• No results found

TNF blockade in daily practice Bijl, A.E. van der

N/A
N/A
Protected

Academic year: 2021

Share "TNF blockade in daily practice Bijl, A.E. van der"

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)TNF blockade in daily practice Bijl, A.E. van der. Citation Bijl, A. E. van der. (2010, March 9). TNF blockade in daily practice. Retrieved from https://hdl.handle.net/1887/15056 Version:. Corrected Publisher’s Version. License:. Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden. Downloaded from:. https://hdl.handle.net/1887/15056. Note: To cite this publication please use the final published version (if applicable)..

(2) Chapter 1: introduction. Rheumatoid arthritis Rheumatoid arthritis (RA) is a chronic inflammatory auto-immune disease especially involving the joints. The disease is characterised by chronic inflammation of the synovium resulting in damage of the articular structures e.g. cartilage and bone. Patients affected with RA suffer from pain, swelling and limited movement of the affected joints. Rheumatoid arthritis has a worldwide distribution and affects 0.8% of the population (prevalence)(1) from which ~70% is female. The annual incidence is 36/100 000 in women and 14/100 000 in men(1). For reasons unknown, the incidence, prevalence as well as mortality of RA appear to have fallen in women in the last 50 year(2). Besides involvement of the synovium, this systemic disease can lead to clinical symptoms outside the joints, the so-called extra-articular manifestation. Many patients have non-specific complaints including fatigue, anorexia, weight loss and increased body temperature. Another relative common clinical feature is nodulosis. This subcutaneous swelling is predominately located on the bony prominences and consists of inflammatory cells with central necrosis. Other, more severe extra-articular manifestations with considerable morbidity and mortality are mononeuritis (multiplex), serositis, vasculitis as well as eye-involvement such as panuveitis. These severe extra-articular manifestations, as well as distinct destruction of the joint, occur most often in longstanding and active RA. Patients suffering from RA have decreased life expectancy due to cardiovascular complications, infections, (lympho-proliferative) malignancy or gastroenterological disease(3). Besides the direct clinical symptoms, RA is a severe disease associated with substantial individual and societal costs. Health care, loss of productivity and benefit payments for work disability contribute to increased expenses(4;5). The diagnosis RA is based on the clinical findings in combination with specific laboratory and radiographic features. The American College of Rheumatology (ACR 1987) criteria(6) (see table) describe the classification criteria, including signs and symptoms that are associated with relatively advanced disease. At the first presentation, recognition of RA by the clinical symptoms may be difficult since the start of the disease can be diverse ranging from slowly progressive to acutely disabling(7). One third of patients who present with undifferentiated arthritis (UA) progress to the rheumatoid arthritis, yet, the clinical phenotypes of these patients can not be distinguished from those who do not progress to RA or who go into spontaneous remission (approximately 50%)(8;9). The clinical outcomes of the disease appear not to depend on the tempo in which the clinical symptoms develop(7). For this reason, prediction of the clinical course of the disease based on the initial clinical picture is difficult. The presence of the auto-antibodies, e.g. rheumatoid factor (RF) and anti-citrullinated protein/ peptide antibodies (ACPA), may help making an early diagnosis. These auto-antibodies occur much more frequent in UA patients who progress to RA(10) and appear to develop already years prior to the first symptoms of arthritis(11). Recently, ACPA results were integrated in a prediction model that was developed to estimate the chance of progression to RA in individual patients presenting with UA(12).. Chapter 1.   

(3)  

(4)

(5)

(6) . 7. 

(7)

(8)

(9) .

(10) American College of Rheumatology classification criteria RA: the presence of 4 or more of the following criteria: morning stiffness morning stiffness in and around joints lasting for at least 1 hour before maximal improvement poly-arthritis soft tissue swelling (arthritis) of 3 or more joint areas observed by a physician arthritis of the hand joints swelling (arthritis) of the proximal interphalangeal, metacarpophalangeal, or wrist joints symmetric arthritis rheumatoid nodules subcutaneous nodules over bony permanencies, extensor surfaces or in juxta-articular region rheumatoid factor radiographic erosions The criteria 1 through 4 must have been present for at least 6 weeks.. pathogenesis The cause of rheumatoid arthritis is still unknown. Both genetic and environmental factors play a role in the development of the disease. Genetic factors probably account for ~60% of disease susceptibility and expression. It has been suggested that the composition of the peptide-binding site by the shared epitope (SE) region on the HLA-DRB1 gene(13;14) resulted in presentation of arthritogenic peptides to T cells contributing to the pathogenesis of RA(15). In the resulting inflammation activated (T- and B-) lymphocytes, macrophages, cytokines (including TNF-Ơ) as well as synoviocytes play an important role. The presence of the shared epitope is a risk factor for the development of ACPA(16) and presence of ACPA is correlated with the severity of the disease(17). Previous observational studies indicated smoking as an important environmental risk factor contributing to the development of RA(18). Further investigations have shown that tobacco exposure is a risk factor for development of ACPA only in patients who carry HLA-DRB1 SE alleles(13;19). In patients with ACPA-negative rheumatoid arthritis HLA-DR3 was more frequent present indicating a different pathogenetic mechanisms underling ACPA-positive and ACPA-negative RA(20).. treatment Untreated or insufficiently treated RA will result in joint destruction and permanent disability and an at least 2-fold increase in mortality(3;21). Mortality appears to be increased due to specific causes such as infections, lympho-proliferative malignancies, gastro-enterological diseases and the inflammatory character of the disease itself(3). More recently it has been shown that patients with rheumatic arthritis have an increased risk of cardiovascular disease (CVD) which is not fully explained by the traditional. 8.   

(11)  

(12)

(13)

(14) . Chapter 1. 

(15)

(16)

(17) .

(18) risk factors of CVD(22). Active inflammation and secondary disability are two of the major risk factors for CVD in patients with active RA(23). Recent data indicate that suppression of inflammation reduces the risk of CVD morbidity and mortality in patients with severe RA(22). Nowadays, the goal of treatment is to relieve symptoms, to prevent joint damage and to decrease the excess of mortality. Treatment of RA has changed considerably over the last decades. Before the 1980s patients were treated according to a “pyramid schedule” starting with non-steroid anti-inflammatory drugs (NSAIDs) for all patients and the introduction of disease modifying anti-rheumatic drugs (DMARDs) was reserved for ‘severe cases’ expressing rapid joint destruction. Subsequently, it became obvious that (early) introduction of DMARDs was important to delay the progression of joint damage(24;25). Nevertheless, it proved to be very difficult to stop or alter the course of the disease with the available drugs. In the next years, newer DMARDs and especially DMARD-combination therapy were initiated. Therapy strategies starting with, or moving quickly on to, a combination of drugs resulted in more and earlier effective suppression of the inflammation and the clinical symptoms as well as of radiological damage progression(26-30).. monitoring of disease activity In order to register disease activity in RA patients a Disease Activity Score (DAS) has been designed. The DAS can be calculated by a formula using the following parameters: number of tender and swollen joints, erythrocyte sedimentation rate (ESR) and a global health assessment. The score was first designed for clinical research(31), however, the DAS has become an important measurement for clinicians in order to adjust therapy(32). Currently, the most commonly used disease scores are the original DAS based on evaluation of 44 joints, and the DAS28, describing disease activity in 28 joints (see figure).. Chapter 1.   

(19)  

(20)

(21)

(22) . 9. 

(23)

(24)

(25) .

(26) DAS-guided therapy Besides the application of intensive therapy in patients with (early) RA, DAS-guided therapy can further improve the clinical outcome of a patient with RA. Recently, a survey of clinical trials using tight control demonstrated a higher rate of remission compared to earlier trials and/or control groups(33). The TICORA trial demonstrated that compared to ‘routine care’, DAS-guided outpatient management is superior in improving the symptoms of RA and in preventing radiographic damage progression(30). Also in the CAMERA trial, intensive and frequent surveillance of patients aimed at achieving a set goal of low disease activity(27). The remarkable results of the DAS guided BeSt study(34) -more than 80% of patients in all 4 treatment arms achieve the goal of low disease activity, more than 40% achieved remission- demonstrate that rheumatologists have to target themselves and their patients higher goals in terms of aiming low disease activity or even remission, and use the available drugs until that goal is achieved. Once low disease is achieved the probability of maintaining a low DAS without further treatment adjustments is high as again demonstrated in the BeSt(35).. window of opportunity In recent years the treatment of RA has become more focused on optimal intervention in the early phase of the disease(36). It has been demonstrated that patients treated in this period have higher response rates to therapy than those receiving delayed treatment(25) and experienced less radiographic damage over time(24). A meta-analysis confirmed the perspective that early treatment was associated with sustained benefit in terms of less radiographic progression for up to even 5 years(37). It is hypothesized that intensive treatment in the early phase of the disease, the so-called “window of opportunity”, may not only result in a rapid and effective induction of low disease activity but may also effectively change the clinical course of the disease(38). The terms ‘remission’ and ‘remission induction’, both previously unknown in the vocabulary of rheumatologist and patients, have recently been introduced as possible treatment goals. Several studies gave support to the “window of opportunity” hypothesis. The BeSt demonstrated the superiority of initial intensive combination therapy -either with prednisone or infliximab- resulting in earlier achievement of low disease activity and remission and less progression of radiographic damage(26). This trial was also the first that allowed patients with permanent low disease activity to discontinue all medication after a prolonged period of drug tapering. Depending on the therapy, drug free disease remission (defined as DAS<1.6) was achieved in up to 18% of the patients(34). Both clinical remission and radiographic non-progression were achieved in 80% of patients with early severe rheumatoid arthritis after 1 year of combined treatment with etanercept plus MTX (COMET)(39). In another approach, the PROMPT trial showed that, in ACPA positive patients with UA, the clinical diagnosis of RA could be postponed by treating them with MTX(40). Unfortunately, most patients still developed RA after MTX was discontinued, indicating that MTX in these patients is not the right drug to induce remission during the “window of opportunity”.. 10.   

(27)  

(28)

(29)

(30) . Chapter 1. 

(31)

(32)

(33) .

(34) The timing and duration of the “window of opportunity” is not known and the optimal therapy in order to induce disease remission is still under debate(41). Ideally, optimal timing and therapy would collaborate in a cure for early RA. It appears however, that the symptoms of joint inflammation may be a relative late manifestation of a disease process that may have started with auto-antibody formation(11) prior to the clinical picture defined as RA. Also the high rate of early erosions -up to 80%- in trials especially focused on early RA like BeSt and COBRA(26;28), suggests already longstanding inflammation of the joint prior to establishing the diagnosis. Identification of risk factors for the development of clinical diagnosis RA remains a challenge and it is currently unknown if intervention at the stage of asymptomatic RA would be superior to safe, effective and affordable treatment of early symptomatic RA.. anti-rheumatic drugs Several “conventional” DMARDs are currently available for the treatment of RA, such as methotrexate (MTX), sulphasalazine (SASP), leflunomide, gold salts, hydroxychloroquine (HCQ), azathioprine and cyclosporine. These drugs are moderately to well tolerated(42) and to some extent effective in suppressing the rheumatoid process(43), but this last claim depends on the definition of ‘effective’, which over time has become more stringent(44). Several comparative drug studies have shown that, in the majority of patients, anti-TNF therapy results in earlier and lower disease activity as well as better improvement of functional ability than treatment with MTX, either after previous failure on DMARDs or in untreated early RA(26;39;45-48). Furthermore, with the “conventional” DMARDs, prevention of joint damage progression seems inferior to that achieved with anti-TNF. Unfortunately, some patients do not benefit (enough) from treatment with these agents. Only between 46% and 67% of patients with advanced RA show a 50% improvement in clinical outcomes, and between 20% and 30% of patients show no significant improvement at all(49-51). In a systematic review the efficacy of the different TNF blocking agents was not different(52). In case of (secondary) inefficacy and/or a (allergic) side effect, in many countries patients may switch to a second TNF-blocking agent or other biologicals such as anti-B cell therapy or anti-activated T-cell therapy. However, study results on switching biological therapies are difficult to interpret because the definitions of the reasons for switching are often as vague as the definition of ‘success’ or ‘failure’ on the second drug. The efficacy of DMARDs can be increased by adding corticosteroids. In literature, most reports describe combination therapy with oral prednisolone, however, from a practical point of view, in daily practice parenteral administration of corticosteroids, intra-muscular or intra-articular, is used frequently. Initially corticosteroids were welcomed as a wonder drug in the late 1940s-1950s. Cortisone proved very effective in suppressing the inflammation of rheumatoid arthritis(53,54), however, later reports on side effects of long term and relatively high dose corticosteroids led to increasing reservations against using these drugs, both in physicians and in patients(55). These reservations remain to this day, although physicians and patients appear to be concerned about different side effects(56-58). Despite these limitations, it is adequately. Chapter 1.   

(35)  

(36)

(37)

(38) . 11. 

(39)

(40)

(41) .

(42) proven that combination therapy with DMARDs and prednisolone is very effective as treatment for rheumatoid arthritis(59,60) and that during the use of low dose and short term the clinically significant side effects appear to be absent or manageable. When in the BeSt study, initial combination therapy with methotrexate, sulphasalazine and tapered high dose of prednisone was head to head compared to initial combination therapy with methotrexate and infliximab, a TNF-blocker, patients in both arms showed equal improvement and suppression of radiological damage over the first 2 years of follow up(61), although patients treated with infliximab showed greater improvement in quality of life(van der Hout ACR 2009), and appeared to be happier with the treatment received than patients treated with prednisone(62). Three TNF-blocking agents are currently registered in the Netherlands for the treatment of RA. Two monoclonal antibodies are used: infliximab (Remicade®; chimeric mouse-human antibody) and adalimumab (Humira®; humanised antibody). The third anti-TNF is a fusion-protein: etanercept (Enbrel®). TNF-blocking agents are administrated via intravenous infusion (infliximab) or subcutaneous injection (etanercept/ adalimumab). Infliximab and adalimumab are both monoclonal antibodies targeting free as well as bound TNF. In contrast, etanercept is a TNF-receptor only binding to free TNF. To date, in the Netherlands patients are reimbursed for treatment with a TNF-blocker only if at least 2 conventional DMARDs, including MTX, are unsuccessfully tried. Since TNF is produced locally in the synovium, intra-articular injections of infliximab and etanercept have been tried with varying results(63). For a long time, the optimal timing for the introduction of a TNF-blocking agent in RA had not been defined. The BeSt study evaluated the use of infliximab in four different treatment strategies (see below for details of the study design) and showed that initial treatment with a combination of drugs, including infliximab or prednisone, was more effective than initial treatment with monotherapy(26). Furthermore, initial treatment with infliximab was better than delayed treatment with anti-TNF after failure on conventional DMARDs(64). The higher costs of initial infliximab can probably be compensated by less indirect societal costs as patients treated with initial infliximab have less sick leave and work more paid hours(65). A French study suggests that even a period of three months methotrexate monotherapy, before treatment with anti-TNF, resulted in a delay of clinical improvement in the majority of patients but not in significant less usage of anti-TNF(66).. working mechanism of anti-TNF The 17 kDa polypeptide cytokine Tumor Necrosis Factor (TNF) is an important mediator of inflammation. The name stems from the fact that TNF, when administered locally in high dose, induces tumor necrosis. However, produced chronically it may in fact act as a tumor promoter(67;68). Induction of TNF by pathogenic stimuli induces a cascade of other inflammatory proteins such as cytokines and chemokines resulting in recruitment and activating of a range of cells at the site of the infection or tissue damage(69). A negative feedback mechanism controls this process. In auto-immune diseases, such as RA, there appears to be an imbalance between pro- and anti-inflam-. 12.   

(43)  

(44)

(45)

(46) . Chapter 1. 

(47)

(48)

(49) .

(50) matory stimuli. TNF plays an important role(70). TNF is produced by macrophage cells as response to cytokines produced by activated CD4+ T-cells. Besides TNF, the macrophages produce other pro-inflammatory cytokines such as IL-1 and interferon. TNF binding to the target cell results in increased number of adhesion molecules of the endothelium (resulting in an increase of inflammatory cells), release of metalloproteïnasen (resulting in destruction of extracellular matrix) and production of proinflammatory cytokines. In RA the net effect is pro-inflammatory excess resulting in persistent inflammation with all its clinical characteristics. In humans the TNF-blocking agent infliximab demonstrated for the first time a reduction in disease activity and erosive damage(71). Infliximab binds to TNFƠ which in turn prevents the binding of TNFƠ to its receptors on the cell surface, prohibiting the start of the intracellular signal cascade that leads to more pro-inflammatory activity. In vitro, infliximab was found to bind to TNFƠ already bound to the cell receptors, which resulted in cell death(72;73). Whether this also happens in the living organism is unknown. Due to its mode of action, the anti-inflammatory effect of treatment with infliximab may therefore in theory be associated with an increased susceptibility to infections.. adverse events during treatment with anti-TNF: infectious Already in the 90’s, clinical observations demonstrated an increased risk for infection during the use of “conventional” DMARDs(74;75). Based on the theoretic risk, it cannot be ruled out that some of the reports on an increased number of serious infections in relation to anti-inflammatory drugs are subject to expectation bias. It is likely that physicians, and also patients after instruction, are more aware of possible signs and symptoms of infections, will investigate these more thoroughly, and are more prone to treat with intravenous antibiotics. On the other hand, such awareness might lead to an underestimation of the infectious risk because patients are instructed to come in earlier and may be treated with oral antibiotics, or other beneficiary treatment, in the earliest stages of what otherwise might have become a serious infection. In any case, increased numbers of (opportunistic) infections have been reported(76-79), and on the other hand, large database studies appear to demonstrate no significant increase of infections compared to MTX(80) or only an increase of non-serious infections of the skin and soft tissues(81). Annalists of the British Society for Rheumatology Biologics Register found that the adjusted incidence rate ratio for a serious infection in patients receiving anti-TNF compared to patients receiving “conventional” DMARDs was not increased(81). Interestingly, limiting the follow up to 90 days revealed a 4.6 times higher incidence ratio. The increased incidence ratio may be explained by the interruption of anti-TNF after a serious side effect, such as infection or allergic reaction. In RA patients the overall risk for infections -resulting in hospitalisation- seemed raised due to various associated risk factors such as leucopenia, extra-articular manifestation, increased age, co-morbidity and steroid-usage(83;84). Other factors influencing the number of infections may be differences in presence or virulence of specific pathogens in the general population. An example is the high background presence of latent tuberculosis in many countries in Europe, compared to the US (see below).. Chapter 1.   

(51)  

(52)

(53)

(54) . 13. 

(55)

(56)

(57) .

(58) tuberculosis After the introduction of TNF-blocking agents their was an increase in the incidence of granulomatous infectious diseases, especially mycobacterium tuberculosis, in patients treated with these agents, and the clinical presentation also seemed more severe than in patients without anti-TNF(85-89). The rise in tuberculosis infections seemed mainly the result of reactivation of latent tuberculosis, which is present in a considerable part of the population in certain parts of Europe(89;90). After an infection with mycobacterium the micro-organisms that survive are sequestered in granuloma(91). TNF is required for the development and persistence of this granuloma. During anti-TNF therapy this defence mechanism is obstructed resulting in an active infection. Following these observations, screening for (latent) tuberculosis has been recommended by medical society committees such as the Dutch Society for Rheumatology(87;89;92;93). Prior to the start of a TNF blocking agent the patient should be screened by medical history, clinical examination, chest X-ray and a PPD (purified protein derivative) skin test. In patients over age 65 years an initially negative test should be repeated (twostep PPD). In case of positive findings for latent tuberculosis the patient should receive prophylactic therapy prior to the start of anti TNF. Patients with active tuberculosis are excluded from use of TNF blocking agents. influenza Influenza is a common viral infection with a high disease burden, causing significant morbidity and mortality in both immunocompetent and immunodeficient hosts(94). Although data on an increased morbidity and mortality due to influenza in patients treated with anti-TNF are lacking, guidelines recommend that patients at risk for complications of influenza, including those treated with anti-TNF, should be annually vaccinated against influenza(95;96). The use of immunosuppressive therapy can reduce the response upon vaccination. Data on the immunologic response to influenza vaccination during anti-TNF therapy are scarce and conflicting(97-100) and data on the clinical relevance of this vaccination in these patients are altogether lacking.. adverse events during treatment with anti-TNF: neurological A pivotal role for TNF in the pathogenesis of inflammatory demyelinating disease of the central nervous system has been suggested in several studies of MS in humans and in experimental autoimmune encephalomyelitis(101). TNF is overproduced in the serum and cerebrospinal fluid of MS patients and by resident and infiltrating cells at sites of CNS injury. There were high expectations that the use of anti-TNF-alpha in MS patients could suppress progression, but a randomized trial was adjourned prematurely because of an increase in exacerbations(102). On the other hand, new-onset neurologic signs and symptoms with signs of demyelination in the central nervous system have been reported in patients using TNF-blocking agents(103). Also case reports were published of patients with optic neuritis which developed during treatment with anti-TNF(104). How demyelination would be a result to use of anti-TNF is unknown, although several explanations have been proposed(105). Large database research, however, did not show an increase in neurological symptoms in patients treated with. 14.   

(59)  

(60)

(61)

(62) . Chapter 1. 

(63)

(64)

(65) .

(66) TNF-blockers compared to the general population, which puts a direct link between the use of TNF-alpha blockade and demyelination under debate(103;106;107). Still many agree that it seems prudent to avoid the use of anti-TNF in patients with a history of demyelination. In contrast to possible negative side effects of TNF blocking agents, from clinical experience it certainly appears that many patients experience an improvement in quality of life and wellbeing that rises above the degree of clinical improvement in the signs and symptoms of RA. On the other hand it has been suggested that inflammatory responses, including an increased TNF, may have an important role in the pathophysiology of depression(108;109). Also, an association of a TNF-308G/A promoter polymorphism with schizophrenia and bipolar affective disorder in a Polish population has been identified(110). This has resulted in speculation on a possible role of TNF and TNF blockade in mental processes.. discontinuation of anti-TNF in treatment of RA: Serious adverse events would of course be a reason to discontinue anti-TNF, as would be lack of response (see below). Discontinuation because of a good response was rarely tried in daily practice, since it was demonstrated that discontinuation led to exacerbation of the inflammatory process(111). Quinn et al were the first to demonstrate that in patients with early RA, initial treatment with anti-TNF could be discontinued without loss of efficacy(112). This was again shown on a larger scale in the BeSt study(26). The BeSt study is a randomized trial comparing the clinical and radiographic outcomes of 4 different treatment strategies: sequential monotherapy (group 1; start with MTX, subsequent steps for patients with an insufficient response were: SASP monotherapy, leflunomide monotherapy, MTX with infliximab, further steps), stepup combination therapy (group 2; start MTX, subsequent steps for patients with an insufficient response were: SASP added, followed by the addition of HCQ and then by prednisone, MTX with infliximab, further steps), initial combination therapy with MTX and tapered high-dose prednisone (group 3, subsequent steps for patients with an insufficient response were combination of MTX with ciclosporin and prednisone, MTX with infliximab, further steps), and initial combination therapy with MTX and infliximab (group 4, subsequent steps for patients with an insufficient response were SASP, leflunomide, further steps). The common goal in all strategies was to reduce disease activity rapidly and persistently by tight monitoring and immediate adjustment of therapy in the case of an insufficient response. Treatment adjustments were based on the DAS that was measured every 3 months and 1 to 2 weeks prior to each infliximab infusion by a research nurse who remained blinded for the allocated treatment strategy. Good clinical response to therapy was defined as a DAS ≤2.4, indicating low disease activity. If this was not achieved, the next step in the protocol was accomplished. According to the protocol, in case of persistently low disease activity (DAS ≤2.4 for at least 6 consecutive months) treatment intensity was reduced. In case of combinations of drugs, the drugs were discontinued one by one as long as a DAS ≤2.4 was maintained. The results of the BeSt study showed that more patients had earlier and. Chapter 1.   

(67)  

(68)

(69)

(70) . 15. 

(71)

(72)

(73) .

(74) better clinical response, and less joint damage progression, after initial treatment with combination therapy, either including prednisone or infliximab, than after initial treatment with MTX monotherapy (and subsequent steps if necessary). More than 50% of patients who had started treatment with MTX and infliximab and achieved a DAS ≤2.4 could permanently discontinue infliximab without loosing the clinical response. These results demonstrated that “window of opportunity”, where the RA process can be effectively arrested or even reversed, probably exists and that infliximab can be effective as ‘remission induction therapy’, with the additional benefits of reduced exposure time, less risks for side effects and less costs in the long term. Another indication for the “window of opportunity” is the experience in the BeSt that using MTX plus infliximab as initial treatment for recent onset RA patients is more effective than reserving MTX plus infliximab for patients who failed on traditional DMARDs(64).. switching anti-TNF As pointed out before, in clinical trials up to 70% of RA patients showed at least some improvement after treatment with anti-TNF. Approximately 30% did not achieve clinical improvement. Predictors of non-response were smoking and high disease activity and no relation between the efficacy of TNF blocking agents and age, disease duration, rheumatoid factor or number of DMARD was demonstrated(113). In daily clinical practice, as registered in an open database of anti-TNF use by rheumatologists joined in the DREAM (Dutch Rheumatoid Arthritis Monitoring), the response rate appeared to be lower compared to previous randomised controlled trials(114). This was possibly due to the inclusion of patients with different disease characteristics at the onset of anti-TNF therapy compared to the ones included in randomised clinical trials. Although the net result for all TNF-blockers is the same, which is inhibition of TNF, the mechanisms of action are slightly different. This suggests that, in case of primary inefficacy -immediate insufficient clinical response to an anti-TNF-, switching to another TNF-blocker may be successful(115). However, the results of the ‘switch studies’ are sometimes difficult to interpret because the reasons for discontinuation of the first TNF-blocker were not always clear and the criteria for claiming ‘success’ were not always very strict. Secondary inefficacy -a loss of the original response to a TNF-blocker after some time- is thought to be associated with the development of antibodies against the TNF-blocking agent(116). Reports on switching to another TNF-blocker in these patients suggest that this could be as successful as in primary inefficacy. However, a recent study suggests that patients who produced antibodies against one TNF-blocker have a higher likelihood to produce antibodies to a second one(117). The development of these antibodies affects the clinical response to treatment. It is therefore sometimes suggested that after failure on a TNF-blocker, other targeted therapies may be superior to another TNF-blocker(118;119).. 16.   

(75)  

(76)

(77)

(78) . Chapter 1. 

(79)

(80)

(81) .

(82) other targeted therapy In recent years, anti-B-cell treatment (rituximab), anti IL-6 (tocilizumab) and regulation of T-cell interaction (abatacept) have been introduced for the treatment of RA, and are generally marketed for patients who failed on anti-TNF. Initial trials have shown promising response rates(118;120;121), but in daily practice experience with these drugs is still limited. Other biological therapies are in advanced stages of development and testing: new TNF-blockers golimumab and certolizumab (pegolated anti-TNF), ofatumumab (anti-CD20) and oral JAK and Syk inhibitors(122-126). These drugs give hope for patients who have failed on anti-TNF but may also in due time find a place in the treatment of patients with early RA.. outline of the thesis The current thesis investigates some practical aspects of the use of anti-TNF in daily practice. In chapter 2 we describe the efficacy and safety of a second TNF-Ơ blocking agent, adalimumab, in patients who had an inadequate response or a side effect on infliximab causing them to stop that drug. During anti-TNF-alpha therapy, neurologic symptoms ascribed to demyelination have been reported. There are also links between pro-inflammatory cytokines, including TNF, and behavioural disorders, notably depression(109;127). In chapter 3 we describe the effect of anti-TNF-alpha on the brain parenchyma by using magnetic resonance (MR). National and international guidelines advise influenza as well as pneumococcal vaccination in immunocompromised patients, including patients using TNF-blocking agents. In chapters 4 and 5 we report on the efficacy of influenza and pneumococcal vaccination in patients using anti-TNF-Ơ therapy compared to that in healthy individuals and in patients using other (non-biological) immunosuppressive therapy. Chapter 6 is a report on the use of a combination of infliximab plus methotrexate as initial therapy in patients with recent onset RA. The impact of intensive therapy with infliximab in early RA provides new insights in the clinical course of the disease and could change the way the new biological drugs are positioned in the treatment of RA. We speculate on the possibility of remission induction treatment with infliximab. Currently the use of anti-TNF-Ơ is focused on systemic treatment of patients with a poly-arthritis such as in patients with RA. In cases of mono-arthritis, intra-articular treatment with corticosteroids is the advocated approach. However, recurrent inflammations are frequent, and a more effective therapy would be desirable. Theoretically, intra-articular therapy with anti-TNF-Ơ could be very effective. In chapter 7 we evaluated the effect of infliximab intra-articular in patients with a recurrent and/or chronic gonarthritis. Advanced, insufficiently controlled rheumatoid inflammation affects organs and body tissues outside the joints. This is a condition that is called rheumatic vasculitis. This condition has a high morbidity and mortality, and for the traditional therapy with high dose corticosteroids and immunosuppressants, the same is true. Anti-TNF is probably a safer therapy and has been tried, with various successes, as treatment in patients with rheumatoid vasculitis. In chapter 8 we report a case of a patient treated with infliximab.. Chapter 1.   

(83)  

(84)

(85)

(86) . 17. 

(87)

(88)

(89) .

(90) References (1) Gabriel SE. The epidemiology of rheumatoid arthritis. Rheum Dis Clin North Am 2001; 27(2):26981. (2) Symmons DP. Looking back: rheumatoid arthritis--aetiology, occurrence and mortality. Rheumatology (Oxford) 2005; 44 Suppl 4:iv14-iv17. (3) Wolfe F, Mitchell DM, Sibley JT, Fries JF, Bloch DA, Williams CA et al. The mortality of rheumatoid arthritis. Arthritis Rheum 1994; 37(4):481-94. (4) Clarke AE, Zowall H, Levinton C, Assimakopoulos H, Sibley JT, Haga M et al. Direct and indirect medical costs incurred by Canadian patients with rheumatoid arthritis: a 12 year study. J Rheumatol 1997; 24(6):1051-60. (5) Schadlich PK, Zeidler H, Zink A, Gromnica-Ihle E, Schneider M, Straub C et al. Modelling cost effectiveness and cost utility of sequential DMARD therapy including leflunomide in rheumatoid arthritis in Germany: I. Selected DMARDs and patient-related costs. Pharmacoeconomics 2005; 23(4):37793. (6) 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(3):315-24. (7) van Aken J, van Dongen H, le Cessie S, Allaart CF, Breedveld FC, Huizinga TW. Comparison of long term outcome of patients with rheumatoid arthritis presenting with undifferentiated arthritis or with rheumatoid arthritis: an observational cohort study. Ann Rheum Dis 2006; 65(1):20-5. (8) Verpoort KN, van DH, Allaart CF, Toes RE, Breedveld FC, Huizinga TW. Undifferentiated arthritis-disease course assessed in several inception cohorts. Clin Exp Rheumatol 2004; 22(5 Suppl 35):S12S17. (9) van der Helm-van Mil AH, Verpoort KN, Breedveld FC, Toes RE, Huizinga TW. Antibodies to citrullinated proteins and differences in clinical progression of rheumatoid arthritis. Arthritis Res Ther 2005; 7(5):R949-R958. (10) van Gaalen FA, Linn-Rasker SP, van Venrooij WJ, de Jong BA, Breedveld FC, Verweij CL et al. Autoantibodies to cyclic citrullinated peptides predict progression to rheumatoid arthritis in patients with undifferentiated arthritis: a prospective cohort study. Arthritis Rheum 2004; 50(3):709-15. (11) Nielen MM, van SD, Reesink HW, van de Stadt RJ, van der Horst-Bruinsma IE, de Koning MH et al. Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis Rheum 2004; 50(2):380-6. (12) van der Helm-van Mil AH, Detert J, le CS, Filer A, Bastian H, Burmester GR et al. Validation of a prediction rule for disease outcome in patients with recent-onset undifferentiated arthritis: moving toward individualized treatment decision-making. Arthritis Rheum 2008; 58(8):2241-7. (13) van der Helm-van Mil AH, Huizinga TW, de Vries RR, Toes RE. Emerging patterns of risk factor makeup enable subclassification of rheumatoid arthritis. Arthritis Rheum 2007; 56(6):1728-35. (14) Turesson C, Matteson EL. Genetics of rheumatoid arthritis. Mayo Clin Proc 2006; 81(1):94-101. (15) Gregersen PK, Silver J, Winchester RJ. The shared epitope hypothesis. An approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum 1987; 30(11):1205-13. (16) van der Helm-van Mil AH, Verpoort KN, Breedveld FC, Huizinga TW, Toes RE, de Vries RR. The HLADRB1 shared epitope alleles are primarily a risk factor for anti-cyclic citrullinated peptide antibodies and are not an independent risk factor for development of rheumatoid arthritis. Arthritis Rheum 2006; 54(4):1117-21. (17) De Rycke L, Peene I, Hoffman IE, Kruithof E, Union A, Meheus L et al. Rheumatoid factor and antici-. 18.   

(91)  

(92)

(93)

(94) . Chapter 1. 

(95)

(96)

(97) .

(98) (18) (19). (20). (21) (22) (23) (24). (25). (26). (27). (28). (29). (30). (31) (32) (33) (34). trullinated protein antibodies in rheumatoid arthritis: diagnostic value, associations with radiological progression rate, and extra-articular manifestations. Ann Rheum Dis 2004; 63(12):1587-93. Albano SA, Santana-Sahagun E, Weisman MH. Cigarette smoking and rheumatoid arthritis. Semin Arthritis Rheum 2001; 31(3):146-59. Linn-Rasker SP, van der Helm-van Mil AH, van Gaalen FA, Kloppenburg M, de Vries RR, le CS et al. Smoking is a risk factor for anti-CCP antibodies only in rheumatoid arthritis patients who carry HLADRB1 shared epitope alleles. Ann Rheum Dis 2006; 65(3):366-71. Verpoort KN, van Gaalen FA, van der Helm-van Mil AH, Schreuder GM, Breedveld FC, Huizinga TW et al. Association of HLA-DR3 with anti-cyclic citrullinated peptide antibody-negative rheumatoid arthritis. Arthritis Rheum 2005; 52(10):3058-62. Scott DL, Symmons DP, Coulton BL, Popert AJ. Long-term outcome of treating rheumatoid arthritis: results after 20 years. Lancet 1987; 1(8542):1108-11. Turesson C, Jacobsson LT, Matteson EL. Cardiovascular co-morbidity in rheumatic diseases. Vasc Health Risk Manag 2008; 4(3):605-14. Turesson C, Matteson EL. Cardiovascular risk factors, fitness and physical activity in rheumatic diseases. Curr Opin Rheumatol 2007; 19(2):190-6. van Aken J, Lard LR, le Cessie S, Hazes JM, Breedveld FC, Huizinga TW. Radiological outcome after four years of early versus delayed treatment strategy in patients with recent onset rheumatoid arthritis. Ann Rheum Dis 2004; 63(3):274-9. Lard LR, Visser H, Speyer I, vander Horst-Bruinsma IE, Zwinderman AH, Breedveld FC et al. Early versus delayed treatment in patients with recent-onset rheumatoid arthritis: comparison of two cohorts who received different treatment strategies. Am J Med 2001; 111(6):446-51. Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, van ZD, Kerstens PJ, Hazes JM et al. Comparison of treatment strategies in early rheumatoid arthritis: a randomized trial. Ann Intern Med 2007; 146(6):406-15. Verstappen SM, Jacobs JW, van d, V, 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(11):1443-9. Landewe RB, Boers M, Verhoeven AC, Westhovens R, van de Laar MA, Markusse HM et al. COBRA combination therapy in patients with early rheumatoid arthritis: long-term structural benefits of a brief intervention. Arthritis Rheum 2002; 46(2):347-56. Makinen H, Kautiainen H, Hannonen P, Mottonen T, Leirisalo-Repo M, Laasonen L et al. Sustained remission and reduced radiographic progression with combination disease modifying antirheumatic drugs in early rheumatoid arthritis. J Rheumatol 2007; 34(2):316-21. 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(9430):263-9. van der Heijde DM, van ‘t HM, van Riel PL, van de Putte LB. Development of a disease activity score based on judgment in clinical practice by rheumatologists. J Rheumatol 1993; 20(3):579-81. Ranganath VK, Khanna D, Paulus HE. ACR remission criteria and response criteria. Clin Exp Rheumatol 2006; 24(6 Suppl 43):S-21. Bakker MF, Jacobs JW, Verstappen SM, Bijlsma JW. Tight control in the treatment of rheumatoid arthritis: efficacy and feasibility. Ann Rheum Dis 2007; 66 Suppl 3:iii56-iii60. Van Der Kooij SM, Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Guler-Yuksel M, Zwinderman. Chapter 1.   

(99)  

(100)

(101)

(102) . 19. 

(103)

(104)

(105) .

(106) (35). (36) (37) (38) (39). (40). (41) (42). (43). (44). (45). (46). (47). (48). AH, Kerstens PJ et al. Drug-free remission, functioning and radiographic damage after 4 years of response-driven treatment in patients with recent onset rheumatoid arthritis. Ann Rheum Dis 2008. van der Kooij SM, Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Peeters AJ, van Krugten MV, Breedveld FC et al. Probability of continued low disease activity in patients with recent onset rheumatoid arthritis treated according to the disease activity score. Ann Rheum Dis 2008; 67(2):266-9. Quinn MA, Cox S. The evidence for early intervention. Rheum Dis Clin North Am 2005; 31(4):57589. Finckh A, Liang MH, van Herckenrode CM, de PP. Long-term impact of early treatment on radiographic progression in rheumatoid arthritis: A meta-analysis. Arthritis Rheum 2006; 55(6):864-72. Quinn MA, Emery P. Potential for altering rheumatoid arthritis outcome. Rheum Dis Clin North Am 2005; 31(4):763-72. 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(9636):375-82. van Dongen H, van Aken J, Lard LR, Visser K, Ronday HK, Hulsmans HM et al. Efficacy of methotrexate treatment in patients with probable rheumatoid arthritis: a double-blind, randomized, placebo-controlled trial. Arthritis Rheum 2007; 56(5):1424-32. Nurmohamed MT, Dijkmans BA. Are biologics more effective than classical disease-modifying antirheumatic drugs? Arthritis Res Ther 2008; 10(5):118. Aletaha D, Stamm T, Kapral T, Eberl G, Grisar J, Machold KP et al. Survival and effectiveness of leflunomide compared with methotrexate and sulfasalazine in rheumatoid arthritis: a matched observational study. Ann Rheum Dis 2003; 62(10):944-51. Jones G, Halbert J, Crotty M, Shanahan EM, Batterham M, Ahern M. The effect of treatment on radiological progression in rheumatoid arthritis: a systematic review of randomized placebo-controlled trials. Rheumatology (Oxford) 2003; 42(1):6-13. Aletaha D, Landewe R, Karonitsch T, Bathon J, Boers M, Bombardier C et al. Reporting disease activity in clinical trials of patients with rheumatoid arthritis: EULAR/ACR collaborative recommendations. Arthritis Rheum 2008; 59(10):1371-7. Klareskog L, van der HD, 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(9410):675-81. Maini R, St Clair EW, Breedveld F, Furst D, Kalden J, Weisman M et al. Infliximab (chimeric antitumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. ATTRACT Study Group. Lancet 1999; 354(9194):1932-9. Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka K, van VR 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(1):26-37. 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(11):3432-43.. 20.   

(107)  

(108)

(109)

(110) . Chapter 1. 

(111)

(112)

(113) .

(114) (49). (50). (51). (52). (53). (54) (55). (56). (57). (58). (59). (60). (61). (62). Lipsky PE, van der Heijde DM, St Clair EW, 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(22):1594-602. 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(4):253-9. 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(1):35-45. 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(2):124-34. Hench PS, Kendall EC. The effect of a hormone of the adrenal cortex (17-hydroxy-11-dehydrocorticosterone; compound E) and of pituitary adrenocorticotropic hormone on rheumatoid arthritis. Mayo Clin Proc 1949;24:181-97. Lundberg IE, Grundtman C, Larsson E, Klareskog L. Best Pract Res Clin Rheumatol 2004;18:7-19. Saag KG, Koehnke R, Caldwell JR, Brasington R, Burmeister LF, Zimmerman B, Kohler JA, Furst DE. Low dose long-term corticosteroid therapy in rheumatoid arthritis: an analysis of serious adverse events. Am J Med 1994; 96: 115-23. Fardet L, Blanchon T, Perdoncini-Roux A, Kettaneh A, Tiev K, Turbelin C, Dorleans Y, Cabane J, Hanslik T. [Internal medicine physicians’ perception of frequency and impact of corticosteroid-induced adverse events] .Rev Med Interne 2009; 30:113-8. van Tuyl LH, Plass AM, Lems WF, Voskuyl AE, Kerstens PJ, Dijkmans BA, Boers M. Discordant perspectives of rheumatologists and patients on COBRA combination therapy in rheumatoid arthritis. Rheumatology (Oxford) 2008; 47: 1571-6. Fardet L, Flahault A, Kettaneh A, Tiev KP, Généreau T, Tolédano C, Lebbé C, Cabane J. Corticosteroid-induced clinical adverse events: frequency, risk factors and patient’s opinion. Br J Dermatol 2007; 157: 142-8. van Everdingen AA, Jacobs JW, Siewertsz Van Reesema DR, Bijlsma JW. Low-dose prednisone therapy for patients with early active rheumatoid arthritis: clinical efficacy, disease-modifying properties, and side effects: a randomized, double-blind, placebo-controlled clinical trial. Ann Int Med 02; 136: 1-12. van Tuyl LH, Boers M, Lems WF, Landewé RB, Han H, van der Linden S, van der Laar MA, Westhovens R, Van Denderen JC, Westedt ML, Peeters AJ, Jacobs P, Huizinga TW, van den Brink H, Dijkmans BA, Voskuyl AE van Tuyl LH. Survival, comorbidities and joint damage 11 years after the COBRA combination therapy trial in early rheumatoid arthritis. Ann Rheum Dis. 2009 May 17. Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, van Zeben D, Kerstens PJ, Hazes JM, Zwinderman AH, Peeters AJ, de Jonge-Bok JM, Mallée C, de Beus WM, de Sonnaville PB, Ewals JA, Breedveld FC, Dijkmans BA. Comparison of treatment strategies in early rheumatoid arthritis: a randomized trial. Ann Intern Med. 2007; 146(6): 406-15. Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, Kerstens PJ, Grillet BA, de Jager MH, Han KH, Speyer I, van der Lubbe PA, Seys PE, Breedveld FC, Dijkmans BA. Patient preferences for treatment: report from a randomised comparison of treatment strategies in early rheumatoid arthritis (BeSt trial). Ann Rheum Dis. 2007; 66(9): 1227-32.. Chapter 1.   

(115)  

(116)

(117)

(118) . 21. 

(119)

(120)

(121) .

(122) (63). (64). (65) (66) (67) (68) (69) (70) (71). (72). (73). (74). (75) (76). (77) (78) (79). (80). van der Bijl AE, Teng YK, van Oosterhout M, Breedveld FC, Allaart CF, Huizinga TW. Efficacy of intraarticular infliximab in patients with chronic or recurrent gonarthritis: a clinical randomized trial. Arthritis Rheum. 2009; 61(7): 974-8. van der Kooij SM, le CS, Goekoop-Ruiterman YP, de Vries-Bouwstra JK, van Zeben D, Kerstens PJ et al. Clinical and radiological efficacy of initial versus delayed treatment with infliximab plus methotrexate in patients with early rheumatoid arthritis. Ann Rheum Dis. 2009; 68(7): 1153-8. vdHout, Arthritis Care and Research, in press Soubrier M, Puechal X, Sibilia J, Mariette X, Meyer O, Combe B et al. Guepard study. American College of Rheumatology, 2008 annual scientific meeting, abstract 1640. Lejeune F, Lienard D, Eggermont A. Regional Administration of Recombinant Tumour Necrosis Factor-alpha in Cancer, with Special Reference to Melanoma. BioDrugs 1998; 9(3):211-8. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet 2001; 357(9255):53945. Beutler BA. The role of tumor necrosis factor in health and disease. J Rheumatol Suppl 1999; 57:1621. Feldmann M, Maini RN. Lasker Clinical Medical Research Award. TNF defined as a therapeutic target for rheumatoid arthritis and other autoimmune diseases. Nat Med 2003; 9(10):1245-50. Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, Macfarlane JD et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41(9):1552-63. Siegel SA, Shealy DJ, Nakada MT, Le J, Woulfe DS, Probert L et al. The mouse/human chimeric monoclonal antibody cA2 neutralizes TNF in vitro and protects transgenic mice from cachexia and TNF lethality in vivo. Cytokine 1995; 7(1):15-25. Barone D KCLDMKMK. Comparative analysis of the ability of etanercept and infliximab to lyse TNF-expressing cells in a complement dependent fashion [abstract]. Arthritis Rheum 1999, 42(suppl):S90. Hernandez-Cruz B, Cardiel MH, Villa AR, Cocer-Varela J. Development, recurrence, and severity of infections in Mexican patients with rheumatoid arthritis. A nested case-control study. J Rheumatol 1998; 25(10):1900-7. van der Veen MJ, van der HA, Kruize AA, Bijlsma JW. Infection rate and use of antibiotics in patients with rheumatoid arthritis treated with methotrexate. Ann Rheum Dis 1994; 53(4):224-8. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA 2006; 295(19):2275-85. Listing J, Strangfeld A, Kary S, Rau R, von HU, Stoyanova-Scholz M et al. Infections in patients with rheumatoid arthritis treated with biologic agents. Arthritis Rheum 2005; 52(11):3403-12. Bernatsky S, Hudson M, Suissa S. Anti-rheumatic drug use and risk of serious infections in rheumatoid arthritis. Rheumatology (Oxford) 2007; 46(7):1157-60. Curtis JR, Patkar N, Xie A, Martin C, Allison JJ, Saag M et al. Risk of serious bacterial infections among rheumatoid arthritis patients exposed to tumor necrosis factor alpha antagonists. Arthritis Rheum 2007; 56(4):1125-33. Schneeweiss S, Setoguchi S, Weinblatt ME, Katz JN, Avorn J, Sax PE et al. Anti-tumor necrosis factor alpha therapy and the risk of serious bacterial infections in elderly patients with rheumatoid arthritis. Arthritis Rheum 2007; 56(6):1754-64.. 22.   

(123)  

(124)

(125)

(126) . Chapter 1. 

(127)

(128)

(129) .

(130) (81). (82). (83) (84). (85) (86) (87) (88). (89). (90) (91) (92) (93) (94) (95). (96) (97). (98). (99). Dixon WG, Watson K, Lunt M, Hyrich KL, Silman AJ, Symmons DP. Rates of serious infection, including site-specific and bacterial intracellular infection, in rheumatoid arthritis patients receiving antitumor necrosis factor therapy: results from the British Society for Rheumatology Biologics Register. Arthritis Rheum 2006; 54(8):2368-76. Dixon WG, Symmons DP, Lunt M, Watson KD, Hyrich KL, Silman AJ. Serious infection following antitumor necrosis factor alpha therapy in patients with rheumatoid arthritis: lessons from interpreting data from observational studies. Arthritis Rheum 2007; 56(9):2896-904. Doran MF, Crowson CS, Pond GR, O’Fallon WM, Gabriel SE. Predictors of infection in rheumatoid arthritis. Arthritis Rheum 2002; 46(9):2294-300. Doran MF, Crowson CS, Pond GR, O’Fallon WM, Gabriel SE. Frequency of infection in patients with rheumatoid arthritis compared with controls: a population-based study. Arthritis Rheum 2002; 46(9):2287-93. Wallis RS, Broder MS, Wong JY, Hanson ME, Beenhouwer DO. Granulomatous infectious diseases associated with tumor necrosis factor antagonists. Clin Infect Dis 2004; 38(9):1261-5. Wallis RS, Broder M, Wong J, Beenhouwer D. Granulomatous infections due to tumor necrosis factor blockade: correction. Clin Infect Dis 2004; 39(8):1254-5. Winthrop KL. Risk and prevention of tuberculosis and other serious opportunistic infections associated with the inhibition of tumor necrosis factor. Nat Clin Pract Rheumatol 2006; 2(11):602-10. Askling J, Fored CM, Brandt L, Baecklund E, Bertilsson L, Coster L et al. Risk and case characteristics of tuberculosis in rheumatoid arthritis associated with tumor necrosis factor antagonists in Sweden. Arthritis Rheum 2005; 52(7):1986-92. Carmona L, Gomez-Reino JJ, Rodriguez-Valverde V, Montero D, Pascual-Gomez E, Mola EM et al. Effectiveness of recommendations to prevent reactivation of latent tuberculosis infection in patients treated with tumor necrosis factor antagonists. Arthritis Rheum 2005; 52(6):1766-72. Wallis RS. Tumour necrosis factor antagonists: structure, function, and tuberculosis risks. Lancet Infect Dis 2008; 8(10):601-11. Randhawa PS. Lymphocyte subsets in granulomas of human tuberculosis: an in situ immunofluorescence study using monoclonal antibodies. Pathology 1990; 22(3):153-5. Hamilton CD. Tuberculosis in the cytokine era: what rheumatologists need to know. Arthritis Rheum 2003; 48(8):2085-91. NVR richtlijn screening tuberculose bij antiTNF. june 2003 Nicholson KG, Wood JM, Zambon M. Influenza. Lancet 2003; 362(9397):1733-45. Bridges CB, Winquist AG, Fukuda K, Cox NJ, Singleton JA, Strikas RA. Prevention and control of influenza: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2000; 49(RR-3):1-38. Sands BE, Cuffari C, Katz J, Kugathasan S, Onken J, Vitek C et al. Guidelines for immunizations in patients with inflammatory bowel disease. Inflamm Bowel Dis 2004; 10(5):677-92. Kaine JL, Kivitz AJ, Birbara C, Luo AY. Immune responses following administration of influenza and pneumococcal vaccines to patients with rheumatoid arthritis receiving adalimumab. J Rheumatol 2007; 34(2):272-9. Gelinck LB, van der Bijl AE, Beyer WE, Visser LG, Huizinga TW, van Hogezand RA et al. The effect of anti-tumour necrosis factor alpha treatment on the antibody response to influenza vaccination. Ann Rheum Dis 2008; 67(5):713-6. Fomin I, Caspi D, Levy V, Varsano N, Shalev Y, Paran D et al. Vaccination against influenza in rheumatoid arthritis: the effect of disease modifying drugs, including TNF alpha blockers. Ann Rheum. Chapter 1.   

(131)  

(132)

(133)

(134) . 23. 

(135)

(136)

(137) .

(138) Dis 2006; 65(2):191-4. (100) Kapetanovic MC, Saxne T, Nilsson JA, Geborek P. Influenza vaccination as model for testing immune modulation induced by anti-TNF and methotrexate therapy in rheumatoid arthritis patients. Rheumatology (Oxford) 2007; 46(4):608-11. (101) Kollias G, Douni E, Kassiotis G, Kontoyiannis D. The function of tumour necrosis factor and receptors in models of multi-organ inflammation, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease. Ann Rheum Dis 1999; 58 Suppl 1:I32-I39. (102) TNF neutralization in MS: results of a randomized, placebo-controlled multicenter study. The Lenercept Multiple Sclerosis Study Group and The University of British Columbia MS/MRI Analysis Group. Neurology 1999; 53(3):457-65. (103) Mohan N, Edwards ET, Cupps TR, Oliverio PJ, Sandberg G, Crayton H et al. Demyelination occurring during anti-tumor necrosis factor alpha therapy for inflammatory arthritides. Arthritis Rheum 2001; 44(12):2862-9. (104) Simsek I, Erdem H, Pay S, Sobaci G, Dinc A. Optic neuritis occurring with anti-tumour necrosis factor alpha therapy. Ann Rheum Dis 2007; 66(9):1255-8. (105) Robinson WH, Genovese MC, Moreland LW. Demyelinating and neurologic events reported in association with tumor necrosis factor alpha antagonism: by what mechanisms could tumor necrosis factor alpha antagonists improve rheumatoid arthritis but exacerbate multiple sclerosis? Arthritis Rheum 2001; 44(9):1977-83. (106) Magnano MD, Robinson WH, Genovese MC. Demyelination and inhibition of tumor necrosis factor (TNF). Clin Exp Rheumatol 2004; 22(5 Suppl 35):S134-S140. (107) Hyrich KL, Silman AJ, Watson KD, Symmons DP. Anti-tumour necrosis factor alpha therapy in rheumatoid arthritis: an update on safety. Ann Rheum Dis 2004; 63(12):1538-43. (108) Maes M. The cytokine hypothesis of depression: inflammation, oxidative & nitrosative stress (IO&NS) and leaky gut as new targets for adjunctive treatments in depression. Neuro Endocrinol Lett 2008; 29(3):287-91. (109) Raison CL, Capuron L, Miller AH. Cytokines sing the blues: inflammation and the pathogenesis of 8; 12(2):171-83.. 24.   

(139)  

(140)

(141)

(142) . Chapter 1. 

(143)

(144)

(145) .

(146)

Referenties

GERELATEERDE DOCUMENTEN

This prospective open-label pilot study evaluated the effectiveness and safety of ada- limumab and the relationship to antibodies against infliximab (IFX) in adult patients with

Seven patients with a systemic inflammatory disease (5 rheumatoid arthritis, 2 psoria- tic arthritis) had Magnetization Transfer Imaging, Diffusion Weighted Imaging (DWI) and

The present study demonstrates adequate protection rates against influenza after influenza vaccination in patients treated with anti-TNF, despite lower postvaccination antibody

The main finding of the present study is the synergistic immunosuppressive effect of combined methotrexate and anti-TNF use on the antibody response upon pneumo- coccal

Patients who required continued infliximab treatment achieved low disease activity, but it was not as persistent as for the Responders, and possibly as a consequence, they had more

Successful treatment of refractory mononeuritis multiplex secondary to rheumatoid arthritis with the anti-tumour necrosis factor alpha monoclonal antibody infliximab.

hoofdstuk 4: het effect van anti-TNF op de effectiviteit van griep vaccinatie Richtlijnen adviseren griepvaccinatie bij immuungecompromitteerde patiënten, zoals bij het gebruik

Alle reumatologen (in opleiding) van de afdeling reumatologie van het LUMC en verwijzende reumatologen uit andere klinieken voor het doorsturen van patiënten en de reumatologen van