Notes
Article history
The research reported in this issue of the journal was funded by the HTA programme as project number 06/303/84. The contractual start date was in April 2007. The draft report began editorial review in February 2013 and was accepted for publication in September 2013. The authors have been wholly responsible for all data collection, analysis and interpretation, and for writing up their work. The HTA editors and publisher have tried to ensure the accuracy of the authors’ report and would like to thank the reviewers for their constructive comments on the draft document. However, they do not accept liability for damages or losses arising from material published in this report.
Declared competing interests of authors
none
Permissions
Copyright statement
© Queen’s Printer and Controller of HMSO 2014. This work was produced by Scott et al. under the terms of a commissioning contract issued by the Secretary of State for Health. This issue may be freely reproduced for the purposes of private research and study and extracts (or indeed, the full report) may be included in professional journals provided that suitable acknowledgement is made and the reproduction is not associated with any form of advertising. Applications for commercial reproduction should be addressed to: NIHR Journals Library, National Institute for Health Research, Evaluation, Trials and Studies Coordinating Centre, Alpha House, University of Southampton Science Park, Southampton SO16 7NS, UK.
Chapter 1 Introduction
Rheumatoid arthritis
Key impacts
Rheumatoid arthritis (RA), one of the commonest disabling diseases in the UK, remains a major health-care problem. 1–3 It affects almost 1% of UK adults and is more common in women. There are two peak ages of onset, early adulthood (mainly women) and later life (equal sex distribution). There are internationally accepted classification criteria for RA; from time to time these been revised and modernised. 3–6
Its main impacts are increasing disability and reduced quality of life. 7 Both are substantial and persistent and reflect the combined effects of persisting joint inflammation, progressive joint damage and extra-articular features of RA. 8 Another significant impact of RA is reduced life expectancy, which is mainly due to associated comorbidities such as coronary artery disease. 9 The final major impact of RA is the substantial costs in terms of medical and social care and lost employment. 10
Disease course and outcomes
The primary clinical feature of RA is chronic, usually persistent, inflammatory synovitis, initially mainly affecting the small joints of the hands and feet but subsequently spreading to involve multiple other joints. 7 Without adequate treatment many patients will develop joint damage, classically erosions, but also joint space loss and secondary osteoarthritis. 11 In addition, RA may be associated with extra-articular features, such as nodules and interstitial lung disease,8 and with comorbidities, such as the increased risk of cardiovascular disease and infection. 12
Diagnosis combines clinical features with laboratory tests such as acute phase markers [erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP)],13 rheumatoid factor, anticyclic citrullinated peptide (anti-CCP) antibodies14–16 and imaging [ultrasound, magnetic resonance imaging (MRI) and/or radiography]. 17–19 Definitive differentiation from other forms of inflammatory arthritis is difficult in early arthritis but usually uncontroversial in established disease.
The outcome of RA is highly variable, ranging from mild disease with limited impact on a patient’s life to severe unremitting disease unresponsive to treatment. Some features are known genetically and epidemiologically to be associated with a poorer outcome, including specific human leucocyte antigen genotypes, smoking and the presence of anti-CCP antibodies. 20–22 However, it has proved difficult to develop an outcome predictor at the level of the individual patient, which would be required to develop tailor-made individual treatment regimens.
Disease costs
Rheumatoid arthritis results in high medical and social costs,23 with drug costs a significant part of the economic cost. Conventional drugs are relatively inexpensive whereas newer biological agents are very expensive; over time, drug costs have risen substantially. A second cost component is other medical care. These costs are modest in the short term but rise substantially when surgical treatment or supportive long-term medical treatment is needed for disabling severe RA or for comorbid disease. The final costs are societal costs. These include loss of work, support from family and carers and costs of care within the community. These societal costs usually exceed medical expenses and rise with disease duration and severity.
Historically, in the period before biological treatments were available, the direct and indirect costs were estimated to be in the region of £55–70M per million of the population,24 with a total disease cost of £4B for the UK as a whole. 25 Since the introduction of biological treatments, drug costs have increased substantially. A report by the National Audit Office in 2009 estimated that RA costs the NHS around £560M a year in health-care costs, with the majority of this in the acute sector. 26 This report estimated that the costs to the NHS of biologics for treating RA were around £160M annually. As biologics prescribing for RA has continued to increase, the current costs are likely to be substantially higher but may be balanced by reductions in other medical costs, such as orthopaedic interventions for RA, if high-cost drug treatments improve medical outcomes. The National Audit Office report also estimated that the additional cost to the UK economy of sick leave and work-related disability for RA is £1.8B a year.
Assessments
Assessments in RA mainly look at joint inflammation. Clinical-based assessments include swollen and tender joint counts and global assessment, which estimates overall disease activity and health status. Standard joint counts focus on 28 joints in the hands, upper limbs, and knees. Some experts prefer extended 66 and 68 joint counts; these include the feet. Laboratory measures include the ESR, CRP or both. Patient-based measures span pain, global assessment and disability. 27–29 The Health Assessment Questionnaire (HAQ) measures disability. 30 Other areas, such as fatigue and depression,31,32 are very relevant to patients but are not always formally assessed. Patient-based measures are especially important because they measure an individual’s perspective of the burden of their RA.
A number of combined indices amalgamate individual assessments. A widely used combined index is the Disease Activity Score for 28 Joints (DAS28), which combines the numbers of swollen and tender joints (hands, arms and knees) out of a total of 28, a patient’s global assessment and the ESR to indicate a patient’s current status. 33 As calculating the DAS28 involves a complex mathematical formula, simplified variants have been devised. 34 The Simplified Disease Activity Index uses the number of tender and swollen joints (out of a total of 28), doctors’ and patients’ global assessments and CRP level. The Clinical Disease Activity Index is similar but omits CRP level. The American College of Rheumatology (ACR) improvement criteria, which gauge change in status in clinical trials, include falls in joint counts and several other measures (patients’ and doctors’ global assessments, ESR, pain and HAQ). They record 20% (ACR20), 50% (ACR50) and 70% (ACR70) improvements in five of the seven measures. 35
Juxta-articular erosions characterise progressive, established RA and are usually irreversible. They can be readily identified on radiographic images of the hands and feet. Two typical erosions are sufficient for diagnosis. 36 Extensive damage seen on radiographs suggests that RA is inadequately controlled. Rapid progression of joint damage needs intensive treatment. 37 Several scoring systems are used to quantify damage seen on radiographs in research studies. Although new imaging modalities such as ultrasound and MRI can assess structural changes, they are not yet widely used except in research. 38
Treatment goals
The overall treatment goal is making patients feel better and minimising the impact of RA on their lives. 39 The main immediate treatment goal over the last two decades has been to reduce disease activity. Reducing joint and systemic inflammation is beneficial in itself. Crucially, it is also associated with other benefits including decreased disability, improved quality of life and reduced progression of joint damage. A dominant theme has been to treat patients with active RA; in the main, current treatments mean that few patients now have persisting active disease.
More recently there has been a shift towards making remission the main goal. An ideal treatment would result in the majority of patients achieving remission with no active joint inflammation and no functional deterioration or erosive progression. 40 Although 10–50% of patients with early RA can achieve remission,41 only a small minority of established RA patients achieve sustained remission. An associated difficulty in determining the frequency of remission depends on how it is defined and the intensity of treatment. 42
Relatively cheap, readily available disease-modifying antirheumatic drugs (DMARDs) such as methotrexate have made major inroads into managing active RA. DMARDs were initially given as monotherapies but in recent years there has been greater emphasis on using combinations of two or more DMARDs as this has been shown to be more effective in disease control. 43
Since the mid-1990s a new treatment approach has been developed – the use of targeted biological treatments. They are usually given in combination with methotrexate or other DMARDs. Biologics have revolutionised the treatment of severe RA, for which they appear highly effective. A major limiting factor is their high cost. 44
Reducing disease activity appears a clear-cut well-defined goal. However, the degree of reduction required for a good ultimate outcome is not yet known. Intensive treatment aimed at inducing remission45 appears an inevitable next step. However, it is not clear whether or not this is appropriate for every patient. Furthermore, there remains uncertainty about the appropriate definition of remission in RA. 46
Synopsis of specific drug treatment
Conventional disease-modifying antirheumatic drugs
Disease-modifying antirheumatic drugs are a diverse range of drugs. 47 They form a single group because they both improve symptoms and also, to a greater or lesser extent, modify the course of the disease. This means that they reduce the progression of erosive joint damage and decrease disability. 48,49
Many drugs have some features of DMARDs but only a few have been accepted into clinical practice. The use of DMARDs varies, with a small number being particularly favoured. The current situation is summarised in Table 1. At present, methotrexate is the dominant DMARD because of its greater efficacy and retention compared with other DMARDs. 50 As the most widely used DMARD, methotrexate is now considered by regulatory agencies as a benchmark against which new agents must be tested. The majority of RA patients treated with DMARDs in most UK specialist units either are currently taking or have previously received methotrexate. Sulfasalazine, leflunomide and hydroxychloroquine (the last largely as part of a combination regime) are the only other DMARDs used to any appreciable extent in the UK. 51
Theme | DMARDs |
---|---|
Range of DMARDs | |
Commonly used | Methotrexate, leflunomide, sulfasalazine |
Infrequently used | Hydroxychloroquine/chloroquine, injectable gold, azathioprine |
Rarely used | Ciclosporin, auranofin, cyclophosphamide |
Combinations of DMARDs | |
Methotrexate-based | Methotrexate, sulfasalazine, hydroxychloroquine |
Methotrexate, leflunomide | |
Methotrexate, ciclosporin | |
Methotrexate, gold | |
Methotrexate, sulfasalazine | |
Methotrexate, azathioprine | |
Other DMARDs | Leflunomide, sulfasalazine |
Gold, hydroxychloroquine | |
Steroid based | Steroids, methotrexate, sulfasalazine |
Steroids, methotrexate, ciclosporin | |
Steroids, methotrexate |
The efficacy of DMARDs involves reduced features of joint inflammation, such as fewer swollen joints and a lower ESR, a reduction in the progression of joint damage, particularly erosive damage, decreased levels of disability and improved quality of life. The harms, or adverse events, related to DMARDs include common problems seen with most DMARDs such as low white cell or platelet counts and unique toxicities with specific DMARDs. There is a reasonable evidence base for their use as monotherapies. 52–58
Steroids
The commonest use of steroids in RA is as adjunctive agents to control disease flares; they may be given intra-articularly, intramuscularly or orally. Because in early disease it has been suggested that steroids exert a disease-modifying effect, they form an initial but temporary component of several early arthritis combination regimens. They are also widely used as part of intensive DMARD combination therapy regimes in patients with uncontrolled established disease. There is a reasonably strong evidence base for their use. 59–61
Disease-modifying antirheumatic drug combinations with and without steroids
Disease-modifying antirheumatic drugs can be used in combination (see Table 1). This approach, initially advocated by McCarty,62 has been examined in many clinical trials. Initial studies evaluated combinations that turned out to have excessive toxicity (gold–hydroxychloroquine)63 or limited efficacy (methotrexate–azathioprine). 64 This toxicity led early reviews to suggest that risk/benefit ratios were unfavourable compared with monotherapy. 65
However, the situation changed when randomised controlled trials (RCTs) of methotrexate–ciclosporin,66 methotrexate–sulfasalazine–hydroxychloroquine67 and methotrexate–sulfasalazine–steroids68 reported improved disease control in active RA with mild or no excess toxicity; similar results were obtained in subsequent combination therapy studies. Combination DMARDs (cDMARDs) may not be required for all RA patients. In the only RCT of mild, early RA patients on stable DMARD monotherapy, they did not add benefit. 69
Overall, from our 2005 systematic review,70 and as suggested by a gradual expansion of its use in routine practice,71 the benefits of combination therapy are now thought to outweigh the risks in patients with active disease not controlled by monotherapy. They are recommended in UK national guidelines72 for early RA patients with active disease to avoid delay in bringing the disease under control, which is known to be associated with a poor outcome.
The RCT evidence for using DMARD combinations is of crucial importance to the Tumour necrosis factor inhibitors Against Combination Intensive Therapy (TACIT) trial. It is summarised in detail for both early and established RA in two systematic reviews (see Chapter 3).
Tumour necrosis factor inhibitors
These agents were developed in the late 1980s to target tumour necrosis factor (TNF)-α, a cytokine of central importance in the pathogenesis of RA, which exerts its effects by binding to type 1 and 2 receptors on immune, inflammatory and endothelial cells in the lymphoid system and joints and in less well-studied systems such as the central nervous system. 73
The proof of principle for inhibiting this cytokine came from an open-label clinical study in which patients with RA received a single infusion of a tumour necrosis factor inhibitor (TNFi). Patients showed a rapid response, including an early fall in CRP level. However, the anti-inflammatory effect lasted only 6−12 weeks and was followed by a return of active disease. 74 As a result, patients were retreated with further infusions; these showed responses of similar magnitude and duration. 75 The scene was set for a major clinical development programme.
There are currently five TNFis available to treat inflammatory arthropathies, summarised in Table 2. All have been shown to be effective in large clinical trials, which have been collated in systematic reviews. 76–80 These TNFis can be subdivided into first-generation agents (comprising etanercept, infliximab and adalimumab) and second-generation agents (comprising certolizumab and golimumab). In RA, all of these agents are licensed for use in routine clinical care; they are also approved by National Institute for Health and Care Excellence (NICE) for use in the NHS although in some cases this has required a financial risk-sharing agreement. 81–83
TNFi | Site of action | Dosing | Methotrexate |
---|---|---|---|
Infliximab | Binds soluble/transmembrane TNF-α and inhibits binding of TNF-α to receptors | Intravenous administration every 4–8 weeks | Essential to co-prescribe |
Etanercept | Binds TNF-α and lymphotoxin and competitive inhibitor of TNF receptor | Subcutaneous twice weekly | Optional to co-prescribe |
Adalimumab | Binds soluble/transmembrane TNF-α and inhibits binding of TNF-α to receptors | Subcutaneous fortnightly | Optional to co-prescribe |
Certolizumab | Binds soluble/transmembrane TNF-α and inhibits binding of TNF-α to receptors | Subcutaneous fortnightly | Optional to co-prescribe |
Golimumab | Binds soluble/transmembrane TNF-α and inhibits binding of TNF-α to receptors | Subcutaneously monthly | Optional to co-prescribe |
There is no clear-cut evidence that any one of these agents is superior to any other, and practical issues, including cost, determine which is chosen. There have been network meta-analyses of the efficacy and toxicity of different TNFis and these suggest potential minor differences in efficacy and adverse event risks. 84–86
Infliximab must be given concurrently with methotrexate (or another DMARD in methotrexate-intolerant patients) to prevent the formation of human antichimeric antibodies. 87 The licence for adalimumab also requires concomitant methotrexate unless the patient is intolerant. Although concomitant treatment is not required for etanercept, substantial data suggest that combination treatment is more effective, especially in terms of the effect on bone erosion. Therefore, all three drugs are almost always given with methotrexate or another DMARD. 88
The RCT evidence for using TNFis in combination with methotrexate and other DMARDs is also of crucial importance to the TACIT trial. This evidence is also summarised in detail in the systematic reviews in Chapter 3.
The question of what to do when a TNFi failed was a crucial question, particularly in the early 2000s when other biologics were not available. There is only limited information about the relative merits of switching from one TNFi to another. The only RCT studied golimumab in patients who had failed another TNFi; this showed some benefit from the switch. 89 The relative benefits of switching TNFis in patients who, for one reason or another, have not responded to their first biologic has also been addressed using observational data from registries and similar studies. Again, these studies provided some evidence that switching TNFis can give clinically useful improvements although response rates for second and subsequent TNFis are lower than for first-time use. 90
More recently, several trials evaluating non-TNF-targeted biologics, including abatacept, rituximab and tocilizumab, have provided convincing evidence that non-TNF-targeted biologics are effective in patients who have failed with TNFis, and this is increasingly the preferred approach. 91,92
Other new agents
A number of other biological treatments have been licensed, and in some cases approved by NICE, for treating RA. An early agent, anakinra, which is an interleukin-1 (IL1) receptor protein, is relatively ineffective93 and is not often used for treating RA. It is, however, highly effective in a range of other disorders including acute gout, some forms of juvenile arthritis and some familial periodic fevers. Further anti-IL1 agents are in late-stage development, currently for these indications.
Rituximab targets B cells and is highly effective in active RA. 94 Its mechanism of action is controversial as the presence of rheumatoid factor is not essential for its efficacy. Tocilizumab targets IL-6 and is also highly effective in active RA. 95 The third effective biological treatment, abatacept, targets costimulatory molecules on T lymphocytes. 96 Although some of these other biological treatments are licensed to be used as first-line treatment in methotrexate incomplete responders with RA, network meta-analysis and similar comparative studies84,85 show that these different biologics have comparable efficacy. TNFis are the most widely used treatment in methotrexate incomplete responders. Therefore, comparing cDMARDs with TNFis will provide results of general interest.
Several new non-biological agents such as kinase inhibitors97,98 are being developed. One of these agents, tofacitinib, has been licensed in the USA99 but is not yet approved for use in Europe. Depending on their cost, and relative efficacy and toxicity, such orally active agents may also change the treatment pathways for RA. However, for the present their roles are uncertain.
Non-TNFi biologics and new oral DMARDs are not part of the TACIT trial. Most of their actual or projected use is for patients who have failed to respond to both conventional DMARDs and TNFis. This late-stage treatment pathway for RA, which is complex and less well defined than the earlier management stages for RA, is not the key focus of the TACIT trial. We have therefore not reviewed it in detail.
Treatment strategies
Supportive and symptomatic treatment
As with all long-term disorders the management of RA requires multiple inputs from a range of health-care professionals from primary and secondary care. Patients need to be fully informed about their condition and be able to access advice; this is one of the key roles of the specialist nurses. Patients need effective treatment for pain, using analgesics and non-steroidal anti-inflammatory drugs,100,101 and their comorbidities, notably ischaemic heart disease, need to be appropriately managed. 102 Finally, they need access to physiotherapists and in some cases occupational therapists and need to be encouraged to take regular exercise. 103,104 The appropriate use of all of these treatments is crucial to ensure a good outcome. However, they are outside the focus of the TACIT trial and so have not been considered in detail.
Treat to target
There is evidence that intensive treatment is important in early RA both to suppress disease activity105–109 and to maintain low disease activity when it has been reduced. Welsing et al. 110 investigated the longitudinal relationship between disease activity and radiological progression in two independent follow-up cohorts. Both showed significant relationships between disease activity and radiological progression, but only in patients seropositive for rheumatoid factor. The results support systematic monitoring to achieve persistent low disease activity. This approach, termed ‘tight control’ or ‘treat to target’, includes several standard procedures such as:
-
a predefined treatment protocol to which the treatment of individual patients is adjusted
-
being able to assess whether or not the treatment chosen is necessary and effective
-
incorporating measures to ensure that patients are not overtreated.
Many groups have reported on aspects of tight control. 111–114 Most used the Disease Activity Score (DAS) or DAS28 to guide treatment or as the primary end point. Overall clinical and radiological outcomes were more favourable in patients receiving tight-control regimens; in particular, remission rates were generally higher with tight control than with conventional therapy. These improved clinical and radiological outcomes did not appear to be at the cost of increased drug toxicity.
Access to high-cost treatments
Different countries have taken divergent approaches to the use of high-cost treatments such as biologics. A range of international groups, specialist societies and regulatory bodies recommends TNFis for patients with active RA who have failed to respond to conventional DMARDs. 115–118 The current UK consensus recommends that TNFis are started only in patients who have a DAS28 of > 5.1115 and who have failed to respond to adequate therapeutic trials of two standard DMARDs including methotrexate. 119 Some UK experts believe that the threshold for active RA should be reduced to a DAS28 of > 3.2 with at least three or more tender joints and three or more swollen joints. 120 There are major national differences in the guidelines followed by rheumatologists for starting biological treatments and considerable diversity in biological treatment use across Europe. 121–124
Economic modelling and biological treatments
Economic modelling conventionally extends beyond conventional RCTs,125,126 bringing together cost and outcome evidence from a range of sources. Several modelling methods are used including simple decision trees, Markov models and individual sampling models. Most economic studies evaluate the impact of biological treatments on quality-adjusted life-years (QALYs). In the absence of direct QALY measures, values may be inferred from other available outcomes. 127 Recent systematic reviews of health economic studies in RA highlight the different conclusions reached based on assessments of the much the same set of published evidence. Schoels et al. 128 identified 21 relevant studies of biological treatments and, based on willingness-to-pay thresholds of US$50,000–100,000 per QALY, they concluded that combinations of TNFis with methotrexate were cost-effective after conventional DMARD failure. The sequential use of TNFis has been a difficult problem to resolve; however, Brennan et al. 129 reported favourable incremental cost-effectiveness ratios (ICERs) for using a second TNFi compared with DMARD treatment. A different perspective was taken in a systematic review by van der Velde et al. 130 They concluded that the economic evidence suggests that biological treatments are not cost-effective compared with DMARDs for RA in adults at a cost-effectiveness threshold of C$50,000 per QALY and that there is mixed evidence of cost-effectiveness in selected populations at a willingness-to-pay threshold of C$100,000 per QALY.
Rationale for the Tumour necrosis factor inhibitors Against Combination Intensive Therapy trial
Alternative approaches for accessing high-cost biological treatments in rheumatoid arthritis
Different groups of experts have reached widely differing views on when biological treatments in general and TNFis in particular should be started in RA. Almost all experts recommend usually using them in combination with methotrexate or other DMARDs. There is also universal agreement that they should be reserved for active RA, although there is uncertainty about what constitutes active disease.
Most experts recommend that they are used in patients who have failed to respond fully to methotrexate and who continue to have active RA. Many trials have been undertaken in such patients with positive findings. Most countries in continental Europe and North America have adopted this approach. The UK is more conservative and TNFis are generally used after patients have failed two DMARDs and still have active disease.
There are many alternative ways in which TNFis could be used in RA. One approach, which might be the most effective and cost-effective, is to reserve them until RA patients have tried and failed to respond to intensive DMARD combination treatments. Such an approach would place TNFis slightly lower down the therapeutic cascade; however, this might be of greater overall benefit to the health service by optimising the use of resources without causing patients any problems. This option is specifically explored in the TACIT trial.
Limiting the use of high-cost treatments has always been a component of Western health care. No country allows universal access to all high-cost treatments for all patients who would like to have them. It is equally important to ensure that patients are not denied effective treatments. If intensive cDMARDs are effective in some patients and the use of biological treatments can be changed so that they are given to patients most likely to show substantial benefits, the needs of both patients and health-care funders can be met. TNFis are not universally effective in RA; they have a response failure rate in the region of 20–30% of patients. 131–134 Some patients who would fail TNFis may respond to intensive cDMARD regimens. As the number of treatment options is limited in RA, and as patients rarely move from biological to non-biological treatment, there is a sound logic in exhausting treatment options in an organised sequence. If TNFis were curative such an argument would be misplaced; however, the balance of evidence indicates that DMARDs and biological treatments are both suppressive therapies that do not appear to alter the long-term clinical phenotype.
Systematic reviews of intensive treatments
The two treatment strategies used in the TACIT trial – cDMARDs and TNFis given in combination with methotrexate or another DMARD – have both been studied in RCTs in RA. Systematically reviewing these previous trials is crucial for both designing the TACIT trial and interpreting its findings.
We have previously published three systematic reviews of combination therapy trials: one looked at all combinations at all time points;70 the second compared DMARD combinations and TNFi/methotrexate combinations in early RA;135 and the third specifically examined the toxicity of combination therapies. 136 A number of other groups have also published systematic reviews of both cDMARDs and TNFis. 65,85,137–144 These systematic reviews all show that both DMARD combinations and TNFi/methotrexate combinations are effective in both active early and established RA. Their effectiveness in clinical trials is broadly comparable, although there are insufficient head-to-head clinical trials of these two treatment strategies. Overall, these published systematic reviews provide strong clinical support for undertaking a head-to-head trial such as the TACIT trial.
In early RA the relative clinical effectiveness and cost-effectiveness of conventional DMARD combinations have been considered in detail in guidance from NICE. 72 This guidance recommends cDMARDs, including methotrexate, as first-line treatment for early active RA. Biological treatments were excluded as first-line therapy by NICE because they were not considered cost-effective in these patients. Not all guidance accepts this conclusion; for example, the ACR advises the use of biological treatments (TNFis) combined with methotrexate as one initial therapy for active early RA patients with a poor prognosis118 as an alternative to cDMARDs.
In patients with established RA disease the overall value of intensive DMARD combination regimens compared with TNFi/methotrexate combinations is less certain, particularly as these patients will usually have failed one or more previous DMARDs. The TACIT trial is aimed at these patients because there is genuine uncertainty about the relative merits of cDMARDs in such cases.
Aims and trial design
The TACIT trial focuses on the treatment of patients with active RA who have failed two DMARDs and who meet the current NICE criteria for starting TNFis. These NICE criteria are based partly on evidence from RCTs, partly on economic modelling and partly on expert opinion. Our alternative view is that many of these patients will do equally well on intensive combination therapy with conventional DMARDs.
Agreeing the research hypothesis and designing a RCT to test the hypothesis required considering the following three crucial issues:
-
the key outcome
-
the duration of the trial
-
minimising the risk that patients randomised to receive cDMARDs are disadvantaged.
Our previous research has shown that the HAQ is a sensitive patient-assessed outcome measure in active RA trials of DMARDs. 145,146 It also has a crucial role in the economic modelling that is used to justify prescribing biological treatments. The HAQ was also the primary outcome measure in the Behandel Strategieen (BeSt) trial;147 the only previous trial involving comparisons between cDMARDs and biological treatments published before the start of the TACIT trial, albeit in early RA. We therefore decided that changes in HAQ score should be the primary outcome measure.
The trial duration was more straightforward. Six months is probably too short a period of time to judge both clinical effectiveness and cost-effectiveness. A duration longer than 12 months appeared to be impractical and had no obvious advantage. As a consequence we decided that 12 months was the optimal time. This was also the time point at which the BeSt trial was first analysed. 147
The final issue, about minimising risks to patients randomised to cDMARDs, was more complex. There were two potential risks. The first was that cDMARDs may have excessive toxicity. This risk would be minimised by independent oversight of the trial by the Data Monitoring and Ethics Committee. The other risk was inefficacy. We considered that if patients showed no response to cDMARDs after 6 months of treatment they should then be offered TNFis. We also considered that a response should adopt the same criterion that NICE recommends for maintaining patients on TNFis – a change in DAS28 of ≥ 1.20.
The final issue for the TACIT trial was whether it could be a placebo-controlled trial or an open-label strategy trial. As cDMARDs need to be individualised it would be impractical to deliver a placebo-controlled trial; instead, we considered that the trial had to be open label.
Hypothesis
The TACIT trial was designed to test the hypothesis that patients with active RA who meet the NICE criteria for treatment with TNFis will gain equivalent benefit over 12 months at substantially less expense and without increased toxicity from starting treatment with intensive combination therapy with DMARDs.
Primary and secondary outcomes
As a result of these various considerations the TACIT trial used the following outcome measures:
-
primary outcome measure: HAQ, the key patient-completed disability measure in RA
-
secondary outcome measures: joint damage, quality of life, disease activity, withdrawal rates, adverse effects, costs, QALYs, cost-effectiveness and cost–utility.
Testing the hypothesis
The TACIT hypothesis would be rejected if the primary outcome measure – the HAQ – showed substantial clinically important improvements at 12 months in patients randomised to receive TNFis. The TACIT trial was designed to confirm or refute the equivalence of treatment with TNFis and cDMARDs in improving HAQ scores over 12 months.
The TACIT hypothesis would also be either rejected or substantially weakened if economic evaluations at 12 months – including health and social care costs, societal costs, cost-effectiveness and cost–utility – showed disadvantages in patients randomised to receive cDMARDs or if the adverse event profile was substantially worse with cDMARDs.
The TACIT trial also collected a range of secondary outcomes to help evaluate the clinical usefulness of cDMARDs in these patients. These included assessing joint damage, quality of life, disease activity using the DAS28 and retention rates on such DMARD treatment. We considered also collecting other outcomes such as ACR response rates and responses on other indexes such as the Simplified Disease Activity Index. However, we concluded that measuring the same outcome in multiple ways, particularly using methods not followed in the UK where the trial is based to inform routine practice, would be counterproductive.
Systematic reviews
In the last few years more trials have been published about DMARD combinations and TNFi/methotrexate combinations. To ensure that the results of the TACIT trial can be placed into an appropriate context it is essential to provide an updated systematic review.
As there are different issues in early RA and established RA we have undertaken two reviews of these different aspects of RA treatment. As it is crucial to define whether or not there are differences in these two clinical settings in the relative efficacy of cDMARDs and TNFis, we used similar methods in both reviews. Our analytical approach is also comparable.
Consequently, the two systematic reviews assess the efficacy and toxicity of combination treatment with both cDMARDs and TNFis with methotrexate in the two groups of RA patients. The first group was patients with early disease, which is disease of < 3 years’ duration. The second group was patients with established disease who have failed one or more DMARDs. The reviews evaluate treatments in trials that compared (1) cDMARDs with DMARD monotherapy; (2) TNFis plus methotrexate with methotrexate monotherapy; and (3) cDMARDs with TNFis plus methotrexate. The trials that enrolled patients with early RA were analysed separately from the trials that enrolled patients with established RA.
Chapter 2 Methods
Trial design
The TACIT trial was an open-label, pragmatic, randomised, multicentre, two-arm trial. Patients were allocated to each arm in equal numbers. The duration of the TACIT trial was 12 months.
The trial compared intensive cDMARDs with TNFis given together with methotrexate or another DMARD in active established RA. Patients who failed to respond to cDMARDs were eligible to receive TNFis after 6 months; this period was considered optimal to judge responsiveness to DMARDs. Patients in the TNFi arm were assessed for response to their first TNFi at 6 months, reflecting NICE guidance. Those who did not respond tried another TNFi. If they failed they were offered alternative treatment such as cDMARDs.
The trial was unblinded because individually optimised intensive cDMARD therapy cannot be given blind. Many previous RCTs in RA using such treatments have been unblinded. This approach provided the closest possible approximation to routine clinical care. The disadvantage of unblinded studies – that clinicians have excessive influence on the results – was ameliorated because the primary outcome measure, the HAQ, was a patient self-completed questionnaire. In addition, another key outcome, radiographic changes, was measured without knowledge of treatment group.
The TACIT trial raised a number of ethical issues related to whether or not patients were being potentially denied access to highly effective treatments. These are considered in detail in the discussion.
Eligibility criteria
The trial was aimed at patients with RA attending outpatient rheumatology clinics in England who met the current NICE criteria for receiving TNFis. 81
Inclusion criteria
-
Men and women aged > 18 years.
-
Established RA according to the 1987 criteria of the ACR. 5
-
Disease duration of at least 12 months.
-
Meet NICE criteria for being prescribed TNFis:81 DAS28 > 5.1; failure to respond to two DMARDs including methotrexate; no contraindications to TNFis (including possibility of pregnancy).
Exclusion criteria
-
Unable or unwilling to give informed consent.
-
Failure of, or contraindications to, all proposed DMARD combinations (including possibility of pregnancy).
-
Serious intercurrent illness.
-
On high-dose steroids (in excess of 10 mg prednisolone or equivalent per day at trial entry).
Settings, locations and patient identification
Patients were recruited from rheumatology clinics in England and Wales, divided into three sectors: London/south, the Midlands and the north. The trial was undertaken in a routine outpatient setting with patient management shared between rheumatology specialist nurses and consultant rheumatologists. The supervising consultant was responsible for all aspects of patient care within the trial.
Before starting recruitment the primary care trusts (health-care commissioners) associated with each collaborating centre were contacted to ensure that they were informed about the nature and purpose of the trial and understood its clinical and economic implications. Collaborating rheumatologists and their specialist nurses were fully briefed to ensure that they had a good understanding of the rationale behind the study and of the principles and practice of using combinations of DMARDs in an intensive regimen. These processes were designed to allowed unhindered recruitment into the study.
Patients likely to be eligible to receive TNFis were managed in the following way (in line with NICE guidance):
-
ensure that they have failed adequate treatment with two DMARDs and have a DAS28 > 5.1.
-
negative screen for tuberculosis including chest radiography (and other local measures such as Mantoux testing where applicable)
-
repeat the DAS28 assessment 4 weeks after the initial DAS28 assessment to ensure that it remains > 5.1.
Patients were eligible to enter the trial only at this stage, when they received full information about the trial and informed consent was obtained. Patients had adequate time and information to decide whether or not they wished to participate.
Patients were pre-screened and the following data were collected:
-
number of patients who were potentially eligible to receive TNFis
-
reasons why patients chose not to enter the trial (insufficient disease activity, consent not obtained and other reasons).
-
numbers of patients randomised.
Trial interventions
The TACIT trial compared two treatment algorithms, one for TNFis and one for cDMARDs. Treatments were individualised and depended on patients’ responses.
Tumour necrosis factor inhibitors
The three licensed agents available when the trial started – adalimumab, etanercept and infliximab – were allowed at standard doses (British National Formulary148). The choice of TNFi reflected patient preference and local circumstances. Methotrexate was also given to maximise efficacy and (in the case of infliximab) reduce the formation of antichimeric antibodies. Patients intolerant to methotrexate took another DMARD. DAS28 at 3 and 6 months defined responses to therapy.
Patients had their TNFi stopped for one or more of three reasons:
-
lack of effect as defined by the NICE criterion,81 that is, a change in DAS28 of < 1.2 at 3 or 6 months
-
an adverse event that, in the opinion of the supervising specialist, necessitated treatment withdrawal
-
patients could stop therapy for any reason should they wish (reasons to be specified if patient willing).
Patients in whom one TNFi was stopped were able to start another. This option represented current UK practice when the trial started. Patients who failed two TNFis for whatever reason were not able to start a third agent and required alternative treatment such as cDMARDs.
The principles of the treatment algorithm were as follows:
-
start a TNFi of choice on the basis of local circumstances and patient preference
-
assess at 3 months: no change if good response (change in DAS28 ≥ 1.2); change to second TNFi if change in DAS28 is < 1.2
-
assess at 6 months: no change if good response (change in DAS28 ≥ 1.2); change to second TNFi if change in DAS28 is < 1.2; if two biologics already given and DAS28 change is < 1.2, TNFi stopped and patient offered DMARD combination or other therapy.
Combination disease-modifying antirheumatic drugs
Those cDMARDs with proven efficacy over DMARD monotherapy in RCTs were used, including:
-
triple therapy with methotrexate (methotrexate–sulfasalazine–hydroxychloroquine)
-
other methotrexate combinations (methotrexate–ciclosporin, methotrexate–leflunomide and methotrexate–gold)
-
a sulfasalazine combination (sulfasalazine–leflunomide)
-
additional monthly steroids [intramuscular Depo-Medrone (120 mg stat) or equivalent] were used if needed.
The DMARD combinations were stopped for three reasons: adverse events, patient-initiated withdrawals (which are identical to those reasons for stopping a TNFi) and lack of effect (change in DAS28 < 1.2), which is similar to that for TNFis but was implemented only at 6 months.
The principles of the treatment algorithm were:
-
initially: maximise initial DMARD/optimise administration (e.g. parenteral methotrexate); start second/third DMARD; give intramuscular Depo-Medrone® (methylprednisolone, Pfizer) (whenever possible)
-
second step: maximise dose of second/third DMARD
-
third step: change combination (repeated if needed)
-
additional option: continue with intramuscular Depo-Medrone monthly short term if RA remains active
-
assess monthly and change treatment if change in DAS28 is < 1.2 or DAS28 is > 3.2
-
at 6 months start a TNFi if change in DAS28 is < 1.2.
The target doses of different DMARDs used in combinations were as follows:
-
methotrexate: 25 mg weekly – preferably by intramuscular injections although could be oral (achieved by 5-mg increments)
-
sulfasalazine: 3 g daily (starting at 500 mg daily and increasing by 500-mg increments)
-
hydroxychloroquine: 400 mg daily (starting at 200 mg and increasing as one increment)
-
ciclosporin: 3.5 mg/kg (starting at 2 mg/kg and increasing incrementally depending on creatinine levels)
-
leflunomide: 20 mg/day (staring at 10 mg/day and not increasing if used in combination with methotrexate)
-
gold: 50 mg/month (starting with test dose, then 50 mg/week for 20 weeks, then 50 mg/month)
-
intramuscular Depo-Medrone: 120 mg/month for 3 months; further courses were given if the RA was still active.
Dose adjustments to all drugs depended on both disease activity and evidence of adverse events. Decisions about changes in treatment were made by the supervising rheumatologist but were reviewed by the principal investigator (DS) or deputy to ensure that the algorithm was followed.
Concomitant therapy
Non-opiate analgesics and non-steroidal anti-inflammatory drugs were used as needed at standard doses. Patients taking methotrexate also received folic acid (5 mg/week) to limit adverse events. Patients taking steroids received bone protection (e.g. alendronate and calcium/vitamin D). Other drugs (e.g. antihypertensives) were used as needed. Patients taking oral prednisolone up to 10 mg at entry stayed on treatment. Intra-articular steroids were used as required.
Trial outcomes
Primary clinical outcome
Patient self-assessed outcomes were used in the TACIT trial. 152 The HAQ was the primary clinical outcome measure. 153 Although it is sometimes termed the HAQ Disability Index, most reports refer to it simply as the HAQ. The HAQ is a self-assessed questionnaire that is completed by patients. It primarily measures disability and is the dominant disability assessment in RA. 154 Scores range from 0 to 3, with higher scores indicating greater disability. It has established reliability and validity and has been used in many published RCTs in RA. HAQ scores were measured initially and at 6 and 12 months.
Secondary clinical outcomes
The European Quality of Life-5 Dimensions (EQ-5D)155 and the Short Form questionnaire-36 items (SF-36)156 were measured initially and at 6 and 12 months. These are also self-assessed questionnaires completed by patients. They measure health-related quality of life and can be used to estimate health utility and have been extensively studied in RA. 157–160
Plain radiographs of the hands (including the wrists) and the feet were taken initially and at 6 and 12 months. These are widely used outcome measures. 161 Digital images of the radiographs were read at the end of the trial by a single observer (DS) experienced in reading radiographs using the Larsen score,162 modified for minor changes. 163 The radiographs were assessed in known date order without knowledge of the treatments that patients had received.
Disease activity scores for 28 joint counts were measured initially and every month throughout the trial. 164 Scores are calculated using tender joint counts and swollen joint counts for 28 joints assessed by trained specialist nurses, the ESR and patients’ global assessments of their disease activity recorded on a 100-mm visual analogue scale (VAS). DAS28 was used to guide treatment based on the predefined treatment targets and to assess responses to treatment.
Adverse effects were recorded each month by patient reporting. Specific events such as hospital admission were also recorded following international guidance. 165
International recommendations
These outcome measures have all been recommended by international bodies including the European Medicines Agency166 and the US Food and Drug Administration. 167
Health economic assessments
An adapted version of the Client Service Receipt Inventory (CSRI) was used168 to measure individual-level resource use. It covered sociodemographics, the use of (all-cause) secondary and community-based health and social care services, time off work because of illness, receipt of social security benefits and medication prescribed in addition to the study treatments. The CSRI has been previously used successfully in trials in arthritis. 169 We also measured take-up rates (without intention to attribute costs) for NHS/social services contributions towards more exceptional resources, such as special mobility equipment, adaptations to the home or transport to health care. The CSRI was administered as a self-complete questionnaire retrospectively for the previous 3-month period at three assessment points: baseline, 6 months after randomisation and 12 months after randomisation. Use of trial medications was recorded separately and prospectively by the clinical/research teams over the entire study period in the form of medication name, dose, frequency and duration of use.
As discussed earlier, health-related quality of life was assessed by self-report questionnaires at baseline, and 6 and 12 months using the EQ-5D and SF-36 for the purpose of estimating QALY gains.
Data collection
Patient details and outcomes, with the exception of radiographic outcomes, were collected using an academic online database system with direct entry of data into the electronic case record form (see www.medscinet.net/tacit). This electronic data capture (EDC) system collected information anonymously using patients’ initials and date of birth as identifiers.
Sample size
The TACIT trial sought to show equivalence between treatment strategies; in this setting the calculation of sample size is more complex than in conventional trials intended to show that one treatment is superior. One specific issue is that high-cost treatments such as TNFis can be justified only if they show substantial benefits over conventional inexpensive treatments. Key issues in this respect are the extent to which a difference in HAQ score (the primary outcome) between groups is clinically relevant, the degree of certainty in avoiding a type II error and the degree of conservatism in the statistical approach taken. The final sample size calculation has taken into account these various considerations.
This sample size was defined by the trial hypothesis that treating active RA patients who have failed to respond to two DMARDs with intensive conventional treatment using cDMARDs and steroids gives equivalent results to treatment with TNFis.
The sample size calculation was based on changes in HAQ scores in:
-
the ATTRACT (Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy) trial (infliximab versus placebo in RA patients receiving concomitant methotrexate), in which the mean HAQ score at baseline was 1.7, which was reduced after treatment by 25%; the standard deviation (SD) of the change in HAQ score was 0.4170
-
the CARDERA (Combination Anti-Rheumatic Drugs in Early Rheumatoid Arthritis) trial, an Medical Research Council-funded UK trial of 464 patients in which the mean HAQ score at baseline was 1.6, which was reduced after treatment by 31%; the SD of the change in HAQ score was 0.6. 171
We used the average SD for change in HAQ score in these two trials, estimated at 0.5.
Most experts consider that the minimal clinically important change in HAQ score in RA is considered to be ≥ 0.22. 48,172–175 The trial was therefore designed under the assumption that cDMARDs and TNFis produce equivalent reductions in HAQ score and that a difference of < 0.22 would be regarded as equivalence.
Formally, the trial was designed to test the null hypothesis of a difference > 0.22. With a (one-sided) testing level of 5%, a sample size of 176 was required to achieve 90% power. To allow for a dropout rate of 5–7%, we planned to recruit 190 patients.
Recruitment method, randomisation and baseline assessments
Recruitment
Patients were recruited from rheumatology clinics in England, which were divided into three sectors: London and the south of England, the Midlands and the north of England.
Potentially eligible patients were identified by rheumatologists and clinic nurses at the participating centres. Rheumatologists approached potentially eligible patients and outlined the trial to them. Patients interested in participating were given the patient information sheet and were then contacted by telephone at least 24 hours after receiving the patient information sheet to see whether or not they were interested in participating. If they were, a screening assessment was arranged.
Screening involved making the following checks against NICE guidance to ensure that patients were eligible to receive TNFis:
-
ensure that patient has failed adequate treatment with two DMARDs and has a DAS28 > 5.1
-
negative screen for tuberculosis including chest radiography (and other local measures such as Mantoux testing where applicable)
-
repeat DAS28 assessment 4 weeks after the initial DAS28 assessment to ensure that it remains > 5.1.
The screening assessment pages were collected anonymously using the EDC system. Once complete and eligible, the EDC system automatically assigned consecutive patient numbers to patients in chronological order.
Randomisation
Randomisation numbers were formed of four numbers and prefixed by the region identifier (i.e. 1 for London and the south, 2 for the Midlands and 3 for the north). The allocation sequence for randomisation was generated by the EDC system. Block randomisation was used in blocks of four with allocation balancing. Randomisation was stratified by region. Formally, the trial was designed to test the null hypothesis of a difference of > 0.22. With a (one-sided) testing level of 5%, a sample size of 176 was required to achieve 90% power. To allow for a dropout rate of 5–7%, we planned to recruit 190 patients. The clinicians at each of the trial centres and the trial co-ordinator were unaware of the allocation sequence.
Once a randomisation number was allocated, the EDC system automatically informed the researcher at the individual centre and the trial co-ordinator by e-mail. The trial co-ordinator informed the pharmacy at site of the randomisation. The patient was then informed that they had been recruited to the trial and the baseline assessment was arranged.
Data collected during screening
As part of screening data were collected on:
-
number of patients who were potentially eligible to receive TNFis
-
reasons why patients chose not to enter the clinical trial (insufficient disease activity, consent not given and other reasons)
-
numbers of patients randomised.
Baseline assessment
Delays between screening and baseline were anticipated because of the pragmatic nature of the trial and local practices relating to the supply and delivery of TNFis. It was recognised that patients may therefore require additional treatment between the screening assessment and the baseline assessment, which would usually be an intramuscular steroid injection. The following rules were therefore applied:
-
patients were given an appropriate dose of intramuscular steroid if needed
-
the baseline assessment was delayed for 1 month after the date of injection.
Eligibility based on a DAS28 of > 5.1 at screening was not required to be maintained at the baseline assessment as this is not a requirement for receiving TNFis in routine practice.
Blinding
The TACIT trial was not blinded and both clinicians and patients knew to which treatment strategy patients had been allocated.
Statistical methods
Recruitment and follow-up patterns
Recruitment was recorded by year and region. The numbers of patients enrolled – excluding patients who had been withdrawn from therapy and who were unwilling to continue follow-up – were reported by treatment arm. The numbers of patients who withdrew from therapy, who were lost to follow-up or who died while taking part in the study were also reported by treatment arm.
Baseline comparability
Baseline characteristics were summarised by randomised group. Summary measures for the baseline characteristics of each group have been presented as means and SDs for continuous (approximate) normally distributed variables, medians and interquartile ranges (IQRs) for non-normally distributed variables, and frequencies and percentages for categorical variables.
Intention-to-treat population
Except for enrolled patients who withdrew consent or who were found to be ineligible at the baseline visit and so never received any treatment and for whom no data were therefore available, analyses on an intention-to-treat (ITT) basis reflect the randomisation process. We also carried out two additional analyses on the following populations:
-
a complete-case population: these were observations that subjects completed without missing data or violation of the protocol; this analysis is therefore referred to as a ‘complete-case analysis’ throughout this report
-
a per-protocol population: these were observations that excluded those patients who were found to deviate from the protocol.
The allowed variations to the protocol are shown in Appendix 1. The results of the per-protocol analysis were similar to those of the ITT analysis. Therefore, the results of the ITT and complete-case analyses have been presented in this report.
Imputing missing data
All participants had observations at baseline. However, some subjects had missing data on the outcome variables at 6 months, 12 months or both. The outcome variables that were measured at baseline and 6 and 12 months (HAQ, SF-36, EQ-5D and Larsen score) were imputed under different assumptions from those used for the DAS28 and its components, because DAS28 was measured monthly.
All missing data were imputed regardless of the reasons why it was missing. For the subjects who had missing outcomes, the baseline outcomes and other explanatory covariates (treatment group, sex, age, ethnicity, region and disease duration) were used to impute the missing data, assuming that unobserved measurements were missing at random.
For the subjects who had missing outcomes at 6 months, under the monotone assumption, baseline outcomes and explanatory covariates were used to impute these missing values. Then, for those patients who had missing outcomes at 12 months, baseline and 6-month outcomes with explanatory covariates were used to impute the missing values at 12 months. If the outcome variables were missing at 6 and 12 months then the outcome variables at 6 months were imputed first followed by the outcome variables at 12 months.
The DAS28 and its component were imputed using multivariate sequential imputation using chained equations. First, all missing values were filled in by simple random sampling with replacement from the observed values. The first variable with missing values, say DAS28 at month 1, was regressed on all other variables, DAS28–0, DAS28–2 through to DAS28–12, restricted to individuals with the observed DAS28–1. Missing values for DAS28–1 were replaced by simulated data points drawn from the corresponding posterior predictive distribution of DAS28–1. Then, the next variable with missing values was replaced using the same cycle. 176
The imputation was 20 cycles; at the end of the first cycle one imputed data set was created and the process was then repeated to create 20 imputed data sets. The 20 data sets were combined using Rubin’s rules;177–179 therefore, the estimates and standard errors presented here are the combined ones. As an additional check of the robustness of the analyses performed to the missing at random assumption, we further analysed the individual HAQ scores, EQ-5D scores, Larsen scores and DAS28 and its components) using the linear increments method of Diggle et al. 180 to handle the missingness. As the results obtained using this approach were qualitatively the same as those of the multiple imputation approach adopted, we report only the findings from the standard multiple imputation analyses.
Adjustment for design factors
Randomisation was stratified by region and therefore analyses of outcomes in the univariate or multivariable analyses were adjusted for region.
Outcomes assessed every 6 months
The primary outcome (HAQ score) and three of the secondary outcomes (EQ-5D score, SF-36 summary scores and Larsen score) were measured at baseline and 6 and 12 months. As there were not a significant number of zero values for the HAQ and other outcomes during follow-up, a linear regression model was used to analyse the change in these outcomes at 6 and 12 months. Thus, change was defined as either 12- or 6-month score minus baseline score. The unadjusted univariate analysis (model 1) was adjusted for region to account for design effect. The adjusted multivariable model (model 2) included sex, ethnicity, age, region, disease duration and baseline covariate as explanatory variables. Interactions between treatment and sex were assessed in the adjusted model 2 using the Wald test. The sex-specific interactions were not significant (for all outcomes p > 0.70). The treatment regression coefficient provided an estimate of the mean differences in HAQ, EQ-5D, SF-36 and Larsen scores.
For individual components of the SF-36 we used generalised estimating equations (GEEs) to estimate the effect of treatment including baseline values as a covariate for these outcomes. Working correlation matrices were unstructured, which was not unduly restrictive given that measurements were taken only at three time points. As the data were analysed longitudinally, time was included as a covariate in models 1 and 2. A final model tested specifically for interactions between treatment and sex and treatment and time using the Wald test. The sex-specific interactions were not significant (for all outcomes p > 0.50 in the overall test of all interaction terms). However, the interaction term between time and treatment was of borderline significance for some SF-36 domains (physical functioning, general health perception). We therefore report the period-specific treatment effect for those variables that had significant interaction terms.
Outcomes assessed every month
The DAS28 and its components were measured monthly and were therefore analysed separately. Changes in DAS28 and its components were analysed using GEEs to estimate the effect of treatment including baseline values as a covariate. Working correlation matrices were autoregressive with lag 1. In this analysis interactions between time and treatment and sex and treatment were also assessed and were found to be non-significant. Treatment effects were examined as subanalyses in two periods (1–6 months and 7–12 months). The estimates were presented as mean treatment effects (beta coefficients) with 95% confidence intervals (CIs). The sandwich estimator of error was used with the aim of obtaining robust estimates of precision. Statistical significance was determined at the 5% level using a two-sided test throughout. These analyses were based on the assumption that patients stayed in their original randomised treatment arm and thus ignored subsequent treatment switches.
Exploratory analyses
The patients randomised to start cDMARDs fell into two categories. The first category included those patients who remained on cDMARDs throughout the TACIT trial. The second category included those patients who switched to a TNFi after ≥ 6 months because they had not fully responded to cDMARDs. The outcomes of these two categories of patients have been compared in a series of exploratory analyses, recognising that these are non-randomised in their original treatment arm. These analyses were carried out for all populations (ITT, complete case and per protocol).
Four additional analyses used all observed data only and missing data were not imputed. This approach was taken when patients were divided into discrete response categories. These analyses comprised: (a) changes in Larsen score; (b) the development of new erosions shown by categorical increases in Larsen score; (c) clinical response to treatment indicated by a decrease in DAS28 of ≥ 1.2; and (d) achieving remission indicated by a DAS28 of ≤ 2.6. Analytical approaches used specifically with all observed data were the construction of Kaplan–Meier plots and a comparison of treatments using the log-rank test.
Toxicity
The proportion of serious adverse events was compared across randomised groups using Fisher’s exact test as appropriate.
Software specification
All data management and analyses were carried out using Stata version 12.0 (StataCorp LP, College Station, TX, USA) and the R statistical package (The R Foundation for Statistical Computing, Vienna, Austria181).
Economic evaluation methods
Costs
Unit costs were applied to resource use data to calculate cost per participant. Unit cost estimates, their sources and any assumptions made for their estimation are detailed in Appendix 2. Medication unit costs were converted into cost per mg based on the most cost-efficient pack size, choosing maintenance prices over initial treatment prices and generic prices over branded ones to obtain conservative estimates.
Total costs were computed for each participant at each assessment point from two perspectives: health and social care and societal. Health and social care costs included the costs of inpatient services, outpatient services, primary care services, other community-based services, social services, trial medications and other prescribed medications. Two sets of societal costs were calculated, one that included health and social care costs plus participant lost productivity because of absence from work and one that included health and social care costs, participant lost productivity because of absence from work and, additionally, the cost of social security benefit payments received.
For the economic evaluation, costs generated from the 3-month CSRI data were extrapolated (multiplied by 2) to cover the full 6 months before each follow-up point. All costs are reported in pounds sterling at 2010/11 prices. Discounting was not necessary as all costs were related to a 1-year period.
Outcome measures
Cost-effectiveness analyses were based on the primary outcome measure (the HAQ). Cost–utility analyses were based on QALYs derived from both the SF-36 and the EQ-5D. Utility weights appropriate to each measure were attached to the SF-36- and EQ-5D-produced health states at baseline and 6 and 12 months. 182,183 QALY gains between baseline and 6 months and between 6 months and 12 months were then calculated using the total area under the curve approach with linear interpolation between assessment points (and baseline adjustment for comparisons184).
Analyses, missing data and sensitivity analyses
Data were analysed using IMB Statistical Product and Service Solutions (SPSS) Statistics for Windows (version 20; IBM Coporation, Armonk, NY, USA) and Stata (version 11.2). Participants had individual unit costs applied based on the exact medication that they were prescribed, not on which arm they were in. Therefore, appropriate costs were applied regardless of switching during the trial.
Costs and outcomes were compared at 6 and 12 months and are presented as means and SDs. Mean differences and 95% CIs were obtained using non-parametric bootstrap regressions (1000 repetitions) to account for the non-normal distribution commonly found in economic data, with adjustment for region as this was a stratification factor in the randomisation process. Although this was a RCT and participants in all groups were expected to be balanced at baseline, baseline costs and outcomes could be predictors of follow-up costs. To provide more relevant treatment effect estimates,185 regressions to calculate mean differences in costs at follow-up included covariates for baseline cost from the same cost perspective, baseline HAQ score, duration of illness, age, sex, region and ethnicity. Outcome comparisons (for the economic evaluation) at follow-up included covariates for baseline values of the same outcome plus baseline HAQ score, duration of illness, age, sex, region and ethnicity.
Data were entered via an EDC system using the MedSciNet database (MedSciNet AB, Stockholm, Sweden; http://medscinet.com) which was programmed to disallow individual-item non-response on the CSRI service use section. There was thus no item non-response for this part of the CSRI. For lost employment data, if the CSRI indicated that this was positive but the amount was missing, the mean lost employment cost for that arm at that time point (only for those who had lost employment and had valid data) was substituted. For social security benefit data, if the CSRI indicated that this was positive but the amount was missing, unit costs for specified benefits were applied. When receipt of benefits was positive but specific benefits were unspecified, the mean benefit cost for that arm at that time point (only for those who received benefits and had valid data) was substituted. For non-trial medication data, if the medication name was missing but other information (e.g. dose) indicated some use, an average prescription cost (from Department of Health prescription cost analyses; see http://data.gov.uk/dataset/prescription_cost_analysis_england) was assumed. If a medication name was provided but usage quantity was missing, an average prescription cost for that particular medication was assumed.
Analyses were based on available cases for each analysis, that is, they excluded non-responders to the CSRI, HAQ, EQ-5D or SF-36 at each time point if there were any. To explore the potential impact of excluding non-responders we examined the sociodemographic and clinical characteristics of those included in the analyses and those in the full sample. We also carried out an ITT analysis, imputing missing 6- and 12-month total costs and outcomes using the multiple imputation command in Stata (version 11). Imputations of missing 6- and 12-month costs were based on variables expected to predict follow-up costs: baseline HAQ score, duration of illness, age, sex, region, ethnicity, trial arm and equivalent baseline cost (and equivalent cost at 6 months for 12-month imputations). Imputations of HAQ scores at 6 and 12 months were based on baseline HAQ score, duration of illness, age, sex, region, ethnicity and trial arm (and HAQ score at 6 months for 12-month imputations). Imputations of missing QALYs at 6 and 12 months were based on baseline HAQ score, duration of illness, age, sex, region, ethnicity, trial arm and equivalent baseline utility score (and utility score at 6 months for 12-month imputations). Cost and outcome data for the resulting imputed full sample were analysed and presented as per the base (available) case data.
Cost-effectiveness and cost–utility analyses
Accounting for the three cost perspectives and three outcomes, there were nine possible cost–outcome combinations to consider in the economic evaluation. ICERs were calculated for any combination that showed both significantly higher costs and better outcomes in either the intervention group or the control group.
Uncertainty around cost-effectiveness/cost–utility was explored using cost-effectiveness acceptability curves (CEACs) based on the net-benefit approach. This followed the Bayesian approach to cost-effectiveness analysis outlined by Briggs. 186 These curves address some of the problems associated with examining ICERs and show the probability that one intervention is cost-effective compared with another other for a range of values that a decision-maker would be willing to pay for an additional unit of each outcome (i.e. per additional QALY or per additional point improvement in HAQ score). Net benefits for each participant were calculated using the following formula, where λ is the willingness to pay for one additional unit of outcome:
A series of net benefits were calculated for each individual for λ values ranging between £0 and £50,000 per QALY gained and per point improvement on the HAQ. After calculating net benefits for each participant for each value of λ, coefficients of differences in net benefits between the trial arms were obtained through a series of bootstrapped linear regressions (1000 repetitions) of group upon net benefit, which included the baseline values of the same cost category and the same outcome as covariates plus baseline HAQ score, duration of illness, age, sex, region and ethnicity. The resulting coefficients were then examined to calculate for each value of λ the proportion of times that the cDMARDs group had a greater net benefit than the TNFi group. These proportions were then plotted to generate CEACs for all three outcomes from the health and social care perspective at 6 and 12 months.
Systematic review methods
The systematic reviews were carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2009 checklist. 187
Search strategies
Ovid MEDLINE and EMBASE were searched from 1946 to 2013. The terms used in the search strategies are found in Appendix 5. This was then limited to English language and clinical trials. The Cochrane Library was also searched using the terms ‘early rheumatoid arthritis and combination therapy’ or ‘early rheumatoid arthritis and anti-TNF’ for the early RA search and ‘rheumatoid arthritis and combination therapy’ or ‘rheumatoid arthritis and anti-TNF’ for the established RA search. The titles and abstracts were then assessed by two reviewers (MM, DS) independently. If there were any doubts regarding the eligibility of a particular study it was discussed between the two reviewers until agreement was reached.
Selection criteria
Early rheumatoid arthritis
The following criteria were used to select studies for evaluation:
-
the study was a RCT
-
patients fulfilled the ACR classification criteria for RA
-
disease duration was < 3 years (this threshold was chosen to maximise the number of studies included in this systematic review)
-
the ‘treatment’ arms comprised one or other or both of cDMARDs and TNFi/methotrexate
-
the ‘control’ arm comprised DMARD monotherapy.
Established rheumatoid arthritis
The following criteria were used to select studies for evaluation:
-
the study was a RCT
-
patients fulfilled the ACR classification criteria for RA
-
patients were treatment resistant to at least one previous DMARD given for at least 3 months
-
the ‘treatment’ arms comprised one or other or both of cDMARDs and TNFi/methotrexate; when more than one dosage of TNFi was used the treatment arm that mirrored clinical practice the closest was chosen
-
the ‘control’ arm comprised DMARD monotherapy.
Assessing the risk of bias
The risk of bias was assessed using criteria recommended by Viswanathan et al. 188
Outcome measures
American College of Rheumatology responses and patient withdrawals because of inefficacy and toxicity
These dichotomous outcomes were used to calculate random-effects odds ratios (ORs) with 95% CIs. Patient withdrawals because of inefficacy and toxicity are routinely reported in clinical trials and are increasingly used as outcome measures. They have face validity and are used to assess effectiveness in clinical practice.
Health Assessment Questionnaire
Our meta-analysis included only studies reporting mean changes in HAQ scores; these were used to calculate random-effects weighted mean differences (WMDs).
Radiological progression
This was variously expressed as mean or median values, as changes over time or as final values. The meta-analysis included only studies reporting mean changes in radiological scores. To allow for different periods of observation, the annual rate of radiological progression was calculated (mean change in radiological score divided by duration of follow-up in years). As different radiological scoring systems were used (Sharp score, van der Heijde modified Sharp score and Larsen–Dale score), these were standardised as per cent maximal change (annual rate of progression divided by maximum possible score expressed as a percentage). The mean percentage annual changes were used to calculate random-effects WMDs.
Statistical analysis
Results were analysed using Review Manager 5.1.6 (The Cochrane Collaboration, The Nordic Cochrane Centre, Copenhagen, Denmark). The random-effects model based on DerSimonian and Laird’s method189 was used to estimate the pooled effect sizes; this gives more equal weighting to studies of different precision than a simple inverse variance weighted approach, so accommodating between-study heterogeneity. For all meta-analyses we performed Cochran’s chi-squared test to assess between-study heterogeneity and quantified I2-statistics. 190,191 We considered a p-value < 0.05 as statistically significant.
In RCTs with two or more combination arms, the treatment arm with the best outcome was used. We also carried out sensitivity analyses using methotrexate monotherapy as the comparator arm. Methotrexate is the most commonly used DMARD monotherapy and is considered the most effective DMARD. Therefore, including this DMARD as a comparator ensures that trials had an effective comparator agent.
Chapter 3 Results
Introduction
Results of the Tumour necrosis factor inhibitors Against Combination Intensive Therapy trial
The results of the TACIT trial are presented in five sections. The first section describes screening, randomisation, patients studied and the treatments that they received. The second section describes the impact of treatment on disability, quality of life and erosive damage; these outcomes were assessed every 6 months and include the HAQ, which was the primary outcome. The third section describes the impact of treatment on disease activity; it focuses on the DAS28 and its individual components, with outcomes assessed every month. The fourth section describes adverse effects encountered during the trial and the final section reports the economic evaluation.
The report presents the results from the ITT and complete-case populations. The results of the per-protocol population were similar to those of the ITT population and these findings are considered only in summary form. However, detailed tables of the complete-case analyses are presented in Appendix 3.
Results of the systematic reviews
The two systematic reviews focus on early RA and established RA and these are reported after the trial results.
The Tumour necrosis factor inhibitors Against Combination Intensive Therapy trial: screening, randomisation, patients studied and drug treatments
Screening and randomisation
Between September 2008 and December 2010 432 patients were screened at 24 rheumatology clinics in England. In total, 218 patients were excluded, 196 because they did not consent to participate and a further 20 because they were not eligible to participate; in addition, for two patients no reasons were recorded. The remaining 214 patients were randomised: 107 to receive cDMARDs and 107 to receive TNFis (Figure 1). The final assessment for the TACIT trial was in December 2011.
After randomisation three patients in the cDMARD group did not receive the intervention; this was because the patients changed their minds about participating in the trial after randomisation but before receiving treatment. There were also six patients in the TNFi group who did not receive the intervention. In three cases this was also because the patients changed their minds about taking part in the trial after randomisation but before receiving treatment. In one patient new information about a previous non-melanotic skin cancer resulted in the supervising rheumatologist considering that the patient should not receive biological treatment; in another patient the supervising rheumatologist changed his opinion about the suitability of the patient for intensive treatment; and the final patient mistakenly self-injected the TNFi, which had been delivered before the formal baseline assessment had been carried out.
Patients studied
Although 214 patients were randomised (107 to receive cDMARDs and 107 to receive TNFis), only 104 patients started cDMARDs and 101 patients started TNFis and the trial report focuses on these 205 treated patients. Their baseline characteristics are summarised in Table 3. Both groups had similar demographic characteristics including region, sex, ethnicity, disease duration, weight and height. Most clinical variables were also similar between groups including DAS28, the individual components of the DAS28, HAQ and EQ-5D scores, and SF-36 domain and summary scores. The only variable to show a baseline difference was Larsen score, with a mean score of 45.1 (SD 41.9) in the cDMARD group and 37.9 (SD 38.8) in the TNFi group.
Variable | cDMARDs (N = 104), n (%) | TNFis (N = 101), n (%) |
---|---|---|
Demographic variables | ||
Region | ||
London/south | 65 (63) | 63 (62) |
Midlands | 9 (9) | 7 (7) |
North | 30 (29) | 31 (31) |
Age (years), mean (SD) | 58 (13) | 57 (11) |
Sex | ||
Female | 73 (70) | 79 (78) |
Male | 31 (30) | 22 (22) |
Ethnic group | ||
White | 89 (86) | 92 (91) |
Black (African, Caribbean, black other) | 6 (6) | 2 (2) |
Asian (Bangladeshi/Indian, Pakistani) | 8 (8) | 6 (6) |
Chinese | 0 (0) | 1 (1) |
Other/mixed ethnic group | 1 (1) | 0 (0) |
Disease duration (years), median (IQR) | 4.4 (1.6–9.9) | 5.9 (2.2–13.4) |
Height (m), mean (SD) | 1.64 (0.11) | 1.66 (0.09) |
Weight (kg), mean (SD) | 78.3 (19.5) | 80.6 (16.9) |
BMI (kg/m2), median (IQR) | 28.5 (23.8–32.8) | 29.0 (25.0–32.4) |
Clinical variables | ||
DAS28, mean (SD) | 6.21 (0.92) | 6.30 (0.81) |
Tender joint count, mean (SD) | 16.4 (7.1) | 17.5 (6.74) |
Swollen joint count, mean (SD) | 10.5 (6.1) | 10.8 (6.74) |
ESR (mm/hour), mean (SD) | 33.1 (26.1) | 30.1 (22.84) |
VAS, mean (SD) | 68.1 (19.7) | 68.2 (21.30) |
HAQ score, mean (SD) | 1.80 (0.59) | 1.90 (0.67) |
Larsen score, mean (SD) | 45.1 (41.9) | 37.9 (38.8) |
EQ-5D utility score, mean (SD) | 0.39 (0.31) | 0.35 (0.31) |
SF-36 scores, mean (SD) | ||
Physical functioning | 30.1 (22.6) | 24.6 (21.0) |
Role physical | 14.9 (30.1) | 12.4 (26.1) |
Pain | 28.1 (16.3) | 26.3 (17.8) |
General health perception | 35.8(18.2) | 31.4 (16.8) |
Vitality | 30.3 (21.4) | 26.6 (19.0) |
Social functioning | 50.2 (25.2) | 42.1 (25.3) |
Role emotion | 43.9 (44.9) | 35.3 (44.9) |
Mental health | 61.9 (20.2) | 58.8 (23.1) |
PCS | 28.4 (6.8) | 27.3 (7.0) |
MCS | 43.4 (12.4) | 40.7 (12.3) |
During the 12 months of the trial 16 out of the 205 treated patients (8%) were lost to follow-up, nine patients in the cDMARD arm and seven in the TNFi arm (see Figure 1). A further 42 out of the 205 patients (20%) discontinued their intervention but remained under follow-up; these comprised 23 patients in the cDMARD arm and 19 patients in the TNFi arm.
In total, 10 out of 104 patients (10%) in the cDMARD arm stopped treatment because of toxicity, one (1%) stopped because of disease progression and 21 (15%) stopped for other reasons, including because a patient decided to stop treatment. In the TNFi arm six out of 101 (6%) patients stopped treatment because of toxicity, four (4%) stopped because of disease progression and 16 (16%) stopped for other reasons, including because a patient decided to stop treatment.
The main analysis evaluated 205 patients in the ITT group (104 in the cDMARD group and 101 in the TNFi group). The complete-case analysis evaluated the 147 completers (72 in the cDMARD group and 75 in the TNFi group).
Drug treatments
General
Drug treatments have been considered in two different ways: first, by describing treatments received by individual patients and, second, by reporting the numbers of prescriptions issued.
Patient received drug treatments in the TACIT trial using standard NHS prescribing mechanisms. DMARDs and steroids were prescribed from hospital pharmacies and were usually immediately available. TNFis were either delivered to patients’ homes (adalimumab and etanercept) by a private company (Healthcare at Home, Burton on Trent, Staffordshire, UK) or given as day-case infusions (infliximab) in hospital. In both circumstances there were variable delays between making the clinical decision to initiate TNFis and starting treatment so that home deliveries or infusions could be arranged. These delays varied from weeks to months. In the group randomised to receive TNFis, the timing of the baseline assessment was adjusted to enable this initial assessment to coincide with delivery of the TNFis or the first infusion. However, when patients switched to start a TNFi (in the group randomised to start cDMARDs) or changed TNFis (in the group randomised to start TNFis), then the delay in starting a TNFi occurred with the trial time frame.
Individual treatments in the combination disease-modifying antirheumatic drug group
Most patients received combinations of two or three DMARDs during the course of the trial (Table 4). A minority had four or five DMARDs. As patients were receiving DMARDs when enrolled, combinations were usually given in a step-up approach. The most common combination was methotrexate and leflunomide. Other frequently used combinations included methotrexate and ciclosporin, methotrexate, sulfasalazine and hydroxychloroquine and methotrexate and gold. A variety of other combinations were used occasionally.
Therapies | No. of patients |
---|---|
No. of DMARDs combined | |
One | 0 |
Two | 46 |
Three | 48 |
Four | 8 |
Five | 2 |
Total | 104 |
DMARD combinations | |
Methotrexate/leflunomide | 62 |
Methotrexate/ciclosporin | 17 |
Methotrexate/sulfasalazine/hydroxychloroquine | 13 |
Methotrexate/gold | 10 |
Other | 2 |
Total | 104 |
Use of steroids | |
Oral prednisolone | 24 |
Depo-Medrone injections | 3 |
Total | 27 |
Switched to TNFis | |
Adalimumab | 25 |
Etanercept | 14 |
Infliximab | 4 |
Withdrew before startinga | 3 |
Total | 46 |
In total, 27 patients received glucocorticoids (steroids). In 24 patients these were given as oral prednisolone. Most of these patients receiving steroids (16 cases) had oral prednisolone combined with two DMARDs. An additional three patients received Depo-Medrone injections.
After 6 months, patients who had failed to achieve a decrease in DAS28 of ≥ 1.2 could receive a TNFi. Altogether, 46 out of 104 patients (44%) were recommended to switch to a TNFi. Three patients withdrew from the trial before starting a TNFi and therefore 43 out of 104 patients (41%) actually switched to a TNFi. The majority of these patients received adalimumab (see Table 4). The times at which the new treatments were actually started (as opposed to when they were recommended) are shown in Figure 2. TNFis were started after an average of 9 months (range 7–12 months).
Individual treatments in the tumour necrosis factor inhibitor group
Most patients received adalimumab with etanercept and infliximab being used less often (Table 5). All patients received DMARDs: these comprised methotrexate (82 patients), sulfasalazine (13 patients), leflunomide (10 patients) and hydroxychloroquine (eight patients). In 13 patients the initial DMARD treatment involved a combination of two or more DMARDs. These combinations reflected pretrial treatment regimens and they were reduced to monotherapies during the trial.
Therapies | No. of patients |
---|---|
Initial TNFi | |
Adalimumab | 58 |
Etanercept | 34 |
Infliximab | 9 |
Total | 101 |
Second TNFi | |
Adalimumab | 7 |
Etanercept | 9 |
Infliximab | 0 |
Total | 16 |
Use of steroids | |
Oral prednisolone | 19 |
Depo-Medrone injections | 0 |
Total | 19 |
In total, 19 patients received glucocorticoids (steroids). All of these patients received oral steroids with none receiving Depo-Medrone injections.
After 6 months 16 patients received a second TNFi. Four patients were subsequently recommended to switch to DMARDs; however, none of these patients completed the trial.
Numbers of prescriptions
Overall, 4608 prescriptions were issued in the TACIT trial, 2418 for patients in the DMARD group and 2190 for patients in the TNFi group (Table 6). The most widely used DMARD was methotrexate, followed by leflunomide, hydoxychloroquine and sulfasalazine. The most widely used TNFi was adalimumab.
Treatment | cDMARDs (n = 104) | TNFis (n = 101) | Total (n = 205) |
---|---|---|---|
Methotrexate | 810 | 830 | 1640 |
Leflunomide | 435 | 75 | 510 |
Hydroxychloroquine | 355 | 72 | 427 |
Sulfasalazine | 226 | 97 | 323 |
Ciclosporin | 128 | 0 | 128 |
Gold injections | 61 | 0 | 61 |
Penicillamine | 26 | 0 | 26 |
Azathioprine | 1 | 2 | 3 |
Prednisolone | 199 | 134 | 333 |
Depo-Medrone | 3 | 0 | 3 |
Adalimumab | 112 | 580 | 692 |
Etanercept | 49 | 331 | 380 |
Infliximab | 13 | 69 | 82 |
Total | 2418 | 2190 | 4608 |
Disability, quality of life and erosive damage (assessed every 6 months)
The outcome measures specifically collected every 6 months include the primary outcome measure, HAQ score, and three secondary outcome measures – EQ-5D score, SF-36 scores and Larsen score for radiological progression.
Changes in Health Assessment Questionnaire scores
Primary outcome in intention-to-treat population
Initial HAQ scores were similar in patients randomised to receive cDMARDs (mean 1.80, 95% CI 1.68 to 1.91) and TNFis (mean 1.90, 95% CI 1.77 to 2.03) (Table 7).
Measure | cDMARDs (n = 104) | TNFis (n = 101) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | |
HAQ score | 1.80 (1.68 to 1.91) | 1.52 (1.39 to 1.65) | 1.35 (1.20 to 1.50) | 0.28 (0.18 to 0.38) | 0.45 (0.34 to 0.55) | 1.90 (1.77 to 2.03) | 1.55 (1.39 to 1.71) | 1.59 (1.43 to 1.76) | 0.35 (0.23 to 0.46) | 0.30 (0.19 to 0.42) |
EQ-5D score | 0.39 (0.33 to 0.45) | 0.53 (0.48 to 0.59) | 0.59 (0.53 to 0.65) | −0.14 (−0.20 to −0.08) | −0.20 (−0.27 to −0.13) | 0.35 (0.28 to 0.41) | 0.52 (0.46 to 0.58) | 0.49 (0.43 to 0.55) | −0.17 (−0.23 to −0.11) | −0.14 (−0.21 to −0.08) |
Larsen score | 45.1 (37.0 to 53.2) | 45.9 (37.7 to 54.0) | 46.3 (38.1 to 54.5) | −0.78 (−1.65 to −0.02) | −1.26 (−2.34 to −0.19) | 37.9 (30.2 to 45.6) | 38.7 (30.81 to 46.6) | 39.3 (31.2 to 47.4) | −0.81 (−1.65 to 0.02) | −1.37 (−2.48 to −0.26) |
After 12 months HAQ scores had changed by a mean of 0.45 (95% CI 0.34 to 0.55) in patients randomised to cDMARDs and by a mean of 0.30 (95% CI 0.19 to 0.42) in patients randomised to TNFis (see Table 7). The unadjusted and adjusted linear regression analyses (Table 8) showed that patients randomised to start cDMARDs had a greater reduction in HAQ score than those randomised to start TNFis. The unadjusted coefficient (adjusted for region only) was 0.14 (95% CI −0.01 to 0.29). After adjusting for demographic factors (age, sex, ethnicity, disease duration and region) and baseline score the adjusted coefficient was 0.15 (95% CI −0.003 to 0.31). The unadjusted linear regression analysis showed that the reduction in HAQ score was of borderline statistical significance in patients randomised to cDMARDs (p = 0.075). After adjusting for demographic factors and baseline score, the reduction in HAQ score was of stronger statistical significance in patients in the cDMARDs group (p = 0.046) (see Table 8).
Outcome | Model 1 (unadjusted): treatment + region | Model 2 (adjusted): treatment + demographics + baseline score | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
Change in HAQ score | ||||
12 months | 0.14 (−0.01 to 0.29) | 0.075 | 0.15 (0.00 to 0.31) | 0.047 |
6 months | −0.07 (−0.22 to .08) | 0.360 | −0.08 (−0.23 to 0.07) | 0.311 |
Change in EQ-5D score | ||||
12 months | −0.06 (−0.15 to 0.04) | 0.245 | −0.11 (−0.18 to −0.03) | 0.009 |
6 months | 0.03 (−0.06 to 0.11) | 0.500 | 0.01 (−0.07 to 0.08) | 0.882 |
Change in Larsen score | ||||
12 months | 0.11 (−1.45 to 1.67) | 0.891 | 0.35 (−1.37 to 2.06) | 0.689 |
6 months | 0.03 (−1.09 to 1.15) | 0.958 | 0.24 (−1.02 to 1.51) | 0.704 |
Change in SF-36 PCS | ||||
12 months | −0.23 (−3.26 to 2.79) | 0.880 | −1.40 (−4.22 to 1.41) | 0.327 |
6 months | 2.66 (1.50 to 3.83) | <0.001 | 1.75 (0.64 to 2.86) | 0.002 |
Change in SF-36 MCS | ||||
12 months | 0.42 (−3.51 to 4.35) | 0.832 | −1.73 (−5.07 to 1.61) | 0.307 |
6 months | 0.68 (−3.17 to 4.54) | 0.728 | −1.62 (−4.94 to 1.70) | 0.336 |
The minimum clinically detectable difference in HAQ score is 0.22. The difference in HAQ change scores from 0 to 12 months between patients starting cDMARDs and those starting TNFis was 0.15 and the 95% CIs fell within 0.22 of this difference. The TACIT trial therefore provides no evidence of a clinically important difference in 12-month HAQ scores between groups.
Six-month outcomes in the intention-to-treat population
At 6 months the HAQ score decreased by a mean of 0.28 (95% CI 0.18 to 0.38) in patients randomised to cDMARDs and by a mean of 0.35 (95% CI 0.23 to 0.46) in patients randomised to TNFis (see Table 7). This difference was not significant in either the unadjusted or the adjusted model (see Table 8). The overall pattern of change is shown in Figure 3.
The effects of patients in the combination disease-modifying antirheumatic drug group switching to tumour necrosis factor inhibitors
In total, 58 of the 104 patients in the cDMARD group remained on cDMARDs and 46 switched to TNFis after 6 months. Over 12 months both sets of patients showed similar changes in HAQ score and there was no evidence of a difference between groups by linear regression analysis (Table 9 and Figure 4). Comparing changes in HAQ scores in both of these groups using general estimating equations provided no evidence that there were any significant differences between the two groups in both an unadjusted and an adjusted model (Table 10).
Measure | Stayed on cDMARDs (n = 58) | Changed to TNFis (n = 46) | ||||||
---|---|---|---|---|---|---|---|---|
Initial | 6 months | 12 months | Change 0–12 months | Initial | 6 months | 12 months | Change 0–12 months | |
HAQ score | 1.82 (1.65 to 1.98) | 1.42 (1.23 to 1.60) | 1.38 (1.17 to 1.60) | 0.43a (0.29 to 0.58) | 1.77 (1.62 to 1.92) | 1.64 (1.46 to 1.82) | 1.31 (1.10 to 1.51) | 0.46 (0.30 to 0.62) |
EQ-5D score | 0.35 (0.27 to 0.43) | 0.57 (0.50 to 0.64) | 0.61 (0.53 to 0.69) | −0.26b (−0.35 to −0.17) | 0.44 (0.35 to 0.52) | 0.49 (0.40 to 0.57) | 0.57 (0.48 to 0.65) | −0.13 (−0.24 to −0.02) |
Larsen score | 44.7 (33.6 to 55.8) | 45.3 (34.1 to 56.5) | 45.9 (34.7 to 57.0) | −1.13c (−2.63 to 0.38) | 45.5 (33.4 to 57.6) | 46.5 (34.3 to 58.7) | 46.9 (34.6 to 59.3) | −1.43 (−2.92 to 0.06) |
Outcome | Model 1 (unadjusted): treatment + region + time | Model 2 (adjusted): treatment + demographics + baseline score + time | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
Change in HAQ score | ||||
12 months | 0.14 (−0.03 to 0.31) | 0.103 | 0.12 (−0.05 to 0.29) | 0.185 |
Complete-case analysis
Initial HAQ scores and changes in HAQ scores were similar between patients randomised to cDMARDs and those randomised to TNFis. There was no evidence of any significant differences between groups (see Appendix 3, Tables 50 and 52 and Figure 28). However, in the longitudinal analysis (see Appendix 3, Table 55), there was some evidence of a treatment difference in the unadjusted and adjusted models.
Changes in European Quality of Life-5 Dimensions scores
Intention-to-treat population
Initial EQ-5D scores were similar in patients randomised to receive cDMARDs (mean 0.39, 95% CI 0.33 to 0.45) and patients randomised to receive TNFis (mean 0.35, 95% CI 0.28 to 0.41) (see Table 7).
At 12 months EQ-5D scores changed by a mean of −0.20 (95% CI −0.27 to −0.13) in patients randomised to cDMARDs and by a mean of −0.14 (95% CI −0.21 to −0.08) in patients randomised to TNFis (see Table 7). There was no significant difference between groups in the unadjusted model (see Table 8). The adjusted model, in which the coefficient was −0.11 (95% CI −0.18 to −0.03), showed a significant change in EQ-5D score in patients randomised to cDMARDs compared with those randomised to TNFis (p = 0.009).
At 6 months EQ-5D scores changed by a mean of −0.14 (95% CI −0.20 to −0.08) in patients randomised to cDMARDs and by −0.17 (95% CI −0.23 to −0.11) in patients randomised to TNFis (see Table 7). The difference between treatment groups was not significant in either the unadjusted or the adjusted model (see Table 8). The overall pattern of change is shown in Figure 3.
The effects of patients in the combination disease-modifying antirheumatic drug group switching to tumour necrosis factor inhibitors
Over 12 months both sets of patients saw changes in EQ-5D scores (see Table 9 and Figure 4). In patients remaining on cDMARDs, EQ-5D scores improved by a mean of −0.26 (95% CI −0.35 to −0.17) and in patients switching to TNFis, EQ-5D scores improved by a mean of −0.13 (95% CI −0.24 to −0.02). Comparing these changes in EQ-5D scores over 12 months by linear regression (see Table 9) showed that the difference was of borderline significance (p = 0.069).
Complete-case analysis
Initial EQ-5D scores and changes in EQ-5D scores were similar between patients randomised to cDMARDs and those randomised to TNFis. There was no evidence of any significant differences between groups (see Appendix 3, Tables 50 and 52 and Figure 28).
Changes in Short Form Questionnaire-36 items scores
Intention-to-treat population
Changes in the SF-36 profiles and physical component score (PCS) and mental component score (MCS) are summarised in Table 11. There was a complex pattern of change. There were large mean changes (> 20) in the role physical domain at both 6 and 12 months in both groups. At 12 months physical functioning, pain, vitality, social functioning and role emotion showed changes of between 10 and 20 in both groups. General health perception and mental health showed smaller changes over 12 months (< 10) in both groups. We have not undertaken an in-depth statistical analysis of changes in the individual domains. However, longitudinal analyses assessing changes in these SF-36 domains at both 6 and 12 months (Table 12) mainly showed no significant differences between treatment groups in the unadjusted model or in the adjusted model.
Domain | cDMARDs (n = 104) | TNFis (n = 101) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | |
Physical functioning | 30.1 (25.8 to 34.5) | 36.7 (31.3 to 42.2) | 42.1 (36.4 to 47.7) | −6.58 (−12.2 to −0.9) | −11.9 (−17.5 to −6.3) | 24.6 (20.5 to 28.7) | 40.0 (34.4 to 45.5) | 37.8 (31.9 to 43.6) | −15.4 (−20.8 to −10.1) | −13.2 (−18.9 to −7.5) |
Role physical | 14.9 (9.1 to 20.7) | 36.2 (28.0 to 44.3) | 37.2 (28.2 to 46.1) | −21.3 (−30.7 to −11.8) | −22.3 (−31.9 to −12.6) | 12.4 (7.3 to 17.5) | 37.6 (29.2 to 46.1) | 33.1 (24.4 to 41.8) | −25.2 (−33.6 to −16.9) | −20.7 (−29.5 to −11.9) |
Pain | 28.1 (25.0 to 31.3) | 41.2 (37.4 to 45.1) | 46.4 (41.5 to 51.2) | −13.1 (−17.5 to −8.7) | −18.2 (−23.5 to −12.9) | 26.3 (22.8 to 29.8) | 45.5 (40.9 to 50.0) | 44.7 (40.0 to 49.4) | −19.2 (−24.1 to −14.3) | −18.4 (−24.0 to −12.9) |
General health perception | 35.8 (32.3 to 39.3) | 40.9 (37.1 to 44.8) | 44.6 (39.8 to 49.4) | −5.2 (−9.3 to −1.1) | −8.9 (−13.7 to −4.1) | 31.4 (28.1 to 34.7) | 44.1 (39.9 to 48.3) | 39.6 (35.3 to 44.0) | −12.7 (−17.1 to −8.3) | −8.2 (−12.8 to −3.7) |
Vitality | 30.3 (26.2 to 34.5) | 36.8 (32.5 to 41.2) | 40.4 (35.3 to 45.5) | −6.5 (−11.3 to −1.7) | −10.1 (−14.9 to −5.2) | 26.6 (22.9 to 30.3) | 40.4 (35.9 to 44.9) | 40.1 (35.4 to 44.8) | −13.8 (−18.4 to −9.2) | −13.5 (−18.5 to −8.4) |
Social functioning | 50.2 (45.4 to 55.1) | 61.6 (56.4 to 66.8) | 66.2 (60.6 to 71.8) | −11.4 (−16.6 to −6.1) | −16.0 (−22.0 to −9.9) | 42.1 (37.1 to 47.0) | 58.9 (53.6 to 64.3) | 59.8 (54.0 to 65.5) | −16.8 (−22.8 to −10.9) | −17.7 (−24.2 to −11.1) |
Role emotion | 43.9 (35.2 to 52.6) | 58.3 (49.3 to 67.3) | 60.4 (50.8 to 70.0) | −14.4 (−25.1 to −3.6) | −16.5 (−28.0 to −5.0) | 35.3 (26.5 to 44.1) | 50.9 (41.7 to 60.1) | 52.1 (42.7 to 61.5) | −15.6 (−26.8 to −4.3) | −16.8 (−28.3 to −5.3) |
Mental health | 61.9 (58.0 to 65.8) | 68.1 (64.2 to 72.0) | 70.4 (66.3 to 74.5) | −6.2 (−10.6 to −1.8) | −8.5 (−13.3 to −3.7) | 58.8 (54.3 to 63.3) | 65.8 (61.4 to 70.2) | 67.8 (63.7 to 71.9) | −7.0 (−12.4 to −1.6) | −9.0 (−14.1 to −4.0) |
PCS | 28.4 (27.1 to 29.7) | 32.6 (30.7 to 34.4) | 34.4 (32.2 to 36.5) | −4.2 (−6.2 to −2.1) | −6.0 (−8.1 to −3.8) | 27.3 (25.9 to 28.7) | 34.9 (32.9 to 36.9) | 33.0 (31.1 to 35.0) | −7.6 (−9.5 to −5.8) | −5.8 (−7.9 to −3.7) |
MCS | 43.4 (41.0 to 45.8) | 47.0 (44.6 to 49.4) | 48.4 (46.0 to 50.8) | −3.6 (−6.1 to −1.1) | −5.0 (−7.8 to −2.2) | 40.7 (38.3 to 43.1) | 45.0 (42.4 to 47.6) | 46.1 (43.7 to 48.6) | −4.3 (−7.2 to −1.4) | −5.4 (−8.2 to −2.7) |
Variable | Model 1 (unadjusted): treatment + region | Model 2 (adjusted): treatment + demographics + baseline score | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
No period-specific treatment effects | ||||
SF-36 role physical | −0.77 (−6.52 to 4.97) | 0.793 | 0.40 (−4.74 to 5.53) | 0.879 |
SF-36 pain | −1.50 (−4.76 to 1.76) | 0.368 | −0.21 (−2.95 to 2.53) | 0.880 |
SF-36 vitality | −2.92 (−6.05 to 0.21) | 0.067 | −1.76 (−4.54 to 1.02) | 0.215 |
SF-36 social functioning | −1.81 (−5.66 to 2.03) | 0.356 | 1.80 (−1.43 to 5.04) | 0.274 |
SF-36 role emotion | −0.43 (−7.64 to 6.78) | 0.907 | 3.98 (−1.58 to 9.54) | 0.160 |
SF-36 mental health | −0.31 (−3.46 to 2.85) | 0.848 | 1.35 (−1.06 to 3.76) | 0.272 |
Period-specific treatment effects | ||||
Period (1−6 months) | ||||
SF-36 physical functioning | 8.69 (1.04 to 16.34) | 0.026 | 5.52 (−1.74 to 12.77) | 0.136 |
SF-36 general health perception | 7.37 (1.43 to 13.30) | 0.015 | 4.20 (−0.78 to 9.18) | 0.098 |
Period (7–12 months) | ||||
SF-36 physical functioning | 1.16 (−6.49 to 8.81) | 0.767 | −3.12 (−10.44 to 4.19) | 0.403 |
SF-36 general health perception | −0.79 (−7.24 to 5.66) | 0.81 | −4.14 (−10.05 to 1.76) | 0.169 |
Initial PCS scores were similar in the two groups: in patients randomised to cDMARDs the mean score was 28.4 (95% CI 27.1 to 29.7) and in patients randomised to TNFis the mean score was 27.3 (95% CI 25.9 to 28.7). At 12 months PCS scores changed by a mean of −6.0 (95% CI −8.1 to −3.8) in patients randomised to cDMARDs and by a mean of −5.8 (95% CI −7.9 to −3.7) in patients randomised to TNFis (see Table 11). There was no significant difference between the groups in the unadjusted or adjusted model on linear regression analysis (see Table 8). At 6 months PCS scores changed by a mean of −4.2 (95% CI −6.2 to −2.1) in patients randomised to cDMARDs and by a mean of −7.6 (95% CI −9.5 to −5.8) in patients randomised to TNFis (see Table 11). This difference was significant on linear regression analysis in both the unadjusted model (2.66, 95% CI 1.50 to 3.83; p < 0.001) and the adjusted model (−1.75, 95% CI 0.64 to 2.86; p = 0.002) (see Table 8).
Initial MCS scores were similar in the two groups: in patients randomised to cDMARDs the mean score was 43.4 (95% CI 41.0 to 45.8) and in patients randomised to TNFis the mean score was 40.7 (95% CI 38.3 to 43.1). At 12 months MCS scores changed by a mean of −5.0 (95% CI −7.8 to −2.2) in patients randomised to cDMARDs and by −5.4 (95% CI −8.2 to −2.7) in patients randomised to TNFis (see Table 11). There was no significant difference between the groups in the unadjusted or adjusted model (see Table 8). At 6 months MCS scores changed by a mean of −3.6 (95% CI −6.1 to −1.1) in patients randomised to cDMARDs and by a mean of −4.3 (95% CI −7.2 to −1.4) in patients randomised to TNFis (see Table 11). There was no significant difference between treatment groups in the unadjusted or adjusted model in linear regression analysis (see Table 8).
Complete-case analysis
Changes in SF-36 profiles and initial scores and changes in scores for the PCS and MCS were similar between patients randomised to cDMARDs and those randomised to TNFis. There was no evidence of a significant difference between groups in the longitudinal analysis (see Appendix 3, Tables 51 and 53).
Changes in Larsen scores
Intention-to-treat population
The initial Larsen scores differed between groups (see Table 7): in the cDMARD group the initial mean score was 45.1 (95% CI 37.0 to 53.2) and in the TNFi group it was 37.9 (95% CI 30.2 to 45.6). The Larsen score was the only clinical variable to show baseline differences and no clinical significance was attached to this difference.
Progression over 12 months was similar between the groups (Figure 5). With cDMARDs the Larsen score increased by 1.26 and with TNFis it increased by 1.37. Progression over 6 months was also similar. These differences between the treatment groups were not statistically significant (see Tables 8 and 12).
An exploratory analysis examined individual changes over 12 months using all observed data for both groups (Figure 6); this showed no evidence of a different pattern of progression between the groups.
Another exploratory analysis evaluated the development of one (increase in Larsen score of 2–5) or many new erosions (increase in Larsen score of > 5) using all observed data for both groups; this is summarised in Figure 7, showing that there were no differences between the groups. By the end of the trial, 23 out of 91 patients (25%) randomised to receive cDMARDs developed one new erosion and 12 out of 91 patients (13%) developed two or more erosions. In the group randomised to receive TNFis, 19 out of 93 patients (20%) developed one new erosion and 13 out of 93 patients (14%) developed two or more erosions.
The effects of patients in the combination disease-modifying antirheumatic drug group switching to tumour necrosis factor inhibitors
Over 12 months both sets of patients showed small increases in Larsen scores (see Table 9). In patients remaining on cDMARDs Larsen scores increased by a mean of −1.13 (95% CI −2.63 to 0.38) and in patients switching to TNFis Larsen scores increased by a mean of −1.43 (95% CI −2.92 to 0.06). Comparing these changes in Larsen scores over 12 months by linear regression provided no evidence that the difference was significant (see Table 11). We also examined individual changes over 12 months for both sets of patients using all observed data (Figure 8); this showed no evidence of a different pattern of progression between the two groups.
Complete-case analysis
Changes in Larsen scores were similar between patients randomised to cDMARDs and those randomised to TNFis. There was no evidence of a significant difference between groups (see Appendix 3, Tables 50 and 53).
Disease activity scores (assessed every month)
Outcomes that were collected monthly comprised the DAS28 and its components – tender joint count, swollen joint count, ESR and VAS patient global assessments. We assessed changes in DAS28 and changes in its components in the ITT population. We also assessed the occurrence of a clinical response (decrease in DAS28 of ≥ 1.2) and low DAS28 indicative of remission (DAS28 of ≤ 2.6) using all observed data; imputation was not undertaken for these summary data because evaluating clinical responses and DAS28 remissions were exploratory analyses rather than predefined analyses as explained in the statistical analysis plan (see Chapter 2).
Changes in Disease Activity Score for 28 Joints scores
Intention-to-treat population
Initial DAS28 were similar in both groups. In the cDMARD group the initial mean DAS28 was 6.21 (95% CI 6.04 to 6.39) and in the TNFi group the initial mean score was 6.30 (95% CI 6.14 to 6.46). The patterns of change are shown in Table 13 and Figure 9.
Month of assessment | cDMARDs (n = 104) | TNFis (n = 101) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
DAS28 | Tender joint count | Swollen joint count | ESR (mm/hour) | VAS | DAS28 | Tender joint count | Swollen joint count | ESR (mm/hour) | VAS | |
0 | 6.21 (6.04 to 6.39) | 16.38 (15.02 to 17.75) | 10.51 (9.34 to 11.68) | 33.14 (28.10 to 38.19) | 68.13 (64.32 to 71.95) | 6.30 (6.14 to 6.46) | 17.48 (16.15 to 18.80) | 10.79 (9.47 to 12.11) | 30.13 (25.65 to 34.61) | 68.18 (64.00 to 72.36) |
1 | 5.32 (5.05 to 5.59) | 11.81 (10.07 to 13.56) | 6.76 (5.55 to 7.97) | 32.44 (27.43 to 37.46) | 54.87 (49.90 to 59.84) | 4.67 (4.38 to 4.95) | 10.69 (8.93 to12.46) | 5.76 (4.53 to 6.99) | 19.56 (15.78 to 23.34) | 46.15 (41.09 to 51.20) |
2 | 5.02 (4.76 to 5.27) | 9.85 (8.36 to 11.34) | 5.72 (4.72 to 6.72) | 30.23 (25.51 to 34.95) | 51.96 (47.22 to 56.70) | 4.30 (3.98 to 4.61) | 7.84 (6.47 to 9.21) | 5.02 (3.74 to 6.31) | 21.54 (17.27 to 25.81) | 43.18 (38.14 to 48.22) |
3 | 4.92 (4.63 to 5.21) | 9.97 (8.36 to 11.59) | 5.37 (4.26 to 6.47) | 30.10 (24.81 to 35.39) | 50.94 (45.74 to 56.15) | 4.28 (3.95 to 4.60) | 7.71 (6.12 to 9.31) | 4.43 (3.27 to 5.59) | 23.44 (19.08 to 27.80) | 43.16 (38.10 to 48.23) |
4 | 4.73 (4.45 to 5.02) | 8.40 (7.03 to 9.78) | 4.91 (3.85 to 5.97) | 31.20 (26.05 to 36.36) | 45.38 (39.69 to 51.07) | 4.31 (3.97 to 4.64) | 8.16 (6.54 to 9.78) | 4.07 (2.95 to 5.20) | 22.23 (18.31 to 26.15) | 45.40 (39.48 to 51.31) |
5 | 4.66 (4.36 to 4.96) | 8.51 (6.98 to 10.04) | 5.44 (4.33 to 6.55) | 29.87 (24.95 to 34.79) | 43.62 (37.87 to 49.37) | 4.13 (3.81 to 4.44) | 7.75 (6.06 to 9.45) | 4.38 (3.15 to 5.62) | 20.51 (16.96 to 24.06) | 40.60 (35.02 to 46.18) |
6 | 4.78 (4.45 to 5.12) | 10.16 (8.39 to 11.93) | 6.11 (4.74 to 7.49) | 29.25 (24.16 to 34.33) | 48.25 (42.42 to 54.09) | 4.23 (3.89 to 4.58) | 8.57 (6.87 to 10.27) | 4.66 (3.41 to 5.90) | 21.80 (17.64 to 25.96) | 40.51 (34.98 to 46.03) |
7 | 4.57 (4.27 to 4.87) | 8.34 (6.68 to 10.01) | 5.76 (4.53 to 7.00) | 28.21 (23.03 to 33.39) | 42.51 (36.95 to 48.07) | 4.17 (3.81 to 4.53) | 8.01 (6.35 to 9.67) | 4.64 (3.34 to 5.93) | 21.01 (17.18 to 24.85) | 40.73 (34.72 to 46.73) |
8 | 4.25 (3.92 to 4.59) | 7.42 (5.98 to 8.85) | 4.54 (3.43 to 5.64) | 24.14 (19.29 to 28.99) | 41.30 (35.45 to 47.14) | 4.05 (3.69 to 4.41) | 6.62 (5.14 to 8.09) | 4.23 (3.04 to 5.42) | 21.77 (17.57 to 25.97) | 42.56 (36.91 to 48.22) |
9 | 4.21 (3.87 to 4.56) | 6.93 (5.40 to 8.47) | 3.99 (2.98 to 5.00) | 25.47 (20.57 to 30.37) | 41.56 (35.78 to 47.35) | 4.08 (3.73 to 4.42) | 6.68 (5.14 to 8.22) | 4.30 (3.07 to 5.52) | 22.92 (18.62 to 27.21) | 40.86 (35.36 to 46.36) |
10 | 4.05 (3.73 to 4.37) | 6.17 (4.74 to 7.60) | 3.69 (2.78 to 4.61) | 25.42 (20.44 to 30.40) | 38.33 (33.02 to 43.65) | 3.90 (3.56 to 4.24) | 6.51 (4.90 to 8.12) | 3.42 (2.33 to 4.51) | 21.48 (17.34 to 25.62) | 39.33 (33.64 to 45.03) |
11 | 4.03 (3.74 to 4.31) | 6.67 (5.09 to 8.25) | 3.27 (2.31 to 4.24) | 23.28 (18.69 to 27.87) | 40.64 (34.69 to 46.59) | 3.84 (3.48 to 4.20) | 6.16 (4.57 to 7.76) | 3.50 (2.25 to 4.75) | 22.01 (17.73 to 26.28) | 37.69 (31.82 to 43.56) |
12 | 4.04 (3.74 to 4.34) | 6.32 (4.88 to 7.77) | 3.39 (2.63 to 4.14) | 25.03 (20.41 to 29.65) | 39.21 (33.23 to 45.19) | 3.89 (3.53 to 4.24) | 6.81 (5.22 to 8.40) | 3.20 (2.25 to 4.14) | 20.32 (16.04 to 24.59) | 43.03 (36.79 to 49.27) |
By 6 months the DAS28 had fallen in the cDMARDs group to 4.78 (95% CI 4.45 to 5.12) and in the TNFis group to 4.23 (95% CI 3.89 to 4.58). By 12 months the DAS28 had fallen further in the cDMARDs group to 4.04 (95% CI 3.74 to 4.34) and in the TNFis group to 3.89 (95% CI 3.53 to 4.24). The initial change in DAS28 was greater in patients randomised to TNFis and there was a significant difference between groups within the first month of treatment. After 1 month the mean DAS28 in the cDMARDs group fell to 5.32 (95% CI 5.05 to 5.59) and the mean score in the TNFis group fell to 4.67 (95% CI 4.38 to 4.95, p = 0.001).
Longitudinal analysis (Table 14) showed that there was a significant difference between treatment groups over the whole 12-month period. Patients randomised to TNFis achieved greater overall reductions in DAS28 than those randomised to cDMARDs in both the unadjusted model (−0.48, 95% CI −0.79 to −0.17; p = 0.002) and the adjusted model (−0.40, 95% CI −0.69 to −0.10; p = 0.009). Comparing the initial and final treatment periods in the adjusted model showed a difference in the pattern of change. In the first 6 months there was a greater reduction in DAS28 in patients randomised to TNFis than in patients randomised to cDMARDs, with a coefficient of −0.63 (95% CI −0.93 to −0.34, p < 0.001). In the second period there was no difference between groups, with a coefficient of −0.19 (95% CI −0.55 to 0.18, p = 0.317).
Time period | Variable | Model 1 (unadjusted): treatment + region + time | Model 2 (adjusted): treatment + demographics + baseline score | ||
---|---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | ||
Months 1–6 | DAS28 | −0.68 (−0.99 to −0.37) | < 0.001 | −0.63 (−0.93 to −0.34) | < 0.001 |
Tender joint count | −2.42 (−4.22 to −0.63) | 0.008 | −1.79 (−3.31 to −0.26) | 0.022 | |
Swollen joint count | −1.35 (−2.76 to 0.07) | 0.062 | −1.16 (−2.20 to −0.12) | 0.029 | |
ESR (mm/hour) | −6.46 (−10.23 to −2.68) | 0.001 | −7.18 (−10.60 to −3.76) | < 0.001 | |
VAS | −6.97 (−13.10 to −0.84) | 0.026 | −6.41 (−11.66 to −1.15) | 0.017 | |
Months 7–12 | DAS28 | −0.31 (−0.69 to 0.07) | 0.111 | −0.19 (−0.55 to 0.18) | 0.317 |
Tender joint count | −1.10 (−3.22 to 1.01) | 0.307 | −0.13 (−1.79 to 1.53) | 0.879 | |
Swollen joint count | −0.69 (−2.27 to 0.88) | 0.388 | −0.31 (−1.36 to 0.75) | 0.570 | |
ESR (mm/hour) | −1.63 (−5.88 to 2.62) | 0.452 | −2.15 (−5.73 to 1.44) | 0.241 | |
VAS | 0.60 (−6.47 to 7.67) | 0.867 | 2.04 (−4.08 to 8.17) | 0.513 | |
Months 1–12 | DAS28 | −0.48 (−0.79 to −0.17) | 0.002 | −0.40 (−0.69 to −0.10) | 0.009 |
Tender joint count | −1.69 (−3.50 to 0.11) | 0.066 | −0.93 (−2.36 to 0.51) | 0.205 | |
Swollen joint count | −0.86 (−2.27 to 0.55) | 0.233 | −0.63 (−1.57 to 0.31) | 0.186 | |
ESR (mm/hour) | −4.04 (−7.67 to −0.40) | 0.029 | −4.62 (−7.77 to −1.47) | 0.004 | |
VAS | −2.83 (−8.85 to 3.20) | 0.358 | −1.96 (−7.04 to 3.11) | 0.448 |
Complete-case analysis
Mean DAS28 fell in both groups with treatment (see Appendix 3, Table 56 and Figure 30). Longitudinal analysis using GEEs showed that the decreases were significantly greater with TNFis (see Appendix 3, Table 57) in both the unadjusted model (p < 0.001) and the adjusted model (p < 0.001).
Changes in Disease Activity Score for 28 Joints components
Intention-to-treat population
Baseline tender joint counts, swollen joint counts, ESR and patient global assessments were similar in both groups and they all improved when patients received either cDMARDs or TNFis. The patterns of change are shown in Table 13 and Figures 10 and 11.
Longitudinal analysis (see Table 14) showed that in the overall adjusted model changes in ESR were significantly different between patients randomised to cDMARDs and those randomised to TNFis; the decrease in ESR was significantly larger in the TNFis group (coefficient −4.62, 95% CI −7.77 to −1.47; p = 0.004). There were no significant differences between treatment groups in the other components over the whole 6 months.
In the first 6 months of treatment the adjusted mean treatment effects for all of the components were significantly greater in patients randomised to TNFis than in those randomised to cDMARDs. In the second 6 months there were no statistically significant differences between the groups.
The speed of onset of changes was particularly marked for the ESR in patients randomised to TNFis. With cDMARDs the ESR fell from an initial mean of 33.1 (95% CI 28.1 to 38.2) mm/hour to 32.4 (95% CI 27.4 to 37.5) mm/hour by 1 month. With TNFis the ESR fell from an initial mean of 30.1 (95% CI 25.7 to 34.6) mm/hour to 19.6 (95% CI 15.8 to 23.3) mm/hour by 1 month.
Complete-case analysis
Tender joint counts, swollen joint counts, ESR and patient global assessments improved in both patients receiving cDMARDs and patients receiving TNFis (see Appendix 3, Table 56). Longitudinal analysis (see Appendix 3, Table 57) showed that the improvements were significantly greater in the TNFis group for tender joint counts, swollen joint counts and ESR in both the unadjusted model and the adjusted model.
The effects of patients in the combination disease-modifying antirheumatic drug group switching to tumour necrosis factor inhibitors
Patients were selected to switch from cDMARDs to TNFis after 6 months if the change in DAS28 was < 1.2. As a consequence, the mean DAS28 for the switchers would be expected to be more than that in those who remained on cDMARDs. This difference is shown in Table 15, together with changes in the individual components of the DAS28, and is also illustrated in Figure 12. The difference is confirmed to be significant in the longitudinal analysis shown in Table 16. The adjusted model showed a significant reduction in DAS28 in the switchers. The same effect was seen for the components of the DAS28 and was most marked for tender joint count and patient global VAS score.
Month of assessment | Stayed on cDMARDs (n = 58) | Changed to TNFis (n = 46) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
DAS28 | Tender joint count | Swollen joint count | ESR (mm/hour) | VAS | DAS28 | Tender joint count | Swollen joint count | ESR (mm/hour) | VAS | |
0 | 6.10 (5.84 to 6.35) | 15.05 (13.11 to16.99) | 9.95 (8.45 to 11.44) | 34.07 (27.29 to 40.85) | 69.50 (64.19 to 74.81) | 6.36 (6.12 to 6.60) | 18.07 (16.24 to 19.89) | 11.22 (9.34 to 13.10) | 31.98 (24.26 to 39.70) | 66.41 (60.88 to 71.95) |
1 | 5.06 (4.69 to 5.42) | 10.14 (7.78 to 12.49) | 5.72 (4.39 to 7.06) | 32.76 (26.01 to 39.50) | 52.23 (45.54 to 58.92) | 5.66 (5.26 to 6.05) | 13.92 (11.39 to 16.46) | 8.07 (5.93 to 10.21) | 32.05 (24.35 to 39.74) | 58.21 (50.74 to 65.68) |
2 | 4.74 (4.40 to 5.07) | 8.17 (6.33 to 10.00) | 4.70 (3.49 to 5.92) | 30.11 (24.05 to 36.18) | 49.69 (43.34 to 56.05) | 5.37 (4.99 to 5.75) | 11.97 (9.64 to 14.30) | 7.01 (5.38 to 8.64) | 30.38 (22.77 to 37.98) | 54.81 (47.50 to 62.13) |
3 | 4.64 (4.27 to 5.01) | 8.54 (6.58 to 10.50) | 4.59 (3.08 to 6.09) | 28.94 (21.86 to 36.03) | 48.80 (41.44 to 56.16) | 5.27 (4.81 to 5.72) | 11.77 (9.12 to 14.42) | 6.35 (4.69 to 8.00) | 31.55 (23.45 to 39.66) | 53.65 (46.32 to 60.98) |
4 | 4.51 (4.15 to 4.87) | 7.04 (5.44 to 8.64) | 3.80 (2.59 to 5.00) | 31.11 (23.79 to 38.43) | 44.47 (36.92 to 52.02) | 5.01 (4.54 to 5.49) | 10.13 (7.78 to 12.47) | 6.32 (4.52 to 8.12) | 31.32 (24.17 to 38.47) | 46.52 (37.63 to 55.41) |
5 | 4.27 (3.90 to 4.65) | 6.43 (4.64 to 8.23) | 3.64 (2.46 to 4.81) | 29.31 (22.87 to 35.76) | 39.13 (31.94 to 46.32) | 5.15 (4.68 to 5.62) | 11.13 (8.73 to 13.53) | 7.72 (5.85 to 9.59) | 30.57 (22.88 to 38.27) | 49.28 (40.01 to 58.55) |
6 | 4.01 (3.61 to 4.40) | 6.27 (4.35 to 8.19) | 3.20 (1.78 to 4.62) | 27.66 (20.92 to 34.40) | 37.26 (29.75 to 44.78 | 5.76 (5.32 to 6.19) | 15.07 (12.47 to 17.66) | 9.78 (7.65 to 11.91) | 31.24 (23.35 to 39.14) | 62.11 (54.51 to 69.71) |
7 | 4.09 (3.71 to 4.46) | 5.85 (3.79 to 7.90) | 3.91 (2.40 to 5.41) | 28.87 (21.57 to 36.18) | 38.04 (30.16 to 45.92) | 5.18 (4.74 to 5.61) | 11.49 (8.97 to 14.01) | 8.11 (6.25 to 9.97) | 27.37 (20.00 to 34.74) | 48.15 (40.36 to 55.93) |
8 | 4.13 (3.68 to 4.58) | 6.26 (4.51 to 8.00) | 3.55 (2.23 to 4.86) | 24.88 (18.17 to 31.58) | 42.35 (34.19 to 50.50) | 4.41 (3.91 to 4.92) | 8.87 (6.54 to 11.21) | 5.79 (3.97 to 7.60) | 23.20 (16.01 to 30.40) | 39.97 (31.60 to 48.34) |
9 | 4.15 (3.69 to 4.62) | 5.75 (3.87 to 7.63) | 3.46 (2.12 to 4.81) | 26.58 (20.08 to 33.07) | 43.31 (34.89 to 51.73) | 4.29 (3.78 to 4.80) | 8.43 (6.02 to 10.84) | 4.64 (3.03 to 6.26) | 24.07 (16.40 to 31.74) | 39.36 (31.28 to 47.44) |
10 | 3.91 (3.48 to 4.34) | 5.23 (3.51 to 6.96) | 3.35 (2.11 to 4.59) | 25.67 (18.55 to 32.80) | 37.16 (29.52 to 44.81) | 4.23 (3.75 to 4.72) | 7.35 (5.03 to 9.67) | 4.12 (2.77 to 5.48) | 25.10 (18.29 to 31.92) | 39.81 (32.33 to 47.29) |
11 | 3.87 (3.48 to 4.25) | 5.44 (3.54 to 7.33) | 2.63 (1.40 to 3.85) | 22.65 (16.69 to 28.61) | 40.56 (31.84 to 49.27) | 4.23 (3.77 to 4.69) | 8.22 (5.66 to 10.78) | 4.09 (2.53 to 5.65) | 24.08 (16.79 to 31.38) | 40.74 (32.36 to 49.13) |
12 | 3.91 (3.52 to 4.31) | 5.37 (3.66 to 7.08) | 2.87 (1.83 to 3.91) | 26.39 (20.45 to 32.33) | 38.33 (29.89 to 46.78 | 4.19 (3.73 to 4.66) | 7.52 (5.08 to 9.97) | 4.04 (2.93 to 5.15) | 23.33 (15.89 to 30.76) | 40.32 (31.89 to 48.75) |
Variable | Model 1 (unadjusted): treatment + region + time | Model 2 (adjusted): treatment + demographics + baseline score | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
DAS28 | 0.35 (−0.03 to 0.74) | 0.071 | 0.51 (0.16 to 0.86) | 0.005 |
Tender joint count | 0.69 (−1.47 to 2.85) | 0.532 | 2.42 (0.75 to 4.10) | 0.004 |
Swollen joint count | 0.97 (−1.04 to 2.97) | 0.344 | 1.94 (0.65 to 3.22) | 0.003 |
ESR (mm/hour) | 2.54 (−2.65 to 7.73) | 0.338 | 2.44 (−1.97 to 6.84) | 0.278 |
VAS | 10.26 (2.70 to 17.83) | 0.008 | 8.29 (2.14 to 14.44) | 0.008 |
Achieving a clinical response
Time to achieve a response
An exploratory analysis examined the time taken to achieve a clinically meaningful response – a decrease in DAS28 of ≥ 1.2. The time to achieve a clinically meaningful response was compared between the groups using Kaplan–Meier plots (Figure 13). In total, 98 of 104 patients (94%) randomised to receive cDMARDs and 94 of 101 patients (93%) randomised to receive TNFis achieved such responses. The responses occurred sooner in the patients randomised to TNFis and this difference was significant in a log-rank test (p = 0.035). Patients randomised to receive cDMARDs who had a clinically meaningful DAS28 response achieved it within a mean of 3 months. Patients randomised to receive TNFis who had a clinically meaningful DAS28 response achieved it within a mean of 2 months.
Persistence of response
There was a complex pattern of achieving responses. In some patients the response was persistent and in others it was unsustained. Examples of these variations are shown in Figure 14 for four patients randomised to the TNFi group. As a consequence of these variations we evaluated the frequency of response each month in the two treatment groups (Figure 15). There was a different pattern of response in the two groups. Patients randomised to cDMARDs showed a gradual increase in the rate of response from ≤ 45% at 3 months or earlier to > 70% by 10 months. By contrast, patients randomised to TNFis achieved a response rate of > 70% by 2 months and the response rate remained at > 70% thereafter, with the highest response rate achieved in this group being 84% (achieved at month 11).
Impact of switching from combination disease-modifying antirheumatic drugs to tumour necrosis factor inhibitors
There was a difference in response rate between the patients randomised to cDMARDs who remained on cDMARDs and those who were randomised to cDMARDs but who switched to TNFis (Figure 16). Patients remaining on cDMARDs had a response rate of > 50% from 2 months onwards and after 6 months the response rate increased to > 70%. Those patients who switched to TNFis had an initial response rate of < 50% and the response rate did not increase to 70% until 10 months.
Achieving a Disease Activity Score for 28 Joints of ≤ 2.6
Time to achieve a Disease Activity Score for 28 Joints of ≤ 2.6
The time taken to achieve remission (DAS28 of ≤ 2.6) was compared using Kaplan–Meier plots (Figure 17). In total, 36 out of 104 patients (35%) randomised to receive cDMARDs and 44 out of 101 patients (44%) randomised to receive TNFis achieved remission at any time. There was no evidence that the speed of onset of remission was significantly different between groups (p = 0.085). Those patients randomised to receive both cDMARDs and TNFis who achieved DAS28 remission achieved it within a mean of 4 months.
Persistence of Disease Activity Score for 28 Joints of ≤ 2.6
There was a complex pattern of achieving remission. In some patients remission was persistent and in others it was unsustained. Examples of these variations are shown in Figure 18 for four patients randomised to the TNFi group. As a consequence of these variations we have also evaluated the frequency of response each month for each group (Figure 19). There was a different pattern of response between the groups. Patients randomised to cDMARDs showed a gradual increase in the rate of response from ≤ 5% at 3 months or earlier to a maximum of 20% by 12 months. By contrast, those patients randomised to TNFis had achieved a remission rate of 16% by 3 months, which gradually increased to a maximum of 32% by 11 months.
Impact of switching from combination disease-modifying antirheumatic drugs to tumour necrosis factor inhibitors
There was a difference in response rate between the patients randomised to cDMARDs who remained on cDMARDs and those randomised to cDMARDs who switched to TNFis (Figure 20). In both groups < 10% of patients achieved a DAS28 of ≤ 2.6 at ≤ 5 months. From 6 to 12 months between 13% and 24% of patients remaining on cDMARDs and between 5% and 21% of patients who switched to TNFis achieved a DAS28 of ≤ 2.6.
Adverse events
Serious adverse events
In total, 10 patients in the cDMARDs group had a serious adverse event, eight in the first 6 months and two in the second 6 months (Table 17). Seven of these serious adverse events involved or prolonged inpatient treatment. In the TNFis group, 19 patients had a serious adverse event, six in the first 6 months and 13 in the second 6 months; 13 of these serious adverse events involved or prolonged inpatient treatment. One patient in the TNFi group died from pneumonia and multiple organ failure during the second 6 months of treatment. Cardiovascular, respiratory, digestive and genitourinary systems were most commonly involved. Although there were more serious adverse events in the TNFi group, there was no evidence of major clinically important differences between the treatment groups and the frequency of adverse events was not significantly different (Fisher’s exact test p = 0.110).
Adverse event | cDMARDs | TNFis | ||||
---|---|---|---|---|---|---|
0–6 months | 6–12 months | Total | 0–6 months | 6–12 months | Total | |
Cardiovascular | 2 | 0 | 2 | 1 | 1 | 2 |
Digestive | 0 | 0 | 0 | 2 | 2 | 4 |
Ear, nose and throat | 0 | 0 | 0 | 0 | 1 | 1 |
Endocrine/metabolic | 0 | 0 | 0 | 1 | 0 | 1 |
Genitourinary | 3 | 0 | 3 | 0 | 1 | 1 |
Haematological | 1 | 0 | 1 | 0 | 1 | 1 |
Mental | 0 | 0 | 0 | 0 | 0 | 0 |
Musculoskeletal | 0 | 0 | 0 | 0 | 1 | 1 |
Nervous system | 0 | 1 | 1 | 1 | 1 | 2 |
Ophthalmological | 0 | 0 | 0 | 0 | 0 | 0 |
Respiratory | 2 | 1 | 3 | 1 | 2 | 3 |
Skin | 0 | 0 | 0 | 0 | 2 | 2 |
Total | 8 | 2 | 10 | 6 | 12 | 18 |
Patient died | 0 | 0 | 0 | 0 | 1 | 1 |
Involved/prolonged inpatient hospitalisation | 5 | 2 | 7 | 5 | 8 | 13 |
Life-threatening | 1 | 0 | 1 | 0 | 1 | 1 |
Stopping treatment because of adverse events
In total, 10 out of 104 patients (10%) in the cDMARD arm and six out of 101 patients (6%) in the TNFi arm stopped treatment because of toxicity (see Figure 1). Although more patients withdrew from treatment because of toxicity in the cDMARDs arm, there was no evidence of major clinically important differences between the treatment groups and the frequency of withdrawals because of adverse events as this was not significantly different (Fisher’s exact test p = 0.441).
Individual adverse events
There were 635 different adverse events reported by patients in the cDMARD group. The most frequent events are listed in Table 18 and they are grouped by system involved in Table 19. All reported events are listed in Appendix 4.
Adverse event | No. of events | Percentage of total no. of adverse events |
---|---|---|
cDMARDs group | ||
Diarrhoea | 30 | 4.7 |
Headache | 30 | 4.7 |
Nausea | 26 | 4.1 |
Vomiting | 26 | 4.1 |
Chest infection | 19 | 3.0 |
Flare of RA | 17 | 2.7 |
Sore throat | 15 | 2.4 |
Cold | 12 | 1.9 |
Ulcers – mouth | 12 | 1.9 |
Fatigue | 11 | 1.7 |
Dizziness | 9 | 1.4 |
Elevated alanine aminotransferase | 7 | 1.1 |
Flu | 7 | 1.1 |
High blood pressure | 7 | 1.1 |
Itchy skin | 7 | 1.1 |
Low white cell count | 7 | 1.1 |
TNFis group | ||
Chest infection | 27 | 5.8 |
Cold | 16 | 3.4 |
Elevated alanine aminotransferase | 16 | 3.4 |
Headache | 15 | 3.2 |
Flare of RA | 14 | 3.0 |
Sore throat | 13 | 2.8 |
Diarrhoea | 12 | 2.6 |
Urinary tract infection | 9 | 1.9 |
Nausea | 8 | 1.7 |
Breathlessness | 7 | 1.5 |
Cold sore | 7 | 1.5 |
Shoulder pain | 6 | 1.3 |
Upper respiratory tract infection | 6 | 1.3 |
Chest pain | 5 | 1.1 |
Cough – productive | 5 | 1.1 |
Fatigue | 5 | 1.1 |
Injection site reaction | 5 | 1.1 |
Vaginal thrush | 5 | 1.1 |
Knee pain | 5 | 1.1 |
Adverse event | cDMARDs group | TNFis group |
---|---|---|
Cardiovascular | 22 | 17 |
Digestive | 148 | 60 |
Ear, nose and throat | 88 | 76 |
Endocrine/metabolic | 7 | 7 |
Genitourinary | 28 | 27 |
Haematological | 25 | 10 |
Mental | 24 | 15 |
Musculoskeletal | 104 | 94 |
Nervous system | 61 | 41 |
Ophthalmological | 12 | 5 |
Respiratory | 59 | 66 |
Skin | 57 | 47 |
Total | 635 | 465 |
There were 465 different adverse events reported by patients in the TNFis group. The most frequent events are listed in Table 18 and they are grouped by system involved in Table 19. All reported events are listed in Appendix 4.
Chest infections (46 events), headaches (45 events), diarrhoea (42 events), nausea (34 events), sore throats (28 events), colds (28 events), elevated liver enzymes (alanine aminotransferase) (23 events) and fatigue (16 events) were the most common adverse events across both groups. Some types of adverse events spanned systems, in particular infections, which accounted for 112 adverse events in the cDMARDs group and 117 in the TNFis group.
There was no evidence of any major clinically important differences between the two treatment groups. However, the cDMARDs group had 37% more adverse events overall (635 vs. 465). This difference was mainly due to there being 88 more adverse events related to the digestive system (148 vs. 60) and 20 more adverse events related to the nervous system (61 vs. 41) in the cDMARDs group.
Economic evaluation
Response rates
The response rates for the CSRI and outcome questionnaires and the availability of trial medication data are summarised in Tables 20–22 respectively. These were > 90% and were similar for all of the questionnaires at baseline and 6 and 12 months and across both trial arms.
Group | Baseline | 6 months | 12 months | |||
---|---|---|---|---|---|---|
n | % | n | % | n | % | |
TNFis (n = 101) | 101 | 100 | 97 | 96 | 93 | 92 |
cDMARDs (n = 104) | 104 | 100 | 94 | 90 | 95 | 91 |
Total (n = 205) | 205 | 100 | 191 | 93 | 188 | 92 |
Group | Baseline | 6 months | 12 months | |||
---|---|---|---|---|---|---|
n | % | n | % | n | % | |
HAQ | ||||||
TNFis (n = 101) | 101 | 100 | 97 | 96 | 94 | 93 |
cDMARDs (n = 104) | 104 | 100 | 94 | 90 | 95 | 91 |
Total (n = 205) | 205 | 100 | 191 | 93 | 189 | 92 |
EQ-5D | ||||||
TNFis (n = 101) | 101 | 100 | 97 | 96 | 93 | 92 |
cDMARDs (n = 104) | 104 | 100 | 94 | 90 | 94 | 90 |
Total (n = 205) | 205 | 100 | 191 | 93 | 187 | 91 |
SF-36 | ||||||
TNFis (n = 101) | 101 | 100 | 97 | 96 | 94 | 93 |
cDMARDs (n = 104) | 104 | 100 | 94 | 90 | 95 | 91 |
Total (n = 205) | 205 | 100 | 191 | 93 | 189 | 92 |
Group | 6 months | 12 months | ||
---|---|---|---|---|
n | % | n | % | |
TNFis (n = 101) | 97 | 96 | 94 | 93 |
cDMARDs (n = 104) | 97 | 93 | 96 | 92 |
Total (n = 205) | 194 | 95 | 190 | 93 |
Table 23 summarises the joint availability of both cost and outcome data (a requirement for the construction of CEACs) by outcome measure. In total, 191 of the 205 study participants (93%) had both cost and outcome data at 6 months’ follow-up and 186–188 of the 205 study participants (91–92%) had both cost and outcome data at 12 months’ follow-up. There were thus very few cases excluded from the available case analyses.
Group | 6 months | 12 months | ||
---|---|---|---|---|
n | % | n | % | |
HAQ | ||||
TNFis (n = 101) | 97 | 96 | 93 | 92 |
cDMARDs (n = 104) | 94 | 90 | 95 | 91 |
Total (n = 205) | 191 | 93 | 188 | 92 |
EQ-5D | ||||
TNFis (n = 101) | 97 | 96 | 92 | 91 |
cDMARDs (n = 104) | 94 | 90 | 94 | 90 |
Total (n = 205) | 191 | 93 | 186 | 91 |
SF-36 | ||||
TNFis (n = 101) | 97 | 96 | 93 | 92 |
cDMARDs (n = 104) | 94 | 90 | 95 | 91 |
Total (n = 205) | 191 | 93 | 188 | 92 |
Tables 24–26 suggest that there were no notable differences in characteristics between the subsamples included in the available case analyses and the full sample.
Characteristic | Full sample (N = 205) | Subsample with 6-month cost and HAQ data (N = 191) | Subsample with 12-month cost and HAQ data (N = 188) | |||
---|---|---|---|---|---|---|
n | % | n | % | n | % | |
Sex | ||||||
Male | 53 | 26 | 45 | 24 | 46 | 25 |
Female | 152 | 74 | 146 | 76 | 142 | 76 |
Ethnicity | ||||||
White | 181 | 88 | 168 | 88 | 164 | 87 |
Other | 24 | 12 | 23 | 12 | 24 | 13 |
Region | ||||||
London and south | 128 | 62 | 127 | 67 | 121 | 64 |
Midlands | 16 | 8 | 13 | 7 | 13 | 7 |
North | 61 | 30 | 51 | 27 | 54 | 29 |
Mean | SD | Mean | SD | Mean | SD | |
Age (years) | 57.34 | 11.97 | 57.11 | 11.94 | 56.91 | 12.02 |
Duration of illness (years) | 8.20 | 8.82 | 8.35 | 8.98 | 8.24 | 8.88 |
HAQ score at baseline | 1.85 | 0.63 | 1.86 | 0.63 | 1.85 | 0.64 |
Characteristic | Full sample (N = 205) | Subsample with 6-month cost and EQ-5D data (N = 191) | Subsample with 12-month cost and EQ-5D data (N = 186) | |||
---|---|---|---|---|---|---|
n | % | n | % | n | % | |
Sex | ||||||
Male | 53 | 26 | 45 | 24 | 45 | 24 |
Female | 152 | 74 | 146 | 76 | 141 | 76 |
Ethnicity | ||||||
White | 181 | 88 | 168 | 88 | 162 | 87 |
Other | 24 | 12 | 23 | 12 | 24 | 13 |
Region | ||||||
London and south | 128 | 62 | 127 | 67 | 121 | 65 |
Midlands | 16 | 8 | 13 | 7 | 11 | 6 |
North | 61 | 30 | 51 | 27 | 54 | 29 |
Mean | SD | Mean | SD | Mean | SD | |
Age (years) | 57.34 | 11.97 | 57.11 | 11.94 | 56.84 | 12.08 |
Duration of illness (years) | 8.20 | 8.82 | 8.35 | 8.98 | 8.25 | 8.92 |
HAQ score at baseline | 1.85 | 0.63 | 1.86 | 0.63 | 1.85 | 0.64 |
EQ-5D-based utility at baseline | 0.37 | 0.31 | 0.37 | 0.31 | 0.37 | 0.31 |
Characteristic | Full sample (N = 205) | Subsample with 6-month cost and EQ-5D data (N = 191) | Subsample with 12-month cost and EQ-5D data (N = 186) | |||
---|---|---|---|---|---|---|
n | % | n | % | n | % | |
Sex | ||||||
Male | 53 | 26 | 45 | 24 | 46 | 25 |
Female | 152 | 74 | 146 | 76 | 142 | 76 |
Ethnicity | ||||||
White | 181 | 88 | 168 | 88 | 164 | 87 |
Other | 24 | 12 | 23 | 12 | 24 | 13 |
Region | ||||||
London and south | 128 | 62 | 127 | 67 | 121 | 64 |
Midlands | 16 | 8 | 13 | 7 | 13 | 7 |
North | 61 | 30 | 51 | 27 | 54 | 29 |
Mean | SD | Mean | SD | Mean | SD | |
Age (years) | 57.34 | 11.97 | 57.11 | 11.94 | 56.91 | 12.02 |
Duration of illness (years) | 8.20 | 8.82 | 8.35 | 8.98 | 8.24 | 8.88 |
HAQ score at baseline | 1.85 | 0.63 | 1.86 | 0.63 | 1.85 | 0.64 |
SF-36-based utility at baseline | 0.54 | 0.11 | 0.54 | 0.11 | 0.54 | 0.11 |
Resource use
Resource use differences between the groups were not compared statistically, first, because the economic evaluation was focused on costs and cost-effectiveness/utility and, second, to avoid problems associated with multiple testing. Therefore, resource use patterns are described in Tables 27–29 without statistical comparisons. Use of services appeared similar in both groups at all three time points. General practitioner (GP) surgery visits, practice nurse surgery visits, repeat prescription requests and hospital outpatient appointments were common in both groups at all time points, with other service use being relatively rare. The number of participants using non-trial medications was also similar in both groups at all time points.
Resource use | Unit | cDMARDs group (n = 94) | TNFis group (n = 97) | ||||
---|---|---|---|---|---|---|---|
No. of users | Mean usea | SD | No. of users | Mean usea | SD | ||
GP | |||||||
At surgery | Visit | 70 | 2 | 2 | 73 | 3 | 2 |
At home | Visit | 3 | 1 | 1 | 3 | 1 | 1 |
Telephone call | Call | 16 | 2 | 1 | 15 | 2 | 1 |
Repeat prescription request without GP contact | Prescription | 93 | 3 | 2 | 92 | 3 | 2 |
Nurse | |||||||
At surgery | Visit | 42 | 3 | 4 | 50 | 2 | 2 |
Telephone call | Call | 6 | 1 | < 1 | 7 | 1 | < 1 |
Physiotherapist | |||||||
At hospital | Unit | 7 | 2 | 2 | 9 | 2 | 1 |
At home | Visit | 0 | – | – | 1 | 3 | – |
At GP surgery | Visit | 2 | 11 | 13 | 1 | 3 | – |
Elsewhere | Visit | 0 | – | – | 0 | – | – |
Occupational therapist | |||||||
At hospital | Unit | 5 | 4 | 5 | 5 | 1 | 1 |
At home | Visit | 3 | 2 | 1 | 5 | 1 | 1 |
At GP surgery | Visit | 0 | – | – | 0 | – | – |
Elsewhere | Visit | 0 | – | – | 2 | 2 | < 1 |
Hospital services | |||||||
A&E | Unit | 9 | 1 | < 1 | 6 | 1 | < 1 |
Hospital stay overnight | Night | 4 | 2 | 1 | 3 | 12 | 6 |
Outpatient appointment | Unit | 77 | 3 | 2 | 85 | 3 | 2 |
Social services | |||||||
Meals on Wheels | Meal | 0 | – | – | 0 | – | – |
Home help | Visit | 0 | – | – | 1 | 90 | – |
Social worker | Hour | 0 | – | – | 2 | 3 | 1 |
Social worker telephone call | Call | 0 | – | – | 3 | 1 | < 1 |
Other health or social service | Contact | 1 | 1 | < 1 | 4 | 3 | 2 |
Non-trial medication | Medication | 102 | – | – | 100 | – | – |
Resource use | Unit | cDMARDs group (n = 94) | TNFis group (n = 97) | ||||
---|---|---|---|---|---|---|---|
No. of users | Mean usea | SD | No. of users | Mean usea | SD | ||
GP | |||||||
At surgery | Visit | 42 | 2 | 1 | 55 | 2 | 1 |
At home | Visit | 2 | 1 | < 1 | 3 | 2 | 1 |
Telephone call | Call | 9 | 2 | 1 | 14 | 1 | 1 |
Repeat prescription request without GP contact | Prescription | 63 | 3 | 1 | 70 | 3 | 1 |
Nurse | |||||||
At surgery | Visit | 31 | 3 | 3 | 31 | 3 | 4 |
Telephone call | Call | 2 | 2 | 1 | 2 | 1 | < 1 |
Physiotherapist | |||||||
At hospital | Unit | 8 | 4 | 3 | 4 | 3 | 1 |
At home | Visit | 0 | – | – | 0 | – | – |
At GP surgery | Visit | 2 | 3 | < 1 | 1 | 1 | – |
Elsewhere | Visit | 0 | – | – | 2 | 2 | 1 |
Occupational therapist | |||||||
At hospital | Unit | 4 | 2 | 1 | 3 | 1 | 1 |
At home | Visit | 2 | 1 | < 1 | 4 | 1 | < 1 |
At GP surgery | Visit | 0 | – | – | 0 | – | – |
Elsewhere | Visit | 1 | 1 | – | 0 | – | – |
Hospital services | |||||||
A&E | Unit | 4 | 1 | < 1 | 9 | 1 | < 1 |
Hospital stay overnight | Unit/night | 4 | 4 | 5 | 5 | 7 | 5 |
Outpatient appointment | Unit | 55 | 3 | 2 | 58 | 3 | 1 |
Social services | |||||||
Meals on Wheels | Meal | 1 | 60 | – | 0 | – | – |
Home help | Visit | 1 | 1 | – | 2 | 46 | 63 |
Social worker | Hour | 3 | 1 | 1 | 3 | 1 | 1 |
Social worker telephone call | Call | 1 | 2 | – | 1 | 3 | – |
Other health or social service | Contact | 3 | 31 | 51 | 3 | 14 | 11 |
Non-trial medication | Medication | 88 | – | – | 94 | – | – |
Resource use | Unit | cDMARDs group (n = 104) | TNFis group (n = 101) | ||||
---|---|---|---|---|---|---|---|
No. of users | Mean usea | SD | No. of users | Mean usea | SD | ||
GP | |||||||
At surgery | Visit | 60 | 2 | 1 | 58 | 2 | 2 |
At home | Visit | 4 | 2 | 1 | 3 | 1 | 1 |
Telephone call | Call | 16 | 1 | 1 | 13 | 1 | 1 |
Repeat prescription request without GP contact | Prescription | 68 | 3 | 2 | 61 | 2 | 1 |
Nurse | |||||||
At surgery | Visit | 24 | 2 | 1 | 31 | 2 | 2 |
Telephone call | Call | 2 | 1 | < 1 | 5 | 2 | 1 |
Physiotherapist | |||||||
At hospital | Unit | 11 | 5 | 6 | 7 | 3 | 2 |
At home | Visit | 0 | – | – | 0 | – | – |
At GP surgery | Visit | 1 | 8 | – | 2 | 3 | 3 |
Elsewhere | Visit | 1 | 1 | – | 1 | 2 | – |
Occupational therapist | |||||||
At hospital | Unit | 6 | 2 | 1 | 1 | 1 | – |
At home | Visit | 1 | 1 | – | 1 | 1 | – |
At GP surgery | Visit | 0 | – | – | 0 | – | – |
Elsewhere | Visit | 1 | 1 | – | 1 | 3 | – |
Hospital services | |||||||
A&E | Unit | 10 | 1 | < 1 | 5 | 1 | 1 |
Hospital stay overnight | Unit/night | 5 | 2 | 1 | 2 | 11 | 13 |
Outpatient appointment | Unit | 56 | 2 | 1 | 55 | 3 | 2 |
Social services | |||||||
Meals on Wheels | Meal | 0 | – | – | 0 | – | – |
Home help | Visit | 0 | – | – | 3 | 31 | 51 |
Social worker | Hour | 1 | 1 | – | 2 | 2 | < 1 |
Social worker telephone call | Call | 2 | 2 | 1 | 1 | 1 | – |
Other health or social service | Service | 2 | 19 | 16 | 2 | 1 | < 1 |
Non-trial medication | Contact | 90 | – | – | 91 | – | – |
Data on the use of NHS/social services-funded transport, equipment and home adaptations (costs of which are excluded from cost calculations) are presented in Table 30.
Resource use | cDMARDs group | TNFis group | ||||
---|---|---|---|---|---|---|
No. of users/total no. | No. paid by NHS | No. paid by social services | No. of users/total no. | No. paid by NHS | No. paid by social services | |
Baseline | ||||||
Transport | 5/104 | 4 | 1 | 3/101 | 2 | 1 |
Equipment | 4/104 | 1 | 3 | 2/101 | 0 | 2 |
Home adaptations | 4/104 | 1 | 3 | 1/101 | 0 | 1 |
Other | 2/104 | 1 | 1 | 3/101 | 1 | 2 |
6 months | ||||||
Transport | 6/94 | 6 | 0 | 4/97 | 3 | 1 |
Equipment | 2/94 | 0 | 2 | 4/97 | 0 | 4 |
Home adaptations | 4/94 | 2 | 2 | 3/97 | 0 | 3 |
Other | 0/94 | 0 | 0 | 2/97 | 0 | 2 |
12 months | ||||||
Transport | 2/95 | 2 | 0 | 6/93 | 6 | 0 |
Equipment | 3/95 | 1 | 2 | 2/93 | 0 | 2 |
Home adaptations | 1/95 | 1 | 0 | 3/93 | 0 | 3 |
Other | 1/95 | 0 | 1 | 1/93 | 0 | 1 |
Costs
Cost components at baseline, 6 months and 12 months are summarised in Table 31. Costs for both groups were equivalent at baseline. Costs of social security benefits and employment losses are small compared to the cost of health and social care. At 6 and 12 months’ follow-up all cost components remained equivalent between groups except for the cost of trial medications, which was significantly lower in the cDMARDs group (6-month adjusted mean difference −£3637, 95% CI −£3838 to −£3420; 12-month adjusted mean difference −£1894, 95% CI −£2320 to −£1427). The additional trial medication cost in the TNFis group overshadowed all other cost components in that group.
Cost component | TNFis group (N = 101) | cDMARDs group (N = 104) | Unadjusted differencea | Adjusted differenceb | ||||||
---|---|---|---|---|---|---|---|---|---|---|
Valid n | Mean cost (£) | SD (£) | Valid n | Mean cost (£) | SD (£) | Mean (£) | 95% CI (£) | Mean (£) | 95% CI (£) | |
Costs at baseline | ||||||||||
Health and social care, excluding trial medicationc | 101 | 736 | 1082 | 104 | 601 | 476 | −131 | −379 to 97 | – | – |
Employment lossesc | 101 | 60 | 262 | 104 | 84 | 440 | 24 | −66 to 131 | – | – |
Social security benefitsc | 101 | 71 | 76 | 104 | 63 | 67 | −9 | −29 to 12 | – | – |
Costs at 6 months | ||||||||||
Health and social care, excluding trial medicationc | 97 | 536 | 947 | 94 | 511 | 705 | −27 | −262 to 202 | 6 | −217 to 206 |
Employment lossesc | 97 | 71 | 405 | 94 | 35 | 310 | −35 | −127 to 67 | −35 | −115 to 59 |
Social security benefitsc | 97 | 77 | 75 | 94 | 74 | 77 | −2 | −21 to 21 | 3 | −15 to 19 |
Trial medicationd | 97 | 4166 | 1012 | 97 | 510 | 356 | −3660e | −3855 to −3432 | −3637e | −3838 to −3420 |
Costs at 12 months | ||||||||||
Health and social care, excluding trial medicationc | 95 | 659 | 1699 | 93 | 583 | 634 | −74 | −486 to 255 | −24 | −363 to 230 |
Employment lossesc | 93 | 19 | 132 | 95 | 2 | 18 | −16 | −46 to 2 | −17 | −42 to 2 |
Social security benefitsc | 93 | 85 | 83 | 95 | 77 | 84 | −6 | −32 to 16 | 5 | −12 to 23 |
Trial medicationd | 96 | 3546 | 1631 | 94 | 1547 | 1547 | −1988e | −2458 to −1555 | −1894e | −2320 to −1427 |
The increase in trial medication costs in the cDMARDs group between 6 and 12 months was due to a significant proportion of this group switching to the more expensive TNFis at 6 months because of non-response to cDMARDs by 6 months. Switching in the reverse direction was uncommon (a total of four participants) and so trial medication costs in the TNFis group did not fall a great deal between 6 and 12 months.
Table 32 shows total costs at 6 and 12 months from a health and social care perspective and the two societal perspectives that we adopted (with and without social security benefit costs). All figures (including those for trial medication) represent a 3-month period. The cDMARDs group has significantly lower total costs from all perspectives at both follow-up points. The difference is greater at 6 months than at 12 months because of the greater trial medication cost differential before switching taking place. Costs from each of the societal perspectives are similar to those from a health and social care perspective because of the dominance of trial medication costs.
Perspective | TNFis group (N = 101) | cDMARDs group (N = 104) | Unadjusted differencea | Adjusted differenceb | ||||||
---|---|---|---|---|---|---|---|---|---|---|
Valid n | Mean cost (£) | SD (£) | Valid n | Mean cost (£) | SD (£) | Mean (£) | 95% CI (£) | Mean (£) | 95% CI (£) | |
Costs at 6 months | ||||||||||
Health and social care perspective, including trial medication | 97 | 2547 | 1083 | 94 | 793 | 703 | −1757c | −2006 to −1500 | −1708c | −1973 to −1483 |
Societal perspective, including trial medication but excluding social security benefits | 97 | 2617 | 1145 | 94 | 828 | 791 | −1793c | −2050 to −1519 | −1742c | −2024 to −1506 |
Societal perspective, including trial medication and social security benefits | 97 | 2694 | 1148 | 94 | 902 | 802 | −1794c | −2055 to −1515 | −1742c | −2023 to −1506 |
Costs at 12 months | ||||||||||
Health and social care perspective, including trial medication | 93 | 2411 | 1608 | 95 | 1493 | 1089 | −907c | −1327 to −524 | −817c | −1170 to −481 |
Societal perspective, including trial medication but excluding social security benefits | 93 | 2430 | 1645 | 95 | 1494 | 1088 | −924c | −1351 to −540 | −840c | −1205 to −501 |
Societal perspective, including trial medication and social security benefits | 93 | 2515 | 1637 | 95 | 1571 | 1100 | −930c | −1363 to −541 | −841c | −1200 to −508 |
For the purpose of combining cost and outcome data for the cost-effectiveness/cost–utility analyses, all costs were equivalised to 6-month values. Trial medication costs were available for the 0- to 6-month and 7- to 12-month periods so all other costs were multiplied by 2 to represent 6-month rather than 3-month periods. The extrapolated figures are shown in Table 33. Imputing missing cost data (based on the extrapolated costs) for those lost to follow-up confirmed the findings from the unimputed available case analysis (Table 34).
Perspective | TNFis group (N = 101) | cDMARDs group (N = 104) | Unadjusted differencea | Adjusted differenceb | ||||||
---|---|---|---|---|---|---|---|---|---|---|
Valid n | Mean cost (£) | SD (£) | Valid n | Mean cost (£) | SD (£) | Mean (£) | 95% CI (£) | Mean (£) | 95% CI (£) | |
Costs at 6 months | ||||||||||
Health and social care perspective, including trial medication | 97 | 5238 | 2093 | 94 | 1538 | 1393 | −3703c | −4175 to −3199 | −3615c | −4104 to −3182 |
Societal perspective, including trial medication but excluding social security benefits | 97 | 5379 | 2236 | 94 | 1607 | 1569 | −3774c | −4298 to −3230 | −3683c | −4198 to −3195 |
Societal perspective, including trial medication and social security benefits | 97 | 5533 | 2241 | 94 | 1755 | 1591 | −3778c | −4303 to −3230 | −3684c | −4199 to −3194 |
Costs at 12 months | ||||||||||
Health and social care perspective, including trial medication | 93 | 4866 | 3147 | 95 | 2718 | 1890 | −2129c | −2941 to −1417 | −1930c | −2599 to −1301 |
Societal perspective, including trial medication but excluding social security benefits | 93 | 4904 | 3218 | 95 | 2722 | 1890 | −2162c | −2977 to −1449 | −1974c | −2648 to −1334 |
Societal perspective, including trial medication and social security benefits | 93 | 5073 | 3208 | 95 | 2876 | 1914 | −2175c | −2991 to −1465 | −1977c | −2644 to −1338 |
Perspective | TNFis group (N = 101) | cDMARDs group (N = 104) | Unadjusted differencea | Adjusted differenceb | ||||||
---|---|---|---|---|---|---|---|---|---|---|
Valid n | Mean cost (£) | SD (£) | Valid n | Mean cost (£) | SD (£) | Mean (£) | 95% CI (£) | Mean (£) | 95% CI (£) | |
Costs at 6 monthsc | ||||||||||
Health and social care perspective, including trial medication | 101 | 5234 | 2052 | 104 | 1520 | 1329 | −3717e | −4205 to −32556 | −3615e | −4067 to −3198 |
Societal perspective, including trial medication but excluding social security benefits | 101 | 5373 | 2192 | 104 | 1594 | 1496 | −3780e | −4341 to −3288 | −3688e | −4195 to −3232 |
Societal perspective, including trial medication and social security benefits | 101 | 5527 | 2197 | 104 | 1743 | 1518 | −3784e | −4348 to −3298 | −3691e | −4194 to −3246 |
Costs at 12 monthsd | ||||||||||
Health and social care perspective, including trial medication | 101 | 4874 | 3023 | 104 | 2729 | 1816 | −2137e | −2838 to −1516 | −1937e | −2612 to −1353 |
Societal perspective, including trial medication but excluding social security benefits | 101 | 4910 | 3092 | 104 | 2728 | 1818 | −2173e | −2895 to −1535 | −1971e | −2648 to −1377 |
Societal perspective, including trial medication and social security benefits | 101 | 5080 | 3082 | 104 | 2887 | 1840 | −2182e | −2885 to −1543 | −1976e | −2668 to −1368 |
Outcomes
The cDMARDs arm had an advantage of four points based on the SF-36-based utility scores at baseline but this did not carry through as an advantage in (baseline-adjusted) utility scores at either of the two follow-up points or in the resulting QALY estimates (Table 35). The cDMARDs group did, however, show advantages in terms of the HAQ and EQ-5D-based utility scores at 12 months, although the latter did not translate into an advantage in terms of the QALYs estimated from the EQ-5D. As with cost data, imputing missing outcome data for those lost to follow-up did not alter the conclusions from the available case analyses (Table 36).
Outcome | TNFis group | cDMARDs group | Unadjusted differencea | Adjusted differenceb | ||||||
---|---|---|---|---|---|---|---|---|---|---|
Valid n | Mean | SD | Valid n | Mean | SD | Mean | 95% CI | Mean | 95% CI | |
Utilities and HAQ score | ||||||||||
Baseline | ||||||||||
SF-36 utility | 101 | 0.52 | 0.11 | 104 | 0.56 | 0.10 | 0.04c | 0.01 to 0.07 | – | – |
EQ-5D utility | 101 | 0.35 | 0.31 | 104 | 0.39 | 0.31 | 0.04 | −0.04 to 0.12 | – | – |
HAQ score | 101 | 1.90 | 0.67 | 104 | 1.80 | 0.59 | −0.10 | −0.28 to 0.07 | – | – |
6 months | ||||||||||
SF-36 utility | 97 | 0.59 | 0.13 | 94 | 0.62 | 0.12 | 0.03 | −0.01 to 0.06 | 0.00 | −0.03 to 0.03 |
EQ-5D utility | 97 | 0.53 | 0.30 | 94 | 0.56 | 0.26 | 0.03 | −0.05 to 0.10 | −0.01 | −0.08 to 0.06 |
HAQ score | 97 | 1.55 | 0.83 | 94 | 1.52 | 0.65 | −0.03 | −0.22 to 0.19 | 0.07 | −0.08 to 0.21 |
12 months | ||||||||||
SF-36 utility | 94 | 0.60 | 0.14 | 94 | 0.64 | 0.13 | 0.04c | 0.01 to 0.08 | 0.03 | −0.00 to 0.07 |
EQ-5D utility | 93 | 0.50 | 0.31 | 94 | 0.60 | 0.28 | 0.10c | 0.02 to 0.19 | 0.10 | 0.02 to 0.18c |
HAQ score | 94 | 1.60 | 0.84 | 95 | 1.33 | 0.77 | −0.27c | −0.51 to −0.04 | −0.16c | −0.32 to −0.01 |
QALYs | ||||||||||
6 months | ||||||||||
SF-36 QALYs | 97 | 0.28 | 0.05 | 94 | 0.30 | 0.05 | 0.02 | 0.00 to 0.03 | 0.00 | −0.01 to 0.01 |
EQ-5D QALYs | 97 | 0.22 | 0.14 | 94 | 0.24 | 0.12 | 0.02 | −0.02 to 0.05 | 0.00 | −0.02 to 0.02 |
12 months | ||||||||||
SF-36 QALYs | 93 | 0.30 | 0.06 | 87 | 0.32 | 0.05 | 0.02 | −0.00 to 0.03 | 0.01 | −0.00 to 0.02 |
EQ-5D QALYs | 92 | 0.26 | 0.13 | 88 | 0.29 | 0.11 | 0.03 | −0.01 to 0.06 | 0.02 | −0.01 to 0.05 |
Outcome | TNFis group | cDMARDs group | Adjusted differencea | Adjusted differenceb | ||||||
---|---|---|---|---|---|---|---|---|---|---|
Valid n | Mean | SD | Valid n | Mean | SD | Mean | 95% CI | Mean | 95% CI | |
6 months | ||||||||||
HAQ scorec | 101 | 1.55 | 0.82 | 104 | 1.51 | 0.64 | −0.04 | −0.24 to 0.16 | 0.07 | −0.07 to 0.21 |
SF-36 QALYsd | 101 | 0.28 | 0.05 | 104 | 0.29 | 0.05 | 0.02 | 0.00 to 0.03 | 0.00 | −0.01 to 0.01 |
EQ-5D QALYsd | 101 | 0.22 | 0.14 | 104 | 0.24 | 0.12 | 0.02 | −0.02 to 0.05 | −0.00 | −0.02 to 0.02 |
12 months | ||||||||||
HAQ scorec | 101 | 1.59 | 0.83 | 104 | 1.35 | 0.74 | −0.25e | −0.45 to −0.03 | −0.16e | −0.30 to −0.02 |
SF-36 QALYsd | 101 | 0.30 | 0.06 | 104 | 0.32 | 0.06 | 0.02 | 0.00 to 0.03 | 0.01 | −0.00 to 0.02 |
EQ-5D QALYsd | 101 | 0.26 | 0.13 | 104 | 0.29 | 0.11 | 0.03 | −0.00 to 0.06 | 0.02 | −0.00 to 0.05 |
Cost-effectiveness and cost–utility analyses
Table 37 presents the ICERs for the cost-effectiveness and cost–utility analyses based on costs from each perspective (based on extrapolations representing 6-month periods) and outcomes at 6 and 12 months. For the ICERs, the mean difference in the HAQ score was reversed (negatives turned to positives and vice versa) as a reduction in HAQ score indicates a better outcome. Of the 18 cost–outcome combinations, three showed statistically significant between-group differences for both costs and outcomes: at 12 months, the cDMARDs group dominated with the group having better outcomes (mean difference −0.16, 95% CI −0.32 to −0.01) and lower costs from a health-care perspective (mean difference −£1930, 95% CI −£2599 to −£1301), societal perspective excluding benefits (mean difference −£1974, 95% CI −£2648 to −£1334) and societal perspective including benefits (mean difference −£1977, 95% CI −£2644 to −£1338). These translated into ICERs of −£12,063, −£12,338 and −£12,356 per QALY respectively. All other cost–outcome combinations suggest that the cDMARDs group is superior, with equivalent outcomes achieved at a significantly lower cost. The conclusions remained the same when costs and outcomes for those lost to follow-up were imputed. It was not necessary to compute any ICERs as none of the combinations suggested a significantly better outcome at a significantly lower cost.
Cost per additional point improvement on the HAQ (£), cDMARDs vs. TNFis | Cost per additional QALY (SF-36 based) (£), cDMARDs vs. TNFis | Cost per additional QALY (EQ-5D based) (£), cDMARDs vs. TNFis | |
---|---|---|---|
6 months | |||
Health and social care perspective | 51,643 | −3615 | −3615 |
Societal perspective excluding benefits | 52,614 | −3683 | −3683 |
Societal perspective including benefits | 52,629 | −3684 | −3684 |
12 months | |||
Health and social care perspective | cDMARDs dominate: −12,063 | −193,000 | −96,500 |
Societal perspective excluding benefits | cDMARDs dominate: −12,338 | −197,400 | −98,700 |
Societal perspective including benefits | cDMARDs dominate: −12,356 | −197,700 | −98,850 |
Figures 21 and 22 show the probability that the cDMARDs group is cost-effective compared with the TNFis group for each outcome from a health and social care perspective at 6 and 12 months respectively. Both EQ-5D- and SF-36-based QALYs at each time point suggest that the probability that the cDMARDs group is cost-effective is ≥ 99% at all willingness-to-pay thresholds that were examined.
The probability that the cDMARDs group is cost-effective at 6 months based on the HAQ is 100% for willingness-to-pay thresholds of up to £10,000 per point improvement on the HAQ but decreases at higher willingness-to-pay thresholds. At 12 months the probability of cost-effectiveness is 100% for willingness-to-pay thresholds of up to £10,000 per point improvement on the HAQ and remains at 99% up to a threshold of £50,000.
Systematic reviews
Early rheumatoid arthritis
Trials
The preliminary search identified 463 papers, of which 36 were potentially relevant trials and were selected for full-text review (Figure 23). Of these, four trials were excluded: one included patients with a disease duration of > 3 years, two used treatment strategies in which the same approaches were included in both arms and one used steroids with methotrexate in the control monotherapy arm. The remaining 32 trials68,111,147,171,192–219 formed the basis of this systematic review.
The baseline characteristics of the 32 RCTs are summarised in Table 38. The trials randomised between 20 and 1049 patients and enrolled over 8400 patients. In total, 19 trials compared cDMARDs with methotrexate,68,111,171,192–207 10 trials compared TNFis/methotrexate with methotrexate monotherapy208–217 and three trials compared cDMARDs with TNFis/methotrexate directly (head-to-head trials). 147,218,219 The Optimal Protocol for Methotrexate and Adalimumab Combination Therapy in Early Rheumatoid Arthritis (OPTIMA)217 and High Induction Therapy with Anti-Rheumatic Drugs (HIT-HARD)216 studies withdrew anti-TNF treatment from 24 and 26 weeks, respectively; therefore, only outcomes at 24 and 26 weeks, respectively, were considered. The BeSt220 and Swedish Farmacotherapy (Swefot)221 trials initially published 12-month results and subsequently 24-month results.
Study | Year | Design | Cases | Max RA duration | Quality assessments | Follow-up | Combinations | |||
---|---|---|---|---|---|---|---|---|---|---|
Allocation | Blinding | Analysed in original groups | Analysed for bias | |||||||
cDMARDs | ||||||||||
Boers et al.68 | 1997 | Step-down | 156 | 2 years | Unstated | Double | Yes | Yes | 1 year | SSZ/MTX/prednisolone vs. SSZ |
Haagsma et al.192 | 1997 | Parallel | 105 | 1 year | Unstated | Double | Yes | No | 1 year | MTX/SSZ vs. SSZ vs. MTX |
van den Borne et al.193 | 1998 | Step-up | 88 | 3 years | Central block randomisation | Double | Yes | No | 0.5 years | Chloroquine/CsA vs. chloroquine |
Dougados et al.194 | 1999 | Parallel | 209 | 1 year | Unstated | Double | Yes | No | 1 year | MTX/SSZ vs. SSZ vs. MTX |
Mottonen et al.195 | 1999 | Step-down | 199 | 2 years | Cards | Open | Yes | Yes | 2 years | SSZ/MTX/HCQ/prednisolone vs. SSZ |
Proudman et al.196 | 2000 | Parallel | 82 | 1 year | Centralised randomisation list | Open | Yes | Yes | 1 year | CsA/MTX/IA methylprednisolone vs. SSZ |
Ferraccioli et al.197 | 2002 | Step-up | 126 | – | Unstated | Open | Yes | No | 1.5 years | MTX/CsA vs. SSZ |
Gerards et al.198 | 2003 | Parallel | 120 | 3 years | Computer generated | Double | Yes | No | 1 year | CsA/MTX vs. CsA |
Marchesoni et al.199 | 2003 | Parallel | 61 | 2 years | Randomisation list | Open | Yes | No | 1 year | CsA/MTX vs. MTX |
Capell et al.200 | 2004 | Parallel | 167 | 3 years | Unstated | Double | Yes | No | 2 years | SSZ/prednisolone vs. SSZ |
Grigor et al.111 | 2004 | Step-up | 111 | 5 years | Computer generated | Single | Yes | No | 1.5 years | Intensive combinations vs. routine care |
Miranda et al.201 | 2004 | Parallel | 149 | 2 years | Computer generated | Double | Yes | No | 1 year | CsA/chloroquine vs. CsA |
Ichikawa et al.202 | 2005 | Parallel | 71 | 2 years | Randomised by test drug number | Double | Yes | No | 1.8 years | MTX/bucillamine vs. MTX vs. bucillamine |
Sarzi-Puttini et al.203 | 2005 | Parallel | 105 | 3 years | Unstated | Open | Yes | Yes | 1 year | CsA/HCQ vs. CsA/MTX vs. CsA |
Svensson et al.204 | 2005 | Parallel | 259 | 1 year | Computer generated | Open | Yes | No | 2 years | Prednisolone/DMARD vs. DMARD |
Wassenberg et al.205 | 2005 | Parallel | 192 | 2 years | Computer generated | Double | Yes | No | 2 years | Prednisolone/DMARD vs. DMARD |
Hetland et al.206 | 2006 | Parallel | 163 | 6 years | Computer generated | Double | Yes | Yes | 1 years | MTX/CsA/IA beclamethasone vs. MTX/IA beclamethasone |
O’Dell et al.207 | 2006 | Parallel | 66 | 1 year | Cards | Double | Yes | No | 2 years | Doxycycline/MTX vs. MTX |
Choy et al.171 | 2008 | Step-up | 467 | 2 years | Computer generated | Double | Yes | Yes | 2 years | MTX vs. MTX + CsA vs. MTX + prednisolone vs. triple |
TNFi/MTX | ||||||||||
Breedveld et al.208 | 2004 | Parallel | 82 | 3 years | Unstated | Double | Yes | No | 2 years | Infliximab/MTX |
St Clair et al.209 | 2004 | Parallel | 1049 | 3 years | Interactive voice response | Double | Yes | Yes | 1 year | Infliximab/MTX |
Taylor et al.210 | 2004 | Parallel | 24 | 3 years | Pharmacist randomisation | Double | Yes | No | 1 year | Infliximab/MTX |
Quinn et al.211 | 2005 | Parallel | 20 | 1 year | Adaptive stratified randomisation technique | Double | Yes | No | 1 year | Infliximab/MTX |
Breedveld et al.212 | 2006 | Parallel | 799 | 3 years | Unstated | Double | Yes | No | 2 years | Adalimumab/MTX |
Durez et al.213 | 2007 | Parallel | 44 | 1 year | Unstated | Open | Yes | No | 1 year | Infliximab/MTX |
Emery et al.214 | 2008 | Parallel | 542 | 2 years | Computer generated | Double | Yes | Yes | 1 year | Etanercept/MTX |
Soubrier et al.215 | 2009 | Parallel | 65 | 0.5 years | Unstated | Open | Yes | Yes | 1 year | Adalimumab/MTX |
Detert et al.216 | 2013 | Parallel | 172 | 1 year | Unstated | Double | Yes | Yes | 0.5 years | Adalimumab/MTX |
Kavanaugh et al.217 | 2013 | Parallel | 1032 | 1 year | Interactive voice response | Double | Yes | Yes | 0.5 years | Adalimumab/MTX |
Direct comparisons | ||||||||||
Goekoop-Ruiterman et al.147 | 2005 | All three | 508 | 2 years | Variable block randomisation | Open | Yes | Yes | 1 year | Step-up vs. step-down vs. infliximab |
van Vollenhoven et al.218 | 2009 | Step-up | 487 | 1 year | Computer generated | Open | Yes | No | 1 year | MTX/SSZ/HCQ vs. infliximab/MTX |
Moreland et al.219 | 2012 | Parallel | 755 | 3 years | Computer generated | Double | Yes | Yes | 2 years | MTX/SSZ/HCQ vs. etanercept/MTX |
Baseline characteristics
The baseline characteristics of the patients are summarised in Table 39. The average age ranged from 46 to 55 years in the TNFis/methotrexate RCTs, from 37 to 59 years in the cDMARDs RCTs and from 49 to 54 years in the direct comparison trials. Mean disease duration ranged from 0.5 to 3 years in the TNFis/methotrexate RCTs, from 0.5 to 3 years in the cDMARDs RCTs and from 1 to 3 years in the direct comparison trials. Not all trials reported initial DAS28; in the 14 trials in which this was recorded the mean score ranged from 4.8 to 6.7 with an overall average score of 5.58.
Study | Cases | Age (years), mean (SD) | Female (%) | Baseline DAS28, mean (SD) |
---|---|---|---|---|
Indirect comparisons | ||||
Boers et al.68 | 76 | 50 (11.9) | 66 | Not stated |
Breedveld et al.208 | 82 | 50a | 79 | Not stated |
Breedveld et al.212 | 268 | 51.9 (14) | 72 | 6.3 (0.9) |
Capell et al.200 | 84 | 55 (range 25–76) | 65 | Not stated |
Choy et al.171 | 116 | 55 | 67 | 5.6 (1.2) |
Detert et al.216 | 87 | 47 (12) | 70 | 6.2 (0.8) |
Dougados et al.194 | 68 | 52 | 77 | DAS: 4.23 |
Durez et al.213 | 15 | 50 (9.9) | 67 | DAS28-CRP: 5.3 (1.1) |
Emery et al.214 | 265 | 50.5 (0.9) | 74 | 6.5 (1.0) |
Ferraccioli et al.197 | 42 | 59 (7.7) | 86 | Not stated |
Gerards et al.198 | 60 | 53 (10.6) | 62 | Not stated |
Grigor et al.111 | 55 | 51 (15) | 71 | DAS: 4.9 (0.9) |
Haagsma et al.192 | 36 | 57 (12.2) | 66 | DAS: 5.0 (0.8) |
Hetland et al.206 | 60 | 53.2a | 64 | 5.31 (1.34) |
Ichikawa et al.202 | 24 | 49.1 (12.9) | 71 | Not stated |
Kavanaugh et al.217 | 515 | 50.7 (14.5) | 74 | DAS28-CRP: 6.0 (1.0) |
Marchesoni et al.199 | 30 | 46.6 (10.5) | 93 | 5.2 (1.2) |
Miranda et al.201 | 75 | 37 (11) | 92 | Not stated |
Mottonen et al.195 | 97 | 47 (range 23–65) | 58 | Not stated |
O’Dell et al.207 | 24 | 49.5 | 67 | Not stated |
Proudman et al.196 | 40 | 51 (13.7) | 65 | 5.4 (1) |
Quinn et al.211 | 10 | 51.3 (9.5) | Not stated | Not stated |
Sarzi-Puttini et al.203 | 30 | 53 (10) | 63 | Not stated |
Soubrier et al.215 | 65 | 46.3 (16.3) | 79 | 6.31 (0.78) |
St Clair et al.209 | 363 | 50 (13) | 68 | 6.7 (1.0) |
Svensson et al.204 | 119 | 51 (14) | 65 | 5.28 (1.11) |
Taylor et al.210 | 12 | 55 (11.8) | Not stated | 5.4 (1.1) |
van den Borne et al.193 | 30 | 51 (11.1) | 73 | Not stated |
Wassenberg et al.205 | 80 | 53 (12.6) | 75 | Not stated |
Direct comparisons | ||||
Goekoop et al. (DMARDs)147 | 133 | 55 (14) | 65 | DAS44: 4.4 (0.9) |
Goekoop et al. (TNFi/methotrexate)147 | 128 | 54 (14) | 66 | DAS44: 4.3 (0.9) |
Moreland et al. (DMARDs)219 | 132 | 48.8 (12.7) | 77 | 5.8 (1.1) |
Moreland et al. (TNFi/methotrexate)219 | 244 | 50.7 (13.4) | 74 | 5.8 (1.1) |
van Vollenhoven et al. (DMARDs)218 | 130 | 52.9 (13.9) | 78 | 4.79 (1.05) |
van Vollenhoven et al. (TNFi/methotrexate)218 | 128 | 51.1 (13.3) | 76 | 5.91 (0.93) |
American College of Rheumatology responses and withdrawals for inefficacy
Indirect comparisons showed that in trials of DMARD combinations (Table 40 and Figure 24) more patients achieved ACR20–70 responses with combination therapy (OR 1.76–2.81) and less patients withdrew because of inefficacy with combination therapy (OR 0.47, 95% CI 0.34 to 0.64). In trials of TNFi/methotrexate combinations more patients achieved ACR20–70 responses with combination therapy (OR 1.88–2.22) and fewer patients withdrew because of inefficacy with combination therapy (OR 0.44, 95% CI 0.22 to 0.85). Sensitivity analysis of trials using only methotrexate monotherapy showed similar results.
Outcome | Treatment regimen | Studies | Random-effects analyses |
---|---|---|---|
OR (95% CI) | |||
Categorical outcomes | |||
ACR20 | DMARD combinations | 14 | 1.76 (1.26 to 2.46) |
DMARD combinations (methotrexate only) | 5 | 2.01 (1.08 to 3.72) | |
TNFi/methotrexate | 8 | 1.88 (1.61 to 2.19) | |
ACR50 | DMARD combinations | 13 | 2.34 (1.40 to 3.91) |
DMARD combinations (methotrexate only) | 5 | 1.64 (1.15 to 2.34) | |
TNFi/methotrexate | 7 | 2.09 (1.80 to 2.43) | |
ACR70 | DMARD combinations | 8 | 2.81 (1.48 to 5.33) |
DMARD combinations (methotrexate only) | 4 | 2.00 (1.32 to 3.02) | |
TNFi/methotrexate | 7 | 2.22 (1.78 to 2.76) | |
Inefficacy withdrawals | DMARD combinations | 15 | 0.47 (0.34 to 0.64) |
DMARD combinations (methotrexate only) | 7 | 0.52 (0.33 to 0.82) | |
TNF/methotrexate | 9 | 0.44 (0.22 to 0.85) | |
Toxicity withdrawals | DMARD combinations | 15 | 1.86 (1.42 to 2.44) |
DMARD combinations (methotrexate only) | 7 | 2.69 (1.49 to 4.83) | |
TNFi/methotrexate | 9 | 1.42 (0.87 to 2.34) | |
WMD (95% CI) | |||
Continuous outcomes | |||
Disability (HAQ) | DMARD combinations | 7 | −0.03 (−0.12 to 0.07) |
DMARD combinations (methotrexate only) | 2 | −0.17 (−0.33 to −0.01) | |
TNFi/methotrexate | 2 | −0.16 (−0.24 to −0.08) | |
Radiological progression | DMARD combinations | 7 | −0.99% (−1.11% to −0.87%) |
DMARD combinations (methotrexate only) | 4 | −1.21% (−1.37% to −1.04%) | |
TNFi/methotrexate | 4 | −0.61% (−0.79% to −0.43%) |
Direct comparisons showed that there were no differences between DMARD combinations (Table 41) and TNFi/methotrexate with regard to ACR20 outcomes or patient withdrawals because of inefficacy. However, fewer patients achieved ACR50 and ACR70 responses using cDMARDs than using TNFi/methotrexate (ORs 0.54 and 0.53 respectively). A more detailed analysis of data from each of these trials is shown in Figure 25. Overall, there were small differences in favour of TNFi/methotrexate compared with cDMARDs at most time points but these were not always significant. There were also marked differences in response rates in the different trials.
Outcome | Studies | Random-effects analyses |
---|---|---|
OR (95% CI) | ||
Categorical outcomes | ||
ACR20 | 2 | 0.74 (0.42 to 1.29) |
ACR50 | 1 | 0.54 (0.33 to 0.90) |
ACR70 | 2 | 0.53 (0.36 to 0.79) |
Inefficacy withdrawals | 3 | 3.28 (0.51 to 21.28) |
Toxicity withdrawals | 3 | 1.63 (0.78 to 3.43) |
WMD (95% CI) | ||
Continuous outcome | ||
Radiological progression | 2 | 0.22 (−0.02 to 0.45) |
Disability
In the indirect comparisons there were greater improvements in HAQ scores with both combination regimens when compared with DMARD monotherapy (OR −0.15, 95% CI −0.23 to −0.07) or methotrexate monotherapy (OR −0.17, 95% CI −0.33 to −0.01) (see Table 40). No RCTs that made a direct comparison between cDMARDs and TNFi/methotrexate reported HAQ outcomes.
Toxicity
Indirect comparisons (see Table 40) showed that more patients withdrew with DMARD combinations because of toxicity than with DMARD monotherapy (OR 1.50, 95% CI 1.11 to 2.03) or with methotrexate monotherapy (OR 2.69, 95% CI 1.49 to 4.83). There were no differences between TNFi/methotrexate and methotrexate monotherapy in terms of withdrawals because of toxicity. The direct comparisons showed no differences in patient withdrawal because of toxicity (see Table 41).
Radiological progression
Indirect comparisons showed less erosive progression with both combination regimens compared with DMARD monotherapy (see Table 40). Sensitivity analysis of cDMARDs including only those trials in which the comparator was methotrexate monotherapy showed similar results. The direct comparison showed that there was no difference in radiological progression between cDMARDs and TNFi/methotrexate (see Table 41).
Heterogeneity
The cDMARD trials showed evidence of heterogeneity in ACR20 scores (p < 0.007), ACR50 scores (p < 0.0001) and ACR70 scores (p = 0.02). In contrast, the TNFi trials showed no heterogeneity. There was also no heterogeneity in the head-to-head trials.
Established rheumatoid arthritis
Trials
The preliminary search identified 3642 papers, of which 28 were potentially relevant and were selected for full-text review (Figure 26). Of these, nine studies were excluded: in four patients were treatment naive, in two patients had not received DMARDs for > 3 months, two did not specify previous DMARD treatment and one was a duplicate of an included study. The remaining 19 studies66,67,170,222–237 were included in the systematic review and are summarised in Table 42. In total, 10 trials compared cDMARDs with DMARD monotherapy,66,67,222–229 of which six used methotrexate monotherapy as the control arm,66,67,222,223,227,229 and eight compared TNFi/methotrexate with methotrexate monotherapy,170,230–236 with one involving infliximab,170 two etanercept,230,232 one adalimumab,231 two golimumab233,236 and two certolizumab pegol. 234,235 For the Trial of Etanercept and Methotrexate with Radiographic Patient Outcomes (TEMPO)232 and Rheumatoid Arthritis Prevention of Structural Damage 1 (RAPID1)234 trials, 2-year follow-up data were subsequently published. 238,239 Finally, one trial made a direct comparison between methotrexate/sulfasalazine/hydroxychloroquine and etanercept/methotrexate. 237
Study | Year | Cases | Refractory to treatment | Quality assessments | Follow-up | Treatment arms | |||
---|---|---|---|---|---|---|---|---|---|
Allocation | Blinding | Analysed in original groups | Analysed for bias | ||||||
cDMARDs | |||||||||
Ferraz et al.222 | 1994 | 82 | Failed more than one DMARD | Unstated | Double | Yes | No | 0.5 years | MTX vs. MTX/chloroquine |
Tugwell et al.66 | 1995 | 148 | MTX > 3 months | Unstated | Double | Yes | No | 0.5 years | MTX vs. CsA/MTX |
Willkens et al.223 | 1995 | 209 | Resistant to penicillamine/gold | Permutated blocks | Double | Yes | Yes | 1 year | MTX vs. MTX/AZA |
Bendix et al.224 | 1996 | 40 | Insufficient response to gold | Computerised | Double | Yes | No | 0.5 years | PGT vs. PGT/CsA |
O’Dell et al.67 | 1996 | 102 | More than one DMARD | Cards | Double | Yes | Yes | 2 years | MTX vs. MTX/SSZ/HCQ |
Kremer et al.225 | 2002 | 263 | MTX > 6 months | Computerised | Double | Yes | Yes | 0.5 years | Leflunamide vs. leflunamide/MTX |
Dougados et al.226 | 2005 | 106 | Inadequate response to leflunomide | Unstated | Double | Yes | No | 0.5 years | SSZ vs. leflunamide/SSZ |
Lehman et al.227 | 2005 | 65 | MTX > 12 weeks | Random number table | Double | Yes | Yes | 1 year | MTX vs. MTX/IM gold |
Karanikolas et al.228 | 2006 | 106 | Refractory to more than one DMARD | Unstated | Open | Yes | No | 1 year | Leflunamide vs. CsA/leflunamide |
Capell et al.229 | 2007 | 191 | SSZ > 6 months | Computerised | Double | Yes | No | 1 year | MTX vs. MTX/SSZ |
TNFis/MTX | |||||||||
Weinblatt et al.230 | 1999 | 89 | MTX > 6 months | Unstated | Double | Yes | No | 0.5 years | MTX vs. etanercept/MTX |
Lipsky et al.170 | 2000 | 428 | MTX > 6 months | Unstated | Double | Yes | No | 1 year | MTX vs. 3 mg infliximab/MTX |
Weinblatt et al.231 | 2003 | 271 | MTX > 6 months | Unstated | Double | Yes | No | 0.5 years | MTX vs. 40 mg adalimumab/MTX |
Klareskog et al.232 | 2004 | 686 | More than one DMARD other than MTX | Centralised telephone | Double | Yes | Yes | 0.5 years | MTX vs. etanercept/MTX |
Kay et al.233 | 2008 | 172 | MTX > 3 months | Unstated | Double | Yes | No | 0.3 years | MTX vs. 50 mg golimumab/MTX |
Keystone et al.234 | 2008 | 982 | MTX > 6 months | Unstated | Double | Yes | No | 1 year | MTX vs. 200 mg certolizumab/MTX |
Smolen et al.235 | 2009 | 619 | MTX > 6 months | Unstated | Double | Yes | Yes | 0.5 years | MTX vs. 200 mg certolizumab/MTX |
Kremer et al.236 | 2010 | 643 | MTX > 3 months | Interactive voice response | Double | Yes | Yes | 0.3 years | MTX vs. 2 mg/kg golimumab/MTX |
Direct comparison | |||||||||
O’Dell et al.237 | 2013 | 353 | MTX > 3 months | Unstated | Double | Yes | No | 1 year | MTX/SSZ/HCQ vs. etanercept/MTX |
Baseline characteristics
The baseline characteristics of the patients are summarised in Table 43. The trials enrolled between 40 and 982 patients; overall, > 5500 patients were randomised. The average age ranged from 49 to 59 years in the TNFi/methotrexate trials and from 44 to 56 years in the cDMARD trials. Mean disease duration ranged from 6.7 to 13 years in the TNFi/methotrexate trials and from 1–12.7 years in the cDMARDs trials. Disease activity, assessed using the DAS28 or its component parts, was reported in the majority of trials and showed active disease. In the six trials reporting initial DAS28, these ranged from a mean of 3.6 to a mean of 7.0, with an average of 5.75.
Study | Year | Treatment | Age (years) | RF (% positive) | Disease duration (years) | DAS28 | ESR (mm/hour) | SJC | TJC | PGAa |
---|---|---|---|---|---|---|---|---|---|---|
cDMARDs | ||||||||||
Ferraz et al.222 | 1994 | MTX vs. MTX/chloroquine | 50 | 71 | 9 | – | – | – | – | – |
Tugwell et al.66 | 1995 | MTX vs. CsA/MTX | 55 | – | 11 | – | – | 17 | 23 | 62 |
Wilkens et al.223 | 1995 | MTX vs. MTX/AZA | 54 | – | 8 | – | – | – | – | – |
Bendix et al.224 | 1996 | PGT vs. PGT/CsA | 55 | 81 | 11 | – | – | – | – | – |
O’Dell et al.67 | 1996 | MTX vs. MTX/SSZ/HCQ | 50 | 84 | 10 | – | 36 | 27 | 29 | 60 |
Kremer et al.225 | 2002 | MTX vs. leflunamide/MTX | 56 | 79 | 11 | – | – | – | – | – |
Dougados et al.226 | 2005 | SSZ vs. leflunamide/SSZ | 56 | 78 | 6 | 6.2 | – | – | – | – |
Lehman et al.227 | 2005 | MTX vs. MTX/IM gold | 51 | 67 | 3 | – | 29 | 11 | 21 | 42 |
Karanikolas et al.228 | 2006 | Leflunamide vs. CsA/leflunamide | – | – | 7 | – | – | – | – | – |
Capell et al.229 | 2007 | MTX vs. MTX/SSZ | 56 | 68 | 1 | 3.6 | – | – | – | – |
TNFis/methotrexate | ||||||||||
Weinblatt et al.230 | 1999 | MTX vs. etanercept/MTX | 48 | 84 | 13 | – | – | – | – | – |
Lipsky et al.170 | 2000 | MTX vs. 3 mg infliximab/MTX | 54 | 84 | 10 | – | 49 | 22 | 32 | 70 |
Weinblatt et al.231 | 2003 | MTX vs. 40 mg adalimumab/MTX | 57 | 369b | 12 | – | – | 17 | 28 | 55 |
Klareskog et al.232 | 2004 | MTX vs. etanercept/MTX | 53 | 76 | 7 | 5.5 | – | 22 | 34 | – |
Kay et al.233 | 2008 | MTX vs. 50 mg golimumab/MTX | 57 | – | 6 | 6.4 | – | 14 | 28 | 70 |
Keystone et al.234 | 2008 | MTX vs. 200 mg certolizumab/MTX | 52 | 84 | 6 | 7.0 | 44 | 22 | 31 | – |
Smolen et al.235 | 2009 | MTX vs. 200 mg certolizumab/MTX | 52 | 76 | 6 | – | 29 | 21 | 30 | 61 |
Kremer et al.236 | 2010 | MTX vs. 2 mg/kg golimumab/MTX | 50 | – | 8 | – | – | 16 | 27 | 60 |
Direct comparison | ||||||||||
O’Dell et al.237 | 2013 | MTX/SSZ/HCQ | 58 | 66 | 6 | 5.8 | 27 | 11 | 13 | 54 |
O’Dell et al.237 | 2013 | Etanercept/MTX | 56 | 70 | 4.9 | 5.9 | 30 | 11 | 13 | 56 |
American College of Rheumatology responses and withdrawals for inefficacy
In trials of DMARD combinations more patients achieved ACR20–70 responses with combination therapy (OR 2.75–5.07), as shown in Table 44 and Figure 27. More patients withdrew with combination therapy (OR 1.51, 95% CI 1.02 to 2.25). Sensitivity analysis of RCTs that included a methotrexate monotherapy arm showed that more patients achieved ACR20–70 responses with combination therapy (OR 3.55–4.74) but few patients withdrew because of inefficacy (OR 0.34, 95% CI 0.20 to 0.59).
Outcome | Treatment | Studies | Random-effects analyses |
---|---|---|---|
OR (95% CI) | |||
Categorical outcomes | |||
ACR20 | DMARDs (all) | 6 | 2.75 (1.79 to 4.22) |
DMARDs (methotrexate only) | 4 | 3.55 (2.43 to 5.17) | |
TNFi/methotrexate | 8 | 5.32 (3.03 to 9.34) | |
ACR50 | DMARDs (all) | 6 | 5.07 (3.10 to 8.29) |
DMARDs (methotrexate only) | 4 | 4.70 (2.40 to 9.20) | |
TNFi/methotrexate | 8 | 8.13 (4.26 to 15.52) | |
ACR70 | DMARDs (all) | 5 | 4.85 (2.34 to 10.05) |
DMARDs (methotrexate only) | 3 | 4.74 (1.65 to 13.61) | |
TNFi/methotrexate | 8 | 5.36 (2.92 to 9.83) | |
Inefficacy withdrawals | DMARDs (all) | 10 | 0.38 (0.24 to 0.62) |
DMARDs (methotrexate only) | 7 | 0.34 (0.20 to 0.59) | |
TNFi/methotrexate | 8 | 0.12 (0.06 to 0.25) | |
Toxicity withdrawals | DMARDs (all) | 10 | 1.51 (1.02 to 2.25) |
DMARDs (methotrexate only) | 7 | 1.58 (0.97 to 2.59) | |
TNFi/methotrexate | 8 | 0.94 (0.62 to 1.41) | |
WMD (95% CI) | |||
Continuous outcome | |||
Disability (HAQ) | DMARDs (all) | 3 | −0.19 (−0.27 to −0.10) |
DMARDs (methotrexate only) | 1 | −0.30 (−0.42 to −0.18) | |
TNFi/methotrexate | 1 | −0.35 (−0.56 to −0.14) |
In trials of TNFi/methotrexate combinations more patients achieved ACR20–70 responses with combination therapy (OR 5.32–8.13), as shown in Table 44. Fewer patients withdrew because of inefficacy with combination therapy (OR 0.12, 95% CI 0.06 to 0.25).
The trial comparing triple DMARD therapy with etanercept/MTX237 showed no statistical difference between groups in ACR20 (57% vs. 66%), ACR50 (35% vs. 43%) and ACR70 (18% vs. 26%). This study did not report patient withdrawals for inefficacy.
Disability
Five randomised trials of cDMARDs reported change in HAQ scores (Table 45). 224–227,229 Only three of these trials reported both mean changes and SDs for these changes. 224–226 A combined analysis of these three trials’ HAQ scores (see Table 45) showed that, overall, there were greater improvements with cDMARDs than with DMARD monotherapy (WMD −0.19, 95% CI −0.27 to −0.10). Only one of these RCTs used methotrexate as the monotherapy (see Table 45);225 this trial also showed greater improvement with cDMARDs (WMD −0.30, 95% CI −0.42 to −0.18).
Study | Year | Treatment | Patients | HAQ change reported as | HAQ change scores |
---|---|---|---|---|---|
cDMARDs | |||||
Ferraz et al.222 | 1994 | MTX vs. MTX/chloroquine | 34 vs. 34 | – | |
Tugwell et al.66 | 1995 | MTX vs. CsA/MTX | 73 vs. 75 | – | |
Willkens et al.223 | 1995 | MTX vs. MTX/AZA | 67 vs. 29 | – | |
Bendix et al.224 | 1996 | PGT/placebo vs. PGT/CsA | 21 vs. 19 | Mean change (SD) | 0 (0.4) vs. 0.1 (0.4) |
O’Dell et al.67 | 1996 | MTX vs. MTX/SSZ/HCQ | 36 vs. 31 | – | |
Kremer et al.225 | 2002 | MTX vs. leflunomide/MTX | 133 vs. 130 | Mean change (SD) | 0.10 (0.43) vs. 0.40 (0.53) |
Dougados et al.226 | 2005 | SSZ vs. leflunomide/SSZ | 50 vs. 56 | Mean change (SD) | 0.02 (0.36) vs. 0.09 (0.32) |
Lehman et al.227 | 2005 | MTX vs. MTX/IM gold | 27 vs. 38 | Mean % change (SE) | 25 (7) vs. 38 (6) |
Karanikolas et al.228 | 2006 | CsA/leflunomide vs. CsA/leflunomide | 36 vs. 35 | – | |
Capell et al.229 | 2007 | MTX vs MTX/SSZ | 54 vs. 56 | Median change (IQR) | 0.2 (−0.13 to 10.3) vs. 0.5 (−0.10 to −10.3) |
TNFis/methotrexate | |||||
Weinblatt et al.230 | 1999 | MTX vs. etanercept/MTX | 30 vs. 59 | – | |
Lipsky et al.170 | 2000 | MTX vs. infliximab/MTX | 88 vs. 86 | – | |
Weinblatt et al.231 | 2003 | MTX vs. 40 mg adalimumab/MTX | 62 vs. 67 | Mean change (SD) | 0.27 (0.57) vs. 0.62 (0.63) |
Klareskog et al.232 | 2004 | MTX vs. etanercept/MTX | 228 vs. 231 | Mean change (95% CI) | 1.7 (1.6 to 1.8) vs. 1.8 (1.7 to 1.8) to 1.1 (1.0 to 1.1) vs. 0.8 (0.7 to 0.9) |
Kay et al.233 | 2008 | MTX vs. 50 mg golimumab/MTX | 35 vs. 35 | – | |
Keystone et al.234 | 2008 | MTX vs. 200 mg certolizumab/MTX | 199 vs. 393 | Median improvement (IQR) | 0.25 (0.00 to 0.75) vs. 0.63 (0.25 to 0.88) |
Smolen et al.235 | 2009 | MTX vs. 200 mg certolizumab/MTX | 127 vs. 246 | Mean change (SE) | 0.14 (0.04) vs. −0.5 (0.03) |
Kremer et al.236 | 2010 | MTX vs. 2 mg/kg golimumab/MTX | Mean % change | 9.7 vs. 32.6 |
For TNFi/methotrexate combinations five trials reported change in HAQ scores (see Table 45). 231,232,234–236 In all of these trials there was an improvement in HAQ score in the combination arm. One trial reported mean (SD) change in HAQ score (WMD −0.35, 95% CI −0.56 to −0.14). 231
The trial that made a direct comparison between methotrexate/sulfasalazine/hydroxychloroquine and etanercept/methotrexate reported mean HAQ scores at 48 weeks. 237 There was no difference in HAQ scores between triple DMARD therapy (0.93 ± 0.85) and etanercept/methotrexate (0.83 ± 0.81).
Toxicity
For cDMARDs, all 10 trials66,67,222–229 reported patient withdrawals because of toxicity. The overall OR for withdrawal with combination therapy was 1.51 (95% CI 1.02 to 2.25). Seven66,67,222,223,225,227,229 of these studies used methotrexate as the monotherapy arm; the OR for withdrawal was 1.58 (95% CI 0.97 to 2.59).
For TNFi/methotrexate combinations, eight trials170,230–236 reported patient withdrawals because of toxicity. There were no significant differences between treatments, with an OR of 0.94 (95% CI 0.62 to 1.41).
The direct comparison trial237 did not report patient withdrawals because of toxicity.
Heterogeneity
The cDMARD trials showed no evidence of heterogeneity in ACR20–70 scores. In contrast, the TNFi trials showed significant heterogeneity in ACR20 scores (p < 0.00001) and ACR50 scores (p < 0.0002) and borderline heterogeneity in ACR70 scores (p = 0.06).
Chapter 4 Discussion
The Tumour necrosis factor inhibitors Against Combination Intensive Therapy trial
Key findings
Patients with active established RA who meet current NICE criteria to receive TNFis achieve equivalent reductions in disability and improvements in quality of life over 12 months by treating initially with cDMARDs and reserving TNFis for cDMARD non-responders. The cDMARD strategy costs substantially less. However, neither treatment strategy was ideal. The majority of patients in both groups failed to achieve a DAS28 of ≤ 2.6, which is often considered to indicate remission.
The DAS28 improved more rapidly in patients receiving TNFis. Overall, monthly DAS28 were lower in patients receiving TNFis and more patients receiving TNFis achieved a DAS28 response (decrease in score of ≥ 1.2) within the first 6 months. This benefit of TNFis with regard to the DAS28 response particularly reflected rapid and sustained reductions in ESR in this group. However, the benefits of TNFis with regard to the DAS28 response were small and did not result in improvements in disability or quality of life. There was also no evidence that patients who received cDMARDs had more erosive progression. Larsen scores showed that both groups had comparable, minimal radiological progression.
Serious adverse events and withdrawals because of toxicity were equally common with cDMARDs and TNFis. However, the total number of adverse events, spanning both serious and more minor events, was higher with cDMARDs. This was most marked for adverse reactions involving the digestive system.
As TNFis are more expensive than cDMARDs, the economic evaluation showed that the cDMARD group was substantially more cost-effective, whatever approach was taken to assessing costs and relevant outcomes. This included incorporating societal costs such as lost time from work and social security benefit claims into the calculations.
In total, 44% of the patients in the cDMARD group were recommended to switch to a TNFi because their disease activity had not improved after 6 months of treatment. However, there was no evidence that patients who switched in this way had a worse quality of life, more disability or more erosive progression. There was therefore no evidence that these ‘switchers’ had any long-term disadvantages from taking cDMARDs for 6 months.
Limitations and sources of bias
Not all patients invited to participate agreed to do so; overall, 192 out of 432 patients (44%) declined to take part in the trial. We cannot be certain that the patients who did not consent to the trial would have responded in the same way as those who took part. 240 However, this is only one of a number of causes of bias in trials of long-term diseases241 and does not seem a crucial factor compared with the range of issues influencing such trials. In addition, patient choice is of crucial importance and accepting that not everyone will agree to participate is an inevitable consequence of informed choice around clinical trials. In addition, as considered below, TACIT patients receiving TNFis were similar to those in the UK national register (see pp. 90).
Those patients who did not respond to cDMARDs in the TACIT trial were treated with TNFis. It could therefore be argued that over time the two arms of the trial become very similar if not identical. However, only a minority of patients were involved in switching, with 44% of patients who started cDMARDs switching to TNFis in the second 6 months. As a consequence, the two trial arms remained sufficiently different to make this a genuine comparison. Furthermore, the 6-month comparisons did not include any patients randomised to cDMARDs who had received TNFis (because no-one switched until after 6 months). Therefore, at 6 months there was a genuine head-to-head comparison within the TACIT trial. Switching between treatment strategies is normal clinical practice and over time many RA patients starting DMARDs or biologics will switch to other treatments.
The cDMARD treatment was not standardised and it could be argued that the therapy given was too heterogeneous, making it an intervention that could be difficult to reproduce. This is an intellectual challenge as the only way to standardise cDMARD treatment is to study early RA patients who are DMARD naive or study methotrexate non-responders. These patients do not meet existing NICE criteria for receiving TNFis. If anything, the cDMARD treatments used were too conservative. We had hoped that patients would receive more intensive treatment and more short-term steroids in the cDMARD arm. However, supervising clinicians and patients placed more emphasis on slowly changing treatment to limit toxicity rather that giving maximal-dose therapy as soon as possible. Over time we anticipate that the use of cDMARDs will increase and concerns about toxicity may consequently lessen. We also accept that some combinations may be more effective, although this could not be resolved in the TACIT trial. More trials of different cDMARDs would be needed to answer this question.
Steroid use in the cDMARD group, including intramuscular injections, was less than anticipated when designing the TACIT trial based on our previous experience with steroids in established RA. 242 UK rheumatologists may have concerns about treating many patients with steroids because of the risk of adverse events. However, the relatively limited use of steroids would serve to reduce rather than magnify the impact of cDMARDs. More intensive steroid use could make cDMARD treatment even more effective.
It could be argued that the same results could have been obtained from starting another DMARD monotherapy and that the use of intensive DMARD combinations in the TACIT trial was not needed. This is a theoretical rather than a practical issue as there is no reason to stop one DMARD and start another in active RA in the absence of adverse events. DMARDs often have long half-lives, particularly agents such as leflunomide. Consequently, washing out current DMARDs and then starting a new DMARD monotherapy has limitations for patients as well as being of limited interest as the toxicity of modern DMARDs used in combination is not excessive.
The use of DMARDs other than methotrexate in combination with TNFis could have reduced the efficacy of these treatments in some patients. However, to do otherwise would be to move away from current UK practice, in which a range of DMARDs are given with TNFis. There is some evidence supporting the use of these different DMARDs in combination, as shown in our systematic reviews.
The use of the HAQ as the primary outcome measure might be viewed by some experts as being inappropriate, as the opportunity to reverse HAQ scores decreases with increasing disease duration. 243,244 Although this is theoretically correct, both of our groups showed clinically relevant reductions in HAQ scores over 12 months. In addition, the disease duration of patients in the TACIT trial (median duration of < 6 years in both groups) was below that in the Phase III trials that have led to the approval of the different TNFis. Furthermore, the degree of reduction in HAQ scores in the TACIT trial was similar to that reported in previous trials of biologics. In our view, if TNFis do not substantially reduce HAQ scores compared with other treatments then their potential clinical value is limited.
The TACIT trial was not a blinded trial. It could be argued that being unblinded influenced patients and clinicians to favour cDMARDs inappropriately. However, it was impractical to have a fully blinded treatment strategy involving multiple drugs, all of which need careful monitoring. Given the enthusiasm of most clinicians and most patients for receiving high-cost biological treatment, we consider that unblinding would, if anything, benefit TNFis. The issue of blinding is related to a number of ethical matters; these are considered in detail in the subsequent section on ethical issues.
The TACIT trial and similar trials of efficacy are not able to completely assess the relative impacts of different types of adverse event on clinical outcomes. Many adverse events reported in patients taking cDMARDs in the TACIT trial, particularly gastroenterological events, may have been relatively minor. Many patients might have felt that the treatment was ‘worth it’, irrespective of these adverse events. Although we collected detailed information about such adverse events, we did not assess their specific impacts on patient outcomes and whether they affected patients’ quality of life. We are therefore not able to fully determine the clinical consequences of such adverse events. Large long-term observational studies are needed to fully assess the impact of adverse events on treatment outcomes. Despite this limitation, the data that we collected in the TACIT trial on serious adverse events and adverse events linked to stopping treatment provided no evidence that there were more such events in the cDMARDs group than in the TNFis group.
The trial involved dividing patients into two groups after 6 months based on change in DAS28: responders and non-responders. This approach resulted in 46 out of 104 (44%) patients switching to TNFis after starting with intensive DMARDs. However, more than half (56%) of patients did not switch. This simple concept hides a more complex problem. Patients can fall on one side and then the other of such an arbitrary response without there being a major change in their condition, and the duration that patients remain within a defined state is not captured using such as approach, a problem discussed by Farewell and Su. 245 This general issue applies whenever arbitrary cut-offs are used at single time points in longitudinally collected data. It shows the difficulty of comparing the extent to which patients benefited from treatment.
Analytical issues
The TACIT trial was analysed on an ITT basis using multiple imputations and the primary outcome was compared using logistic regression methods. The HAQ is a complex assessment and it does not invariably behave as a conventional numerical scale. 246 There are identical issues with regard to the linearity of the scales of other key outcome measures, including the EQ-5D and Larsen scores. As both trial arms gave very similar outcomes using the HAQ, EQ-5D and Larsen scores, there is little merit in such an argument. In addition, the overwhelming balance of advice that we received favoured the analytical method that we preselected.
Not all of the outcomes confirmed equivalence. Changes in DAS28 and ESR favoured TNFis, particularly within the first 3–6 months. In part, this reflects the rapid onset of response with TNFis and the slow onset of response with DMARDs, which historically, and probably more accurately, used to be known as slow-acting drugs. Most DMARDs show maximal effects only by 6 months.
Measurement issues
There are several relevant measurement issues when measuring HAQ scores, radiological progression and DAS28. The HAQ was the primary outcome measure and its validity as an assessment instrument is therefore of most concern. The validity, reliability and responsiveness of the HAQ were reviewed by Linde et al. 158 Overall, the HAQ has appropriate measurement properties for a patient-generated outcome measure. It is not necessarily a simple scale and Rasch analysis by Wolfe247 and Tennant et al. 246 have highlighted some relative weaknesses. Our own previous research contributions suggest that the HAQ is the best available measure to use in trials such as the TACIT trial. 145,248,249
There is debate about the minimum clinically important difference in HAQ score in routine practice as opposed to clinical trials. Pope et al. 250 suggest that this is smaller than the difference that is considered important in trials. From this perspective the change in HAQ score in patients randomised to receive cDMARDs, which was overall 0.15 greater than that seen in patients randomised to receive TNFis, might be clinically relevant. However, this perspective appears questionable. We predetermined the minimum clinically important difference in HAQ score in trial settings and believe that it is inappropriate to change it retrospectively. It is also a theoretical rather than a practical issue as our aim was to show that the TNFi strategy was not better than the cDMARD strategy; showing that the cDMARD strategy has benefits is an identical conclusion in terms of its influence in clinical practice.
The use of Larsen scores to assess erosive damage merits consideration. The radiographs were all scored using the modified Larsen method by one investigator (DS), who has contributed to a number of published trials using this method and has achieved appropriate reproducibility of scoring. 171,242,251–260 There is debate about the relative merits of different approaches to scoring radiographs. The TACIT trial followed the approach taken in the British Rheumatoid Outcome Study Group (BROSG) trial in established RA261 and we have no reason to doubt its validity or appropriateness.
The TACIT trial used the DAS28, which was scored by many clinicians in different clinics. Each centre received detailed information during initiation on the methods of scoring the DAS28. 262 However, we did not give either explicit standardisation training nor did we retrain observers periodically to assess whether or not they maintained consistent standards. There are challenges in assessing patients using the DAS28. 263 Although training increases clinicians’ short-term agreement when measuring joint counts,264 the overall benefit of such training is uncertain. 265 Training is useful within the national context to deliver high-quality care, but its value in an individual clinical trial is limited. As the TACIT trial is a strategy trial within routine care settings we consider that it needs to replicate standard methods and should not adopt more stringent approaches because these would limit its generalisability.
Strengths of the Tumour necrosis factor inhibitors Against Combination Intensive Therapy trial
The TACIT trial was undertaken in outpatient rheumatology clinics in England in conditions that, as far as is possible within a clinical trial, mirrored routine practice. The patients enrolled were typical of those treated within England and included patients from a range of ethnicities and levels of deprivation. They are similar to those reported in the British Society for Rheumatology (BSR) Biologics Register. 266 A comparison between patients enrolled in the BSR Biologics Register and those enrolled in the TACIT trial is shown in Table 46. As there is evidence that the patients enrolled in the BSR Biologics Register have changed over time, data from all available years are shown. For the HAQ and DAS28, patients in the TACIT trial had similar initial scores and similar changes in scores (in the TNFi group) to those of patients most recently enrolled in the BSR Biologics Register.
Outcome | Time | BSR Biologics Register (by year)235 | TACIT TNFi cases | ||||||||
---|---|---|---|---|---|---|---|---|---|---|---|
2001 | 2002 | 2004 | 2004 | 2005 | 2006 | 2007 | 2008 | ITT | Completers | ||
n | 119 | 1206 | 2930 | 3138 | 1553 | 1056 | 782 | 432 | 101 | 75 | |
Mean HAQ score | Baseline | 2.21 | 2.14 | 2.10 | 2.04 | 1.98 | 1.95 | 1.87 | 1.87 | 1.90 | 1.84 |
6-month change | 0.26 | 0.33 | 0.32 | 0.32 | 0.34 | 0.33 | 0.33 | 0.32 | 0.35 | 0.41 | |
12-month change | 0.31 | 0.33 | 0.33 | 0.33 | 0.34 | 0.35 | 0.34 | 0.37 | 0.30 | 0.38 | |
Mean DAS | Baseline | 6.77 | 6.75 | 6.67 | 6.56 | 6.51 | 6.41 | 6.34 | 6.38 | 6.30 | 6.28 |
6-month change | 2.08 | 2.20 | 2.17 | 2.33 | 2.33 | 2.29 | 2.26 | 2.31 | 2.07 | 2.35 | |
12-month change | 2.03 | 2.33 | 2.35 | 2.41 | 2.46 | 2.38 | 2.46 | 2.32 | 2.41 | 2.84 |
The TACIT trial focused on patient-centred outcomes. We consider this vital because such patient-centred outcomes have a central place in clinical trials in RA. Changes in measures such as the ESR, although of interest to clinicians, are of limited value to patients. There are also concerns about the interobserver reproducibility of assessing joint counts.
The TACIT trial was of sufficient size to provide robust assessments of the changes in measures. In addition, it showed benefits favouring cDMARD treatment. In other words, cDMARDs give somewhat better outcomes than just achieving equivalence. Although we do not think that the trial shows that cDMARDs are preferable, the chance of the conclusions being incorrect and of TNFis being better appears remote.
The TACIT trial showed that only a minority of patients randomised to TNFis achieved DAS28 of ≤ 2.6 and the use of cDMARDs also resulted in relatively few DAS28 of ≤ 2.6. These low scores are often considered to reflect remission although, as discussed earlier, defining remission is an ongoing challenge. The frequency of such ‘remission scores’ in the TACIT trial is similar to that reported by both the BSR Biologics Register266 and other international registers of patients receiving TNFis in routine clinical practice267–271 (Table 47). In addition to achieving few single low DAS28 of ≤ 2.6, few TACIT trial patients achieve sustained remission. There is a need for more research on the nature and predictors of sustained low DAS28 and other indicators of remission, but this problem lies outside the remit of the TACIT trial.
Registry | TACIT | |||||||
---|---|---|---|---|---|---|---|---|
BSR BR266 | CORONA267 | DANBIO268 | DREAM269 | GISEA270 | RABBIT271 | Overall | ||
Patients, n | 11,216 | 326 | 1839 | 1531 | 591 | 775 | 16,278 | 101 |
6-month DAS28 remission rate (%)b | 14 | 25 | 21 | 27 | 26 | – | 17a | 15 |
12-month DAS28 remission rate (%)b | 16 | 29 | – | – | – | 16 | 17a | 23 |
Economic evaluation
Key findings
The economic evaluation indicates that initiating treatment with cDMARDs produces similar HAQ and QALY outcomes at 6 months as initiating treatment with TNFis, at a significantly lower cost (from all cost perspectives). By 12 months, the cDMARD approach additionally brings advantages as measured using the HAQ (−0.16, 95% CI −0.32 to −0.01), although a difference of this size is not considered to be clinically significant and so this approach can thus be regarded as being clinically similar to the TNFi approach. The cost advantage in the cDMARDs group is almost entirely due to cDMARD medications being cheaper than TNFis.
In the cDMARDs group, costs at 12 months are significantly larger than at 6 months because of the high proportion of the group who switched from cDMARDs to TNFis at 6 months. Given that there is no outcome disadvantage in the cDMARD arm at 6 or 12 months, there may be some merit in a strategy of initiating treatment with cDMARDs as this incurs lower costs for those who remain on that treatment and delays the additional costs associated with TNFis for those who go on to switch treatment.
These findings are likely to be robust because of the breadth of the cost perspectives taken and the individual-level nature of the data, which represents the variation in the sample. A pragmatic trial design performed within NHS settings also makes the findings applicable to the NHS.
Limitations
The economic evaluation has one notable limitation. Taking a broader cost perspective and a multicentre approach necessitated collating data by self-report questionnaires, which carries the risk of recall bias. Although we collected the CSRI data at 6 and 12 months, we limited the recall period to the previous 3 months to guard against recall inaccuracies. The disadvantage of this approach was that it necessitated extrapolating cost data to represent the full 6-month periods. This approach may not accurately reflect any variations that may exist across the measured and non-measured periods. However, we did have data for trial medication use over the entire period of follow-up and any biases associated with recall of other resource use would not be expected to impact on the findings given the dominance of trial medication costs. We also have no reason to believe that any such recall bias would differ by randomisation group. We accept that there was a theoretical possibility that patients switching from cDMARDs to TNFis might have resulted in the introduction of some bias, although many patients did not switch treatment until month 9. It could also be argued that we should also have reviewed patients’ medical records to ensure that no major health-care costs were missed. However, this would be unachievable over multiple sites that were dispersed throughout England and which collect clinical data in a variety of different ways. In any case, there is no evidence to suggest that the groups completed the CSRI in different ways. In addition, the differences in costs between groups were almost entirely accounted for by the treatment costs and all trial medication use was directly recorded each month for all patients in the trial.
Economic modelling
Our economic analysis used data from within the trial only. It could be argued that long-term modelling is needed to make a more convincing case for different treatment strategies including both the more extensive use of cDMARDs and the role of TNFis in non-responders to conventional DMARDs with active RA.
Marra et al. 126 have outlined the reasons for carrying out long-term modelling studies to justify the use of high-cost treatments including TNFis and other biologics in RA. Barton et al. 272 provide similar justifications. The key points are as follows:
-
Randomised controlled trials of biological treatments for RA are too short to capture relevant long-term costs and outcomes. Decision models that extrapolate the evidence from RCTs to longer-term outcomes are needed to meet the requirements of policy-makers.
-
Such models can link intermediate end points such as HAQ score with final health outcomes such as death, morbidity and employability.
-
Because only a minority of RCTs collect relevant data about both costs and health-related quality of life, it is important to use economic models to relate clinical benefits to economic outcomes.
-
The constraints inherent in all RCTs limit the generalisability of their findings for routine clinical care. Modelling can help translate their findings into different clinical settings.
Without the use of long-term modelling it would be challenging to justify the use of high-cost biologics such as TNFis in RA. However, this approach has been accepted and the balance of opinion, summarised by Bathon and McMahon,273 is that TNFis are now the preferred next step when methotrexate and DMARD monotherapy have proved insufficient.
The economic case for using conventional DMARDs more intensively, including assessments of potential long-term benefits, is somewhat different. It has been reviewed by Fautrel,274 who stressed the following key points:
-
As RA medical costs increase with rising disability levels, delaying sustained disability by better disease control will have economic benefits. 275
-
These benefits will be largest when outcomes can be improved by optimising low-cost treatments, including conventional DMARDs and glucocorticoids.
-
Early combination treatment with cDMARDs and glucocorticoids gives better efficacy at lower costs and its benefits extend over time. 276,277
-
Increasing the intensity of low-cost treatment, for example by adding modified-release prednisone to synthetic DMARDs, reduces the proportion of patients eligible for biologics, resulting in substantial cost savings. These economic benefits increase when the effects of modified-release prednisone persist. 278
Other groups have provided evidence that cDMARDs are cost-effective in RA. 279,280 The evidence is strongest in early RA with there being less information about established disease.
Showing that the use of cDMARDs before the use of biologics such as TNFis is cost-effective in the long term requires different sorts of modelling studies from those required to establish the cost-effectiveness of TNFis. It requires information about the persisting effects of cDMARDs beyond 12 months, including evidence that their clinical benefits are sustained and more information about their potential long-term risks. None of this crucial information is currently available. The duration of both RCTs and observational studies is usually too short to provide definitive assessments of medium- to long-term RA outcomes. Despite the potential importance of long-term modelling, it falls outside the scope of our research goals in the TACIT trial. As cDMARDs have been used for many years without major toxicity concerns and as the TACIT trial shows that their use is cost-effective in the short term, we are unconvinced that there is a need to measure their overall long-term cost-effectiveness compared with biologics. Although we cannot estimate the overall extent of any savings, all of the evidence suggests that their use along the lines adopted in the TACIT trial is effective and cost-effective and has no identifiable risks.
Finally, we analysed patients from the economic perspective within their original groups. Although some patients in the cDMARD group switched to TNFis after 6 months, we have not analysed these separately from the economic perspective, although we have provided a separate analysis for clinical outcomes. From the clinical perspective we wished to make certain that there was no disadvantage for patients from either remaining on cDMARDs or switching to TNFis. Our analysis provided no evidence that this was the case in terms of disability, quality of life, disease activity or erosive damage. From the economic analysis it is unclear that comparisons of patients who remained on cDMARDs with those who switched to TNFis would provide relevant information. If all patients switched to TNFis this would clearly have meant that the cDMARDs strategy was not a viable strategy. However, enough patients remained on cDMARDs to ensure that there was still an economic benefit from the cDMARD strategy in the second 6 months of treatment. Analysing data for patients who remained on cDMARDs separately from data for those who switched to TNFis in the second 6-month period would show that remaining on cDMARDs cost less; however, this inevitable finding might overemphasise the benefits of cDMARDs.
Clinical implications
This economic evaluation suggests that, at 6 months and 12 months following randomisation, cDMARDs are a more cost-effective treatment approach for RA as the cDMARDs group achieved similar outcomes as the TNFi group at a significantly lower cost.
Systematic reviews
Key findings
Compared with DMARD monotherapy, including methotrexate monotherapy, these systematic reviews show that combination treatment regimens involving either cDMARDs or TNFis with methotrexate were both superior in terms of ACR responses, reduced withdrawals because of lack of effect, reduced disability scores and reduced erosive progression. The findings are similar in early and established RA. The three head-to-head trials in early RA gave similar findings.
In our systematic review, HAQ scores showed a WMD in favour of cDMARDs, including methotrexate, of −0.30 and a WMD in favour of TNFis combined with methotrexate of −0.35. Interestingly, a systematic review of clinical trials of leflunomide in active RA,281 one of the most intensively studied DMARDs, showed a WMD in favour of leflunomide at 6 months of −0.43 (95% CI −0.52 to −0.33) compared with placebo in three trials involving 387 patients receiving leflunomide and 292 control patients. 281 These results with leflunomide suggest that starting a new DMARD is likely to have the same impact on HAQ scores as starting a new biological treatment.
Compared with the substantial benefits of combination treatments, there appeared to be relatively little to choose between cDMARDs and TNFis combined with methotrexate. There are some potential benefits of TNFi/methotrexate combinations in that there appear to be fewer withdrawals for toxicity and there was some evidence that ACR responses occurred more rapidly with biological treatments. Not all evaluations have drawn a similar conclusion; some reviews conclude that biologics have more advantages than cDMARDs and others are less optimistic about combination treatment approaches in general. Of interest are two observational studies of the impact of DMARDs compared with TNFis. Analysis of data on DMARDs from a national register collected in Germany271 and from an observational study in the UK282 resulted in very different conclusions. The German register considered that DMARDs were relatively ineffective whereas the UK study found that they were often highly effective in patients who met the criteria for receiving TNFis. Non-randomised studies can often result in very different conclusions.
The main implication from our systematic review was the need for head-to-head studies that directly compare the clinical effectiveness and cost-effectiveness of cDMARDs with those of biologics in established RA. The TACIT trial fitted exactly with this requirement. A broader outstanding challenge is to identify how best to integrate the use of DMARD combinations with biologics so that the maximal number of patients achieve the best possible response in an effective and cost-effective manner. These issues extend beyond the TACIT trial and indicate the need for an ongoing research programme in the field.
Limitations
Our systematic reviews have several limitations. First, only three RCTs directly compared cDMARDs with TNFi/methotrexate combinations and all of these were in early RA. Although we have relied more on indirect comparisons, these are invariably less informative than direct comparisons. Second, there was diversity in the range of cDMARDs used and some are not commonly used in clinical practice, for example bucillamine and doxycycline. However, we felt that these agents should be included in the meta-analysis to avoid bias and because we are examining the concept of DMARD combinations rather than the effects of specific combinations. Third, there were differences in the designs of the trials. Some trials, such as BeSt, used a tight-control regimen whereas others, such as the CARDERA trial, used a step-down design.
There are also technical limitations in assessing RA outcomes in trials. First, not all studies report the same measures in the same way. Such variability in reporting affected HAQ scores and radiological assessments. Second, not all studies had the same duration of treatment; early findings may mask longer-term limitations and therefore some comparisons may be flawed. Third, erosive damage was conventionally assessed using plain radiographs, and newer imaging methods, particularly ultrasound and MRI, may show benefits of one form of combination therapy that are not seen using conventional radiography. Fourth, reporting of adverse events is variable and assessing withdrawals because of side effects is open to criticism as a crude evaluation of a complex issue. A fifth problem is that conventional clinical measures may miss important improvements with biologics, for example work disability may be particularly reduced by TNFis. Finally, the studies may have recruited patients with different initial disease activities. We examined the entry criteria and baseline measures in all studies. Although they showed a degree of variability, all patients had active disease and there was no evidence that patients receiving cDMARDs had less active disease than those receiving TNFi/methotrexate combinations.
We mainly focused on trials in which methotrexate monotherapy was used as the control treatment. We considered that this provided the best approach for comparing cDMARDs with TNFi/methotrexate. On balance, we believe that it preferable to standardise different DMARD combinations against methotrexate monotherapy, especially in early RA, as trials with less effective monotherapy arms create uncertainty when interpreting the findings.
Strengths
Both systematic reviews were large and they each enrolled > 5000 patients. They also showed very similar findings and the head-to-head trials gave similar supportive results. The findings in the TACIT trial are replicated in the different trials comparing cDMARDs and TNFi/methotrexate combinations with DMARD monotherapy, indicating that the trial results are likely to be generalisable.
Ethical issues
Trial design
The last decade has seen substantial progress in trial designs. 283 Regulatory pathways that demonstrate efficacy of new therapeutic agents have been agreed. The use of pure placebo treatment beyond 12–16 weeks is no longer considered ethical and consequently background therapy and early rescue has become regular practice. Identification of rare adverse events associated with new therapies has resulted in intensive safety evaluation during RCTs, a greater focus on post-marketing surveillance and use of registries, particularly for biological agents such as TNFis.
A crucial question before starting the TACIT trial was whether or not the trial was genuinely ethical. This is because the TACIT trial potentially involved restricting access to TNFis in patients with RA who met the criteria to receive these agents. This section deals with the relevant ethical issues. Although observational studies often provide similar results to those of randomised trials,284 these similarities are not universal,285 and clinical practice is unlikely to change in the absence of clinical trials that establish equivalence between cDMARDs and TNFis
Risks and anticipated benefits for trial participants and society
Equipoise, or the uncertainty principle, is a key requirement for RCTs in which the best treatment must be unknown so that participants do not suffer harm by assignment to one particular arm. 286,287 Alternative ethical approaches to RCTs have not gained universal acceptance and strategies such as equipoise-stratified randomisation are not widely used. 288–291
Equipoise in individual patients reflects not only the scientific probabilities of particular outcomes (known to trial clinicians) but also the value that individuals place on particular outcomes or risks (known only to individual patients). The solution to varying individual patient equipoise is genuine consultation with each patient about the choices using clear written information; similar approaches can be used with referring clinicians. It is difficult to ascertain the level of equipoise across the patient community for two reasons. First, patients hold highly variable views on the impact of and risks from adverse events to drug treatments. Second, patients also have highly variable views on the severity and ultimate outcome of RA. 292,293
Community equipoise is essential for a RCT to be ethical. We considered that there was sufficient equipoise among rheumatologists on when to use TNFis to justify our proposed RCT. Rheumatologists vary markedly in prescribing these agents. Discussion with consultant rheumatologists suggests that TNFis are felt to be highly effective but there is debate on which patients should receive them (in terms of severity and stage of disease), on their long-term risks and benefits and on their advantages over maximal existing therapy. There is also uncertainty, especially among public health clinicians, about their cost-effectiveness.
Concerns for patients entering this trial
Apart from general concerns about randomisation, especially for individuals who do not perceive true equipoise between treatments, there was a specific emotive concern about ‘entitlement’ to anti-TNF agents. Initially, many UK patients believed that, compared with the USA and continental Europe, they were deprived of these agents on financial grounds. This was exacerbated by intense pharmaceutical company involvement with clinicians and some patient organisations and by media presentation of these agents as ‘miracle cures’. Alternatives such as cDMARDs, which are relatively inexpensive and can be prescribed generically, have not received the same amount of attention either in the general media or in information provided to patient groups. As access to TNFis remains variable, patients and clinicians may perceive the proposed trial as an additional means of inhibiting access. However, a strategy is needed as biologics cannot be given ‘on demand’ in our resource-limited health system, because of their long-term costs (reflecting high production costs), the need for indefinite treatment, their uncertain cost-effectiveness and the many new biologics coming on stream (e.g. abatacept and rituximab).
Public issues and concerns
In the authors’ opinion a national strategy for using TNFis is required, taking into account the extensive new emerging information about these treatments. The adoption of new agents goes through several phases. Initially, they are considered safe and effective. Adverse events are underestimated at this stage, reflecting selective recruitment to clinical trials, careful patient follow-up in trials, the expertise of the research clinicians and the small numbers of patients treated; efficacy is overestimated for similar reasons. The next phase of drug adoption involves a reaction against the agent precipitated by unexpected side effects and the recognition that the agent does not fulfil all of its initial promise. TNFis are leaving the initial phase as many patients do not respond, those who do respond require continual treatment and large studies have been published describing more accurately rare, serious complications such as infection and cancer. They now need to enter the final stage of drug adoption, in which their advantages and disadvantages are seen in a balanced light. We believe that the TACIT trial is therefore timely from the perspective of both patients and recruiting clinicians.
Informing potential participants of benefits and risks
Potential participants were identified by rheumatologists and specialist nurses in routine clinics at participating centres. They received a brief summary of relevant information about the trial including information on the key risks and benefits. Those patients who were interested received a full patient information sheet explaining in plain English the purpose of the study and the actual and potential risks and benefits of DMARD combination therapy compared with the risks and benefits of treatment with TNFis. The patient information sheet was drawn up by the investigators and patient representatives based on the analysis of risks and benefits in this application. Advice was sought from the full trial patient representatives group and the Trial Steering Committee before submission to the relevant research ethics committee.
Clinical implications
General implications
Tumour necrosis factor inhibitors and other biological treatments have revolutionised the treatment of RA and other inflammatory immune disorders. The TACIT trial underlines the need for patients to have ongoing access to these treatments. There is no evidence in the TACIT trial to indicate that TNFis do not have a crucial role in the treatment of RA.
Clinical implications
A range of leading experts helped devise existing NICE guidance for the use of TNFis in active RA,81 which was based on extensive reviews of RCTs and associated observational studies. The rationale for using TNFis is mainly derived from extrapolating the results of these placebo-controlled trials using modelling studies that examine the health economic benefits of TNFis, with the help of historical data from observational studies. Before the TACIT trial there have been no head-to-head trials comparing TNFis with effective alternative treatments in established RA.
There have been three head-to-head trials of cDMARDs compared with TNFis in early RA. These trials all show that treatment strategies starting with cDMARDs or with TNFis give equivalent results over 12–24 months. As a consequence, there is no strong indication to start TNFis in preference to cDMARDs in early RA patients. Current NICE guidance, in our view, correctly recommends that cDMARDs are used in active early RA. 72
The balance of current evidence suggests that the key role of TNFis in RA is in active disease that is not fully controlled by DMARDs. Placebo-controlled trials have established the efficacy of TNFis. Observational studies in registries have confirmed their safety. However, neither approach has identified how best to use them. We consider that defining their optimal use requires undertaking head-to-head trials of different treatment strategies. Although more than a decade has passed since their introduction, we still do not know their value as short-term tapered treatments or whether they should be given to selected subsets of patients.
If TNFis were low-cost treatments there would be little concern about their optimal use. However, they are among the most expensive of those treatments that are used for relatively common diseases. As a consequence, the payers for health care wish to ensure that their use delivers true ‘value for money’.
If TNFis ensured that most patients with active RA who received them entered a period of sustained remission, there would be relatively little difficulty defending their widespread use. However, the TACIT trial and all other trials and observational studies show that only a minority of patients with active RA who receive TNFis achieve sustained remission.
Tumour necrosis factor inhibitors are usually simple for patients to take, adverse events are relatively uncommon and the onset of their effect is usually fairly rapid. Therefore, if cost was not an issue most patients would probably prefer to take TNFis rather than try cDMARDs. 294 However, this is probably the wrong question to ask. As neither strategy in the TACIT trial ensured that most patients with active RA enter remission, the real need is to identify more effective and more cost-effective treatment strategies.
The TACIT trial therefore shows that the current approach to using TNFis in established RA, encapsulated within current NICE guidance, does not necessarily result in cost-effective outcomes in all patients. We do not consider that using cDMARDs followed by TNFis represents an ideal approach. Instead, further research is needed to identify more effective treatment strategies. For the present it appears preferable to ensure that patients with active established RA receive the most clinically effective and cost-effective treatment possible. From this perspective offering cDMARDs before TNFis appears to be appropriate and sensible.
The model of care used in the TACIT trial assumed that all patients with active established RA should be offered similar treatment. Using this approach some patients achieved a very good response with TNFis, a slightly small number of patients achieved a very good response with cDMARDs, a few patients achieved a very good response when they received TNFis after failing to respond to cDMARDs and most patients had a relatively poor response to all treatments. Universal treatment strategies do not appear to be very effective. The most sensible approach would be to individualise care. 295,296
Research implications
Most clinicians consider that TNFis are highly effective treatments for active RA. However, we have found them to be no better than intensive cDMARDs for many patients. One reason for clinicians favouring them is their rapid onset of action. Another reason is that patients enrolled in early trials of biologics had more severe RA than is normally seen in current routine practice. 297 As a consequence, the benefits of biologics in these trials may have appeared greater that the benefit that would be likely to occur when they are used in routine practice settings. In addition, there is extensive evidence, at least in some countries, that patients starting biologics in clinical practice have far milder disease than patients in clinical trials,298–301 making the translation of research findings into practice recommendations particularly challenging.
The TACIT trial was a strategy trial that required patients to attend outpatient clinics for monthly review and involved substantial efforts from both patients and the rheumatologists and specialist nurses in the collaborating centres. Before the start of the trial there were concerns about the ethics of asking patients to wait for biological treatments and whether or not patients would wish to participate. One important conclusion from the TACIT trial is that comparative trials of high-cost treatments are feasible in long-term disorders such as RA. Patients and clinicians are willing to take part in such trials and when they are undertaken in routine clinic settings they can deliver results of potential clinical relevance.
The TACIT trial involved giving patients intensive cDMARD treatments that were organised by specialist nurses and supervised by rheumatologists. Although some training was provided in the specific organisation of the trial, this did not include detailed training about how to deliver intensive DMARD combinations. Nevertheless, specialist nurses achieved this without any difficulties being encountered. A second general conclusion therefore is that rheumatology specialist nurses have sufficiently high levels of clinical skills to deliver more intensive DMARD combination therapy. It would therefore be possible to deliver this management strategy within existing specialist centres using currently available staff.
Finally, the costs of undertaking the TACIT trial merit consideration. The trial was funded by a substantial grant from the National Institute for Health Research Health Technology Assessment programme and without this grant it could not have been undertaken. However, the savings from not prescribing TNFis within the TACIT trial to patients who met the criteria for receiving theses biologics but who received cheaper DMARDs meant that the overall cost of the TACIT trial to the NHS was relatively small. Therefore, we consider that it is possible to undertake further strategy trials of high-cost treatments such as TNFis for minimal additional costs to the NHS as a whole. Many NHS patients receiving high-cost biologics for arthritis could be enrolled in strategy trials such as the TACIT trial to help the NHS identify the most effective and cost-effective ways to use high-cost treatments.
The TNFis used in the TACIT trial and a number of other biological agents in RA are licensed within Europe and North America for treating active RA. The Phase II and Phase III development programmes for these agents have all been funded by their manufacturers and have used broadly similar trial methods, focusing on patients who have failed to respond to treatments such as methotrexate either remaining on this treatment or taking an additional biologic. Such trial designs are efficient in establishing whether or not the biologics are effective. However, the regulatory process does not involve head-to-head comparisons of biologics with effective standard treatments. It is likely that the widespread adoption of the current approach by regulatory agencies might have overemphasised the benefits of biologics compared with other less expensive forms of treatment. Clearly this a complex issue as there is a balance between the complexity and duration of the regulatory process and the need to obtain full information about the relative value of new treatments. In our view there are advantages in placing head-to-head trials with effective comparators at some point in the regulatory pathway, an assessment that has been made by others. 302,303
Tumour necrosis factor inhibitors achieve rapid improvements in the ESR and other measures of disease activity compared with conventional DMARDs. Indeed, some licensed DMARDs, such as ciclosporin, have little impact on the ESR. The use of composite measures to assess treatment response in RA, such as the DAS28 and ACR response, is likely, in our opinion, to unduly favour TNFis. The impact of TNFis on measures such as the HAQ and EQ-5D, which are more reflective of patients’ overall status, is less marked. We are unconvinced that the disproportionate impact of TNFis on laboratory measures such as the ESR is of clinical consequence, and it may lead to an overemphasis on improving laboratory as opposed to clinical measures. In the TACIT trial we found that this rapid improvement in ESR was not immediately related to decreases in clinical measures of direct importance to patients, such as falls in tender and swollen joint counts. The development of the current assessment methods in clinical trials in RA, which date back to the 1990s, is based on expert opinion rather than direct evidence. Although the approach is likely to reduce sample sizes in trials, it may favour some forms of treatment over others. One way of minimising this risk is to ensure that trials use a wide range of measures. Using changes in some measures, such as the DAS28, to model changes in other measures, such as the HAQ and EQ-5D, seems particularly inappropriate.
The economic case for using biologics such as TNFis in RA depends on extrapolating the results of placebo-controlled trials and using historical data from observational cohorts of previously treated patients. This approach involves two challenges. First, it is difficult to be certain how non-biological treatments would affect RA patients over time. Many of the models assume that they would not do well but there is limited evidence to support this view. Second, the data used for modelling are often historical and changes in the severity and natural history of treated RA may mean that these historical data have limited relevance to current patients. We consider that the economic rationale for using biological treatments should involve more emphasis on directly collected information from clinical trials and give less emphasis to theoretical modelling over long time frames.
Chapter 5 Conclusions
Key finding
The TACIT trial showed that RA patients who have failed to respond to methotrexate and another DMARD show clinically important improvements over 12 months if initially treated with cDMARDs, reserving TNFis for non-responders to these combinations. These improvements were equivalent to those achieved by starting all patients on TNFis with methotrexate or another DMARD monotherapy. The cost of the approach focused on using cDMARDs initially is approximately half the cost of using TNFis during the first 12 months of treatment. The equivalence of cDMARDs and TNFis is confirmed in systematic reviews of published trials in both early and established RA.
Health-care implications
The results from the TACIT trial, together with the results of the systematic reviews of previous trials of intensive cDMARDs and TNFis in active early and established RA, suggest that the following points could be considered when deciding how best to treat patients with active established RA who have not responded to methotrexate:
-
There is an extensive body of direct and indirect evidence which shows that giving such patients intensive cDMARD therapy and reserving TNFis for 6-month non-responders is clinically effective. Both EQ-5D- and SF-36-based QALY assessments suggest that cDMARDs are also cost-effective. A 6-month period of cDMARD therapy is sufficient to assess its effectiveness and there is no evidence that patients have any long-term disadvantages in terms of future disability, quality of life or joint damage from taking DMARD combinations for 6 months, even if they fail to respond.
-
In active established RA, starting treatment with either cDMARDs or TNFis results in equivalent clinically relevant improvements in disability and quality of life over 12 months. Immediately starting TNFis gives rapid early reductions in disease activity compared with starting cDMARDs but these improvements do not result in larger reductions in disability. There is no evidence that either strategy is associated with substantial erosive damage; radiological progression was minimal with both cDMARDs and TNFis.
-
Only a minority of patients achieve sustained remission with cDMARDs or TNFis. This suggests that neither approach should be considered an ideal long-term treatment strategy for all RA patients. Instead, they appear to be therapeutic options that decrease disability and reduce disease activity in some patients with active established RA.
Research implications
The TACIT trial raises many questions as well as providing some answers. There are a number of research areas that need to be taken forward. The following issues appear to be particularly important:
-
Identifying predictors of response to cDMARDs and TNFis will enable a move towards individualised treatment. This is of crucial importance as some patients respond well to cDMARDs whereas others respond well to TNFis, and prospectively identifying potential good responders should optimise treatment outcomes. In essence, there is a need to move away from the conventional ‘one size fits all’ approach to a more personalised clinical care approach. Research needs to focus on identifying predictors of response to these different treatment approaches. One possible implication is that national guidance on treatment decisions for specific interventions given to individual patients may not represent the most effective way of planning the delivery of care. Guidance might be most appropriate if it is moved from the general to the specific.
-
We need to define the most effective ways of using current treatments, including undertaking more strategy trials to examine novel ways of using high-cost treatments. Examples include identifying the benefits of short courses of biologics in early RA, in which the rapid effects of biologics may be beneficial, and redefining the optimal duration of TNFi treatment in established RA. Currently, once started, TNFis are continued if patients respond. However, this approach is based on custom and practice and has not been tested in clinical trials. The TACIT trial suggests that TNFis have dramatic immediate benefits but that, as currently used, these major improvements are present by 2 or 3 months and patients do not generally improve further. It is possible that short-term ‘induction therapy’ might be particularly useful with these treatments. Such an approach would change the cost base of using biological treatments.
-
There should be a greater emphasis on head-to-head trials when defining the overall benefits of high-cost treatments in RA. Extrapolating the results of short-term placebo-controlled trials and using observational studies to model economic benefits are less helpful in determining treatment pathways. Only head-to-head trials of treatment strategies including economic analyses can help drive forward innovative, cost-effective treatment approaches involving biologics. The results of the TACIT trial do not indicate that there should be less overall use of TNFis but that they are not being used in a highly effective manner.
-
A range of new non-biological treatments, particularly kinase inhibitors, is being developed for RA and some of these agents may soon be introduced into clinical practice. It is too early to judge the potential impact of these new treatments but it is likely that the treatment paradigm will change as a result. The TACIT trial highlights the limitations of our current treatment paradigm and therefore strengthens the case for developing new approaches to disease management.
The biologics revolution following the introduction of TNFis into routine clinical practice has changed RA care and, in our view, has benefited patients substantially. Clinicians and patients were keen to have access to these treatments when they first became available. As with all new treatments this is likely to have resulted in a relative overestimation of their clinical and economic benefits. Time and experience usually temper the initial enthusiasm for new treatments and this is likely to be the case with biologics for RA during the next decade. Trials such as the TACIT trial should help modify previous potential overenthusiasm for biologics in RA. However, the development of new agents is more likely to have a major impact, as novelty is a potent driver for changing behaviour. In our view it does not matter so much what drives change; the crucial point is to realise that some changes are needed.
Acknowledgements
Contributors to the trial
The TACIT trial involved a very large number of staff, who ran the trial, served on its various committees and were involved in recruitment and patient care within the trial. We are very grateful for all of their help.
Trial office staff
Dr Kim Mahood.
Dr Kelly Gormley.
Rebecca Brendell.
Dominic Stringer.
Dr Anna Kowalczyk.
Mrs Beverley White-Alao.
Janice Jimenez.
Trial Steering Committee
Professor Peter Maddison (chairperson).
Dr Ernest Choy.
Dr Gabrielle Kingsley.
Professor Howard Bird.
Professor Bhaskar Dasgupta.
Professor Anisur Rahman.
Mrs Sylvia Greinig.
Mrs Sally Wilson.
Ms Diane Home.
Dr Taher Mahmud.
Dr Khalid Ahmed.
Dr Clive Kelly.
Dr Selwyn Richards.
Dr Sanjeev Patel.
Data Monitoring and Ethics Committee
Professor Deborah Symmons (chairperson).
Professor Peter Taylor.
Mrs Caroline Doré.
Principal investigators at clinical sites
Dr Khalid Ahmed, Princess Alexandra Hospital, Harlow.
Dr Sandra Green, Weston General Hospital, Weston-super-Mare.
Dr Anurag Bharadwaj, Basildon University Hospital.
Dr Fraser Birrell, Wansbeck General Hospital, Ashington.
Professor Kuntal Chakravarty, Queens Hospital, Romford.
Dr Gerald Coakley, Queen Elizabeth Hospital, Woolwich.
Professor Andrew Cope, Guy’s Hospital, London.
Dr Christopher Deighton, Derby City General Hospital.
Dr Karen Douglas, Russell’s Hall Hospital, Dudley.
Dr Sarah Medley, Orpington Hospital.
Dr Tania Gordon, Southend University Hospital.
Dr Clive Kelly, Queen Elizabeth Hospital, Gateshead.
Dr Namita Kumar, University Hospital of North Durham, Durham.
Dr Ramasharan Laxminarayan, Queen’s Hospital, Burton-on-Trent.
Dr Jon Packham, Haywood Hospital, Stroke-on-Trent.
Dr Ira Pande, Queens Medical Centre, Nottingham.
Dr Michael Plant, James Cook University Hospital, Middlesbrough.
Dr Selwyn Richards, Poole Hospital.
Dr Euthalia Roussou, King George’s Hospital, London.
Dr Thomas Sheeran, Cannock Chase Hospital, Cannock.
Dr Karen Walker-Bone, Royal Sussex County Hospital, Brighton.
Dr Christopher Erdhardt, Orpington Hospital.
Dr Atheer Al-Ansari, Weston General Hospital, Weston-super-Mare.
Other site staff (including research nurses and co-investigators)
Mrs Doris Butawan, Queens Hospital, Romford.
Ms Christina Blanco-Gil, Guy’s Hospital, London.
Mrs Alison Booth, Royal Derby Hospital.
Mrs Liz Dragonetti, Orpington Hospital.
Ms Julie Edwards, Cannock Chase Hospital, Cannock.
Mr Andy Georgiou, King George’s Hospital, London.
Ms Donna Gray, Wansbeck General Hospital, Ashington.
Ms Jane Hollywood, Southend University Hospital.
Ms Debbie Johnson, University Hospital Lewisham.
Mr Hugh Lloyd-Jones, Weston General Hospital, Weston-super-Mare.
Ms Lucy Kadiki, Russell’s Hall Hospital, Dudley.
Ms Ann Barcroft, Haywood Hospital, Stroke-on-Trent.
Mrs Margot Lilley, Freeman Hospital, Newcastle upon Tyne.
Ms Febisola Akinboyewa, Queen Elizabeth Hospital, Woolwich, London.
Ms Rashidat Adeniba, Basildon University Hospital.
Ms Marie-Josephe Pradere, Queens Medical Centre, Nottingham.
Ms Susan Pugmire, Queen Elizabeth Hospital, Gateshead.
Ms Pamela Race, University Hospital of North Durham, Durham.
Mrs Rosaria Salerno, Kings College Hospital, London.
Ms Michele Powell, Royal Sussex County Hospital, Brighton.
Mrs Jane Solomon, Princess Alexandra Hospital, Harlow.
Ms Annie Baker, Poole Hospital.
Ms Jane Whitmore, Queen’s Hospital, Burton upon Trent.
Dr Laith Al-Sweedan, Queen’s Hospital, Romford.
Kerry Goodsell, Basildon University Hospital.
Bernard Hadebe, Basildon University Hospital.
Nhlanhla Mguni, Basildon University Hospital.
Dr Amel Ginawi, Basildon University Hospital.
Maxwell Masuku, Basildon University Hospital.
Dr Nagui Gendi, Basildon University Hospital.
Julie Edwards, Cannock Chase Hospital, Cannock.
Sarah Stevenson, Cannock Chase Hospital, Cannock.
Sharon Murphy, Cannock Chase Hospital, Cannock.
Elaine Taylor, Cannock Chase Hospital, Cannock.
Dr Thomas Price, Cannock Chase Hospital, Cannock.
Joanne Logan, Cannock Chase Hospital, Cannock.
Dr Venkata Chalam Cannock Chase Hospital, Cannock.
Dr Abdul Baker, Cannock Chase Hospital, Cannock.
Sally Giles, Cannock Chase Hospital, Cannock.
Annette Wilkinson, Cannock Chase Hospital, Cannock.
Jacqueline Peake, Cannock Chase Hospital, Cannock.
Deborah Lloyd, Cannock Chase Hospital, Cannock.
Judith Bellaby, Cannock Chase Hospital, Cannock.
Dr Diarmuid Mulherin, Cannock Chase Hospital, Cannock.
Dr Tina Ding, Royal Derby Hospital, Derby.
Jo Morris, Royal Derby Hospital, Derby.
Sandra Robinson, Freeman Hospital, Newcastle upon Tyne.
Heather Fogo, Freeman Hospital, Newcastle upon Tyne.
Dr Pamela Peterson, Freeman Hospital, Newcastle upon Tyne.
Laura Blackler, Guy’s Hospital, London.
Dr Toby Garrood, Guy’s Hospital, London.
Dr Margaret Ma, Guy’s Hospital, London.
Dr Edward Roddy, Haywood Hospital, Stroke-on-Trent.
Dr Sanjeet Kamath, Haywood Hospital, Stroke-on-Trent.
Dr Samantha Hider, Haywood Hospital, Stroke-on-Trent.
Ann Brownfield, Haywood Hospital, Stroke-on-Trent.
Julie Gray, Haywood Hospital, Stroke-on-Trent.
Dr Kamran Naraghi, James Cook University Hospital, Middlesbrough.
Val Lunn, James Cook University Hospital, Middlesbrough.
Kathleen Bell, James Cook University Hospital, Middlesbrough.
Joanne Dobson, King’s College Hospital, London.
Radka Chura, King’s College Hospital, London.
Gayle Porter, King’s College Hospital, London.
Aderonke Olatunji, King’s College Hospital, London.
Aysha Khanom, King’s College Hospital, London.
Dr Nicola Gullick, King’s College Hospital, London.
Nestor Salazar, King’s College Hospital, London.
Dr Richard Campbell, King’s College Hospital, London.
Dr Sarah Levy, King’s College Hospital, London.
Dr Myles Lewis, King’s College Hospital, London.
Dr Sophia Steer, King’s College Hospital, London.
Dr Ernest Choy, King’s College Hospital, London.
Dr Ian Scott, Lewisham Hospital, London.
Dr Ian Gaywood, Lewisham Hospital, London.
Jenny Berrington, Orpington Hospital, London.
Dr Amit Saha, Orpington Hospital, London.
Dr Pauline Pitt, Orpington Hospital, London.
Dr Khaldoun Chaabo, Orpington Hospital, London.
Julia Taylor, Poole Hospital, Dorset.
Dr Sarah Westlake, Poole Hospital, Dorset.
Dr Fouz Rahmeh, Poole Hospital, London.
Dr Paul Thompson, Poole Hospital, London.
Dr Melonie Sriranganathan, Poole Hospital, London.
Charlottle Mahuma, Princess Alexandra Hospital, Harlow.
Dr Sarah Farrow, Princess Alexandra Hospital, Harlow.
Ed Ekanem, Princess Alexandra Hospital, Harlow.
Carol Ann Keel, Princess Alexandra Hospital, Harlow.
Manju Joy, Princess Alexandra Hospital, Harlow.
Julie Leggett, Queen’s Hospital, Burton-on-Trent.
Dr Subhashini Arthanari, Queen’s Hospital, Burton-on-Trent.
Dr Mohamed Nisar, Queen’s Hospital, Burton-on-Trent.
Dr Vadivelu Saravanan, Queen Elizabeth Hospital, Gateshead.
Jennifer Hamilton, Queen Elizabeth Hospital, Gateshead.
Carol Heycock, Queen Elizabeth Hospital, Gateshead.
Julie Dodds, Queen Elizabeth Hospital, Gateshead.
Dr Louise Dolan, Queen Elizabeth Hospital, Woolwich.
Dr Amit Saha, Queen Elizabeth Hospital, Woolwich.
Ratidzo Maboreke, Queen Elizabeth Hospital, Woolwich.
Dr Cathy Mathews, Queen Elizabeth Hospital, Woolwich.
Leah Irungu, Queen Elizabeth Hospital, Woolwich.
Grace Bonnici, Queen’s Hospital, Romford.
Avani Shukla, Queen’s Hospital, Romford.
Dr Inam Haq, Royal Sussex County Hospital, Brighton.
Professor Kevin Davies, Royal Sussex County Hospital, Brighton.
Mel Smith, Royal Sussex County Hospital, Brighton.
Wendy Harman, Royal Sussex County Hospital, Brighton.
Kate Trivedi, Royal Sussex County Hospital, Brighton.
Professor George Kitas, Russell’s Hall Hospital, Dudley.
Tracy Toms, Russell’s Hall Hospital, Dudley.
Daljit Kaur, Russell’s Hall Hospital, Dudley.
Kirsty Baron, Russell’s Hall Hospital, Dudley.
Professor Bhaskar Dasgupta, Southend University Hospital, Essex.
Dr Nada Hassan, Southend University Hospital, Essex.
Dr Dimitrios Christidis, Southend University Hospital, Essex.
Pam Long, Southend University Hospital, Essex.
Victoria Katsande, Southend University Hospital, Essex.
Kirstie Walker, Wansbeck General Hospital, Ashington.
Dr Tehseen Ahmed, Weston General Hospital, Weston-super-Mare.
Dr Matthew Roy, Weston General Hospital, Weston-super-Mare.
Glenn Saunders, Weston General Hospital, Weston-super-Mare.
Dawn Simmons, Weston General Hospital, Weston-super-Mare.
Donna Cotterill, Weston General Hospital, Weston-super-Mare.
Pharmacists
Peter Croot, Basildon Hospital, Essex.
Omolara Ejiwuumi, Basildon Hospital, Essex.
Andreas Muenstedt, Basildon Hospital, Essex.
Ann Bentley, Basildon Hospital, Essex.
Susan Price, Basildon Hospital, Essex.
Sharon Hanson, Basildon Hospital, Essex.
Peter Fox, Royal Derby Hospital, Derby.
Margaret Harper, Royal Derby Hospital, Derby.
Wendy Abbott, Royal Derby Hospital, Derby.
Maria Allen, Freeman Hospital, Newcastle upon Tyne.
Sarah Lynn Robertson, Freeman Hospital, Newcastle upon Tyne.
Maureen Foreman, Freeman Hospital, Newcastle upon Tyne.
Julie Stephenson, Freeman Hospital, Newcastle upon Tyne.
Deirdre Wood, Guy’s Hospital, London.
Negood Baggash, Guy’s Hospital, London.
Eve Wisdom, Guy’s Hospital, London.
Chi Kai Tam, Guy’s Hospital, London.
Shane Artis, Haywood Hospital, Stroke-on-Trent.
Susan Rachel Abell, Haywood Hospital, Stroke-on-Trent.
Agnieszka Skotnicka, James Cook University Hospital, Middlesbrough.
Helen Carver, James Cook University Hospital, Middlesbrough.
Senait Haile, King’s College Hospital, London.
Gabrielle Ellis, King’s College Hospital, London.
Joanne Gordon, King’s College Hospital, London.
Gaynor Notcheva, King’s College Hospital, London.
Fatima El-Oulidi, King’s College Hospital, London.
Madhavi Dudheiya, King’s College Hospital, London.
Donna Palmer, King’s College Hospital, London.
Asia Flanagan, King’s College Hospital, London.
Jacqueline Ricketts, King George Hospital, Ilford, London.
Carla Hunt, King George Hospital, Ilford, London.
Sharon Hoyte, Lewisham Hospital.
Jagdev Bains, Lewisham Hospital.
Sheila Hodgson, Queen’s Medical Centre, Nottingham.
Joyce Handley, Queen’s Medical Centre, Nottingham.
Adam Henderson, Queen’s Medical Centre, Nottingham.
Lisa Humphries, Queen’s Medical Centre, Nottingham.
Bernie Cook, Queen’s Medical Centre, Nottingham.
Anthony Mazzei, Queen’s Medical Centre, Nottingham.
Lorraine Jaundrill, Queen’s Medical Centre, Nottingham.
Betty Chan, Orpington Hospital, London.
Alison John, Orpington Hospital, London.
Deryck Burton, Poole Hospital, Dorset.
Sharon Power, Poole Hospital, Dorset.
Cherise Sweatland, Poole Hospital.
Evelyn Holmes, Princess Alexandra Hospital, Harlow.
Chris Curtus, Queen’s Hospital, Burton-on-Trent.
Ann Adams, Queen’s Hospital, Burton-on-Trent.
Gavin Mankin, Queen Elizabeth Hospital, Gateshead.
Namrita Sen, Queen Elizabeth Hospital, Woolwich.
Yousaf Razzak, Queen’s Hospital, Romford.
Parveen Dugh, Queen’s Hospital, Romford.
Tenesa Sargent, Royal Sussex County Hospital, Brighton.
Amy Murray, Royal Sussex County Hospital, Brighton.
Jodie Smith, Royal Sussex County Hospital, Brighton.
Hayley Pearson, Russell’s Hall Hospital, Dudley.
Stuart Chandler, Southend Hospital, Essex.
Rebecca Palmer, Southend Hospital, Essex.
Michael Donaghy, Southend Hospital, Essex.
Terry Dowling, Southend Hospital, Essex.
Andrew Parker, University Hospital of North Durham, Durham.
Penny Gamble, University Hospital of North Durham, Durham.
Richard Copeland, Wansbeck General Hospital, Ashington.
Sarah Jobling, Wansbeck General Hospital, Ashington.
Sharon Stothart, Wansbeck General Hospital, Ashington.
Lesley Barnfather, Wansbeck General Hospital, Ashington.
Gillian Kincaid, Wansbeck General Hospital, Ashington.
Andrew Mckendrick, Weston Hospital, Weston-super-Mare.
Kathy Beard, Weston Hospital, Weston-super-Mare.
Sally Squire, Weston Hospital, Weston-super-Mare.
Contributions of authors
David L Scott (Professor of Clinical Rheumatology) designed the study, contributed to clinical data acquisition, contributed to the systematic reviews and drafted the final report.
Fowzia Ibrahim (Statistician) undertook the primary analysis of the trial data, contributed to the systematic reviews and contributed to the final report.
Vern Farewell (Professor and Scientific Programme Leader, Statistics) designed the study, oversaw the statistical analysis and contributed to the final report.
Aidan G O’Keeffe (Statistician) supported the analysis of the trial data, provided independent imputation of missing data and reviewed the final report.
Margaret Ma (Clinical Research Fellow, Rheumatology) undertook the systematic reviews and contributed to the final report.
David Walker (Consultant Rheumatologist) designed the study, contributed to clinical trial data acquisition and reviewed the final report.
Margaret Heslin (Research Assistant, Health Economics) contributed to the economic analysis and the final report.
Anita Patel (Reader in Health Economics) designed the study, led the economic analysis and contributed to the final report.
Gabrielle Kingsley (Professor of Clinical Rheumatology) designed the study, contributed to clinical trial data acquisition and reviewed the final report.
Disclaimers
This report presents independent research funded by the National Institute for Health Research (NIHR). The views and opinions expressed by authors in this publication are those of the authors and do not necessarily reflect those of the NHS, the NIHR, NETSCC, the HTA programme or the Department of Health. If there are verbatim quotations included in this publication the views and opinions expressed by the interviewees are those of the interviewees and do not necessarily reflect those of the authors, those of the NHS, the NIHR, NETSCC, the HTA programme or the Department of Health.
References
- Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet 2010;376:1094-108. http://dx.doi.org/10.1016/S0140-6736(10)60826-4.
- Klarenbeek NB, Kerstens PJ, Huizinga TW, Dijkmans BA, Allaart CF. Recent advances in the management of rheumatoid arthritis. BMJ 2010;341. http://dx.doi.org/10.1136/bmj.c6942.
- McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011;365:2205-19. http://dx.doi.org/10.1056/NEJMra1004965.
- Ropes MW, Bennett GA, Cobb S, Jacox R, Jessar RA. 1958 revision of diagnostic criteria for rheumatoid arthritis. Arthritis Rheum 1959;2:16-20. http://dx.doi.org/10.1002/1529-0131(195902)2:1<16::AID-ART1780020104>3.0.CO;2-9.
- Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1988;31:315-24. http://dx.doi.org/10.1002/art.1780310302.
- Aletaha D, Neogi T, Silman A, Funovits J, Felson D, Bingham CO, et al. The 2010 American College of Rheumatology/European League against Rheumatism classification criteria for rheumatoid arthritis. Ann Rheum Dis 2010;69:1580-8. http://dx.doi.org/10.1136/ard.2010.138461.
- Scott DL, Steer S. The course of established rheumatoid arthritis. Best Pract Res Clin Rheumatol 2007;21:943-67. http://dx.doi.org/10.1016/j.berh.2007.05.006.
- Young A, Koduri G. Extra-articular manifestations and complications of rheumatoid arthritis. Best Pract Res Clin Rheumatol 2007;21:907-27. http://dx.doi.org/10.1016/j.berh.2007.05.007.
- Sokka T, Abelson B, Pincus T. Mortality in rheumatoid arthritis: 2008 update. Clin Exp Rheumatol 2008;26:S35-61.
- Allaire S, Wolfe F, Niu J, LaValley MP, Zhang B, Reisine S. Current risk factors for work disability associated with rheumatoid arthritis: recent data from a US national cohort. Arthritis Rheum 2009;61:321-8. http://dx.doi.org/10.1002/art.24281.
- Smolen JS, van der Heijde DM, Aletaha D, Xu S, Han J, Baker D, et al. Progression of radiographic joint damage in rheumatoid arthritis: independence of erosions and joint space narrowing. Ann Rheum Dis 2009;68:1535-40. http://dx.doi.org/10.1136/ard.2008.094128.
- Michaud K, Wolfe F. Comorbidities in rheumatoid arthritis. Best Pract Res Clin Rheumatol 2007;21:885-906. http://dx.doi.org/10.1016/j.berh.2007.06.002.
- Dougados M, Aletaha D, van Riel P. Disease activity measures for rheumatoid arthritis. Clin Exp Rheumatol 2007;25:S22-9.
- Nishimura K, Sugiyama D, Kogata Y, Tsuji G, Nakazawa T, Kawano S, et al. Meta-analysis: diagnostic accuracy of anti-cyclic citrullinated peptide antibody and rheumatoid factor for rheumatoid arthritis. Ann Intern Med 2007;146:797-808. http://dx.doi.org/10.7326/0003-4819-146-11-200706050-00008.
- van der Linden MP, van der Woude D, Ioan-Facsinay A, Levarht EW, Stoeken-Rijsbergen G, Huizinga TW, et al. Value of anti-modified citrullinated vimentin and third-generation anti-cyclic citrullinated peptide compared with second-generation anti-cyclic citrullinated peptide and rheumatoid factor in predicting disease outcome in undifferentiated arthritis and rheumatoid arthritis. Arthritis Rheum 2009;60:2232-41. http://dx.doi.org/10.1002/art.24716.
- Whiting PF, Smidt N, Sterne JA, Harbord R, Burton A, Burke M, et al. Systematic review: accuracy of anti-citrullinated peptide antibodies for diagnosing rheumatoid arthritis. Ann Intern Med 2010;152:456-64. http://dx.doi.org/10.7326/0003-4819-152-7-201004060-00010.
- Yazici Y, Sokka T, Pincus T. Radiographic measures to assess patients with rheumatoid arthritis: advantages and limitations. Rheum Dis Clin North Am 2009;35:723-9. http://dx.doi.org/10.1016/j.rdc.2009.10.005.
- Boutry N, Morel M, Flipo RM, Demondion X, Cotten A. Early rheumatoid arthritis: a review of MRI and sonographic findings. Am J Roentgenol 2007;189:1502-9. http://dx.doi.org/10.2214/AJR.07.2548.
- Kubassova O, Boesen M, Peloschek P, Langs G, Cimmino MA, Bliddal H, et al. Quantifying disease activity and damage by imaging in rheumatoid arthritis and osteoarthritis. Ann N Y Acad Sci 2009;1154:207-38. http://dx.doi.org/10.1111/j.1749-6632.2009.04392.x.
- Jacob N, Jacob CO. Genetics of rheumatoid arthritis: an impressionist perspective. Rheum Dis Clin North Am 2012;38:243-57. http://dx.doi.org/10.1016/j.rdc.2012.05.001.
- Sugiyama D, Nishimura K, Tamaki K, Tsuji G, Nakazawa T, Morinobu A, et al. Impact of smoking as a risk factor for developing rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis 2010;69:70-81. http://dx.doi.org/10.1136/ard.2008.096487.
- van Venrooij WJ, van Beers JJ, Pruijn GJ. Anti-CCP antibodies: the past, the present and the future. Nat Rev Rheumatol 2011;7:391-8. http://dx.doi.org/10.1038/nrrheum.2011.76.
- Pugner KM, Scott D, Holmes JW, Hieke K. The costs of rheumatoid arthritis: an international long-term view. Semin Arthritis Rheum 2000;29:305-20. http://dx.doi.org/10.1016/S0049-0172(00)80017-7.
- Bansback N, Marra CA, Finckh A, Anis A. The economics of treatment in early rheumatoid arthritis. Best Pract Res Clin Rheumatol 2009;23:83-92. http://dx.doi.org/10.1016/j.berh.2008.11.005.
- Nixon RM, O’Hagan A, Oakley J, Madan J, Stevens JW, Bansback N, et al. The Rheumatoid Arthritis Drug Development Model: a case study in Bayesian clinical trial simulation. Pharm Stat 2009;8:371-89. http://dx.doi.org/10.1002/pst.368.
- Services for People with Rheumatoid Arthritis. London: The Stationery Office; 2009.
- van Riel PL, Fransen J. Established rheumatoid arthritis: clinical assessments. Best Pract Res Clin Rheumatol 2007;21:807-25. http://dx.doi.org/10.1016/j.berh.2007.06.001.
- Wells GA. Patient-driven outcomes in rheumatoid arthritis. J Rheumatol Suppl 2009;82:33-8. http://dx.doi.org/10.3899/jrheum.090129.
- Sokka T, Rannio T, Khan NA. Disease activity assessment and patient-reported outcomes in patients with early rheumatoid arthritis. Rheum Dis Clin North Am 2012;38:299-310. http://dx.doi.org/10.1016/j.rdc.2012.04.005.
- Bruce B, Fries JF. The Health Assessment Questionnaire (HAQ). Clin Exp Rheumatol 2005;23:S14-18.
- Wolfe F. Fatigue assessments in rheumatoid arthritis: comparative performance of visual analog scales and longer fatigue questionnaires in 7760 patients. J Rheumatol 2004;31:1896-902.
- Gettings L. Psychological well-being in rheumatoid arthritis: a review of the literature. Musculoskeletal Care 2010;8:99-106. http://dx.doi.org/10.1002/msc.171.
- Fransen J, van Riel PL. The Disease Activity Score and the EULAR response criteria. Rheum Dis Clin North Am 2009;35:745-57. http://dx.doi.org/10.1016/j.rdc.2009.10.001.
- Aletaha D, Smolen JS. The Simplified Disease Activity Index and Clinical Disease Activity Index to monitor patients in standard clinical care. Rheum Dis Clin North Am 2009;35:759-72. http://dx.doi.org/10.1016/j.rdc.2009.10.006.
- Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith C, et al. American College of Rheumatology preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995;38:727-35. http://dx.doi.org/10.1002/art.1780380602.
- Thabet MM, Huizinga TW, van der Heijde DM, van der Helm-van Mil AH. The prognostic value of baseline erosions in undifferentiated arthritis. Arthritis Res Ther 2009;11. http://dx.doi.org/10.1186/ar2832.
- Scott DL. Radiological progression in established rheumatoid arthritis. J Rheumatol Suppl 2004;69:55-6.
- Baillet A, Gaujoux-Viala C, Mouterde G, Pham T, Tebib J, Saraux A, et al. Comparison of the efficacy of sonography, magnetic resonance imaging and conventional radiography for the detection of bone erosions in rheumatoid arthritis patients: a systematic review and meta-analysis. Rheumatology 2011;50:1137-47. http://dx.doi.org/10.1093/rheumatology/keq437.
- Keystone EC, Smolen J, van Riel P. Developing an effective treatment algorithm for rheumatoid arthritis. Rheumatology 2012;51:48-54. http://dx.doi.org/10.1093/rheumatology/kes122.
- Felson D. Defining remission in rheumatoid arthritis. Ann Rheum Dis 2012;71:86-8. http://dx.doi.org/10.1136/annrheumdis-2011-200618.
- Ma MH, Scott IC, Kingsley GH, Scott DL. Remission in early rheumatoid arthritis. J Rheumatol 2010;37:1444-53. http://dx.doi.org/10.3899/jrheum.091131.
- Thiele K, Huscher D, Bischoff S, Späthling-Mestekemper S, Backhaus M, Aringer M, et al. Performance of the 2011 ACR/EULAR preliminary remission criteria compared with DAS28 remission in unselected patients with rheumatoid arthritis. Ann Rheum Dis 2013;72:1194-9. http://dx.doi.org/10.1136/annrheumdis-2012-201821.
- Bakker MF, Jacobs JW, Verstappen SM, Bijlsma JW. Tight control in the treatment of rheumatoid arthritis: efficacy and feasibility. Ann Rheum Dis 2007;66:iii56-60. http://dx.doi.org/10.1136/ard.2007.078360.
- Scott DL. Biologics-based therapy for the treatment of rheumatoid arthritis. Clin Pharmacol Ther 2012;91:30-43. http://dx.doi.org/10.1038/clpt.2011.278.
- Boers M, van Tuyl L, van den Broek M, Kostense PJ, Allaart CF. Meta-analysis suggests that intensive non-biological combination therapy with step-down prednisolone (COBRA strategy) may also ‘disconnect’ disease activity and damage in rheumatoid arthritis. Ann Rheum Dis 2013;72:406-9. http://dx.doi.org/10.1136/annrheumdis-2012-202333.
- Westhovens R, Verschueren P. Rheumatoid arthritis: defining remission in patients with RA in clinical practice. Nat Rev Rheumatol 2012;8:445-7. http://dx.doi.org/10.1038/nrrheum.2012.111.
- Jurgens MS, Jacobs JW, Bijlsma JW. The use of conventional disease-modifying anti-rheumatic drugs in established RA. Best Pract Res Clin Rheumatol 2011;25:523-33. http://dx.doi.org/10.1016/j.berh.2011.10.006.
- Strand V, Scott DL, Emery P, Kalden JR, Smolen JS, Cannon GW, et al. Leflunomide Rheumatoid Arthritis Investigators Groups. Physical function and health related quality of life: analysis of 2-year data from randomized, controlled studies of leflunomide, sulfasalazine, or methotrexate in patients with active rheumatoid arthritis. J Rheumatol 2005;32:590-601.
- Finckh A, Liang MH, van Herckenrode CM, de Pablo P. Long-term impact of early treatment on radiographic progression in rheumatoid arthritis: a meta-analysis. Arthritis Rheum 2006;55:864-72. http://dx.doi.org/10.1002/art.22353.
- Visser K, Katchamart W, Loza E, Martinez-Lopez JA, Salliot C, Trudeau J, et al. 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. http://dx.doi.org/10.1136/ard.2008.094474.
- Jobanputra P, Wilson J, Douglas K, Burls A. A survey of British rheumatologists’ DMARD preferences for rheumatoid arthritis. Rheumatology 2004;43:206-10. http://dx.doi.org/10.1093/rheumatology/keh003.
- Gaujoux-Viala C, Smolen JS, Landewé R, Dougados M, Kvien TK, Mola EM, et al. Current evidence for the management of rheumatoid arthritis with synthetic disease-modifying antirheumatic drugs: a systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2010;69:1004-9. http://dx.doi.org/10.1136/ard.2009.127225.
- Mouterde G, Baillet A, Gaujoux-Viala C, Cantagrel A, Wendling D, Le Loët X, et al. Optimizing methotrexate therapy in rheumatoid arthritis: a systematic literature review. Joint Bone Spine 2011;78:587-92. http://dx.doi.org/10.1016/j.jbspin.2011.01.010.
- Suarez-Almazor ME, Belseck E, Shea B, Wells G, Tugwell P. Sulfasalazine for rheumatoid arthritis. Cochrane Database Syst Rev 2000;2.
- Osiri M, Shea B, Robinson V, Suarez-Almazor M, Strand V, Tugwell P, et al. Leflunomide for the treatment of rheumatoid arthritis: a systematic review and metaanalysis. J Rheumatol 2003;30:1182-90.
- Suarez-Almazor ME, Belseck E, Shea B, Homik J, Wells G, Tugwell P, et al. Antimalarials for treating rheumatoid arthritis. Cochrane Database Syst Rev 2000;4.
- Clark P, Tugwell P, Bennet K, Bombardier C, Shea B, Wells G, et al. Injectable gold for rheumatoid arthritis. Cochrane Database Syst Rev 2000;2.
- Suarez-Almazor ME, Spooner C, Belseck E. Azathioprine for treating rheumatoid arthritis. Cochrane Database Syst Rev 2000;4.
- Gotzsche PC, Johansen HK. Short-term low-dose corticosteroids vs placebo and nonsteroidal antiinflammatory drugs in rheumatoid arthritis. Cochrane Database Syst Rev 2004;3.
- Kirwan JR, Bijlsma JW, Boers M, Shea BJ. Effects of glucocorticoids on radiological progression in rheumatoid arthritis. Cochrane Database Syst Rev 2007;1.
- Gorter SL, Bijlsma JW, Cutolo M, Gomez-Reino J, Kouloumas M, Smolen JS, et al. Current evidence for the management of rheumatoid arthritis with glucocorticoids: a systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2010;69:1010-14. http://dx.doi.org/10.1136/ard.2009.127332.
- McCarty DJ. Treating intractable rheumatoid arthritis. N Engl J Med 1981;305:1009-11. http://dx.doi.org/10.1056/NEJM198110223051710.
- Porter DR, Capell HA, Hunter J. Combination therapy in rheumatoid arthritis – no benefit of addition of hydroxychloroquine to patients with a suboptimal response to intramuscular gold therapy. J Rheumatol 1993;20:645-9.
- Willkens RF, Urowitz MB, Stablein DM, McKendry RJ, Berger RG, Box JH, et al. Comparison of azathioprine, methotrexate, and the combination of both in the treatment of rheumatoid arthritis. A controlled clinical trial. Arthritis Rheum 1992;35:849-56. http://dx.doi.org/10.1002/art.1780350802.
- Felson DT, Anderson JJ, Meenan RF. The efficacy and toxicity of combination therapy in rheumatoid arthritis. A meta-analysis. Arthritis Rheum 1994;37:1487-91. http://dx.doi.org/10.1002/art.1780371012.
- Tugwell P, Pincus T, Yocum D, Stein M, Gluck O, Kraag G, et al. Combination therapy with cyclosporine and methotrexate in severe rheumatoid arthritis. The Methotrexate–Cyclosporine Combination Study Group. N Engl J Med 1995;333:137-41. http://dx.doi.org/10.1056/NEJM199507203330301.
- O’Dell JR, Haire CE, Erikson N, Drymalski W, Palmer W, Eckhoff PJ, et al. Treatment of rheumatoid arthritis with methotrexate alone, sulfasalazine and hydroxychloroquine, or a combination of all three medications. N Engl J Med 1996;334:1287-91. http://dx.doi.org/10.1056/NEJM199605163342002.
- Boers M, Verhoeven AC, Markusse HM, van de Laar MA, Westhovens R, van Denderen JC, et al. Randomised comparison of combined step-down prednisolone, methotrexate and suphasalazine with sulphasalazine alone in early rheumatoid arthritis. Lancet 1997;350:309-18. http://dx.doi.org/10.1016/S0140-6736(97)01300-7.
- van Eijk IC, Nielen MM, van der Horst-Bruinsma I, Tijhuis GJ, Boers M, Dijkmans BA, et al. Aggressive therapy in patients with early arthritis results in similar outcome compared with conventional care: the STREAM randomized trial. Rheumatology 2012;51:686-94. http://dx.doi.org/10.1093/rheumatology/ker355.
- Choy EH, Smith C, Dore CJ, Scott DL. A meta-analysis of the efficacy and toxicity of combining disease-modifying anti-rheumatic drugs in rheumatoid arthritis based on patient withdrawal. Rheumatology 2005;44:1414-21. http://dx.doi.org/10.1093/rheumatology/kei031.
- Kiely P, Williams R, Walsh D, Young A. Early Rheumatoid Arthritis Network. Contemporary patterns of care and disease activity outcome in early rheumatoid arthritis: the ERAN cohort. Rheumatology 2009;48:57-60. http://dx.doi.org/10.1093/rheumatology/ken406.
- Deighton C, O’Mahony R, Tosh J, Turner C, Rudolf M. Guideline Development Group . Management of rheumatoid arthritis: summary of NICE guidance. BMJ 2009;338. http://dx.doi.org/10.1136/bmj.b702.
- Feldmann M. Development of anti-TNF therapy for rheumatoid arthritis. Nat Rev Immunol 2002;2:364-71. http://dx.doi.org/10.1038/nri802.
- Elliott MJ, Maini RN, Feldmann M, Long-Fox A, Charles P, Katsikis P, et al. Treatment of rheumatoid arthritis with chimeric monoclonal antibodies to tumor necrosis factor alpha. Arthritis Rheum 1993;36:1681-90. http://dx.doi.org/10.1002/art.1780361206.
- Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C, Smolen JS, et al. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet 1994;344:1105-10. http://dx.doi.org/10.1016/S0140-6736(94)90628-9.
- Zintzaras E, Dahabreh IJ, Giannouli S, Voulgarelis M, Moutsopoulos HM. Infliximab and methotrexate in the treatment of rheumatoid arthritis: a systematic review and meta-analysis of dosage regimens. Clin Ther 2008;30:1939-55. http://dx.doi.org/10.1016/j.clinthera.2008.11.007.
- Wiens A, Correr CJ, Pontarolo R, Venson R, Quinalha JV, Otuki MF. A systematic review and meta-analysis of the efficacy and safety of etanercept for treating rheumatoid arthritis. Scand J Immunol 2009;70:337-44. http://dx.doi.org/10.1111/j.1365-3083.2009.02296.x.
- Wiens A, Correr CJ, Venson R, Otuki MF, Pontarolo R. A systematic review and meta-analysis of the efficacy and safety of adalimumab for treating rheumatoid arthritis. Rheumatol Int 2010;30:1063-70. http://dx.doi.org/10.1007/s00296-009-1111-4.
- Singh JA, Noorbaloochi S, Singh G. Golimumab for rheumatoid arthritis: a systematic review. J Rheumatol 2010;37:1096-104. http://dx.doi.org/10.3899/jrheum.091466.
- Ruiz Garcia V, Jobanputra P, Burls A, Cabello JB, Gálvez Muñoz JG, Saiz Cuenca ES, et al. Certolizumab pegol (CDP870) for rheumatoid arthritis in adults. Cochrane Database Syst Rev 2011;2. http://dx.doi.org/10.1002/14651858.CD007649.pub2.
- Adalimumab, Etanercept and Infliximab for the Treatment of Rheumatoid Arthritis. London: NICE; 2007.
- Certolizumab Pegol for the Treatment of Rheumatoid Arthritis. London: NICE; 2010.
- National Institute for Health and Care Excellence . NICE Provisionally Recommends New Drug for Rheumatoid Arthritis n.d. www.nice.org.uk/media/E4C/E5/2011081GolimumabForRAFAD.pdf (accessed April 2014).
- Singh JA, Christensen R, Wells GA, Suarez-Almazor ME, Buchbinder R, Lopez-Olivo MA, et al. A network meta-analysis of randomized controlled trials of biologics for rheumatoid arthritis: a Cochrane overview. CMAJ 2009;181:787-96. http://dx.doi.org/10.1503/cmaj.091391.
- Orme ME, Macgilchrist KS, Mitchell S, Spurden D, Bird A. Systematic review and network meta-analysis of combination and monotherapy treatments in disease-modifying antirheumatic drug-experienced patients with rheumatoid arthritis: analysis of American College of Rheumatology criteria scores 20, 50, and 70. Biologics 2012;6:429-64. http://dx.doi.org/10.2147/BTT.S36707.
- Singh JA, Wells GA, Christensen R, Tanjong Ghogomu E, Maxwell L, Macdonald JK, et al. Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev 2011;2. http://dx.doi.org/10.1002/14651858.CD008794.pub2.
- Svenson M, Geborek P, Saxne T, Bendtzen K. Monitoring patients treated with anti-TNF-alpha biopharmaceuticals: assessing serum infliximab and anti-infliximab antibodies. Rheumatology 2007;46:1828-34. http://dx.doi.org/10.1093/rheumatology/kem261.
- Strangfeld A, Hierse F, Kekow J, von Hinueber U, Tony HP, Dockhorn R, et al. Comparative effectiveness of tumour necrosis factor alpha inhibitors in combination with either methotrexate or leflunomide. Ann Rheum Dis 2009;68:1856-62. http://dx.doi.org/10.1136/ard.2008.098467.
- Smolen JS, Kay J, Doyle MK, Landewé R, Matteson EL, Wollenhaupt J, et al. Golimumab in patients with active rheumatoid arthritis after treatment with tumour necrosis factor alpha inhibitors (GO-AFTER study): a multicentre, randomised, double-blind, placebo-controlled, phase III trial. Lancet 2009;374:210-21. http://dx.doi.org/10.1016/S0140-6736(09)60506-7.
- Lloyd S, Bujkiewicz S, Wailoo AJ, Sutton AJ, Scott D. The effectiveness of anti-TNF-alpha therapies when used sequentially in rheumatoid arthritis patients: a systematic review and meta-analysis. Rheumatology 2010;49:2313-21. http://dx.doi.org/10.1093/rheumatology/keq169.
- Malottki K, Barton P, Tsourapas A, Uthman AO, Liu Z, Routh K, et al. Adalimumab, etanercept, infliximab, rituximab and abatacept for the treatment of rheumatoid arthritis after the failure of a tumour necrosis factor inhibitor: a systematic review and economic evaluation. Health Technol Assess 2011;15. http://dx.doi.org/10.3310/hta15140.
- Bergman GJ, Hochberg MC, Boers M, Wintfeld N, Kielhorn A, Jansen JP. Indirect comparison of tocilizumab and other biologic agents in patients with rheumatoid arthritis and inadequate response to disease-modifying antirheumatic drugs. Semin Arthritis Rheum 2010;39:425-41. http://dx.doi.org/10.1016/j.semarthrit.2009.12.002.
- Mertens M, Singh JA. Anakinra for rheumatoid arthritis: a systematic review. J Rheumatol 2009;36:1118-25. http://dx.doi.org/10.3899/jrheum.090074.
- Lee YH, Bae SC, Song GG. The efficacy and safety of rituximab for the treatment of active rheumatoid arthritis: a systematic review and meta-analysis of randomized controlled trials. Rheumatol Int 2011;31:1493-9. http://dx.doi.org/10.1007/s00296-010-1526-y.
- Singh JA, Beg S, Lopez-Olivo MA. Tocilizumab for rheumatoid arthritis: a Cochrane systematic review. J Rheumatol 2011;38:10-2. http://dx.doi.org/10.3899/jrheum.100717.
- Maxwell LJ, Singh JA. Abatacept for rheumatoid arthritis: a Cochrane systematic review. J Rheumatol 2010;37:234-45. http://dx.doi.org/10.3899/jrheum.091066.
- Scott DL. Role of spleen tyrosine kinase inhibitors in the management of rheumatoid arthritis. Drugs 2011;71:1121-32. http://dx.doi.org/10.2165/11591480-000000000-00000.
- Riese RJ, Krishnaswami S, Kremer J. Inhibition of JAK kinases in patients with rheumatoid arthritis: scientific rationale and clinical outcomes. Best Pract Res Clin Rheumatol 2010;24:513-26. http://dx.doi.org/10.1016/j.berh.2010.02.003.
- Traynor K. FDA approves tofacitinib for rheumatoid arthritis. Am J Health Syst Pharm 2012;69. http://dx.doi.org/10.2146/news120088.
- Wienecke T, Gøtzsche PC. Paracetamol versus non-steroidal anti-inflammatory drugs for rheumatoid arthritis. Cochrane Database Syst Rev 2004;1.
- Chen YF, Jobanputra P, Barton P, Bryan S, Fry-Smith A, Harris G, et al. Cyclooxygenase-2 selective non-steroidal anti-inflammatory drugs (etodolac, meloxicam, celecoxib, rofecoxib, etoricoxib, valdecoxib and lumiracoxib) for osteoarthritis and rheumatoid arthritis: a systematic review and economic evaluation. Health Technol Assess 2008;12.
- Haraoui B, Liu PP, Papp KA. Managing cardiovascular risk in patients with chronic inflammatory diseases. Clin Rheumatol 2012;31:585-94. http://dx.doi.org/10.1007/s10067-011-1921-0.
- Christie A, Jamtvedt G, Dahm KT, Moe RH, Haavardsholm EA, Hagen KB. Effectiveness of non-pharmacological and nonsurgical interventions for patients with rheumatoid arthritis: an overview of systematic reviews. Phys Ther 2007;87:1697-715. http://dx.doi.org/10.2522/ptj.20070039.
- Hurkmans E, van der Giesen FJ, Vliet Vlieland TP, Schoones J, Van den Ende EC. Dynamic exercise programs (aerobic capacity and/or muscle strength training) in patients with rheumatoid arthritis. Cochrane Database Syst Rev 2009;4. http://dx.doi.org/10.1002/14651858.CD006853.pub2.
- Schoels M, Knevel R, Aletaha D, Bijlsma JW, Breedveld FC, Boumpas DT, et al. Evidence for treating rheumatoid arthritis to target: results of a systematic literature search. Ann Rheum Dis 2010;69:638-43. http://dx.doi.org/10.1136/ard.2009.123976.
- Haraoui B, Smolen JS, Aletaha D, Breedveld FC, Burmester G, Codreanu C, et al. Treating Rheumatoid Arthritis to Target: multinational recommendations assessment questionnaire. Ann Rheum Dis 2011;70:1999-2002. http://dx.doi.org/10.1136/ard.2011.154179.
- de Wit MP, Smolen JS, Gossec L, van der Heijde DM. Treating rheumatoid arthritis to target: the patient version of the international recommendations. Ann Rheum Dis 2011;70:891-5. http://dx.doi.org/10.1136/ard.2010.146662.
- Schoels M, Smolen JS. Treating rheumatoid arthritis to target: evidence-based recommendations for enhanced disease management. Rheumatol Clin 2012;8:1-2.
- Sen D, Brasington R. Tight disease control in early RA. Rheum Dis Clin North Am 2012;38:327-43. http://dx.doi.org/10.1016/j.rdc.2012.04.004.
- Welsing PM, Landewé RB, van Riel PL, Boers M, van Gestel AM, van der Linden S, et al. The relationship between disease activity and radiologic progression in patients with rheumatoid arthritis: a longitudinal analysis. Arthritis Rheum 2004;50:2082-93. http://dx.doi.org/10.1002/art.20350.
- Grigor C, Capell H, Stirling A, McMahon AD, Lock P, Vallance R, et al. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet 2004;364:263-9. http://dx.doi.org/10.1016/S0140-6736(04)16676-2.
- Fransen J, Moens HB, Speyer I, van Riel PL. Effectiveness of systematic monitoring of rheumatoid arthritis disease activity in daily practice: a multicentre, cluster randomised controlled trial. Ann Rheum Dis 2005;64:1294-8. http://dx.doi.org/10.1136/ard.2004.030924.
- Verstappen SM, Jacobs JW, van der Veen MJ, Heurkens AH, Schenk Y, ter Borg EJ, et al. Intensive treatment with methotrexate in early rheumatoid arthritis: aiming for remission. Computer Assisted Management in Early Rheumatoid Arthritis (CAMERA, an open-label strategy trial). Ann Rheum Dis 2007;66:1443-9. http://dx.doi.org/10.1136/ard.2007.071092.
- Rantalaiho V, Korpela M, Laasonen L, Kautiainen H, Järvenpää S, Hannonen P, et al. Early combination disease-modifying antirheumatic drug therapy and tight disease control improve long-term radiologic outcome in patients with early rheumatoid arthritis: the 11-year results of the Finnish Rheumatoid Arthritis Combination Therapy trial. Arthritis Res Ther 2010;12. http://dx.doi.org/10.1186/ar3060.
- Ledingham J, Deighton C. Update on the British Society for Rheumatology guidelines for prescribing TNFalpha blockers in adults with rheumatoid arthritis (update of previous guidelines of April 2001). Rheumatology 2005;44:157-63. http://dx.doi.org/10.1093/rheumatology/keh464.
- Fautrel B, Flipo RM, Saraux A. Eligibility of rheumatoid arthritis patients for anti-TNF-alpha therapy according to the 2005 recommendations of the French and British Societies for Rheumatology. Rheumatology 2008;47:1698-703. http://dx.doi.org/10.1093/rheumatology/ken348.
- Smolen JS, Landewé R, Breedveld FC, Dougados M, Emery P, Gaujoux-Viala C, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs. Ann Rheum Dis 2010;69:964-75. http://dx.doi.org/10.1136/ard.2009.126532.
- Singh JA, Furst DE, Bharat A, Curtis JR, Kavanaugh AF, Kremer JM, 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. http://dx.doi.org/10.1002/acr.21641.
- Guidelines for Prescribing TNF-α Blockers in Adults with Rheumatoid Arthritis. London: British Society for Rheumatology; 2001.
- Deighton C, Hyrich K, Ding T, Ledingham J, Lunt M, Luqmani R, et al. BSR and BHPR rheumatoid arthritis guidelines on eligibility criteria for the first biological therapy. Rheumatology 2010;49:1197-9. http://dx.doi.org/10.1093/rheumatology/keq006a.
- Jönsson B, Kobelt G, Smolen J. The burden of rheumatoid arthritis and access to treatment: uptake of new therapies. Eur J Health Econ 2008;8:S61-86. http://dx.doi.org/10.1007/s10198-007-0089-7.
- Orlewska E, Ancuta I, Anic B, Codrenau C, Damjanov N, Djukic P, et al. Access to biologic treatment for rheumatoid arthritis in Central and Eastern European (CEE) countries. Med Sci Monit 2011;17:SR1-13. http://dx.doi.org/10.12659/MSM.881697.
- Hoebert JM, Mantel-Teeuwisse AK, van Dijk L, Bijlsma JW, Leufkens HG. Do rheumatoid arthritis patients have equal access to treatment with new medicines?: tumour necrosis factor-alpha inhibitors use in four European countries. Health Policy 2012;104:76-83. http://dx.doi.org/10.1016/j.healthpol.2011.10.011.
- Laires PA, Exposto F, Mesquita R, Martins AP, Cunha-Miranda L, Fonseca JE. Patients’ access to biologics in rheumatoid arthritis: a comparison between Portugal and other European countries. Eur J Health Econ 2013;14:875-85. http://dx.doi.org/10.1007/s10198-012-0432-5.
- National Institute for Health and Care Excellence . Assumptions Used in Estimating a Population Benchmark 2012. www.nice.org.uk/usingguidance/commissioningguides/biologicaltherapies/AssumptionsUsedInEstimatingAPopulationBenchmark.jsp (accessed April 2012).
- Marra CA, Bansback N, Anis AH, Shojania K. Introduction to economic modelling for clinical rheumatologists: application to biologic agents in rheumatoid arthritis. Clin Rheumatol 2011;30:S9-18. http://dx.doi.org/10.1007/s10067-010-1635-8.
- Madan J, Ades AE, Welton NJ. An overview of models used in economic analyses of biologic therapies for arthritis – from current diversity to future consensus. Rheumatology 2011;50:iv10-18. http://dx.doi.org/10.1093/rheumatology/ker240.
- Schoels M, Wong J, Scott DL, Zink A, Richards P, Landewé R, et al. Economic aspects of treatment options in rheumatoid arthritis: a systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2010;69:995-1003. http://dx.doi.org/10.1136/ard.2009.126714.
- Brennan A, Bansback N, Nixon R, Madan J, Harrison M, Watson K, et al. Modelling the cost effectiveness of TNF-alpha antagonists in the management of rheumatoid arthritis: results from the British Society for Rheumatology Biologics Registry. Rheumatology 2007;46:1345-54. http://dx.doi.org/10.1093/rheumatology/kem115.
- van der Velde G, Pham B, Machado M, Ieraci L, Witteman W, Bombardier C, et al. Cost-effectiveness of biologic response modifiers compared to disease-modifying antirheumatic drugs for rheumatoid arthritis: a systematic review. Arthritis Care Res 2011;63:65-78. http://dx.doi.org/10.1002/acr.20338.
- Hyrich KL, Watson KD, Silman AJ, Symmons DP. Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Rheumatology 2006;45:1558-65. http://dx.doi.org/10.1093/rheumatology/kel149.
- Smith N, Ding T, Butt S, Gadsby K, Deighton C. The importance of the baseline Disease Activity Score 28 in determining responders and non-responders to anti-TNF in UK clinical practice. Rheumatology 2008;47:1389-91. http://dx.doi.org/10.1093/rheumatology/ken233.
- Desroches M, Louis G, Gleizes A, Krzysiek R, Emilie D. Treatment failure with antagonists of TNF-α: mechanisms and implications for the care of patients. Eur Cytokine Netw 2010;21:226-31. http://dx.doi.org/10.1684/ecn.2010.0212.
- McGeough CM, Berrar D, Wright G, Mathews C, Gilmore P, Cunningham RT, et al. Killer immunoglobulin-like receptor and human leukocyte antigen-C genotypes in rheumatoid arthritis primary responders and non-responders to anti-TNF-α therapy. Rheumatol Int 2012;32:1647-53. http://dx.doi.org/10.1007/s00296-011-1838-6.
- Ma MH, Kingsley GH, Scott DL. A systematic comparison of combination DMARD therapy and tumour necrosis inhibitor therapy with methotrexate in patients with early rheumatoid arthritis. Rheumatology 2010;49:91-8. http://dx.doi.org/10.1093/rheumatology/kep331.
- Ma MHY, Cope AC, Scott DL. Safety of combination therapies in early RA: a systematic comparison between combination DMARDs and TNF inihibitors with methotrexate. Int J Clin Rheumatol 2010;5:547-54. http://dx.doi.org/10.2217/ijr.10.44.
- Verhoeven AC, Boers M, Tugwell P. Combination therapy in rheumatoid arthritis: updated systematic review. Br J Rheumatol 1998;37:612-19. http://dx.doi.org/10.1093/rheumatology/37.6.612.
- Chen YF, Jobanputra P, Barton P, Jowett S, Bryan S, Clark W, et al. A systematic review of the effectiveness of adalimumab, etanercept and infliximab for the treatment of rheumatoid arthritis in adults and an economic evaluation of their cost-effectiveness. Health Technol Assess 2006;10.
- Kristensen LE, Christensen R, Bliddal H, Geborek P, Danneskiold-Samsøe B, Saxne T. The number needed to treat for adalimumab, etanercept, and infliximab based on ACR50 response in three randomized controlled trials on established rheumatoid arthritis: a systematic literature review. Scand J Rheumatol 2007;36:411-17. http://dx.doi.org/10.1080/03009740701607067.
- Donahue KE, Gartlehner G, Jonas DE, Lux LJ, Thieda P, Jonas BL, et al. Systematic review: comparative effectiveness and harms of disease-modifying medications for rheumatoid arthritis. Ann Intern Med 2008;148:124-34. http://dx.doi.org/10.7326/0003-4819-148-2-200801150-00192.
- 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. http://dx.doi.org/10.1136/ard.2008.099861.
- Katchamart W, Trudeau J, Phumethum V, Bombardier C. Methotrexate monotherapy versus methotrexate combination therapy with non-biologic disease modifying anti-rheumatic drugs for rheumatoid arthritis. Cochrane Database Syst Rev 2010;4. http://dx.doi.org/10.1002/14651858.CD008495.
- Nam JL, Winthrop KL, van Vollenhoven RF, Pavelka K, Valesini G, Hensor EM, et al. Current evidence for the management of rheumatoid arthritis with biological disease-modifying antirheumatic drugs: a systematic literature review informing the EULAR recommendations for the management of RA. Ann Rheum Dis 2010;69:976-86. http://dx.doi.org/10.1136/ard.2009.126573.
- Aaltonen KJ, Virkki LM, Malmivaara A, Konttinen YT, Nordström DC, Blom M. Systematic review and meta-analysis of the efficacy and safety of existing TNF blocking agents in treatment of rheumatoid arthritis. PLoS One 2012;7. http://dx.doi.org/10.1371/journal.pone.0030275.
- Scott DL, Strand V. The effects of disease-modifying anti-rheumatic drugs on the Health Assessment Questionnaire score. Lessons from the leflunomide clinical trials database. Rheumatology 2002;41:899-90. http://dx.doi.org/10.1093/rheumatology/41.8.899.
- Strand V, Cohen S, Crawford B, Smolen JS, Scott DL. Leflunomide Investigators Groups . Patient-reported outcomes better discriminate active treatment from placebo in randomized controlled trials in rheumatoid arthritis. Rheumatology 2004;43:640-7. http://dx.doi.org/10.1093/rheumatology/keh140.
- Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, van Zeben D, Kerstens PJ, Hazes JM, et al. Clinical and radiographic outcomes of four different treatment strategies in patients with early rheumatoid arthritis (the BeSt study): a randomized, controlled trial. Arthritis Rheum 2005;52:3381-90. http://dx.doi.org/10.1002/art.21405.
- British National Formulary. London: BMA and RPS; 2007.
- British Society for Rheumatology . National Guidelines for the Monitoring of Second Line Drugs n.d. www.rheumatology.org.uk/includes/documents/cm_docs/2009/m/monitoring_second_line_drugs.pdf (accessed August 2014).
- Chakravarty K, McDonald H, Pullar T, Taggart A, Chalmers R, Oliver S, et al. BSR/BHPR guideline for disease-modifying anti-rheumatic drug (DMARD) therapy in consultation with the British Association of Dermatologists. Rheumatology 2008;47:924-5. http://dx.doi.org/10.1093/rheumatology/kel216a.
- Ding T, Ledingham J, Luqmani R, Westlake S, Hyrich K, Lunt M, et al. BSR and BHPR rheumatoid arthritis guidelines on safety of anti-TNF therapies. Rheumatology 2010;49:2217-19. http://dx.doi.org/10.1093/rheumatology/keq249a.
- Scott DL, Garrood T. Quality of life measures: use and abuse. Baillieres Best Pract Res Clin Rheumatol 2000;14:663-87. http://dx.doi.org/10.1053/berh.2000.0106.
- Bruce B, Fries JF. The Stanford Health Assessment Questionnaire: dimensions and practical applications. Health Qual Life Outcomes 2003;1. http://dx.doi.org/10.1186/1477-7525-1-20.
- Wolfe F. A reappraisal of HAQ disability in rheumatoid arthritis. Arthritis Rheum 2000;43:2751-61. http://dx.doi.org/10.1002/1529-0131(200012)43:12<2751::AID-ANR15>3.0.CO;2-6.
- The EuroQol Group . EuroQol – a new facility for the measurement of health-related quality of life. Health Policy 1990;16:199-208. http://dx.doi.org/10.1016/0168-8510(90)90421-9.
- Ware JE. SF-36 health survey update. Spine 2000;25:3130-9. http://dx.doi.org/10.1097/00007632-200012150-00008.
- Khanna D, Tsevat J. Health-related quality of life – an introduction. Am J Manag Care 2007;13:S218-23.
- Linde L, Sørensen J, Ostergaard M, Hørslev-Petersen K, Hetland ML. Health-related quality of life: validity, reliability, and responsiveness of SF-36, 15D, EQ-5D, RAQoL, and HAQ in patients with rheumatoid arthritis. J Rheumatol 2008;35:1528-37.
- Wolfe F, Michaud K, Li T, Katz RS. EQ-5D and SF-36 quality of life measures in systemic lupus erythematosus: comparisons with rheumatoid arthritis, noninflammatory rheumatic disorders, and fibromyalgia. J Rheumatol 2010;37:296-304. http://dx.doi.org/10.3899/jrheum.090778.
- Oude Voshaar MA, ten Klooster PM, Taal E, van de Laar MA. Measurement properties of physical function scales validated for use in patients with rheumatoid arthritis: a systematic review of the literature. Health Qual Life Outcomes 2011;9. http://dx.doi.org/10.1186/1477-7525-9-99.
- Scott DL, Smith C, Kingsley G. Joint damage and disability in rheumatoid arthritis: an updated systematic review. Clin Exp Rheumatol 2003;21:S20-7.
- Larsens A, Dale K, Eek M. Radiographic evaluation of rheumatoid arthritis and related conditions by standard reference films. Acta Radiol Diagn 1977;8:481-91.
- Scott DL, Houssien DA, Laassonen L. Proposed modification to Larsen’s scoring methods for hand and wrist radiographs. Br J Rheumatol 1995;34. http://dx.doi.org/10.1093/rheumatology/34.1.56.
- Prevoo ML, van‘T Hof M, Kuper HH, Van Leeuwen MA, Van de Putte LB, Van Riel PL. Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with RA. Arthritis Rheum 1995;38:44-8. http://dx.doi.org/10.1002/art.1780380107.
- Lassere MN, Johnson KR, Boers M, Carlton K, Day RO, de Wit M, et al. Standardized assessment of adverse events in rheumatology clinical trials: summary of the OMERACT 7 drug safety module update. J Rheumatol 2005;32:2037-41.
- European Medicines Agency . Guideline on Clinical Investigation of Medicinal Products Other Than NSAIDs for Treatment of Rheumatoid Arthritis 2011. www.ema.europa.eu/ema/index.jsp (accessed April 2014).
- US Food and Drug Administration . Guidance for Industry. Rheumatoid Arthritis: Developing Drug Products for Treatment 2013. www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm (accessed April 2014).
- Beecham, Knapp M., Thornicroft G, Brewin C, Wing JK. Measuring Mental Health Needs. London: Gaskell; 1992.
- Hurley MV, Walsh NE, Mitchell H, Nicholas J, Patel A. Long-term outcomes and costs of an integrated rehabilitation program for chronic knee pain: a pragmatic, cluster randomized, controlled trial. Arthritis Care Res 2012;64:238-47. http://dx.doi.org/10.1002/acr.20642.
- Lipsky PE, van der Heijde D, St Clair E, Furst DE, Breedveld FC, Kalden JR, et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med 2000;343:1594-602. http://dx.doi.org/10.1056/NEJM200011303432202.
- Choy EH, Smith CM, Farewell V, Walker D, Hassell A, Chau L, et al. Factorial randomised controlled trial of glucocorticoids and combination disease modifying drugs in early rheumatoid arthritis. Ann Rheum Dis 2008;67:656-63. http://dx.doi.org/10.1136/ard.2007.076299.
- Wells GA, Tugwell P, Kraag GR, Baker PR, Groh J, Redelmeier DA. Minimum important difference between patients with rheumatoid arthritis: the patient’s perspective. J Rheumatol 1993;20:557-60.
- Redelmeier DA, Lorig K. Assessing the clinical importance of symptomatic improvements. An illustration in rheumatology. Arch Intern Med 1993;153:1337-42. http://dx.doi.org/10.1001/archinte.1993.00410110045008.
- Kosinski M, Zhao SZ, Dedhiya S, Osterhaus JT, Ware JE. Determining minimally important changes in generic and disease-specific health-related quality of life questionnaires in clinical trials of rheumatoid arthritis. Arthritis Rheum 2000;43:1478-87. http://dx.doi.org/10.1002/1529-0131(200007)43:7<1478::AID-ANR10>3.0.CO;2-M.
- Strand V, Tugwell P, Bombardier C, Maetzel A, Bombardier C, Crawford B, et al. Function and health-related quality of life: results from a randomized controlled trial of leflunomide versus methotrexate or placebo in patients with active rheumatoid arthritis. Leflunomide Rheumatoid Arthritis Investigators Group. Arthritis Rheum 1999;42:1870-8. http://dx.doi.org/10.1002/1529-0131(199909)42:9<1870::AID-ANR11>3.0.CO;2-D.
- White IR, Royston P, Wood AM. Multiple imputation using chained equations: issues and guidance for practice. Stat Med 2011;30:377-99. http://dx.doi.org/10.1002/sim.4067.
- Rubin DB. Inference and missing data. Biometrika 1976;63:581-90. http://dx.doi.org/10.1093/biomet/63.3.581.
- Little RJA, Rubin DB. Statistical Analysis with Missing Data. Hoboken, NJ: John Wiley; 2002.
- Schafer JL. Analysis of Incomplete Multivariate Data. London: Chapman and Hall; 1997.
- R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2012.
- Diggle P, Farewell D, Henderson R. Analysis of longitudinal data with drop-out: objectives, assumptions and a proposal. J R Stat Soc C Appl Stat 2007;56:499-550. http://dx.doi.org/10.1111/j.1467-9876.2007.00590.x.
- Brazier J, Roberts J, Deverill M. The estimation of a preference-based measure of health from the SF-36. J Health Econ 2002;21:271-92. http://dx.doi.org/10.1016/S0167-6296(01)00130-8.
- Dolan P, Gudex C, Kind P, Williams A. A Social Tariff for EuroQol: Results from a UK Population Survey. York: University of York; 1995.
- Manca A, Hawkins N, Schulpher MJ. Estimating mean QALYs in trial-based cost-effectiveness analysis: the importance of controlling for baseline utility. Health Econ 2005;14:487-96. http://dx.doi.org/10.1002/hec.944.
- Assmann SF, Pocock SJ, Enos LE, Kasten LE. Subgroup analysis and other (mis)uses of baseline data in clinical trials. Lancet 2000;355:1064-9. http://dx.doi.org/10.1016/S0140-6736(00)02039-0.
- Briggs AH. A Bayesian approach to stochastic cost-effectiveness analysis. Health Econ 1999;8:257-61. http://dx.doi.org/10.1002/(SICI)1099-1050(199905)8:3<257::AID-HEC427>3.0.CO;2-E.
- Moher D, Liberati A, Tetzlaff J, Altman DG. The PRISMA group . Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the PRISMA statement. PLoS Med 2009;6. http://dx.doi.org/10.1371/journal.pmed.1000097.
- Viswanathan M, Ansari MT, Berkman ND, Chang S, Hartling L, McPheeters LM, et al. Methods Guide for Comparative Effectiveness Review. Rockville, MD: Agency for Healthcare Research and Quality; 2012.
- DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials 1986;7:177-88. http://dx.doi.org/10.1016/0197-2456(86)90046-2.
- Hardy RJ, Thompson SG. Detecting and describing heterogeneity in meta-analysis. Stat Med 1998;17:841-56. http://dx.doi.org/10.1002/(SICI)1097-0258(19980430)17:8<841::AID-SIM781>3.0.CO;2-D.
- Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327:557-60. http://dx.doi.org/10.1136/bmj.327.7414.557.
- Haagsma CJ, van Riel PL, de Jong AJ, van de Putte LB. Combination of sulphasalazine and methotrexate versus the single components in early rheumatoid arthritis: a randomized, controlled, double-blind, 52 week clinical trial. Br J Rheumatol 1997;36:1082-8. http://dx.doi.org/10.1093/rheumatology/36.10.1082.
- van den Borne BE, Landewé RB, Goei The HS, Rietveld JH, Zwinderman AH, Bruyn GA, et al. Combination therapy in recent onset rheumatoid arthritis: a randomized double blind trial of the addition of low dose cyclosporine to patients treated with low dose chloroquine. J Rheumatol 1998;25:1493-8.
- Dougados M, Combe B, Cantagrel A, Goupille P, Olive P, Schattenkirchner M, et al. Combination therapy in early rheumatoid arthritis: a randomised, controlled, double blind 52 week clinical trial of sulphasalazine and methotrexate compared with the single components. Ann Rheum Dis 1999;58:220-5. http://dx.doi.org/10.1136/ard.58.4.220.
- Mottonen T, Hannonen P, Leirisalo-Repo M, Nissilä M, Kautiainen H, Korpela M, et al. Comparison of combination therapy with single-drug therapy in early rheumatoid arthritis: a randomised trial. FIN-RACo trial group. Lancet 1999;353:1568-73. http://dx.doi.org/10.1016/S0140-6736(98)08513-4.
- Proudman SM, Conaghan PG, Richardson C, Griffiths B, Green MJ, McGonagle D, et al. Treatment of poor-prognosis early rheumatoid arthritis. A randomized study of treatment with methotrexate, cyclosporin A, and intraarticular corticosteroids compared with sulfasalazine alone. Arthritis Rheum 2000;43:1809-19. http://dx.doi.org/10.1002/1529-0131(200008)43:8<1809::AID-ANR17>3.0.CO;2-D.
- Ferraccioli GF, Gremese E, Tomietto P, Favret G, Damato R, Di Poi E. Analysis of improvements, full responses, remission and toxicity in rheumatoid patients treated with step-up combination therapy (methotrexate, cyclosporin A, sulphasalazine) or monotherapy for three years. Rheumatology 2002;41:892-8. http://dx.doi.org/10.1093/rheumatology/41.8.892.
- Gerards AH, Landewe RB, Prins AP, Bruyn GA, Goei Thé HS, Laan RF, et al. Cyclosporin A monotherapy versus cyclosporin A and methotrexate combination therapy in patients with early rheumatoid arthritis. Ann Rheum Dis 2003;62:291-6. http://dx.doi.org/10.1136/ard.62.4.291.
- Marchesoni A, Battafarano N, Arreghini M, Panni B, Gallazzi M, Tosi S. Radiographic progression in early rheumatoid arthritis: a 12-month randomized controlled study comparing the combination of cyclosporin and methotrexate with methotrexate alone. Rheumatology 2003;42:1545-9. http://dx.doi.org/10.1093/rheumatology/keg394.
- Capell HA, Madhok R, Hunter JA, Porter D, Morrison E, Larkin J, et al. Lack of radiological and clinical benefit over two years of low dose prednisolone for rheumatoid arthritis: results of a randomised controlled trial. Ann Rheum Dis 2004;63:797-803. http://dx.doi.org/10.1136/ard.2003.014050.
- Miranda JM, Alvarez-Nemegyei J, Saavedra MA, Terán L, Galván-Villegas F, García-Figueroa J, et al. A randomized, double-blind, multicenter, controlled clinical trial of cyclosporine plus chloroquine vs. cyclosporine plus placebo in early-onset rheumatoid arthritis. Arch Med Res 2004;35:36-42. http://dx.doi.org/10.1016/j.arcmed.2003.07.008.
- Ichikawa Y, Saito T, Yamanaka H, Akizuki M, Kondo H, Kobayashi S, et al. Therapeutic effects of the combination of methotrexate and bucillamine in early rheumatoid arthritis: a multicenter, double-blind, randomized controlled study. Mod Rheumatol 2005;15:323-8. http://dx.doi.org/10.3109/s10165-005-0420-z.
- Sarzi-Puttini P, D’Ingianna E, Fumagalli M, Scarpellini M, Fiorini T, Chérié-Lignière EL, et al. An open, randomized comparison study of cyclosporine A, cyclosporine A + methotrexate and cyclosporine A + hydroxychloroquine in the treatment of early severe rheumatoid arthritis. Rheumatol Int 2005;25:15-22. http://dx.doi.org/10.1007/s00296-003-0384-2.
- Svensson B, Boonen A, Albertsson K, van der Heijde D, Keller C, Hafström I. Low-dose prednisolone in addition to the initial disease-modifying antirheumatic drug in patients with early active rheumatoid arthritis reduces joint destruction and increases the remission rate: a two-year randomized trial. Arthritis Rheum 2005;52:3360-70. http://dx.doi.org/10.1002/art.21298.
- Wassenberg S, Rau R, Steinfeld P, Zeidler H. Very low-dose prednisolone in early rheumatoid arthritis retards radiographic progression over two years: a multicenter, double-blind, placebo-controlled trial. Arthritis Rheum 2005;52:3371-80. http://dx.doi.org/10.1002/art.21421.
- Hetland ML, Stengaard-Pedersen K, Junker P, Lottenburger T, Ellingsen T, Andersen LS, et al. Combination treatment with methotrexate, cyclosporine, and intraarticular betamethasone compared with methotrexate and intraarticular betamethasone in early active rheumatoid arthritis: an investigator-initiated, multicenter, randomized, double-blind, parallel-group, placebo-controlled study. Arthritis Rheum 2006;54:1401-9. http://dx.doi.org/10.1002/art.21796.
- O’Dell JR, Elliott JR, Mallek JA, Mikuls TR, Weaver CA, Glickstein S, et al. Treatment of early seropositive rheumatoid arthritis: doxycycline plus methotrexate versus methotrexate alone. Arthritis Rheum 2006;54:621-7. http://dx.doi.org/10.1002/art.21620.
- Breedveld FC, Emery P, Keystone E, Patel K, Furst DE, Kalden JR, et al. Infliximab in active early rheumatoid arthritis. Ann Rheum Dis 2004;63:149-55. http://dx.doi.org/10.1136/ard.2003.013961.
- St Clair EW, van der Heijde DM, Smolen JS, Maini RN, Bathon JM, Emery P, et al. Combination of infliximab and methotrexate therapy for early rheumatoid arthritis: a randomized, controlled trial. Arthritis Rheum 2004;50:3432-43. http://dx.doi.org/10.1002/art.20568.
- Taylor PC, Steuer A, Gruber J, Cosgrove DO, Blomley MJ, Marsters PA, et al. Comparison of ultrasonographic assessment of synovitis and joint vascularity with radiographic evaluation in a randomized, placebo-controlled study of infliximab therapy in early rheumatoid arthritis. Arthritis Rheum 2004;50:1107-16. http://dx.doi.org/10.1002/art.20123.
- Quinn MA, Conaghan PG, O’Connor PJ, Karim Z, Greenstein A, Brown A, et al. Very early treatment with infliximab in addition to methotrexate in early, poor-prognosis rheumatoid arthritis reduces magnetic resonance imaging evidence of synovitis and damage, with sustained benefit after infliximab withdrawal: results from a twelve-month randomized, double-blind, placebo-controlled trial. Arthritis Rheum 2005;52:27-35. http://dx.doi.org/10.1002/art.20712.
- Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka K, van Vollenhoven R, et al. The PREMIER study: a multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum 2006;54:26-37. http://dx.doi.org/10.1002/art.21519.
- Durez P, Malghem J, Nzeusseu Toukap A, Depresseux G, Lauwerys BR, Westhovens R, et al. Treatment of early rheumatoid arthritis: a randomized magnetic resonance imaging study comparing the effects of methotrexate alone, methotrexate in combination with infliximab, and methotrexate in combination with intravenous pulse methylprednisolone. Arthritis Rheum 2007;56:3919-27. http://dx.doi.org/10.1002/art.23055.
- Emery P, Breedveld FC, Hall S, Durez P, Chang DJ, Robertson D, et al. Comparison of methotrexate monotherapy with a combination of methotrexate and etanercept in active, early, moderate to severe rheumatoid arthritis (COMET): a randomised, double-blind, parallel treatment trial. Lancet 2008;372:375-82. http://dx.doi.org/10.1016/S0140-6736(08)61000-4.
- Soubrier M, Puechal X, Sibilia J, Mariette X, Meyer O, Combe B, et al. Evaluation of two strategies (initial methotrexate monotherapy vs its combination with adalimumab) in management of early active rheumatoid arthritis: data from the GUEPARD trial. Rheumatology 2009;48:1429-34. http://dx.doi.org/10.1093/rheumatology/kep261.
- Detert J, Bastian H, Listing J, Weiß A, Wassenberg S, Liebhaber A, et al. Induction therapy with adalimumab plus methotrexate for 24 weeks followed by methotrexate monotherapy up to week 48 versus methotrexate therapy alone for DMARD-naive patients with early rheumatoid arthritis: HIT HARD, an investigator-initiated study. Ann Rheum Dis 2013;72:844-50. http://dx.doi.org/10.1136/annrheumdis-2012-201612.
- Kavanaugh A, Fleischmann RM, Emery P, Kupper H, Redden L, Guerette B, et al. Clinical, functional and radiographic consequences of achieving stable low disease activity and remission with adalimumab plus methotrexate or methotrexate alone in early rheumatoid arthritis: 26-week results from the randomised, controlled OPTIMA study. Ann Rheum Dis 2013;72:64-71. http://dx.doi.org/10.1136/annrheumdis-2011-201247.
- van Vollenhoven RF, Ernestam S, Geborek P, Petersson IF, Cöster L, Waltbrand E, et al. Addition of infliximab compared with addition of sulfasalazine and hydroxychloroquine to methotrexate in patients with early rheumatoid arthritis (Swefot trial): 1-year results of a randomised trial. Lancet 2009;374:459-66. http://dx.doi.org/10.1016/S0140-6736(09)60944-2.
- Moreland LW, O’Dell JR, Paulus HE, Curtis JR, Bathon JM, St Clair EW, et al. TEAR Investigators. A randomized comparative effectiveness study of oral triple therapy versus etanercept plus methotrexate in early aggressive rheumatoid arthritis: the treatment of Early Aggressive Rheumatoid Arthritis Trial. Arthritis Rheum 2012;64:2824-35. http://dx.doi.org/10.1002/art.34498.
- Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, van Zeben D, Kerstens PJ, Hazes JM, et al. Comparison of treatment strategies in early rheumatoid arthritis: a randomized trial. Ann Intern Med 2007;146:406-15. http://dx.doi.org/10.7326/0003-4819-146-6-200703200-00005.
- van Vollenhoven RF, Geborek P, Forslind K, Albertsson K, Ernestam S, Petersson IF, et al. Swefot study group. Conventional combination treatment versus biological treatment in methotrexate-refractory early rheumatoid arthritis: 2 year follow-up of the randomised, non-blinded, parallel-group Swefot trial. Lancet 2012;379:1712-20. http://dx.doi.org/10.1016/S0140-6736(12)60027-0.
- Ferraz MB, Pinheiro GR, Helfenstein M, Albuquerque E, Rezende C, Roimicher L, et al. Combination therapy with methotrexate and chloroquine in rheumatoid arthritis. A multicenter randomized placebo-controlled trial. Scand J Rheumatol 1994;23:231-6. http://dx.doi.org/10.3109/03009749409103721.
- Willkens RF, Sharp JT, Stablein D, Marks C, Wortmann R. Comparison of azathioprine, methotrexate, and the combination of the two in the treatment of rheumatoid arthritis. A forty-eight-week controlled clinical trial with radiologic outcome assessment. Arthritis Rheum 1995;38:1799-806. http://dx.doi.org/10.1002/art.1780381213.
- Bendix G, Bjelle A. Adding low-dose cyclosporin A to parenteral gold therapy in rheumatoid arthritis: a double-blind placebo-controlled study. Br J Rheumatol 1996;35:1142-9. http://dx.doi.org/10.1093/rheumatology/35.11.1142.
- Kremer JM, Genovese MC, Cannon GW, Caldwell JR, Cush JJ, Furst DE, et al. Concomitant leflunomide therapy in patients with active rheumatoid arthritis despite stable doses of methotrexate. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 2002;137:726-33. http://dx.doi.org/10.7326/0003-4819-137-9-200211050-00007.
- Dougados M, Emery P, Lemmel EM, Zerbini CA, Brin S, van Riel P. When a DMARD fails, should patients switch to sulfasalazine or add sulfasalazine to continuing leflunomide?. Ann Rheum Dis 2005;64:44-51. http://dx.doi.org/10.1136/ard.2003.016709.
- Lehman AJ, Esdaile JM, Klinkhoff AV, Grant E, Fitzgerald A, Canvin J. METGO study group . A 48-week, randomized, double-blind, double-observer, placebo-controlled multicenter trial of combination methotrexate and intramuscular gold therapy in rheumatoid arthritis: results of the METGO study. Arthritis Rheum 2005;52:1360-70. http://dx.doi.org/10.1002/art.21018.
- Karanikolas G, Charalambopoulos D, Andrianakos A, Antoniades C, Katsilambros N. Combination of cyclosporine and leflunomide versus single therapy in severe rheumatoid arthritis. J Rheumatol 2006;33:486-9.
- Capell HA, Madhok R, Porter DR, Munro RA, McInnes IB, Hunter JA, et al. Combination therapy with sulfasalazine and methotrexate is more effective than either drug alone in patients with rheumatoid arthritis with a suboptimal response to sulfasalazine: results from the double-blind placebo-controlled MASCOT study. Ann Rheum Dis 2007;66:235-41. http://dx.doi.org/10.1136/ard.2006.057133.
- Weinblatt ME, Kremer JM, Bankhurst AD, Bulpitt KJ, Fleischmann RM, Fox RI, et al. A trial of etanercept, a recombinant tumor necrosis factor receptor: Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate. N Engl J Med 1999;340:253-9. http://dx.doi.org/10.1056/NEJM199901283400401.
- Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA, et al. Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum 2003;48:35-4. http://dx.doi.org/10.1002/art.10697.
- Klareskog L, van der Heijde D, de Jager JP, Gough A, Kalden J, Malaise M, et al. Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet 2004;363:675-81. http://dx.doi.org/10.1016/S0140-6736(04)15640-7.
- Kay J, Matteson EL, Dasgupta B, Nash P, Durez P, Hall S, et al. Golimumab in patients with active rheumatoid arthritis despite treatment with methotrexate: a randomized, double-blind, placebo-controlled, dose-ranging study. Arthritis Rheum 2008;58:964-75. http://dx.doi.org/10.1002/art.23383.
- Keystone E, Heijde DV, Mason D, Landewé R, Vollenhoven RV, Combe B, et al. Certolizumab pegol plus methotrexate is significantly more effective than placebo plus methotrexate in active rheumatoid arthritis: findings of a fifty-two-week, phase III, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Arthritis Rheum 2008;58:3319-29. http://dx.doi.org/10.1002/art.23964.
- Smolen J, Landewé RB, Mease P, Brzezicki J, Mason D, Luijtens K, et al. Efficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the RAPID 2 study. A randomised controlled trial. Ann Rheum Dis 2009;68:797-804. http://dx.doi.org/10.1136/ard.2008.101659.
- Kremer J, Ritchlin C, Mendelsohn A, Baker D, Kim L, Xu Z, et al. Golimumab, a new human anti-tumor necrosis factor alpha antibody, administered intravenously in patients with active rheumatoid arthritis: forty-eight-week efficacy and safety results of a phase III randomized, double-blind, placebo-controlled study. Arthritis Rheum 2010;62:917-28. http://dx.doi.org/10.1002/art.27348.
- O’Dell JR, Mikuls TR, Taylor TH, Ahluwalia V, Brophy M, Warren SR, et al. Therapies for active rheumatoid arthritis after methotrexate failure. N Engl J Med 2013;369:307-18. http://dx.doi.org/10.1056/NEJMoa1303006.
- van der Heijde D, Klareskog L, Rodriguez-Valverde V, Codreanu C, Bolosiu H, Melo-Gomes J, et al. Comparison of etanercept and methotrexate, alone and combined, in the treatment of rheumatoid arthritis: two-year clinical and radiographic results from the TEMPO study, a double-blind, randomized trial. Arthritis Rheum 2006;54:1063-74. http://dx.doi.org/10.1002/art.21655.
- Keystone EC, Combe B, Smolen J, Strand V, Goel N, van Vollenhoven R, et al. Sustained efficacy of certolizumab pegol added to methotrexate in the treatment of rheumatoid arthritis: 2-year results from the RAPID 1 trial. Rheumatology 2012;51:1628-38. http://dx.doi.org/10.1093/rheumatology/kes082.
- Marcus SM. Assessing non-consent bias with parallel randomized and nonrandomized clinical trials. J Clin Epidemiol 1997;50:823-8. http://dx.doi.org/10.1016/S0895-4356(97)00068-1.
- Berger VW, Vali B. Intent-to-randomize corrections for missing data resulting from run-in selection bias in clinical trials for chronic conditions. J Biopharm Stat 2011;21:263-70. http://dx.doi.org/10.1080/10543406.2011.550107.
- Choy EH, Kingsley GH, Khoshaba B, Pipitone N, Scott DL. Intramuscular Methylprednisolone study group . A two year randomised controlled trial of intramuscular depot steroids in patients with established rheumatoid arthritis who have shown an incomplete response to disease modifying antirheumatic drugs. Ann Rheum Dis 2005;64:1288-93. http://dx.doi.org/10.1136/ard.2004.030908.
- Aletaha D, Smolen J, Ward MM. Measuring function in rheumatoid arthritis: identifying reversible and irreversible components. Arthritis Rheum 2006;54:2784-92. http://dx.doi.org/10.1002/art.22052.
- Aletaha D, Strand V, Smolen JS, Ward MM. Treatment-related improvement in physical function varies with duration of rheumatoid arthritis: a pooled analysis of clinical trial results. Ann Rheum Dis 2008;67:238-43. http://dx.doi.org/10.1136/ard.2007.071415.
- Farewell VT, Su L. A multistate model for events defined by prolonged observation. Biostatistics 2011;12:102-11. http://dx.doi.org/10.1093/biostatistics/kxq041.
- Tennant A, Hillman M, Fear J, Pickering A, Chamberlain MA. Are we making the most of the Stanford Health Assessment Questionnaire?. Br J Rheumatol 1996;35:574-8. http://dx.doi.org/10.1093/rheumatology/35.6.574.
- Wolfe F. Which HAQ is best? A comparison of the HAQ, MHAQ and RA-HAQ, a difficult 8 item HAQ (DHAQ), and a rescored 20 item HAQ (HAQ20): analyses in 2,491 rheumatoid arthritis patients following leflunomide initiation. J Rheumatol 2001;28:982-9.
- Scott DL, Garrood T. Quality of life measures: use and abuse. Baillieres Best Pract Res Clin Rheumatol 2000;14:663-87. http://dx.doi.org/10.1053/berh.2000.0106.
- Pollard L, Choy EH, Scott DL. The consequences of rheumatoid arthritis: quality of life measures in the individual patient. Clin Exp Rheumatol 2005;23:S43-52.
- Pope JE, Khanna D, Norrie D, Ouimet JM. The minimally important difference for the Health Assessment Questionnaire in rheumatoid arthritis clinical practice is smaller than in randomized controlled trials. J Rheumatol 2009;36:254-9. http://dx.doi.org/10.3899/jrheum.080479.
- Scott DL, Grindulis KA, Struthers GR, Coulton BL, Popert AJ, Bacon PA. Progression of radiological changes in rheumatoid arthritis. Ann Rheum Dis 1984;43:8-17. http://dx.doi.org/10.1136/ard.43.1.8.
- Scott DL, Greenwood A, Bryans R, Huskisson EC. Progressive joint damage during penicillamine therapy for rheumatoid arthritis. Rheumatol Int 1988;8:135-9. http://dx.doi.org/10.1007/BF00272436.
- Scott DL, Greenwood A, Davies J, Maddison PJ, Maddison MC, Hall ND. Radiological progression in rheumatoid arthritis: do D-penicillamine and hydroxychloroquine have different effects?. Br J Rheumatol 1990;29:126-7. http://dx.doi.org/10.1093/rheumatology/29.2.126.
- Scott DL, Hanly JG, Moriarty M, Bresnihan B. The effect of lymphoid irradiation on the progression of joint damage in intractable rheumatoid arthritis. Arthritis Rheum 1988;31:147-8. http://dx.doi.org/10.1002/art.1780310124.
- Soden M, Hassan J, Scott DL, Hanly JG, Moriarty M, Whelan A, et al. Lymphoid irradiation in intractable rheumatoid arthritis. Long-term follow-up of patients treated with 750 rads or 2000 rads. Arthritis Rheum 1989;32:523-30. http://dx.doi.org/10.1002/anr.1780320503.
- Grindulis KA, Scott DL, Struthers GR. The assessment of radiological changes in the hands and wrists in rheumatoid arthritis. Rheumatol Int 1983;3:39-42. http://dx.doi.org/10.1007/BF00541231.
- Scott DL, Coulton BL, Bacon PA, Popert AJ. Methods of X-ray assessment in rheumatoid arthritis: a re-evaluation. Br J Rheumatol 1985;24:31-9. http://dx.doi.org/10.1093/rheumatology/24.1.31.
- Edmonds J, Saudan A, Lassere M, Scott D. Introduction to reading radiographs by the Scott modification of the Larsen method. J Rheumatol 1999;26:740-2.
- Lassere M, Houssein D, Scott D, Edmonds J. Reference curves of radiographic damage in patients with rheumatoid arthritis: application of quantile regression and fractional polynomials. J Rheumatol 1997;24:1288-94.
- Scott DL, Laasonen L, Priolo F, Houssien DA, Bacarini L, Cerase A, et al. The radiological assessment of rheumatoid arthritis. Clin Exp Rheumatol 1997;15:S53-61.
- Symmons D, Tricker K, Roberts C, Davies L, Dawes P, Scott DL. The British Rheumatoid Outcome Study Group (BROSG) randomised controlled trial to compare the effectiveness and cost-effectiveness of aggressive versus symptomatic therapy in established rheumatoid arthritis. Health Technol Assess 2005;9.
- van Riel PLCM, Scott DL. EULAR Handbook of Clinical Assessment in Rheumatoid Arthritis. Alphen Aan Den Rijn, the Netherlands: Van Zuiden Communications; 2000.
- Scott DL, Antoni C, Choy EH, Van Riel PC. Joint counts in routine practice. Rheumatology 2003;42:919-23. http://dx.doi.org/10.1093/rheumatology/keg235.
- Scott DL, Choy EH, Greeves A, Isenberg D, Kassinor D, Rankin E, et al. Standardising joint assessment in rheumatoid arthritis. Clin Rheumatol 1996;15:579-82. http://dx.doi.org/10.1007/BF02238547.
- Stamp LK, Harrison A, Frampton C, Corkill MM. Does a joint count calibration exercise make a difference? Implications for clinical trials and training. J Rheumatol 2012;39:877-8. http://dx.doi.org/10.3899/jrheum.111224.
- Hyrich KL, Watson KD, Lunt M, Symmons DP. Changes in disease characteristics and response rates among patients in the United Kingdom starting anti-tumor necrosis factor therapy for rheumatoid arthritis between 2001 and 2008. Rheumatology 2011;50:117-23. http://dx.doi.org/10.1093/rheumatology/keq209.
- Greenberg JD, Reed G, Decktor D, Harrold L, Furst D, Gibofsky A, et al. A comparative effectiveness study of adalimumab, etanercept and infliximab in biologically naive and switched rheumatoid arthritis patients: results from the US CORRONA registry. Ann Rheum Dis 2012;71:1134-42. http://dx.doi.org/10.1136/annrheumdis-2011-150573.
- Hetland ML, Christensen IJ, Tarp U, Dreyer L, Hansen A, Hansen IT, et al. Direct comparison of treatment responses, remission rates, and drug adherence in patients with rheumatoid arthritis treated with adalimumab, etanercept, or infliximab: results from eight years of surveillance of clinical practice in the nationwide Danish DANBIO registry. Arthritis Rheum 2010;62:22-3. http://dx.doi.org/10.1002/art.27227.
- de Punder YM, Fransen J, Kievit W, Houtman PM, Visser H, van de Laar MA, et al. The prevalence of clinical remission in RA patients treated with anti-TNF: results from the Dutch Rheumatoid Arthritis Monitoring (DREAM) registry. Rheumatology 2012;51:1610-17. http://dx.doi.org/10.1093/rheumatology/kes078.
- Mancarella L, Bobbio-Pallavicini F, Ceccarelli F, Falappone PC, Ferrante A, Malesci D, et al. Good clinical response, remission, and predictors of remission in rheumatoid arthritis patients treated with tumor necrosis factor-alpha blockers: the GISEA study. J Rheumatol 2007;34:1670-3.
- Listing J, Strangfeld A, Rau R, Kekow J, Gromnica-Ihle E, Klopsch T, et al. Clinical and functional remission: even though biologics are superior to conventional DMARDs overall success rates remain low – results from RABBIT, the German biologics register. Arthritis Res Ther 2006;8. http://dx.doi.org/10.1186/ar1933.
- Barton P, Bryan S, Robinson S. Modelling in the economic evaluation of health care: selecting the appropriate approach. J Health Serv Res Policy 2004;9:110-18. http://dx.doi.org/10.1258/135581904322987535.
- Bathon JM, McMahon DJ. Making rational treatment decisions in rheumatoid arthritis when methotrexate fails. N Engl J Med 2013;369:384-5. http://dx.doi.org/10.1056/NEJMe1306381.
- Fautrel B. Economic benefits of optimizing anchor therapy for rheumatoid arthritis. Rheumatology 2012;51:iv21-6. http://dx.doi.org/10.1093/rheumatology/kes088.
- Kobelt G, Jönsson L, Lindgren P, Young A, Eberhardt K. Modeling the progression of rheumatoid arthritis: a two-country model to estimate costs and consequences of rheumatoid arthritis. Arthritis Rheum 2002;46:2310-19. http://dx.doi.org/10.1002/art.10471.
- Korthals-de Bos I, Van Tulder M, Boers M, Verhoeven AC, Adèr HJ, Bibo J, et al. Indirect and total costs of early rheumatoid arthritis: a randomized comparison of combined step-down prednisolone, methotrexate, and sulfasalazine with sulfasalazine alone. J Rheumatol 2004;31:1709-16.
- Verhoeven AC, Bibo JC, Boers M, Engel GL, van der Linden S. Cost-effectiveness and cost–utility of combination therapy in early rheumatoid arthritis: randomized comparison of combined step-down prednisolone, methotrexate and sulphasalazine with sulphasalazine alone. Br J Rheumatol 1998;37:1102-9. http://dx.doi.org/10.1093/rheumatology/37.10.1102.
- Bakker MF, Jacobs JW, Welsing PM, Verstappen SM, Tekstra J, Ton E, et al. Low-dose prednisone inclusion in a methotrexate-based, tight control strategy for early rheumatoid arthritis: a randomized trial. Ann Intern Med 2012;156:329-39. http://dx.doi.org/10.7326/0003-4819-156-5-201203060-00004.
- Choi HK, Seeger JD, Kuntz KM. A cost-effectiveness analysis of treatment options for patients with methotrexate-resistant rheumatoid arthritis. Arthritis Rheum 2000;43:2316-27. http://dx.doi.org/10.1002/1529-0131(200010)43:10<2316::AID-ANR20>3.0.CO;2-6.
- Tosh JC, Wailoo AJ, Scott DL, Deighton CM. Cost-effectiveness of combination nonbiologic disease-modifying antirheumatic drug strategies in patients with early rheumatoid arthritis. J Rheumatol 2011;38:1593-600. http://dx.doi.org/10.3899/jrheum.101327.
- Osiri M, Shea B, Robinson V, Suarez-Almazor M, Strand V, Tugwell P, et al. Leflunomide for treating rheumatoid arthritis. Cochrane Database Syst Rev 2003;1.
- Bingham SJ, Buch MH, Tennant A, Emery P. The impact of escalating conventional therapy in rheumatoid arthritis patients referred for anti-tumour necrosis factor-alpha therapy. Rheumatology 2004;43:364-8. http://dx.doi.org/10.1093/rheumatology/keh057.
- Strand V, Sokolove J. Randomized controlled trial design in rheumatoid arthritis: the past decade. Arthritis Res Ther 2009;11. http://dx.doi.org/10.1186/ar2555.
- Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. N Engl J Med 2000;342:1887-92. http://dx.doi.org/10.1056/NEJM200006223422507.
- Kievit W, Fransen J, Oerlemans AJ, Kuper HH, van der Laar MA, de Rooij DJ, et al. The efficacy of anti-TNF in rheumatoid arthritis, a comparison between randomised controlled trials and clinical practice. Ann Rheum Dis 2007;66:1473-8. http://dx.doi.org/10.1136/ard.2007.072447.
- Fries JF, Krishnan E. Equipoise, design bias, and randomized controlled trials: the elusive ethics of new drug development. Arthritis Res Ther 2004;6:250-5. http://dx.doi.org/10.1186/ar1170.
- Lilford RJ. Ethics of clinical trials from a Bayesian and decision analytic perspective: whose equipoise is it anyway?. BMJ 2003;326:980-1. http://dx.doi.org/10.1136/bmj.326.7396.980.
- Djulbegovic B, Lacevic M, Cantor A, Fields KK, Bennett CL, Adams JR, et al. The uncertainty principle and industry-sponsored research. Lancet 2000;356:635-8. http://dx.doi.org/10.1016/S0140-6736(00)02605-2.
- Felson DT, Glantz L. A surplus of positive trials: weighing biases and reconsidering equipoise. Arthritis Res Ther 2004;6:117-19. http://dx.doi.org/10.1186/ar1189.
- Ashcroft RE. Ethics of randomised controlled trials – not yet time to give up on equipoise. Arthritis Res Ther 2004;6:237-9. http://dx.doi.org/10.1186/ar1442.
- Lavori PW, Rush AJ, Wisniewski SR, Alpert J, Fava M, Kupfer DJ, et al. Strengthening clinical effectiveness trials: equipoise-stratified randomization. Biol Psychiatry 2001;50:792-801. http://dx.doi.org/10.1016/S0006-3223(01)01223-9.
- Lempp H, Scott DL, Kingsley GH. Patients’ views on the quality of health care for rheumatoid arthritis. Rheumatology 2006;45:1522-8. http://dx.doi.org/10.1093/rheumatology/kel132.
- Lempp H, Scott DL, Kingsley GH. The personal impact of rheumatoid arthritis on patients’ identity. Chronic Illn 2006;2:109-20. http://dx.doi.org/10.1177/17423953060020020601.
- Lempp H, Hofmann D, Hatch SL, Scott DL. Patients’ views about treatment with combination therapy for rheumatoid arthritis: a comparative qualitative study. BMC Musculoskelet Disord 2012;13. http://dx.doi.org/10.1186/1471-2474-13-200.
- Isaacs JD, Ferraccioli G. The need for personalised medicine for rheumatoid arthritis. Ann Rheum Dis 2011;70:4-7. http://dx.doi.org/10.1136/ard.2010.135376.
- Miossec P, Verweij CL, Klareskog L, Pitzalis C, Barton A, Lekkerkerker F, et al. Biomarkers and personalised medicine in rheumatoid arthritis: a proposal for interactions between academia, industry and regulatory bodies. Ann Rheum Dis 2011;70:1713-18. http://dx.doi.org/10.1136/ard.2011.154252.
- Rahman MU, Buchanan J, Doyle MK, Hsia EC, Gathany T, Parasuraman S, et al. Changes in patient characteristics in anti-tumor necrosis factor clinical trials for rheumatoid arthritis: results of an analysis of the literature over the past 16 years. Ann Rheum Dis 2011;70:1631-40. http://dx.doi.org/10.1136/ard.2010.146043.
- Sokka T, Pincus T. Eligibility of patients in routine care for major clinical trials of anti-tumor necrosis factor alpha agents in rheumatoid arthritis. Arthritis Rheum 2003;48:313-18. http://dx.doi.org/10.1002/art.10817.
- Sokka T, Pincus T. Most patients receiving routine care for rheumatoid arthritis in 2001 did not meet inclusion criteria for most recent clinical trials or American College of Rheumatology criteria for remission. J Rheumatol 2003;30:1138-46.
- Zink A, Strangfeld A, Schneider M, Herzer P, Hierse F, Stoyanova-Scholz M, et al. Effectiveness of tumor necrosis factor inhibitors in rheumatoid arthritis in an observational cohort study: comparison of patients according to their eligibility for major randomized clinical trials. Arthritis Rheum 2006;54:3399-407. http://dx.doi.org/10.1002/art.22193.
- Tunis SR, Stryer DB, Clancy CM. Practical clinical trials: increasing the value of clinical research for decision making in clinical and health policy. JAMA 2003;290:624-32. http://dx.doi.org/10.1001/jama.290.12.1624.
- Garattini S, Bertele’ V. How can we regulate medicines better?. BMJ 2007;335:803-5. http://dx.doi.org/10.1136/bmj.39281.615706.94.
- Stafford RS, Wagner TH, Lavori PW. New, but not improved? Incorporating comparative-effectiveness information into FDA labeling. N Engl J Med 2009;361:1230-3. http://dx.doi.org/10.1056/NEJMp0906490.
Appendix 1 Types of protocol deviations and permitted flexibilities
Deviation type number | Protocol deviation | Protocol criteria | Data Monitoring and Ethics Committee flexibility |
---|---|---|---|
1 | Multiple DMARDs (while on TNFi) | Patients randomised to the TNFi arm are permitted to take one DMARD only (methotrexate unless contraindicated) | No flexibility |
2 | High-dose steroids | Not on high-dose steroids (in excess of 10 mg of prednisolone or equivalent per day at trial entry) | No flexibility |
3 | Trial medication given before baseline | Not expressly stated, but trial medication should commence immediately after the baseline outcome data are collected | No flexibility |
4 | Baseline outcome data (questionnaires) collected 3 months after starting trial medication | Not expressly stated, but trial medication should commence immediately after the baseline outcome data are collected | No flexibility |
5 | > 8 weeks off trial medication | A temporary interruption in trial medication of up to 8 weeks (consecutive) will be permitted if an adverse event or other unforeseen circumstance, deemed by the principal investigator to require stoppage of trial medication, has occurred | No flexibility |
6 | Ineligible – history of serious illness | No serious intercurrent illness | No flexibility |
7 | Changed treatment at 6 months despite improvement in DAS of > 1.2 | At 6 months: no change in treatment if good response (≥ 1.2 fall in DAS) | No flexibility |
8 | Steroid injections given between screening and baseline | If a steroid injection is given before baseline, the baseline assessment should be delayed for 1 month after the date of the injection | Include in the ITT analysis and the per-protocol analysis but baseline screening assessment should be used rather than the one immediately following the steroid injection |
9 | Milestone assessments performed outside the visit window | Milestone assessments (6 and 12 months) must be performed within ±14 days of the estimated date of assessment; this was not defined in the protocol but as part of the TACIT Working Practice | Milestone assessments must be performed within ±31 days of the estimated date of assessment |
10 | Insufficient medication at baseline | At baseline, patients must be started on cDMARDs if in the DMARD arm and on a TNFi with accompanying DMARD if in the TNFi arm | Allow up to 1 month from baseline for the introduction of the second trial medication |
11 | Chest radiography not carried out prior to randomisation | Negative screen for tuberculosis (including chest radiography) | Local methods can be used |
12 | Patient not switched at 6 months | Patients assessed at 6 months: no change if good response (≥ 1.2 fall in DAS); change treatment from 6-month assessment if < 1.2 fall in DAS (change to second TNFi if in TNFi arm; change to first TNFi if in cDMARD arm) | Switch permitted at up to 9 months; however, the decision to switch is still based on the 6-month time point |
Appendix 2 Health economic costs
This appendix contains the source data for costs used in the economic analysis.
Item | Unit | Unit cost (£) (2010/11 prices) | Assumptions |
---|---|---|---|
GP | |||
At surgerya | Consultation | 30 | Per surgery consultation lasting 11.7 minutes. Includes direct care staff costs; excludes qualification costs |
At homea | Home visit | 99 | Per home visit lasting 23.4 minutes. Includes direct care staff costs; excludes qualification costs |
Telephone calla | Call | 18 | Per telephone consultation lasting 7.1 minutes. Includes direct care staff costs; excludes qualification costs |
Repeat prescription request without GP contacta | Prescription | 19 | Assuming 5 minutes of GP time. Includes direct care staff costs; excludes qualification costs |
Nurse | |||
At surgerya | Visit | 11 | Based on cost per hour of face-to-face contact. Excludes qualifications and assumes that each consultation lasts 15.5 minutes |
Telephone calla | Call | 7 | Assumes that ratio of time spent on telephone consultation to time spent on face-to-face consultation is same as for GP (60.68%) |
Physiotherapist | |||
At hospitalb | Attendance | 38 | Physiotherapy Total Attendances – Adult (19 and Over) – service code 650A – Total OPATT table |
At homea,c | Visit | 58 | Based on 2010/11 prices but with time estimates from 2009/10. Excludes qualification costs |
At GP surgerya,c | Visit | 27 | Based on 2010/11 prices but with time estimates from 2009/10. Excludes qualification costs |
Elsewherea | Visit | 27 | Assumes same cost as physiotherapist at GP surgery as conservative estimate |
Occupational therapist | |||
At hospitalb | Attendance | 56 | Occupational Therapy Total Attendances – Adult (19 and Over) – service code 651A – Total OPATT table |
At homea,c | Visit | 57 | Based on 2010/11 prices but with time estimates from 2009/10. Excludes qualification costs |
At GP surgerya,c | Visit | 20 | Based on 2010/11 prices but with time estimates from 2009/10. Excludes qualification costs |
Elsewherea | Visit | 20 | Assumes same cost as occupational therapist at GP surgery as conservative estimate |
Hospital services | |||
A&Eb | Attendance | 108 | Accident and Emergency Services: Not leading to Admitted – Index table |
Hospital stay 1 nightb | Bed-day | 568 | Non-Elective Inpatient (Short Stay) HRG Data – Index table (per 1 night) |
Hospital stay > 1 nightb | Bed-day | 426 | Weighted average of all Non-Elective Inpatient (Long Stay) HRG Data |
Outpatient appointmentb | Attendance | 105 | Total – Outpatient Attendances – Index table |
Social services | |||
Meals on Wheelsa | Meal | 6 | Average cost per local authority meal on wheels |
Home helpa | Visit | 12 | Assumes 30-minute visits. Based on cost per hour of face-to-face contact. Weighted average accounting for different rates for day/evening/weekday/weekends |
Social workera | Hour | 152 | Adult services – cost per hour of face-to-face contact. Excludes qualifications |
Social worker telephone calla | Call | 38 | Assumes face-to-face consultation lasting 15 minutes |
Other health or social service | |||
Community and outreach nurseb | Contact | 50 | Community and Outreach Nursing Services: Specialist Nursing – Index table – TCSCNSN |
Dentistb | Attendance | 78 | TOCS tab: Community Dental Services – CN20 |
District nursea | Contact | 64 | Assumes home visit lasting 1 hour |
Orthoticsa | Contact | 16 | Assumes podiatrist |
Osteopathd | Contact | 43 | Assumes mid-point cost per session from range of £35–50 per 30- to 40-minute contact |
Paramedicb | Contact | 119 | TPARO – Index table |
Podiatrista | Contact | 16 | Assumes 30-minute appointment |
Medicatione | Milligrams | Range < 0.01 to 296 | |
Social security benefits | |||
Attendance Allowancef | Benefit | 60 | Based on mean of high and low attendance allowance |
Disability Living Allowancef | Benefit | 41 | Based on mean of care and mobility component |
Council Tax Benefitf | Benefit | 65 | Based on single person aged 25+ years |
Housing Benefitf | Benefit | 65 | Based on single person aged 25+ years |
Incapacity Benefitf | Benefit | 75 | Based on the mean of lower and higher rates of short-term incapacity benefit under state pension age |
Income Supportf | Benefit | 65 | Based on single person aged 25+ years |
Jobseeker’s Allowancef | Benefit | 65 | Based on contribution-based jobseeker’s allowance personal rates, aged 25+ years |
Severe Disablement Allowancef | Benefit | 59 | Basic rate only |
Statutory Sick Payf | Benefit | 79 | |
Working Tax Creditg | Benefit | 37 | Based on basic element only, £1920 per year divided by 52 weeks |
Carer Allowancef | Benefit | 54 | |
Child Benefitg | Benefit | 20 | Assumes one child only |
Child Tax Creditg | Benefit | 10 | Based on basic element only, £545 per year divided by 52 weeks |
Disability Working Allowanceg | Benefit | 51 | Disabled person’s tax credit replaced disability working allowance (see www.hmrc.gov.uk/dptctables/index.htm), £2650 per year divided by 52 weeks |
Family Tax Creditg | Benefit | 10 | Child tax credit family element |
Pensionf | Benefit | 98 | Category A or B pension |
Pension Creditf | Benefit | 10 | The maximum for a single person is £20.52 so divide by 2 for an arbitrary figure |
Tax Creditg | Benefit | 37 | Assumes working tax credit |
Medication | Preparation | Cost per mg (£)a | Cost per day if dose missing (£)b | Cost per medication if medication taken as required (£, assuming medication used for 1 month)b |
---|---|---|---|---|
Adalimumab | Injection | 8.94 | 1399.73 | 1399.73 |
Azathioprine | Oral | 0.01 | 0.28 | 8.27 |
Ciclosporin | Oral | 0.02 | 3.09 | 92.67 |
Depo-Medrone | Injection | 0.07 | 4.76 | 4.76 |
Etanercept | Injection | 3.58 | 1002.12 | 1002.12 |
Folic acid | Oral | 0.01 | 0.04 | 1.06 |
Gold injections | Injection | 0.22 | 27.69 | 27.69 |
Hydroxychloroquine | Oral | < 0.00 | 0.17 | 4.95 |
Infliximab | Injection | 4.20 | 419.62 | 419.62 |
Kenalog (Triamcinolone, Bristol-Myers Squibb) | Injection | 0.04 | 1.92 | 1.92 |
Leflunomide | Oral | 0.17 | 2.36 | 70.87 |
Methotrexate | Oral | 0.05 | 5.01 | 5.01 |
Methotrexate | Injection | 1.98 | 5.01 | 5.01 |
Methylprednisolone | Injection | 0.04 | 47.64 | 47.64 |
Penicillamine | Oral | < 0.00 | 1.00 | 29.94 |
Prednisolone | Oral | 0.06 | 0.09 | 2.58 |
Sulfasalazine | Oral | < 0.00 | 0.49 | 14.66 |
Appendix 3 Complete-case population analysis
This appendix contains the tables and figures from the complete-case analysis.
Outcome | cDMARDs (n = 72) | TNFis (n = 75) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | |
HAQ score | 1.85 (1.71 to 1.99) | 1.50 (1.34 to 1.66) | 1.33 (1.16 to 1.51) | 0.35 (0.23 to 0.48) | 0.52 (0.41 to 63) | 1.84 (1.68 to 2.00) | 1.43 (1.24 to 1.63) | 1.47 (1.27 to 1.66) | 0.41 (0.26 to 0.55) | 0.38 (0.24 to 0.51) |
EQ-5D score | 0.40 (0.32 to 0.47) | 0.53 (0.47 to 0.60) | 0.62 (0.56 to 0.69) | −0.14 (−0.21 to −0.06) | −0.22 (−0.31 to −0.14) | 0.36 (0.29 to 0.43) | 0.57 (0.50 to 0.63) | 0.53 (0.46 to 0.60) | −0.21 (−0.27 to −0.14) | −0.17 (−0.24 to −0.10) |
Larsen Score | 47.3 (37.6 to 57.1) | 48.1 (38.3 to 58.0) | 47.4 (37.4 to 57.4) | −0.8 (−1.7 to 0.1) | −1.4 (−2.7 to −0.2) | 34.7 (25.9 to 43.5) | 34.4 (25.5 to 43.4) | 35.1 (25.9 to 44.3) | −0.3 (−1.1 to 0.6) | −0.7 (−1.8 to 0.4) |
Outcome | cDMARDs (n = 72) | TNFis (n = 75) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | Initial | 6 months | 12 months | Change 0–6 months | Change 0–12 months | |
Physical functioning | 30.8 (25.6 to 36.0) | 36.5 (30.1 to 43.0) | 42.8 (36.2 to 49.4) | −5.7 (−12.7 to 1.3) | −11.9 (−18.6 to −5.3) | 25.8 (21.1 to 30.6) | 44.3 (37.8 to 50.8) | 40.3 (33.6 to 46.9) | −18.4 (−24.4 to −12.5) | −14.4 (−20.5 to −8.3) |
Role physical | 13.5 (7.1 to 20.0) | 38.2 (28.4 to 48.0) | 37.2 (26.8 to 47.5) | −24.7 (−35.6 to −13.7) | −23.6 (−34.3 to −13.0) | 14.3 (7.8 to 20.9) | 44.7 (34.7 to 54.6) | 36.0 (26.2 to 45.9) | −30.3 (−40.8 to −19.9) | −21.7 (−31.7 to −11.7) |
Pain | 26.0 (22.6 to 29.4) | 41.5 (36.8 to 46.1) | 48.9 (43.5 to 54.2) | −15.5 (−20.9 to −10.1) | −22.9 (−28.7 to −17.0) | 28.4 (24.3 to 32.4) | 49.3 (44.3 to 54.3) | 46.2 (40.7 to 51.7) | −21.0 (−26.8 to −15.1) | −17.9 (−24.2 to −11.5) |
General health perception | 34.7 (30.6 to 38.8) | 40.0 (35.3 to 44.7) | 45.1 (39.5 to 50.7) | −5.4 (−10.0 to −0.7) | −10.8 (−16.2 to −5.3) | 31.6 (27.7 to 35.5) | 47.3 (42.5 to 52.1) | 40.7 (35.5 to 45.8) | −15.7 (−20.9 to −10.6) | −9.1 (−14.2 to −3.9) |
Vitality | 27.6 (23.1 to 32.0) | 36.6 (31.3 to 41.9) | 38.2 (32.4 to 44.0) | −9.0 (−14.7 to −3.4) | −10.6 (−16.1 to −5.1) | 28.1 (23.6 to 32.6) | 43.5 (38.3 to 48.7) | 39.9 (34.3 to 45.5) | −15.5 (−21.0 to −10.0) | −11.8 (−17.8 to −5.8) |
Social functioning | 50.0 (44.4 to 55.6) | 61.3 (54.9 to 67.7) | 68.0 (61.7 to 74.2) | −11.3 (−17.6 to −5.0) | −18.1 (−25.0 to −11.2) | 42.7 (36.9 to 48.4) | 61.2 (54.9 to 67.5) | 61.2 (54.6 to 67.7) | −18.5 (−25.7 to −11.3) | −18.5 (−26.4 to −10.6) |
Role emotion | 44.9 (34.2 to 55.7) | 62.5 (51.9 to 73.1) | 63.4 (53.0 to 73.9) | −17.6 (−31.0 to −4.2) | −18.5 (−31.8 to −5.2) | 35.6 (25.2 to 45.9) | 56.0 (45.2 to 66.8) | 54.7 (43.9 to 65.4) | −20.4 (−34.0 to −6.9) | −19.1 (−33.2 to −5.1) |
Mental health | 61.6 (56.9 to 66.2) | 68.3 (63.5 to 73.1) | 71.9 (67.2 to 76.7) | −6.7 (−12.1 to −1.4) | −10.4 (−16.0 to −4.8) | 59.8 (54.7 to 64.9) | 68.5 (63.6 to 73.4) | 68.4 (63.5 to 73.2) | −8.7 (−14.4 to −3.0) | −8.6 (−14.4 to −2.8) |
PCS | 27.9 (26.3 to 29.5) | 32.3 (30.0 to 34.6) | 34.3 (31.7 to 36.9) | −4.4 (−6.9 to −1.8) | −6.3 (−9.0 to −3.7) | 27.9 (26.2 to 29.6) | 36.7 (34.4 to 39.0) | 34.0 (31.7 to 36.2) | −8.8 (−10.9 to −6.7) | −6.1 (−8.4 to −3.7) |
MCS | 43.2 (40.4 to 46.0) | 47.7 (44.7 to 50.6) | 48.9 (46.2 to 51.6) | −4.5 (−7.6 to −1.4) | −6.0 (−9.4 to −2.8) | 41.0 (38.2 to 43.7) | 46.2 (43.3 to 49.2) | 46.5 (43.8 to 49.2) | −5.3 (−8.5 to −2.0) | −5.5 (−8.9 to −2.2) |
Outcome | Model 1 (unadjusted): treatment + region | Model 2 (adjusted): treatment + demographics + baseline score | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
Change in HAQ score | ||||
12 months | 0.14 (−0.03 to 0.32) | 0.108 | 0.15 (−0.03 to 0.32) | 0.094 |
6 months | −0.06 (−0.25 to 0.13) | 0.542 | −0.08 (−0.271 to 0.10) | 0.362 |
Outcome | Model 1 (unadjusted): treatment + region + time | Model 2 (adjusted): treatment + demographics + baseline score + time | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
Larsen score | −0.58 (−1.86 to 0.69) | 0.370 | −0.46 (−1.91 to 0.98) | 0.531 |
SF-36 role physical | 1.39 (−10.75 to 13.52) | 0.822 | 2.29 (−8.22 to 12.80) | 0.669 |
SF-36 vitality | 3.84 (−2.69 to 10.37) | 0.249 | 3.66 (−1.82 to 9.14) | 0.190 |
SF-36 social functioning | 3.55 (−4.95 to 12.05) | 0.413 | −2.49 (−9.27 to 4.29) | 0.472 |
SF-36 role emotion | 1.11 (−15.37 to 17.59) | 0.895 | −7.00 (−18.40 to 4.39) | 0.229 |
SF-36 mental health | −0.08 (−6.78 to 6.63) | 0.982 | −1.77 (−6.69 to 3.15) | 0.480 |
SF-36 MCS | 0.07 (−3.82 to 3.97) | 0.971 | −1.68 (−4.64 to 1.29) | 0.268 |
Outcome | Stayed on cDMARDs (n = 35) | Changed to TNFis (n = 37) | ||||||
---|---|---|---|---|---|---|---|---|
Initial | 6 months | 12 months | Change 0–12 months | Initial | 6 months | 12 months | Change 0–12 months | |
HAQ score | 1.91 (1.69 to 2.14) | 1.35 (1.11 to 1.58) | 1.39 (1.13 to 1.65) | 0.53a (0.37 to 0.68) | 1.79 (1.62 to 1.96) | 1.64 (1.42 to 1.85) | 1.28 (1.04 to 1.53) | 0.51 (0.34 to 0.67) |
Larsen score | 54.2 (38.1 to 70.3) | 54.8 (38.6 to 70.9) | 55.5 ( 39.5 to 71.5) | −1.3b (−3.0 to 0.5) | 40.8 (29.3 to 52.3) | 41.8 (30.1 to 53.6) | 39.4 (27.4 to 51.4) | −1.6 (−3.4 to 0.3) |
EQ-5D score | 0.38 (0.27 to 0.49) | 0.63 (0.55 to 0.71) | 0.68 (0.61 to 0.76) | −0.30c (−0.40 to −0.20) | 0.41 (0.31 to 0.51) | 0.44 (0.34 to 0.54) | 0.56 (0.46 to 0.67) | −0.15 (−0.28 to −0.02) |
Outcome | Model 1 (unadjusted): treatment + region + time | Model 2 (adjusted): treatment + demographics + baseline score + time | ||
---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | |
Change in HAQ score | ||||
12 months | 0.22 (0.03 to 0.41) | 0.025 | 0.20 (0.01 to 0.39) | 0.039 |
Month of assessment | cDMARDs (n = 72) | TNFis (n = 75) | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
DAS28 | Tender joint count | Swollen joint count | ESR (mm/hour) | VAS | DAS28 | Tender joint count | Swollen joint count | ESR (mm/hour) | VAS | |
0 | 6.20 (5.98 to 6.42) | 15.96 (14.47 to 17.45) | 10.01 (8.58 to 11.45) | 34.17 (28.04 to 40.30) | 67.42 (62.74 to 72.10) | 6.28 (6.10 to 6.47) | 18.17 (16.66 to 19.68) | 11.28 (9.66 to 12.90) | 26.12 (21.72 to 30.52) | 67.56 (62.97 to 72.15) |
1 | 5.36 (5.05 to 5.68) | 11.43 (9.44 to 13.41) | 6.80 (5.32 to 8.28) | 33.40 (27.50 to 39.30) | 54.54 (48.47 to 60.60) | 4.56 (4.24 to 4.88) | 10.92 (8.89 to 12.95) | 5.18 (3.90 to 6.46) | 16.51 (12.91 to 20.10) | 44.07 (38.34 to 49.79) |
2 | 5.01 (4.70 to 5.31) | 9.30 (7.65 to 10.95) | 5.89 (4.67 to 7.10) | 31.03 (25.14 to 36.92) | 50.09 (44.22 to 55.95) | 3.99 (3.64 to 4.33) | 7.48 (5.98 to 8.98) | 4.44 (3.05 to 5.83) | 16.79 (12.27 to 21.31) | 41.14 (35.32 to 46.96) |
3 | 4.91 (4.55 to 5.27) | 9.48 (7.61 to 11.35) | 5.58 (4.20 to 6.95) | 30.65 (24.58 to 36.72) | 49.99 (43.70 to 56.27) | 4.06 (3.70 to 4.41) | 7.20 (5.52 to 8.88) | 4.03 (2.75 to 5.30) | 18.68 (15.04 to 22.32) | 41.12 (35.32 to 46.92) |
4 | 4.69 (4.37 to 5.01) | 8.38 (6.71 to 10.05) | 4.76 (3.44 to 6.09) | 30.93 (25.30 to 36.55) | 41.93 (35.47 to 48.38) | 4.14 (3.75 to 4.52) | 8.19 (6.36 to 10.03) | 3.88 (2.70 to 5.05) | 18.00 (14.33 to 21.67) | 41.65 (35.21 to 48.09) |
5 | 4.61 (4.25 to 4.97) | 8.37 (6.62 to 10.12) | 5.73 (4.30 to 7.16) | 28.74 (23.26 to 34.22) | 41.91 (35.10 to 48.73) | 3.87 (3.52 to 4.22) | 7.03 (5.15 to 8.90) | 3.75 (2.62 to 4.88) | 16.21 (13.13 to 19.29) | 39.01 (32.96 to 45.07) |
6 | 4.70 (4.29 to 5.11) | 9.78 (7.70 to 11.86) | 5.97 (4.28 to 7.66) | 28.61 (22.68 to 34.55) | 46.76 (39.59 to 53.93) | 3.93 (3.56 to 4.30) | 7.80 (5.88 to 9.72) | 3.96 (2.67 to 5.25) | 17.77 (13.94 to 21.61) | 36.64 (30.78 to 42.50) |
7 | 4.55 (4.19 to 4.91) | 8.54 (6.61 to 10.46) | 5.83 (4.38 to 7.28) | 28.44 (22.33 to 34.54) | 40.96 (34.46 to 47.46) | 3.82 (3.44 to 4.19) | 7.00 (5.23 to 8.77) | 3.76 (2.44 to 5.09) | 17.47 (13.51 to 21.44) | 37.63 (31.17 to 44.08) |
8 | 4.20 (3.84 to 4.57) | 7.29 (5.62 to 8.96) | 4.46 (3.18 to 5.74) | 25.14 (19.41 to 30.87) | 39.43 (33.15 to 45.71) | 3.57 (3.21 to 3.93) | 5.24 (3.77 to 6.70) | 3.13 (1.97 to 4.28) | 16.90 (13.02 to 20.78) | 37.63 (31.17 to 44.08) |
9 | 4.10 (3.74 to 4.45) | 6.50 (4.94 to 8.06) | 3.82 (2.75 to 4.88) | 26.25 (20.51 to 31.99) | 37.93 (31.89 to 43.97) | 3.76 (3.41 to 4.11) | 5.95 (4.36 to 7.53) | 3.54 (2.27 to 4.81) | 17.18 (13.96 to 20.40) | 37.66 (32.12 to 43.20) |
10 | 3.94 (3.61 to 4.27) | 5.97 (4.42 to 7.52) | 3.64 (2.66 to 4.62) | 26.17 (20.54 to 31.80) | 35.90 (30.37 to 41.43) | 3.54 (3.19 to 3.90) | 5.51 (3.85 to 7.16) | 2.93 (1.80 to 4.06) | 16.97 (13.15 to 20.79) | 34.75 (28.83 to 40.67) |
11 | 3.95 (3.63 to 4.28) | 6.35 (4.65 to 8.06) | 3.25 (2.23 to 4.28) | 24.01 (18.71 to 29.31) | 37.73 (31.37 to 44.10) | 3.54 (3.17 to 3.91) | 5.23 (3.68 to 6.79) | 3.25 (1.89 to 4.61) | 17.49 (13.66 to 21.33) | 32.25 (26.14 to 38.35) |
12 | 3.92 (3.58 to 4.26) | 5.82 (4.16 to 7.48) | 3.14 (2.28 to 4.00) | 25.03 (19.27 to 30.79) | 36.38 (29.76 to 42.99) | 3.44 (3.09 to 3.79) | 5.59 (3.93 to 7.24) | 2.55 (1.66 to 3.43) | 15.07 (11.36 to 18.78) | 36.97 (30.38 to 43.57) |
Time period | Variable | Model 1 (unadjusted): treatment + region+ time | Model 2 (adjusted): treatment + demographics + baseline score + time | ||
---|---|---|---|---|---|
Coefficient (95% CI) | p-value | Coefficient (95% CI) | p-value | ||
Months 1–6 | DAS28 | −1.06 (−1.41 to −0.71) | < 0.001 | −1.02 (−1.37 to −0.68) | < 0.001 |
Tender joint count | −4.37 (−6.40 to −2.35) | < 0.001 | −3.14 (−4.99 to −1.29) | 0.001 | |
Swollen joint count | −3.33 (−4.95 to −1.71) | < 0.001 | −2.52 (−3.70 to −1.33) | < 0.001 | |
ESR (mm/hour) | −5.95 (−10.33 to −1.56) | 0.008 | −9.20 (−13.02 to −5.37) | < 0.001 | |
VAS | −10.08 (−17.08 to −3.07) | 0.005 | −9.07 (−15.36 to −2.78) | 0.005 | |
Months 6–12 | DAS28 | −0.69 (−1.11 to −0.27) | 0.001 | −0.58 (−0.97 to −0.18) | 0.004 |
Tender joint count | −3.65 (−5.93 to −1.37) | 0.002 | −2.12 (−3.92 to −0.32) | 0.021 | |
Swollen joint count | −2.81 (−4.71 to −0.91) | 0.004 | −1.71 (−2.84 to −0.57) | 0.003 | |
ESR (mm/hour) | −2.71 (−7.38 to 1.96) | 0.255 | −5.01 (−9.06 to −0.95) | 0.016 | |
VAS | −2.73 (−11.16 to 5.71) | 0.527 | −1.47 (−8.53 to 5.60) | 0.684 | |
Months 1–12 | DAS28 | −0.92 (−1.29 to −0.55) | < 0.001 | −0.82 (−1.18 to −0.46) | < 0.001 |
Tender joint count | −4.26 (−6.42 to −2.11) | < 0.001 | −2.71 (−4.50 to −0.92) | 0.003 | |
Swollen joint count | −3.11(−4.85 to −1.37) | < 0.001 | −2.05 (−3.13 to −0.97) | < 0.001 | |
ESR (mm/hour) | −5.20 (−9.52 to −0.88) | 0.018 | −7.96 (−11.58 to −4.35) | < 0.001 | |
VAS | −6.55(−14.17 to 1.07) | 0.092 | −5.55 (−11.90 to 0.81) | 0.087 |
Appendix 4 Adverse events
This appendix contains details of all of the adverse events reported in the trial.
Adverse event | Frequency | Per cent |
---|---|---|
Abdominal discomfort | 3 | 0.47 |
Ache – arms | 1 | 0.16 |
Ache – back | 1 | 0.16 |
Ache – ear | 1 | 0.16 |
Ache – entire body | 3 | 0.47 |
Ache – foot (left) | 1 | 0.16 |
Ache – hands | 1 | 0.16 |
Anaemia | 1 | 0.16 |
Bilateral lower lobe bronchiectasis | 1 | 0.16 |
Bladder irritation | 2 | 0.31 |
Blepharitis – eye (right) | 1 | 0.16 |
Blurred vision | 3 | 0.47 |
Blurred vision (right eye) | 2 | 0.31 |
Breast lump (right) | 1 | 0.16 |
Breathlessness | 4 | 0.63 |
Bruising – arm (left) | 1 | 0.16 |
Bruising – arm (right) | 1 | 0.16 |
Bruising – flank (left) | 1 | 0.16 |
Bruising – thigh (left) | 1 | 0.16 |
Bruising – thigh (right) | 1 | 0.16 |
Burning on micturition | 1 | 0.16 |
Burning sensation – arm (right) | 1 | 0.16 |
Burning sensation – shoulder (right) | 1 | 0.16 |
Cardiac palpitations | 2 | 0.31 |
Carpal tunnel syndrome | 1 | 0.16 |
Cellulitis – shin (right) | 1 | 0.16 |
Chest infection | 19 | 2.99 |
Chest tightness | 3 | 0.47 |
Cold | 12 | 1.89 |
Cold sore | 1 | 0.16 |
Collapse | 1 | 0.16 |
Constipation | 4 | 0.63 |
Cough | 5 | 0.79 |
Cough – dry | 4 | 0.63 |
Cough – productive | 6 | 0.94 |
Cramp – legs | 3 | 0.47 |
Cramp – stomach | 2 | 0.31 |
Cutaneous vasculitis | 1 | 0.16 |
Cyst – breast (right) | 1 | 0.16 |
Cyst – kidney (right) | 1 | 0.16 |
Depression | 2 | 0.31 |
Diarrhoea | 30 | 4.72 |
Diverticular disease | 1 | 0.16 |
Dizziness | 9 | 1.42 |
Dry mouth | 3 | 0.47 |
Dry skin | 1 | 0.16 |
Ear infection (left) | 1 | 0.16 |
Eczema – lower legs | 1 | 0.16 |
Elevated alkaline phosphatase | 2 | 0.31 |
Elevated alanine aminotransferase | 7 | 1.1 |
Elevated aspartate aminotransferase | 1 | 0.16 |
Elevated cholesterol | 2 | 0.31 |
Elevated creatinine | 3 | 0.47 |
Elevated CRP | 3 | 0.47 |
Elevated ESR | 2 | 0.31 |
Elevated gamma-glutamyl transferase | 3 | 0.47 |
Elevated globulin | 1 | 0.16 |
Elevated liver enzyme | 3 | 0.47 |
Enlarged axillary lymph nodes (left) | 1 | 0.16 |
Epigastric pain | 1 | 0.16 |
Exacerbation of hypertension | 1 | 0.16 |
Exhaustion | 1 | 0.16 |
Fall | 4 | 0.63 |
Fatigue | 11 | 1.73 |
Fever | 6 | 0.94 |
Fibromyalgia | 1 | 0.16 |
Flare of RA | 17 | 2.68 |
Flatulence | 1 | 0.16 |
Flexor tendonitis | 2 | 0.31 |
Flu | 7 | 1.1 |
Flushed | 2 | 0.31 |
Funny taste in mouth | 1 | 0.16 |
Gallstones | 1 | 0.16 |
Gastroenteritis | 1 | 0.16 |
Gingivitis | 1 | 0.16 |
Gum disease | 1 | 0.16 |
Haematemesis | 1 | 0.16 |
Haematuria | 1 | 0.16 |
Haematuria – macroscopic | 1 | 0.16 |
Haematuria – microscopic | 1 | 0.16 |
Haemoptysis | 1 | 0.16 |
Hair loss | 1 | 0.16 |
Headache | 30 | 4.72 |
Head cold | 3 | 0.47 |
Hearing – diminished | 1 | 0.16 |
Heartburn | 4 | 0.63 |
Hiatus hernia – moderate size | 1 | 0.16 |
High blood pressure | 7 | 1.1 |
Hot flush | 4 | 0.63 |
Hot flushes | 1 | 0.16 |
Hypoglycaemia | 2 | 0.31 |
Increased frequency of defecation | 2 | 0.31 |
Increased urine frequency | 1 | 0.16 |
Indigestion | 3 | 0.47 |
Infected eyes | 1 | 0.16 |
Infection – foot (left) | 1 | 0.16 |
Inflamed eye (right) | 1 | 0.16 |
Injection site reaction | 1 | 0.16 |
Insomnia | 1 | 0.16 |
Intermittent visual disturbance – eye | 1 | 0.16 |
Itchy skin | 7 | 1.1 |
Joint pain – feet | 1 | 0.16 |
Joint pain – generalised | 3 | 0.47 |
Laryngitis | 1 | 0.16 |
Lesion – spleen | 1 | 0.16 |
Lethargy | 2 | 0.31 |
Leucocytes in urine | 2 | 0.31 |
Lightheadedness | 3 | 0.47 |
Loose stools | 3 | 0.47 |
Loss of appetite | 5 | 0.79 |
Low blood pressure | 1 | 0.16 |
Lower back pain | 1 | 0.16 |
Low creatinine | 1 | 0.16 |
Low haemoglobin | 2 | 0.31 |
Low iron level | 1 | 0.16 |
Low platelet count | 3 | 0.47 |
Low white cell count | 7 | 1.1 |
Lymphadenopathy lungs | 1 | 0.16 |
Lymphopenia | 4 | 0.63 |
Migraine | 4 | 0.63 |
Multiple liver cysts | 1 | 0.16 |
Muscle ache – generalised | 1 | 0.16 |
Muscle ache – shins | 3 | 0.47 |
Muscle ache – shoulder | 1 | 0.16 |
Nausea | 26 | 4.09 |
Neutropenia | 1 | 0.16 |
Night sweat | 4 | 0.63 |
Nitrites in urine | 2 | 0.31 |
Nosebleed | 2 | 0.31 |
Oedema – feet | 1 | 0.16 |
Pain – abdominal | 4 | 0.63 |
Pain – ankle (left) | 2 | 0.31 |
Pain – arm (left) | 1 | 0.16 |
Pain – arm (right) | 2 | 0.31 |
Pain – back | 5 | 0.79 |
Pain – entire body | 3 | 0.47 |
Pain – eye (left) | 1 | 0.16 |
Pain – feet | 4 | 0.63 |
Pain – first metatarsophalangeal joint (right) | 1 | 0.16 |
Pain – foot (left) | 3 | 0.47 |
Pain – groin (left) | 1 | 0.16 |
Pain – hand (right) | 1 | 0.16 |
Pain – hip (left) | 1 | 0.16 |
Pain – hip (right) | 2 | 0.31 |
Pain – knee (left) | 1 | 0.16 |
Pain – knee (right) | 3 | 0.47 |
Pain – left side | 1 | 0.16 |
Pain – leg (right) | 1 | 0.16 |
Pain – neck | 5 | 0.79 |
Pain – shoulder (left) | 4 | 0.63 |
Pain – shoulder (right) | 5 | 0.79 |
Pain – shoulders | 4 | 0.63 |
Pain – wrist (left) | 1 | 0.16 |
Pain – wrist (right) | 3 | 0.47 |
Parotid enlargement (bilateral) | 1 | 0.16 |
Peripheral neuropathy | 1 | 0.16 |
Pins and needles – arms (both) | 1 | 0.16 |
Pneumonia | 2 | 0.31 |
Protein in urine | 5 | 0.79 |
Pruritus – arms | 1 | 0.16 |
Pulmonary fibrosis | 1 | 0.16 |
Raised temperature | 2 | 0.31 |
Rash – arm (left) | 2 | 0.31 |
Rash – arm (right) | 1 | 0.16 |
Rash – arms | 3 | 0.47 |
Rash – back | 4 | 0.63 |
Rash – cheek (right) | 1 | 0.16 |
Rash – entire body | 6 | 0.94 |
Rash – face | 3 | 0.47 |
Rash – heel (right) | 1 | 0.16 |
Rash – leg (left) | 2 | 0.31 |
Rash – leg (right) | 2 | 0.31 |
Rash – legs | 4 | 0.63 |
Rash – neck | 2 | 0.31 |
Rash – torso | 4 | 0.63 |
Rectal bleeding | 2 | 0.31 |
Redness – eye (right) | 2 | 0.31 |
Reduced appetite | 2 | 0.31 |
Rigours | 1 | 0.16 |
Runny nose | 1 | 0.16 |
Shaking | 1 | 0.16 |
Shingles | 3 | 0.47 |
Shortness of breath | 1 | 0.16 |
Sinusitis | 3 | 0.47 |
Skin infection – breast (right) | 1 | 0.16 |
Skin lesions | 1 | 0.16 |
Sore gums | 2 | 0.31 |
Sore lips | 1 | 0.16 |
Sore mouth | 4 | 0.63 |
Sore throat | 15 | 2.36 |
Stomach ache | 1 | 0.16 |
Sweating | 1 | 0.16 |
Swelling – ankle (both) | 2 | 0.31 |
Swelling – calf (left) | 1 | 0.16 |
Swelling – face (right side) | 1 | 0.16 |
Swelling – upper lip | 1 | 0.16 |
Swine flu | 1 | 0.16 |
Swollen gums | 4 | 0.63 |
Swollen temporomandibular joint | 1 | 0.16 |
Synovitis – metatarsophalangeal joint | 1 | 0.16 |
Tachycardia | 1 | 0.16 |
Taste – metallic | 1 | 0.16 |
Tendonitis – achilles | 1 | 0.16 |
Thrush – mouth | 2 | 0.31 |
Tingling – mouth area | 3 | 0.47 |
Tonsillitis | 1 | 0.16 |
Tooth extraction | 5 | 0.79 |
Tooth infection | 2 | 0.31 |
Torn muscle – lower back | 1 | 0.16 |
Transient ischaemic attack | 1 | 0.16 |
Ulcerative skin lesion – breast (right) | 1 | 0.16 |
Ulcers – mouth | 12 | 1.89 |
Urinary incontinence | 1 | 0.16 |
Urinary tract infection | 6 | 0.94 |
Verruca | 1 | 0.16 |
Vomiting | 26 | 4.09 |
Weight gain | 1 | 0.16 |
Weight loss | 5 | 0.79 |
Total | 635 | 100 |
Adverse event | Frequency | Per cent |
---|---|---|
Abnormal vaginal bleeding | 1 | 0.22 |
Abscess – axilla (left) | 2 | 0.43 |
Abscess – tooth | 2 | 0.43 |
Ache – back | 2 | 0.43 |
Ache – ear | 2 | 0.43 |
Ache – ear (left) with discharge | 1 | 0.22 |
Ache – entire body | 4 | 0.86 |
Ache – hand (right) | 1 | 0.22 |
Ache – neck | 1 | 0.22 |
Ache – tooth | 3 | 0.65 |
Anxiety | 1 | 0.22 |
Baker’s cyst | 2 | 0.43 |
Blocked eustachian tube (right) | 1 | 0.22 |
Blurred vision | 1 | 0.22 |
Breast lump (right) | 1 | 0.22 |
Breathlessness | 7 | 1.51 |
Broken arm (right) | 1 | 0.22 |
Bruising – limbs | 1 | 0.22 |
Bruising – thigh (left) | 1 | 0.22 |
Bruising – thigh (right) | 1 | 0.22 |
Burning sensation – neck (right hand side) | 1 | 0.22 |
Cardiac palpitations | 2 | 0.43 |
Carpal tunnel syndrome | 3 | 0.65 |
Cellulitis – leg (right) | 2 | 0.43 |
Chest infection | 27 | 5.81 |
Chest pain | 5 | 1.08 |
Choking – on waking up | 1 | 0.22 |
Cold | 16 | 3.44 |
Cold hands and feet | 1 | 0.22 |
Cold sore | 7 | 1.51 |
Congested ears | 1 | 0.22 |
Constipation | 2 | 0.43 |
Cough | 4 | 0.86 |
Cough – dry | 4 | 0.86 |
Cough – productive | 5 | 1.08 |
Cramp – abdominal | 1 | 0.22 |
Cyst – liver | 1 | 0.22 |
Deep vein thrombosis | 1 | 0.22 |
Depression | 1 | 0.22 |
Diarrhoea | 12 | 2.58 |
Diverticular disease | 1 | 0.22 |
Dizziness | 4 | 0.86 |
Drop in estimated glomerular filtration rate | 1 | 0.22 |
Dry feet with flaky skin and blotches | 1 | 0.22 |
Dry throat | 3 | 0.65 |
Dyspareunia | 1 | 0.22 |
Ear infection (left) | 2 | 0.43 |
Eczema – all over body | 1 | 0.22 |
Elevated alkaline phosphatase | 2 | 0.43 |
Elevated alanine aminotransferase | 16 | 3.44 |
Elevated cholesterol | 2 | 0.43 |
Elevated gamma-glutamyl transferase | 1 | 0.22 |
Elevated globulin | 1 | 0.22 |
Elevated liver enzyme | 1 | 0.22 |
Elevated potassium | 2 | 0.43 |
Elevated protein (blood) | 1 | 0.22 |
Exacerbation of bronchiectasis | 1 | 0.22 |
Exhaustion | 1 | 0.22 |
Fall | 2 | 0.43 |
Fatigue | 5 | 1.08 |
Fatty liver | 2 | 0.43 |
Fever | 2 | 0.43 |
Flare of RA | 14 | 3.01 |
Flexor tendonitis | 2 | 0.43 |
Flu | 4 | 0.86 |
Folliculitis | 1 | 0.22 |
Fracture – foot (left) | 1 | 0.22 |
Fungal infection – nail (big toes) | 1 | 0.22 |
Gastroenteritis | 1 | 0.22 |
Gum infection | 1 | 0.22 |
Haematuria – macroscopic | 2 | 0.43 |
Haematuria – microscopic | 1 | 0.22 |
Haemostatis | 1 | 0.22 |
Hair loss | 1 | 0.22 |
Headache | 15 | 3.23 |
Head cold | 1 | 0.22 |
Hearing – diminished | 1 | 0.22 |
Heartburn | 3 | 0.65 |
Hot flushes | 1 | 0.22 |
Impaired walking | 1 | 0.22 |
Increased urine frequency | 1 | 0.22 |
Indigestion | 1 | 0.22 |
Infected eyes | 2 | 0.43 |
Infection – big toe nail (right) | 1 | 0.22 |
Injection site reaction | 5 | 1.08 |
Insect bite | 3 | 0.65 |
Insomnia | 1 | 0.22 |
Intermittent flashing lights – both eyes | 1 | 0.22 |
Intermittent headache | 1 | 0.22 |
Itchy skin | 3 | 0.65 |
Joint stiffness – generalised | 1 | 0.22 |
Lethargy | 4 | 0.86 |
Leucocytes in urine | 1 | 0.22 |
Loose stools | 1 | 0.22 |
Lower back pain | 2 | 0.43 |
Low platelet count | 1 | 0.22 |
Low white cell count | 1 | 0.22 |
Malaise | 1 | 0.22 |
Medial epicondylitis – (left) | 1 | 0.22 |
Medial epicondylitis – (right) | 1 | 0.22 |
Muscle ache – shoulder | 1 | 0.22 |
Nasal congestion | 2 | 0.43 |
Nausea | 8 | 1.72 |
Neck – stiff | 3 | 0.65 |
Neutropenia | 3 | 0.65 |
Nose – left axillary lymphadenopathy | 1 | 0.22 |
Numbness – legs | 1 | 0.22 |
Numbness – lips | 1 | 0.22 |
Numbness – toes | 1 | 0.22 |
Oedema – feet | 2 | 0.43 |
Oedema – foot (right) | 2 | 0.43 |
Ovarian cyst | 1 | 0.22 |
Pain – abdominal | 3 | 0.65 |
Pain and redness – index distal interphalangeal joint (right) | 1 | 0.22 |
Pain – ankle (left) | 3 | 0.65 |
Pain – arm (left) | 1 | 0.22 |
Pain – back | 4 | 0.86 |
Pain – entire body | 2 | 0.43 |
Pain – eye (left) | 1 | 0.22 |
Pain – foot (left) | 1 | 0.22 |
Pain – foot (right) | 4 | 0.86 |
Pain – hand (right) | 1 | 0.22 |
Pain – hip (left) | 3 | 0.65 |
Pain – hip (right) | 3 | 0.65 |
Pain – knee (left) | 3 | 0.65 |
Pain – knee (right) | 5 | 1.08 |
Pain – leg (left) | 1 | 0.22 |
Pain – legs | 1 | 0.22 |
Pain – neck | 2 | 0.43 |
Pain – ribs (left) | 1 | 0.22 |
Pain – shoulder (left) | 3 | 0.65 |
Pain – shoulder (right) | 6 | 1.29 |
Pain – wrist (left) | 1 | 0.22 |
Photosensitivity | 1 | 0.22 |
Pins and needles – feet | 1 | 0.22 |
Pins and needles – hands (left and right) | 1 | 0.22 |
Plantar fasciitis | 3 | 0.65 |
Pleurisy | 1 | 0.22 |
Pneumonia | 2 | 0.43 |
Protein in urine | 1 | 0.22 |
Pruritus | 1 | 0.22 |
Raised temperature | 1 | 0.22 |
Raised white blood cell count | 1 | 0.22 |
Rash – arms | 2 | 0.43 |
Rash – back | 1 | 0.22 |
Rash – entire body | 1 | 0.22 |
Rash – face | 2 | 0.43 |
Rash – hand (right) | 1 | 0.22 |
Rash – legs | 1 | 0.22 |
Rash – torso | 4 | 0.86 |
Red and sore eyes (both) | 1 | 0.22 |
Restless legs | 2 | 0.43 |
Rigours | 1 | 0.22 |
Runny nose | 3 | 0.65 |
Scalp rash | 2 | 0.43 |
Shaking | 1 | 0.22 |
Shingles | 3 | 0.65 |
Shortness of breath | 1 | 0.22 |
Sinusitis | 1 | 0.22 |
Skin infection – ankle (left) | 1 | 0.22 |
Skin lesions | 2 | 0.43 |
Skin nodules – back | 1 | 0.22 |
Skin nodules – face | 1 | 0.22 |
Soft tissue nodule – both feet | 1 | 0.22 |
Sore mouth | 1 | 0.22 |
Sore throat | 13 | 2.8 |
Streptococcus A infection – vaginal | 1 | 0.22 |
Superficial thrombophlebitis | 1 | 0.22 |
Swelling – ankle (both) | 2 | 0.43 |
Swelling – ankle (left) | 1 | 0.22 |
Swelling – knee (left) | 2 | 0.43 |
Swelling – lower jaw | 1 | 0.22 |
Swelling – wrist (right) | 1 | 0.22 |
Thrush – mouth | 2 | 0.43 |
Thrush – vaginal | 5 | 1.08 |
Tonsillitis | 4 | 0.86 |
Tooth extraction | 2 | 0.43 |
Tooth infection | 3 | 0.65 |
Trace glucose in urine | 1 | 0.22 |
Ulcers – mouth | 4 | 0.86 |
Ulcers – vascular | 1 | 0.22 |
Upper respiratory tract infection | 6 | 1.29 |
Urinary incontinence | 1 | 0.22 |
Urinary tract infection | 9 | 1.94 |
Uterus fibroid | 1 | 0.22 |
Vaginal dryness | 1 | 0.22 |
Vasovagal attack | 4 | 0.86 |
Vomiting | 3 | 0.65 |
Weight gain | 1 | 0.22 |
Whitening of nails | 1 | 0.22 |
Total | 465 | 100 |
Appendix 5 Systematic review search strategies
Search strategy for early rheumatoid arthritis
-
Early Rheumatoid Arthritis.mp
-
Early RA.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
infliximab.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
etanercept.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
adalimumab.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
golimumab.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
certolizumab [mp=ti, ab, ot, nm, hw, kf, ps, rs, ui, an, sh, tn, dm, mf, dv, kw]
-
anti-TNF.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
biological products.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
methotrexate.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
cyclosporin.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
sulphasalazine.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
hydroxychloroquine.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
prednisolone.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
immunosuppressive agents.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combination DMARDs.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combinatin atreatment.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combination anti-rheumatic drugs.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combination therapy.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
leflunomide.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
1 or 2
-
3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12 or 13 or 14 or 15 or 18 or 19
-
20 and 21
-
remove duplicates from 23
-
limit 24 to clinical trial
-
limit 25 to english language
Search strategy for established rheumatoid arthritis
-
Rheumatoid Arthritis.mp
-
RA.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
infliximab.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
etanercept.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
adalimumab.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
golimumab.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
certolizumab [mp=ti, ab, ot, nm, hw, kf, ps, rs, ui, an, sh, tn, dm, mf, dv, kw]
-
anti-TNF.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
biological products.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
methotrexate.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
cyclosporin.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
sulphasalazine.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
hydroxychloroquine.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
prednisolone.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
immunosuppressive agents.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combination DMARDs.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combinatin atreatment.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combination anti-rheumatic drugs.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
combination therapy.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
leflunomide.mp. [mp=ti, ab, sh, hw, tn, ot, dm, mf, dv, kw, nm, ps, rs, an, ui]
-
1 or 2
-
3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12 or 13 or 14 or 15 or 18 or 19
-
20 and 21
-
remove duplicates from 23
-
limit 24 to clinical trial
-
limit 25 to english language
List of abbreviations
- ACR
- American College of Rheumatology
- anti-CCP
- anticyclic citrullinated peptide
- BeSt
- Behandel Strategieen
- BSR
- British Society for Rheumatology
- CARDERA
- Combination Anti-Rheumatic Drugs in Early Rheumatoid Arthritis
- cDMARD
- combination disease-modifying antirheumatic drug
- CEAC
- cost-effectiveness acceptability curve
- CI
- confidence interval
- CRP
- C-reactive protein
- CSRI
- Client Service Receipt Inventory
- DAS28
- Disease Activity Score for 28 Joints
- DAS
- Disease Activity Score
- DMARD
- disease-modifying antirheumatic drug
- EDC
- electronic data capture
- EQ-5D
- European Quality of Life-5 Dimensions
- ESR
- erythrocyte sedimentation rate
- GEE
- generalised estimating equation
- GP
- general practitioner
- HAQ
- Health Assessment Questionnaire
- ICER
- incremental cost-effectiveness ratio
- IL
- interleukin
- IQR
- interquartile range
- ITT
- intention to treat
- MCS
- Mental Component summary
- MRI
- magnetic resonance imaging
- NICE
- National Institute for Health and Care Excellence
- OR
- odds ratio
- PCS
- Physical Component summary
- QALY
- quality-adjusted life-year
- RA
- rheumatoid arthritis
- RCT
- randomised controlled trial
- SD
- standard deviation
- SF-36
- Short Form Questionnaire-36 items
- Swefot
- Swedish Farmacotherapy
- TACIT
- Tumour necrosis factor inhibitors Against Combination Intensive Therapy
- TNFi
- tumour necrosis factor inhibitor
- VAS
- visual analogue scale
- WMD
- weighted mean difference