Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2016

Open Access 01.12.2016 | Research article

Comparative effectiveness of treatment with the first TNF antagonist in monotherapy, the first TNF antagonist plus one conventional synthetic disease-modifying antirheumatic drug, and the first TNF antagonist plus two or more conventional synthetic disease-modifying antirheumatic drugs in patients with rheumatoid arthritis

verfasst von: Blanca Hernández-Cruz, Esther Márquez-Saavedra, Rafael Caliz-Caliz, Federico Navarro-Sarabia

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2016

Abstract

Background

Rheumatoid arthritis (RA) patients are treated with a mean of 3–4 conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) with or without glucocorticoids (GCs), before the first biologic prescription. The main reasons for change are inefficacy in 30–40 % of patients, and toxicity ≈ 10 %. Thus, they are treated with the first TNF antagonists in monotherapy. The aim of this study was to analyse the csDMARD and GC prescription patterns before and during treatment with the first TNF antagonist, and compare their effectiveness in three groups of patients.

Methods

An observational, prospective, multicentre study in common clinical practice was designed. Treating rheumatologists recorded patient variables, including previous and concomitant csDMARDs and GCs in a database. The data were analysed using descriptive, inferential and multivariate statistics.

Results

There were 1136 patients included; 21 % received the first TNF antagonist in monotherapy, 67 % received the first TNF antagonist plus one csDMARD, and 12 % the first TNF antagonist plus two or more csDMARDs. Most patients were female (73 %), RF+, and ACPA+, and had erosions; mean age was 53.2 (±13.0) years, and duration of disease was 9.1 (±7.6) years. They had high activity with DAS28 of 5.8 ± 1.1, and poor physical function with HAQ of 1.43 ± 0.63, and significant differences between groups in clinical variables and comorbidities; 94 % had received treatment with GCs, MTX, LFN, or SSZ at any time before the first TNF antagonist, 5 % (n = 52) had been treated with CLQ or HCLQ, and 1 % (n = 13) had received neither GCs nor csDMARDs. Before the first TNF antagonist, the drugs most commonly used were GCs (78 %), MTX (50 %), LFN (44 %), and SSZ (21 %). Concomitantly with the first TNF antagonist, 977 patients (85 %) were receiving GCs, MTX, LFN, or SSZ; 15 % (n = 173) received their first TNF antagonist without any concomitant GCs or csDMARDs, true monotherapy, and 6 % received their first TNF antagonist with GCs. The drug most commonly used at the time of first TNF antagonist initiation was MTX (58 %). All treatment groups had clinically and statistically significant improvements in DAS and HAQ scores. Effectiveness analysis (controlling for confounders) showed mean drug survival of 16.7, 20.1 and 11.7 months in each group, respectively (p < 0.001). The model that best explained a good EULAR response included the baseline and 6-month DAS28.

Conclusions

The three groups of patiernts, have different comorbidities and disease characteristics. Treatment with low or very low doses of GCs is common. True monotherapy with the first TNF antagonist without prednisone or csDMARDs is infrequent. After controlling for potential confounders, effectiveness was a little different.
Abkürzungen
ACPA
anti-citrullinated protein antibodies
bDMARDs
biologic disease-modifying anti-rheumatic drugs
CCT
controlled clinical trials
CLQ
chloroquine
csDMARDs
conventional synthetic disease-modifying anti-rheumatic drugs
DAS28
disease activity score with 28 count joints
DAS28-4v-ESR score
disease activity score with four variables and erythrocyte sedimentation rate
DMARD
disease-modifying anti-rheumatic drug
ESR
erythrocyte sedimentation rate
EULAR
European League Against Rheumatism
GCs
glucocorticoids
HAQ
health assessment questionnaire
HCLQ
hydroxychloroquine
LFN
leflunomide
MTX
methotrexate
PCR
protein C reactive
PDN
prednisone
PPD skin reaction
purified protein derivative skin reaction
RA
rheumatoid arthritis
RF
rheumatoid factor
SSZ
sulfasalazine
TCZ
tocilizumab
TNF
tumor necrosis factor
VAS
visual analogue scale

Background

Conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) with or without glucocorticoids (GCs) are the first line of the treatment for rheumatoid arthritis (RA) in the T2T strategy and in most of the recommendations for RA treatment [1, 2], and see https://​www.​nice.​org.​uk/​guidance/​cg79/​chapter/​Recommendations#pharmacological-management [3]. They can be used as sequential monotherapy or in combination, and in all of them, methotrexate (MTX) is the “anchor drug” [4].
Before the first biologic drug is prescribed, the patients are treated with a mean of 3–4 csDMARDs, mainly MTX, leflunomide (LFN), sulfasalazine (SSZ) and antimalarial drugs [5]. The main reasons for changing this therapy are inefficacy in 30–40 % of patients with early RA [6], and toxicity in about 10 % of patients [47]. Thus, between 10 % and 30 % of patients seen in clinical practice are on inefficacious treatment or have serious comorbidities or toxicity that rule out the use of MTX, LFN, and SSZ [8]. Some of these comorbidities are hepatitis B or C virus infections, abnormal liver function, severe anaemia, leukopenia or thrombocytopenia, severe or recurrent infections, past history of cancer, or multiple sclerosis. Thus, up to a third of patients are treated with biologic disease-modifying antirheumatic drugs (bDMARDs) in monotherapy (i.e., without any concomitant csDMARD) [814].
Summaries of the product characteristics of bDMARDs, with the exception of tocilizumab (TCZ), and clinical practice guidelines, clinical trials meta-analyses, and clinical practice registries recognise that a TNF antagonist plus csDMARD combination therapies are associated with better outcomes and with greater retention rates. [1, 814]. In controlled clinical trials (CCT), bDMARD monotherapy (except for tocilizumab (mainly anti-TNF)) had lower efficacy and survival rates when compared with combination therapies [13, 14]. European League Against Rheumtism (EULAR) recommendations are clear on this point, and emphasize the complexity of patient management in RA and the benefits of GCs at low doses [1, 2, 15]. GCs at low or very low doses are commonly used in clinical practice in combination with csDMARDs, particularly in patients with early disease, and there is a growing body of evidence supporting their role as cost-effective csDMARDs [16, 17].
As TNF antagonist are frequently used in monotherapy for reasons of safety in patients with adult-onset RA treated within the Andalusian Health Service, a registry was designed with the following objectives:
1.
To understand and compare clinical features of patients who receive the first TNF antagonist monotherapy, against the patients who receive the first TNF antagonist plus one csDMARD, against the patients who receive the first TNF antagonist plus two or more csDMARDs.
 
2.
To determine the prescription patterns of csDMARDs with high efficacy (MTX, LFN, and SSZ) and GCs before the first TNF antagonist prescription.
 
3.
To identify csDMARD and GC prescription patterns at initiation of the first TNF antagonist.
 
4.
To assess the effectiveness of the first TNF antagonist in the three groups of patients, using EULAR response, and improvement in health assessment questionnaire (HAQ) scores, survival curves and multiple logistic regression analysis.
 

Methods

Design

This was an observational, prospective, multicentre, analytical study.

Inclusion criteria

Inclusion criteria were patients over 18 years of age, able to complete follow-up questionnaires, fulfilling criteria for RA published in 1987 [18], and treated with at least one dose of any TNF antagonist (adalimumab, infliximab, etanercept) approved for use in patients with moderately active RA in accordance with the Second Consensus Document on the Use of Biologic Agents in RA of the Spanish Rheumatology Society [19]. This consensus document states that a patient should have failed treatment with two or more csDMARDs for at least 3 months, or have developed toxicity to the administered drugs in order to initiate therapy with a TNF antagonist.
Patients with diagnoses other than RA, duplicated registration, or those whose data were insufficient to calculate clinically relevant outcomes were excluded from the study. The study was approved by the Ethical and Human Research Committee of the Virgen de la Macarena Hospital Area, and conducted in accordance with The Declaration of Helsinki [20] and the Good Clinical Practice Guidelines (http://​www.​ich.​org/​products/​guidelines/​efficacy/​efficacy-single/​article/​good-clinical-practice.​html). The SAS, the public healthcare system in the Autonomous Community of Andalusia, prescribes and reimburses approximately 80 % of bDMARDS, while the remaining 20 % are prescribed by private medical clinics. Registry completion was mandatory to obtain the bDMARD from the SAS. All rheumatology departments of Andalusia were invited to take part in the registry. Participating rheumatologists received training on the study objectives and on the different operative definitions used in this project. Inefficacy was defined according to the judgement of the treating rheumatologist. Serious adverse events were defined as death, hospitalization, or life-threatening for the patient. Non-serious adverse events were defined as non-serious clinical conditions leading to discontinuation of therapy with DMARDs.
A treating rheumatologist and/or a research fellow at each centre had access to a specifically designed website, and prospectively recorded the following information: sociodemographic and RA-related variables, with particular emphasis on the patient’s disease activity, physical function, previous and concomitant therapy with csDMARDs and GCs, current therapy with csDMARDs and GCs at initiation of therapy with the biologic agent, reasons for discontinuation of csDMARDs or GCs or switch, and comorbidities. The relevant comorbidities were: secondary Sjögren’s syndrome, interstitial lung disease, rheumatoid vasculitis, amyloidosis, latent or active tuberculosis, past history of tuberculosis, purified protein derivative skin reaction (PPD) test, hepatitis B or C infection, HIV infection, other severe or recurrent infections, previous cancer, history of heart failure, and demyelinating disease. The number of comorbidities and mortality were also registered.
Patients were divided into three groups according to the type of treatment they were receiving: first TNF antagonist monotherapy; first TNF antagonist plus one csDMARD; and a first TNF antagonist plus two or more csDMARDs. From the website the data were recorded in an Excel spreadsheet. The Excel spreadsheet was cleaned and the data were verified with the clinical charts, for subsequent processing using STATA v 10.0 software.
Central tendency and dispersion measures were calculated in the first phase of the analysis and graphic analysis was performed. The three groups were subsequently compared using analysis of variance (ANOVA) for quantitative variables with a Gaussian distribution and/or the Kruskal − Wallis test for quantitative variables with a non-Gaussian distribution or ordinal variables. In the case of significant differences, the Mann–Whitney U test or Student’s t test with Bonferroni correction for multiple comparisons was used to analyse specific differences between two of the three groups. For nominal variables, contingency tables were produced and the chi-squared or fisher exact test was applied. In all cases the most pragmatic statistical analysis was performed.
For effectiveness analyses the primary outcome was good/moderate EULAR response with a first anti-TNF therapy, which was defined as the length of time the patients continued to receive their first anti-TNF therapy; patients were censored at the treatment stop date, date of death or date of the last follow up, whichever came first. Kaplan–Meier survival curves were used to describe persistence with anti-TNF therapy. Also logistic regression was used to identify predictive models of EULAR good response. Survival analysis and logistic regression were performed in a crude way, and adjusted by age, sex, disease duration, rheumatoid factor (RF) positivity, anti-citrullinated protein antibodies (ACPA) positivity, baseline disease activity score in 28 joints (DAS28), baseline health assessment questionnaire (HAQ), erosions, and number of comorbidities. In a third phase a propensity score was created using the same variables in order to adjust for severity of the disease.
Missing values were imputed using three techniques: (1) using the mean value of the previous observations, (2) using the value of the last observation, and (3) looking at the pattern of missing values in the time series, and calculating these values.

Results

The registry was active from 15 May 2008 to 31 March 31 2012. During this period, 20 physicians from 18 rheumatology departments, and 2 internal medicine departments recorded data from 1237 patients treated with their first biologic agent. There were 101 patients (8 %) excluded from the registry for various reasons: 47 patients did not initiate treatment, 28 had duplicate registrations, 14 patients started rituximab, 6 patients had received abatacept as their first biologic agent, and 6 were excluded due to lack of indication. Results of 1136 patients with adult-onset RA are presented. Of these patients, 21 % received their first TNF antagonist in monotherapy, 67 % received their first TNF antagonist plus one csDMARD, and 12 % were treated with a first TNF antagonist plus two or more csDMARDs (Table 1).
Table 1
Rheumatoid arthritis characteristics at baseline
 
First TNF antagonist monotherapy
First TNF antagonist plus one csDMARD
First TNF antagonist plus two or more csDMARDs
Total
P*
n, %
234
21
766
67
136
12
1.136
100
 
Gender, female n, %
175
75
557
73
99
73
831
73
0.8
Rheumatoid factor (RF)+ n, %
183
78
619
81
104
77
906
80
0.4
ACPA + n, % *
47/71
66
330/431
77
47/70
67
424/ 572
74
0.06
RF- and ACPA- n, %
12
5
64
8
14
10
90
8
0.07
Erosions n, %
195
83
622
81
100
74
917
81
0.05
Rheumatoid nodules n, %
50
21
189
25
18
13
257
23
0.01
Joint arthroplasty n, %
15
6
66
9
3
2
84
7
0.02
Number of comorbidities n, %
 0
67
30
159
22
51
42
277
26
0.003
 1
32
14
72
10
12
10
116
11
 ≥2
124
56
481
68
57
48
662
63
Anti-hepatitis B antibody+ n, %
2/47
4
7/194
4
0/26
0
9/267
3
0.2
Anti-hepatitis C antibody+ n, %
6/46
13
6/195
3
2/26
8
14/267
5
0.05
PPD skin reaction + (≥5 mm) n, %
24
10
120
16
17
13
161/1.112
14
0.09
Secondary Sjögren’s syndrome n, %
44
19
128
17
17
12
189
17
0.2
Interstitial lung disease n, %
16
7
25
3
1
0.7
42
4
0.007
Recurrent infections n, %
5
2
11
1
1
0.7
17
2
0.54
Past cancer n, %
4
2
5
0.6
0
0
9
0.8
0.1
Mean ± SD
         
Age (years) mean ± SD
55.9
±13.7
52.8
±13.4
50 .7
±11.1
53.2
±13.3
0.0001
Duration of disease (years) mean ± SD
9.4
±8.0
9.0
±7.5
8.8
±7.3
9.1
±7.6
0.8
Number of comorbidities mean ± SD
2.2
±2.3
2.7
±2.4
1.9
±2.3
2.5
±2.4
0.0001
Tender joint count (0–28) mean ± SD
10.9
±6.4
10.3
±6.6
11.6
±6.9
10.6
±6.6
0.0003
Swollen joint count (0–28) mean ± SD
9.0
±6.1
7.9
±5.5
8.5
±5.8
8.2
±5.7
0.02
Pain visual analogue scale (0–100) mean ± SD
61.9
±19.3
63.5
±22.2
66.0
±20.8
63.4
±21.5
0.01
DAS28-ESR mean ± SD
5.9
±1.0
5.7
±1.1
5.7
±1.0
5.8
±1.1
0.03
HAQ score (0–3) mean ± SD
1.40
±0.58
1.44
±0.65
1.6
±0.59
1.46
±0.63
0.0003
ESR (mm) mean ± SD
45.7
±24.7
42.1
±25.8
32.7
±21.5
41.8
±25.3
0.0001
C-reactive protein (mg/L) mean ± SD
19.3
±21.4
15.7
±21.5
15.8
±19.4
16.5
±21.2
0.0001
csDMARD conventional synthetic disease-modifying antirheumatic drug, ACPA anti-citrullianted protein antibodies, PPD purified protein derivative skin reaction, DAS28-ESR disease activity score in 28 joints-erythrocyte sedimentation rate, HAQ health assessment questionnaire.
*This variable was not assessed in all patients
Differences between groups were observed in patient characteristics, percentages of patients with joint arthroplasty, number of comorbidities, positive hepatitis virus antibodies, and positive tuberculin test results (Table 1). In addition to the comorbidities shown in the table, there were 13 patients (1 %) with rheumatoid vasculitis, of whom 3 (1 %) were patients in the group treated with the first TNF antagonist in monotherapy, 9 (1 %) were in the group treated with the first TNF antagonist plus one csDMARD, and 1 (0.7 %) was in the group treated with the first TNF antagonist plus two or more csDMARDs (p = 0.8); in the same groups respectively, there were 2 patients (0.8 %), 4 patients (0.5 %), and 0 patients (0 %) with amyloidosis, respectively p = 0.5), and 1 patient (0.4 %), 4 patients (0.5 %), 0 patients (0 %) with previous heart failure (p = 0.6). Previous tuberculosis was identified in 6 patients (3 %), 24 patients (3 %) and 0 patients (0 %), respectively (p = 0.08). In the group treated with the first TNF antagonist plus one csDMARD there was one patient with HIV infection and one with demyelinating disease. Neither of these patients died.
Types of first TNF antagonist are shown on Fig. 1. As expected, and in accordance with the summaries of product characteristics and treatment recommendations, most patients received etanercept as their first TNF antagonist monotherapy, and significant differences were observed in the type of TNF antagonist used within each treatment group, p < 0.0001.
Among all patients, 94 % had received treatment with GCs, MTX, LFN, or SSZ at any time before the TNF antagonist prescription (Table 2); 6 % had not received these drugs, 5 % (n = 52) had been treated with antimalarial agents, and only 1 % (n = 13) had received neither GCs nor csDMARDs. Most patients had received combination therapies (n = 761; 66 %). The drugs most commonly used in monotherapy or in combination were GCs, either prednisone (PDN) or equivalent, used in 78 % of patients, followed by MTX (50 %), LFN (44 %), and SSZ (21 %). The most frequent combination was GC plus MTX, LFN, or SSZ, used in 31 % (n = 357) of patients, followed by a GC plus two csDMARD 23 % (n = 267). Combinations of csDMARDs without GCs were less frequent: 4 % of patients were treated with MTX + SSZ + LFN + PDN. Inefficacy of the drug was the primary reason for MTX, LFN, and SSZ discontinuation in over half of the patients. Non-serious adverse events were the second reason for discontinuation of these csDMARDs in 40 %, 36 %, and 40 % of patients, respectively. The frequency of serious adverse events was very low.
Table 2
Prescription patterns of csDMARDs (MTX, LFN y SSZ) and GCs before the firs TNF antagonist
Treatment
First TNF antagonist monotherapy
First TNF antagonist plus one csDMARD
First TNF antagonist plus two or more csDMARDs
Total
P*
 
n
%
n
%
n
%
n
%
 
 
234
21
766
67
136
12
1.136
100
 
Previous use of DMARDs (MTX, LFN, SSZ) and/or PDN) (n , %)
229
98
712
93
130
96
1,071
94
 
None (n, %)
5
2
54
7
6
4
65
6
0.0001
One (n, %)
        
< 0.001
 PDN
8
3
139
18
76
56
223
20
 MTX + PDN
47
20
122
16
9
7
178
16
 LFN + PDN
17
7
106
14
8
6
131
11
 LFN
1
0.4
41
5
2
1
44
4
 SSZ + PDN
3
1
29
4
12
9
44
4
 MTX
5
2
33
4
1
0.7
39
3
 SSZ
4
2
11
1
1
0.7
16
1
 Total (MTX, LFN, SSZ)
77
33
342
45
33
24
452
40
 Total PDN
75
32
396
52
105
77
576
51
Two (n, %)
        
< 0.001
 LFN + MTX + PDN
67
29
89
12
12
9
168
15
 LFN + MTX
16
7
26
3
2
1
44
4
 SSZ + MTX + PDN
15
6
31
4
3
2
49
4
 SSZ + LFN + PDN
4
2
38
5
2
1
44
4
 SSZ + MTX
8
3
14
2
1
0.7
23
2
 SSZ + LFN
1
0.4
6
0.7
0
0
7
0.6
 Total (MTX, LFN, SSZ)
111
48
204
27
20
15
335
30
 Total PDN
86
37
158
21
17
12
218
20
Three (n, %)
        
< 0.001
 MTX + SSZ + LFN
6
2
6
0.7
1
0.7
13
1
 MTX + SSZ + LFN + PDN
27
12
21
3
0
0
48
4
 Total (MTX, LFN, SSZ)
33
14
27
3
1
0.7
61
5
 Total PDN
27
12
21
3
0
3
48
4
Previous exposure to MTX (n, %)
191
83
343
45
29
21
563
50
0.03
 Reason for discontinuation
        
 Inefficacy
92
48
212
62
16
55
320
57
 Non-serious adverse event
91
48
121
35
13
45
225
40
 Serious adverse event
8
4
10
3
0
0
18
3
Previous exposure to LFN (n, %)
139
60
338
44
27
20
505
44
< 0.0001
 Reason for discontinuation
        
 Inefficacy
64
46
229
68
16
59
309
62
 Non-serious adverse event
69
50
104
31
11
41
184
36
 Serious adverse event
6
4
5
1
0
0
11
2
Previous exposure to SSZ (n, %)
68
29
157
20
20
15
245
21
0.0001
Reason for discontinuation (n, %)
        
 Inefficacy
17
50
46
63
1
33
64
26
 Non-serious adverse event
18
47
26
36
2
66
46
19
 Serious adverse event
0
0
1
1
0
0
2
0.8
Previous exposure to PDN ( n,%)
188
80
575
75
122
90
885
78
< 0.0001
csDMARD conventional synthetic disease-modifying antirheumatic drug, DMARD disease-modifying antirheumatic drug, MTX methotrexate, LFN leflunomide, SSZ sulfasalazine, PDN prednisone
MTX, LFN, SSZ, and GC prescription patterns at the time of initiation of the first TNF antagonist are shown on Table 3. At initiation of the first TNF antagonist, 977 patients (85 %) were receiving concomitant GCs, MTX, LFN, or SSZ: 15 % (173 patients) received their first TNF antagonist without any concomitant GC or csDMARD, i.e., true monotherapy, and 6 % received their first TNF antagonist with GCs. The drug most commonly used at the time of first TNF antagonist initiation was MTX alone or in combination (58 %), followed by PDN (45 %), LFN (24 %), SSZ (5 %), and anti-malarial agents (1 %). Over 80 % of patients were using non-steroidal anti-inflammatory drugs (NSAIDs). Mean doses of PDN, MTX, and LFN were the commonly used doses. Mean duration of the previous courses with csDMARD therapies before the first TNF antagonist were 2 years for MTX, and less than 1 year for LFN.
Table 3
Prescription patterns of concomitant MTX, LFN, SSZ, and PDN with the first TNF antagonist
Current treatment with synthetic DMARDs plus the first TNF antagonist
First TNF antagonist monotherapy
First TNF antagonist plus one csDMARD
First TNF antagonist plus two or more csDMARDs
Total
P*
 
n
%
n
%
n
%
n
%
 
 
234
21
766
67
136
12
1.136
100
 
None (n, %)
172
73
       
PDN (n, %)
62
26
336
44
115
84
513
45
0.0001
MTX (n, %)
  
364
47
    
< 0.0001
MTX + PDN (n, %)
  
189
25
    
< 0.0001
LFN + PDN (n, %)
  
129
17
    
< 0.0001
LFN (n, %)
  
57
7
    
< 0.0001
SSZ + PDN (n, %)
  
9
1
    
< 0.0001
SSZ (n, %)
  
5
0.6
    
< 0.0001
Combinations (n, %)
        
< 0.0001
 MTX + LFN + PDN
    
66
48
  
 MTX + SSZ + PDN
    
24
17
  
 MTX + SSZ
    
12
9
  
 LFN + CLQ + PDN
    
8
6
  
 MTX + SSZ + HCLQ + PDN
    
5
4
  
 MTX + LFN
    
4
3
  
 Others
    
17
13
  
NSAIDs (n, %)
201
86
673
88
127
93
1.013
88
0.09
 
Mean
SD
Mean
SD
Mean
SD
Mean
SD
 
Current PDN dose, mg/day (mean ± SD)
10.7
±5.6
9.6
±6.7
10.0
±7.5
9.8
±6.6
0.002
Current MTX dose, mg/week (mean ± SD)
0
0
15.3
±4.8
16.0
4.7
15.5
4.8
0.08
Duration of current treatment with MTX (months) (mean ± SD)
  
31.0
32.2
21.5
23.6
27.6
28.6
0.0001
Current LFN dose, mg/day (mean ± SD)
0
0
18.8
3.1
15.6
4.9
17.8
4.0
< 0.0001
Duration of current treatment with LFN (months) (mean ± SD)
  
12.9
19.0
9.4
9.9
12.0
16.5
0.5
Follow up, (years) (mean ± SD)
2.1
1.0
1.8
1.0
1.6
1.1
1.8
1.1
0.0001
csDMARD conventional synthetic disease-modifying antirheumatic drug, DMARD disease-modifying antirheumatic drug, MTX methotrexate, LFN leflunomide, SSZ sulfasalazine, PDN prednisone, NDAID non-steroidal anti-inflammatory drug
Effectiveness data are shown in Table 4. With regard to disease activity, all treatment groups showed clinically significant improvements in final vs. baseline DAS. Physical function data assessed by the HAQ scores were significantly different between groups at the final vs. the baseline assessment, and the TNF antagonist plus two or more csDMARDs group had the poorest physical function. The percentage of patients with completed visits were 56 %, 51 %, and 60 % in each group, respectively; p = 0.4. The number and causes of loss of follow up were the same in the three groups except for adverse events. The adverse events were more frequent in the patients treated with the first TNF antagonist plus one csDMARD, with marginal statistical significance. Loss of follow up due to patient´s decision were due to intention to become pregnant, change of city, addition of other inter-current disease, and in some cases the reasons were unknown.
Table 4
Rresponse in patients with rheumatoid arthritis at 6 months and follow-up data
Treatment with synthetic DMARDs
First TNF antagonist monotherapy
First TNF antagonist plus one csDMARD
First TNF antagonist plus two or more csDMARDs
Total
Pp*
n, %
234
21
766
67
136
12
1.136
100
 
Baseline DAS28 score (mean ± SD)
5.9
1.0
5.7
1.1
5.7
1.0
5.8
1.1
0.2
Final DAS28 score (mean ± SD)
3.56
1.54
3.39
1.52
3.6
1.3
3.4
1.5
0.8
Difference in DAS28 score (mean ± SD)
1.6
0.7
1.6
0.7
1.7
0.6
1.6
0.7
0.4
Baseline HAQ score (mean ± SD)
1.40
0.58
1.44
0.65
1.63
0.59
1.461
0.633
0.002
Final HAQ score (mean ± SD)
0.83
0.71
0.93
0.76
1.08
0.74
0.930
0.752
0.005
Difference in HAQ score (mean ± SD)
0.60
0.69
0.51
0.75
0.60
0.70
0.545
0.734
0.1
Follow up
 Completed last visit (n, %)
132
56
391
51
82
60
605
53
0.46
 Loss of follow up (n, %)
102
44
375
49
54
40
531
47
0.07
  Reasons
         
  Adverse event
33
32
174
46
16
30
223
42
0.001
  Inefficacy
30
29
87
23
12
22
129
24
0.5
  Patient decision
7
7
15
4
3
5
25
5
0.6
  Unknown
32
32
99
27
23
43
154
29
0.4
*P value for between-group comparisons. Differences between groups in baseline vs. final disease activity score in 28 joints (DAS) and health assessment questionnaire (HAQ) scores were statistically significant, p < 0.001. csDMARD conventional synthetic disease-modifying antirheumatic drug, DMARD disease-modifying antirheumatic drug
Percentages of patients with good EULAR response were 29 % in the first TNF antagonist monotherapy group, 29 % in the first TNF antagonist plus one csDMARD group, and 43 % in the first TNF antagonist plus two or more csDMARDs group (p = 0.07). Percentages of patients with moderate EULAR response were 50 %, 52 %, and 51 %, respectively, and marginal statistically significant differences were observed (p = 0.07) (Fig. 2). Percentages of patients with a final HAQ score ≤ 1 were 73 %, 63 %, and 54 %, respectively (p = 0.01) (Fig. 3).
On crude analysis of drug survival, for TNF antagonist survival the mean (25th to 75th percentiles) was 33.3 (16.8–40.7) months in the TNF antagonist monotherapy group, 30.4 (16.6–41.4) months in the TNF antagonist plus one csDMARD group, and 34.2 (8.1–40.8) months in the TNF antagonist plus two or more csDMARDs group (p = 0.07) (Fig. 4). When this analysis was adjusted for age, gender, number of comorbidities, and disease duration, thus, controlling for these potential confounders, there were statistical differences in survival between the three groups, with mean TNF survival of 16.7 (11.8–24.1), 20.1 (11.3–31.3), and 10,5 (6.1–29.6) months, respectively (p < 0.001) (Fig. 5).
Finally, in the logistic regression the model that best explained a good EULAR response included the baseline DAS28 and the 6-month DAS28 (p <0.0001 and R 2 0.8). The model was not modified after adjustment for age, gender, disease duration, and treatment group. The model followed a similar pattern using the different techniques for handle missing values (data not shown).

Discussion

This was an observational, prospective, multicentre study of a cohort of patients with RA treated with their first TNF antagonist in standard clinical practice within the Andalusian Health Services. The registry included 90 % of prescriptions of biologic agents issued in this community. We observed differences in the characteristics of patients who received their first TNF antagonist in monotherapy or in combination with csDMARDs, and found that prescriptions were largely influenced not only by accessibility to biological drugs within the health system, but also by the clinical characteristics of the patients, particularly comorbidities and drug intolerance, and by the previous experience of the rheumatologist [14].
Patients in the three groups had differences in the type and number of comorbidities. As we know since the time of Hippocrates, there are patients, not diseases, and this is the art of medicine reflected in DMARDs prescription patterns, in spite of treatment schedules that are defined in Spain in summaries of product characteristics and their respective Clinical Practice Guidelines [1, 2], (https://​www.​nice.​org.​uk/​guidance/​cg79/​chapter/​Recommendations#pharmacological-management), [3, 19]. The data are in agreement with data recently reported by other registries and publications [5, 814].
In our study, before initiation of their first TNF antagonist, only 1 % of patients had not received GCs, MTX, LFN, SSZ, and/or CLQ/HCLQ. PDN, either as monotherapy or in combination with other csDMARDs was the most commonly prescribed drug (78 %), followed by MTX (50 %), and LFN (44 %); SSZ (21 %) was only used in a small number of cases. Rheumatologists prefer combinations of csDMARDs with PDN, and the most frequently prescribed for combination therapy is MTX [47, 1215]. This high level of prescription of PDN at low or very low doses, in monotherapy or in combination with csDMARDs or a TNF antagonist, is a consequence of the effectiveness of this drug for reducing signs and symptoms of the disease. The advantages of PDN include improved patient-reported outcomes and lower disease activity levels, its benefit on radiological evidence of disease progression, and its very favourable efficacy/toxicity ratio producing very low rates of serious adverse events, and no serious adverse events related to this low or very low dose.
According to the literature, low or very low doses of steroids are used the same way as any other csDMARDs, and should be considered as such [812, 16, 17]. The frequency of baseline use of steroids found in several registries of patients with RA receiving bDMARDs are the following: 27 % in the Swiss Registry [9], 22 % in the Danish Registry [10], and in accordance with a review including various countries [11], figures range from 38 % to 84 % in Germany (84 %), Spain (52 %), Sweden (51 %), UK (44 %), and USA (38 %) (CORRONA database). Considering these data, TNF antagonists are not really given in monotherapy, due to the high frequency of GC use in patients treated with TNF antagonists in monotherapy. Unfortunately, this study did not include data on radiographic progression, so the real value of GCs as csDMARDs cannot be assessed.
The low use of SSZ in Spain may be explained by the enteric coating of the formulation. This is different from the coating used in other European countries, and its efficacy is lower than that reported in studies conducted in Northern Europe. As a consequence, SSZ is only used after other csDMARDs, namely MTX, LFN, and CLQ/HCLQ [21]. Our results may be influenced by our requirement that during the period in which the study was conducted, patients had to have failed to respond to at least two csDMARDs in order to be prescribed a bDMARD [3, 19]. A lack of efficacy, followed by mild csDMARD toxicity (30–40 %), were the primary reasons for the participating rheumatologists to request bDMARDs. Severe toxicity was very infrequent (< 3 %).
With regard to GCs and csDMARDs prescription patterns, at the time of initiation of the first TNF antagonist most patients were receiving monotherapy or combination therapy with MTX, followed by PDN, LFN, and SSZ. The decreased frequency of GC treatment, from 78 % to 45 %, needs to be emphasized. Only 15 % of patients received the first TNF antagonist without concomitant csDMARDs or GCs. Almost one third (27 %) of patients who received their first TNF antagonist in monotherapy were being treated with PDN as the sole medication. Again, the data show that low or very low doses of PDN were a commonly used treatment at the time of initiation of the first TNF antagonist. GC, MTX, and LFN doses were similar to those recorded in other registries and observational studies [5, 812, 14]. It should be noted that a smaller subgroup of patients (those who received their first TNF antagonist plus two or more csDMARDs) were treated with different GCs, and csDMARD combinations. The most commonly used regimen was combination treatment with PDN plus one or more csDMARDs.
It should be emphasized that patients achieved adequate responses irrespective of the treatment they received. There were no differences between groups in improvement in disease activity after starting the biologic agent, but poorer levels of physical function were observed in the baseline and final assessments of those patients treated with the first TNF antagonist plus two or more csDMARDs. After adjustment for potential confounders, the survival analysis showed the best survival rates in the group treated with the first TNF antagonist plus one csDMARD.
This study had advantages that should also be pointed out. First, the study was based on data from standard clinical practice that included 90 % of biologic drug prescriptions. This prospective study was conducted in the setting of specialized rheumatology care and was specially designed to determine prescription patterns of GCs, csDMARDs, and the first TNF antagonist. Furthermore, patients’ clinical characteristics were similar to those observed in other European registries of patients with RA.
Unfortunately, due to the large number of patients lost to follow up, effectiveness data should be viewed with reservation, even though no statistically or clinically relevant differences were found in a comparison of sociodemographic and RA disease characteristics between patients lost to follow up vs. patients who continued (data not shown). The different statistical techniques used to handle missing values also showed different results. We know that the effectiveness data are not fully robust, as they might reflect a cohort of survivors. Another limitation is that, currently, many of these patients who cannot receive csDMARDs are treated with TCZ; thus, the external validity of the data is limited to patients who cannot receive TCZ. The third handicap is the lack of evaluation of radiographic outcome.

Conclusions

Patients receiving the first TNF antagonist monotherapy, the first TNF antagonist plus one csDMARD, and the first TNF antagonist plus two or more csDMARDs are patients with different comorbidities and disease characteristics, which influence the type of drugs they receive. Low or very low doses of GCs are frequently used. Combination treatments with PDN plus csDMARDs are more frequently used by rheumatologists, compared to csDMARD monotherapy or other combinations. In most cases, these agents are discontinued due to lack of efficacy, requiring the introduction of TNF antagonist therapy. With the first TNF antagonist, PDN in combination with one csDMARD is used in almost half of the cases. Monotherapy with a TNF antagonist without PDN or csDMARDs is uncommon. In spite of their different characteristics, patients receiving the first TNF antagonist monotherapy, the first TNF antagonist plus one csDMARD, and the first TNF antagonist plus two or more csDMARDs obtain clinical responses in terms of activity and physical function measured by an acceptable HAQ score. Survival differed in the three groups of patients.

Acknowledgements

We want to acknowledge the contribution of the Andalusian Registry of Biologic Therapies Working Group; the Rheumatologists and their co-investigators: Dolores Bejarano Rojas, Pharmaceutical Supplies and Services, Central Services, Andalucian Health Services, Seville, Spain. Hospital Regional Virgen del Rocío: Clara Aguilera Cross, Rheumatologist; Juan Povedano Gómez, Rheumatologist, Seville, Spain. Hospital Regional Virgen de las Nieves: Miguel Angel Ferrer, Rheumatologist; Antonio García, Rheumatologist; Alfonso González Utrilla, Rheumatologist, Granada, Spain. Hospital Regional Virgen Macarena: Carmen Vargas Lebrón, Rheumatologist; Carmen López Martín, Pharmaceutical, Seville, Spain. Hospital Regional Reina Sofía: Maria del Carmen Castro Villegas, Rheumatologist; Eduardo Collantes Estevez, Rheumatologist, Córdoba, Spain. Hospital Regional Carlos Haya: Sara Manrique Arija, Rheumatologist; Antonio Fernández Nebro, Rheumatologist, Málaga, Spain.
Hospital de Especialidades Virgen de Valme: Julia Uceda, Rheumatologist; Jose Luis Marenco de la Fuente, Rheumatologist. Seville, Spain. Hospital de Especialidades Puerta del Mar: Inmaculada Macías Fernández, Rheumatologist; Sergio García Pérez, Rheumatologist. Cadiz, Spain. Hospital de Especialidades de Jerez de la Frontera: Carmen Carrasco Cubero, Rheumatologist; Raúl Menor Almagro, Rheumatologist; Jose Pérez Venegas, Rheumatologist, Jerez de la Frontera, Spain. Hospital de Especialidades San Cecilio: Enrique Raya, Rheumatologist; Juan Salvatierra, Rheumatologist; Jesús Dominguez, Rheumatologist, Granada, Spain. Hospital de Especialidades Juan Ramón Jiménez: Manuel Riesco Diaz, Rheumatologist, Huelva, Spain. Hospital de Especialidades Torrecárdenas Julio García Feito, Rheumatologist, Almería, Spain. Complejo Hospitalario de Jaén: Celia García Giménez, Rheumatologist, Jaen, Spain. Hospital de Especialidades Virgen de la Victoria: Rosa García Portales, Rheumatologist, Málaga, Spain.

Funding

This work was funding by a limited grant from the Andalucian Foundation of Rheumatology.

Availability of data and materials

The clinical data for the patients and the database were verified by two of the investigators (BHC and EMS). The data were introduced into a excel file and then they were converted into a STATA file, *.dta. These data files are kept in an e-file in the Rheumatology Service.

Authors’ contributions

BHC conceived of the study, participated in the design and coordination of the study, did the statistical analysis, verified the clinical data and the database, helped to draft the manuscript, and created the webpage and the database. EMS participated in the design and coordination of the study, verified the clinical data and the database, completed and cleaned the final database, and helped to draft the manuscript. RCC conceived of the study, participated in the design and coordination of the study, did the statistical analysis, helped to draft the manuscript, and created the webpage and the database. FNS conceived of the study, participated in the design and coordination of the study, and helped to draft the manuscript. All the authors participated in the interpretation of results. BHC and RCC drafted the manuscript. All authors read and approved the final manuscript.

Competing interests

Blanca Hernández Cruz is a rheumatologist at the Rheumatology Clinical Unit, Virgen de la Macarena University Hospital, Andalusian Health Services, and a researcher at the HUVM i + D + I. She has declared no conflicts of interest for this paper. In the past year she conducted activities as investigator, adviser, and lecturer for the Spanish Society of Rheumatology, and for the Andalusian Society of Rheumatology. She is a regular speaker in activities organised by patients’ associations. She has been a lecturer for Lilly, Amgen, Bristol, Gebro Pharma, and Roche; has received financial support from private bodies (Andalusian Society of Rheumatology) for her participation in research projects and has received financial support from the following pharmaceutical companies: Lilly, Amgen, Abbie, Bristol, Gebro Pharma, and Roche for her participation in research projects. Esther Márquez Saavedra has declared no conflicts of interest. Federico Navarro Sarabia has declared no conflicts of interest. Rafael Caliz Caliz has declared no conflicts of interest.
Not applicable.
The study was approved by the Ethical and Human Research Committee of the Virgen de la Macarena Hospital Area (number 948), on 10 December 10 2004, and was conducted in accordance with Good Clinical Practice Guidelines. The Ethical Approval Committee letter is included in the submission.
In agreement with the local and European regulation, this study was observational and in real-life clinical practice, so written consent to participate was not required. Confidentiality was maintained as local protection of data laws demand.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted 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. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M, Emery P, Kvien TK, Navarro-Compán MV, Oliver S, Schoels M, Scholte-Voshaar M, Stamm T, Stoffer M, Takeuchi T, Aletaha D, Andreu JL, Aringer M, Bergman M, Betteridge N, Bijlsma H, Burkhardt H, Cardiel M, Combe B, Durez P, Fonseca JE, Gibofsky A, Gomez-Reino JJ, Graninger W, Hannonen P, Haraoui B, Kouloumas M, Landewe R, Martin-Mola E, Nash P, Ostergaard M, Östör A, Richards P, Sokka-Isler T, Thorne C, Tzioufas AG, van Vollenhoven R, de Wit M, van der Heijde D. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75(1):3–15. doi:10.1136/annrheumdis-2015-207524. PubMed PMID: 25969430; PubMed Central PMCID: PMC4717393.CrossRefPubMed Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M, Emery P, Kvien TK, Navarro-Compán MV, Oliver S, Schoels M, Scholte-Voshaar M, Stamm T, Stoffer M, Takeuchi T, Aletaha D, Andreu JL, Aringer M, Bergman M, Betteridge N, Bijlsma H, Burkhardt H, Cardiel M, Combe B, Durez P, Fonseca JE, Gibofsky A, Gomez-Reino JJ, Graninger W, Hannonen P, Haraoui B, Kouloumas M, Landewe R, Martin-Mola E, Nash P, Ostergaard M, Östör A, Richards P, Sokka-Isler T, Thorne C, Tzioufas AG, van Vollenhoven R, de Wit M, van der Heijde D. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75(1):3–15. doi:10.​1136/​annrheumdis-2015-207524. PubMed PMID: 25969430; PubMed Central PMCID: PMC4717393.CrossRefPubMed
2.
Zurück zum Zitat Singh JA, Saag KG, Bridges Jr SL, Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, Curtis JR, Furst DE, Parks D, Kavanaugh A, O'Dell J, King C, Leong A, Matteson EL, Schousboe JT, Drevlow B, Ginsberg S, Grober J, St Clair EW, Tindall E, Miller AS, McAlindon T. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol. 2016;68(1):1–26. doi:10.1002/art.39480.CrossRefPubMed Singh JA, Saag KG, Bridges Jr SL, Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, Curtis JR, Furst DE, Parks D, Kavanaugh A, O'Dell J, King C, Leong A, Matteson EL, Schousboe JT, Drevlow B, Ginsberg S, Grober J, St Clair EW, Tindall E, Miller AS, McAlindon T. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol. 2016;68(1):1–26. doi:10.​1002/​art.​39480.CrossRefPubMed
4.
Zurück zum Zitat Visser K, Katchamart W, Loza E, Martinez-Lopez JA, Salliot C, Trudeau J, Bombardier C, Carmona L, van der Heijde D, Bijlsma WJ, Boumpas DT, Canhao H, Edwards CJ, Hamuryudan V, Kvien TK, Leeb BF, Martín Mola E, Mielants H, Müller-Ladner U, Murphy G, Østergaard M, Pereira JA, Ramos-Remus C, Valentini G, Zochling J, Dougados M. Multinational evidence-based recommendations for the use of methotrexate in rheumatic disorders with a focus on rheumatoid arthritis: integrating systematic literature research and expert opinion of a broad international panel of rheumatologists in the 3E Initiative. Ann Rheum Dis. 2009;68:1086–93. doi:10.1136/ard.2008.094474.CrossRefPubMed Visser K, Katchamart W, Loza E, Martinez-Lopez JA, Salliot C, Trudeau J, Bombardier C, Carmona L, van der Heijde D, Bijlsma WJ, Boumpas DT, Canhao H, Edwards CJ, Hamuryudan V, Kvien TK, Leeb BF, Martín Mola E, Mielants H, Müller-Ladner U, Murphy G, Østergaard M, Pereira JA, Ramos-Remus C, Valentini G, Zochling J, Dougados M. Multinational evidence-based recommendations for the use of methotrexate in rheumatic disorders with a focus on rheumatoid arthritis: integrating systematic literature research and expert opinion of a broad international panel of rheumatologists in the 3E Initiative. Ann Rheum Dis. 2009;68:1086–93. doi:10.​1136/​ard.​2008.​094474.CrossRefPubMed
5.
Zurück zum Zitat Zink A, Listing J, Kary S, Ramlau P, Stayanova-Scholz M, Babinsky K, van Hinueber A, Gromnica-Ihle E, Wassenberg S, Antoni C, Herzer P, Kekow J, Shneider M, Rau R. Treatment continuation in patients receiving biological agents or conventional DMARD therapy. Ann Rheum Dis. 2005;64:1274–9. doi:10.1136/ars.2004.031476.CrossRefPubMedPubMedCentral Zink A, Listing J, Kary S, Ramlau P, Stayanova-Scholz M, Babinsky K, van Hinueber A, Gromnica-Ihle E, Wassenberg S, Antoni C, Herzer P, Kekow J, Shneider M, Rau R. Treatment continuation in patients receiving biological agents or conventional DMARD therapy. Ann Rheum Dis. 2005;64:1274–9. doi:10.​1136/​ars.​2004.​031476.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Katchamart W, Trudeau J, Phumethum V, Bombardier C. Efficacy and toxicity of methotrexate (MTX) monotherapy versus MTX combination therapy with non-biological disease-modifying antirheumatic drugs in rheumatoid arthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2009;68:1105–12. doi:10.1136/ard.2008.099861.CrossRefPubMed Katchamart W, Trudeau J, Phumethum V, Bombardier C. Efficacy and toxicity of methotrexate (MTX) monotherapy versus MTX combination therapy with non-biological disease-modifying antirheumatic drugs in rheumatoid arthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2009;68:1105–12. doi:10.​1136/​ard.​2008.​099861.CrossRefPubMed
7.
Zurück zum Zitat Wevers-de Boer K, Visser K, Heimans L, Ronday HK, Molenaar E, Groenendael JH, et al. Remission induction therapy with methotrexate and prednisone in patients with early rheumatoid and undifferentiated arthritis (the IMPROVED study). Ann Rheum Dis. 2012;71(9):1472–7. PubMed: 22402145.CrossRefPubMed Wevers-de Boer K, Visser K, Heimans L, Ronday HK, Molenaar E, Groenendael JH, et al. Remission induction therapy with methotrexate and prednisone in patients with early rheumatoid and undifferentiated arthritis (the IMPROVED study). Ann Rheum Dis. 2012;71(9):1472–7. PubMed: 22402145.CrossRefPubMed
8.
Zurück zum Zitat Soliman MM, Ashcroft DM, Watson KD, Lunt M, Symmons DP, Hyrich KL, British Society for Rheumatology Biologics Register. 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.CrossRefPubMedPubMedCentral Soliman MM, Ashcroft DM, Watson KD, Lunt M, Symmons DP, Hyrich KL, British Society for Rheumatology Biologics Register. 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.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Gabay C, Riek M, Scherer A, Finckh A, on behalf of the SCQM collaborating physicians. Effectiveness of biologic DMARDs in monotherapy versus in combination with synthetic DMARDs in rheumatoid arthritis: data from the Swiss Clinical Quality Management Registry. Rheumatology. 2015;54:16641672. doi:10.1093/rheumatology/kev019. Advance Access publication 27 April 2015.CrossRef Gabay C, Riek M, Scherer A, Finckh A, on behalf of the SCQM collaborating physicians. Effectiveness of biologic DMARDs in monotherapy versus in combination with synthetic DMARDs in rheumatoid arthritis: data from the Swiss Clinical Quality Management Registry. Rheumatology. 2015;54:16641672. doi:10.​1093/​rheumatology/​kev019. Advance Access publication 27 April 2015.CrossRef
10.
Zurück zum Zitat Jørgensen TS, Kristensen LE, Christensen R, et al. Effectiveness and drug adherence of biologic monotherapy in routine care of patients with rheumatoid arthritis: a cohort study of patients registered in the Danish biologics registry. Rheumatology (Oxford). 2015 [Epub ahead of print]. Jørgensen TS, Kristensen LE, Christensen R, et al. Effectiveness and drug adherence of biologic monotherapy in routine care of patients with rheumatoid arthritis: a cohort study of patients registered in the Danish biologics registry. Rheumatology (Oxford). 2015 [Epub ahead of print].
11.
Zurück zum Zitat Curtis JR, Jain A, Askling J, et al. A comparison of patient characteristics and outcomes in selected European and U.S. rheumatoid arthritis registries. Semin Arthritis Rheum. 2010;40:2–14.CrossRefPubMedPubMedCentral Curtis JR, Jain A, Askling J, et al. A comparison of patient characteristics and outcomes in selected European and U.S. rheumatoid arthritis registries. Semin Arthritis Rheum. 2010;40:2–14.CrossRefPubMedPubMedCentral
12.
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 Jt Dis. 2008;66:77–85.PubMed 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 Jt Dis. 2008;66:77–85.PubMed
13.
Zurück zum Zitat Buckley F, Finckh A, Huizinga TW, Dejonckheere F, Jansen JP. Comparative efficacy of novel DMARDs as monotherapy and in combination with methotrexate in rheumatoid arthritis patients with inadequate response to conventional DMARDs: a network meta-analysis. J Manag Care Spec Pharm. 2015;21:409–23.CrossRefPubMed Buckley F, Finckh A, Huizinga TW, Dejonckheere F, Jansen JP. Comparative efficacy of novel DMARDs as monotherapy and in combination with methotrexate in rheumatoid arthritis patients with inadequate response to conventional DMARDs: a network meta-analysis. J Manag Care Spec Pharm. 2015;21:409–23.CrossRefPubMed
14.
Zurück zum Zitat Hyrich K, Symmons D, Watson K, Silman A, BSRBR Control Centre Consortium; British Society for Rheumatology Biologics Register. Baseline comorbidity levels in biologic and standard DMARD treated patients with rheumatoid arthritis: results from a national patient register. Ann Rheum Dis. 2006;65:895–8.CrossRefPubMed Hyrich K, Symmons D, Watson K, Silman A, BSRBR Control Centre Consortium; British Society for Rheumatology Biologics Register. Baseline comorbidity levels in biologic and standard DMARD treated patients with rheumatoid arthritis: results from a national patient register. Ann Rheum Dis. 2006;65:895–8.CrossRefPubMed
15.
Zurück zum Zitat Smolen JS, Landewé R, Breedveld F, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying anti rheumatic drugs. Ann Rheum Dis. 2010;69:964–75.CrossRefPubMedPubMedCentral Smolen JS, Landewé R, Breedveld F, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying anti rheumatic drugs. Ann Rheum Dis. 2010;69:964–75.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Wailoo A, Hernández Alava M, Scott IC, Ibrahim F, Scott DL. Cost-effectiveness of treatment strategies using combination disease-modifying anti-rheumatic drugs and glucocorticoids in early rheumatoid arthritis. Rheumatology Oxford. 2014;53:1773–7.CrossRefPubMed Wailoo A, Hernández Alava M, Scott IC, Ibrahim F, Scott DL. Cost-effectiveness of treatment strategies using combination disease-modifying anti-rheumatic drugs and glucocorticoids in early rheumatoid arthritis. Rheumatology Oxford. 2014;53:1773–7.CrossRefPubMed
17.
Zurück zum Zitat Kavanaugh A, Wells AF. Benefits and risks of low-dose glucocorticoid treatment in the patient with rheumatoid arthritis. Rheumatology Oxford. 2014;53:1742–51.CrossRefPubMedPubMedCentral Kavanaugh A, Wells AF. Benefits and risks of low-dose glucocorticoid treatment in the patient with rheumatoid arthritis. Rheumatology Oxford. 2014;53:1742–51.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–24.CrossRefPubMed Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–24.CrossRefPubMed
19.
Zurück zum Zitat Rodríguez Valverde V, Álvaro Gracia JM, Andrew JL, Batlle E, Tornero JL. Segunda actualización del Consenso de la Sociedad Española de Reumatología sobre la Terapia Biológica en la Artritis Reumatoide. Rev Esp Reumatol. 2004;31:394–401. Rodríguez Valverde V, Álvaro Gracia JM, Andrew JL, Batlle E, Tornero JL. Segunda actualización del Consenso de la Sociedad Española de Reumatología sobre la Terapia Biológica en la Artritis Reumatoide. Rev Esp Reumatol. 2004;31:394–401.
21.
Zurück zum Zitat Blanco FJ, Ballina J, Carbonell J, et al. Estudio descriptivo de la utilización de los FAMES en los pacientes con artritis reumatoide o artritis persistente que inician tratamiento farmacológico en España. (ESTUDIO FIRST). Reumatol Clin. 2011;7:88–9.CrossRefPubMed Blanco FJ, Ballina J, Carbonell J, et al. Estudio descriptivo de la utilización de los FAMES en los pacientes con artritis reumatoide o artritis persistente que inician tratamiento farmacológico en España. (ESTUDIO FIRST). Reumatol Clin. 2011;7:88–9.CrossRefPubMed
Metadaten
Titel
Comparative effectiveness of treatment with the first TNF antagonist in monotherapy, the first TNF antagonist plus one conventional synthetic disease-modifying antirheumatic drug, and the first TNF antagonist plus two or more conventional synthetic disease-modifying antirheumatic drugs in patients with rheumatoid arthritis
verfasst von
Blanca Hernández-Cruz
Esther Márquez-Saavedra
Rafael Caliz-Caliz
Federico Navarro-Sarabia
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2016
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-016-1137-4

Weitere Artikel der Ausgabe 1/2016

Arthritis Research & Therapy 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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