizumab 120mg or placebo at Weeks 0, 2, 4, and Q4W thereafter. This analysis was performed using data from the pooled intention-to-treat population across the double-blind periods of both studies. We assessed the severity of protocol-defined relapses (PDRs) by comparing patients’ Expanded Disability Status Scale (EDSS) score at PDR vs their score prior to relapse (last scheduled study visit). A similar analysis on optic neuritis PDRs was performed using visual Functional Systems Score (FSS). A PDR was categorised as severe if it resulted in a change of ≥2 points on the EDSS or visual FSS (optic neuritis analysis). Kaplan-Meier analyses were performed to assess time to first severe PDR. Additionally, the number of patients receiving acute therapy for any relapse was compared between treatment groups.

Results

Overall, 178 patients were included in the analyses. In the satralizumab group, 27 of 104 patients (26%) experienced a PDR vs 34 of 74 patients (46%) in the placebo group. The proportion of PDRs that were severe was lower in patients receiving satralizumab vs placebo (5 of 27 events [19%] vs 12 of 34 events [35%]). Similarly, the proportion of optic neuritis PDRs that were severe was lower in patients receiving satralizumab vs placebo (2 of 8 events [25%] vs 5 of 13 events [39%]). Across all patients, there was a 79% reduction in severe PDR risk with satralizumab vs placebo (hazard ratio [95% CI]; 0.21 [0.07–0.61]; p=0.002). A lower proportion of patients receiving satralizumab were prescribed acute relapse therapy vs placebo (38% vs 58%; odds ratio [95% CI] 0.46 [0.25–0.86], p=0.015).

Conclusions

Patients treated with satralizumab had a lower risk of severe relapse, and were less likely to receive acute relapse therapy compared with placebo. The number of patients with severe PDRs was low, so results should be interpreted with caution.

Collapse

Author Of 3 Presentations

Microbiome Poster Presentation

P0677 - Possible association between miRNAs and gut microbiota in pathogenesis of Multiple sclerosis (ID 1624)

Speakers
Presentation Number
P0677
Presentation Topic
Microbiome

Abstract

Background

Multiple Sclerosis (MS) is an autoimmune demyelinating disease of CNS, involving interplay of multiple genetic and environmental factors, which leads to a chronic activation of the immune cells targeting CNS autoantigen. Involvement of microbiota has been correlated with the pathogenesis of MS, further mechanistic analyses are needed. Circulating extracellular vesicles(EVs) including exosomes play an important role in many signaling pathway by regulating gene expression, and the role of exosomal microRNA in MS has been demonstrated in our previous study (Kimura et al. Nature Comm 2018). Here we explored if extracellular vesicles including exosomes can be a communication tool between gut microbiota and the host immune system.

Objectives

Role of gut microbiome in generation of circulating exosomes and miRNAs.

Elucidate the relationship between circulating miRNA and gut microbiota in the pathogenesis of MS, using EAE model.

Methods

We generated gut microbiome dysbiosis model mice by oral administration of non-absorbing antibiotics cocktail (ABX) containing kanamycin, vancomycin and colistin. The model mice were subjected to Experimental autoimmune encephalomyelitis (EAE) by injecting MOG35-55 peptide in CFA. Exosome concentrations in the sera were quantified using enzymatic analysis. MOG tetramer35-55 reactive CD4+ T cells (%) were evaluated for lymphocytes isolated from the CNS, spleen and blood. For cell-free miRNA analysis, total RNA was isolated from the plasma by a Plasma/Serum. Circulating and exosomal RNA Purification kit and miRNA expression analysis was performed by a DNA chip of mouse miRNAs and TaqMan miRNA assay.

Results

Dysbiosis of gut microbiome was shown to ameliorate signs of EAE, along with a notable reduction in the migration of total T cells and MOG tetramer35-55 reactive CD4+ T cells to the CNS. We also revealed substantial changes in the circulating exosome and significant decreases in the expression of exosomal miRNAs, including miR-21a-5p, miR-146-5p, and miR-223-3p. Notably, these miRNAs can directly bind to the 3` UTR region of major genes that controls the T cell migration to the CNS in EAE. The results indicate that gut microbiome would significantly influence the T cell trafficking through exosomal miRNAs in EAE.

Conclusions

Our data support that exosomes and exosomal miRNAs could be the major source of communication between gut microbiome and host immune response.

Collapse
Neuromyelitis Optica and Anti-MOG Disease Poster Presentation

P0711 - Efficacy of satralizumab in neuromyelitis optica spectrum disorder (NMOSD): Results from open-label extension periods of SAkuraSky and SAkuraStar (ID 1319)

Speakers
Presentation Number
P0711
Presentation Topic
Neuromyelitis Optica and Anti-MOG Disease

Abstract

Background

Satralizumab, a humanized, monoclonal recycling antibody that targets the interleukin-6 receptor, reduced patients’ risk of NMOSD relapse in the double-blind (DB) periods of two randomized, phase 3 clinical trials in NMOSD: SAkuraSky (satralizumab in combination with baseline immunosuppressants; NCT02028884), and SAkuraStar (satralizumab monotherapy; NCT02073279).

Objectives

To assess the efficacy of satralizumab over a longer period of treatment, using data from the SAkura studies’ open-label extension (OLE) periods.

Methods

Patients entering SAkuraSky/Star were randomized to receive satralizumab 120mg or placebo at Weeks 0, 2, 4, and Q4W thereafter. After completing the DB period or experiencing a relapse, patients could enter the OLE period (same satralizumab dosing as DB period). The primary endpoint of both studies was time to first protocol-defined relapse (PDR) in the DB period, adjudicated by a Clinical Endpoint Committee (CEC). In this analysis, which includes OLE data (CEC adjudication unavailable), we assessed time to first investigator-reported PDR (any relapse considered by the investigator to meet PDR criteria) in the combined DB+OLE periods, using a pooled population from both studies.

Results

Overall, 179 patients were randomized to treatment (satralizumab n=105; placebo n=74), of whom 166 received ≥1 dose of satralizumab in the combined DB+OLE period. The median (range) satralizumab exposure in the DB period was 96.1 (8–224) weeks, and in the combined DB+OLE was 131.9 (13–276) weeks.

In the combined DB+OLE, patients originally randomized to satralizumab had a 51% lower risk of investigator-reported PDR vs those originally randomized to placebo (HR [95% CI] 0.49 [0.31–0.79]; P=0.002); the risk reduction was more pronounced in AQP4-IgG seropositive patients (66% risk reduction; HR [95% CI] 0.34 [0.19–0.62]; P<0.001). Patients who switched from placebo to satralizumab upon entry into the OLE period were included in the placebo group for this analysis, which likely reduced the observed treatment difference between satralizumab and placebo compared with the DB period.

No patients randomized to satralizumab withdrew from the OLE period due to a relapse, vs four patients who were originally randomized to placebo. The safety profile of satralizumab in the OLE was consistent with the DB period.

Conclusions

Across the DB and OLE periods of the SAkura studies, patients randomized to satralizumab had a significantly reduced risk of relapse vs placebo.

Collapse
Neuromyelitis Optica and Anti-MOG Disease Poster Presentation

P0753 - Safety of satralizumab based on pooled data from phase 3 studies in patients with neuromyelitis optica spectrum disorder (ID 1375)

Speakers
Presentation Number
P0753
Presentation Topic
Neuromyelitis Optica and Anti-MOG Disease

Abstract

Background

Interleukin-6 (IL-6) is implicated in the immunopathology of neuromyelitis optica spectrum disorder (NMOSD). Satralizumab, a humanized recycling monoclonal antibody that inhibits the IL-6 receptor, demonstrated a reduction in NMOSD relapse risk in two phase 3 studies: SAkuraSky (satralizumab in combination with baseline immunosuppressants; NCT02028884), and SAkuraStar (satralizumab monotherapy; NCT02073279).

Objectives

To evaluate the safety of satralizumab vs placebo in a pooled population of NMOSD patients from the SAkura studies, including the latest data from the open-label extension (OLE) period of the studies.

Methods

SAkuraStar and SAkuraSky are randomized studies comprising a double-blind (DB) period (satralizumab 120mg Q4W vs placebo) followed by an OLE period (satralizumab only). The combined DB and extension period was defined as the overall satralizumab treatment (OST) period (cut-off: 7 Jun 2019). Safety was evaluated in the DB and OST periods and reported as adverse event (AE) rates per 100 patient-years (PY).

Results

The pooled DB population included 178 patients (satralizumab, n=104; placebo, n=74), and 166 patients received satralizumab in the OST. Median duration of safety observation with satralizumab was 96.1 weeks in the DB period and 131.9 weeks in the OST period. Rates of AEs and serious AEs were comparable between satralizumab and placebo groups in the DB period (AEs: 478.49 vs 506.51 events/100PY, respectively; serious AEs: 14.97 vs 17.98 events/100PY, respectively), and were consistent in the OST period. In the DB period, four patients (3.8%) in the satralizumab group and five (6.8%) in the placebo group discontinued treatment due to AEs. Serious infection rates were comparable between the satralizumab and placebo groups in the DB period (4.13 vs 6.99 events/100PY) and remained stable in the OST (3.88 events/100PY). No opportunistic infections were observed in the satralizumab group. The injection-related reaction (IRR) rate was higher with satralizumab vs placebo in the DB period (17.03 vs 8.99 events/100PY); IRRs were mostly mild-to-moderate and did not lead to treatment discontinuation. Laboratory abnormalities were in line with those expected with IL-6 receptor antagonists in the DB and OST period. No deaths or anaphylactic reactions were reported.

Conclusions

In NMOSD patients, satralizumab was well tolerated and showed a favorable safety profile. Results from the OST period were consistent with the DB period.

Collapse