Next Article in Journal
Deciphering the Metabolic Pathways of Pitaya Peel after Postharvest Red Light Irradiation
Next Article in Special Issue
The Lipolysome—A Highly Complex and Dynamic Protein Network Orchestrating Cytoplasmic Triacylglycerol Degradation
Previous Article in Journal
Antifungal Drugs
Previous Article in Special Issue
TGF-ß1 Induces Changes in the Energy Metabolism of White Adipose Tissue-Derived Human Adult Mesenchymal Stem/Stromal Cells In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Interrelations between Biological and Targeted Synthetic Agents Used in Inflammatory Joint Diseases, and Obesity or Body Composition

by
Eric Toussirot
1,2,3,4,5
1
INSERM CIC-1431, CHU de Besançon, Centre d’Investigation Clinique Biothérapie, Pôle Recherche, 25000 Besançon, France
2
Fédération Hospitalo-Universitaire INCREASE, CHU de Besançon, 25000 Besançon, France
3
CHU de Besançon, Rhumatologie, Pôle PACTE (Pathologies Aiguës Chroniques Transplantation Éducation), 25000 Besançon, France
4
Département Universitaire de Thérapeutique, Université de Bourgogne Franche-Comté, UFR des Sciences Médicales et Pharmaceutiques de Besançon, CS 71806, 25030 Besançon Cedex, France
5
INSERM UMR1098, Relations Hôte Greffon Tumeurs, ingénierie cellulaire et génique, Université de Bourgogne Franche-Comté, 25000 Besançon, France
Metabolites 2020, 10(3), 107; https://doi.org/10.3390/metabo10030107
Submission received: 2 February 2020 / Revised: 8 March 2020 / Accepted: 10 March 2020 / Published: 13 March 2020
(This article belongs to the Special Issue Adipose Tissue and Metabolic Health)

Abstract

:
Obesity is a comorbidity that plays a role in the development and severity of inflammatory joint diseases, including rheumatoid arthritis, psoriatic arthritis and axial spondyloarthritis. The relationships between obesity and adipose tissue and the treatments given for inflammatory joint diseases are bidirectional. In fact, biological agents (bDMARDs) and targeted synthetic agents (tsDMARDs) may influence body weight and body composition of treated patients, while obesity in turn may influence clinical response to these agents. Obesity is a prevalent comorbidity mainly affecting patients with rheumatoid arthritis (RA) and psoriatic arthritis (PsA) with specific phenotypes. Tumour necrosis factor alpha (TNFα) inhibitors have been associated with changes in body composition by improving lean mass, but also by significantly increasing fat mass, which localized toward the abdominal/visceral region. The IL-6 inhibitor tocilizumab is associated with an increase in lean mass without change in fat mass. The clinical response to TNFα inhibitors is attenuated by obesity, an effect that is less pronounced with IL-6 inhibitors and the B-cell depletion agent rituximab. Conversely, body weight has no influence on the response to the costimulation inhibitor abatacept. These effects may be of help to the physician in personalized medicine, and may guide the therapeutic choice in obese/overweight patients.

1. Introduction

Inflammatory joint diseases (IJD) are complex disorders characterized by chronic inflammation of the joints and related skeletal structures. They comprise a wide range of disorders of which rheumatoid arthritis (RA) and spondyloarthritis (SpA) are the most common.
Rheumatoid arthritis (RA) is a systemic immune-mediated disease with chronic symmetrical joint inflammation, leading to progressive clinical deformations, erosive radiographic changes and disability. Pro-inflammatory cytokines such as IL-1β, TNFα and IL-6, but also IL-17A play a major role in the joint inflammation of RA [1]. Spondyloarthritis encompasses a group of inflammatory disorders mainly affecting the axial skeleton, i.e., the spine and the sacroiliac joints, but also specific anatomical structures such as the entheses. Peripheral arthritis may also be observed in SpA. According to the main skeletal involvement, SpA is classified as axial or peripheral disease. The most common forms of SpA are ankylosing spondylitis (AS), the prototypical form of axial SpA (ax-SpA), and psoriatic arthritis (PsA). Shared clinical manifestations in SpA are axial joint inflammation, enthesitis, dactylitis and peripheral oligoarthritis. Axial disease in SpA may lead to ligamentous ossifications, reduced spinal mobility and thus progressive ankylosis and disability [2]. TNFα and IL-17A have been identified as key mediators driving inflammation in ax-SpA, while IL-23 and IL-17A, together with TNFα are involved in PsA [3].
Extra-articular features may also be observed in RA and SpA. In RA, systemic inflammation can affect different organs including the eyes, skin, lungs and kidneys. However, it is well established that the cardiovascular (CV) system is predominantly involved in RA with the development of atherosclerotic CV disease [4]. This increased CV risk may be explained by the combination of traditional CV risk factors, but also by the influence of systemic inflammation. In SpA, extra-articular involvement involves the eyes (acute anterior uveitis), the skin (psoriasis) and the bowels (inflammatory bowel diseases [IBD]). However, the CV system is also often affected, and like RA, ax-SpA and PsA are also characterized by a high mortality rate linked to CV comorbidities [5]. Systemic inflammation also plays a predominant role in this enhanced CV risk.
Inflammatory joint diseases are further characterized by obesity and changes in body composition. The interest garnered by obesity in IJD can be explained by the influence of obesity on the development of arthritis, but also by the contribution to systemic inflammation that adipose tissue may provide [6]. In turn, adipose tissue may impact on the therapeutic response to specific targeted drugs that are commonly used in the management of these diseases, i.e., biological disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs).
In this review, we aim to discuss the interrelations between obesity, fat mass/adipose tissue and the specific treatments that are used in clinical practice in RA and SpA. We will also discuss the influence that biological and targeted synthetic drugs may have on body composition and conversely, the role that body mass index (BMI) may play in therapeutic response to these agents. The influence of weight or BMI on the response to conventional synthetic (cs) DMARDs is not examined in this review.

2. Obesity, Body Composition and Inflammatory Joint Diseases

Obesity and overweight are defined by the World Health Organization (WHO) as abnormal or excessive fat mass that presents a risk to health. Body mass index is the most common anthropometric measurement to assess and define obesity. According to this definition, BMI categorizes subjects into 3 groups: normal weight (BMI < 25 kg/m2), overweight (BMI between 25 and 30 kg/m2) and obesity (BMI > 30 kg/m2). However, BMI gives an evaluation of total weight, but does not distinguish between fat and lean mass, and thus, is an approximation of fat percentage. In addition, there are different phenotypes of obesity, the most commonly described being android and gynoid obesity. Waist circumference is a valid measurement for abdominal, or android, obesity, a fat distribution strongly associated with CV complications [7]. Body mass index is also influenced by several parameters, including age, gender, ethnicity and inflammation, as observed in IJD [8]. Fat mass, and other tissues, namely skeletal muscle, solid organs, skin and bone, collectively termed fat-free mass or lean mass, may be evaluated by specific techniques, given body composition measurements. The two methods that are widely used for the measurement of body composition compartments are bioelectric impedance and dual X-ray absorptiometry (DEXA) [8]. DEXA is more accurate and thus considered to be the reference technique for body composition measurements. In addition, DEXA provides measurements of total lean and fat mass, as well as the precise anatomical localisations, such as the android and gynoid regions [9].
Body composition is altered in IJD. In RA, a condition with systemic inflammation and release of pro-inflammatory cytokines with catabolic properties, a loss of body cell mass and lean mass was initially reported under the term “rheumatoid cachexia” [10]. Body cell mass includes muscle mass and visceral mass as well as immune cells. Loss of body cell mass in RA has been estimated at between 13% and 15%, therefore influencing muscle strength, energy metabolism and susceptibility to infections. TNFα has been identified as a key mediator for body mass depletion [10]. Initial reports focused on depletion of lean body mass and cachexia but more recent research has drawn attention to excess fat mass or obesity in RA [6,8]. According to the WHO definition, obesity was reported to be prevalent in 18–31% of patients and overweight in 60% of patients [6]. In fact, loss of body cell mass is usually accompanied by increased fat mass and thus stable or slightly increased body weight, leading to a phenotype of cachectic obesity in RA. Abnormal body composition, characterized by lean body mass deficit and fat mass excess has been described in patients with RA. Body composition measurements by DEXA in patients with RA showed that fat distribution was altered, with a clear shift to the abdominal region and a parallel decrease in lean mass [11]. The parallel loss of skeletal muscle and increase in fat defines body composition phenotypes such as sarcopenia, overfat and sarcopenic obesity. The different studies evaluating body composition in RA found that such phenotypes were very common in patients with long disease duration [12]. They showed abnormal values for all measures of adiposity, including abdominal fat. In a case-control study, we reported that women with RA had increased visceral (intra-abdominal) adipose tissue [13], a fat accumulation that is strongly associated with adverse CV events [7].
Obesity in SpA has been less widely investigated. A recent study performed in the Netherlands found that obesity was prevalent in 22% of patients, and overweight in 37% [14]. Conversely, the link between obesity and psoriasis, with or without arthritis, is well established: obesity is a factor that plays a role in the development of psoriasis and contributes to the severity of this skin disease [15]. Several studies have consistently underlined a significant link between BMI and psoriasis, while additional studies provide some evidence that increased adiposity is a risk factor for incident psoriasis [16]. An increased prevalence of obesity has also been reported in PsA as compared to the general population. The prevalence of obesity was found to be higher in patients with PsA compared to patients with psoriasis [17], and even higher than that observed in RA and psoriasis (respectively 37%, 27% and 29%) [18]. There is additional evidence linking obesity to the risk of incident PsA [19]. Collectively, obesity is associated with both psoriasis and PsA and is considered to be a contributing factor to the development of both diseases. A limited number of studies have evaluated body composition in patients with psoriasis using DEXA. In a series of Brazilian patients with psoriasis, total body fat was similar between psoriatic patients and healthy controls. In that study, patients with PsA had more evidence of adiposity with a significantly higher body fat percentage than psoriatic patients and healthy controls. In these patients, the excess of fat tissue was located in the android region [20]. Using bio-impedance, post-menopausal women with PsA were characterized by sarcopenia and increased total fat mass [21].

3. Influence of bDMARDs and tsDMARDS on Body Composition in Inflammatory Joint Diseases

We are now in the era of targeted drugs for the treatment of many immune-mediated diseases including IJD. These agents may be classified as biological agents (bDMARDs) or targeted synthetic agents (tsDMARDs). bDMARDs control the biological properties of specific pro-inflammatory cytokines (TNFα, IL-6, IL-17A, IL-23 or IL-1) or the function of a specific cellular subset (B lymphocyte or T lymphocyte co-stimulation). Available agents are TNFα inhibitors (TNFi: infliximab, etanercept, adalimumab, certolizumab and golimumab), a B-cell depletion agent (rituximab), a co-stimulation blocking agent (abatacept), IL-6 inhibitors (IL-6i: tocilizumab and sarilumab), IL-17A inhibitors (IL-17Ai: secukinumab and ixekizumab) and an IL-23p19 inhibitor (IL-23p19i: ustekinumab) [22]. Beside bDMARDs, tsDMARDs were recently introduced on the market, and especially, kinase inhibitors that have been licensed in the treatment of different immune-mediated diseases. Targeted synthetic DMARDs differ from csDMARDs by the selectivity of their action, targeting specific signaling pathways. The Janus kinase family is a small family of receptor-associated tyrosine kinases that are essential for the signal cascade of type I and type II cytokine receptors, and thus, Janus kinase inhibitors (JAKi) block the cytokine-mediated signaling pathway [23]. JAKi controls the production of several cytokines involved in immune reaction and they have proven to be highly effective in the treatment of RA and PsA. Currently, two JAKi are available, namely tofacitinib and baricitinib [24]. In RA and PsA, both bDMARDs (TNFi, IL-6i, rituximab and abatacept for RA; TNFi, IL-17Ai, IL-23p19i for PsA) and tsDMARDs (JAKi: tofacitinib and baricitinib for RA; tofacitinib for PsA) are very effective in the control of joint inflammation and progression of the disease. In ax-SpA, only bDMARDs (TNFi and IL-17Ai) are approved, leading to effective control of joint symptoms and certain extra-articular manifestations. These agents control the inflammatory process and the biological functions of specific cytokines with catabolic properties (for instance, TNFα), and may therefore potentially influence weight, BMI and/or body composition.

3.1. bDMARDs

3.1.1. TNF Inhibitors

Initial reports showed that patients with IBD or psoriasis gained weight while receiving TNFi [25,26]. The weight gain ranged between 1.5 and 3 kg, and rarely exceeded 5 kg [25]. Weight gain was observed during the first six months of treatment (but as early as 4 weeks) and described with the initially approved TNFi, infliximab and etanercept, then with adalimumab. In parallel, BMI also increased. This phenomenon was then described in different immune-mediated diseases, RA, ax-SpA and PsA (Table 1).
Numerous studies have examined the impact of TNFi on body composition in IJD (Table 1) [27,28,29,30,31,32,33,34,35,36,37,38]. The question of weight/BMI/body composition changes during TNFi was examined in patients with RA in 7 studies [27,28,29,30,31,32,33], in patients with SpA in 3 studies [35,36,37] and in patients with PsA in one study [38]. In one study, patients had RA or SpA [34]. Only two studies were randomized trials [27,30] while the others were open-labeled [28,29,31,32,33,34,35,36,37,38]. Collectively, a weight gain/BMI increase was reported by 6 studies [31,32,33,35,36,38], while fat mass increase was described in 7 [28,30,31,34,36,37,38]. No changes in fat mass were reported in 3 studies [27,34,36] with or without a parallel increase in lean mass [27,35,36]. In a randomized study comparing the effects of methotrexate (MTX), a csDMARD, and etanercept on body composition, there was no overall change in weight, BMI or body composition. However, in the etanercept group, 6/13 patients gained weight that was explained by an increase in lean mass [27]. Conversely, the study by Engvall et al. described increased body fat mass in patients with early RA while receiving infliximab while no similar changes were observed in the control group under csDMARDs [30]. Finally, a prospective observational study followed patients with RA starting a TNFi or MTX. After 24 months of therapy, weight gain was observed only among patients treated by TNFi [33].

3.1.2. IL-6 Inhibitors

There are a limited number of studies examining changes in weight, BMI and body composition with IL-6i (Table 2) [39,40,41,42]. Tocilizumab, an anti-IL-6 receptor, was associated with weight gain during randomized controlled trials, and this has been confirmed in observational studies [39]. Three studies examined changes in body composition during TCZ treatment in patients with RA. One study with a short (3 month) follow-up found no variation in body composition parameters [40]. On the contrary, the two other studies reported an increase in weight and BMI that was explained by a significant increase in lean mass. Conversely, no change in fat mass was observed in these studies [41,42].

3.1.3. Abatacept and Rituximab

There is no specific study examining this question, but neither abatacept nor ritixumab was associated with bodyweight changes during clinical trials. Bodyweight, BMI, and waist circumference did not change under abatacept IV given for 6 months in 15 patients with RA [43]. A study evaluated the changes in BMI in patients with antineutrophil cytoplasmic antibody-associated vasculitis while receiving RTX. Patients received 4 weekly infusions of RTX 375 mg/m2 followed by a placebo for a period of 18 months. The results showed that BMI significantly increased at 6 months (+1.1 ± 2.2 kg/m2), then remained stable without returning to baseline values. However, this increase in BMI was multifactorial and resulted from the improvement of disease activity, but also from glucocorticoid exposure and RTX administration. It was concluded that RTX also had effects on BMI independently of its impact on disease activity [44]. No similar data are available in patients with RA.

3.1.4. Anti-IL-23 and Anti-IL-17A Agents

Ustekinumab, an anti-IL-23 p19i, has been associated with weight changes in patients with psoriasis. Indeed, this treatment increased BMI by 3.5% [45]. However, results are conflicting. In a prospective study comparing the effects of infliximab, a TNFi, and ustekinumab on body weight and BMI, only infliximab was associated with changes in these parameters [46]. DEXA assessment during ustekinumab treatment was not available, but a study evaluated body composition using bioelectrical impedance: 53 patients with psoriasis were evaluated at baseline and at 6 and 12 months. Decreases in body weight, BMI and fat mass were observed at month 6, while there was an increase at month 12 in body cell mass and phase angle, two indicators of nutritional status [47]. Secukinumab was not associated with changes in body weight or BMI in patients with psoriasis followed for 7 months after initiating the treatment [48]. However, a substantial weight gain (+10 kg) was reported in a patient with type 2 diabetes and PsA after 3 months of secukinumab treatment [49].

3.2. tsDMARDs

The effects of tofacitinib on anthropometric measurements and body composition have rarely been examined. In female mice, tofacitinib had no influence on body weight or body composition [50]. In a small series of female patients with RA (N = 4), it was also observed that tofacitinib (5 mg twice a day) did not affect body weight, while fat mass (evaluated by bioelectric impedance) slightly increased after 3 months of treatment [50]. In a larger series of patients with RA (N = 31) followed for 12 months after starting tofacitinib, a weight increase was observed (+4.2%), while BMI did not change significantly. Interestingly, this study showed that the visceral adiposity index, a mathematical model of visceral adipose tissue estimation, decreased during tofacitinib treatment [51]. There is no available study examining the changes in body composition under tofacitinib or baricitinib using DEXA.

4. Influence of Weight or Body Mass Index on the Therapeutic Response to bDMARDs or tsDMARDs Used to Treat Inflammatory Joint Diseases

The association with obesity concerns different IJD [6], but this comorbidity mainly affects patients with PsA [52]. The impact of obesity, and body weight in general, on the therapeutic response to conventional, but also targeted drugs is a relevant issue [53]. The first data on the potential effects of obesity on the therapeutic response to bDMARDs came from psoriatic patients and showed that obese patients had a poor skin response to TNFi [54]. This question was further examined in different studies involving patients with RA and SpA, including PsA (Table 3 and Table 4). The therapeutic response to bDMARDs according to BMI was mainly examined with TNFi, and less with other bDMARDs. Limited data have been published with tsDMARDs. Results stem from retrospective studies, observational cohort studies or alternatively, from a limited number of randomized controlled trials.

4.1. bDMARDs in Rheumatoid Arthritis

4.1.1. TNFi

Results are available for infliximab, etanercept, and to a lesser extent, for adalimumab and the other TNFi (Table 3). Patients had established disease except in one study [55].
The duration of follow-up varied from 4 to 12 months. Reported clinical outcomes were remission according to different definitions (disease activity score-28 joints (DAS-28), simplified disease activity index [SDAI] or clinical disease activity index (CDAI)), variation in DAS28 score, or time to discontinuation of the drug. All together, published studies show that in RA, obesity was present in 10% to 40%, and overweight in 32% to 57% [55,56,57,58,59,60,61]. There was clearly a worse clinical response to the different TNFi among obese or overweight patients compared to patients in the normal BMI group. This effect was independent of the route of administration of the biological drug (intravenously [IV] or subcutaneously [SC]) and observed for both fixed (etanercept and adalimumab) and weight-based doses (infliximab). The impaired response to TNFi in overweight/obese subjects was described regardless of the clinical outcome (remission or variation in DAS28). In one study, the lowest percentage of remission was observed with infliximab compared to etanercept or adalimumab [58]. One study examined the drug retention of a second line bDMARD following the failure of the first TNFi in obese patients. This study showed that the survival of the first TNFi was lower in obese (39.4%) compared to non-obese patients (49.1%). However, for patients who started a second non-TNFi biological drug, the persistence on therapy at a median period of 57 months was statistically lower in obese (43.5%) than in non-obese patients (80%). In that study, obese status was identified as a risk factor for discontinuing TNFi as first-line (Hazard Ratio (HR) and 95% CI: 1.64 (1.02–2.62)) and as second-line biological drug (HR: 2.9 1.08–8.45)) [60]. Only one study did not report a loss of efficacy of TNFi in obese patients. This was an analysis from the Veterans Affairs administrative database of US veterans with RA. In this large retrospective cohort including 23,669 subjects with RA, severe obesity (defined as a BMI > 35 kg/m2) was not associated with treatment discontinuation of the first SC administered TNFi, after adjustment for important covariables. On the contrary, patients with low (<20 kg/m2) or normal (20–25 kg/m2) BMI were more likely to discontinue a TNFi (and hydroxychoroquine and MTX as well) compared to overweight patients [61].
Data are more limited for the other bDMARDs.

4.1.2. IL-6i

Two retrospective studies did not show any influence of body weight/BMI on the therapeutic response to tocilizumab (Table 3) [62,63].

4.1.3. B- Cell Depletion Agent

For rituximab, a retrospective study also found no influence of BMI on the clinical outcome as evaluated by the decrease in DAS28 or percentage of good responders and remission [64]. However, in the study by Iannone et al., obese patients receiving rituximab as second-line bDMARD responded less well to this agent compared to normal-weight patients (Table 3) [60].

4.1.4. Abatacept

Data stem from two retrospective studies and large prospective phase IV trials or registries (Table 3) [65,66,67,68,69]. Taken together, these studies showed that body weight or BMI did not influence response to abatacept given by the SC or IV route.

4.2. bDMARDs in Spondyloarthritis and Psoriatic Arthritis

In ax-SpA, analysis of the therapeutic response according to body weight was limited to TNFi (Table 4). Four retrospective studies were published, and as for RA, the therapeutic response (as evaluated by Ankylosing Spondylitis Disease Activity Score (ASDAS) or rate of Assessment of Spondyloarthritis Society (ASAS) responders) was impaired in obese or overweight patients [70,71,72,73]. Results were mainly given for infliximab. The duration of the studies ranged from 6 to 12 months.
Four studies examined therapeutic response in patients with PsA taking into account their body weight/BMI (Table 4) [74,75,76,77]. Clinical outcomes were the achievement of minimal disease activity (MDA), DAS28 (or SDAI) response and the European League Against Rheumatism (EULAR) response. The duration of follow-up was 6 to 36 months. As previously observed in RA and ax-SpA, BMI influenced the outcome, with fewer responders in the obese category. However, in one study, the disease remission rate based on DAS28 or SDAI did not differ between obese, overweight and normal-weight patients [75].
For ustekinumab, the IL-23p19i, data from psoriatic patients showed that obesity impaired the skin response as evaluated by the psoriasis area severity index (PASI) [78,79]. Thus, the recommended dose for patients weighing 100 kg or more has been fixed at 90 mg per injection [80]. There are no data on the efficacy of ustekinumab and secukinumab in patients with PsA according to body weight. In a pooled analysis of phase III trials in psoriasis evaluating secukinumab, response rates were analyzed by body-weight quartiles. The results showed that responses remained meaningful in all weight groups, although there was a trend towards a lower response by increasing body weight [81,82]. However, secukinumab 300 mg dose demonstrated a greater benefit than 150 mg dose.
Abatacept is also licensed for the treatment of PsA. In a post hoc analysis of phase III trials, it was concluded that BMI did not affect clinical or radiographic response to SC abatacept in patients wih PsA [83].
4.3/data on bDMARDs response from meta-analysis and registry: Two meta-analyses examined the association between obesity and response to TNFi or different bDMARDs in IJD [84,85]. The first was based on 54 cohorts and included patients with various immune-mediated diseases, such as IJD but also psoriasis and IBD. Only the response to TNFi was analyzed. This analysis showed that patients in the obese category had a 60% higher risk of failing therapy (Odds Ratio (OR) (95% CI):1.6 (1.39–1.83)). A dose-response relationship was observed, i.e., the higher the body weight, the lower the response. These effects were observed in the different immune-mediated diseases, except for IBD [84]. The second meta-analysis examined the efficacy of all bDMARDs used in IJD and was based on 24 articles, including 10 on RA, 4 on ax-SpA, 2 on PsA and 8 on psoriasis and Crohn’s disease. This meta-analysis showed that the odds of achieving a good response or remission were lower in obese than in non-obese patients who were treated by TNFi (good response in RA, OR: 0.34 (0.18–0.64); remission in RA: OR: 0.36 (0.21–0.59); Bath Anlylosing Spondylitis Disease Activity Index 50 (BASDAI50) in ax-SpA: OR 0.41 (0.21–083)). No significant difference between obese and non-obese patients was found for those treated by abatacept or tocilizumab [85].
Lastly, data from the German biologic registry RABBITT were recently published. This registry included data from 10,593 patients with RA and the influence of obesity was analyzed after adjustment for potential confounders. Obesity was found to have a negative effect on the improvement of DAS28 for women receiving a csDMARD or a TNFi, but also for women and men receiving tocilizumab. Conversely, obesity did not impact on treatment efficacy with rituximab and abatacept [86].
Table 4. Response to bDMARDs in obese/overweight patients with spondyloarthritis.
Table 4. Response to bDMARDs in obese/overweight patients with spondyloarthritis.
Author (Reference)Number of SubjectsDiseaseSex Ratio M/F (%)Disease Duration (Years)Obese/Overweight Subjects (%)bDMARDsOutcomeMain Results
Ottaviani [70]155AS63.3/36.78overweight: 35
obese: 25
IFX (5 mg/kg)BASDAI50 at month 6Fewer responders in obese/overweight groups
Gremese [71]170ax-SpA69.4/ 30.616.3overweight: 32.4
obese: 13.5
IFX (5 mg/kg) or ETA or ADABASDAI 50 month 6Rate of responders lower in obese and overweight patients
Micheroli [72]624ax-SpA62.2/ 37.813overweight: 32.7
obese: 14.1
all TNFiRate of ASAS40 responders at one yearRate of responders lower in obese and overweight patients
Ibanez Vodnizza [73]41AS61/3914.6overweight: 36.6
obese: 12.2
ETA or ADABASDAI or ASDAS-CRP change at month 6Higher body fat associated with worse response to TNFi
Di Minno [74]270PsA62/389.2obese: 50IFX (5 mg/kg)
ETA or ADA
MDA at month 12Rate of MDA < obese patients vs non obese
Iannone [75]135PsA50.4/49.6NDoverweight: 34.8
obese: 33
IFX (5 mg/kg)
ETA or ADA
DAS28 or SDAI responseNo difference in rate of remission according to DAS28 or SDAI
Eder [76]557PsA58.4/41.615overweight: 36.2
obese: 35.4
TNFi without precisionMDA at month 12Less MDA in obese category
Hojgaard [77]1943PsA44.5/55.54obese: 34.6all TNFiEULAR response at month 6EULAR response lower in the obese category
Mc Innes [83]422PsAfemale % according to BMI categories: - placebo: 41% to 64%
abatacept: 42.9 to 67.7%
NAPlacebo group:
overweight: 27.1%
obese: 54.3%
Abatacept group:
overweight: 36.3%
obese: 49%
abatacept SCACR20no difference in the rate of responders between obese/overweight and normal weight patients
(TNF inhibitor, IL-23 inhibitor and IL-17A inhibitor) in axial spondyloarthritis and psoriatic arthritis according to body weight or body mass index (M: male; F: female; BMI: body mass index; bDMARD: biological disease-modifying antirheumatic drug; TNFi: TNF inhibitor; IFX: infliximab; ETA: etanercept; ADA: adalimumab; AS: ankylosing spondylitis; SpA: spondyloarthritis; ax-SpA: axial spondyloarthritis; PsA: psoriatic arthritis; BASDAI: Bath Ankylosing Spondylitis Disease Activity Index; ASAS: Assessment of SpondyloArthritis Society; EULAR: European League Against Rheumatism; MDA: minimal disease activity; ACR: American College of Rhematology; NA: not available).

4.3. tsDMARDs

In a post hoc analysis of baricitinib trials in RA, certain baseline patient characteristics were identified to have an effect on the drug response. They included specific ethnic group (non-Asian and non-white), negativity for rheumatoid factors and anti-citrullinated protein antibodies but also weight more than 100 Kg [87].

5. Discussion

The treatments that are routinely given in IJD may have specific consequences on body weight, BMI or body composition. This impact is well described with certain bDMARDs, and less so with tsDMARDs. The most demonstrative data are well illustrated by weight gain, and BMI and fat mass changes observed under TNFi. This effect is observed with the different TNFi (albeit mainly with infliximab) and in the different forms of IJD [88]. Weight gain is predominantly explained by a gain of fat that localized toward the android/abdominal (visceral) region [35,36]. In parallel, there was also a gain of lean mass, which is less sizable [28]. TNFα is a catabolic cytokine that plays a crucial role in the development of cachexia, a state that is well described in RA [10]. Indeed, TNFα, in concert with other pro-inflammatory cytokines, induces proteolysis and lipolysis. The administration of TNFi thus controls the negative effects of TNFα on cell mass and muscle mass. In this sense, the randomized trial by Marcora et al. clearly demonstrated a specific effect of TNFα inhibition on fat-free mass [27]. The increase in lean mass is modest and can be explained by the control of inflammation and disease activity, resulting in improved well-being and physical activity. Modulation of appetite and gut/growth hormones may also play a role [89]. The predominant and significant gain of fat mass under TNFi may also be explained by the control of inflammation and disease activity. The fat mass increase was small, but significant and located specifically towards the abdominal/visceral region, a type of adipose tissue strongly associated with enhanced CV risk [7]. This effect is drug-specific, since it was not observed with a csDMARD (MTX) in the study by Engvall et al. [30]. Indeed, visceral fat mass is associated with metabolic complications, atherosclerosis development and adverse CV events [7]. These changes in body composition during TNFα inhibition with the increase in abdominal/visceral fat raised questions about the ultimate effect on CV risk in patients receiving this class drug. However, reassuring data have now emerged regarding the long term protective effects of TNFi on CV diseases [88,89,90]. Indeed, it is thought that the effective control of disease activity and thus of chronic inflammation in IJD have a favorable impact on the CV prognostic of the patients. This is well suggested by the reduced risk of CV events for patients with RA under TNFi [91].
The IL-6i tocilizumab has been associated with a significant gain in lean mass without modification of fat, counteracting the process of cachexia in patients with RA [41,42]. This effect may be explained by the specific properties of IL-6 on muscle physiology. Thus, IL-6 inhibition may have anabolic effects on myocytes [92]. IL-6, in conjunction with TNFα may promote muscle loss and is associated with a decline in muscle mass and muscle strength, especially in older people. Indeed, elevated serum IL-6 has been associated with frailty and physical function in the aging population [93]. In addition, muscle atrophy has been observed in IL-6 transgenic mice and this was reversed by IL-6 receptor blockade [94].
Co-stimulatory inhibition with abatacept does not induce changes in weight/BMI, while for the B-cell depletion agent rituximab, the question remains open. Indeed, in vasculitis, this latter agent was associated with an increase in BMI that may be multifactorial [44]. For the IL-23/th17 pathway and related cytokine inhibitors, both ustekinumab and anti-IL-17Ai seem to have neutral effects on weight and BMI, but more detailed evaluations of body composition with DEXA are warranted.
The JAKi tofacitinib induces weight gain and seems to have a beneficial effect on fat mass, by reducing visceral adipose tissue [51]. However, specific studies examining body composition and fat mass under the different JAKi, tofacitinib and baricitinib, are required.
In parallel, obesity has a clear impact on disease outcomes in IJD, and on the therapeutic response to various treatments. bDMARDs are used at a weight adapted-dose only for those given by the IV route (infliximab, tocilizumab or abatacept), while for subcutaneously administered biologics, a fixed-dose is recommended. However, for ustekinumab and golimumab, there are recommendations to increase the drug dosage (respectively from 45 to 90 mg and from 50 to 100 mg) for patients weighing more than 100 kg. There is robust clinical evidence from observational studies and the real-life setting that biological agents are less effective in obese and even overweight patients and this is especially well demonstrated for TNFi [84,85,86]. This negative effect was reported in the different forms of IJD, mainly with infliximab, and was then reported with etanercept and adalimumab. Results on the specific response to certolizumab and golimumab according to body weight are less well documented. Based on the above literature review and the 2 meta-analyses, some key observations related to the influence of body weight/obesity can be made. The clinical response to TNFi in IJD was globally impaired in obese subjects, regardless of the outcome measurement. There was a dose-dependent effect in the sense that each unit increase in BMI is associated with an increase in the odds of failing therapy (by a factor of 6.5 according to the meta-analysis by Singh et al.) [84]. This resistance to TNFi is independent of the route of administration of the treatment [84]. The results for other bDMARDs are more limited, but indicate that response to tocilizumab is probably not influenced by body weight [62,63]. However, the SUMMACTA trial showed that tocilizumab given SC is less effective in obese compared to non-obese patients [95]. Furthermore, in the German RABBIT registry, response to tocilizumab was also attenuated by obesity [86]. Additional studies are thus required for this biological agent. For abatacept, results are robust for both the IV and SC routes, and show no influence of bodyweight/BMI on the efficacy of the drug [65,66,67,68,69]. For the B cell depletion agent rituximab, results are contradictory, and showed no effect of weight in one retrospective analysis [64], while the opposite was reported in a second study [60]. For ustekinumab and IL-17A agents, data examining the influence of obesity on the therapeutic efficacy are scare and even lacking in IJD [96]. Data come from psoriatic studies and reported a clear effect of obesity on ustekinumab response. This explains why the dose of this drug must be adapted for patients weighing more than 100 kg [80]. The clinical response to the anti-IL-17A agent secukinumab in IJD does not seem to be influenced by body weight but more data are needed. According to these results, for patients with high BMI or obese status, the clinician may be orientated for selecting a weight-adapted targeted DMARD. Actually, TNFi does not appear to be the best choice. In RA, abatacept or rituximab may be proposed or alternatively an IL6i. In ax-SpA, IL-17Ai may be a preferred choice. In PsA, the prescribed dose may be adapted for ustekinumab and golimumab, but IL-17Ai may be another option.
All these data show that obesity may be a factor for resistance to TNFi, an effect that is less pronounced for tocilizumab and rituximab, and not observed for abatacept. The reasons why the TNFi biological class selectively fails to respond in obese patients are unknown. Multiple explanations have been proposed such as the pharmacokinetic properties and the volume of distribution of the drugs, taking account of their lipophilic properties [6,53]. Obesity is a state that modifies the pharmacokinetics of TNFi by increasing the clearance of the drug, resulting in a shorter half-life and lower serum trough drug concentrations [84]. In parallel, obesity and overfat are conditions that may perpetuate chronic systemic low-grade inflammation, through increased levels of cytokines, chemokines and adipokines [6]. Adipokines such as leptin and adiponectin are involved in immune response and they may contribute to the inflammatory process in various immune-mediated diseases [97]. Therapeutic resistance to TNFi is independent of the route of administration and observed even for weighted-dose biologics such as infliximab. Infliximab also seems to work less well compared to fixed-dose administered drugs such as etanercept or adalimumab [58]. One potential explanation is that omental adipocytes have high expression of Fc receptors and the presence of this receptor modulates the efficacy of infliximab in both in vitro and ex vivo models [6]. A specific analysis of the therapeutic response to certolizumab (a TNFi without an Fc fragment) according to body weight and fat mass is thus a relevant issue. One weakness of the different studies into the association between obesity and therapeutic response is that the analyses were performed according to body weight (or BMI) and not fat mass. The study by Ibanez Vodnizza et al. specifically analyzed the TNFi clinical response in patients with ax-SpA according to fat mass [73]. This study confirmed that fat mass was associated with impaired clinical response: higher body weight, BMI, body fat percentage, fat mass and fat mass index at baseline were all associated with a lower chance of having a clinical improvement according to ASDAS-CRP change. The multivariate analysis confirmed that fat mass was a significant negative predictor of achieving a clinical response to TNFi in ax-SpA.
Finally, we have some information on the impact of diet interventions in obese patients with IJD or other immune-mediated diseases. Such interventions are beneficial, showing that weight loss improves treatment response to TNFi. In obese/overweight patients with PsA starting a TNFi, a hypocaloric diet was associated with more patients in MDA than patients with a free managed diet. Weight loss of more than 5% was predictive of the achievement of MDA [98]. In addition, weight loss in patients with PsA or psoriasis under stable treatment improved the different measurements of (skin and joint) disease activity [99,100]. Bariatric surgery also improves the disease activity of patients with RA [101].
We focus our review on the most common IJD. However, the crosstalk between adipose tissue and response to therapy also involved other immune-mediated diseases such as Crohn’s disease or psoriasis [54,84]. Obesity worsens the course of systemic lupus erythematosus. However, limited information is available on the relationship between the therapeutic response to conventional treatments and BMI [102].

6. Conclusions

The relationships between obesity/adipose tissue and bDMARDs/tsDMARDs in IJD are bidirectional. On the one hand, certain bDMARDs induce changes in body composition, especially TNFi, with an increase in fat mass and minor changes in lean mass. Tocilizumab is also involved by selectively increasing lean mass. On the other hand, therapeutic response to bDMARDs is hampered in obese patients, especially with TNFi. Obesity is thus a relevant comorbidity to consider in patients with IJD. Obesity may be accentuated by TNF inhibition, at least to a moderate degree, but this drug class favors visceral adiposity. On the contrary, IL-6 inhibition has a more beneficial effect by increasing lean mass and skeletal muscle, counteracting cachexia. More data on the effects of the other bDMARDs and JAKi on body composition are needed. The finding that obesity compromises the therapeutic efficacy of certain bDMARDs (TNFi and, with less evidence, rituximab or tocilizumab) but not others (abatacept), may help the physician with personalized medicine and guide the therapeutic choice in obese/overweight patients. Weight management also appears to be crucial in obese patients to improve disease activity. Specific studies using DEXA examining the direct impact of obesity, and, ideally, of fat mass, on the therapeutic response of recently licensed bDMARDs and tsDMARDs are required.

Funding

This work was supported by grants from CHU de Besancon, Roche Chugai and Agence Nationale de la Recherche (ANR) under the program ‘Investissements d’Avenir’ with reference ANR-11-LABX-0021-LipSTIC and by the Region Bourgogne Franche-Comté (support to LipSTIC LabEX 2017).

Acknowledgments

To Fiona Ecarnot, MSc, EA3920, Department of Cardiology, University Hospital Besançon, France for her help in preparing the manuscript.

Conflicts of Interest

The author declared no conflict of interest.

References

  1. McInnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef] [PubMed]
  2. Sieper, J.; Poddubnyy, D. Axial spondyloarthritis. Lancet 2017, 390, 73–84. [Google Scholar] [CrossRef]
  3. Mease, P.J. Psoriatic arthritis: Update on pathophysiology, assessment and management. Ann. Rheum. Dis. 2011, 70 (Suppl. 1), i77–i84. [Google Scholar] [CrossRef] [PubMed]
  4. Nurmohamed, M.T. Cardiovascular risk in rheumatoid arthritis. Autoimmun. Rev. 2009, 8, 663–667. [Google Scholar] [CrossRef]
  5. Peters, M.J.; van der Horst-Bruinsma, I.E.; Dijkmans, B.A.; Nurmohamed, M.T. Cardiovascular risk profile of patients with spondylarthropathies, particularly ankylosing spondylitis and psoriatic arthritis. Semin. Arthritis Rheum. 2004, 34, 585–592. [Google Scholar] [CrossRef] [PubMed]
  6. Gremese, E.; Tolusso, B.; Gigante, M.R.; Ferraccioli, G. Obesity as a risk and severity factor in rheumatic diseases. Front. Immunol. 2014, 5, 576. [Google Scholar] [CrossRef] [Green Version]
  7. Després, J.P. Body fat distribution and risk of cardiovascular disease: An update. Circulation 2012, 126, 1301–1313. [Google Scholar] [CrossRef] [Green Version]
  8. Stavropoulos-Kalinoglou, A.; Metsios, G.S.; Koutedakis, Y.; Kitas, G.D. Obesity in rheumatoid arthritis. Rheumatology 2011, 50, 450–462. [Google Scholar] [CrossRef] [Green Version]
  9. Xia, Y.; Ergun, D.L.; Wacker, W.K.; Wang, X.; Davis, C.E.; Kaul, S. Relationship between dual-energy X-ray absorptiometry volumetric assessment and X-ray computed tomography-derived single-slice measurement of visceral fat. J. Clin. Densitom. 2014, 17, 78–83. [Google Scholar] [CrossRef] [Green Version]
  10. Rall, L.C.; Roubenoff, R. Rheumatoid cachexia: Metabolic abnormalities, mechanisms and interventions. Rheumatology 2004, 43, 1219–1223. [Google Scholar] [CrossRef] [Green Version]
  11. Westhovens, R.; Nijs, J.; Taelman, V.; Dequeker, J. Body composition in rheumatoid arthritis. Br. J. Rheumatol. 1997, 36, 444–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Giles, J.T.; Ling, S.M.; Ferrucci, L.; Bartlett, S.J.; Andersen, R.E.; Towns, M.; Muller, D.; Fontaine, K.R.; Bathon, J.M. Abnormal body composition phenotypes in older rheumatoid arthritis patients: Association with disease characteristics and pharmacotherapies. Arthritis Rheumatol. 2008, 59, 807–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Toussirot, E.; Grandclément, B.; Gaugler, B.; Michel, F.; Wendling, D.; Saas, P.; Dumoulin, G.; CBT-506. Serum adipokines and adipose tissue distribution in rheumatoid arthritis and ankylosing spondylitis. A comparative study. Front. Immunol. 2013, 4, 453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Maas, F.; Arends, S.; van der Veer, E.; Wink, F.; Efde, M.; Bootsma, H.; Brouwer, E.; Spoorenberg, A. Obesity Is Common in Axial Spondyloarthritis and Is Associated with Poor Clinical Outcome. J. Rheumatol. 2016, 43, 383–387. [Google Scholar] [CrossRef] [PubMed]
  15. Toussirot, E.; Aubin, F.; Dumoulin, G. Relationships between Adipose Tissue and Psoriasis, with or without Arthritis. Front. Immunol. 2014, 5, 368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hercogová, J.; Ricceri, F.; Tripo, L.; Lotti, T.; Prignano, F. Psoriasis and body mass index. Dermatol. Ther. 2010, 23, 152–154. [Google Scholar] [CrossRef]
  17. Husted, J.A.; Thavaneswaran, A.; Chandran, V.; Eder, L.; Rosen, C.F.; Cook, R.J.; Gladman, D.D. Cardiovascular and other comorbidities in patients with psoriatic arthritis: A comparison with patients with psoriasis. Arthritis Care Res. 2011, 63, 729–1735. [Google Scholar] [CrossRef]
  18. Bhole, V.M.; Choi, H.K.; Burns, L.C.; Vera Kellet, C.; Lacaille, D.V.; Gladman, D.D.; Dutz, J.P. Differences in body mass index among individuals with PsA, psoriasis, RA and the general population. Rheumatology 2012, 51, 552–556. [Google Scholar] [CrossRef] [Green Version]
  19. Love, T.J.; Zhu, Y.; Zhang, Y.; Wall-Burns, L.; Ogdie, A.; Gelfand, J.M.; Choi, H.K. Obesity and the risk of psoriatic arthritis: A population-based study. Ann. Rheum. Dis. 2012, 71, 1273–1277. [Google Scholar] [CrossRef] [Green Version]
  20. Pedreira, P.G.; Pinheiro, M.M.; Szejnfeld, V.L. Bone mineral density and body composition in postmenopausal women with psoriasis and psoriatic arthritis. Arthritis Res. Ther. 2011, 13, R16. [Google Scholar] [CrossRef] [Green Version]
  21. Krajewska-Włodarczyk, M.; Owczarczyk-Saczonek, A.; Placek, W. Changes in body composition and bone mineral density in postmenopausal women with psoriatic arthritis. Reumatologia 2017, 55, 215–221. [Google Scholar] [CrossRef] [Green Version]
  22. Aletaha, D.; Smolen, J.S. Diagnosis and Management of Rheumatoid Arthritis: A Review. JAMA 2018, 320, 1360–1372. [Google Scholar] [CrossRef] [PubMed]
  23. Schwartz, D.M.; Kanno, Y.; Villarino, A.; Ward, M.; Gadina, M.; O’Shea, J.J. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat. Rev. Drug Discov. 2017, 16, 843–862. [Google Scholar] [CrossRef]
  24. O’Shea, J.J.; Laurence, A.; McInnes, I.B. Back to the future: Oral targeted therapy for RA and other autoimmune diseases. Nat. Rev. Rheumatol. 2013, 9, 173–182. [Google Scholar] [CrossRef] [PubMed]
  25. Franchimont, D.; Roland, S.; Gustot, T.; Quertinmont, E.; Toubouti, Y.; Gervy, M.C.; Deviere, J.; Van Gossum, A. Impact of infliximab on serum leptin levels in patients with Crohn’s disease. J. Clin. Endocrinol. Metab. 2005, 9, 3510–3516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gisondi, P.; Cotena, C.; Tessari, G.; Girolomoni, G. Anti-tumour necrosis factor-alpha therapy increases body weight in patients with chronic plaque psoriasis: A retrospective cohort study. J. Eur. Acad. Dermatol. Venereol. 2008, 22, 341–344. [Google Scholar] [CrossRef]
  27. Marcora, S.M.; Chester, K.R.; Mittal, G.; Lemmey, A.B.; Maddison, P.J. Randomised phase 2 trial of anti-tumor necrosis factor therapy for cachexia in patients with early rheumatoid arthritis. Am. J. Clin. Nutr. 2006, 84, 463–472. [Google Scholar] [CrossRef]
  28. Metsios, G.S.; Stavropoulos-Kalinoglou, A.; Douglas, K.M.; Koutedakis, Y.; Nevill, A.M.; Panoulas, V.F.; Kita, M.; Kitas, G.D. Blockade of tumour necrosis factor-alpha in rheumatoid arthritis: Effects on components of rheumatoid cachexia. Rheumatology 2007, 46, 1824–1827. [Google Scholar] [CrossRef] [Green Version]
  29. Serelis, J.; Kontogianni, M.D.; Katsiougiannis, S.; Bletsa, M.; Tektonidou, M.G.; Skopouli, F.N. Effect of anti-TNF treatment on body composition and serum adiponectin levels of women with rheumatoid arthritis. Clin. Rheumatol. 2008, 27, 795–797. [Google Scholar] [CrossRef]
  30. Engvall, I.L.; Tengstrand, B.; Brismar, K.; Hafström, I. Infliximab therapy increases body fat mass in early rheumatoid arthritis independently of changes in disease activity and levels of leptin and adiponectin: A randomised study over 21 months. Arthritis Res. Ther. 2010, 12, R197. [Google Scholar] [CrossRef] [Green Version]
  31. Kopec-Medrek, M.; Kotulska, A.; Widuchowska, M.; Adamczak, M.; Więcek, A.; Kucharz, E.J. Plasma leptin and neuropeptide Y concentrations in patients with rheumatoid arthritis treated with infliximab, a TNF-α antagonist. Rheumatol. Int. 2012, 32, 3383–3389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Chen, C.Y.; Tsai, C.Y.; Lee, P.C.; Lee, S.D. Long-term etanercept therapy favors weight gain and ameliorates cachexia in rheumatoid arthritis patients: Roles of gut hormones and leptin. Curr. Pharm. Des. 2013, 19, 1956–1964. [Google Scholar] [CrossRef] [PubMed]
  33. Sfriso, P.; Caso, F.; Filardo, G.S.; Botsios, C.; Costa, L.; Scarpa, R.; Todesco, S.; Spinella, P.; Oliviero, F.; Punzi, L. Impact of 24 months of anti-TNF therapy versus methotrexate on body weight in patients with rheumatoid arthritis: A prospective observational study. Clin. Rheumatol. 2016, 35, 1615–1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Toussirot, E.; Mourot, L.; Dehecq, B.; Wendling, D.; Grandclément, É.; Dumoulin, G.; CBT-506. TNFα blockade for inflammatory rheumatic diseases is associated with a significant gain in android fat mass and has varying effects on adipokines: A 2-year prospective study. Eur. J. Nutr. 2014, 53, 951–961. [Google Scholar] [CrossRef] [PubMed]
  35. Briot, K.; Garnero, P.; Le Henanff, A.; Dougados, M.; Roux, C. Body weight, body composition, and bone turnover changes in patients with spondyloarthropathy receiving anti-tumour necrosis factor {alpha} treatment. Ann. Rheum. Dis. 2005, 64, 1137–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Briot, K.; Gossec, L.; Kolta, S.; Dougados, M.; Roux, C. Prospective assessment of body weight, body composition, and bone density changes in patients with spondyloarthropathy receiving anti-tumor necrosis factor-alpha treatment. J. Rheumatol. 2008, 35, 855–861. [Google Scholar]
  37. Hmamouchi, I.; Roux, C.; Paternotte, S.; Kolta, S.; Dougados, M.; Briot, K. Early increase of abdominal adiposity in patients with spondyloarthritis receiving anti-tumor necrosis factor-α treatment. J. Rheumatol. 2014, 41, 1112–1117. [Google Scholar] [CrossRef]
  38. Renzo, L.D.; Saraceno, R.; Schipani, C.; Rizzo, M.; Bianchi, A.; Noce, A.; Esposito, M.; Tiberti, S.; Chimenti, S.; De Lorenzo, A. Prospective assessment of body weight and body composition changes in patients with psoriasis receiving anti-TNF-α treatment. Dermatol. Ther. 2011, 24, 446–451. [Google Scholar] [CrossRef] [Green Version]
  39. Younis, S.; Rosner, I.; Rimar, D.; Boulman, N.; Rozenbaum, M.; Odeh, M.; Slobodin, G. Weight change during pharmacological blockade of interleukin-6 or tumor necrosis factor-α in patients with inflammatory rheumatic disorders: A 16-week comparative study. Cytokine 2013, 61, 353–355. [Google Scholar] [CrossRef]
  40. Hugo, M.; Pierreisnard, A.; Mehsen-Cetre, N.; Gin, H.; Schaeverbeke, T.; Rigalleau, V. Action of tocilizumab on energy expenditure in rheumatoid arthritis: A prospective study using calorimetry and actimetry. Jt. Bone Spine 2017, 84, 751–752. [Google Scholar] [CrossRef]
  41. Tournadre, A.; Pereira, B.; Dutheil, F.; Giraud, C.; Courteix, D.; Sapin, V.; Frayssac, T.; Mathieu, S.; Malochet-Guinamand, S.; Soubrier, M. Changes in body composition and metabolic profile during interleukin 6 inhibition in rheumatoid arthritis. J. Cachexia Sarcopenia Muscle 2017, 8, 639–646. [Google Scholar] [CrossRef]
  42. Toussirot, E.; Marotte, H.; Mulleman, D.; Cormier, G.; Coury-Lucas, F.; Gaudin, P.; Dernis, E.; Bonnet, C.; Damade, R.; Grauer, J.L.; et al. Increased High Molecular Weight Adiponectin and Lean Mass During Tocilizumab Treatment in Patients with Rheumatoid Arthritis: A 12 Month Multicenter Study (Abstract). Arthritis Rheumatol. 2019, 71 (Suppl. 10). Available online: https://acrabstracts.org/abstract/ (accessed on 12 March 2020).
  43. Ursini, F.; Russo, E.; Letizia Hribal, M.; Mauro, D.; Savarino, F.; Bruno, C.; Tripolino, C.; Rubino, M.; Naty, S.; Grembiale, R.D. Abatacept improves whole-body insulin sensitivity in rheumatoid arthritis: An observational study. Medicine 2015, 94, e888. [Google Scholar] [CrossRef]
  44. Wallace, Z.S.; Miloslavsky, E.M.; Cascino, M.; Unizony, S.H.; Lu, N.; Hoffman, G.S.; Kallenberg, C.G.M.; Langford, C.A.; Merkel, P.A.; Monach, P.A.; et al. Effect of Disease Activity, Glucocorticoid Exposure, and Rituximab on Body Composition During Induction Treatment of Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Care Res. 2017, 69, 1004–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Owczarczyk-Saczonek, A.; Placek, W.; Rybak-d’Obyrn, J.; Wygonowska, E. Influence of ustekinumab on body weight of patients with psoriasis: An initial report. Postepy Dermatol. Alergol. 2014, 31, 29–31. [Google Scholar] [CrossRef] [PubMed]
  46. Gisondi, P.; Conti, A.; Galdo, G.; Piaserico, S.; De Simone, C.; Girolomoni, G. Ustekinumab does not increase body mass index in patients with chronic plaque psoriasis: A prospective cohort study. Br. J. Dermatol. 2013, 168, 1124–1127. [Google Scholar] [CrossRef] [PubMed]
  47. Galluzzo, M.; D’Adamio, S.; Pastorino, R.; Andreoli, A.; Servoli, S.; Bianchi, L.; Talamonti, M. Effect of anti IL-12/23 on body composition: Results of bioelectrical impedance analysis in Caucasian psoriatic patients. Expert Opin. Biol. Ther. 2018, 18, 229–235. [Google Scholar] [CrossRef] [PubMed]
  48. Takamura, S.; Takahashi, A.; Inoue, Y.; Teraki, Y. Effects of tumor necrosis factor-α, interleukin-23 and interleukin-17A inhibitors on bodyweight and body mass index in patients with psoriasis. J. Dermatol. 2018, 45, 1130–1134. [Google Scholar] [CrossRef]
  49. Nakamura, T.; Iwasaki, Y.; Yamane, S.; Ogura, M.; Yasoda, A.; Nagashima, K.; Inagaki, N. Adiposity induced by interleukin-17A blockade. Diabetes Metab. 2017, 43, 93–94. [Google Scholar] [CrossRef]
  50. Chikugo, M.; Sebe, M.; Tsutsumi, R.; Iuchi, M.; KIshi, J.; Kuroda, M.; Harada, N.; Nishioka, Y.; Sakaue, H. Effect of Janus kinase inhibition by tofacitinib on body composition and glucose metabolism. J. Med. Investig. 2018, 65, 166–170. [Google Scholar] [CrossRef] [Green Version]
  51. Novikova, D.S.; Udachkina, H.V.; Markelova, E.I.; Kirillova, I.G.; Misiyuk, A.S.; Demidova, N.V.; Popkova, T.V. Dynamics of body mass index and visceral adiposity index in patients with rheumatoid arthritis treated with tofacitinib. Rheumatol. Int. 2019, 39, 1181–1189. [Google Scholar] [CrossRef] [PubMed]
  52. Cañete, J.D.; Mease, P. The link between obesity and psoriatic arthritis. Ann Rheum. Dis. 2012, 71, 1265–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Iannone, F.; Lopalco, G.; Rigante, D.; Orlando, I.; Cantarini, L.; Lapadula, G. Impact of obesity on the clinical outcome of rheumatologic patients in biotherapy. Autoimmun. Rev. 2016, 15, 447–450. [Google Scholar] [CrossRef] [PubMed]
  54. Puig, L. Obesity and psoriasis: Body weight and body mass index influence the response to biological treatment. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 1007–1011. [Google Scholar] [CrossRef] [PubMed]
  55. Heimans, L.; van den Broek, M.; le Cessie, S.; Siegerink, B.; Riyazi, N.; Han, K.H.; Kerstens, P.J.; Huizinga, T.W.; Lems, W.F.; Allaart, C.F. Association of high body mass index with decreased treatment response to combination therapy in recent-onset rheumatoid arthritis patients. Arthritis Care Res. 2013, 65, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
  56. Klaasen, R.; Wijbrandts, C.A.; Gerlag, D.M.; Tak, P.P. Body mass index and clinical response to infliximab in rheumatoid arthritis. Arthritis Rheum. 2011, 63, 359–364. [Google Scholar] [CrossRef]
  57. Smolen, J.; Szumski, A.; Koenig, A.S.; Jones, T.V. Impact of body mass index on response to etanercept therapy in subject with moderately active rheumatoid arthritis in the PRESERVE trial (Abstract). Arthritis Rheum. 2011, 63 (Suppl.), S154. [Google Scholar]
  58. Gremese, E.; Carletto, A.; Padovan, M.; Atzeni, F.; Raffeiner, B.; Giardina, A.R.; Favalli, E.G.; Erre, G.L.; Gorla, R.; Galeazzi, M.; et al. Obesity and reduction of the response rate to anti-tumor necrosis factor α in rheumatoid arthritis: An approach to a personalized medicine. Arthritis Care Res. 2013, 65, 94–100. [Google Scholar] [CrossRef]
  59. Ottaviani, S.; Gardette, A.; Tubach, F.; Roy, C.; Palazzo, E.; Gill, G.; Meyer, O.; Dieudé, P. Body mass index and response to infliximab in rheumatoid arthritis. Clin. Exp. Rheumatol. 2015, 33, 478–483. [Google Scholar]
  60. Iannone, F.; Fanizzi, R.; Notarnicola, A.; Scioscia, C.; Anelli, M.G.; Lapadula, G. Obesity reduces the drug survival of second line biological drugs following a first TNF-α inhibitor in rheumatoid arthritis patients. Jt. Bone Spine 2015, 82, 187–191. [Google Scholar] [CrossRef]
  61. McCulley, C.B.; Barton, J.L.; Cannon, G.W.; Sauer, B.C.; Teng, C.C.; George, M.D.; Caplan, L.; England, B.R.; Mikuls, T.R.; Baker, J.F. Body mass index and persistence of conventional DMARDs and TNF inhibitors in rheumatoid arthritis. Clin. Exp. Rheumatol. 2019, 37, 422–428. [Google Scholar] [PubMed]
  62. Pers, Y.M.; Godfrin-Valnet, M.; Lambert, J.; Fortunet, C.; Constant, E.; Mura, T.; Pallot-Prades, B.; Jorgensen, C.; Maillefert, J.F.; Marotte, H.; et al. Response to tocilizumab in rheumatoid arthritis is not influenced by the body mass index of the patient. J. Rheumatol. 2015, 42, 580–584. [Google Scholar] [CrossRef] [PubMed]
  63. Gardette, A.; Ottaviani, S.; Sellam, J.; Berenbaum, F.; Lioté, F.; Meyer, A.; Sibilia, J.; Fautrel, B.; Palazzo, E.; Dieudé, P. Body mass index and response to tocilizumab in rheumatoid arthritis: A real life study. Clin. Rheumatol. 2016, 35, 857–861. [Google Scholar] [CrossRef] [PubMed]
  64. Ottaviani, S.; Gardette, A.; Roy, C.; Tubach, F.; Gill, G.; Palazzo, E.; Meyer, O.; Dieudé, P. Body Mass Index and response to rituximab in rheumatoid arthritis. Jt. Bone Spine 2015, 82, 432–436. [Google Scholar] [CrossRef]
  65. Gardette, A.; Ottaviani, S.; Sellam, J.; Berenbaum, F.; Lioté, F.; Fautrel, B.; Palazzo, E.; Meyer, A.; Sibilia, J.; Dieudé, P. Body mass index and response to abatacept in rheumatoid arthritis. Eur. J. Clin. Investig. 2016, 46, 1048–1052. [Google Scholar] [CrossRef]
  66. D’Agostino, M.A.; Alten, R.; Mysler, E.; Le Bars, M.; Ye, J.; Murthy, B.; Heitzmann, J.; Vadanici, R.; Ferraccioli, G. Body mass index and clinical response to intravenous or subcutaneous abatacept in patients with rheumatoid arthritis. Clin. Rheumatol. 2017, 36, 2655–2665. [Google Scholar] [CrossRef] [Green Version]
  67. Mariette, X.; Alten, R.; Nüßlein, H.G.; Galeazzi, M.; Lorenz, H.M.; Cantagrel, A.; Chartier, M.; Poncet, C.; Rauch, C.; Le Bars, M. The effect of body mass index on clinical response to abatacept as a first-line biologic for rheumatoid arthritis: 6-month results from the 2-year, observational, prospective ACTION study. Jt. Bone Spine 2017, 84, 571–576. [Google Scholar] [CrossRef]
  68. Di Carlo, M.; Salaffi, F.; Gremese, E.; Iannone, F.; Lapadula, G.; Ferraccioli, G.; GISEA Study Group. Body mass index as a driver of selection of biologic therapy in rheumatoid arthritis. Results from the US-CLARA study. Eur. J. Intern. Med. 2019, 66, 57–61. [Google Scholar] [CrossRef]
  69. Iannone, F.; Courvoisier, D.S.; Gottenberg, J.E.; Hernandez, M.V.; Lie, E.; Canhão, H.; Pavelka, K.; Hetland, M.L.; Turesson, C.; Mariette, X.; et al. Body mass does not impact the clinical response to intravenous abatacept in patients with rheumatoid arthritis. Analysis from the “pan-European registry collaboration for abatacept (PANABA). Clin. Rheumatol. 2017, 36, 773–779. [Google Scholar] [CrossRef]
  70. Ottaviani, S.; Allanore, Y.; Tubach, F.; Forien, M.; Gardette, A.; Pasquet, B.; Palazzo, E.; Meunier, M.; Hayem, G.; Job-Deslandre, C.; et al. Body mass index influences the response to infliximab in ankylosing spondylitis. Arthritis Res. Ther. 2012, 14, R115. [Google Scholar] [CrossRef] [Green Version]
  71. Gremese, E.; Bernardi, S.; Bonazza, S.; Nowik, M.; Peluso, G.; Massara, A.; Tolusso, B.; Messuti, L.; Miceli, M.C.; Zoli, A.; et al. Body weight, gender and response to TNF-α blockers in axial spondyloarthritis. Rheumatology 2014, 53, 875–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Micheroli, R.; Hebeisen, M.; Wildi, L.M.; Exer, P.; Tamborrini, G.; Bernhard, J.; Möller, B.; Zufferey, P.; Nissen, M.J.; Scherer, A.; et al. Rheumatologists of the Swiss Clinical Quality Management Program. impact of obesity on the response to tumor necrosis factor inhibitors in axial spondyloarthritis. Arthritis Res. Ther. 2017, 19, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ibáñez Vodnizza, S.E.; Nurmohamed, M.T.; Visman, I.M.; van Denderen, J.C.; Lems, W.F.; Jaime, F.; van der Horst-Bruinsma, I.E. Fat Mass Lowers the Response to Tumor Necrosis Factor-α Blockers in Patients with Ankylosing Spondylitis. J. Rheumatol. 2017, 44, 1355–1361. [Google Scholar] [CrossRef] [PubMed]
  74. Di Minno, M.N.; Peluso, R.; Iervolino, S.; Lupoli, R.; Russolillo, A.; Scarpa, R.; di Minno, G. Obesity and the prediction of minimal disease activity: A prospective study in psoriatic arthritis. Arthritis Care Res. 2013, 65, 141–147. [Google Scholar] [CrossRef]
  75. Iannone, F.; Fanizzi, R.; Scioscia, C.; Anelli, M.G.; Lapadula, G. Body mass does not affect the remission of psoriatic arthritis patients on anti-TNF-α therapy. Scand. J. Rheumatol. 2013, 42, 41–44. [Google Scholar] [CrossRef]
  76. Eder, L.; Thavaneswaran, A.; Chandran, V.; Cook, R.J.; Gladman, D.D. Obesity is associated with a lower probability of achieving sustained minimal disease activity state among patients with psoriatic arthritis. Ann. Rheum. Dis. 2015, 74, 813–817. [Google Scholar] [CrossRef]
  77. Højgaard, P.; Glintborg, B.; Kristensen, L.E.; Gudbjornsson, B.; Love, T.J.; Dreyer, L. The influence of obesity on response to tumour necrosis factor-α inhibitors in psoriatic arthritis: Results from the DANBIO and ICEBIO registries. Rheumatology 2016, 55, 2191–2199. [Google Scholar] [CrossRef] [Green Version]
  78. Yanaba, K.; Umezawa, Y.; Ito, T.; Hayashi, M.; Kikuchi, S.; Fukuchi, O.; Saeki, H.; Nakagawa, H. Impact of obesity on the efficacy of ustekinumab in Japanese patients with psoriasis: A retrospective cohort study of 111 patients. Arch. Dermatol. Res. 2014, 306, 921–925. [Google Scholar] [CrossRef]
  79. Del Alcázar, E.; Ferran, M.; López-Ferrer, A.; Notario, J.; Vidal, D.; Riera, J.; Aparicio, G.; Gallardo, F.; Vilarrasa, E.; Alsina, M.; et al. Effectiveness and safety of ustekinumab 90 mg in patients weighing 100 kg or less: A retrospective, observational, multicenter study. J. Dermatolog. Treat. 2019, 18, 1–5. [Google Scholar] [CrossRef]
  80. Lebwohl, M.; Yeilding, N.; Szapary, P.; Wang, Y.; Li, S.; Zhu, Y.; Reich, K.; Langley, R.G.; Papp, K.A. Impact of weight on the efficacy and safety of ustekinumab in patients with moderate to severe psoriasis: Rationale for dosing recommendations. J. Am. Acad. Dermatol. 2010, 63, 571–579. [Google Scholar] [CrossRef]
  81. Lee, J.E.; Wang, J.; Florian, J.; Wang, Y.M.; Kettl, D.; Marcus, K.; Woitach, A. Effect of Body Weight on Risk-Benefit and Dosing Regimen Recommendation of Secukinumab for the Treatment of Moderate to Severe Plaque Psoriasis. Clin. Pharmacol. Ther. 2019, 106, 78–80. [Google Scholar] [CrossRef] [PubMed]
  82. Tiberio, R.; Graziola, F.; Miglino, B.; Veronese, F.; Annali, G.; Savoia, P. Secukinumab for Psoriasis in Obese Patients: Minireview and Clinical Experience. Case Rep. Dermatol. 2019, 11 (Suppl. 1), 29–36. [Google Scholar] [CrossRef]
  83. McInnes, I.B.; Ferraccioli, G.; D’Agostino, M.A.; Le Bars, M.; Banerjee, S.; Ahmad, H.A.; Elbez, Y.; Mease, P.J. Body mass index and treatment response to subcutaneous abatacept in patients with psoriatic arthritis: A post hoc analysis of a phase III trial. RMD Open 2019, 5, e000934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Singh, S.; Facciorusso, A.; Singh, A.G.; Vande Casteele, N.; Zarrinpar, A.; Prokop, L.J.; Grunvald, E.L.; Curtis, J.R.; Sandborn, W.J. Obesity and response to anti-tumor necrosis factor-α agents in patients with select immune-mediated inflammatory diseases: A systematic review and meta-analysis. PLoS ONE 2018, 13, e0195123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Shan, J.; Zhang, J. Impact of obesity on the efficacy of different biologic agents in inflammatory diseases: A systematic review and meta-analysis. Jt. Bone Spine 2019, 86, 173–183. [Google Scholar] [CrossRef] [PubMed]
  86. Schäfer, M.; Meißner, Y.; Kekow, J.; Berger, S.; Remstedt, S.; Manger, B.; Listing, J.; Strangfeld, A.; Zink, A. Obesity reduces the real-world effectiveness of cytokine-targeted but not cell-targeted disease-modifying agents in rheumatoid arthritis. Rheumatology 2019. [Google Scholar] [CrossRef] [Green Version]
  87. Kremer, J.M.; Schiff, M.; Muram, D.; Zhong, J.; Alam, J.; Genovese, M. Response to baricitinib therapy in patients with rheumatoid arthritis with inadequate response to csDMARDs as function of baseline characteristics. RMD Open 2018, 4, e000581. [Google Scholar] [CrossRef]
  88. Marouen, S.; Barnetche, T.; Combe, B.; Morel, J.; Daïen, C.I. TNF inhibitors increase fat mass in inflammatory rheumatic disease: A systematic review with meta-analysis. Clin. Exp. Rheumatol. 2017, 35, 337–343. [Google Scholar]
  89. Hube, F.; Hauner, H. The role of TNF-alpha in human adipose tissue: Prevention of weight gain at the expense of insulin resistance? Horm. Metab. Res. 1999, 31, 626–631. [Google Scholar] [CrossRef]
  90. Sheane, B.J. Tumor necrosis factor inhibition and adipose tissue distribution—Are reported changes relevant to cardiometabolic risk? J. Rheumatol. 2014, 41, 1035–1037. [Google Scholar] [CrossRef] [Green Version]
  91. Roubille, C.; Richer, V.; Starnino, T.; McCourt, C.; McFarlane, A.; Fleming, P.; Siu, S.; Kraft, J.; Lynde, C.; Pope, J.; et al. The effects of tumour necrosis factor inhibitors, methotrexate, non-steroidal anti-inflammatory drugs and corticosteroids on cardiovascular events in rheumatoid arthritis, psoriasis and psoriatic arthritis: A systematic review and meta-analysis. Ann. Rheum. Dis. 2015, 74, 480–489. [Google Scholar] [CrossRef]
  92. VanderVeen, B.N.; Fix, D.K.; Montalvo, R.N.; Counts, B.R.; Smuder, A.J.; Murphy, E.A.; Koh, H.J.; Carson, J.A. The regulation of skeletal muscle fatigability and mitochondrial function by chronically elevated interleukin-6. Exp. Physiol. 2019, 104, 385–397. [Google Scholar] [CrossRef] [PubMed]
  93. Ma, L.; Sha, G.; Zhang, Y.; Li, Y. Elevated serum IL-6 and adiponectin levels are associated with frailty and physical function in Chinese older adults. Clin. Interv. Aging 2018, 13, 2013–2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tsujinaka, T.; Fujita, J.; Ebisui, C.; Yano, M.; Kominami, E.; Suzuki, K.; Tanaka, K.; Katsume, A.; Ohsugi, Y.; Shiozaki, H.; et al. Interleukin 6 receptor antibody inhibits muscle atrophy and modulates proteolytic systems in interleukin 6 transgenic mice. J. Clin. Investig. 1996, 97, 244–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Burmester, G.R.; Rubbert-Roth, A.; Cantagrel, A.; Hall, S.; Leszczynski, P.; Feldman, D.; Rangaraj, M.J.; Roane, G.; Ludivico, C.; Lu, P.; et al. A randomised, double-blind, parallel-group study of the safety and efficacy of subcutaneous tocilizumab versus intravenous tocilizumab in combination with traditional disease-modifying antirheumatic drugs in patients with moderate to severe rheumatoid arthritis (SUMMACTA study). Ann. Rheum. Dis. 2014, 73, 69–74. [Google Scholar]
  96. Costa, L.; Ramonda, R.; Ortolan, A.; Favero, M.; Foti, R.; Visalli, E.; Rossato, M.; Cacciapaglia, F.; Lapadula, G.; Scarpa, R. Psoriatic arthritis and obesity: The role of anti-IL-12/IL-23 treatment. Clin. Rheumatol. 2019, 38, 2355–2362. [Google Scholar] [CrossRef]
  97. Toussirot, E.; Streit, G.; Wendling, D. The contribution of adipose tissue and adipokines to inflammation in joint diseases. Curr. Med. Chem. 2007, 14, 1095–1100. [Google Scholar] [CrossRef]
  98. Di Minno, M.N.; Peluso, R.; Iervolino, S.; Russolillo, A.; Lupoli, R.; Scarpa, R. Weight loss and achievement of minimal disease activity in patients with psoriatic arthritis starting treatment with tumour necrosis factor alpha blockers. Ann. Rheum. Dis. 2014, 73, 1157–1162. [Google Scholar] [CrossRef] [Green Version]
  99. Klingberg, E.; Bilberg, A.; Björkman, S.; Hedberg, M.; Jacobsson, L.; Forsblad-d’Elia, H.; Carlsten, H.; Eliasson, B.; Larsson, I. Weight loss improves disease activity in patients with psoriatic arthritis and obesity: An interventional study. Arthritis Res. Ther. 2019, 21, 17. [Google Scholar] [CrossRef] [Green Version]
  100. Al-Mutairi, N.; Nour, T. The effect of weight reduction on treatment outcomes in obese patients with psoriasis on biologic therapy: A randomized controlled prospective trial. Expert Opin. Biol. Ther. 2014, 14, 749–756. [Google Scholar] [CrossRef]
  101. Sparks, J.A.; Halperin, F.; Karlson, J.C.; Karlson, E.W.; Bermas, B.L. Impact of Bariatric Surgery on Patients With Rheumatoid Arthritis. Arthritis Care Res. 2015, 67, 1619–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Versini, M.; Jeandel, P.Y.; Rosenthal, E.; Shoenfeld, Y. Obesity in autoimmune diseases: Not a passive bystander. Autoimmun. Rev. 2014, 13, 981–1000. [Google Scholar] [CrossRef] [PubMed]
Table 1. Changes in body composition during Tumour necrosis factor (TNF) inhibitor treatment in inflammatory rheumatic diseases.
Table 1. Changes in body composition during Tumour necrosis factor (TNF) inhibitor treatment in inflammatory rheumatic diseases.
Author (Reference)Patients (N)Age, YearsDisease DurationInflammatory Joint DiseaseSex RatioStudy DesignStudy DurationTnfiBody Composition AssessmentResults
Marcora [27]2652<6 monthsRA18 F, 6 Mrandomized phase 2 trial6 monthsETA or MTXDEXANo overall changes. Weight gain in 6/13 patients in ETA group: gain in lean mass
Metsios [28]2061.117.3 yearsRA10 F, 10 Mopen label12 weeksNDbioelectrical impedance↑ truncal fat mass
Serelis [29]19545 monthsRA19 Fopen label1 yearIFX or ADADEXANo changes in fat or lean mass
Engvall [30]4057.55 monthsRA29 F, 11 Mrandomized trial2 yearsIFX or HCQ and SLZDEXA↑ fat mass [+ 3.4 kg) and fat mass index in the IFX group
Kopec- Medrek [31]16ND7.1 yearsRA16 Fopen label12 monthsIFXDEXA↑ weight, BMI and fat mass
Chen [32]2053.86.6 yearsRA18 F, 2 Mcontrol group without ETA12 monthsETAbioelectric impedance↑ weight and BMI in the ETA group. No changes in fat mass
Toussirot [34]2048.69.6 years8 RA, 12 ax-SpA14 F, 6 Mopen label2 yearsIFX, ETA or ADADEXA↑ fat mass, ↑ % fat, ↑ fat android region, and visceral fat
Briot [35]1921–71 (median: 40)16.5 yearsSpA (10 ax-SpA, 4 PsA, 5 IBD)2F, 17 Mopen- label12 monthsIFX 3–5 mg/kgDEXA↑ weight + 2.2 kg, ↑ lean mass + 1.4 kg; no change in fat mass
Briot [36]106381.5 yearsSpA (60 ax-SpA, 46 p-SpA)26 F, 80 Mopen label2 yearsIFX or ETADEXA↑ body weight (+3.5%), ↑ fat mass (+14.5%); ↑ lean mass + 2%)
Hmamouchi [37]6539.313.1ax-SpA22 F, 63 Mopen label2 yearsIFX or ETADEXA↑ subcutaneous and visceral fat
Renzo [38]2042.214.1 yearsPsANDopen label6 monthsIFX or ETADEXA↑ weight (+2.1%) and BMI. ↑ fat mass (+8.9% and lean mass (+2.9%)
(M: male; F: female; BMI: body mass index; TNFi: TNF inhibitor; IFX: infliximab; ETA: etanercept; ADA: adalimumab; MTX: methotrexate; HCQ: hydroxychloroquine; SLZ: sulfasalazine; DEXA: dual X-ray absorptiometry; RA: rheumatoid arthritis; SpA: spondyloarthritis; ax-SpA: axial spondyloarthritis; pSpA: peripheral spondyloarthritis; PsA: psoriatic arthritis; IBD: inflammatory bowel disease).
Table 2. Changes in body composition during tocilizumab treatment in rheumatoid arthritis.
Table 2. Changes in body composition during tocilizumab treatment in rheumatoid arthritis.
Author (Reference)Patients (N)Age (Years)Disease DurationInflammatory Joint DiseaseSex RatioStudy DesignStudy DurationIL-6iBody Composition AssessmentResults
Younis [39]2152NARANAcontrolled trial4 months21 TCZ, 16 IFXNA↑ body weight and BMI (+0.3 unit). No changes under IFX
Hugo [40]1657.213 yearsRA14 F, 2 Mopen label3 monthsTCZDEXANo changes in body weight, body composition
Tournadre [41]2157.88.5 yearsRA16F, 4 Mopen label12 monthsTCZDEXA↑ body weight, ↑ BMI, ↑ lean mass and fat free mass. No change in fat mass
Toussirot [42]10656.69.9 yearsRA78 F, 28 Mopen label12 monthsTCZDEXA↑ BMI, ↑ lean mass. No change in fat mass
(RA: rheumatoid arthritis; BMI: body mass index; M: male; F: female; IL6i: interleukin-6 inhibitor; TCZ: tocilizumab; IFX: infliximab; DEXA: dual X-ray absorptiometry; NA: not available).
Table 3. Response to biological agents (bDMARDs) in obese/overweight patients with rheumatoid arthritis.
Table 3. Response to biological agents (bDMARDs) in obese/overweight patients with rheumatoid arthritis.
Author (Reference)Number of SubjectsSex Ratio M/F (%)Disease Duration (Years)Obese/Overweight Subjects (%)bDMARDsOutcomeMain Results
Heimans [55]50832/680.4obese and overweight: 57.4csDMARDs combination or MTX + IFXDAS≤ 2.4 at one yearRR DAS≤ 2.4 at one year in obese /overweight patients: 2.2 [0.99–4.92]
Klaasen [56]8923/777obese: 16.8IFX [3 mg/kg)∆DAS28 week 16Negative relationship between BMI and ∆DAS28 or remission
Smolen [57]76117/836.9overweight: 35.2
obese: 16.7
ETA (50 mg/week)remission (according to DAS28, CDAI, SDAI) week 36Negative relationship between BMI and remission
Gremese [58]64119/818.4overweight: 32.3
obese: 10.3
IFX(3mg/kg), ETA (50 mg/week) or ADA (40 mg eow)remission (DAS28) at one year% remission in obese/overweight patients < normal BMI
Ottaviani [59]7617/838overweight: 38.2
obese: 28.9
IFX (3 mg/kg)∆DAS28 ≥ 1.2 at 6 monthsFewer responders in overweight/obese patients compared to normal BMI
Iannone [60]29215/8512overweight: 37.3
obese: 22.6
All bDMARDs at the second line of treatmentdrug survival at one yearLess drug persistence in obese patients versus normal weight
McCulley [61]23,66987/13NAoverweight: 36.6
obese: 40.1
All csDMARDs and subcutaneous injectable TNFitime to treatment discontinuationSevere obesity not associated with treatment discontinuation compared to overweight BMI for all except prednisone. Low BMI was associated with TNFi discontinuation
Pers [62]22217.6/82.414overweight: 26
obese: 15.5
TCZ IV (8 mg/kg)EULAR response at month 6Similar response according to BMI
Gardette [63]11584.3/15.711overweight: 32
obese: 22
TCZ IV (8 mg/kg)∆DAS28 ≥ 1.2 at month 6No effect of BMI on TCZ response
Ottaviani [64]11481.5/18.59.6overweight: 35.9
obese: 30
RTX IV (1 g × 2 days 1 and 15)∆DAS28 ≥ 1.2 at month 6No effect of BMI on RTX response
Gardette [65]14182.3/17.712.5overweight: 27
obese: 27.6
ABA IV (500 mg < 60 kg; 750 mg 60–100 kg; 1000 mg > 100 kg)∆DAS28 ≥ 1.2 at month 6No effect of BMI on ABA response
D’Agostino (ACQUIRE trial) [66]145682.5/17.57.6overweight: 34
obese: 30
ABA IV or SCRemission (DAS28 < 2.6) at month 6Rate of remission similar across BMI groups
Mariette (ACTION study) [67]64373.8/26.27.2overweight: 35
obese: 24
ABA IV (500 mg < 60 kg; 750 mg 60–100 kg; 1000 mg > 100 kg)Drug retention at 6 monthsRetention rates similar across BMI groups
Di Carlo [68]13083.8/16.211.2NAABA SC or IVDAS28 remission or Boolean criteria for remissionNo effect of BMI on ABA response
Ianone (PANABA registry) [69]201580.6/19.410.2obese: 18.9ABA IV (500 mg < 60 kg 750 mg 60–100 kg; 1000 mg > 100 kg)Drug retention of ABANo difference in ABA retention between obese and non obese patients
(TNF inhibitor, IL-6 inhibitor, co-stimulatory inhibitor and B-cell depletion agent) in rheumatoid arthritis according to body weight or body mass index (M: male; F: female; BMI: body mass index; bDMARD: biological disease-modifying antirheumatic drug; csDMARD: conventional synthetic disease modifying antirheumatic drug; IFX: infliximab; ETA: etanercept; ADA: adalimumab; TCZ: tocilizumab; ABA: abatacept; RTX: rituximab; MTX: methotrexate; RA: rheumatoid arthritis; DAS28: disease activity score 28 joints; SDAI: simplified disease activity score; CDAI: clinical disease activity index; RR: relative risk; eow: every other week; IV: intravenous; SC: subcutaneous; NA: not available).

Share and Cite

MDPI and ACS Style

Toussirot, E. The Interrelations between Biological and Targeted Synthetic Agents Used in Inflammatory Joint Diseases, and Obesity or Body Composition. Metabolites 2020, 10, 107. https://doi.org/10.3390/metabo10030107

AMA Style

Toussirot E. The Interrelations between Biological and Targeted Synthetic Agents Used in Inflammatory Joint Diseases, and Obesity or Body Composition. Metabolites. 2020; 10(3):107. https://doi.org/10.3390/metabo10030107

Chicago/Turabian Style

Toussirot, Eric. 2020. "The Interrelations between Biological and Targeted Synthetic Agents Used in Inflammatory Joint Diseases, and Obesity or Body Composition" Metabolites 10, no. 3: 107. https://doi.org/10.3390/metabo10030107

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop