Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 9

Received: 10 October 2018  | Revised: 8 January 2019  | Accepted: 22 January 2019

DOI: 10.1111/1756-185X.13511

ORIGINALARTICLE

Real‐world effectiveness and safety of rituximab in the


treatment of rheumatoid arthritis: A single‐center
experience in Taiwan
1,2 1,2,3 1,2
Kai‐Chun Wang  | Hsien‐Tzung Liao  | Wei‐Sheng Chen | Chien‐Chih
1,2,4 1,2 1,2 1,2
Lai  | Chung‐Tei Chou | Ming‐Han Chen  | Chang‐Youh Tsai

1
Division of Allergy, Immunology and
Rheumatology, Department of Medicine,
Abstract
Taipei Veterans General Hospital, Taipei,
Aim: To assess the real‐world effectiveness and safety of rituximab (RTX) at 24
Taiwan
2
Faculty of Medicine, National Yang‐Ming months in patients with established rheumatoid arthritis (RA) and to identify pre‐
University School of Medicine, Taipei, dictors of low disease activity/remission and a good European League Against
Taiwan
Rheumatism (EULAR) response.
3
Division of Allergy, Immunology and
Rheumatology, Department of Internal Methods: Seventy RTX‐treated RA patients were enrolled. Predictors for low
Medicine, School of Medicine, College of disease activity/remission and a good EULAR response at 24 months were identified
Medicine, Taipei Medical University, Taipei,
Taiwan by mul‐ tivariate analyses.
4
Institute of Clinical Medicine, National Results: At 24 months, the mean Disease Activity Score of 28 joints—erythrocyte
Yang‐Ming University School of Medicine,
Taipei, Taiwan sedimentation rate (DAS28‐ESR) decreased from 6.88 ± 0.85 at baseline to 3.47 ±
0.85. Twenty‐nine patients (41.4%) reached low disease activity/remission, while all
Correspondence
Ming‐Han Chen and Chang‐Youh Tsai,
patients had a moderate/good EULAR response. After adjustment by multi‐ variate
Division of Allergy, Immunology, and analyses, we found concomitant methotrexate at a dosage >10 mg/week (odds ratio
Rheumatology, Department of Medicine,
Taipei Veterans General Hospital, Taipei
[OR] 5.17; 95% CI 1.34‐19.93; P = 0.017) predicted low disease activity/ remission,
City, Taiwan. and baseline DAS28 ≤6.5 (OR 4.97; 95% CI 1.22‐20.30; P = 0.026) pre‐ dicted
Emails: mhchen6@vghtpe.gov.tw; cytsai@
good EULAR response at 24 months. The most common adverse events were
vghtpe.gov.tw
infusion‐related (5.7%), and there was no incidence of malignancy or mortality
during the treatment.
Conclusions: RTX was effective and safe in real‐life management of RA patients
with high disease activity. Patients taking concomitant methotrexate and with lower
base‐ line DAS28‐ESR were more likely to benefit from RTX.

KEYWORDS
CD20, DAS‐28, rheumatoid arthritis, rituximab

1 | INTRODUCTION strategy for RA, among which methotrexate (MTX) is the cornerstone
unless contraindicated or not tolerated. Short‐term glucocorticoids would
Rheumatoid arthritis (RA), a systemic autoimmune disease targeting be bridging therapy when switching csDMARDs. Biologic DMARDs
synovial tissues, causes longstanding inflammation and catastrophic (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) can be added
1
sequelae in multiple joints. The resultant progressive deformity and to csDMARDs when remission cannot be achieved.
physical disability impair quality of life. Conventional synthetic disease‐ A presence of rheumatoid factor (RF) and/or anti‐citrullinated
modifying antirheumatic drugs (csDMARDs) are the primary treatment protein antibody (ACPA) could be detected years before onset of

Int J Rheum Dis. 2019;1–9. wileyonlinelibrary.com/journal/apl © 2019 Asia Pacific League of Associations for   |  1
Rheumatology and John Wiley & Sons Australia, Ltd
2  | WANG et al.

2
clinical symptoms of RA, and their titers have been shown to pre‐ Rheumatology Division in Taipei Veterans General Hospital, a ter‐ tiary
dict functional and radiological outcomes. In addition to producing referral center in Taiwan, from October 2008 to January 2016. All
these antibodies, B cells are also highly efficient in antigen presen‐ patients fulfilled the ACR 1987 revised criteria or ACR/EULAR 2010
11,12
tation and can activate auto‐reactive T cells. Rituximab (RTX), a criteria for RA classification, and had previously failed standard
bD‐ MARD of chimeric murine/human monoclonal antibody csDMARDs. There were no disease activity or comorbid‐ ity exclusion
targeting CD20‐positive B cells, has thus been proven effective in criteria. All patients received RTX as accredited by this country's health
randomized clinical trials (RCTs) in patients with active established administration in a 6‐month fixed repetitive proto‐ cols (two 1000 mg
RA resistant to csDMARDs and anti‐tumor necrosis factor α (anti‐ intravenous doses, 14 days apart) as the 2nd line bDMARD with the
3-5
TNFα) bD‐ MARDs. A single course of RTX with two 1000 mg reasons for switching to RTX being recorded. This study was approved
infusions given 2 weeks apart significantly ameliorates disease by the institutional ethics committee of Taipei Veterans General Hospital
burden. As shown previously, at week 24, 51% of patients had an (IRB‐TPEVGH No. 2017‐05‐002AC).
American College of Rheumatology 20% improvement (ACR20)
response, 50% of pa‐ tients had a European League Against 2.2 | Clinical evaluations and laboratory tests
Rheumatism (EULAR) mod‐ erate response and 15% of patients
5
had a EULAR good response. However, the populations in RCTs Clinical parameters including 28 tender joint count (TJC), 28 swol‐
may not accurately reflect the characteristics of the real‐world len joint count (SJC), and DAS28 using erythrocyte sedimentation
patients as strict inclusion and ex‐ clusion criteria were applied to rate (DAS28‐ESR) were assessed at baseline and at 6, 12, 18 and
increase the internal validity but not external generalizability. A 24 months after initiating RTX. The numbers and types of the previ‐
systemic review has demonstrated that characteristics of patients ous anti‐TNFα agents, and numbers and dosages of the
from real‐world observational studies are substantially different from concomitant DMARDs and glucocorticoids were also documented.
6
those enrolled in RCTs, leading to an efficacy‐effectiveness gap. Laboratory tests including ESR, RF and ACPA were periodically
As a result, the real‐world evidence is vital because it is not only assessed. RF was examined using nephelometry (Toshiba TBA™‐
generated from investigations of hetero‐ geneous populations with c16000 device, Toshiba Medical Systems Corporation, Tochigi‐ken,
diverse behaviors in routine practices, but also can validate the Japan; positive if >30 IU/mL) and ACPA was measured using
results in RCTs. Nevertheless, most of the RCTs and observational enzyme‐linked immuno‐ sorbent assay (Phadia EliA™ kit, Phadia
studies showing outcomes of RTX treatments have been conducted AB, Uppsala, Sweden; posi‐ tive if >10 U/mL).
in Western countries and very few studies have examined the
7,8
effectiveness and safety of RTX for RA in ethnic Asians. 2.3 | Definition of clinical response and remission
Several authors have also attempted to identify specific pre‐
dictive factors for RTX treatment response in RA patients to help The clinical remission status was defined by DAS28‐ESR ≤2.6, low
clinical rheumatologists take a more personalized and precise thera‐ disease activity by DAS28‐ESR >2.6 but ≤3.2, moderate disease
peutic approach. The presence of RF and/or ACPA, number of activity by DAS28‐ESR >3.2 but ≤5.1 and high disease activity by
previ‐ ous bDMARDs, baseline Disease Activity Score of 28 joints DAS28‐ESR >5.1. Responses to RTX therapy were assessed using
13
(DAS28), serum immunoglobulin level and biomarkers including type the EULAR criteria. Patients were classified as responders (good
I in‐ terferon and myeloid‐related protein (MRP)8/14 levels, have or moderate) or non‐responders according to DAS28‐ESR levels
9 and their improvement from the baseline. The clinical remission
been shown to affect treatment responses in previous studies. On
the other hand, a low body mass index has not yet been proven to status and responses were evaluated at 6, 12, 18 and 24 months
be a reliable index to predict RTX effectiveness although it has been after the 1st dose of RTX.
10
used so in anti‐TNFα therapy.
Due to lack of studies addressing the real‐life outcomes of RA 2.4 | Safety issues
patients undergoing RTX therapy in Asia, we carried out the pres‐
ent investigation to characterize the real‐world use, effectiveness, Adverse events including infusion reactions, infections, serious in‐
and safety of RTX in patients with RA. Furthermore, we identified fections necessitating admission, malignancies, and mortalities were
possible predictors for achieving low disease activity/remission or a recorded.
EULAR good response in these RTX‐treated RA patients at 24
months of treatment.
2.5 | Statistical analysis

The baseline characteristics were compared with the Chi‐squared


2 | PATIENTS AND METHODS
test or Fisher's exact test for categorical variables and compared
with Mann‐Whitney U tests for the quantitative variables. DAS28‐
2.1 | Patients
ESR, ESR, TJC and SJC were compared with corresponding values
In the present retrospective study, we enrolled 70 adult patients with at baseline using the paired sample t test. Univariate logistic regres‐
RA who underwent RTX treatment at the Allergy, Immunology and sion was performed to identify predictive factors for the low disease
WANG et al.      |  3
TA B LE 1 Demography and characteristics of RA patients (CI). A P value <0.05 was considered significant. Data were
undergoing rituximab therapy analyzed with SPSS software, version 22 (IBM, Armonk, NY, USA).

RA patients
Characteristics (n = 70) 3 | RESULTS
Female gender, n (%) 67 (95.7)
Age at diagnosis, mean (SD), y 54.1 (10.6) 3.1 | Demographic and clinical characteristics
Age at RTX initiation, mean (SD), y 58.9 (10.6)
A total of 70 patients, including 67 women (95.7%), were en‐ rolled
Disease duration, mean (SD), y 4.9 (1.8)
in this study. The mean age at RTX initiation was 58.9 years.
Baseline TJC, mean (SD), n 16.5 (4.8)
Demographic characteristics of patients are shown in Table 1. At
Baseline SJC, mean (SD), n 9.7 (4.8) baseline, patients had a mean disease duration of 4.9 years and a
Baseline ESR, mean (SD), mm/h 59.1 (33.6) sig‐ nificant disease burden with a mean DAS28‐ESR high at 6.88 ±
Baseline DAS28‐ESR, mean (SD) 6.88(0.85) 0.85, mean SJC of 9.7 ± 4.8, and mean TJC of 16.5 ± 4.8. Four
RF positive, n (%) 63 (90.0) patients (5.7%) overlapped with systemic lupus erythematosus, 20
ACPA positive, n/total (%) 38/69 (55.1) patients (28.6%) had Sjögren's syndrome, and 7 patients (10%) had

Comorbidity, n (%)
interstitial lung disease at baseline.

Systemic lupus erythematosus 4 (5.7) Sixty‐six patients (94.3%) had previously used at least 1 anti‐
TNFα agent. The remaining 4 patients took RTX as the first‐line bD‐
Sjögren's syndrome 20 (28.6)
MARD due to history of malignancy (3 had breast cancer and 1 had
Interstitial lung disease 7 (10.0)
schwannoma). During the 24 months of RTX treatment, 69 patients
Number(s) of previous anti‐TNFα, n (%)
(98.6%) received at least 1 concomitant csDMARDs including MTX,
None 4 (5.7)
leflunomide, sulfasalazine, hydroxychloroquine, and cyclosporine; in
One 59 (84.3)
average, 1.9 types of DMARDs were used. Forty‐nine patients
≥2 7 (10.0) (70.0%) received MTX during the 24 months at a mean dosage of
Item(s) of previous anti‐TNFα (specified), n (%) 10.71 ± 3.85 mg/wk. There were also 26 patients (37.1%) taking
Etanercept 49 (74.2) prednisolone along with RTX at a mean dosage of 7.69 ± 2.60 mg/d.
Adalimumab 10 (15.2)
Etanercept and adalimumab 7 (10.6) 3.2 | Improvement of laboratory and
Concomitant medication in 24 mo
clinical parameters by RTX
csDMARDs, mean (SD), n 1.9 (0.6)
csDMARDs other than MTX, mean (SD), n 1.2 (0.6) After initiation of RTX, we observed a significant improvement of mean
DAS28‐ESR at 6, 12, 18, 24 months as compared with baseline levels
MTX usage, n (%) 49(70.0)
(P < 0.001 for each comparison), and the mean decrement of DAS28‐
MTX dosage, mean (SD), mg/wk 10.71 (3.85)
ESR at 24 months was 3.41 ± 0.93 (Figure 1A). The mean ESR level at
Mean ≤7.5 mg/wk, n/total (%) 25/49 (51.0) 24 months significantly decreased when compared to that at baseline
Mean ≤10 mg/wk, n/total (%) 28/49 (57.1) (19.7 ± 17.3 vs 59.1 ± 33.6; P < 0.001; Figure 1B). Similarly, the mean
Prednisolone usage, n (%) 26 (37.1) SJC and TJC were also significantly lower at 24 months, compared to
those at baseline (P < 0.001, P < 0.001, respectively; Figure 1C,D).
Mean prednisolone dosage, mean (SD), mg/d 7.69 (2.6)
All patients were in high disease activity at baseline. After 24 months,
ACPA, anti‐citrullinated protein antibody; CRP, C‐reactive protein; csD‐
none were in high disease activity: 12 patients (17.1%) achieved
MARDs, conventional synthetic disease‐modifying anti‐rheumatic drugs;
DAS28‐ESR, Disease Activity Score of 28 joints—erythrocyte sedimen‐ remission, 17 patients (24.3%) reached low disease activity, and 41
tation rate; ESR, erythrocyte sedimentation rate; MTX, methotrexate; RF, patients (58.5%) had moderate disease activity (Figure 2A).
rheumatoid factor; RTX, rituximab; SD, standard deviation; SJC, swollen
joint count; TJC, tender joint count; TNF, tumor necrosis factor.
3.3 | Predictors for clinical remission at 24 months
activity/remission and good EULAR responders at 24 months. Variables
with a P value ≤0.15 in the univariate analysis were taken into Comparing the 41 patients (58.5%) with moderate disease ac‐ tivity
multivariate logistic regression models using backward selec‐ tions to to the 29 patients (41.4%) with low disease activity/re‐ mission at 24
estimate odds ratios (ORs) for clinical remission status and EULAR months, those who were in low disease activity/ remission at 24
responses. The multivariate models were also adjusted for age, gender, months had a longer duration of disease (5.3 ± 2.1 vs 4.6 ± 1.4
and known predictive factors for RTX response accord‐ ing to a literature years; P = 0.041), and a significantly greater proportion of them took
review, which included the presence of RF and/or ACPA, baseline concomitant MTX at a mean dosage >10 mg/wk (59.1% vs 29.6%; P
9
DAS28‐ESR levels, and numbers of previous biolog‐ ics. Results are = 0.038; Table S1). There were no significant differences in age at
diagnosis, age at RTX initiation, gender, TJC, SJC, DAS28‐ESR,
expressed as ORs with a 95% confidence interval
4  | WANG et al.

FI G U R E 1 Changes in clinical and laboratory parameters in 70 patients with rheumatoid arthritis treated with rituximab, at baseline, 6, 12,
18, and 24 mo. (A) The Disease Activity Score of 28 joints‐ erythrocyte sedimentation rate (DAS28‐ESR) levels; (B) ESR; (C) swollen joint
count (SJC); (D) tender joint count (TJC). All parameters were significantly lower than the corresponding baseline levels (P < 0.001)

13
presence of RF, presence of ACPA, autoimmune comorbidities, pro‐ (17.1%) and 58 moderate responders (82.9%). The good
portion of no more than 1 previous anti‐TNFα, refractoriness to anti‐ respond‐ ers had a longer disease duration (5.5 ± 1.2 vs 4.8 ± 1.8
TNFα, and concomitant uses of csDMARDs and prednisolone. years; P = 0.043), lower baseline DAS28‐ESR levels (41.58 ± 33.30
We then used univariate logistic regression to find predictors for low vs 62.72 ± 32.75; P = 0.043), and a baseline DAS28‐ESR ≤6.5
disease activity/remission at 24 months of RTX therapy. It was identified (66.7% vs 25.9% of respective cohort; P = 0.014; Table S2). No
that a concomitant MTX use (OR 1.17; 95% CI 1.002‐1.37; P = 0.047), significant differences were observed between good and moderate
especially at a level of more than 10 mg/week (OR 3.43; 95% CI 1.05‐ respond‐ ers regarding the age at diagnosis, age at RTX initiation,
11.22; P = 0.041) was a predictor for low disease activity/ remission gender, TJC, SJC, RF/ACPA seropositivity, autoimmune
(Table 2). The multivariate logistic regression model, which was adjusted comorbidities, concomitant use and adjustment of csDMARDs and
for gender, age, disease duration, DAS28‐ESR level, pres‐ ence of predniso‐ lone, previous anti‐TNFα of no more than 1 and
RF/ACPA, no more than 1 previous anti‐TNFα, and concomitant refractoriness to anti‐TNFα.
prednisolone usage revealed the correlation between concomitant mean We analyzed global DAS28‐ESR as well as categorized DAS28‐
MTX dosage of more than 10 mg/wk and low disease activity/re‐ mission ESR at 6, 6.5 and 7 using univariate logistic regression to identify
at 24 months (OR 5.17; 95% CI 1.34‐19.93; P = 0.017; Table 2). predictors of a good EULAR response and found that global DAS28 ‐
ESR (OR 0.28; 95% CI 0.11‐0.69; P = 0.005) and categorization of

3.4 | Predictors for EULAR response at 24 months DAS28‐ESR ≤6.5 (OR 5.73; 95% CI 1.51‐21.82; P = 0.001) were good
predictors. We then analyzed these data further with a multivari‐ ate
As shown in Figure 2B, all patients were EULAR responders at 24 logistic regression model, which was adjusted by gender, age, RF/ACPA
months after RTX treatment, including 12 good responders seropositivity, and number of previous anti‐TNFα ≤1, and
WANG et al.      |  5

FI G U R E 2 (A) Percentages of clinical remission status according to Disease Activity Score of 28 joints‐ erythrocyte sedimentation rate
(DAS28‐ESR) levels and (B) percentages of responses according to European League Against Rheumatism criteria at months 6, 12, 18 and
24 in 70 patients with rheumatoid arthritis receiving rituximab

found that baseline DAS28‐ESR ≤6.5 remained a statistically signifi‐ IGRA received prophylaxis with isoniazid prior to the administration
cant predictor (OR 4.97; 95% CI 1.22‐20.30; P = 0.026; Table 3). of RTX. No reactivation of tuberculosis occurred. At baseline, nine
patients (12.9%) had chronic hepatitis B as defined by the presence
of hepatitis B surface antigen (HBsAg); no reactivation of hepatitis B
3.5 | Safety outcomes
was noted within 24 months. In addition, no patient in our cohort
None of the enrolled patients discontinued RTX therapy during the developed malignancy or died in the 24‐month follow‐up.
follow‐up. There were 4 events of infusion‐related reactions. One
serious infection event occurred, which was cellulitis complicated
with osteomyelitis caused by methicillin‐sensitive Staphylococcus 4 | DISCUSSION
aureus and coagulase‐negative Staphylococcus. Thirteen patients
underwent interferon‐γ release assay (IGRA) at baseline; among In the present observational investigation from a tertiary refer‐ ral
them, six patients had a positive result and two patients had an center in Taiwan, RTX was effective in the treatment of RA pa‐
indeterminate finding. Three out of the six patients with positive tients with high clinical disease activities regardless of response to
6  | WANG et al.

b
TA B LE 2 Predictors of low disease
Univariate OR Multivariate OR
a
Characteristics (95% CI) P value (95% CI) P value activity/remission at 24 mo

Female gender 1.44 (0.12‐16.62) 0.772 0.97 (0.04‐26.85) 0.973


Age at RTX initiation, y 0.98 (0.94‐1.03) 0.407 0.99 (0.91‐1.07) 0.752
Disease duration, y 1.25 (0.93‐1.68) 0.145 1.40 (0.97‐2.02) 0.076
DAS28‐ESR 0.58 (0.32‐1.06) 0.076 0.54 (0.19‐1.59) 0.265
RF positive 0.49 (0.10‐2.40) 0.381 0.12 (0.01‐1.28) 0.078
ACPA positive 1.47 (0.56‐3.90) 0.437 1.09 (0.26‐4.57) 0.904

Previous anti‐TNFα ≤1 0.94 (0.19‐4.54) 0.936 2.90 (0.20‐41.48) 0.432


Refractory to anti‐TNF‐α 0.57 (0.13‐2.51) 0.455
Concomitant medication in 24 mo
csDMARDs, n 1.25 (0.58‐2.70) 0.566
csDMARDs excluding 0.96 (0.43‐2.18) 0.929
MTX, n
MTX usage 1.63 (0.56‐4.74) 0.37
*
Mean dosage of MTX, mg/wk 1.17 (1.002‐1.37) 0.047
* *
>10 mg/wk, n/total 3.43 (1.05‐11.22) 0.041 5.17 (1.34‐19.93) 0.017
Prednisolone usage 0.39 (0.14‐1.05) 0.063 0.59 (0.13‐2.61) 0.485
Mean dosage of predniso‐ 0.84 (0.62‐1.15) 0.286
lone, mg/d

ACPA, anti‐citrullinated protein antibody; CRP, C‐reactive protein; csDMARDs, conventional syn‐
thetic disease‐modifying anti‐rheumatic drugs; DAS28‐ESR, Disease Activity Score of 28 joints—
erythrocyte sedimentation rate; ESR, erythrocyte sedimentation rate; MTX, methotrexate; RF,
rheumatoid factor; RTX, rituximab; TNF, tumor necrosis factor.
a
There are no patients with high disease activities at 24 mo.
b
Factors including age, gender, RF, ACPA, previous number of anti‐TNFα ≤1 and variables with P <
0.15 in univariate logistic regression were analyzed by multivariate logistic regression with back ‐
ward selection. ESR was excluded as it is a component of DAS28‐ESR.
*
P < 0.05

anti‐TNFα. Patients concomitantly taking MTX at a mean dosage of a good EULAR response, in contrast to the 41.1% who reached low
more than 10 mg/wk were more likely to be in low disease activity or disease activity/remission, which may also cause this inconsistency.
remission at 24 months. For EULAR response at 24 months, patients Our mean dosage of MTX has been lower than those prescribed in
with a baseline DAS28‐ESR ≤6.5 were more likely to be good respond‐ Western countries, partly in that Orientals may have a poorer tol‐
ers than to be moderate responders. These outcomes were robust even erance to MTX than Caucasians. As higher dosages are commonly
after adjustment in the multivariate logistic regression models. not tolerated due to adverse events, the maximum dosage of MTX is
16 -18
The present investigation demonstrated that MTX of more than 10 16 mg/wk in Japan and 20 mg/wk in China. Thus, these
mg/wk predicted lower disease activity at 24 months. These results are findings, suggesting maintaining MTX dosage above 10 mg/wk in
important as no clinical trials have demonstrated thera‐ peutic superiority combina‐ tion with RTX can decrease disease activity more
of RTX monotherapy to combination of RTX with csDMARDs up to now, effectively, are important for Orientals receiving RTX therapy.
and various clinical guidelines have recog‐ nized MTX as a preferred We have also shown that patients with lower baseline DAS28‐
csDMARD, either in itself or in combina‐ tion with other csDMARDs, ESR were more likely to be good responders at 24 months. These
1,14
auxiliary to bDMARDs or tsDMARDs. The chimerically designed RTX re‐ sults were compatible with those reported previously which
has been reported to induce anti‐ drug antibodies in 3%‐4% of RA showed 2019 RA patients from the CERERRA initiative, exhibited a
4
patients, which can adversely af‐ fect its efficacy, pharmacokinetics and lower baseline DAS28‐ESR level as a predictor of a good EULAR
19
safety profiles. Combination with MTX may potentially decrease this response at 6 months. We further stratified our patients with
immunogenicity in a pattern similar to those seen in studies with anti‐ different base‐ line DAS28‐ESR levels (at 6, 6.5, and 7) and
15
TNFα. Although this com‐ bination only predicts low disease confirmed with multivar‐ iate analysis that those with baseline
activity/remission but not a good EULAR response in our study, we DAS28‐ESR ≤6.5 were more likely to be good responders.
believe this divergence reflects the inherent differences of these The presence of RF and ACPA in serum has been reported to pre‐
endpoints, which are composed of different factors. In addition, only 20 -22
dict better response to RTX in previous studies. However, the
17.1% of our patients reached present study failed to show such correlation. The reason for this is
WANG et al.      |  7
TA B LE 3 Predictors of good EULAR a
Univariate OR Multivariate OR
response at 24 mo
Characteristics (95% CI) P value (95% CI) P value

Female gender 0.39 (0.03‐4.72) 0.461 0.25 (0.02‐4.11) 0.333


Age at RTX initiation, y 1.05 (0.98‐1.12) 0.176 1.04 (0.96‐1.12) 0.329
Disease duration, y 1.24 (0.90‐1.72) 0.192
*
Baseline DAS28‐ESR 0.28 (0.11‐0.69) 0.005
* *
DAS28‐ESR ≤6.5 5.73 0.010 4.97 (1.22‐20.30) 0.026
(1.51‐21.82)
RF positive 0.22 (0.04‐1.16) 0.075 0.20 (0.03‐1.21) 0.080
ACPA positive 1.52 (0.40‐5.78) 0.535 1.72 (0.36‐8.17) 0.494

Previous n of anti‐TNFα ≤1 0.47 (0.08‐2.78) 0.406 0.91 (0.07‐12.41) 0.941


Refractory to anti‐TNFα 0.48 (0.08‐2.81) 0.415
Concomitant medication in 24 mo
csDMARDs, n 1.01 (0.37‐2.72) 0.988
csDMARDs excluding 0.44 (0.13‐1.42) 0.168
MTX, n
MTX usage 5.79 0.104 3.72 (0.40‐34.14) 0.246
(0.70‐48.11)
MTX mean dosage, mg/wk 1.08 (0.91‐1.29) 0.390
Prednisolone usage 0.36 (0.09‐1.46) 0.151
Prednisolone mean 0.84 (0.53‐1.35) 0.475
dosage, mg/d

ACPA, anti‐citrullinated protein antibodies; csDMARDs, conventional synthetic disease‐modifying


anti‐rheumatic drugs; DAS28‐ESR, Disease Activity Score of 28 joints—erythrocyte sedimentation
rate; ESR, erythrocyte sedimentation rate; EULAR, European League Against Rheumatism; MTX,
methotrexate; RF, rheumatoid factor; RTX, rituximab; TNF, tumor necrosis factor.
a
Factors including age, gender, RF, ACPA, Number of previous anti‐TNFα ≤1 and variables with P <
0.15 in univariate logistic regression are analyzed by multivariate logistic regression with back‐ ward
selection. Swollen joint count (SJC) and tender joint count (TJC) were excluded as they are
components of DAS28‐ESR.
*
P < 0.05.

unclear but might be related to a relatively small sample size, a differ‐ immunity resulting from long‐term administration of RTX is un‐
ent ethnicity and the baseline comorbidities. Also, the average age of our known and needs further studies. In the present investigation, we
patients (58.9 ± 10.6 years) was higher than those enrolled in the clinical have not found reactivation of latent tuberculosis infection (LTBI)
20
trials. Detection of RF increases as patients become older and are although some of the patients did have positive IGRA and under‐
exposed to accumulating kinds of microbes or environmental factors, went prophylactic therapy with isoniazid. Although RA patients have
24
decreasing its accountability for RA. Further studies with longer a 2.7‐fold increase in the risk of developing TB, our findings are in
observation times, larger patient numbers, as well as prospec‐ tive data agreement with those reported previously in that there were no
collection in a real‐world situation would be necessary to demonstrate 25
increased risks of TB infection after RTX therapy. Since RTX has
their predictive usefulness. Similarly, several studies have suggested no significant effect on release of interferon‐γ in RA patients with
that patients previously treated with no more than 1 anti‐TNFα have a LTBI, it is conceivable that RTX exerts it effect through a signal
better response and are more likely to achieve low disease activity or transduction pathway other than Th1 inhibition or innate immune
19,23 8
remission. In contrast, the present study did not show similar results. suppression.
Nevertheless, we adjusted this factor with a presence of RF and ACPA in On the other hand, although our previous study has shown the
the multivariate logistic regression as they were recognized predictors. risk of reactivation of hepatitis B is significantly increased among
RTX treatment appeared to be generally safe in our study. The RTX‐treated RA patients (adjusted hazards ratio, 16.51; 95% CI
26
most frequent adverse events were infusion‐related, which were 1.82‐149.67; P = 0.01) no reactivation of hepatitis B was docu‐
relieved by lowering infusion rates and the administration of glu‐ mented in the present investigation. This was largely because most
cocorticoids and antihistamines. Only one serious infection event of our patients (43, 61.4%) had anti‐hepatitis B surface antibod‐ ies
happened by the end of the observation and there was no fatality. (anti‐HBs) and a negligible HBV viral load at baseline and pos‐ sibly
Whether the infection was predisposed by interference of acquired also because the follow‐up period was not sufficiently long (24
months only).
8  | WANG et al.

Although our present results are encouraging there are still 3. Edwards JC, Szczepański L, Szechiński J, et al. Efficacy of B‐cell‐
tar‐ geted therapy with rituximab in patients with rheumatoid arthritis. N
limitations in the investigation. These include its retrospective and
Engl J Med. 2004;350:2572‐2581.
observational design, a relatively small sample size, and a possible
4. Emery P, Fleischmann R, Filipowicz‐Sosnowska A, et al. The effi‐
selection bias with a strong female preponderance in the RA cohort cacy and safety of rituximab in patients with active rheumatoid arthritis
from only a single referral center. Nevertheless, to our knowledge, despite methotrexate treatment: results of a phase IIB ran‐ domized,
the present investigation is the 1st study in ethnic Asians, trying to double‐blind, placebo‐controlled, dosage‐ranging trial. Arthritis Rheum.
2006;54:1390‐1400.
explore the real‐world data regarding the effectiveness and safety of
5. Cohen SB, Emery P, Greenwald MW, et al. Rituximab for
RTX in active RA patients who failed anti‐TNFα treatment. In ad‐ rheumatoid arthritis refractory to anti‐tumor necrosis factor therapy:
dition, it also provided detailed clinical information in a substantially results of a multicenter, randomized, double‐blind, placebo‐controlled,
long time of observation, as well as a clear and segregated method‐ phase III trial evaluating primary efficacy and safety at twenty‐four
weeks. Arthritis Rheum. 2006;54:2793‐2806.
ology of evaluation on the effect of RTX in a complicated scenario of
6. Kilcher G, Hummel N, Didden EM, et al. GetReal Work Package 4.
tedious and variable combinations of csDMARDs.
Rheumatoid arthritis patients treated in trial and real world set‐ tings:
comparison of randomized trials with registries. Rheumatology (Oxford).
2018;57:354‐369.
5 | CONCLUSION 7. Tan BE, Lim AL, Kan SL, et al. Real‐world clinical experience of
bio‐ logical disease modifying anti‐rheumatic drugs in Malaysia rheuma‐
toid arthritis patients. Rheumatol Int. 2017;37:1719‐1725.
This real‐world study confirms the effectiveness and safety of RTX 8. Chen YM, Chen HH, Lai KL, et al. The effects of rituximab therapy
in well‐documented active RA patients regardless of their prior anti‐ on released interferon‐γ levels in the QuantiFERON assay among RA
patients with different status of Mycobacterium tuberculosis in‐ fection.
TNFα refractoriness. Our results also suggest patients may benefit
Rheumatology (Oxford). 2013;52:697‐704.
more from RTX if maintaining the concomitant MTX at a dosage of
9. Wijbrandts CA, Tak PP. Prediction of response to targeted treat‐
more than 10 mg/wk. ment in rheumatoid arthritis. Mayo Clin Proc. 2017;92:1129‐1143.
10. Ottaviani S, Gardette A, Roy C, et al. Body mass index and re‐
sponse to rituximab in rheumatoid arthritis. Joint Bone Spine.
AC K N OW L E D G E M E N T S 2015;82:432‐436.
11. Arnett FC, Edworthy SM, Bloch DA, et al. The American
The authors would like to thank Dr. Yi‐Syuan Sun for his valuable Rheumatism Association 1987 revised criteria for the classification of
advice and assistance with the statistical analysis of this study. This rheuma‐ toid arthritis. Arthritis Rheum. 1988;31:315‐324.
study was supported by the grants from the Ministry of Science & 12. Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis
clas‐ sification criteria: an American College of Rheumatology/European
Technology (MOST‐107‐2314‐B‐075 ‐051 ‐MY3) and Taipei
League Against Rheumatism collaborative initiative. Arthritis Rheum.
Veterans General Hospital (V107C‐084), Taiwan. 2010;62:2569‐2581.
13. van Gestel AM, Prevoo ML, van 't Hof MA, , et al. Development and
validation of the European League Against Rheumatism re‐ sponse
CONFLICTS OF INTEREST criteria for rheumatoid arthritis. Comparison with the pre‐ liminary
American College of Rheumatology and the World Health
The authors declare no conflicts of interest.
Organization/International League Against Rheumatism Criteria. Arthritis
Rheum. 1996;39:34‐40.
14. Singh JA, Saag KG, Bridges SL Jr, et al. 2015 American College of
ORCID
Rheumatology Guideline for the treatment of rheumatoid arthritis.
Arthritis Rheumatol. 2016;68:1‐26.
Kai‐Chun Wang https://orcid.org/0000-0003-2037-6681
15. Krieckaert CL, Nurmohamed MT, Wolbink GJ. Methotrexate re‐
Hsien‐Tzung Liao https://orcid.org/0000-0001-5930-4840 duces immunogenicity in adalimumab treated rheumatoid ar‐ thritis
patients in a dose dependent manner. Ann Rheum Dis. 2012;71:1914‐
Chien‐Chih Lai https://orcid.org/0000-0002-0546-8695
1915.
Ming‐Han Chen https://orcid.org/0000-0001-7822-2613 16. Atsumi T, Yamamoto K, Takeuchi T, et al. The first double‐blind,
randomised, parallel‐group certolizumab pegol study in methotrex‐ ate‐
Chang‐Youh Tsai https://orcid.org/0000-0002-4154-4018
naïve early rheumatoid arthritis patients with poor prognostic factors, C‐
OPERA, shows inhibition of radiographic progression. Ann Rheum Dis.
2016;75:75‐83.
REFERENCES 17. Li R, Zhao JX, Su Y, et al. High remission and low relapse with pro ‐
longed intensive DMARD therapy in rheumatoid arthritis (PRINT): a
1. Smolen JS, Landewé R, Bijlsma J, et al. EULAR recommendations multicenter randomized clinical trial. Medicine (Baltimore).
for the management of rheumatoid arthritis with synthetic and bi‐ ological 2016;95:e3968.
disease‐modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 18. Kameda H, Fujii T, Nakajima A, et al. Japan College of
2017;76:960‐977. Rheumatology guideline for the use of methotrexate in patients with
2. Nielen MM, van Schaardenburg D, Reesink HW, et al. Specifc rheumatoid arthritis. Mod Rheumatol. 2019;29:31‐40.
autoantibodies precede the symptoms of rheumatoid arthritis: a study of 19. Chatzidionysiou K, Lie E, Nasonov E, et al. Highest clinical effec‐
serial measurements in blood donors. Arthritis Rheum. 2004;50:380‐386. tiveness of rituximab in autoantibody‐positive patients with rheu‐ matoid
arthritis and in those for whom no more than one previous
WANG et al.      |  9

TNF antagonist has failed: pooled data from 10 European registries. 26. Chen MH, Chen MH, Liu CY, et al. Hepatitis B virus reactivation in
Ann Rheum Dis. 2011;70:1575‐1580. rheumatoid arthritis patients undergoing biologics treatment. J Infect Dis.
20. Sellam J, Hendel‐Chavez H, Rouanet S, et al. B cell activation 2017;215:566‐573.
biomarkers as predictive factors for the response to rituximab in
rheumatoid arthritis: a six‐month, national, multicenter, open‐label study.
Arthritis Rheum. 2011;63:933‐938.
21. Tak PP, Rigby WF, Rubbert‐Roth A, et al. Inhibition of joint damage SUPPORTING INFORMATION
and improved clinical outcomes with rituximab plus methotrexate in early
active rheumatoid arthritis: the IMAGE trial. Ann Rheum Dis. Additional supporting information may be found online in the
2011;70:39‐46. Supporting Information section at the end of the article.
22. Isaacs JD, Cohen SB, Emery P, et al. Effect of baseline rheumatoid
factor and anticitrullinated peptide antibody serotype on rituximab clinical
response: a meta‐analysis. Ann Rheum Dis. 2013;72:329‐336.
How to cite this article: Wang K‐C, Liao H‐T, Chen W‐S,
23. Harrold LR, Reed GW, Shewade A, et al. Effectiveness of ritux‐
imab for the treatment of rheumatoid arthritis in patients with prior et al. Real‐world effectiveness and safety of rituximab in the
exposure to anti‐TNF: results from the CORRONA registry. J treatment of rheumatoid arthritis: A single‐center experience
Rheumatol. 2015;42:1090‐1098. in Taiwan. Int J Rheum Dis. 2019;00:1–9. https://doi.
24. Liao TL, Lin CH, Chen YM, et al. Different risk of tuberculosis and
org/10.1111/1756-185X.13511
efficacy of isoniazid prophylaxis in rheumatoid arthritis patients with
biologic therapy: a nationwide retrospective cohort study in Taiwan.
PLoS ONE. 2016;11:e0153217.
25. Arkema EV, Jonsson J, Baecklund E, et al. Are patients with rheuma‐
toid arthritis still at an increased risk of tuberculosis and what is the role of
biological treatments? Ann Rheum Dis. 2015;74:1212‐1217.

You might also like