Skip to main content
Erschienen in: Rheumatology and Therapy 2/2018

Open Access 01.12.2018 | Original Research

Real-World Tocilizumab Use in Patients with Rheumatoid Arthritis in Canada: 12-Month Results From an Observational, Noninterventional Study

verfasst von: Boulos Haraoui, Shahin Jamal, Vandana Ahluwalia, Diana Fung, Tarang Manchanda, Majed Khraishi

Erschienen in: Rheumatology and Therapy | Ausgabe 2/2018

Abstract

Introduction

This study was conducted to observe patterns of use of the interleukin-6 receptor-alpha inhibitor tocilizumab in routine clinical practice in patients with rheumatoid arthritis (RA).

Methods

This was a 12-month noninterventional, observational study in adult patients with RA who initiated tocilizumab in routine practice in Canada according to the local product monograph. The primary end point was the proportion of patients receiving tocilizumab at 6 months. Secondary end points were treatment patterns, effectiveness, and safety of tocilizumab over 12 months.

Results

Of 200 patients who initiated tocilizumab (91.0% at 8 mg/kg), 67 (33.5%) received tocilizumab monotherapy and 133 (66.5%) received tocilizumab combined with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). Kaplan–Meier analysis estimated that 85% (95% CI 74–92%) of monotherapy and 89% (95% CI 82–93%) of combination therapy patients continued to receive tocilizumab at 6 months (log-rank p = 0.0888). During the observation period, 12 (17.9%) monotherapy and 27 (20.3%) combination therapy patients withdrew from the study. At month 12, 58.5% in the monotherapy group and 59.3% in the combination therapy group achieved Disease Activity Score at 28 joints remission (≤ 2.6), 25.6% and 24.7% achieved Simplified Disease Activity Index remission (≤ 3.3), and 18.2% and 22.3% achieved Clinical Disease Activity Index remission (≤ 2.8), respectively. Rates of serious adverse events and serious infections were found in 29.6/100 patient-years (PY) and 3.1/100 PY, respectively, for monotherapy and 19.2/100 PY and 4.8/100 PY, respectively, for combination therapy.

Conclusions

Patients initiating tocilizumab in routine practice had comparable effectiveness and safety outcomes regardless of whether they received tocilizumab as monotherapy or as combination therapy with csDMARDs.

Trial Registration

ClinicalTrials.gov identifier, NCT01613378

Funding

F. Hoffmann-La Roche (Roche) Canada.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1007/​s40744-018-0130-6) contains supplementary material, which is available to authorized users.

Enhanced Digital Features

To view enhanced digital features for this article go to https://​doi.​org/​10.​6084/​m9.​figshare.​7172285.

Introduction

Recommendations from the Canadian Rheumatology Association for the management of rheumatoid arthritis (RA) state that remission or low disease activity (LDA) should be targeted and that therapy should be adjusted every 3–6 months to achieve this goal [1]. Biological disease-modifying antirheumatic drugs (bDMARDs) such as tumor necrosis factor inhibitors (TNFi) abatacept and tocilizumab, in combination with methotrexate (MTX), are recommended to treat patients with poor prognostic factors or patients who do not respond to conventional synthetic DMARDs (csDMARDs) [13]. However, up to 37% of RA patients may not be able to continue treatment with MTX because of intolerance, and approximately one-third of patients receive bDMARD monotherapy [46].
Tocilizumab is a humanized anti–human monoclonal antibody against the interleukin-6 receptor alpha subunit (IL-6Rα), which blocks IL-6 signaling [7, 8]. In Canada, tocilizumab is approved for the treatment of RA and polyarticular and systemic juvenile idiopathic arthritis. The Canadian product monograph states that tocilizumab should be administered in combination with MTX or other csDMARDs or as monotherapy in cases of intolerance or contraindication to MTX. Recommended dosing for intravenous tocilizumab is 4 mg/kg every 4 weeks, increased to 8 mg/kg based on clinical response. In phase 3/4 clinical trials, tocilizumab was effective for treating the signs and symptoms of RA in combination with MTX/csDMARDs [913] and as monotherapy [1315]. Clinical practice studies confirmed the effectiveness and safety of tocilizumab monotherapy and combination therapy in the real world [16, 17].
ACT-UP CARE (ACT-UP Canadian Physician Observance of RA Patients on Tocilizumab) is part of an ongoing, multinational, observational study. Patterns of use of tocilizumab monotherapy or combination therapy with csDMARDs, including persistence on the drug and adherence to the licensed Canadian product monograph [18], were observed in routine clinical practice between February 2012 and October 2014 in RA patients.

Methods

Study Design

ACT-UP CARE, part of a global umbrella study to observe tocilizumab use initiated in routine clinical practice [19], was a 12-month, noninterventional, postmarketing, multicenter, observational study conducted at 26 sites across Canada. No interventional procedures or visits outside routine practice were mandated; tocilizumab and concomitant medications were prescribed, according to the investigator’s judgment and the Canadian product monograph [18], to adult patients with moderate-to-severe RA whose physicians decided to commence treatment with tocilizumab. Enrolled patients provided written informed consent approved by the institutional review boards or independent ethics committees. Patients who provided informed consent and who received at least one dose of tocilizumab in the Canadian arm of ACT-UP were included. Patients were grouped into those who received tocilizumab in combination with csDMARDs and those who received tocilizumab monotherapy. In an exploratory subanalysis, patients were grouped into csDMARD-inadequate responders (IR) and TNFi-IR.

Assessments

The baseline visit was the last valid assessment before the first dose of tocilizumab, which might not have coincided with the enrollment visit. The primary end point was the proportion of patients receiving tocilizumab at 6 months. Because the treatment regimen required infusion every 4 weeks, predefined assessment windows between study days 154 (24 weeks) and 197 (28 weeks) were considered to capture infusions that might have fallen outside a 6-month visit.
The proportion of patients receiving tocilizumab at 12 months was a secondary end point; predefined assessment windows between study days 351 (50 weeks) and 379 (54 weeks) were considered.
Other secondary end points were treatment patterns over time (3 months, 6 months, 9 months, and 12 months), including dose modifications, rates, and reasons for dose modification; physician compliance with the product monograph–recommended dosing regimen and management of adverse events (AEs); proportions of patients receiving tocilizumab monotherapy (3 months, 6 months, 9 months, and 12 months), including reasons for monotherapy at baseline and reasons for csDMARD withdrawal; effectiveness over 52 weeks, including change in C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), swollen joint count, tender joint count, Disease Activity Score based on 28 joints (DAS28), patient-reported outcomes (PROs) (on a visual analog scale [VAS] evaluating pain, fatigue, and morning stiffness), European League Against Rheumatism (EULAR) responses, Clinical Disease Activity Index (CDAI)/Simplified Disease Activity Index (SDAI), prevalence, and severity of extra-articular manifestations; change in concomitant csDMARDs and rationale and compliance with csDMARD and corticosteroid treatment; and safety over 52 weeks, including AEs, serious AEs (SAEs), rates of SAEs, and serious infections per 100 patient-years (PY) of exposure, cardiovascular events, and laboratory abnormalities. SAEs were determined by the investigator and were considered as any experience suggesting a significant hazard, contraindication, side effect, or precaution that resulted in death, was life threatening, necessitated hospitalization, resulted in persistent or significant disability or birth defect, or required intervention to prevent one of these events.

Statistical Analysis

A sample size of 200 was deemed sufficient for valid inferences to Canadian RA patients treated with tocilizumab based on an estimated rate for the primary outcome measure—6-month persistence of treatment—of 75% [95% confidence interval (CI) 69–81%] and estimated rates for effectiveness measures of 73% (95% CI 66–79%) for EULAR good/moderate response and 25% (95% CI 19–31%) for DAS28 < 2.6 according to previous studies [911]. All assessments except safety were performed in the full analysis set, which consisted of all patients enrolled. All results were based on available patient data, and there was no imputation for missing data. The safety population consisted of all patients who received at least one dose of tocilizumab. Continuous variables were summarized using descriptive statistics (number of subjects, mean, standard deviation, or standard error). Categorical variables were summarized using frequencies and percentages. Unless otherwise specified, all statistical tests were two-sided and were performed with a 5% alpha error rate without correction for multiplicity. CIs for the proportions of patients receiving tocilizumab were computed based on the Clopper–Pearson method, and p values were determined using Fisher’s exact test.
Post hoc analysis was conducted to examine baseline demographics and disease characteristics in patients who discontinued tocilizumab for lack of efficacy between baseline and month 6 (primary failure) and between month 6 and month 12 (secondary failure). Primary and secondary failures were defined arbitrarily by the clinician and according to the time of failure.

Results

Patients

Overall, 200 patients were enrolled from 26 sites across Canada and were included in the full analysis set (Fig. 1). The safety population included 198 patients because two patients did not receive tocilizumab. In the full analysis set, 67 patients (33.5%) received tocilizumab monotherapy and 133 patients (66.5%) received tocilizumab in combination with csDMARDs at baseline (19 patients in the combination therapy group discontinued csDMARDs during the study and continued tocilizumab monotherapy but were included in the combination therapy group). Ten patients in each group left the study during the first 6 months; 85.1% of monotherapy patients and 92.5% of combination therapy patients remained in the study at month 6 (including patients who discontinued tocilizumab but continued study observation). Two patients in the monotherapy group and 17 in the combination therapy group left the study between months 6 and 12; 82.1% of monotherapy patients and 79.7% of combination therapy patients remained in the study at month 12. The most common reason for study withdrawal in the monotherapy group was AEs (five patients), whereas the most common reasons for withdrawal in the combination therapy group were lack of efficacy (12 patients) and AEs (six patients).
Baseline characteristics were similar between monotherapy and combination therapy groups except for a higher incidence of previous RA-related surgical procedures and higher Patient Global Assessment of Disease Activity (PtGA) VAS scores in the monotherapy group (Table 1). Patients had advanced RA; the mean (± SD) disease duration at baseline was 13.8 (± 11.1) years in the monotherapy group and 12.0 (± 10.0) years in the combination therapy group. As expected, patients with advanced disease requiring biological treatment had comorbidities, including cardiovascular risk factors. Overall, patients in both groups had stable cholesterol profiles, and nine of 67 (13.4%) patients and 13 of 133 (9.8%) patients in the monotherapy group and combination therapy group, respectively, were concomitantly treated with statins. Baseline demographics and disease characteristics were generally comparable with those of the ACT-UP global study, in which 37.9% of patients received tocilizumab as monotherapy [19]. Framingham risk category at baseline was available for 57 tocilizumab-treated patients; 30 (52.6%) had low risk (< 10%), 21 (36.8%) had moderate risk (10–19%), and six (10.5%) had high risk (≥ 20%).
Table 1
Baseline demographics and disease characteristics
Parameter
Monotherapy
N = 67
Combination therapy, N = 133
p value
Age, years
55.2 (14.0)
55.6 (13.3)
0.841
Female, n/N (%)
53/67 (79.1)
107/133 (80.5)
0.853
Disease duration, years
13.8 (11.1)
n = 65
12.0 (10.0)
n = 132
0.379
TJC28
12.5 (7.0)
n = 65
12.6 (7.2)
n = 129
0.905
SJC28
9.8 (4.8)
n = 65
9.2 (5.5)
n = 129
0.415
PtGA, VAS mm
67.3 (20.8)
n = 67
60.1 (21.4)
n = 132
0.006b
PGA, VAS mm
65.1 (21.4)
n = 67
60.0 (22.7)
n = 132
0.051
CRP, mg/l
15.5 (17.2)
n = 57
21.8 (43.8)
n = 119
0.309
ESR, mm/h
26.9 (20.1)
n = 52
27.3 (23.4)
n = 109
0.623
Pain, VAS mm
65.5 (23.2)
n = 65
62.2 (22.7)
n = 130
0.241
Morning stiffness, VAS mm
60.5 (22.9)
n = 65
57.3 (22.9)
n = 129
0.357
Fatigue, VAS mm
66.1 (23.3)
n = 65
61.1 (24.7)
n = 129
0.096
DAS28
5.7 (1.0)
n = 63
5.6 (1.3)
n = 124
0.477
SDAI
37.3 (11.2)
n = 55
35.6 (14.8)
n = 115
0.268
CDAI
35.7 (11.8)
n = 65
33.8 (13.1)
n = 129
0.282
RF positive, n/N (%)
34/67 (50.7)
76/133 (57.1)
0.600
Anti–CCP positive, n/N (%)
17/67 (25.4)
39/133 (29.3)
0.202
Family history of coronary disease, n/N (%)
18/67 (26.9)
34/129 (26.4)
> 0.999
Current or past smoker, n/N (%)
29/67 (43.3)
60/133 (45.1)
0.885a
Previous RA-related surgical procedure, n/N (%)
23/67 (34.3)
25/133 (18.8)
0.022b
Structural joint damage, n/N (%)
36/67 (53.7)
67/133 (50.4)
0.764
Cardiovascular risk factors, n/N (%)
Cerebrovascular disease
1/67 (1.5)
3/132 (2.3)
> 0.999
Coronary artery disease
6/67 (9.0)
13/132 (9.8)
> 0.999
Diabetes, type 1 or 2
8/67 (11.9)
18/132 (13.6)
0.826
Hyperlipidemia
13/67 (19.4)
30/132 (22.7)
0.716
Hypertension
28/67 (41.8)
45/132 (34.1)
0.351
Data are presented as mean (SD) unless otherwise stated. N’s are the number of patients with baseline data available for each variable
p value was assessed using nonparametric Mann–Whitney U test for continuous variables and Fisher’s exact test for categorical variables
aBetween all smoking status groups: current smoker, past smoker, and non-smoker
bDenotes statistical difference (p < 0.05) between groups
CCP cyclic citrullinated peptide, CDAI Clinical Disease Activity Index, CRP C-reactive protein, DAS28 Disease Activity Score using 28 joints, ESR erythrocyte sedimentation rate, PGA Physician Global Assessment of disease activity, PtGA patient global assessment of disease activity, RA rheumatoid arthritis, RF rheumatoid factor, SDAI Simplified Disease Activity Index, SJC28 swollen joint count at 28 joints, TJC28 tender joint count at 28 joints, VAS visual analog scale

Tocilizumab Treatment

Overall, 173 (87.4%; 95% CI 81.9–91.7%) patients remained in the study and were still receiving tocilizumab infusions at month 6 (primary end point), and 159 (80.3%; 95% CI 74.1–85.6%) remained in the study and were still receiving tocilizumab at month 12. Post hoc analysis of the 13 patients who discontinued tocilizumab because of lack of efficacy between baseline and 12 months revealed that nine discontinued between baseline and month 6 (primary failure) and four discontinued between month 6 and month 12 (secondary failure). Although the patient numbers are too small for statistical comparisons, trends showed that patients with secondary failure were older and experienced longer durations of disease, more structural joint damage, and worse PRO scores—including Patient Pain VAS, PtGA VAS, and Fatigue VAS—than those with primary failure or patients still receiving tocilizumab at month 12. Objective measures, such as disease activity, were comparable between patients with primary failure, patients with secondary failure and patients still receiving tocilizumab at month 12 (Supplementary Table S1).
The most common reason for patients to receive tocilizumab monotherapy rather than combination therapy was intolerance of csDMARDs (34.3%). The next most common reasons were physician decision (26.9%), proven efficacy of monotherapy (9.0%), standard of care (7.5%), biological inadequate response/failure (6.0%), patient decision (6.0%), and other (10.5%).
Proportions of patients who remained in the study and were receiving tocilizumab at month 6 (primary end point) and month 12 were not significantly different regarding monotherapy and combination therapy with csDMARDs. Among monotherapy patients, 56 of 67 (83.5%) continued to receive tocilizumab at month 6 and 54 of 67 (81.0%) continued to receive tocilizumab at month 12. Among combination therapy patients, 117 of 133 (88.0%) continued to receive tocilizumab at month 6 and 105 of 133 (78.9%) continued to receive tocilizumab at month 12. Kaplan–Meier analysis estimated that 85% (95% CI 74–92%) of monotherapy patients and 89% (95% CI 82–93%) of combination therapy patients continued to receive tocilizumab at month 6 (log rank, p = 0.0888).
Most of the total patient population (n = 158; 79.0%) had previously received other biologicals before initiating tocilizumab, and these patients were balanced between the monotherapy (53/67; 79.1%) and the combination therapy (105/133; 78.9%) groups. Among the 53 monotherapy patients, 43.4% previously received one biological, 30.2% received two biologicals, and 26.4% received three or more biologicals. Among the 105 combination therapy patients, the proportions were 52.4, 23.8, and 23.8%, respectively, with no significant difference compared with the monotherapy patients (p = 0.542). The most common previous biological was etanercept, used by 47.8% of monotherapy patients and 34.6% of combination therapy patients (p = 0.091). Other previous biologicals were adalimumab (26.9 vs. 24.1%; p = 0.730), infliximab (22.4 vs. 29.3%; p = 0.317), certolizumab (20.9 vs. 13.5%; p = 0.220), abatacept (19.4 vs. 18.8%; p > 0.999), rituximab (9.0 vs. 9.0%; p > 0.999), and golimumab (1.5 vs. 11.3%; p = 0.014). The most common reason for discontinuing previous biological therapy was lack of efficacy (75.2 vs. 75.9%), followed by intolerance (13.9 vs. 11.5%) and patient decision (2.0 vs. 1.0%); 8.9% of monotherapy and 11.5% of combination therapy patients discontinued for other/unknown reasons.
Among patients who received tocilizumab in combination with previous and ongoing csDMARD/s (n = 125; although eight patients were assigned to the combination therapy group, the type of csDMARD was unknown), 75.2% received MTX, 34.6% received hydroxychloroquine, 15.0% received leflunomide, 8.3% received sulfasalazine, 3.0% received gold, 1.5% received azathioprine, and 0.8% received cyclosporine.
Most patients started tocilizumab at 8 mg/kg (monotherapy group, 92.5%; combination therapy group, 90.2%). Tocilizumab was initiated at 4 mg/kg in four monotherapy patients (6.0%) and 12 combination therapy patients (9.0%). One patient in the monotherapy group reported a starting dose of 7 mg/kg, and one patient in the combination therapy group reported a starting dose of 5 mg/kg, which may reflect logistical reasons for dosing rather than clinical rationale. At month 6, 54 monotherapy patients (96.4%) and 105 combination therapy patients (86.1%) were receiving stable doses. At month 12, 47 monotherapy patients (95.9%) and 90 combination therapy patients (93.8%) were receiving stable doses.

Effectiveness

Both the monotherapy and the combination therapy groups demonstrated a decrease in DAS28 over time (Fig. 2). Mean ± SD DAS28 decreased from baseline (5.7 ± 1.0 monotherapy, 5.6 ± 1.3 combination therapy) to month 3 (3.5 ± 1.6 monotherapy, 3.2 ± 1.5 combination therapy). Further decreases occurred through month 12 (2.7 ± 1.7 monotherapy, 2.4 ± 1.5 combination therapy). Rates of disease activity over time according to DAS28, SDAI, and CDAI criteria are shown in Fig. 3. Similar proportions of monotherapy (58.5%; 24/41) and combination therapy (59.3%; 48/81) patients achieved DAS28 remission (DAS28 ≤ 2.6) by month 12.

Safety

Proportions of patients with treatment-emergent AEs, tocilizumab discontinuations due to AEs, SAEs, and serious infections were similar between the monotherapy and combination therapy groups (Table 2). Rates of SAEs were 29.6/100 PY in the monotherapy group and 19.2/100 PY in the combination therapy group, and rates of serious infection were 3.1/100 PY and 4.8/100 PY, respectively. Shifts in alanine aminotransferase (ALT) or aspartate aminotransferase (AST) levels, or both, from normal at baseline to greater than the upper limit of normal (ULN) during the study were observed in 30.9% of monotherapy and 45.2% of combination therapy patients, respectively. Most increases were from normal at baseline to 1-3 × ULN, with increases in ALT and AST > 3 × ULN not reported in any patients in the monotherapy group but reported in five and three patients, respectively, in the combination therapy group. Neutrophil counts declined from normal at baseline to between the lower limit of normal and 1.0 × 109/l in 13.5% of monotherapy patients and 10.3% of combination therapy patients. Decreases in neutrophil counts to 0.5–1.0 × 109/l were reported in 3.8 and 7.2% of patients, respectively, and shifts to < 0.5 × 109/l were reported in 3.8 and 7.2% of patients, respectively (Table 2). Lipid levels (total cholesterol, low-density lipoprotein–cholesterol, high-density lipoprotein–cholesterol, and triglycerides) remained relatively stable over time (Supplementary Fig. S1). Two patients died during the study; one patient receiving combination therapy died of pneumonia and one patient receiving monotherapy died of cervical instability in the neck, fat embolism, obstructive shock, and hip pseudotumor. Neither death was considered related to tocilizumab by the treating physician. No gastrointestinal perforations were reported.
Table 2
Safety over 12 months (safety population)
 
Monotherapy
n = 67
Combination therapy
N = 131
All tocilizumab-treated patients
N = 198
Adverse events
 Total TEAEs, n
175
380
555
 Patients with ≥ 1 TEAE
60 (89.6)
106 (80.9)
166 (83.8)
 Discontinuation due to AE
7 (10.4)
11 (8.4)
18 (9.1)
 Total SAEs, n (rate per 100 PY)
19 (29.60/100 PY)
24 (19.22/100 PY)
43 (22.75/100 PY)
 Patients with ≥ 1 SAE
13 (19.4)
18 (13.7)
31 (15.7)
 Infection SAEs, n (rate per 100 PY)
2 (3.12/100 PY)
6 (4.80/100 PY)
8 (4.23/100 PY)d
 Patients with ≥ 1 infection SAE
2 (3.0)
5 (3.8)
7 (3.5)
 Deaths
1 (1.5)b
1 (0.8)c
2 (1.0)
Laboratory abnormalities, n/Na (%)
 AST shift from normal to
  1–3 × ULN
4/50 (8.0)
20/94 (21.3)
24/144 (16.7)
  3–5 × ULN
0
2/94 (2.1)
2/144 (1.4)
  > 5 × ULN
0
1/94 (1.1)
1/144 (0.7)
 ALT shift from normal to
  1–3 × ULN
13/55 (23.6)
24/115 (20.9)
37/170 (21.8)
  3–5 × ULN
0
5/115 (4.3)
5/170 (2.9)
  > 5 × ULN
0
0
0
 Neutrophils shift from normal to
  < LLN-1.0 × 109/l
7/52 (13.5)
10/97 (10.3)
17/149 (11.4)
  0.5–1.0 × 109/l
2/52 (3.8)
7/97 (7.2)
9/149 (6.0)
  < 0.5 × 109/l
2/52 (3.8)
7/97 (7.2)
9/149 (6.0)
aBased on available data for patients who had normal levels at baseline
bDeath due to cervical instability, fat embolism, obstructive shock, and hip pseudotumor
cDeath due to pneumonia
dTwo events of pneumonia and one event each of cellulitis, cystitis, postoperative wound infection, pyelonephritis, subcutaneous abscess, and urosepsis
Data are n (%) unless stated otherwise
AE adverse event, ALT alanine aminotransferase, AST aspartate aminotransferase, LLN lower limit of normal, PY patient-years, SAE serious adverse event, TEAE treatment-emergent adverse event, ULN upper limit of normal

Exploratory Analysis in csDMARD-IR and Biological-IR Patients

At baseline, 51 patients (25.5%) were csDMARD-IR when they initiated tocilizumab and 149 patients (74.5%) were biological-IR. After 6 months, 46 csDMARD-IR patients (90.2%) and 134 biological-IR patients (89.9%) remained in the study; after 12 months, 45 (88.2%) and 116 (77.8%) patients, respectively, remained in the study. Among biological-IR patients, 65 of 78 patients (83.3%) whose single previous biological failed and 60 of 80 patients (75.0%) whose multiple previous biologicals failed were still receiving tocilizumab after 12 months. Among the 78 single-previous biological-IR patients, 20 of 23 monotherapy patients (87.0%) and 45 of 55 combination therapy patients (81.8%) were still receiving tocilizumab at month 12. Among the 80 multiple-previous biological-IR patients, 23 of 30 monotherapy patients (76.7%), and 37 of 50 combination therapy patients (74.0%) were still receiving tocilizumab at month 12. Proportions of patients achieving DAS28 remission or LDA at month 12 in the csDMARD-IR and biological-IR subgroups, respectively, were 67.6% (23/34) and 55.7% (49/88) for remission and 5.9% (2/34) and 13.6% (12/88) for LDA. Response rates over time from months 0 to 12 are shown in Fig. 4.
AEs were reported by 47 of 50 csDMARD-IR patients (94.0%) and 119 of 148 biological-IR patients (80.4%). SAEs were reported by 16.0 and 15.5% of patients, respectively. Discontinuation because of AEs was reported for two patients (4.0%) in the csDMARD-IR subgroup and 16 patients (10.8%) in the biological-IR subgroup. No serious infections were reported in csDMARD-IR patients, and eight serious infections were reported in seven biological-IR patients (two pneumonia and one each cellulitis, cystitis, postoperative wound infection, pyelonephritis, subcutaneous abscess, and urosepsis). Both reported deaths occurred in biological-IR patients.

Discussion

The ACT-UP CARE study provides a snapshot of tocilizumab use in routine clinical practice over 12 months in the Canadian RA population between February 2012 and October 2014. Tocilizumab was received as monotherapy in 33.5% of patients in this study, consistent with observational studies estimating that one-third of RA patients treated with biologicals receive them as monotherapy [46]. The most common reason for receiving tocilizumab monotherapy was intolerance of csDMARDs, highlighting the need for biological agents equally effective as monotherapy and combination therapy. Drug survival rates with tocilizumab were high after 6 months and were not statistically significantly different between tocilizumab monotherapy and combination therapy with csDMARDs, with 85% of monotherapy and 89% of combination therapy patients remaining in the study and continuing to receive tocilizumab at 6 months as assessed by Kaplan–Meier analysis (p = 0.0888).
These results are consistent with previous results of tocilizumab monotherapy and combination therapy in randomized controlled trials and in clinical practice. Proportions of patients completing 6 months of tocilizumab were 91.0% for monotherapy and 93.2% for combination therapy in a randomized controlled trial [20] and 87.9 and 87.1%, respectively, in an open-label study resembling clinical practice [16]. However, in the ACT-UP global umbrella study, fewer monotherapy patients (80%) than combination therapy patients (87%) were still receiving tocilizumab after 6 months (p < 0.001 for the comparison) [19]. In RA patients, higher persistence on TNFi was observed when received in combination with csDMARDs than as monotherapy; however, greater persistence with combination therapy may be attributed to increased effectiveness compared with TNFi monotherapy [21]. This is in contrast to the results of the current analyses, which did not demonstrate differences in efficacy between tocilizumab monotherapy and combination therapy. Comparisons between studies are hindered by differences in patient populations and study design; nevertheless, in an observational global comparative effectiveness study in csDMARD-IR RA patients, the 6-month drug survival rate in patients receiving tocilizumab (91%) was higher than in patients receiving TNFi (85%; p < 0.001 for the comparison) [19]. In ACT-UP CARE, the 25.5% of patients who were csDMARD-IR and the 74.5% who were biological-IR when they initiated tocilizumab had good tocilizumab survival rates. These results support those of the previous open-label study of tocilizumab resembling clinical practice that reported 89.8% of DMARD-IR and 83.7% of TNFi-IR patients completed 6 months of tocilizumab treatment [22]. In ACT-UP CARE, 87.0 and 81.8% of monotherapy and combination therapy patients, respectively, were still receiving tocilizumab after 1 year of treatment at comparable rates regardless of whether one or multiple previous biological agents failed.
Effectiveness outcomes, including high hurdle measures such as rates of remission and LDA—targets recommended in American College of Rheumatology (ACR)/EULAR treat-to-target recommendations [23]—did not appear to differ between tocilizumab monotherapy and combination therapy groups in the current study, although statistical comparisons were not performed. After 1 year, DAS28 remission was achieved by 58.5% of monotherapy and 59.3% of combination therapy patients, respectively, SDAI remission by 25.6 and 24.7%, respectively, and CDAI remission by 18.2 and 22.3%, respectively. Canadian Rheumatology Association guidelines recommend a target of remission or LDA within 3 to 6 months [1]. In the current study, the total proportions of patients who achieved DAS28 LDA or remission after 6 months were 69.2% in the monotherapy group and 56.1% in the combination therapy group; for SDAI, the proportions were 62.1 and 39.5%, and for CDAI the proportions were 51.0 and 37.5%, respectively. Assessment of CDAI does not include measurement of CRP or ESR, suggesting that effectiveness was not entirely dependent on normalization of acute phase reactants by tocilizumab. Effectiveness observed in the current study with tocilizumab monotherapy and combination therapy supports results from previous clinical trials [13] and studies in clinical practice [16, 17].
Effectiveness responses in patients naive to biological therapy and patients whose biological therapy failed indicate that substantial proportions of patients who had not responded to previous biological therapy were able to achieve high hurdle responses with tocilizumab treatment: 57.3% of biological-IR patients achieved DAS28 LDA or remission, 43.5% achieved SDAI LDA or remission, and 40.0% achieved CDAI LDA or remission after 6 months of tocilizumab treatment. In the open-label study of tocilizumab resembling clinical practice [22], rates of LDA and remission according to DAS28, SDAI, and CDAI after 6 months of tocilizumab treatment were higher in TNFi-naive patients (75.4, 68.5, and 65.0%, respectively) than in those who previously received TNFi treatment (61.0, 50.6, and 47.1%, respectively). Similarly, in a pooled analysis of long-term data from the tocilizumab phase 3-4 clinical trial program, rates of remission according to DAS28, SDAI, and ACR/EULAR Boolean criteria were lower in TNF-IR patients than in DMARD-IR patients [24].
Patients who experienced secondary failure (discontinuing tocilizumab treatment because of loss of efficacy between months 6 and 12) tended to have more advanced disease and worse PROs than patients who experienced primary failure.
Medication adjustments occurred infrequently during the study, and 94.5% of patients were receiving stable doses of tocilizumab after 12 months. No new safety signals were identified for tocilizumab in this study; SAE and serious infection rates were consistent with those of the tocilizumab phase 3 clinical trial program and long-term extensions at 15.5/100 PY and 4.4/100 PY, respectively, during 12 months of tocilizumab treatment [24].
A limitation of this study was that tocilizumab dose de-escalation was not mandated for responders in the ACT-UP CARE study; future studies should investigate the effect of de-escalation from 8 to 4 mg/kg in patients who achieve remission or LDA. Another limitation is the post hoc nature of the analysis of primary and secondary failure. The subcutaneous formulation of tocilizumab was not approved in Canada when this study was conducted, and future studies should examine the effectiveness of subcutaneous tocilizumab in an observational setting now that it is approved in Canada [18]. Although this study is limited to a single Canadian cohort, it is part of a wider published global study that informs on the generalizability of the findings. In conclusion, treatment with tocilizumab was effective as monotherapy and in combination with csDMARDs in this real-world observational study of RA patients in routine clinical practice in Canada.

Acknowledgements

We thank the patients for their participation in this study.

Funding

This multicenter study was funded by F. Hoffmann-La Roche (Roche) Canada, who participated in the design of the study, interpretation of data, and review and approval of the manuscript. Roche Canada also funded medical writing support and article processing charges. All authors had full access to all the data in this study and take complete responsibility for the integrity of the data and accuracy of the data analysis.

Medical Writing, Editorial, and Other Assistance

Medical writing support was provided by Sara Duggan, PhD, of ApotheCom and was funded by F. Hoffmann-La Roche Ltd.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Disclosures

Boulos Haraoui reports grants and nonfinancial support from Hoffmann-La Roche Canada and has served as an advisor for AbbVie, Amgen, BMS, Eli Lilly, Janssen, Merck, Pfizer, Sanofi, and UCB. Shahin Jamal has served on the advisory boards of Roche, Amgen, AbbVie, BMS, Eli Lilly, Celgene, Merck, Novartis, Pfizer, Sandoz, Sanofi, Janssen, and UCB. Vandana Ahluwalia has served as a consultant or on advisory boards for Amgen, AbbVie, Janssen, Roche, Pfizer, Novartis, Sanofi, Merck, Eli Lilly, and Hospira. Diana Fung is an employee of Hoffmann-La Roch Canada. Tarang Manchanda is an employee of Hoffmann-La Roche Canada. Majed Khraishi has served as a consultant to Amgen, Pfizer, Eli Lilly, and Novartis.

Compliance with Ethics Guidelines

Enrolled patients provided written informed consent approved by the institutional review boards or independent ethics committees.

Data Availability

The data that support the findings of this study are available from the corresponding author on reasonable request and with permission from Hoffmann-La Roche Ltd.

Open Access

This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Bykerk VP, Akhavan P, Hazlewood GS, et al. Canadian Rheumatology Association recommendations for pharmacological management of rheumatoid arthritis with traditional and biologic disease-modifying antirheumatic drugs. J Rheumatol. 2012;39:1559–82.CrossRef Bykerk VP, Akhavan P, Hazlewood GS, et al. Canadian Rheumatology Association recommendations for pharmacological management of rheumatoid arthritis with traditional and biologic disease-modifying antirheumatic drugs. J Rheumatol. 2012;39:1559–82.CrossRef
2.
Zurück zum Zitat Singh JA, Furst DE, Bharat A, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res. 2012;64:625–39.CrossRef Singh JA, Furst DE, Bharat A, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res. 2012;64:625–39.CrossRef
3.
Zurück zum Zitat Smolen JS, Landewe R, Breedveld FC, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis. 2014;73:492–509.CrossRef Smolen JS, Landewe R, Breedveld FC, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis. 2014;73:492–509.CrossRef
4.
Zurück zum Zitat Yazici Y, Shi N, John A. Utilization of biologic agents in rheumatoid arthritis in the United States: analysis of prescribing patterns in 16,752 newly diagnosed patients and patients new to biologic therapy. Bull NYU Hosp Joint Dis. 2008;66:77–85. Yazici Y, Shi N, John A. Utilization of biologic agents in rheumatoid arthritis in the United States: analysis of prescribing patterns in 16,752 newly diagnosed patients and patients new to biologic therapy. Bull NYU Hosp Joint Dis. 2008;66:77–85.
5.
Zurück zum Zitat Lee SJ, Chang H, Yazici Y, et al. Utilization trends of tumor necrosis factor inhibitors among patients with rheumatoid arthritis in a United States observational cohort study. J Rheumatol. 2009;36:1611–7.CrossRef Lee SJ, Chang H, Yazici Y, et al. Utilization trends of tumor necrosis factor inhibitors among patients with rheumatoid arthritis in a United States observational cohort study. J Rheumatol. 2009;36:1611–7.CrossRef
6.
Zurück zum Zitat Soliman MM, Ashcroft DM, Watson KD, Lunt M, Symmons DP, Hyrich KL. Impact of concomitant use of DMARDs on the persistence with anti-TNF therapies in patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis. 2011;70:583–9.CrossRef Soliman MM, Ashcroft DM, Watson KD, Lunt M, Symmons DP, Hyrich KL. Impact of concomitant use of DMARDs on the persistence with anti-TNF therapies in patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis. 2011;70:583–9.CrossRef
7.
Zurück zum Zitat Mihara M, Kasutani K, Okazaki M, et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int Immunopharmacol. 2005;5:1731–40.CrossRef Mihara M, Kasutani K, Okazaki M, et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int Immunopharmacol. 2005;5:1731–40.CrossRef
8.
Zurück zum Zitat Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008;112:3959–64.CrossRef Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008;112:3959–64.CrossRef
9.
Zurück zum Zitat Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008;371:987–97.CrossRef Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008;371:987–97.CrossRef
10.
Zurück zum Zitat Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial. Ann Rheum Dis. 2008;67:1516–23.CrossRef Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial. Ann Rheum Dis. 2008;67:1516–23.CrossRef
11.
Zurück zum Zitat Genovese MC, McKay JD, Nasonov EL, et al. Interleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to disease-modifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy study. Arthritis Rheum. 2008;58:2968–80.CrossRef Genovese MC, McKay JD, Nasonov EL, et al. Interleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to disease-modifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy study. Arthritis Rheum. 2008;58:2968–80.CrossRef
12.
Zurück zum Zitat Kremer JM, Blanco R, Brzosko S, et al. Tocilizumab inhibits structural joint damage in rheumatoid arthritis patients with inadequate responses to methotrexate: results from the double-blind treatment phase of a randomized placebo-controlled trial of tocilizumab safety and prevention of structural joint damage at one year. Arthritis Rheum. 2011;63:609–21.CrossRef Kremer JM, Blanco R, Brzosko S, et al. Tocilizumab inhibits structural joint damage in rheumatoid arthritis patients with inadequate responses to methotrexate: results from the double-blind treatment phase of a randomized placebo-controlled trial of tocilizumab safety and prevention of structural joint damage at one year. Arthritis Rheum. 2011;63:609–21.CrossRef
13.
Zurück zum Zitat Burmester GR, Rigby WF, Van Vollenhoven RF, et al. Tocilizumab in early progressive rheumatoid arthritis: FUNCTION, a randomised controlled trial. Ann Rheum Dis. 2016;75:1081–91.CrossRef Burmester GR, Rigby WF, Van Vollenhoven RF, et al. Tocilizumab in early progressive rheumatoid arthritis: FUNCTION, a randomised controlled trial. Ann Rheum Dis. 2016;75:1081–91.CrossRef
14.
Zurück zum Zitat Jones G, Sebba A, Gu J, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis. 2010;69:88–96.CrossRef Jones G, Sebba A, Gu J, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis. 2010;69:88–96.CrossRef
15.
Zurück zum Zitat Gabay C, Emery P, Van Vollenhoven R, et al. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. Lancet. 2013;381:1540–1.CrossRef Gabay C, Emery P, Van Vollenhoven R, et al. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. Lancet. 2013;381:1540–1.CrossRef
16.
Zurück zum Zitat Bykerk VP, Ostor AJ, Alvaro-Gracia J, et al. Comparison of tocilizumab as monotherapy or with add-on disease-modifying antirheumatic drugs in patients with rheumatoid arthritis and inadequate responses to previous treatments: an open-label study close to clinical practice. Clin Rheumatol. 2015;34:563–71.CrossRef Bykerk VP, Ostor AJ, Alvaro-Gracia J, et al. Comparison of tocilizumab as monotherapy or with add-on disease-modifying antirheumatic drugs in patients with rheumatoid arthritis and inadequate responses to previous treatments: an open-label study close to clinical practice. Clin Rheumatol. 2015;34:563–71.CrossRef
17.
Zurück zum Zitat Weinblatt ME, Kremer J, Cush J, et al. Tocilizumab as monotherapy or in combination with nonbiologic disease-modifying antirheumatic drugs: twenty-four-week results of an open-label, clinical practice study. Arthritis Care Res. 2012;13:362–71. Weinblatt ME, Kremer J, Cush J, et al. Tocilizumab as monotherapy or in combination with nonbiologic disease-modifying antirheumatic drugs: twenty-four-week results of an open-label, clinical practice study. Arthritis Care Res. 2012;13:362–71.
19.
Zurück zum Zitat Haraoui B, Casado G, Czirjak L, et al. Patterns of tocilizumab use, effectiveness and safety in patients with rheumatoid arthritis: core data results from a set of multinational observational studies. Clin Exp Rheumatol 2017 Apr 28 (Epub ahead of print). Haraoui B, Casado G, Czirjak L, et al. Patterns of tocilizumab use, effectiveness and safety in patients with rheumatoid arthritis: core data results from a set of multinational observational studies. Clin Exp Rheumatol 2017 Apr 28 (Epub ahead of print).
20.
Zurück zum Zitat Dougados M, Kissel K, Sheeran T, et al. Adding tocilizumab or switching to tocilizumab monotherapy in methotrexate inadequate responders: 24-week symptomatic and structural results of a 2-year randomized controlled strategy trial in rheumatoid arthritis (ACT-RAY). Ann Rheum Dis. 2013;72:43–50.CrossRef Dougados M, Kissel K, Sheeran T, et al. Adding tocilizumab or switching to tocilizumab monotherapy in methotrexate inadequate responders: 24-week symptomatic and structural results of a 2-year randomized controlled strategy trial in rheumatoid arthritis (ACT-RAY). Ann Rheum Dis. 2013;72:43–50.CrossRef
21.
Zurück zum Zitat Koncz T, Pentek M, Brodszky V, Ersek K, Orlewska E, Gulacsi L. Adherence to biologic DMARD therapies in rheumatoid arthritis. Expert Opin Biol Ther. 2010;10:1367–78.CrossRef Koncz T, Pentek M, Brodszky V, Ersek K, Orlewska E, Gulacsi L. Adherence to biologic DMARD therapies in rheumatoid arthritis. Expert Opin Biol Ther. 2010;10:1367–78.CrossRef
22.
Zurück zum Zitat Bykerk V, Ostor AJK, Alvaro-Gracia J, et al. Tocilizumab in patients with active rheumatoid arthritis and inadequate responses to DMARDs and/or TNF inhibitors: a large, open-label study close to clinical practice. Ann Rheum Dis. 2012;71:1950–4.CrossRef Bykerk V, Ostor AJK, Alvaro-Gracia J, et al. Tocilizumab in patients with active rheumatoid arthritis and inadequate responses to DMARDs and/or TNF inhibitors: a large, open-label study close to clinical practice. Ann Rheum Dis. 2012;71:1950–4.CrossRef
23.
Zurück zum Zitat Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75:3–15.CrossRef Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75:3–15.CrossRef
24.
Zurück zum Zitat Genovese MC, Rubbert-Roth A, Smolen JS, et al. Longterm safety and efficacy of tocilizumab in patients with rheumatoid arthritis: a cumulative analysis of up to 4.6 years of exposure. J Rheumatol 2013;40:768–80.CrossRef Genovese MC, Rubbert-Roth A, Smolen JS, et al. Longterm safety and efficacy of tocilizumab in patients with rheumatoid arthritis: a cumulative analysis of up to 4.6 years of exposure. J Rheumatol 2013;40:768–80.CrossRef
Metadaten
Titel
Real-World Tocilizumab Use in Patients with Rheumatoid Arthritis in Canada: 12-Month Results From an Observational, Noninterventional Study
verfasst von
Boulos Haraoui
Shahin Jamal
Vandana Ahluwalia
Diana Fung
Tarang Manchanda
Majed Khraishi
Publikationsdatum
01.12.2018
Verlag
Springer Healthcare
Erschienen in
Rheumatology and Therapy / Ausgabe 2/2018
Print ISSN: 2198-6576
Elektronische ISSN: 2198-6584
DOI
https://doi.org/10.1007/s40744-018-0130-6

Weitere Artikel der Ausgabe 2/2018

Rheumatology and Therapy 2/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.