ArticlePDF Available

Sustained Low Relapse Rate With Highly Variable B-Cell Repopulation Dynamics With Extended Rituximab Dosing Intervals in Multiple Sclerosis

Authors:

Abstract and Figures

Background and Objectives B cell–depleting therapies are highly effective in relapsing-remitting multiple sclerosis (RRMS) but are associated with increased infection risk and blunted humoral vaccination responses. Extension of dosing intervals may mitigate such negative effects, but its consequences on MS disease activity are yet to be ascertained. The objective of this study was to determine clinical and neuroradiologic disease activity, as well as B-cell repopulation dynamics, after implementation of extended rituximab dosing in RRMS. Methods We conducted a prospective observational study in a specialized-care, single-center setting, including patients with RRMS participating in the COMBAT-MS and MultipleMS observational drug trials, who had received at least 2 courses of rituximab (median follow-up 4.2 years, range 0.1–8.9 years). Using Cox regression, hazard ratios (HRs) of clinical relapse and/or contrast-enhancing lesions on MRI were calculated in relation to time since last dose of rituximab. Results A total of 3,904 dose intervals were accumulated in 718 patients and stratified into 4 intervals: <8, ≥8 to 12, ≥12 to 18, and ≥18 months. We identified 24 relapses of which 20 occurred within 8 months since previous infusion and 4 with intervals over 8 months. HRs for relapse when comparing ≥8 to 12, ≥12 to 18, and ≥18 months with <8 months since last dose were 0.28 (95% CI 0.04–2.10), 0.38 (95% CI 0.05–2.94), and 0.89 (95% CI 0.20–4.04), respectively, and thus nonsignificant. Neuroradiologic outcomes mirrored relapse rates. Dynamics of total B-cell reconstitution varied considerably, but median total B-cell counts reached lower level of normal after 12 months and median memory B-cell counts after 16 months. Discussion In this prospective cohort of rituximab-treated patients with RRMS exposed to extended dosing intervals, we could not detect a relation between clinical or neuroradiologic disease activity and time since last infusion. Total B- and memory B-cell repopulation kinetics varied considerably. These findings, relevant for assessing risk-mitigation strategies with anti-CD20 therapies in RRMS, suggest that relapse risk remains low with extended infusion intervals. Further studies are needed to investigate the relation between B-cell repopulation dynamics and adverse event risks associated with B-cell depletion.
Content may be subject to copyright.
RESEARCH ARTICLE OPEN ACCESS
Sustained Low Relapse Rate With Highly Variable
B-Cell Repopulation Dynamics With Extended
Rituximab Dosing Intervals in Multiple Sclerosis
Chiara Starvaggi Cucuzza, MD, Elisa Longinetti, PhD, Nicolas Ruffin, PhD, Bj¨
orn Evertsson, MD,
Ingrid Kockum, PhD, Maja Jagodic, PhD, Faiez Al Nimer, MD, PhD, Thomas Frisell, PhD,
and Fredrik Piehl, MD, PhD
Neurol Neuroimmunol Neuroinamm 2023;10:e200056. doi:10.1212/NXI.0000000000200056
Correspondence
Dr. Piehl
fredrik.piehl@ki.se
Abstract
Background and Objectives
B celldepleting therapies are highly eective in relapsing-remitting multiple sclerosis (RRMS)
but are associated with increased infection risk and blunted humoral vaccination responses.
Extension of dosing intervals may mitigate such negative eects, but its consequences on MS
disease activity are yet to be ascertained. The objectiveof this study was to determine clinical and
neuroradiologic disease activity, as well as B-cell repopulation dynamics, after implementation of
extended rituximab dosing in RRMS.
Methods
We conducted a prospective observational study in a specialized-care, single-center setting,
including patients with RRMS participating in the COMBAT-MS and MultipleMS observational
drug trials, who had received at least 2 courses of rituximab (median follow-up 4.2 years, range
0.18.9 years). Using Cox regression, hazard ratios (HRs) of clinical relapse and/or contrast-
enhancing lesions on MRI were calculated in relation to time since last dose of rituximab.
Results
A total of 3,904 dose intervals were accumulated in 718 patients and stratied into 4 intervals:
<8, 8 to 12, 12 to 18, and 18 months. We identied 24 relapses of which 20 occurred within
8 months since previous infusion and 4 with intervals over 8 months. HRs for relapse when
comparing 8 to 12, 12 to 18, and 18 months with <8 months since last dose were 0.28 (95%
CI 0.042.10), 0.38 (95% CI 0.052.94), and 0.89 (95% CI 0.204.04), respectively, and thus
nonsignicant. Neuroradiologic outcomes mirrored relapse rates. Dynamics of total B-cell
reconstitution varied considerably, but median total B-cell counts reached lower level of normal
after 12 months and median memory B-cell counts after 16 months.
Discussion
In this prospective cohort of rituximab-treated patients with RRMS exposed to extended dosing
intervals, we could not detect a relation between clinical or neuroradiologic disease activity and
time since last infusion. Total B- and memory B-cell repopulation kinetics varied considerably.
These ndings, relevant for assessing risk-mitigation strategies with anti-CD20 therapies in
RRMS, suggest that relapse risk remains low with extended infusion intervals. Further studies
are needed to investigate the relation between B-cell repopulation dynamics and adverse event
risks associated with B-cell depletion.
MORE ONLINE
COVID-19 Resources
For the latest articles,
invited commentaries, and
blogs from physicians
around the world
NPub.org/COVID19
From the Department of Clinical Neuros cience (C.S.C., E.L., N.R., B.E., I.K ., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K.,
M.J., F.A.N., F.P.), Karolinska University Hospital, Sto ckholm, Sweden; Department of Neurology (B.E., F.P.), Karolinsk a University Hospital, Stockholm, Sweden; Center for Neurolog y
(C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockhol m, Sweden; and Clinical Epidemiology Divis ion (T.F.), Department of Medicine Soln a, Karolinska Institutet,
Stockholm, Sweden.
Go to Neurology.org/NN for full disclosures. Funding information is provided at the end of the article.
The Article Processing Charge was funded by the Swedish Research Council.
This is an open access article distributed under the terms of the Creative Commons Attribution License 4.0 (CC BY), which permits unrestricted use, distribution, and reproduction in any
medium, provided the original work is properly cited.
Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. 1
Multiple sclerosis (MS) is a chronic, inammatory, de-
myelinating disease of the CNS, aecting 2.8 million people
worldwide.
1
Most patients initially present with acute/subacute
episodes of neurologic decit, with variable degree of re-
versibility, followed by a period of clinical stability, thus classied
as relapsing-remitting MS (RRMS). Accumulating evidence
demonstrates that B celldepleting therapies are associated with
strong suppression of RRMS inammatory disease activity.
2-4
So
far, 2 anti-CD20 monoclonal antibodies, ocrelizumab and ofa-
tumumab, have been approved for use in RRMS in the United
States and European Union. In addition, rituximab, an older
chimeric monoclonal antibody approved for rheumatoid arthritis
and other indications, is increasingly being used o-label in some
countries, including Sweden. The safety prole of anti-CD20
therapies in RRMS comprises an increased infection risk,
5
in-
cluding worsened COVID-19 outcomes.
6-8
Furthermore, anti-
CD20s blunt humoral responses to vaccinations,
9
including for
SARS-CoV-2.
10,11
Collectively, these data warrant studies ex-
ploring risk-mitigation strategies to ensure an optimized benet-
risk balance for patients with RRMS treated with anti-CD20s. So
far, however, such eorts comprise relatively small-sized studies
with limited dosing interval prolongation and short observation
time.
12,13
Larger real-world cohort studies with suciently long
observation time to determine time to normalization of B-cell
levels and possible relation to disease activity and risk of adverse
events are thus lacking. Considering an emerging safety signal
regarding infections already before the COVID-19 pandemic, a
pragmatic anti-CD20 dose extension program was initiated at the
Academic Specialist Center in the fall of 2018 and further ex-
tended with the pandemic outbreak in 2020. The aims of this
study were to determine whether prolonged rituximab dosing
intervals increase the risk of RRMS disease activity and to de-
termine B-cell repopulation dynamics in relation to infusions.
Methods
Study Population
The study population at the Academic Specialist Center
(Stockholm, Sweden) included 718 patients with RRMS enrolled
in either of 2 prospective observational drug trials, COMBAT-
MS (n = 658) and MultipleMS (n = 60), who had been exposed
to at least 2 doses of rituximab by September 1, 2021 (Figure 1A).
The COMBAT-MS study included patients with RRMS initiat-
ing a rst disease-modifying therapy (DMT) or doing a
rst DMT switch between January 1, 2011, and October 31,
2018. The MultipleMS study included newly diagnosed,
treatment-naive patients with RRMS who initiated a rst DMT
between April 1, 2018, and September 28, 2020. Data regarding
demographics, disease, and treatment history were extracted from
the Swedish MS registry. Data on total B-cell count and per-
centage of B cells subpopulations were extracted from medical
records from January 1, 2018, to September 1, 2021.
Standard Protocol Approvals, Registrations,
and Patient Consents
COMBAT-MS, EudraCT: 2016-003587-39, Clinicaltrials.gov
identier: NCT03193866; MultipleMS, EudraCT: 2017-
002634-24. Written informed consent was obtained from all
study participants (Stockholm Regional Ethics Board, no.
2017/32-31/4 and 2017/1323-31).
Follow-up, Outcomes, and Covariates
Patients were followed annually with assessment of Expanded
Disability Status Scale (EDSS), relapse history, MRI, and acute
contacts when needed. The MRI protocol has been published
previously and considers contrast administration for follow-up
scans as optional in caseof a stable patient,leaving the decision
to the treating neurologist.
14
COMBAT-MS SMSReg data
have been validated against medical records for completeness,
and both studies are continuously monitored for data com-
pleteness.
15
The outcomes in the main analysis were clinical
relapses and brain or spinal cord contrast-enhancing lesions
(CELs). Because annual MRI scans were not synchronized
with rituximab infusions, it was not possible to attribute the
occurrence of new or enlarged T2 lesions to a specic infusion
interval; hence, data are presented for each participant
according to the longest interval ever experienced.
The eect of dose interval extension was assessed by comparing
relapse rates during <8, 8to12,12 to 18, and 18 months
since last infusion. Treatment interval was analyzed as a time-
varying covariate. As a result, treatment intervals longer than 8
months were spilt in more than 1 category, where for example a
14 months interval since last rituximab infusion contributed
data to the <8, 8and12 monthstime bands. The interval
after the rst rituximab infusion (or after the rst 2 infusions if
<90 days apart) was excluded from the analysis to avoid the
eect of residual disease activity early after treatment start
(eFigure 1, http://links.lww.com/NXI/A770).
The following were considered potential confounders: sex,
age at infusion, EDSS at infusion, number of previous ritux-
imab doses, number of clinical relapses in the year before
rituximab start, number of brain MRI T2 lesions at rituximab
start (categorized as 0, 19, 1020, and >20), and previous
DMTs, classied as none, moderately eective (injectables,
dimethyl fumarate, teriunomide, or daclizumab), highly ef-
fective (ngolimod, natalizumab, or ocrelizumab), and others
(unspecied).
Glossary
CEL = contrast-enhancing lesion; DMT = disease-modifying therapy; EDSS = Expanded Disability Status Scale; HR = hazard
ratio; LLN = lower limit of normal; MS = multiple sclerosis; RR = rate ratio; RRMS = relapsing-remitting MS; SPMS =
secondary progressive MS.
2Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 Neurology.org/NN
B-Cell Data
TotalB-cell(CD3
+
CD19
+
) levels were assessed by
ow cytometry before each rituximab infusion, as per
clinical routine, at the Department of Clinical Immunol-
ogy, Karolinska University Hospital. B memory cell
(CD3
+
CD19
+
CD27+immunoglobulin D (IgD)-, CD27+-
IgD+, and CD27IgD) percentages were determined in
patients with detectable B cells and converted to absolute
numbers using the extracted data. B-cell data were assessed in
relation to time since last infusion (in months). In instances
where multiple measurements had been performed in the
same treatment interval, only the last assessment was in-
cluded. B-cell samples were classied as depleted if below the
detection level (10 cells/μL for total B and 0.05 cells/μL for
total and CD27+IgDmemory B), partially repleted if above
the detection limit, but below lower limit of normal (LLN, 80
cells/μL for total B, 15.2 cells/μL for total memory B, and 5.6
cells/μL for CD27+IgDmemory B), and completely reple-
ted if equal or above LLN.
Statistical Analysis
Cox proportional hazard regression models were used to
calculate hazard ratios (HRs) and corresponding 95% CIs of
clinical relapse or CELs in relation to rituximab dose intervals.
Time since disease onset was the underlying time scale in all
models; thus, HRs were compared across patients with the
same disease duration. Study entry occurred at each rituximab
infusion, with left truncation of follow-up time to avoid im-
mortal time bias (i.e., we used the counting process approach,
with a start and a stop dening each patients each interval).
Every time-to-censoring/outcome after a rituximab dose was
subsequently split in the aforementioned time bands (<8, 8
to 12, 12 to 18, and 18 months), and HRs were calculated
for 8 to 12, 12 to 18, and 18 months since last dose, using
<8 months as reference (for further details, see eMethods,
http://links.lww.com/NXI/A770). A sandwich estimator was
used to account for exposure of the same patient to multiple
rituximab dosing intervals. Models were separately analyzed
as crude and adjusted for confounders (listed above). Cen-
soring events were a subsequent rituximab infusion, conver-
sion to secondary progressive MS (SPMS), emigration, death,
or September 1, 2021, whichever came rst. In the main
analysis, we did not include date of switch to a dierent DMT
as a censoring event in keeping with the hypothesis of a long-
lasting eect of B-cell depletion and to maximize sensitivity
for relapses. However, DMT switch (33 patients out of 718;
4.6%) was considered an additional censoring event in a
sensitivity analysis to restrict the analysis time to exclusive
exposure to rituximab. Negative binomial regression models
were used to calculate repletion rate ratios (RRs) for total B-
and memory B-cell counts per microliter in relation to months
since last infusion. Both total B/memory B-cell counts per
microliter and months since last rituximab infusion were
regarded as numerical, continuous variables in the regression
model. Stratication of the regression model by sex, age, body
mass index (BMI), disease duration, and number of previous
rituximab doses was performed to uncover possible eect
modiers, with continuous variables transformed in binary
variables taking the approximate median value as splitting
point. With this approach, age was categorized as <40 and 40
years, BMI as <24 and 24 kg/m
2
, disease duration as <8 and
8 years since disease onset, and number of previous ritux-
imab doses as 13or4. Analyses were conducted with
STATA/BE software, 17.0.
Figure 1 Study Population
Inclusion criteria flowchart. ASC = Academic Specialist Cen-
ter; DMT = disease-modifying therapy, RRMS = relapsing-re-
mitting MS; RTX = rituximab; SMSReg = Swedish MS Registry.
Neurology.org/NN Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 3
Data Availability
Deidentied or aggregated data will be shared with qualied
investigators on reasonable request and pending a relevant data
transfer agreement, in compliance to European legislation.
Results
Study Participants
We identied 718 unique patients with RRMS enrolled in either
COMBAT-MSorMultipleMSstudies who had been exposed to
at least 2 courses of rituximab treatment, for a total of 4622
treatment episodes (Figure 1A and eFigure 2a, http://links.lww.
com/NXI/A770). Patients were followed from each rituximab
infusion until subsequent infusion, occurrence of clinical relapse
or CELs, emigration (n = 1), death (n = 3), transition to SPMS
(n = 19), or end of follow-up, whichever came rst. After ex-
clusion of the rst treatment course, these individuals had ac-
cumulated 3,904 rituximab dose intervals, for a median follow-up
of 4.2 years (interquartile range 2.75.6 years). The baseline
characteristics of study participants are summarized in Table 1.
The infusion-to-outcome/censoring intervals were distrib-
uted as follows: 2,577 intervals (66%) <8 months, 585 (15%)
812 months, 323 (8%) 1218 months, and 419 (11%)
18 months. Importantly, 95% of patients (n = 683) un-
derwent at least 1 interval extension (8 months), with 87%
(n = 628) and 56% (n = 403) exposed to an interval of 12
months and 18 months, respectively. Of the remaining 35
participants, the main reasons for remaining on a <8 months
treatment schedule were (1) censoring event before imple-
mentation of extension protocol, n = 16 (due to conversion to
SPMS, n = 15, or death, n = 1); (2) receiving a second course
of rituximab after January 1, 2021 (n = 10), consequently too
early to have been exposed to dose interval extension; and (3)
unspecied reasons (n = 9), likely involving personal prefer-
ences of the treating neurologist and/or the patient.
Clinical Relapse Occurrence and Relation With
Time Since Last Rituximab Infusion
During follow-up, a total of 24 relapses were recorded (eFigure 2b,
http://links.lww.com/NXI/A770) with the annualized relapse
rate dropping from a mean of 0.49 (95% CI 0.440.54) in the year
Table 1 Cohort Demographic and Clinical Characteristics
Overall (n = 718)
No. of females (%) 511 (71)
Age at onset, median (Q1Q3) 29 (2436)
Age at diagnosis, median (Q1Q3) 32 (2639)
Age at RTX start, median (Q1Q3) 37 (3045)
Disease duration at RTX start (y), median (Q1Q3) 5.5 (1.910.8)
ARR in the year before RTX start, mean (SD) 0.49 (0.67)
EDSS at RTX start, median (Q1Q3) 2(12.5)
No. of brain T2 lesions at RTX start, no. of patients (%)
0 lesions 7(1)
19 lesions 141 (20)
1020 lesions 196 (28)
>20 lesions 359 (51)
Class of previous DMT, no. of patients (%)
None 258 (36)
Moderately effective
a
273 (38)
Highly effective
b
151 (21)
Others
c
36 (5)
No. of RTX doses, median (Q1Q3) 6(48)
Duration of follow-up (y), median (Q1Q3) 4.2 (2.75.6)
Abbreviations: ARR = annualized relapse rate; DMT = disease-modifying therapy; EDSS = Expanded Disability Status Scale; Q1 = first quartile; Q3 = third
quartile; RTX = rituximab.
a
Injectables, teriflunomide, dimethyl fumarate, and daclizumab.
b
Fingolimod, natalizumab, and ocrelizumab
c
Unspecified or study drug.
4Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 Neurology.org/NN
before rituximab start to 0.03 (95% CI 0.020.05) in the rst
treatment year and 0.01 onward, up to 10 years of follow-up
(eFigure 2c, http://links.lww.com/NXI/A770).
We subsequently determined whether longer time since last
rituximab infusion was associated with increased risk of a
clinical relapse. HRs compared with <8 months since last
infusion were 0.28 (95% CI 0.042.10), 0.38 (95% CI
0.052.94), and 0.89 (95% CI 0.204.04) for 8 to 12, 12 to
18, and 18 months, respectively (Table 2). Adjustment for
potential confounders did not substantially divert from the
crude model. Adjusted HRs for 8 to 12, 12 to 18, and 18
months compared with <8 months since last infusion were
0.30 (95% CI 0.042.32), 0.42 (95% CI 0.053.23), and 0.85
(95% CI 0.183.92), respectively.
When considering date of switch to a dierent DMT as ad-
junctive censoring parameter, 3 relapses were censored,
leaving a total of 21 clinical relapses. In this setting, adjusted
HRs compared with <8 months resulted in 0.32 (95% CI
0.042.38) for 812 months and 0.68 (95% CI 0.076.6) for
>18 months since last rituximab dose, whereas HR for
1218 months could not be calculated due to the absence
of events (Table 2). Overall, these point estimates suggest a
similar or lower risk of relapse with transition from a regular to
an extended dosing interval regimen.
Inclusion of Contrast-Enhancing Lesions
as Outcome
Neuroradiologic assessments conducted with administration of
contrast agent (1,370 brain or spinal cord MRI scans out of 3,075
total scans, 44.6%, resulting in 548 patients with at least 1 follow-
up scan with contrast administration) were evaluated. Among
these, 11 scans revealed CEL events, 4 of which were performed
in the same treatment interval of a registered clinical relapse.
When considering both relapse and/or CELs, 31 disease activity
events were recorded, with HRs of 0.24 (95% CI 0.031.77),
0.66 (95% CI 0.152.93), and 0.72 (95% CI 0.163.35) for 8to
12, 12 to 18, and 18 months, respectively, compared with <8
months since last infusion in the crude model. Similar to the
previous analysis considering only clinical relapses, adjustment
for potential confounders barely altered the associations. HRs
were 0.26 (95% CI 0.031.96), 0.72 (95% CI 0.173.08), and
0.68 (95% CI 0.143.24) for 8 to 12, 12 to 18, and 18
months since last dose, respectively (Table 2).
Overall, the risk of an adverse ecacy outcome did not no-
ticeably change up to 3 years after rituximab infusion, with an
Table 2 Hazard Ratios for (1) Relapse, (2) Relapse and/or Contrast-Enhancing Lesions, and (3) Relapse With Disease-
Modifying Treatment Switch as Adjunctive Censoring Parameter, With Extended Time Since Rituximab Infusion
Compared With <8 Months
No. events Person-years Incidence rate
Cox model HR (95% CI)
a
Crude Adjusted
b
Clinical relapses
<8 months 20 2,119.6 0.009
812 months 1 326.4 0.003 0.28 (0.042.10) 0.30 (0.042.32)
1218 months 1 262.3 0.003 0.38 (0.052.94) 0.42 (0.053.23)
18 months 2 256.3 0.008 0.89 (0.204.04) 0.85 (0.183.92)
Clinical relapses and/or CELs
<8 months 26 2,117.4 0.012 . .
812 months 1 325.8 0.003 0.24 (0.031.77) 0.26 (0.031.96)
1218 months 2 261.3 0.008 0.66 (0.152.93) 0.72 (0.173.08)
18 months 2 249.7 0.008 0.72 (0.163.35) 0.68 (0.143.24)
Clinical relapses DMT switch included in censoring
<8 months 19 2,105.3 0.009 . .
812 months 1 319.9 0.003 0.30 (0.042.26) 0.32 (0.042.38)
1218 months 0 250.7 0 n/a n/a
18 months 1 207.6 0.005 0.65 (0.085.21) 0.68 (0.076.60)
Abbreviations: CEL = contrast-enhancing lesion; HR = hazard ratio.
a
Time since disease onset as underlying time scale of all models.
b
Adjusted for sex, age at infusion, Expanded Disability Status Scale at infusion, previous disease-modifying therapies, number of brain MRI T2 lesions at
rituximab start, annualized relapse rate in the year before rituximab start, and number of previous rituximab doses.
Neurology.org/NN Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 5
event-free survival rate of 0.98 (95% CI 0.970.99)
(Figure 2A). In the same manner, the incidence rate for re-
lapse and/or CELs remained stable across the 4 time bands
(Table 2 and Figure 2B).
New or Enlarging T2 Lesions on MRI Scans
Forty-eight MRI scans in 44 patients (6.9% of 636 patients
with valid follow-up and reference MRI scans) displayed new
or enlarging T2 lesions. As yearly scans were not synchronized
with rituximab infusions, the time period covered between
follow-up and reference MRI scans did not match a specic
treatment interval, so data are reported per individual instead
of per dosing interval, with stratication for the longest
treatment interval ever experienced when increase in T2 le-
sion burden was recorded. With this approach, 25 patients
were in the <8 months group (out of 90 patients in this group,
27.8%), whereas 7 (out of 144, 4.8%), 4 (out of 242, 1.7%),
and 8 (out of 160, 5%) were in the 8 to 12, 12 to 18, and
18 months groups, respectively.
Total B-Cell Repopulation Kinetics
B-cell data were available only after January 1, 2018, resulting
in 1,744 data points for a total of 2,208 infusions after this
date in 648 patients (79% of the infusions; 90% of the patient
cohort). The median of total B-cell counts reached detect-
able levels 6 months since last infusion and the LLN after 12
months (Figure 3A). However, a considerable degree of
variability of total B-cell levels was observed, with variance
increasing over time since last infusion and a small pro-
portion of subjects (3.4%) remaining completely depleted
even with the longest dosing intervals. When considering the
same time bands used in the main analysis, we found that
63.9% of samples remained depleted in the <8 months
group. In contrast, the majority of samples were partially or
completely repleted in the extended-interval groups. The 2
categories combined accounted for 87.1%, 97.9%, and 96.6%
of the samples in the 8 to 12, 12 to 18, and 18 months
groups, respectively (Figure 3B). Negative binomial re-
gression conrmed positive association between B-cell levels
and months since last infusion, with a repletion RR of 1.17
(95% CI 1.141.19, p< 0.0001). In other words, the model
predicted an increase in B-cell counts of 17% every month in
the analyzed data set. When stratifying according to age, sex,
BMI, disease duration, or number of previous rituximab
doses, we did not observe any signicant eect modication
by sex, age, and BMI, whereas longer disease duration and
higher number of previous rituximab doses were associated
with a slower rate of B cell repopulation, as expressed by the
RR (interaction coecients 0.94, 95% CI 0.90.98 and 0.9,
95% CI 0.860.95 for disease duration and number of pre-
vious doses, respectively). The stratied RRs were 1.21 (95%
CI 1.181.25) and 1.14 (95% CI 1.121.16) for disease
duration <8 or 8 years, respectively, and 1.24 (95% CI
1.191.30) and 1.12 (95% CI 1.101.15) for B-cell repo-
pulating after infusions preceded by 13or4 rituximab
doses, respectively (Figure 3, C and D).
Memory B-Cell Repopulation Kinetics
Memory B-cell data were available only for samples with de-
tectable B cells. In this subgroup of 1,157 samples, 580 sam-
ples (46%) also included B-cell subpopulation analysis, for a
total of 489 patients. Compared with total B-cell levels,
memory B cells showed a slower repopulation kinetic: the
median of memory B-cell counts, categorized in 1-month time
bands, was steadily above the limit of detection but reached
LLN only after 16 months (Figure 4A). Thus, memory B cells
were partially repleted in the majority of samples in the rst 2
time bands (79.7% and 59.5% for the <8 and 812 months,
Figure 2 Risk of Clinical Relapse and/or Contrast-Enhancing Lesion Occurrence in Relation to Time Since Last Rituximab
Infusion
(A) Kaplan-Meier curve of event-free time
since last rituximab infusion. (B)Incidence rate
of clinical relapse and/or CELs at <8 months,
8 to 12, 12 to 18, and 18 months since last
rituximab infusion. CEL = contrast-enhancing
lesion; RTX = rituximab.
6Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 Neurology.org/NN
respectively), whereas most samples (60.8%) were normal-
ized after 18 months since last infusion (Figure 4B). Ac-
cordingly, when applying a negative binomial regression
model, the repletion rate ratio was 1.03 (95% CI 1.021.04,
Figure 4C). No eect modication was observed stratifying
samples according to age, sex, BMI, disease duration, or
number of previous rituximab doses (Figure 4D). When
considering separately the 3 subsets composing memory
B cells (CD27+IgD, CD27+IgD+ or double positive, DP,
and CD27IgDor double negative, DN), we noted dif-
ferent repopulation kinetics, with LLN reached in 12 and 7
months for DP and DN subsets, respectively (Figure 5, A
and B), whereas median levels of CD27+IgDmemory
subpopulation persistently remained below LLN for up to
24 months (Figure 5C). As a result, this subset was only
partially repleted in the majority of samples up to 24 months
since last rituximab infusion: 87.5%, 84%, 77%, and 66.9%
partially repleted samples for <8, 8 to 12, 12 to 18, and
18 months, respectively (Figure 5D).
Discussion
We here related inammatory disease activity to time since last
rituximab infusion in a large cohort of patients with RRMS
exposed to dose interval extension with long prospective follow-
up. Alike the ocrelizumab label, the Swedish MS Society rec-
ommends rituximab dosing (500 mg) every 6 months. Because
of an emerging signal for increased infection rate over time,
5
from October 2018, the Academic Specialist Center changed
guidelines to recommend dose extension to 12 months after the
fth infusion for patients with stable disease. With the start of
the COVID-19 pandemic in March 2020, infusion intervals
were further extended to 1224 months or more from the
second infusion regardless of treatment duration, a decision also
inuenced by accumulating evidence suggesting an adverse ef-
fect on COVID-19 outcomes.
6,7
We previously reported the absence of rebound disease ac-
tivity in patients with RRMS who, for various reasons, had
Figure 3 Total B-Cell Repopulation Dynamics in Relation to Time Since Last Rituximab Infusion
(A) Box plot depicting distributions of B-cell count grouping samples into 1-month-time intervals since last rituximab infusion. Continuous red line: B-cell
detection limit; dashed black line: LLN. (B) Total number of observations (y axis) with relative frequencies (bar labels) of depleted (<10 cells/μL), partially
repleted (1080 cells/μL), and completely repleted (80 cells/μL) B-cell counts at <8 months, 8 to 12, 12 to 18, and 18 months since last rituximab infusion.
(C) Scatter plot of total B-cell counts in relation to time since last infusion, with samples collected within 1 month from a relapse/contrast-enhancing lesion
highlighted in red. The predicted repopulation kinetic up to 18 months since last rituximab infusion according to a negative binomial regression model is
depicted by the black curve. Continuous red line: B-cell detection limit; dashed black line: lower limit of normal. (D) Coefficient plot of the repletion rate ratios
(RRs) for B-cell repopulation for the overall model and stratified according to sex, age (<40 and 40 years of age), BMI (<24 and 24 kg/m
2
), disease duration (<8
and 8 years), and number of previous rituximab doses (13 and 4). Among the analyzed potential effect modifiers, disease duration and number of previous
rituximab doses affected B-cell reappearance kinetics (Wald test p< 0.01 for disease duration, **, and p< 0.000 1 for rituximab doses, ****). #BMI data were
available for 86.7% of the patients, 86.5% of the samples.
Neurology.org/NN Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 7
stopped rituximab for more than 12 months.
16
Similar results
were reported for patients with RRMS exposed to low dose
(<1000 mg yearly) or stopping treatment in another Swedish
study.
17
Furthermore, based on data from the phase II trial of
ocrelizumab, which included a safety follow-up of about 12
months without treatment, it has been suggested that treat-
ment intervals could be extended well beyond the regular 6-
month interval.
18
More recently, a retrospective observational
study did not detect signs of disease activity rebound with
extension of ocrelizumab intervals by 4 weeks or more.
12
Fi-
nally, a Dutch prospective study implemented a personalized
dosing interval of ocrelizumab based on B cell counts.
13
In this
study B-cell levels were assessed monthly after 6 months from
the last infusion with retreatment with counts 10 cells/μL,
which occurred at a median of 34 weeks since last infusion.
However, all these studies were relatively small and had
considerably shorter follow-up periods than included here,
with a median structured prospective follow-up of 4.2 years
(interquartile range 2.75.6 years).
Of interest, we could not detect an increased risk of
clinical relapses and/or CELs for any of time bands ana-
lyzed, i.e., 8 to 12, 12 to 18, and 18 months, with point
estimates of relapse risk remaining remarkably stable.
Although the power to detect a dierence was limited
due to the limited number of relapse/CEL events, our
observations suggest that both relapse risk and neuro-
radiologic disease activity remain low well beyond regular
infusion intervals. The fact that HRs with extended dosing
intervals in all cases were below 1 compared with the
regular interval is reassuring, but likely can be inuenced by
overall treatment duration and a small number of subjects
switching treatment or being held on shorter infusion intervals.
It is important to note, however, that a substantial enrichment
of stable patients in the longer time bands is unlikely given the
prospective study design and the high proportion of partici-
pants being exposed to extended intervals. Taken together,
these results are valuable for designing further, suciently
powered conrmatory studies, but meanwhile provide
Figure 4 Memory B-Cell Repopulation Dynamics in Relation to Time Since Last Rituximab Infusion
(A) Box plot showing distributions of memory B-cell count grouping samples into 1-month time intervals since last rituximab infusion. Continuous red line:
memory B-cell detection limit; dashed black line: lower limit of normal. (B) Total number of observations (y axis) with relative frequencies (bar labels) of
depleted (<0.05 cells/μL), partially repleted (0.0515.2 cells/μL), and completely repleted (15.2 cells/μL) levels of memory B cells at <8 months, 8 to 12, 12
to 18, and 18 months since last rituximab infusion. (C) Scatter plot of memory B-cell counts in relation to time since last infusion, with samples collected
within 1 month from a relapse/contrast-enhancing lesion highlighted in red. The repopulation kinetic predicted by a negative binomial regression model up to
24 months since last rituximab infusion is shown by the black curve. Continuous red line: memory B-cell detection limit; dashed black line: lower limitof
normal. (D) Coefficient plot of the repletion rate ratios (RRs) for memory B-cell repopulation for the overall model and stratified according to sex, age, BMI,
disease duration, and number of previous rituximab doses. None of the models showed any effect of the analyzed covariates on memory B-cell repletion
rates.
#
BMI data were available for 88.1% of the patients, 86.9% of the samples. BMI = body mass index
8Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 Neurology.org/NN
preliminary results indicating a viable approach for improving
the benet-risk balance with B celldepleting DMTs. This is
especially relevant in context of increased susceptibility to in-
fections, lowered immunoglobulin levels, scheduling of vacci-
nations, or planning of pregnancy.
5,10,19,20
The striking ecacy of anti-CD20 therapies underscores the
role of B cells in MS disease pathogenesis. Experiences across
a wide spectrum of autoimmune conditions indicate numer-
ous ways by which B cells can contribute to disease, which also
depends on the type of disease process.
21
In MS, B cells have
been shown to produce cytokines with a presumed role in
supporting CNS inammation, with supporting functional
data obtained in experimental autoimmune encephalitis, an
animal model of MS.
22,23
Notably, however, antigen-specic
memory B-cell clones have also been shown to activate
memory T cells with encephalitogenic features in subjects
with RRMS ex vivo.
24
The possible implication of this ob-
servation is that disease-driving cells mainly belong to the
memory B-cell subtype, alike what has been suggested for
neuromyelitis optica, where monitoring of memory B-cell
subset has been suggested to be useful to determine anti-
CD20 infusion intervals.
25,26
However, it is important to
acknowledge that clinical eectiveness of extended dosing
intervals cannot readily be extrapolated due to dierent
pathomechanisms, whereas it is more likely that B-cell repo-
pulation kinetics will be more similar across the 2 conditions.
However, in spite of increasing use of anti-CD20 therapies in
MS, there is only limited information on the kinetics of B-cell
repopulation after depletion and if there is any relation with
return of disease activity. Insights regarding dierent memory
B-cell subsets are also lacking. To this end, we analyzed total
B-cell and memory B-cell counts determined in clinical rou-
tine for our cohort. Although limited by a certain irregularity
of assessments in this real-world setting, our data, neverthe-
less, demonstrate a considerable variability in reconstitution
kinetics of total B-cell numbers, where median counts were
normalized after 12 months, whereas memory B cells
remained below LLN for up 16 months since last dose. In the
prediction model we used, based on negative binomial re-
gression, total B-cell counts increased by 17% every month,
with disease duration and number of previous rituximab
doses negatively aecting repopulation rates, although re-
sults should be interpreted with some caution due to the
variability in timing of sampling. Regarding memory B cells,
the repletion rate was much slower, with a monthly increase
Figure 5 Memory B Subpopulation Repopulation Dynamics
(AC) Box plot showing distributions of the 3 subsets of memory B cell: CD27+IgD(A), CD27+IgD+ or double positive (DP, B), and CD27IgDor double
negative (DN, C). Samples were grouped into 1-month time intervals since last rituximab infusion. Continuous red line: memory B-cell detection limit (0.05
cells/μL for all subsets); dashed black line: lower limit of normal (5.6 cells/μL for CD27+IgDcells, 3.44 cells/μL for DP cells and 1.92 cells/μL for DN cells). (D)
Total number of observations (y axis) with relative frequencies (bar labels) of depleted (<0.05 cells/μL), partially repleted (0.055.6 cells/μL), and completely
repleted (5.6 cells/μL) levels of CD27+IgDmemory B cells at <8 months, 8 to 12, 12 to 18, and 18 months since last rituximab infusion.
Neurology.org/NN Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 9
of only 3%. When considering dierent memory B-cell
subsets, CD27IgDwere the rst to repopulate, followed
by CD27+IgD+, whereas median levels of CD27+IgD
remained consistently below LLN up to 24 months since last
infusion. Overall, these results might hint at dierent roles of
memory B-cell subsets in RRMS pathogenesis. The low
number of adverse ecacy events coupled with lack of sys-
tematic determination of B cells at occurrence of clinical or
radiologic activity meant that a comparison between non-
active and active patients could not be performed. However,
it is evident that a strong signal for return of inammatory
disease activity with repletion of B cells, including of the
memory subtype, is lacking.
Apart from previously mentioned limitations relating to the
real-world nature of this study, including incomplete data
coverage and variability in the structure of data collection, this
study also did not include volumetric MRI data or soluble
biomarkers such as neurolament light chain concentrations.
Thus, we cannot exclude that dose interval extension nega-
tively aects disease processes not reected by relapses or
accrual of focal MRI lesions, and studies exploring the eect of
extended B celldepleting treatment schedules on the pro-
gressive aspects of MS are therefore warranted.
In summary, our ndings suggest that anti-CD20 dose in-
terval extension could be considered in patients with RRMS
with stable disease without incurring risk of return of in-
ammatory disease activity in the short to medium term,
especially in case of treatment-related adverse events or
when planning pregnancy. Further studies are needed to
determine whether dose interval extension is also associated
with a lowered risk of infection, while it has been shown that
vaccination responses are improved with B-cell repopula-
tion,
27
in turn improving benet-risk with anti-CD20
therapies.
Acknowledgment
The authors thank all the patients participating in the
COMBAT-MS and the MultipleMS studies and Simon
Englund, BSc, and Tommaso Piehl for their help with data
collection.
Study Funding
This work was funded by the Patient-Centered Outcomes
Research Institute (PCORI) Award (Combat-MS, MS-
1511-33196), Swedish MRC grant no. 2020-02700, the
European Unions Horizon 2020 Research and Innovation
Programme (MultipleMS, EU RIA 733161), the Knut and
Alice Wallenberg Foundation, the Swedish Research
Council for Health, Working Life, and Welfare (postdoc
Grant No: 2020-0115 to E.L.), and the Swedish Brain
Foundation.
Disclosure
F. Piehl has received research grants from Merck KGaA and
UCB and fees for serving on DMC in clinical trials with
Chugai, Lundbeck, and Roche. C. Starvaggi Cucuzza has
received a travel grant from SanoGenzyme. B. Evertsson
has received travel support from Roche. The remaining
authors declare no competing interests. Go to Neurology.
org/NN for full disclosures.
Publication History
Received by Neurology: Neuroimmunology & Neuroinammation
May 10, 2022. Accepted in nal form September 16, 2022. Submitted
and externally peer reviewed. The handling editor was Friedemann Paul,
MD.
Appendix Authors
Name Location Contribution
Chiara
Starvaggi
Cucuzza, MD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden;
Center for Molecular Medicine,
Karolinska University Hospital,
Stockholm, Sweden
Drafting/revision of the
manuscript for content,
including medical writing
for content; major role in
the acquisition of data;
study concept or design;
and analysis or
interpretation of data
Elisa
Longinetti,
PhD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden
Drafting/revision of the
manuscript for content,
including medical writing
for content; study concept
or design; and analysis or
interpretation of data
Nicolas
Ruffin, PhD
Center for Molecular Medicine,
Karolinska University Hospital,
Stockholm, Sweden; Center for
Molecular Medicine, Karolinska
University Hospital, Stockholm,
Sweden
Drafting/revision of the
manuscript for content,
including medical writing
for content; study concept
or design; and analysis or
interpretation of data
Bj¨
orn
Evertsson,
MD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden;
Department of Neurology,
Karolinska University Hospital,
Stockholm, Sweden
Drafting/revision of the
manuscript for content,
including medical writing
for content, and major
role in the acquisition of
data
Ingrid
Kockum,
PhD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden;
Center for Molecular Medicine,
Karolinska University Hospital,
Stockholm, Sweden; Center for
Neurology, Academic Specialist
Center, Stockholm, Sweden;
Drafting/revision of the
manuscript for content,
including medical writing
for content, and major
role in the acquisition of
data
Maja
Jagodic, PhD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden;
Center for Molecular Medicine,
Karolinska University Hospital,
Stockholm, Sweden; Center for
Neurology, Academic Specialist
Center, Stockholm, Sweden;
Drafting/revision of the
manuscript for content,
including medical writing
for content, and major
role in the acquisition of
data
Faiez Al
Nimer, MD,
PhD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden;
Center for Molecular Medicine,
Karolinska University Hospital,
Stockholm, Sweden; Center for
Neurology, Academic Specialist
Center, Stockholm, Sweden;
Drafting/revision of the
manuscript for content,
including medical writing
for content; major role in
the acquisition of data;
and study concept or
design
10 Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 Neurology.org/NN
References
1. Walton C, King R, Rechtman L, et al. Rising prevalence of multiple sclerosis world-
wide: insights from the Atlas of MS. Mult Scler. 2020;1226(14):1816-1821. doi:
10.1177/1352458520970841.
2. Hauser SL, Waubant E, Arnold DL, et al. B-cell depletion with rituximab in relapsing -
remitting multiple sclerosis. N Engl J Med. 2008;358(7):676-688. doi: 10.1056/
NEJMoa0706383
3. Hauser SL, Bar-Or A, Comi G, et al. Ocrelizumab versus interferon beta-1a in re-
lapsing multiple sclerosis. N Engl J Med. 2017;376(3):221-234. doi: 10.1056/
NEJMoa1601277
4. Hauser SL, Bar-Or A, Cohen JA, et al. Ofatumumab versus teriunomide in multiple
sclerosis. N Engl J Med. 2020;383(6):546-557. doi: 10.1056/NEJMoa1917246
5. Luna G, Alping P, Burman J, et al. Infection risks among patients with multiple
sclerosis treated with ngolimod, natalizumab, rituximab, and injectable therapies.
JAMA Neurol. 2020;77(2):184-191. doi: 10.1001/jamaneurol.2019.3365
6. Simpson-Yap S, De Brouwer E, Kalincik T, et al. Associations of disease-modifying
therapies with COVID-19 severity in multiple sclerosis. Neurology. 2021;97(19):
e1870e1885. doi: 10.1212/WNL.0000000000012753
7. Salter A, Fox RJ, Newsome SD, et al. Outcomes and risk factors associated with SARS-
CoV-2 infection in a north American registry of patients with multiple sclerosis. JAMA
Neurol. 2021;78(6):699-708. doi: 10.1001/jamaneurol.2021.0688
8. Schiavetti I, Ponzano M, Signori A, Bovis F, Carmisciano L, Sormani MP. Severe
outcomes of COVID-19 among patients with multiple sclerosis under anti-CD-20
therapies: a systematic review and meta-analysis. Mult Scler Relat Disord. 2022;57:
103358. doi: 10.1016/j.msard.2021.103358
9. Bar-Or A, Calkwood JC, Chognot C, et al. Eect of ocrelizumab on vaccine responses
in patients with multiple sclerosis: the VELOCE study. Neurology. 2020;95(14):
e1999e2008. doi: 10.1212/WNL.0000000000010380
10. Asplund ogelin K, Run N, Pin E, et al. Development of humoral and cellular
immunological memory against SARS-CoV-2 despite B cell depleting treatment in
multiple sclerosis. iScience. 2021;24(9):103078. doi: 10.1016/j.isci.2021.103078
11. Apostolidis SA, Kakara M, Painter MM, et al. Cellular and humoral immune responses
following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-
CD20 therapy. Nat Med. 2021;27(11):1990-2001. doi: 10.1038/s41591-021-01507-2
12. Rolfes L, Pawlitzki M, Pfeuer S, et al. Ocrelizumab extended interval dosing in
multiple sclerosis in times of COVID-19. Neurol Neuroimmunol Neuroinamm. 2021;
8(5):e1035. doi: 10.1212/NXI.0000000000001035
13. van Lierop ZY, Toorop AA, van Ballegoij WJ, et al. Personalized B-cell tailored dosing
of ocrelizumab in patients with multiple sclerosis during the COVID-19 pandemic.
Mult Scler. 2021;27(11):1990-2001:. doi: 10.1177/13524585211028833
14. VågbergM, Axelsson M, BirganderR, et al. Guidelines for theuse of magnetic resonance
imaging in diagnosing and monitoring the treatment of multiple sclerosis: recommen-
dationsof the Swedish Multiple Sclerosis Association andthe Swedish Neuroradiological
Society. Acta Neurol Scand. 2017;135(1):17-24. do i: 10.1111/ane.12667
15. Alping P, Piehl F, Langer-Gould A, Frisell T; COMBAT-MS Study Group. Validation
of the Swedish multiple sclerosis register: further improving a resource for pharma-
coepidemiologic evaluations. Epidemiology. 2019;30(2):230-233. doi: 10.1097/
EDE.0000000000000948
16. Juto A, Fink K, Al Nimer F, Piehl F. Interrupting rituximab treatment in relapsing-
remitting multiple sclerosis; no evidence of rebound disease activity. Mult Scler Relat
Disord. 2020;37:101468. doi: 10.1016/j.msard.2019.101468
17. Boremalm M, Sundstr¨om P, Salzer J. Discontinuation and dose reduction of rituximab
in relapsing-remitting multiple sclerosis. J Neurol. 2021;268(6):2161-2168. doi:
10.1007/s00415-021-10399-8
18. Baker D, Pryce G, James LK, Marta M, Schmierer K. The ocrelizumab phase II
extension trial suggests the potential to improve the risk: benet balance in multiple
sclerosis. Mult Scler Relat Disord. 2020;44:102279. doi: 10.1016/j.msard.2020.10 2279
19. Vollmer BL, Wallach AI, Corboy JR, Dubovskaya K, Alvarez E, Kister I. Serious safety
events in rituximab-treated multiple sclerosis and related disorders. Ann Clin Transl
Neurol. 2020;7(9):1477-1487. doi: 10.1002/acn3.51136
20. Razaz N, Piehl F, Frisell T, Langer-Gould AM, McKay KA, Fink K. Disease activity in
pregnancy and postpartum in women with MS who suspended rituximab and nata-
lizumab. Neurol Neuroimmunol Neuroinamm. 2020;11(6):7. doi: 10.1212/
NXI.0000000000000903
21. Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune
disease: advances and mechanistic insights. Nat Rev Drug Discov. 2021;20(3):179-199.
doi: 10.1038/s41573-020-00092-2
22. Barr TA, Shen P, Brown S, et al. B cell depletion therapy ameliorates autoimmune
disease through ablation of IL-6-producing B cells. J Exp Med. 2012;209(5):
1001-1010. doi: 10.1084/jem.20111675
23. Li R, Rezk A, Miyazaki Y, et al. Proinammatory GM-CSF-producing B cells in
multiple sclerosis and B cell depletion therapy. Sci Transl Med. 2015;7(310):310ra166.
doi: 10.1126/scitranslmed.aab4176
24. Jelcic I, Al Nimer F, Wang J, et al. Memory B cells activate brain-homing, autoreactive
CD4. Cell. 2018;175(1):85-100.e23. doi: 10.1016/j.cell.2018.08.011
25. Trewin BP, Adelstein S, Spies JM, et al. Precision therapy for neuromyelitis optica
spectrum disorder: a retrospective analysis of the use of class-switched memory B-cells
for individualised rituximab dosing schedules. Mult Scler Relat Disord. 2020;43:
102175. doi: 10.1016/j.msard.2020.102175
26. Novi G, Bovis F, Fabbri S, et al. Tailoring B cell depletion therapy in MS according to
memory B cell monitoring. Neurol Neuroimmunol Neuroinamm. 2020;7(5):e845.
doi: 10.1212/NXI.0000000000000845
27. Asplund ogelin K, Run N, Pin E, et al. B cell repopulation dynamics and drug
pharmacokinetics impact SARS-CoV-2 vaccine ecacy in anti-CD20-treated multiple
sclerosis patients. Eur J Neurol. 2022;29(11):3317-3328. doi: 10.1111/ene.15492
Appendix (continued)
Name Location Contribution
Thomas
Frisell, PhD
Clinical Epidemiology Division,
Department of Medicine Solna,
Karolinska Institutet,
Stockholm, Sweden
Drafting/revision of the
manuscript for content,
including medical writing
for content; study concept
or design; and analysis or
interpretation of data
Fredrik
Piehl, MD,
PhD
Department of Clinical
Neuroscience, Karolinska
Institutet, Stockholm, Sweden;
Center for Molecular Medicine,
Karolinska University Hospital,
Stockholm, Sweden;
Department of Neurology,
Karolinska University Hospital,
Stockholm, Sweden; Center for
Neurology, Academic Specialist
Center, Stockholm, Sweden;
Drafting/revision of the
manuscript for content,
including medical writing
for content; major role in
the acquisition of data;
study concept or design;
and analysis or
interpretation of data
Neurology.org/NN Neurology: Neuroimmunology & Neuroinflammation | Volume 10, Number 1 | January 2023 11
DOI 10.1212/NXI.0000000000200056
2023;10; Neurol Neuroimmunol Neuroinflamm
Chiara Starvaggi Cucuzza, Elisa Longinetti, Nicolas Ruffin, et al.
With Extended Rituximab Dosing Intervals in Multiple Sclerosis
Sustained Low Relapse Rate With Highly Variable B-Cell Repopulation Dynamics
This information is current as of November 21, 2022
Services
Updated Information & http://nn.neurology.org/content/10/1/e200056.full.html
including high resolution figures, can be found at:
References http://nn.neurology.org/content/10/1/e200056.full.html##ref-list-1
This article cites 27 articles, 5 of which you can access for free at:
Permissions & Licensing
http://nn.neurology.org/misc/about.xhtml#permissions
its entirety can be found online at:
Information about reproducing this article in parts (figures,tables) or in
Reprints http://nn.neurology.org/misc/addir.xhtml#reprintsus
Information about ordering reprints can be found online:
Academy of Neurology.. All rights reserved. Online ISSN: 2332-7812.
Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American
Published since April 2014, it is an open-access, online-only, continuous publication journal. Copyright
is an official journal of the American Academy of Neurology.Neurol Neuroimmunol Neuroinflamm
... The results suggest that EID can provide efficient therapies while lowering drug exposure. [5][6][7][8] Several advantages could potentially be derived from lower drug exposure, for example, decreased risk of low IgG levels and infection rates, improved vaccine response, as well as improved patient experience, and lower healthcare expenses. 9,10 In addition, novel studies have suggested therapeutic drug monitoring by measuring the serum concentration of ocrelizumab as a predictor for B-cell repopulation, vaccination response, and disability progression. ...
... B-cell depletion was found in most SID participants similar to previous publications. [5][6][7][8] EID resulted in B-cell repopulation in some participants, while the levels of NfL and GFAP were comparable in both groups and not correlated with levels of B-cells. NfL levels were higher in participants with disease re-activation, possibly reflecting neuroaxonal damage and disease activity. ...
Article
Background This study investigates clinical and biomarker differences between standard interval dosing (SID) and extended interval dosing (EID) of ocrelizumab therapy in multiple sclerosis (MS). Methods This is a prospective, double-arm, open-label, multi-center study in Denmark. Participants diagnosed with MS on ocrelizumab therapy >12 months were included ( n = 184). Clinical, radiological, and blood-based biomarker outcomes were evaluated. MRI disease activity, relapses, worsening of neurostatus, and No Evidence of Disease Activity-3 (NEDA-3) were used as a combined endpoint. Results Out of 184 participants, 107 participants received EID (58.2%), whereas 77 participants received SID (41.8%). The average extension was 9 weeks with a maximum of 78 weeks. When comparing EID to SID, we found higher levels of B-cells, lower serum concentrations of ocrelizumab, and similar levels of age-adjusted NFL and GFAP in the two groups. No difference in NEDA-3 between EID and SID was demonstrated (hazard ratio: 1.174, p = 0.69). Higher levels of NFL were identified in participants with disease activity. Body mass index correlated with levels of ocrelizumab and B-cells. Conclusion Extending one treatment interval of ocrelizumab on average 9 weeks and up to 78 weeks did not result in clinical, radiological, or biomarker evidence of worsening compared with SID.
... In a prospective cohort of RTX-treated RR-MS patients, extended dosing intervals did not lead to the recurrence of clinical or radiological disease activity. 47 In another study, however, an OCR infusion delay of at least 4 weeks was associated with an increased risk of MRI activity. 48 Nonetheless, although B-cell repletion kinetics varied widely, memory B cells seemed to repopulate slower than total B-cell counts, reaching the lower level of normal (LLN) only after 16 months, while the number of total B cells reached the LLN already after 12 months. ...
... 48 Nonetheless, although B-cell repletion kinetics varied widely, memory B cells seemed to repopulate slower than total B-cell counts, reaching the lower level of normal (LLN) only after 16 months, while the number of total B cells reached the LLN already after 12 months. 47 The optimal overall treatment duration of B cell-depleting therapies has not been established. In general, RTX and OCR infusions are tolerated reasonably well, and immediate potential side effects are rare or, if any, usually mild and easily treatable. ...
Article
Full-text available
Objective Repeated intravenous administration of anti‐CD20 depleting monoclonal antibodies 6 months apart is among the highly effective treatment options in multiple sclerosis (MS). Here, we aimed to investigate peripheral immune cell subset depletion kinetics following either rituximab (RTX) or ocrelizumab (OCR) infusions in people with MS (pwMS). Methods We studied pwMS treated de‐novo with either RTX ( n = 7) or OCR ( n = 8). The examinations were scheduled before the initiation of anti‐CD20 therapy and every 12 weeks for up to 15 months. Immunophenotyping of immune cell subsets in peripheral blood was performed by multiparametric fluorescence cytometry. Results A significant, persistent decrease of CD19 ⁺ B cells was observed already with the first anti‐CD20 infusion ( p < 0.0001). A significant proportional reduction of memory B cells within the B‐cell pool was achieved only after two treatment cycles ( p = 0.005). We observed a proportional increase of immature ( p = 0.04) and naive B cells ( p = 0.004), again only after the second treatment cycle. As for the peripheral T‐cell pool, we observed a continuous proportional increase of memory T helper (TH) cells/central memory TH cells ( p = 0.02/ p = 0.008), while the number of regulatory T cells (Treg) decreased ( p = 0.007). The percentage of B‐cell dependent TH17.1 central memory cells dropped after the second treatment cycle ( p = 0.02). No significant differences in the depletion kinetics between RTX and OCR were found. Interpretation Peripheral immune cell profiling revealed more differentiated insights into the prompt and delayed immunological effects of repeated intravenous anti‐CD20 treatment. The observation of proportional changes of some pathogenetically relevant immune cell subsets only after two infusion cycles deserves further attention.
... [3][4][5][6] As compared with cell-trafficking inhibitors, RTX and OCR are not associated with disease reactivation after therapy withdrawal. Conversely, recent studies of adults found sustained control of disease activity after a large extension of the RTX dosing interval, which suggests that renewing the immune system underlies the prolonged effect of RTX/OCR in MS. 7,8 Crucially, early deescalation after RTX/OCR could reduce the risk of infection and hypogammaglobulinemia associated with protracted-maintenance RTX/OCR therapy. 9,10 This schedule could be particularly important for children, with their increased risk of RTX-associated hypogammaglobulinemia as compared with adults. ...
... 9,10 Recent studies of adults suggested that large extended-interval dosing of RTX that leads to a significant reconstitution of circulating B cells is safe and could reduce the risk of infection and improve vaccine efficacy. 7,8,17 To the best of our knowledge, there are no data on the potential efficacy of large extended-interval dosing of RTX/ OCR in patients with PoMS. ...
Article
Recent studies in adults suggested that extended-interval dosing of rituximab/ocrelizumab (RTX/OCR) larger than 12 months was safe and could improve safety. This was an observational cohort study of very active pediatric-onset multiple sclerosis (PoMS) (median (range) age, 16 (12–17) years) treated with RTX/OCR with 6 month standard-interval dosing ( n = 9) or early extended-interval dosing ( n = 12, median (range) interval 18 months (12–25)). Within a median (range) follow-up of 31 (12–63) months after RTX/OCR onset, one patient (standard-interval) experienced relapse and no patient showed disability worsening or new T2-weighted lesions. This study suggests that the effectiveness of RTX/OCR is maintained with a median extended-interval dosing of 18 months in patients with very active PoMS.
... 22,29 While monitoring for declining IgG levels before each infusion and extending dosing intervals and/or reducing dose should continue to be part of standard B-celldepleting therapy practice, clinicians and patients should be aware that the risk of infection increases with increasing use even when IgG levels are normal (i.e., >700 mg/dL). Given mounting evidence that continuous B-cell depletion is not required to maintain efficacy in relapsing MS 36 and demonstration herein that there is a cumulative, dose-dependent increased risk of serious and recurrent outpatient infections, studies to identify the lowest effective dosing regimen of Bcell-depleting therapies are urgently needed. ...
Article
Full-text available
Background and objectives: B-cell-depleting therapies increase the risk of infections and hypogammaglobulinemia. These relationships are poorly understood. The objectives of these analyses were to estimate how much of this rituximab-associated infection risk is mediated by hypogammaglobulinemia and to identify other modifiable risk factors in persons with multiple sclerosis (pwMS). Methods: We conducted a retrospective cohort study of rituximab-treated pwMS from January 1, 2008, to December 31, 2020, in Kaiser Permanente Southern California. Cumulative rituximab dose was defined as ≤2, >2 and ≤4, or >4 g. Serious infections were defined as infections requiring or prolonging hospitalizations, and recurrent outpatient infections as seeking care for ≥3 within 12 months. Exposures, outcomes, and covariates were collected from the electronic health record. Adjusted hazard ratios (aHRs) were estimated using Andersen-Gill hazards models, and generalized estimating equations were used to examine correlates of IgG values. Cross-sectional causal mediation analyses of rituximab and hypogammaglobulinemia were conducted. Results: We identified 2,482 pwMS who were treated with rituximab for a median of 2.4 years (interquartile range = 1.3-3.9). The average age at rituximab initiation was 43.0 years, 71.9% were female, 49.7% were White, non-Hispanic patients, and 29.6% had advanced disability (requiring walker or worse). Seven hundred patients (28.2%) developed recurrent outpatient infections, 155 (6.2%) developed serious infections, and only 248 (10.0%) had immunoglobulin G (IgG) < 700 mg/dL. Higher cumulative rituximab dose (>4 g) was correlated with lower IgG levels (Beta = -58.8, p < 0.0001, ref ≤2 g) and, in models mutually adjusted for hypogammaglobulinemia, both were independently associated with an increased risk of serious (>4 g, aHR = 1.56, 95% CI 1.09-2.24; IgG < 500, aHR = 2.98, 95% CI 1.56-5.72) and outpatient infections (>4 g, aHR = 1.73, 95% CI 1.44-2.06; IgG < 500 aHR = 2.06, 95% CI 1.52-2.80; ref = IgG ≥ 700). Hypogammaglobulinemia explained at most 17.9% (95% CI -47.2-119%) of serious infection risk associated with higher cumulative rituximab exposure but was not significant for outpatient infections. Other independent modifiable risk factors were advanced physical disability for serious (aHR = 5.51, 95% CI 3.71-8.18) and outpatient infections (aHR = 1.24, 95% CI 1.06-1.44) and COPD (aHR = 1.68, 95% CI 1.34-2.11) and obesity (aHR = 1.25, 95% CI 1.09-1.45) for outpatient infections. Discussion: Higher cumulative rituximab doses increase the risk of infections even in this population where 90% of patients maintained normal IgG levels. Clinicians should strive to use minimally effective doses of rituximab and other B-cell-depleting therapies and consider important comorbidities to minimize risks of infections.
... A more recent prospective cohort study demonstrated no difference between clinical or neuroradiologic disease activity in patients stratified into four dosing intervals based on time since last infusion (< 8, ≥ 8-12, ≥ 12-18, and 18 months). Median total B-cell count reconstitution occurred after 12 months and median memory B-cell reconstitution occurred after 16 months [83]. In the initial phase II trials of ocrelizumab, progression and disease activity in the ocrelizumab cohort remained low for up to 18 months after the last infusion, suggesting longer-term efficacy [84][85][86]. ...
Article
Full-text available
Currently, there are four monoclonal antibodies (mAbs) that target the cluster of differentiation (CD) 20 receptor available to treat multiple sclerosis (MS): rituximab, ocrelizumab, ofatumumab, and ublituximab. B-cell depletion therapy has changed the therapeutic landscape of MS through robust efficacy on clinical manifestations and MRI lesion activity, and the currently available anti-CD20 mAb therapies for use in MS are a cornerstone of highly effective disease-modifying treatment. Ocrelizumab is currently the only therapy with regulatory approval for primary progressive MS. There are currently few data regarding the relative efficacy of these therapies, though several clinical trials are ongoing. Safety concerns applicable to this class of therapeutics relate primarily to immunogenicity and mechanism of action, and include infusion-related or injection-related reactions, development of hypogammaglobulinemia (leading to increased infection and malignancy risk), and decreased vaccine response. Exploration of alternative dose/dosing schedules might be an effective strategy for mitigating these risks. Future development of biosimilar medications might make these therapies more readily available. Although anti-CD20 mAb therapies have led to significant improvements in disease outcomes, CNS-penetrant therapies are still needed to more effectively address the compartmentalized inflammation thought to play an important role in disability progression.
Article
Full-text available
Background B-cell depletion displays striking effectiveness in relapsing-remitting multiple sclerosis (RRMS), but is also associated with increased infection risk. To what degree previous treatment history, disease-modifying therapy (DMT) switching pattern and time on treatment modulate this risk is unknown. The objective here was to evaluate previous DMT use and treatment duration as predictors of infection risk with B-cell depletion. Methods We conducted a nationwide RRMS cohort study leveraging data from the Swedish MS registry and national demographic and health registries recording all outpatient-treated and inpatient-treated infections and antibiotics prescriptions from 1 January 2012 to 30 June 2021. The risk of infection during treatment was compared by DMT, treatment duration, number and type of prior treatment and adjusted for a number of covariates. Results Among 4694 patients with RRMS on B-cell depletion (rituximab), 6049 on other DMTs and 20 308 age-sex matched population controls, we found higher incidence rates of inpatient-treated infections with DMTs other than rituximab used in first line (10.4; 95% CI 8.1 to 12.9, per 1000 person-years), being further increased with rituximab (22.7; 95% CI 18.5 to 27.5), compared with population controls (6.6; 95% CI 6.0 to 7.2). Similar patterns were seen for outpatient infections and antibiotics prescriptions. Infection rates on rituximab did not vary between first versus later line treatment, type of DMT before switch or exposure time. Conclusion These findings underscore an important safety concern with B-cell depletion in RRMS, being evident also in individuals with shorter disease duration and no previous DMT exposure, in turn motivating the application of risk mitigation strategies.
Article
Full-text available
BACKGROUND Secondary thyroid autoimmunity, especially Graves’ disease (GD), frequently develops in patients with multiple sclerosis (MS) following alemtuzumab treatment (ALTZ; anti-CD52). Thyroid Eye Disease (TED) can also develop, and rituximab (RTX; anti-CD20) is a suitable treatment. METHODS Immunophenotyping of blood and thyroid-derived lymphocytes in a patient treated with both ALTZ and RTX. RESULTS A 37-year-old woman with MS developed steroid-resistant active moderate-to-severe TED three years after ALTZ, that successfully responded to a single 500 mg dose of i.v. RTX. Before RTX peripheral B-cells were low, and were totally depleted immediately after therapy. Follow-up analysis four years post-ALTZ and one year post-RTX, showed persistent depletion of B cells, and relative reduction of T regulatory cells in both peripheral blood and thyroid tissue obtained at thyroidectomy. CONCLUSIONS RTX therapy successfully inactivated TED in a patient with low B-cell count derived from previous ALTZ treatment. B-cell depletion in both thyroid and peripheral blood was still present one year after RTX, as a likely cumulative effect of both treatments.
Article
Full-text available
Background: Recent findings document a blunted humoral response to SARS-CoV-2 vaccination in patients on anti-CD20 treatment. Although most patients develop a cellular response, it is still important to identify predictors of seroconversion in order to optimize vaccine responses. Methods: We determined antibody responses after SARS-CoV-2 vaccination in a real-world cohort of multiple sclerosis patients (n = 94) treated with anti-CD20, mainly rituximab, with variable treatment duration (median 2.9; range 0.4-9.6 years) and time from last anti-CD20 infusion to vaccination (median 190; range 60-1032 days). Results: We find that presence of B cells and/or rituximab in blood predict seroconversion better than time since last infusion. Using multiple logistic regression, presence of >0.5% B cells increased probability for seroconversion with an odds ratio (OR) of 5.0 (CI 1.0-28.1, p = 0.055), while the corresponding OR for ≥ 6 months since last infusion was 1.45 (CI 0.20-10.15, p = 0.705). In contrast, detectable rituximab levels were negatively associated with seroconversion (OR 0.05; CI 0.002-0.392, p = 0.012). Furthermore, naïve and memory IgG+ B cells correlated with antibody levels. Although re-treatment with rituximab at four weeks or more after booster depleted spike-specific B cells, it did not noticeably affect the rate of decline in antibody titers. Interferon-γ and/or interleukin-13 T cell responses to the spike S1 domain were observed in most patients, but with no correlation to spike antibody levels. Conclusions: These findings are relevant for providing individualized guidance to patients and planning of vaccination schemes, in turn optimizing benefit-risk with anti-CD20.
Article
Full-text available
Background People with multiple sclerosis (MS) are a vulnerable group for severe COVID- 19, particularly those taking immunosuppressive disease-modifying therapies (DMTs). We examined the characteristics of COVID-19 severity in an international sample of people with MS. Methods Data from 12 data-sources in 28 countries were aggregated (sources could include patients from 1-12 countries). Demographic (age, sex), clinical (MS-phenotype, disability), and DMT (untreated, alemtuzumab, cladribine, dimethyl-fumarate, glatiramer acetate, interferon, natalizumab, ocrelizumab, rituximab, siponimod, other DMTs) covariates were queried, alongside COVID-19 severity outcomes, hospitalisation, ICU admission, requiring artificial ventilation, and death. Characteristics of outcomes were assessed in patients with suspected/confirmed COVID-19 using multilevel mixed-effects logistic regression, adjusted for age, sex, MS-phenotype, and EDSS. Results 657(28.1%) with suspected and 1,683(61.9%) with confirmed COVID-19 were analysed. Among suspected+confirmed and confirmed-only COVID-19, 20.9% and 26.9% were hospitalised, 5.4% and 7.2% were admitted to ICU, 4.1% and 5.4% required artificial ventilation, and 3.2% and 3.9% died. Older age, progressive MS-phenotype, and higher disability were associated with worse COVID-19 outcomes. Compared to dimethyl-fumarate, ocrelizumab and rituximab were associated with hospitalisation (aOR=1.56,95%CI=1.01- 2.41; aOR=2.43,95%CI=1.48-4.02) and ICU admission (aOR=2.30,95%CI=0.98-5.39; aOR=3.93,95%CI=1.56-9.89), though only rituximab was associated with higher risk of artificial ventilation (aOR=4.00,95%CI=1.54-10.39). Compared to pooled other DMTs, ocrelizumab and rituximab were associated with hospitalisation (aOR=1.75,95%CI=1.29- 2.38; aOR=2.76,95%CI=1.87-4.07) and ICU admission (aOR=2.55,95%CI=1.49-4.36; aOR=4.32,95%CI=2.27-8.23) but only rituximab with artificial ventilation (aOR=6.15,95%CI=3.09-12.27). Compared to natalizumab, ocrelizumab and rituximab were associated with hospitalisation (aOR=1.86,95%CI=1.13-3.07; aOR=2.88,95%CI=1.68-4.92) and ICU admission (aOR=2.13,95%CI=0.85-5.35; aOR=3.23,95%CI=1.17-8.91), but only rituximab with ventilation (aOR=5.52,95%CI=1.71-17.84). Importantly, associations persisted on restriction to confirmed COVID-19 cases. No associations were observed between DMTs and death. Stratification by age, MS-phenotype, and EDSS found no indications that DMT associations with COVID-19 severity reflected differential DMT allocation by underlying COVID-19 severity. Conclusions Using the largest cohort of people with MS and COVID-19 available, we demonstrated consistent associations of rituximab with increased risk of hospitalisation, ICU admission, and requiring artificial ventilation, and ocrelizumab with hospitalisation and ICU admission. Despite the study’s cross-sectional design, the internal and external consistency of these results with prior studies suggests rituximab/ocrelizumab use may be a risk factor for more severe COVID-19.
Article
Full-text available
SARS-CoV-2 messenger RNA vaccination in healthy individuals generates immune protection against COVID-19. However, little is known about SARS-CoV-2 mRNA vaccine-induced responses in immunosuppressed patients. We investigated induction of antigen-specific antibody, B cell and T cell responses longitudinally in patients with multiple sclerosis (MS) on anti-CD20 antibody monotherapy ( n = 20) compared with healthy controls ( n = 10) after BNT162b2 or mRNA-1273 mRNA vaccination. Treatment with anti-CD20 monoclonal antibody (aCD20) significantly reduced spike-specific and receptor-binding domain (RBD)-specific antibody and memory B cell responses in most patients, an effect ameliorated with longer duration from last aCD20 treatment and extent of B cell reconstitution. By contrast, all patients with MS treated with aCD20 generated antigen-specific CD4 and CD8 T cell responses after vaccination. Treatment with aCD20 skewed responses, compromising circulating follicular helper T (T FH ) cell responses and augmenting CD8 T cell induction, while preserving type 1 helper T (T H 1) cell priming. Patients with MS treated with aCD20 lacking anti-RBD IgG had the most severe defect in circulating T FH responses and more robust CD8 T cell responses. These data define the nature of the SARS-CoV-2 vaccine-induced immune landscape in aCD20-treated patients and provide insights into coordinated mRNA vaccine-induced immune responses in humans. Our findings have implications for clinical decision-making and public health policy for immunosuppressed patients including those treated with aCD20.
Article
Full-text available
B-cell depleting therapies (BCDTs) are widely used as immunomodulating agents for autoimmune diseases such as multiple sclerosis. Their possible impact on development of immunity to SARS-CoV-2 has raised concerns with the COVID-19 pandemic. We here evaluated the frequency of COVID-19-like symptoms and determined immunological responses in participants of an observational trial comprising several multiple sclerosis disease modulatory drugs, (COMBAT-MS; NCT03193866) and in eleven patients after vaccination, with a focus on BCDT. Almost all seropositive and 17.9% of seronegative patients on BCDT, enriched for a history of COVID-19-like symptoms, developed anti-SARS-CoV-2 T-cell memory and T-cells displayed functional similarity to controls producing IFN-γ and TNF. Following vaccination, vaccine-specific humoral memory was impaired, while all patients developed a specific T-cell response. These results indicate that BCDTs do not abrogate SARS-CoV-2 cellular memory and provide a possible explanation as to why the majority of patients on BCDTs recover from COVID-19.
Article
Full-text available
Objective To evaluate the clinical consequences of extended interval dosing (EID) of ocrelizumab in relapsing-remitting multiple sclerosis (RRMS) during the coronavirus disease 2019 (COVID-19) pandemic. Methods In our retrospective, multicenter cohort study, we compared patients with RRMS on EID (defined as ≥4-week delay of dose interval) with a control group on standard interval dosing (SID) at the same period (January to December 2020). Results Three hundred eighteen patients with RRMS were longitudinally evaluated in 5 German centers. One hundred sixteen patients received ocrelizumab on EID (median delay [interquartile range 8.68 [5.09–13.07] weeks). Three months after the last ocrelizumab infusion, 182 (90.1%) patients following SID and 105 (90.5%) EID patients remained relapse free ( p = 0.903). Three-month confirmed progression of disability was observed in 18 SID patients (8.9%) and 11 EID patients (9.5%, p = 0.433). MRI progression was documented in 9 SID patients (4.5%) and 8 EID patients (6.9%) at 3-month follow-up ( p = 0.232). Multivariate logistic regression showed no association between treatment regimen and no evidence of disease activity status at follow-up (OR: 1.266 [95% CI: 0.695–2.305]; p = 0.441). Clinical stability was accompanied by persistent peripheral CD19 ⁺ B-cell depletion in both groups (SID vs EID: 82.6% vs 83.3%, p = 0.463). Disease activity in our cohort was not associated with CD19 ⁺ B-cell repopulation. Conclusion Our data support EID of ocrelizumab as potential risk mitigation strategy in times of the COVID-19 pandemic. Classification of Evidence This study provides Class IV evidence that for patients with RRMS, an EID of at least 4 weeks does not diminish effectiveness of ocrelizumab.
Article
Full-text available
In this observational study, 159 patients with multiple sclerosis received personalized dosing of ocrelizumab incentivized by the COVID-19 pandemic. Re-dosing was scheduled when CD19 B-cell count was ⩾10 cells/µL (starting 24 weeks after the previous dose, repeated 4-weekly). Median interval until re-dosing or last B-cell count was 34 [30–38] weeks. No clinical relapses were reported and a minority of patients showed Expanded Disability Status Scale (EDSS) progression. Monthly serum neurofilament light levels remained stable during extended intervals. Two (1.9%) of 107 patients with a follow-up magnetic resonance imaging (MRI) scan showed radiological disease activity. Personalized dosing of ocrelizumab could significantly extend intervals with low short-term disease activity incidence, encouraging future research on long-term safety and efficacy.
Article
Full-text available
Importance: Emergence of SARS-CoV-2 causing COVID-19 prompted the need to gather information on clinical outcomes and risk factors associated with morbidity and mortality in patients with multiple sclerosis (MS) and concomitant SARS-CoV-2 infections. Objective: To examine outcomes and risk factors associated with COVID-19 clinical severity in a large, diverse cohort of North American patients with MS. Design, setting, and participants: This analysis used deidentified, cross-sectional data on patients with MS and SARS-CoV-2 infection reported by health care professionals in North American academic and community practices between April 1, 2020, and December 12, 2020, in the COVID-19 Infections in MS Registry. Health care professionals were asked to report patients after a minimum of 7 days from initial symptom onset and after sufficient time had passed to observe the COVID-19 disease course through resolution of acute illness or death. Data collection began April 1, 2020, and is ongoing. Exposures: Laboratory-positive SARS-CoV-2 infection or highly suspected COVID-19. Main outcomes and measures: Clinical outcome with 4 levels of increasing severity: not hospitalized, hospitalization only, admission to the intensive care unit and/or required ventilator support, and death. Results: Of 1626 patients, most had laboratory-positive SARS-CoV-2 infection (1345 [82.7%]), were female (1202 [74.0%]), and had relapsing-remitting MS (1255 [80.4%]). A total of 996 patients (61.5%) were non-Hispanic White, 337 (20.8%) were Black, and 190 (11.7%) were Hispanic/Latinx. The mean (SD) age was 47.7 (13.2) years, and 797 (49.5%) had 1 or more comorbidity. The overall mortality rate was 3.3% (95% CI, 2.5%-4.3%). Ambulatory disability and older age were each independently associated with increased odds of all clinical severity levels compared with those not hospitalized after adjusting for other risk factors (nonambulatory: hospitalization only, odds ratio [OR], 2.8 [95% CI, 1.6-4.8]; intensive care unit/required ventilator support, OR, 3.5 [95% CI, 1.6-7.8]; death, OR, 25.4 [95% CI, 9.3-69.1]; age [every 10 years]: hospitalization only, OR, 1.3 [95% CI, 1.1-1.6]; intensive care unit/required ventilator support, OR, 1.3 [95% CI, 0.99-1.7]; death, OR, 1.8 [95% CI, 1.2-2.6]). Conclusions and relevance: In this registry-based cross-sectional study, increased disability was independently associated with worse clinical severity including death from COVID-19. Other risk factors for worse outcomes included older age, Black race, cardiovascular comorbidities, and recent treatment with corticosteroids. Knowledge of these risk factors may improve the treatment of patients with MS and COVID-19 by helping clinicians identify patients requiring more intense monitoring or COVID-19 treatment.
Article
Full-text available
Background Rituximab is safe and effective for treating relapsing–remitting multiple sclerosis (RRMS) according to phase II and observational studies. There are limited data on disease activity after discontinuation and dose reduction. The objective of this study was to evaluate the effects on inflammatory disease activity after discontinuation or dose reduction of rituximab in patients with RRMS or clinically isolated syndrome (CIS). Methods In this retrospective observational study, we included all RRMS and CIS patients ever treated with rituximab at the University Hospital of Umeå who had either; (1) discontinued treatment at any time or (2) reduced the dose to a mean of < 1000 mg yearly. The patients served as their own controls by contributing patient years on full dose, reduced dose, and off treatment. Results A total of 225 patients treated with mean (SD) 6256 (2456) mg rituximab during mean (SD) 6.5 (2.0) years were included. There were no differences regarding the annualized relapse rates during full dose versus reduced dose or off treatment (0.02 versus < 0.01 and 0.02, p = 0.09), neither regarding proportion MRI scans with new or enlarged T2 lesions (0.03 versus 0.01 and 0.03, p = 0.37) or contrast-enhancing lesions (< 0.01 versus 0 and 0.02, p = 0.22). Conclusions This study indicates that rituximab has long-term effects on inflammatory disease activity and that disease reactivation is rare in MS patients who discontinued treatment for any reason. It also suggests that treatment with low-dose rituximab (< 1000 mg yearly) is sufficient to maintain suppression of inflammatory disease activity in patients with stable disease.
Article
Background Covid-19 may spread through various ways ranging from asymptomatic to severe forms, until respiratory failure, critical conditions and death occurs. There is a particular concern for patients affected by multiple sclerosis, especially for those under disease-modifying treatments. Some studies have found an association between anti-CD20 therapies (especially rituximab) and severe Covid-19. However, results were not always clear and thus a systematic review was helpful Methods A systematic literature search was performed independently by two authors on the main search tools considering as key inclusion criterion the presence of data on patients under ocrelizumab or rituximab positive to Covid-19. The quality of the included studies was evaluated based on a modified version of the Dutch Cochrane Centre critical review checklist proposed by MOOSE and in case of missing data an email was sent to the corresponding authors asking for missing information. After excluding case-reports, a random effects meta-analysis of proportions was conducted using the continuity correction and the I² statistic was calculated to measure heterogeneity. Results 29 articles were included in the analysis and the median quality of the articles reached 4/5 after having integrated the additional details provided by the authors. The articles included 5173 patients, of whom 770 (14.8%) and 455 (8.8%) were respectively under ocrelizumab and rituximab. Pooled estimates of hospitalization, pneumonia and intensive care unit admission were 18.1%, 14.8% and 3.3%, respectively, while pooled estimate for death was 1.8% overall and 1.6% and 4.5% respectively for patients under ocrelizumab and rituximab. Conclusion Patients treated with rituximab seem to be at higher risk of severe Covid-19 outcomes compared to patients under other treatments.