OLYMPUS Rituximab

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

ORIGINAL ARTICLES

Rituximab in Patients with Primary


Progressive Multiple Sclerosis
Results of a Randomized Double-Blind Placebo-Controlled
Multicenter Trial
Kathleen Hawker, MD,1 Paul O’Connor, MD,2 Mark S. Freedman, MD,3 Peter A. Calabresi, MD,4
Jack Antel, MD,5 Jack Simon, MD,6 Stephen Hauser, MD,7 Emmanuelle Waubant, MD,7
Timothy Vollmer, MD,8 Hillel Panitch, MD,9 Jiameng Zhang, PhD,10 Peter Chin, MD,10 and
Craig H. Smith, MD,10 for the OLYMPUS trial group

Objective: Rituximab, a monoclonal antibody selectively depleting CD20⫹ B cells, has demonstrated efficacy in reducing
disease activity in relapsing-remitting multiple sclerosis (MS). We evaluated rituximab in adults with primary progressive MS
(PPMS) through 96 weeks and safety through 122 weeks.
Methods: Using 2:1 randomization, 439 PPMS patients received two 1,000mg intravenous rituximab or placebo infusions
every 24 weeks, through 96 weeks (4 courses). The primary endpoint was time to confirmed disease progression (CDP), a
prespecified increase in Expanded Disability Status Scale sustained for 12 weeks. Secondary endpoints were change from baseline
to week 96 in T2 lesion volume and total brain volume on magnetic resonance imaging scans.
Results: Differences in time to CDP between rituximab and placebo did not reach significance (96-week rates: 38.5% placebo,
30.2% rituximab; p ⫽ 0.14). From baseline to week 96, rituximab patients had less ( p ⬍ 0.001) increase in T2 lesion volume;
brain volume change was similar ( p ⫽ 0.62) to placebo. Subgroup analysis showed time to CDP was delayed in rituximab-
treated patients aged ⬍51 years (hazard ratio [HR] ⫽ 0.52; p ⫽ 0.010), those with gadolinium-enhancing lesions (HR ⫽ 0.41;
p ⫽ 0.007), and those aged ⬍51 years with gadolinium-enhancing lesions (HR ⫽ 0.33; p ⫽ 0.009) compared with placebo.
Adverse events were comparable between groups; 16.1% of rituximab and 13.6% of placebo patients reported serious events.
Serious infections occurred in 4.5% of rituximab and ⬍1.0% of placebo patients. Infusion-related events, predominantly mild
to moderate, were more common with rituximab during the first course, and decreased to rates comparable to placebo on
successive courses.
Interpretation: Although time to CDP between groups was not significant, overall subgroup analyses suggest selective B-cell
depletion may affect disease progression in younger patients, particularly those with inflammatory lesions.
Ann Neurol 2009;66:460 – 471

Multiple sclerosis (MS), a chronic inflammatory demy- Relapsing forms of MS are characterized by periods of
elinating disease of the central nervous system, is a neurological deterioration (relapses) and partial or
leading cause of neurologic disability in young adults.1 complete recovery (remission), with (secondary pro-

1
Department of Neurology, The Ohio State University Medical Bayer Healthcare, Eli Lilly, Eisai, Novartis, Genentech; Dr Calabrasi
Center, Columbus, OH; 2Department of Medicine, University of has received honoraria for consulting from: Teva, Biogen-IDEC,
Toronto, Toronto, ON, Canada; 3Department of Medicine, Ottawa Genentech, Serono, Novartis, Amplimmune, Vertex, and Centacor,
Hospital-General Campus, Ottawa, ON, Canada; 4Department of PC has received research support from Bayer, Serono, Biogen-
Neurology, John Hopkins University, Baltimore, MD; 5Department IDEC, Teva, and Genentech; Dr Waubant’s research has been sup-
of Neurology and Neurosurgery, McGill University, Montreal, QC, ported by Biogen-Idec, Pfizer, Sanofi-Aventis, the National MS So-
Canada; 6Department of Neurology, Portland VA Medical Center, ciety, the Immune Tolerance Network and the Nancy Davis
Portland, OR; 7Department of Neurology, University of California, Foundation. Dr Waubant has been a consultant for Artielle and
San Francisco, San Francisco, CA; 8Department of Neurology, Uni- Zymogenetics. She has received honorarium for one educational
versity of Colorado, Denver, CO; 9Department of Neurology, Uni- presentation from Biogen Idec; Dr Hauser, Dr Panitch, Dr Vollmer
versity of Vermont, Burlington, VT; and 10Genentech Inc., South have nothing to disclose.
San Francisco, San Francisco.
Additional Supporting Information may be found in the online ver-
Address correspondence to Dr Hawker, Eli Lilly, 450 South Merid- sion of this article.
ian Street, Indianapolis, IN 46225, E-mail: hawkerks@lilly.com
Other investigators who participated in the OLYMPUS Trial Group
Potential conflict of interest: Dr Hawker has received honoraria are listed in the Appendix on page xxx.
from Biogen IDEC, Teva, Bayer, Genentech and GSK; Dr
O’Connor has received honoraria from Bio MS, BiogenIDEC, Received Aug 5, 2009, and in revised form Aug 19. Accepted for
Teva, Serono, Roche, Genentech, Bayer; Dr Freedman has received publication Sep 1, 2009. Published online in Wiley InterScience
honoraria from Sanofi-Aventis, BiogenIdec, EMD Serono, Teva, (www.interscience.wiley.com). DOI: 10.1002/ana.21867

460 © 2009 American Neurological Association


MRI:

Study
Drug
Dose:

1000 1000 1000 1000


mg x2 mg x2 mg x2 mg x2

Week: -4 -2 0 2 24 26 48 50 72 74 96 122

Treatment Follow-up
Screen Pre-Treatment 96 wks 26 wks
2 wks 2 wks

Fig 1. Study design. The study consisted of 4 periods: a screening period (week ⫺4 to week ⫺2 [days ⫺28 to ⫺15]), a pretreat-
ment period (week ⫺2 to week 0 [days ⫺14 to ⫺1]), a treatment period (week 0 [day 1] to week 96), and a follow-up period
(weeks 97 to 122). The first course of study drug treatment was administered on days 1 and 15. Patients were dosed every 24
weeks, with the last course of treatment administered at weeks 72 and 74. When patients completed the 96-week treatment period,
they entered the follow-up period. Patients who withdrew or discontinued from the study early were followed for collection of safety
data for 48 weeks from the time of their last dose of study drug. Primary endpoint: time to confirmed disease progression during
96-week treatment period (requires 12-week confirmation); safety and tolerability; subgroup analysis for age and gadolinium lesion.
Secondary endpoint: change in T2 lesion volume from baseline to week 96; change in brain volume from baseline to week 96.
MRI ⫽ magnetic resonance imaging.

gressive MS) or without (relapsing remitting MS part a B-cell–mediated disease, we initiated a 96-week
[RRMS]) progression between exacerbations. RRMS is double-blind placebo-controlled trial with rituximab,
the most common type, seen in ⬎80% of patients.2 an anti-CD20 monoclonal antibody. Rituximab (Rit-
The remaining MS patients present with gradually in- uxan) is a genetically engineered chimeric monoclonal
creasing neurological disability, primarily affecting am- antibody that targets the CD20 antigen expressed on B
bulation, without discernable relapses; this type, pri- lymphocytes16 from the pre–B-cell stage through dif-
mary progressive MS (PPMS), has a mean age of onset ferentiation into B cells, but excluding plasma cells.
a decade later than RRMS.1,3 To date, there are no Our objectives in this trial (OLYMPUS) were to as-
proven disease-modifying treatments that slow the sess the efficacy of rituximab relative to placebo, as
course of PPMS. A double-blind placebo-controlled measured by the time to confirmed disease progression
trial with glatiramer acetate4 and small exploratory tri- over a 96-week treatment period, and to evaluate the
als with interferon agents5,6 failed to demonstrate effi- safety and tolerability of rituximab in patients with
cacy in PPMS patients. An effective treatment for PPMS.
PPMS remains an unmet medical need.
In contrast to earlier disease concepts suggesting that Subjects and Methods
pathogenic T cells were sufficient for full expression of
MS, autoimmune B cells and humoral immune mech- Study Design and Randomization
This was a phase II/III randomized, double-blind, placebo-
anisms are now believed to play key roles in disease
controlled trial from 60 centers in the US and Canada de-
pathogenesis.7 A humoral component in MS has been signed jointly by Genentech, Biogen Idec, and the investiga-
implicitly recognized for decades, evidenced by inclu- tors. The protocol was approved by the institutional review
sion of cerebrospinal fluid (CSF) oligoclonal bands and board and the ethics committee of each institution. Written
increased intrathecal immunoglobulin (Ig) G synthesis informed consent was obtained from each patient or the pa-
in MS diagnostic criteria.8 –10 Antibody deposition and tient’s legal guardian. Patients were randomly assigned 2:1 to
immune complement activation associated with vesicu- receive rituximab or placebo (Fig 1), and hierarchically strati-
lar disintegration of the myelin membrane is present in fied according to study site, previous MS therapies with
most MS lesions,11–13 and autoantibody responses interferon-beta or glatiramer acetate (no prior MS therapy,
against many antigens can also be detected in the CSF MS therapies that ended ⬎90 days prior to randomization,
of many MS patients.14 The evidence for diffuse pa- MS therapies that ended within 90 days prior to randomiza-
tion), and baseline disease severity according to the Expanded
thology with demyelination and axonal loss but less
Disability Status Scale (EDSS) score (ⱕ4.0 vs ⬎4.0), an or-
overt inflammation may indicate an antibody-mediated dinal scale ranging from 0 (normal neurologic examination) to
pathogenesis in PPMS.15 10.0 (death) in 0.5-step intervals.17 Patients received 1,000mg
To date, therapies partially effective in relapsing intravenous infusions of rituximab or placebo on study weeks
forms of MS have failed to alter the course of PPMS. 0, 2, 24, 26, 48, 50, 72, and 74 (see Fig 1). Data collected by
Based on what we know about the immunopathology the investigators was held and analyzed by Genentech. Mem-
of MS, and our hypothesis that PPMS is at least in bers of the publication committee had full access to data, and

Hawker et al: Rituximab in PPMS 461


all authors vouch for the veracity and completeness of the data Safety assessments were made on patients who received any
and data analysis. The study was powered to detect a 50% amount of study drug (rituximab or placebo).
relative reduction in the 2-year disease progression rate, with For the primary efficacy analysis, the stratified log-rank
about 90% power and 0.01 type I error level. The assumed test, using pretreatment severity measured by EDSS score
confirmed disease progression rate for the placebo arm was and status of prior interferon-beta or glatiramer acetate ther-
32% at 96 weeks, estimated from the 2-year placebo disease apies for MS as stratifying factors, was applied to compare
progression rate observed in the PROMiSe trial.4 differences in time to CDP between the 2 treatment groups.
The alpha level at the time of the final analysis was adjusted
Patients to 0.0499 for the interim futility analysis at 48 weeks to
Enrollment was limited to patients 18 to 65 years old with a maintain an overall type I error level of 0.05. A Cox propor-
PPMS diagnosis18; disease duration of ⱖ1 year; EDSS base- tional hazard model using the same stratifying factors as the
line between 2.0 and 6.5 points, inclusive; Functional Sys- primary analysis was used to estimate the reduction in hazard
tems scale score of ⱖ2.0 for the pyramidal system or gait associated with treatment compared with placebo. Ranked
impairment due to lower extremity dysfunction; and pres- analysis of variance was used to compare the difference be-
ence of IgG oligoclonal bands or elevated CSF IgG index, or tween the rituximab and placebo group in endpoints related
both, from CSF specimen obtained at screening or docu- to change from baseline.
mented during the previous 24 months. Key exclusion crite- There was no imputation of missing data for assessment of
ria included history of MS exacerbation or neuromyelitis op- time to CDP. If a patient missed a scheduled visit that was
tica; history of myelopathy or neurodegenerative central 84 days after the initial detection of disease progression,
nervous system conditions; systemic autoimmune disorders; CDP was based on the assessment at the next scheduled visit.
recurrent or chronic infections; recent treatment with immu- Patients with an initial disease progression who subsequently
nomodulating or immunosuppressant therapies; and meta- discontinued the study treatment before a subsequent confir-
bolic, hematologic, or immunologic laboratory abnormalities. matory assessment could be obtained were considered to
have CDP. Data from patients who discontinued the study
Study Procedures and Endpoints early or were lost to follow-up and had not shown an initial
The primary efficacy outcome measure was time to confirmed disease progression were censored at time of their last visit.
disease progression (CDP), defined as a sustained EDSS in- Patients who demonstrated an initial disease progression at
crease of ⱖ1.0 point from baseline EDSS if the baseline EDSS the end of the treatment period (week 96) were censored at
was between 2.0 and 5.5 points (inclusive), or an EDSS in- that time and were not considered to have CDP. The EDSS
crease of ⱖ0.5 point if the baseline EDSS was ⬎5.5 points, assessments within 30 days of protocol-defined relapse were
for which change was not attributable to another etiology (eg, excluded for the confirmation of CDP.
fever, concurrent illness, injury, adverse reactions to concur- Hochberg-Bonferroni procedure was used to control the
rent medications, or relapse) sustained for ⱖ12 weeks. Sec- type I error level in testing the 2 secondary endpoints. No
ondary efficacy outcome measures were change from baseline statistical adjustment for multiple comparisons was made for
to week 96 in volume of T2 lesions and change in brain vol- exploratory analyses. For endpoints related to change from
ume, as measured by brain parenchymal fraction, on magnetic baseline, the last-observed value before the treatment period
resonance imaging (MRI) brain scans. Key exploratory out- discontinuation was used to impute the missing values. Sub-
comes included time to CDP sustained for ⱖ24 weeks; group analysis for continuous variables were conducted using
change in Multiple Sclerosis Functional Composite (MSFC) median values as cut-points, as well as additional prespecified
summary and component scale scores; pharmacodynamic as- thresholds. Adverse events were graded according to the Na-
sessments of B-cell counts and immunoglobulin levels; and tional Cancer Institute Common Terminology Criteria for
prespecified subgroup analysis by age, gender, race/ethnicity, Adverse Events, version 3.0.19 Summaries of adverse events,
use of prior therapy, EDSS at baseline, duration since MS serious adverse events, deaths, laboratory test results, vital
symptom onset, and presence of gadolinium-enhancing lesions signs, and antibodies to rituximab were presented.
on brain MRI at baseline. EDSS and MSFC scores were as-
sessed at least quarterly by a neurologist blinded to treatment
and other patient data including imaging and laboratory re- Results
sults. MRI scans were blindly evaluated at the University of Study Population
Colorado Brain Imaging Research Laboratory, an independent A CONSORT diagram showing the flow of partici-
MRI reading facility. An independent Data Monitoring Com- pants through each stage of this randomized, placebo-
mittee met formally every 3 months to review unblinded controlled trial is illustrated in Figure 2. Baseline char-
safety data, including MRI outcomes, clinical outcomes, and acteristics (Table 1) of 439 patients were balanced
adverse events. Patients had an extra EDSS determination and between treatment groups. Patients had longstanding
MRI assessment 4 weeks after completing the first course of
disease, with ⬃65% having no prior MS therapies;
study drug treatment for safety to assess for activation or wors-
ening of MS. An interim futility analysis was carried out after
mean EDSS score was 4.8, and over half had an EDSS
about 50% patients finished 48 weeks of treatment. score of ⱖ4.0. Approximately 50% of study patients
were males, and the median age of patients was 51
Statistical Analysis years. At baseline, 25% of patients had gadolinium le-
Efficacy outcomes were analyzed according to the intention- sions. Of 439 patients, 84.4% in the placebo group
to-treat principle and included all patients randomized. and 82.5% in the rituximab group completed the 96

462 Annals of Neurology Vol 66 No 4 October 2009


608 patients assessed for
eligibility 138 not meeting
inclusion criteria
11 withdrew consent
1 lost to follow up
19 other reasons
439 patients enrolled and
randomized to study groups

292 assigned to receive rituximab 147 assigned to receive placebo


(ITT population) (ITT population)

2 lost to follow up
2 lost to follow up
49 withdraw treatments
8 had adverse event 21 withdraw treatments
27 patient’s decision 1 death
7 physician’s decision 11 patient’s decision
1 init of excluded therapy 4 physician’s decision
1 subject non-compliance 5 disease progression
1 pregnancy
4 disease progression

292 patients included in the 147 patients included in the


analysis analysis

Among them, 241 completed 96 Among them, 124 completed 96


weeks treatment period weeks treatment period

Fig 2. CONSORT diagram showing flow of participants through each stage of the OLYMPUS trial. ITT ⫽ intention-to-treat
population.

weeks (Table 2). Of 365 patients who completed the crease) was similar ( p ⫽ 0.62; see Table 3). The me-
96-week treatment period, 340 completed the 122- dian increase in T2 lesion volume was 301.95 mm3 for
week safety follow-up (93.5% placebo, 92.9% ritux- rituximab and 809.50 mm3 for placebo.
imab). Six (2.05%) patients in the rituximab group
and 5 (3.40%) patients in the placebo group had KEY EXPLORATORY ENDPOINTS
protocol-defined relapses during the trial. Compared with placebo, rituximab patients had less
worsening in the MSFC timed 25-foot walk test ( p ⫽
Efficacy 0.04, p ⫽ 0.076, and p ⫽ 0.015 at weeks 48, 96, and
PRIMARY ENDPOINT 122, respectively, see Table 3). Median increase from
During the 96-week treatment period, there was no ev- baseline to week 96 in the timed 25-foot walk test was
idence of significant difference in time to CDP between 1.48 seconds for placebo patients and 0.9 seconds for
the rituximab and placebo groups ( p ⫽ 0.1442; Table rituximab patients. Results for other key exploratory
3, Fig 3A), although rituximab tended to delay the time endpoints were similar between treatment arms (time
to CDP compared with placebo as measured by a strat- to CDP sustained for ⱖ24 weeks; change of MSFC
ified hazard ratio of 0.77. Week 96 CDP rates were score; 9-Hole Peg Test; Paced Auditory Serial test from
38.5% for the placebo and 30.2% for the rituximab baseline to weeks 48, 96, and 122).
group.
SUBGROUP ANALYSES
SECONDARY ENDPOINTS Planned subgroup analyses of the primary endpoint (see
From baseline to week 96, compared with patients re- Fig 3B) indicated that age and presence of gadolinium
ceiving placebo, those receiving rituximab had signifi- lesions were predictors for treatment effect. Baseline char-
cantly less ( p ⬍ 0.001) increase in T2 lesion volume acteristics of these patients are listed in the Supplemental
on brain MRI scans, whereas brain volume change (de- Table. Compared with placebo, time to CDP was delayed

Hawker et al: Rituximab in PPMS 463


Table 1. Patient Baseline Demographic Characteristics
Demographic Placebo, nⴝ147 Rituximab, nⴝ292 Total, Nⴝ439

Age, mean yr (SD) 49.6 (8.7) 50.1 (9.0) 49.9 (8.9)


Gender, No. (%)
Male 66 (44.9) 152 (52.1) 218 (49.7)
Female 81 (55.1) 140 (47.9) 221 (50.3)
Ethnicity, No. (%)
White 134 (91.2) 268 (91.8) 402 (91.6)
Other 13 (8.8) 24 (8.2) 37 (8.4)
Disease duration, mean yr (SD)
Since onset 9.0 (6.8) 9.2 (6.4) 9.1 (6.6)
Since diagnosis 3.8 (4.2) 4.1 (4.2) 4.0 (4.2)
Prior MS treatment with IFN-beta or glatiramer acetate, No. (%)
No prior MS therapies 96 (65.3) 189 (64.7) 285 (64.9)
Ended ⬎90 days before trial entry 45 (30.6) 87 (29.8) 132 (30.1)
Ended ⱕ90 days before trial entry 6 (4.1) 16 (5.5) 22 (5.0)
EDSS score
Median (min, max) 4.5 (2.0, 6.5) 5.0 (2.0, 6.5) 5.0 (2.0, 6.5)
Mean (SD) 4.7 (1.4) 4.8 (1.4) 4.8 (1.4)
EDSS stratification, No. (%)
ⱕ4.0 68 (46.3) 124 (42.5) 192 (43.7)
⬎4.0 79 (53.7) 168 (57.5) 247 (56.3)
Total gadolinium-enhancing lesion count, No. (%)
⫽0 110 (74.8) 220 (75.9) 330 (75.5)
⬎0 37 (25.2) 70 (24.1) 107 (24.5)
Brain volume, cm3
Median 1,209.5 1,204.0 1,207.0
Minimum–maximum 642.0–1,522.0 712.60–1,508.0 642.0–1,522.0
Mean (SD) 1,210.91 (128.89) 1,202.92 (120.23) 1,205.75 (123.25)
T2 lesion volume, mm3
Median 5,199.50 5,240.50 5,220.70
Minimum–maximum 73.83–74,534.0 174.00–155,303.0 73.83–155,303.0
Mean (SD) 8,850.86 (11,808.95) 9,336.66 (13,744.94) 9,173.25 (13,113.98)
MSFC timed 25-foot walk test, 7.38 8.35 8.08
median seconds
SD ⫽ standard deviation; IFN ⫽ interferon; MS ⫽ multiple sclerosis; EDSS ⫽ Expanded Disability Status Scale; MSFC ⫽ Multiple
Sclerosis Functional Composite.

in rituximab-treated patients aged ⬍51 years (hazard ratio dition, there appeared to be a greater treatment effect of
[HR] ⫽ 0.52; p ⫽ 0.010) or those with gadolinium brain rituximab on T2 lesions for patients aged ⬍51 years with
lesions at baseline MRI (HR ⫽ 0.41; p ⫽ 0.007). More- baseline gadolinium lesions, with a 61.6% ( p ⫽ 0.021)
over, an additive predictive effect of age and gadolinium relative reduction in total T2 lesion volume accumulation
lesions at baseline was found; for patients aged ⬍51 years from baseline to week 96, compared with 50.7% ( p ⫽
with baseline gadolinium lesions treated with placebo, the 0.163) for patients aged ⱖ51 years with baseline gadolin-
risk of having CDP was 3 times higher than for patients ium lesions, ⫺13.1% ( p ⫽ 0.261) for patients aged ⬍51
treated with rituximab (HR ⫽ 0.33; p ⫽ 0.009). In ad- years with no baseline gadolinium lesions, and 34.8%

464 Annals of Neurology Vol 66 No 4 October 2009


Table 2. Patient Disposition, Reasons for Discontinuation, and Safety Follow-up in Intent-to-Treat Patients
Variable Placebo, Rituximab, Total,
nⴝ147, nⴝ292, Nⴝ439,
No. (%) No. (%) No. (%)

Patients randomized 147 (100) 292 (100) 439 (100)


Completed 24 weeks 136 (92.5) 269 (92.1) 405 (92.3)
Completed 48 weeks 130 (88.4) 256 (87.7) 386 (87.9)
Completed 72 weeks 125 (85.0) 246 (84.2) 371 (84.5)
Completed 96 weeks 124 (84.4) 241 (82.5) 365 (83.1)
(completion of treatment
period)
Discontinued treatment period 23 (15.6) 51 (17.5) 74 (16.9)
Reasons for discontinuation of treatment period
Adverse events 0 8 (15.7) 8 (10.8)
a
Death 1 (4.3) 0 1 (1.4)a
Lost to follow-up 2 (8.7) 2 (3.9) 4 (5.4)
Patient’s decision 11 (47.8) 27 (52.9) 38 (51.4)
Physician’s decision 4 (17.4) 7 (13.7) 11 (14.9)
Initiation of excluded 0 1 (2.0) 1 (1.4)
therapy
Subject noncompliance 0 1(2.0) 1(1,4)
Pregnancy 0 1 (2.0) 1 (1.4)
Disease progression 5 (21.7) 4 (7.8) 9 (12.2)
Safety follow-up period
Did not enter 5 (4.0) 5 (2.1) 10 (2.7)
Entered but did not 3 (2.4) 12 (5.0) 15 (4.1)
complete
Completed (week 122) 116 (93.5) 224 (92.9) 340 (93.2)
*This death was due to cardiopulmonary failure.

( p ⫽ 0.022) for patients aged ⱖ51 with no baseline gad- ment period early, 40% recovered peripheral B cells af-
olinium lesions. ter 48 weeks from their last dose. Median CD3 T-cell
Subgroup analyses indicated that patients with a short counts were not appreciably altered by rituximab.
disease duration (ⱕ3 years) receiving rituximab tended to At all times during the study, IgG and IgA levels
have a longer time to CDP compared with the patients were below LLN range in ⬍5% of patients in either
receiving placebo. However, a treatment effect on time to treatment arm. IgM levels were below LLN at all times
CDP was seen with younger patients (⬍51 years) despite during the trial in 31.7% of patients receiving ritux-
a relatively longer disease duration (ⱖ7.7 years), and no imab and 5.9% of patients receiving placebo (Table 4).
effect on CDP was seen in older patients (⬎51 years) There was no evidence of increased incidence of infec-
with relative shorter disease duration (⬍7.7 years). tion or other adverse events in patients with immuno-
globulin levels below LLN.
Pharmacodynamics and Immunogenicity During the treatment or safety follow-up, 20 of 286
Treatment with rituximab was associated with a rapid (7.0%) patients receiving rituximab and 9 of 143
and near-complete depletion (⬎95%) of CD19⫹ (sur- (6.3%) patients receiving placebo were positive for hu-
rogate marker for CD20) peripheral B cells starting at man antichimeric antibodies (HACA). Among them,
week 2 through week 96 of treatment. B-cell counts 18 (7.5%) patients receiving rituximab and 4 (3.4%)
above the lower limit of normal (LLN, 80 cells/␮l) patients receiving placebo were HACA-positive at ap-
were used to measure recovery. At week 122, 35% of proximately 1 year after last infusion (see Table 4).
rituximab-treated patients had recovered peripheral There was no apparent association between HACA
B-cell counts. Of patients who discontinued the treat- positivity and the type of adverse events or efficacy re-

Hawker et al: Rituximab in PPMS 465


Table 3. Efficacy Analysis for Primary, Secondary, and Exploratory Endpoints
Endpoint Placebo, Rituximab, p
nⴝ147 nⴝ292

Primary endpoint: time to CDP


Proportion of patients with CDP (K-M estimates), %a 0.1442b
At week 48 19.3 20.2
At week 96 38.5 30.2
Secondary endpoints: T2 lesion and brain volume
T2 volume change from baseline to week 96
(LOCF)
Mean (SD) 2,205 (4306) 1,507 (3739) ⬍0.001c
Median 809.5 302.0
Brain volume change from baseline to week 96 0.62c
(LOCF)
Mean (SD) ⫺9.9 (37.0) ⫺10.8 (40.3)
Median ⫺14.0 ⫺13.1
Key exploratory endpoints
Proportion of patients with CDP (24-mo 30.4 27.3 0.59b
confirmation) at week 96, %
EDSS change from baseline to week 96 (LOCF) 0.34c
Mean (SD) 0.45 (1.0) 0.33 (1.0)
Median 0 0
Median MSFC change from baseline (LOCF)
To week 48 ⫺0.05 ⫺0.00 0.057c
To week 96 ⫺0.04 ⫺0.05 0.846c
To week 122 ⫺0.10 ⫺0.06 0.089c
Median MSFCS (timed 25-foot walk) change from
baseline (LOCF)
To week 48 ⫺0.05 ⫺0.02 0.037c
To week 96 ⫺0.11 ⫺0.07 0.076c
To week 122 ⫺0.14 ⫺0.08 0.015c
a
Stratified hazard ratio (relative to placebo) with 95% confidence interval: 0.77 (0.55–1.09). bLog-rank test stratifying baseline EDSS
and prior MS treatment. cFriedman ranked analysis of variance model adjusting baseline EDSS and prior MS treatment. CDP ⫽
confirmed disease progression; K-M ⫽ Kaplan-Meier; LOCF ⫽ last observation carried forward; SD ⫽ standard deviation; EDSS ⫽
Expanded Disability Status Scale; MSFCS ⫽ Multiple Sclerosis Functional Composite score; MS ⫽ multiple sclerosis.

sponse. The biologic relevance of HACA has yet to be Infusion-related AEs were more common with ritux-
determined. imab, which were primarily mild to moderate in sever-
ity, decreasing with successive infusions. At week 74,
Safety infusion-related reactions were lower in rituximab
The incidence of adverse events (AEs) was comparable (4.9%) versus placebo (7.2%) patients. Grade 3
between groups (see Table 4). Serious AEs were pro- infusion-associated events were reported in 17 patients.
portionally higher in rituximab patients, attributable to No grade 4 infusion-associated events were reported.
patients who experienced multiple serious AEs. Using Infections reported in ⬎10% of either group in-
the Common Terminology Criteria for Adverse Events cluded upper respiratory infections, urinary tract infec-
grading system, 58.6% of rituximab-treated patients tions, and nasopharyngitis. Of 13 rituximab patients
experienced grade 1-2 (mild-moderate) AEs. Of 292 who reported infection-associated serious AEs, 9 (69%)
rituximab-treated patients, 8.6% reported severe (grade were ⱖ55 years of age.
3) or disabling (grade 4) drug-related AEs. Three deaths occurred. One patient with a history of

466 Annals of Neurology Vol 66 No 4 October 2009


A
70 All Intent-to-Treat Patients (N=439)
60 Placebo
Proportion of Patients

Rituximab
50
P=0.1442
40

30

20

10

0
0 12 24 36 48 60 72 84 96 108
Time to Confirmed Disease Progression (weeks)

B
70 Age ≥51 Placebo 70 Age ≥51
Gd Lesion=0 Rituximab Gd Lesion ≥1
60 N=187 60 N=37
HR: 1.27 HR: 0.52
Proportion of Patients

(95% CI: 0.71–2.27) (95% CI: 0.18–1.52)


50 P=0.4256 50 P=0.2243

40 40

30 30

20 20

10 10

0 0
0 12 24 36 48 60 72 84 96 108 0 12 24 36 48 60 72 84 96 108
Time to Confirmed Disease Progression (weeks) Time to Confirmed Disease Progression (weeks)

70 70
Age <51 Age <51
60
Gd Lesion=0 Gd Lesion ≥1
N=143 60 N=72
Proportion of Patients

HR: 0.63 HR: 0.33


50 (95% CI: 0.34–1.18) 50 (95% CI: 0.14–0.79)
P=0.1427 P=0.0088
40 40

30 30

20 20

10 10

0 0
0 12 24 36 48 60 72 84 96 108 0 12 24 36 48 60 72 84 96 108
Time to Confirmed Disease Progression (weeks) Time to Confirmed Disease Progression (weeks)

Fig 3. Efficacy endpoint. (A) Time to confirmed disease progression, all patients; (B) time to confirmed disease progression by age
and baseline gadolinium-enhancing lesions. HR ⫽ hazard ratio; CI ⫽ confidence interval.

brainstem lesions and aspiration received 2 infusions of contracted pneumonia and died after withdrawing
rituximab and withdrew early from the study after con- from the trial.
tracting pneumonia; he was subsequently hospitalized
multiple times for recurrent aspiration and died ap- Discussion
proximately 10 weeks after the initial event. Two pa- PPMS is thought to incorporate elements of inflamma-
tients receiving placebo died; 1 suffered from cardio- tion and neurodegeneration; however, currently ap-
pulmonary failure during the study, and the other proved first-line MS therapies have not been shown to

Hawker et al: Rituximab in PPMS 467


Table 4. AEs and Immunogenicity in the Safety Population
Events Placebo, nⴝ147, Rituximab, nⴝ292,
No. (%) No. (%)

Total AEs 147 (100) 289 (99.0)


AEs leading to discontinuation of study drug 1 (⬍1.0) 9 (3.1)
a
AE grades
Grade 1 20 (13.6) 44 (15.1)
Grade 2 71 (48.3) 127 (43.5)
Grade 3 51 (34.7) 105 (36.0)
Grade 4 3 (2.0) 12 (4.1)
b
Grade 5 2 (1.4) 1 (⬍1.0)c
Serious AEs 20 (13.6) 48 (16.4)
Infusion-associated events, % patientsd
1st Infusion, week 0 23.1 67.1
2nd Infusion, week 2 15.1 22.6
3rd Infusion, week 24 9.6 20.3
4th Infusion, week 26 8.9 10.9
5th Infusion, week 48 6.2 13.7
6th Infusion, week 50 6.3 7.1
7th Infusion, week 72 5.6 7.8
8th Infusion, week 74 7.2 4.9
Infusion-associated AEs ⱖ5% in either group
Nausea 8 (5.4) 31 (10.6)
Fatigue 6 (4.1) 33 (11.3)
Chills 4 (2.7) 32 (11.0)
Pyrexia 2 (1.4) 28 (9.6)
Headache 4 (2.7) 37 (12.7)
Dizziness 4 (2.7) 22 (7.5)
Throat irritation 0 21 (7.2)
Pharyngolaryngeal pain 0 16 (5.5)
Pruritus 1 (⬍1.0) 54 (18.5)
Rash 3 (2.0) 22 (7.5)
Flushing 6 (4.1) 37 (12.7)
Hypotension 12 (8.2) 27 (9.2)
Infection-associated events
All infection-associated AEs and SAEs 96 (65.3) 199 (68.2)
Infection-associated SAEs 1(⬍1.0) 13 (4.5)e
Specific infection-associated SAEs
Pneumonia 1 (⬍1) 4 (1.4)
Bronchitis 0 1 (⬍1)

substantially alter disease course. Although the primary neurodegeneration in MS, insights that may also assist
endpoint was not met in this trial, our findings suggest in our understanding of other autoimmune disorders.
key insights into the interplay of inflammation and Despite the lack of statistical significance between

468 Annals of Neurology Vol 66 No 4 October 2009


Table 4. Continued
Events Placebo, nⴝ147, Rituximab, nⴝ292,
No. (%) No. (%)

Urinary tract infection 0 2 (⬍1)


Pyelonephritis 0 1 (⬍1)
Cellulitis 0 2 (⬍1)
Diverticulitis 0 1 (⬍1)
Meningitis 0 1 (⬍1)
Escherichia urinary tract infection 0 1 (⬍1)
Eschericia bacteraemia 0 1 (⬍1)
Influenza 0 1 (⬍1)
f
Patients positive for HACA during treatment or follow-up period 9/143 (6.3) 20/286 (7.0%)
Ig below the lower limit of laboratory normal
IgG 7/142 (4.9) 13/274 (4.7)
IgM 8/135 (5.9) 83/262 (31.7)
IgA 1/145 (⬍1.0) 3/207 (1.0)
a
Common Terminology Criteria for Adverse Events classification system. bCardiopulmonary failure at week 74 and pneumonia at week
2. cPneumonia at week 2. dAny AE occurring during or within 24 hours of rituximab infusion. eMultiple occurrences of the same event
for a subject were counted only once. fHACA data collected on 286 patients during treatment period or safety follow-up. AE ⫽ adverse
event; SAE ⫽ adverse event; HACA ⫽ human antichimeric antibodies; Ig ⫽ immunoglobulin.

treatment groups in the overall PPMS population, MRI in OLYMPUS) in the differing gender outcome,
there was a trend in treatment-related delay, as evi- it seems that the rate of progression may be a crucial
denced by confirmed EDSS progression and in the factor in predicting outcomes of therapy over a short
timed 25-foot walk test, a key exploratory endpoint. In time span.
the planned subgroup analyses of both the placebo and These findings provide insight that may change our
rituximab groups, there was a statistically significant concepts of the pathogenesis and treatment of PPMS
treatment effect by both these measures in younger pa- but also of progression in all subtypes of MS. Our cur-
tients (aged ⬍51 years) and particularly in those with rent thinking is that a significant proportion of neuro-
inflammation based on gadolinium-enhancing lesions logical disease progression in PPMS is driven by slow
on MRI. This treatment effect was independent of degeneration of axons after their immune-mediated de-
disease duration in younger patients; however, treat- myelination.20 However, the finding that even a small
ment effects were primarily driven by faster progres- amount of inflammation, as evidenced by gadolinium-
sion rates in the younger cohort (and particularly in enhancing lesions, influenced the rate of progression
those with gadolinium-enhancing lesions) than the and the response to treatment in this trial argues for a
progression rates of older patients. gadolinium scans reversible component of progression as well. This ob-
were done at baseline and week 6 (for safety) only, so servation is supported in a study in which evidence of
the effect on gadolinium lesions of rituximab was not even mild inflammation evident on MRI in PPMS was
assessed. associated with greater disability.21 PPMS patients
In contrast to the PROMiSe trial, in which a trend demonstrate greater MRI enhancement than previously
for a treatment effect was noted in males, gender did believed when scanned earlier in the course of their dis-
not appear to play a role in our trial. The trend toward ease.21–23 More sensitive, quantitative MRI imaging
a treatment effect in PROMiSe was postulated to be techniques and analyses may be helpful to define pa-
driven by the more rapid progression rates in male pa- tient populations at risk for rapid disease progression.
tients.4 Although the results from the 2 trials are at These findings, coupled with our data, suggest sev-
odds in terms of gender, the treatment effect in our eral conclusions. First, early treatment of PPMS, aimed
trial was similarly driven by a more rapidly progressing at decreasing disease progression, may be beneficial, be-
cohort, albeit a cohort characterized by age and gado- fore the advent of irreversible neuronal loss. This has
linium positivity. Although the characteristics of the been well documented in RRMS trials, in which treat-
patients we recruited may have played a role (positive ment at the time of diagnosis was shown to reduce the
CSF and higher prevalence of gadolinium lesions on risk of progressive disability.24,25

Hawker et al: Rituximab in PPMS 469


Second, in our study, the slowing of disability was O’Connor, Hillel Panitch, Mary Ann Picone, Kottil Ram-
driven by an effect on ambulation, as evidenced by the mohan, Anthony T. Reder, Peter Riskind, Syed W. Rizvi,
effect on the timed 25-foot walk. Gadolinium- Howard Rossman, Steven R. Schwid, Stuart Shafer, Virginia
enhancing lesions, reflecting classic focal, macroscopic Simnad, Michael Stein, William H. Stuart, Olaf Stuve, Ben
inflammation, are relatively infrequent in the spinal Thrower, Stephen E. Thurston, Michael (Mihai) Vertino,
cord.21 Thus, one may postulate that other compo- Leslie P. Weiner, Dean Wingerchuk, and Michael M. C.
nents of the immune system, such as dendritic cells as Yeung.
well as B cells, likely play a role in perpetuating the
disease.21,26,27
Third, the treatment effect in younger patients, par- References
ticularly those with evidence of acute blood-brain bar- 1. Hauser SL, Oksenberg JR. The neurobiology of multiple
sclerosis: genes, inflammation, and neurodegeneration. Neuron
rier breakdown on neuroimaging, irrespective of disease 2006;52:61–76.
duration, argues for a change in disease neurobiology 2. Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker
over time. This influence of age as a risk factor for an BG. Multiple sclerosis. N Engl J Med 2000;343:938 –952.
increased relapse rate has also been recently reported 3. Miller DH, Leary SM. Primary-progressive multiple sclerosis.
for RRMS.28 Several factors may be implicated, includ- Lancet Neurol 2007;6:903–912.
ing alteration in the immune system with aging (im- 4. Wolinsky JS, Narayana PA, O’Connor P, et al. Glatiramer ac-
etate in primary progressive multiple sclerosis: results of a mul-
munosenescence29 –33). Thus, identifying patients ame-
tinational, multicenter, double-blind, placebo-controlled trial.
nable to treatment, with a greater risk of progression Ann Neurol 2007;61:14 –24.
and thus an acceptable risk-to-benefit ratio, has impor- 5. Leary SM, Miller DH, Stevenson VL, et al. Interferon beta-1a
tant implications for treating MS as well as other au- in primary progressive MS: an exploratory, randomized, con-
toimmune diseases in which a similar age-related phe- trolled trial. Neurology 2003;60:44 –51.
nomenon has been reported.34 6. Montalban X. Overview of European pilot study of interferon
In conclusion, although our study was negative, cou- beta-Ib in primary progressive multiple sclerosis. Mult Scler
2004;10(suppl 1):S62; discussion 62– 64.
pled with our recent reports of selective B-cell deple-
7. Owens GP, Bennett JL, Gilden DH, Burgoon MP. The B cell
tion in RRMS,35,36 our results provide us with new response in multiple sclerosis. Neurol Res 2006;28:236 –244.
insights into PPMS, and demonstrate the need for ex- 8. Andersson L, Bolling M, Wirestam R, et al. Combined diffu-
ploring the potential of pharmacological depletion of B sion weighting and CSF suppression in functional MRI. NMR
cells in MS patients with or without relapses. Our aim Biomed 2002;15:235–240.
is to reduce the accumulation of disability early in the 9. Link H, Huang Y-M. Oligoclonal bands in multiple sclerosis
progressive phase of the disease, when a targeted anti- cerebrospinal fluid: an update on methodology and clinical use-
fulness. J Neuroimmunol 2007;180:7–28.
inflammatory intervention may still have the potential
10. Siden A. Isoelectric focusing and crossed immunoelectrofocusing
to retard downstream neurodegenerative processes. of CSF immunoglobulins in MS. J Neurol 1979;221:39 –51.
11. Genain CP, Cannella B, Hauser SL, Raine CS. Identification of
autoantibodies associated with myelin damage in multiple scle-
This study was supported by Clinicaltrials.gov under “A Study to rosis. Nat Med 1999;5:170 –175.
Evaluate the Safety and Efficacy of Rituximab in Adults With Pri- 12. O’Connor KC, Appel H, Bregoli L, et al. Antibodies from in-
mary Progressive Multiple Sclerosis” (grant NCT00087529), by Ge- flamed central nervous system tissue recognize myelin oligoden-
nentech, Inc., and by Biogen Idec, Inc. drocyte glycoprotein. J Immunol 2005;175:1974 –1982.
13. O’Connor KC, Chitnis T, Griffin DE, et al. Myelin basic
protein-reactive autoantibodies in the serum and cerebrospinal
We thank Genentech, Inc. for assistance in data anal-
fluid of multiple sclerosis patients are characterized by low-
ysis and preparation of the manuscript. affinity interactions. J Neuroimmunol 2003;136:140 –148.
14. Kanter JL, Narayana S, Ho PP, et al. Lipid microarrays identify
Appendix key mediators of autoimmune brain inflammation. Nat Med
We thank each of the OLYMPUS investigators, including 2006;12:138 –143.
15. Pender MP. The pathogenesis of primary progressive multiple
Drs. Virender Bhan, Michael P. Biber, Staley A. Brod, Peter
sclerosis: antibody-mediated attack and no repair? J Clin Neu-
Calabresi, Denise Campagnolo, Mark Cascione, Nelson
rosci 2004;11:689 – 692.
(Kanter) Cooke, Joanna A. Cooper, John Corboy, Bruce
16. Reff ME, Carner K, Chambers KS, et al. Depletion of B cells in
Cree, Anne Cross, Pierre Duquette, Stanton Elias, Corey vivo by a chimeric mouse human monoclonal antibody to
Ford, Mark S. Freedman, Mitchell S. Freedman, Suzanne K. CD20. Blood 1994;83:435– 445.
Gazda, Francois Grand’Maison, Michael (Lava) Gruenthal, 17. Kurtzke JF. Rating neurologic impairment in multiple sclerosis:
Stuart Hoffman, William Honeycutt, John Huddlestone, an expanded disability status scale (EDSS). Neurology 1983;33:
Bruce Hughes, George J. Hutton, Daniel Jacobs, Francois 1444 –1452.
H. Jacques, Lloyd H. Kasper, Lorne Kastrukoff, Michael D. 18. McDonald WI, Compston A, Edan G, et al. Recommended
Kaufman, Omar Khan, Bhupendra O. Khatri, Mariko Kita, diagnostic criteria for multiple sclerosis: guidelines from the In-
Yves La Pierre, Sharon Lynch, Clyde Markowitz, Michele ternational Panel on the diagnosis of multiple sclerosis. Ann
Mass, David Mattson, Aaron Miller, Harold Moses, Paul Neurol 2001;50:121–127.

470 Annals of Neurology Vol 66 No 4 October 2009


19. National Institutes of Health. Common Terminology Criteria for 27. Moore W. Evidence for Secondary Neurogeneration in MS.
Adverse Events v3.0 online instructions and guidelines. In: Can- Montreal, Canada: ECTRIMS/ACTRIMS; 2008.
cer Therapy Evaluation Program DoCTaDD. Bethesda, MD: 28. Mowry EM, Pesic M, Grimes B, et al. Clinical predictors of
National Cancer Institute, National Institutes of Health; 2006. early second event in patients with clinically isolated syndrome.
https://webapps.ctep.nci.nih.gov/webobjs/ctc/webhelp/welcome_ J Neurol 2009;256:1061–1066.
to_ctcae.htm 29. Aw D, Silva AB, Maddick M, et al. Architectural changes in the
20. Dutta R, Trapp BD. Pathogenesis of axonal and neuronal dam- thymus of aging mice. Aging Cell 2008;7:158 –167.
age in multiple sclerosis. Neurology 2007;68:S22–S31; discus- 30. Chen J, Buchanan JB, Sparkman NL, et al. Neuroinflammation
sion S43–S54. and disruption in working memory in aged mice after acute
21. Ingle GT, Sastre-Garriga J, Miller DH, Thompson AJ. Is in- stimulation of the peripheral innate immune system. Brain Be-
flammation important in early PPMS? A longitudinal MRI hav Immun 2008;22:301–311.
study. J Neurol Neurosurg Psychiatry 2005;76:1255–1258. 31. Godbout JP, Chen J, Abraham J, et al. Exaggerated neuroin-
22. Khaleeli Z, Ciccarelli O, Manfredonia F, et al. Predicting pro-
flammation and sickness behavior in aged mice following acti-
gression in primary progressive multiple sclerosis: a 10-year
vation of the peripheral innate immune system. FASEB J 2005;
multicenter study. Ann Neurol 2008;63:790 –793.
19:1329 –1331.
23. Rovaris M, Judica E, Sastre-Garriga J, et al. Large-scale, multicen-
32. Hasler P, Zouali M. Immune receptor signaling, aging, and au-
tre, quantitative MRI study of brain and cord damage in primary
progressive multiple sclerosis. Mult Scler 2008;14:455– 464. toimmunity. Cell Immunol 2005;233:102–108.
24. Kappos L, Freedman MS, Polman CH, et al. Effect of early 33. Sawada M, Sawada H, Nagatsu T. Effects of aging on neuro-
versus delayed interferon beta-1b treatment on disability after a protective and neurotoxic properties of microglia in neurode-
first clinical event suggestive of multiple sclerosis: a 3-year generative diseases. Neurodegener Dis 2008;5:254 –256.
follow-up analysis of the BENEFIT study. Lancet 2007;370: 34. Prelog M. Aging of the immune system: a risk factor for auto-
389 –397. immunity? Autoimmun Rev 2006;5:136 –139.
25. Kinkel RP, Kollman C, O’Connor P, et al. IM interferon 35. Bar-Or A, Calabresi P, Arnold D, et al. Safety and efficacy of
beta-1a delays definite multiple sclerosis 5 years after a first de- rituximab in adults with relapsing remitting multiple sclerosis: a
myelinating event. Neurology 2006;66:678 – 684. phase I open-label multicenter trial. Ann Neurol 2008;63:
26. Lyons JA, San M, Happ MP, Cross AH. B cells are critical to 395– 400.
induction of experimental allergic encephalomyelitis by protein 36. Hauser SL, Waubant E, Arnold DL, et al. B-cell depletion with
but not by a short encephalitogenic peptide. Eur J Immunol rituximab in relapsing-remitting multiple sclerosis. N Engl
1999;29:3432–3439. J Med 2008;358:676 – 688.

Hawker et al: Rituximab in PPMS 471

You might also like