Abstract
Objective
Immune‐mediated inflammatory arthritis (IMIA) is a heterogeneous group of diseases including rheumatoid arthritis (RA), psoriatic arthritis (PsA), and spondyloarthritis (SpA). Disease‐modifying antirheumatic drugs (DMARDs) target very different cellular components of the disease processes. Characterization of the pathobiological subtypes of IMIA could provide more specific treatment approaches for each disease. For example, RA has been proposed to consist of at least three synovial pathotypes (lymphoid, myeloid, and fibroid), and only a subgroup of RA patients have erosive disease. The objective of this study was to evaluate the effects of various DMARDs on different synovial cell subsets using human ex vivo models of IMIA.
Methods
Synovial fluid and blood samples were obtained from a study population consisting of patients with RA, PsA, or peripheral SpA with at least one swollen joint (n = 18). The DMARDs used in this study were methotrexate, adalimumab, etanercept, tocilizumab, anakinra, ustekinumab, secukinumab, tofacitinib, and baricitinib. Paired synovial fluid mononuclear cells (SFMCs), peripheral blood mononuclear cells (PBMCs), and fibroblast‐like synovial cells (FLSs) were used in three different previously optimized ex vivo models.
Results
In SFMCs cultured for 48 hours, all DMARDs except anakinra decreased the production of monocyte chemoattractant protein (MCP)‐1. In SFMCs cultured for 21 days, only the two tumor necrosis factor alpha (TNFα) inhibitors adalimumab and etanercept decreased the secretion of tartrate‐resistant acid phosphatase (P < 0.01, P < 0.001). In the FLS and PBMC 48‐hour co‐cultures, only tocilizumab (P < 0.001) and the two Janus kinase inhibitors tofacitinib and baricitinib (both P < 0.05) decreased the production of MCP‐1 by around 50%.
Conclusion
TNFα inhibition was effective in preventing inflammatory osteoclastogenesis, whereas tocilizumab, tofacitinib, and baricitinib had superior efficacy in cultures dominated by FLSs. Taken together, this study reveals that responses to cytokine inhibitors associate with cellular composition in models of IMIA. In particular, this study provides new evidence on the differential effect of DMARDs on leukocytes compared with stromal cells.
Introduction
Immune‐mediated inflammatory arthritis (IMIA), including rheumatoid arthritis (RA), psoriatic arthritis (PsA), and spondyloarthritis (SpA), encompasses a group of immune‐mediated inflammatory diseases characterized by synovitis and cartilage and bone damage. Early intervention with disease‐modifying antirheumatic drugs (DMARDs) and the development of therapies targeting specific components of the disease pathogenesis has radically improved the treatment of these diseases 1. However, despite general improvements in treatment options, some patients still do not respond to treatment 2.
Tumor necrosis factor alpha (TNFα) plays a central role in the pathogenesis of all of the IMIA diseases. Thus, TNFα inhibitors have shown efficacy in patients suffering from RA, PsA, and SpA. In contrast, other proinflammatory cytokines are considered to play a central role in only some of these diseases; for example, interleukin (IL)‐6 is important in RA, whereas IL‐17 and IL‐23 play more prominent roles in the pathogenesis of SpA and PsA 3, 4. However, there is still lack of tailored therapy for patients within each disease subgroup. Currently, the first choice of DMARD in RA is mostly dependent on local policies including market pricing, administration route, and side effects. This is perpetuated by the rather similar efficacy profile of the biological DMARDs in the clinical trials 5, 6.
Cytokine profiling 4 and synovial phenotyping 7 holds promise for the future stratification of patients with immune‐mediated inflammatory diseases. The RA synovium can histologically be divided in the three synovial pathotypes: 1) lymphoid, 2) myeloid, and 3) fibroid 8. The fibroid pathotype is believed to include a large proportion of the nonresponders to biologic DMARDs 9, 10. In addition, erosive disease can be seen in patients with combinations of RA, PsA, and SpA 11, 12. There are also some links between pathobiology and DMARD‐specific treatment responses. Thus, IL‐6 inhibition seems to be more efficacious in RA patients with a high C‐reactive protein level 13 and inhibition of lymphocytes with either rituximab or abatacept is more efficacious in anticitrullinated protein antibody–positive RA patients 14. Furthermore, TNFα inhibitors seem to be superior in patients with a CD68‐positive macrophage‐dominated synovium 9 and are most effective in reducing erosive joint damage in RA 15. In PsA, treatment with different DMARDs based on T cell phenotyping was recently shown to be beneficial 16.
The increase in treatment options now requires more definitive studies on how to optimize patient‐tailored therapy in IMIA. Therefore, we used in vitro models that mimic different pathotypes of IMIA to study potential associations between the treatment effect of different cytokine inhibitors and the cellular composition of the cultures. The first model used was synovial fluid mononuclear cell (SFMC) 48‐hour cultures. SFMCs have previously been shown to consist of primarily lymphocytes and cells of the monocyte and macrophage lineage 17. Cultures of SFMCs have been shown to preserve functional characteristics, such as stimulated proliferation and spontaneous production of autoantibodies 18, 19. Therefore, this culture was used as a model of a lymphocyte‐ and monocyte‐dominated environment 20. SFMCs cultured for 21 days can differentiate into osteoclasts without the addition of any inducers in vitro. The cells could differentiate into osteoclasts only because they were primed in vivo in the inflamed joint 21, 22. This model was used to mimic inflammatory osteoclastogenesis. The fibroblast‐like synovial cell (FLS) and peripheral blood mononuclear cell (PBMC) co‐cultures have previously been used to evaluate the contribution of fibroblast‐leukocyte interactions and treatment response of immunosuppressive compounds in IMIA 23, 24, 25. Therefore, this model was used to study immune reactions in an environment dominated by fibroblasts. The model was further optimized by using autologous PBMCs to prevent any contribution of graft versus host reactions to the model. DMARDs included in this study were methotrexate, adalimumab, etanercept, tocilizumab, anakinra, ustekinumab, secukinumab, tofacitinib, and baricitinib. Collectively, the objective of this study was to examine the effect of different DMARDs on different synovial cell subsets using three human ex vivo models of IMIA.
Materials and Methods
Patients and samples
A cross‐sectional, paired set of PBMCs and SFMCs were obtained from patients with chronic RA (n = 10), peripheral SpA (n = 5), or PsA (n = 3) with at least one swollen joint (from where synovial fluid was acquired) (n = 18) at the outpatient clinic at Aarhus University Hospital at the time of therapeutic arthrocentesis (Table 1). Not all patient samples were used in all experiments. The exact number of patients in each experiment is stated in the figure legends. We did not have access to data on treatment for all patients.
Table 1.
Patient characteristicsa
| Diagnosis | RA (n = 10) | SpA (n = 5) | PsA (n = 3) |
|---|---|---|---|
| Disease activity | |||
| VAS (0‐100) | 58.5 (40‐84) | 74.5 (37‐86) | 40.0 (‐) |
| Swollen joint count (0‐28) | 2.0 (1–7) | 1.5 (1‐2) | 1.0 (1‐2) |
| HAQ (0‐3) | 0.3 (0‐1) | 0.5 (0.1‐1.4) | 0.8 (‐) |
| CRP (mg/L) | 31.5 (19‐105) | 29.0 (5‐62) | 36.0 (5‐67) |
| DAS28CRP (0‐10) | 4.2 (4‐6) | 4.0 (3‐5) | 3.0 (‐) |
| Treatment | |||
| MTX (n) | 4 | 1 | 1 |
| TNFα inhibitor (n) | 0 | 2 | 2 |
| Salazopyrin (n) | 1 | 1 | 1 |
Abbreviation: CI, confidence interval; CRP, C‐reactive protein; DAS28CRP, disease activity score 28 based on CRP; HAQ, Health Assessment Questionnaire; MTX, methotrexate, PsA, psoriatic arthritis; RA, rheumatoid arthritis; SpA, spondyloarthritis; TNF, tumor necrosis factor; VAS, Visual Analogue Scale.
Data are expressed as the mean (CI) or percentage were indicated. A few patients had missing treatment data or were treated with multiple compounds. No patients received other biological DMARDs besides TNFi.
Ethics
All samples were obtained after informed written consent was provided according to the Declaration of Helsinki and approved by the Local Ethics Committee (the Central Denmark Region committee on health research ethics, project 20121329) and the Danish Data Protection Agency.
Culture conditions
Human PBMCs, SFMCs, and FLSs were cultured with the DMARDs shown in Table 2. In all experiments, untreated cells or cells treated with dimethyl sulfoxide (DMSO) were used as matched controls, and cells cultured with lipopolysaccharide at 100 ng/ml were included as a positive control. Supernatants were harvested after centrifugation of the culture plates at 1200 rpm for 5 minutes, and the cell‐free supernatant was thereafter stored at −80°C for later analysis. Monocyte chemoattractant protein 1 (MCP‐1) was chosen as a readout because this chemokine is highly increased in both RA and SpA 26, 27. Furthermore, MCP‐1 has been extensively studied in a variety of inflammatory diseases and is known to be produced by many cells, including monocytes and fibroblasts. Finally, we had to choose a readout not directly affected by any of the included drugs 28.
Table 2.
List of disease‐modifying antirheumatic drugs used in the ex vivo cultures
| Category | Drug Names | ||||||||
|---|---|---|---|---|---|---|---|---|---|
| Generic name | Adalimumab | Etanercept | Tocilizumab | Anakinra | Ustekinumab | Secukinumab | Tofacitinib | Baricitinib | Methotrexate |
| Commercial name | Humira | Enbrel | RoActemra | Kineret | Stelara | Cosentyx | Tofacitinib citrate | Baricitinib citrate | Ebetrex |
| Source | AbbVie | Pfizer | Roche | Swedish Orphan Biovitrum | Janssen | Novartis | Selleckchem | Selleckchem | Sandoz |
| Concentration | 5 μg/ml | 5 μg/ml | 5 μg/ml | 10 μg/ml | 5 μg/ml | 5 μg/ml | 200 nM | 200 nM | 0.5 μg/ml |
| Target | TNFα | TNFα | IL‐6R | IL‐1R | IL‐12/23 | IL‐17A | JAK1/JAK2 | JAK1/JAK3 | Not known |
Abbreviation: IL, interleukin; JAK, Janus kinase; TNF, tumor necrosis factor.
SFMC 48‐hour ex vivo model
The SFMC 48‐hour culture is an in vitro model of IMIA dominated by lymphocytes and monocytes 20, 29. SFMCs were isolated by conventional Ficoll‐Paque (GE Healthcare) density‐gradient centrifugation and cryopreserved at −150°C. The cells were then thawed and seeded in Dulbecco's modified Eagle's medium, 10% fetal calf serum, penicillin, streptomycin, and glutamine (culture medium) for 48 hours at a concentration of 1 × 106 cells/ml (n = 14) with or without the DMARDs in Table 2 and were kept in a humidified incubator at 37°C and 5% CO2 as described previously 30. After 48 hours, the cell‐free culture supernatants were analyzed for the concentration of MCP‐1.
SFMC 21‐day ex vivo model
The 21‐day culture is an in vitro model of low‐grade inflammatory osteoclastogenesis containing macrophage‐like synovial cells 21. Long‐term cultures were performed as described for the SFMC 48‐hour model, changing the medium every 2 to 3 days as previously described for a total of 21 days with or without the DMARDs in Table 2 21, 31 (n = 10). Culture supernatants were analyzed for the enzyme activity of tartrate‐resistant acid phosphatase (TRAP).
FLS 48‐hour ex vivo model
The 48‐hour co‐culture with FLSs and PBMCs is an in vitro model of IMIA dominated by FLSs 23. FLSs were grown from SFMCs, and the medium was changed every 2 to 3 days. When the cell layer was 70% confluent, the FLSs were passaged by trypsin/EDTA treatment and used for analyses at passage 4 to 5 as previously described 32, 33. The FLSs were then cultured at 2 × 104 cells/ml with either allogenic PBMCs from a healthy donor (n = 6) or paired autologous PBMCs (n = 6) at 1 to 2 × 106 cells/ml in culture medium for 48 hours. Initially, FLS and PBMC monocultures were used as controls. Culture supernatants were analyzed for the concentration of multiple cytokines (interferon‐gamma [IFN‐γ], MCP‐1, TNFα, interleukin [IL]‐1β, IL‐2, IL‐4, IL‐6, IL‐8, IL‐10, IL‐12p70, and IL‐13). The co‐cultures with FLSs and autologous PBMCs were then grown in culture medium for 48 hours (n = 6) with or without the DMARDs in Table 2. The culture supernatants were analyzed for the concentration of MCP‐1.
MCP‐1 enzyme‐linked immunosorbent assay and TRAP measurement
The concentration of MCP‐1 was analyzed by a commercially available enzyme‐linked immunosorbent assay (Biolegend) according to the manufacturer's instructions. The concentration of TRAP was analyzed by an enzymatic assay (B‐bridge International) according to the manufacturer's instructions.
Electrochemoluminescence‐based sandwich immunoassay
The concentrations of IFN‐γ, TNFα, IL‐1β, IL‐2, IL‐4, IL‐6, IL‐8, IL‐10, IL‐12p70, and IL‐13 were analyzed using multiplex electrochemoluminescence‐based sandwich immunoassay (V‐PLEX Pro‐inflammatory Panel 1, Meso Scale Discovery) according to the manufacturer's instructions.
Statistics
Statistical analyses and graphs were done using GraphPad Prism 7 for Mac (GraphPad Software). All data were transformed to ratios by dividing the value of the samples with the value of either untreated cultures (biological DMARDs and methotrexate) or DMSO‐treated cultures (tofacitinib and baricitinib). Data were analyzed with paired or unpaired t test. A two‐sided P value < 0.05 was considered statistically significant.
Results
Most DMARDs reduced the MCP‐1 production from IMIA SFMCs cultured for 48 hours
We first tested whether the different DMARDs affected production of MCP‐1 by SFMCs. SFMCs were cultured for 48 hours with or without DMARDs (Table 2). All DMARDs except anakinra resulted in a decrease in MCP‐1 production in most cultures. The reduction of MCP‐1 was significantly different from controls in cultures with methotrexate (0.93 mean [confidence interval (CI), 0.86‐1.00; P < 0.05]), adalimumab (0.74 mean [CI, 0.55‐94; P < 0.05]), etanercept (0.77 mean [CI, 0.60‐93; P < 0.01]), ustekinumab (0.80 mean [CI, 0.70‐0.91; P < 0.01]), secukinumab (0.81 mean [CI, 0.72‐0.90; P < 0.001]), and baricitinib (0.75 mean [CI, 0.60‐0.90; P < 0.05]). The decrease seen with tocilizumab and tofacitinib did not reach statistical significance (Figure 1). Furthermore, the response to the different DMARDs was analyzed separately for RA, PsA, and SpA. There were no obvious differences in treatment responses in the three disease subgroups. However, the low sample size in the PsA and SpA subgroups makes it impossible to do statistical analyses (Supplementary Figure 1).
Figure 1.

Secretion of monocyte chemoattractant protein (MCP‐1; n = 14) by synovial fluid mononuclear cells (SFMCs) cultured for 48 hours in culture medium, treated with dimethyl sulfoxide (DMSO), or treated with the panel of different disease‐modifying antirheumatic drugs (DMARDs) in Table 2. A, Data are expressed as concentrations. B, Data are expressed as ratios of the concentration of MCP‐1 in DMARD‐treated cultures divided by the concentration in untreated cultures (biological DMARDs and methotrexate) or DMSO‐treated cultures (tofacitinib and baricitinib). Differences were analyzed using the paired t tests to compare two groups. Bars indicate mean and SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
TNFα inhibitors were the only DMARDs to inhibit TRAP‐positive osteoclastogenesis from adherent synovial cells
We used SFMCs cultured for 21 days as an inflammatory osteoclastogenesis model. Here, adalimumab and etanercept robustly reduced the secretion of TRAP (0.78 mean [CI, 0.63‐0.92]) and (0.67 mean [CI, 0.58‐0.76; P < 0.01 and P < 0.0001]). Anakinra showed a significant but very modest reduction in the secretion of TRAP (0.93 mean [CI, 0.87‐0.99; P < 0.05]). In the SFMCs cultured for 21 days, methotrexate, ustekinumab, secukinumab, tocilizumab, tofacitinib, and baricitinib did not significantly decrease the secretion of TRAP (Figure 2). The responses to the different DMARDs were again analyzed separately for RA, PsA, and SpA without obvious differences (Supplementary Figure 2).
Figure 2.

Production of tartrate‐resistant acid phosphatase (TRAP; n = 10) by synovial fluid mononuclear cells (SFMCs) cultured for 21 days in culture medium, treated with dimethyl sulfoxide (DMSO) control or treated with the panel of different disease‐modifying antirheumatic drugs (DMARDs) in Table 2. A, Data are expressed as TRAP activity. B, Data are expressed as ratios of the activity of TRAP in DMARD‐treated cultures divided by the concentration in untreated cultures (biological DMARDs and methotrexate) or DMSO‐treated cultures (tofacitinib and baricitinib). Differences were analyzed using the paired t tests to compare two groups. Bars indicate mean and SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Co‐cultures with FLSs and PBMCs produced higher levels of MCP‐1 than monocultures
We next evaluated the effect of the DMARDs in an inflammatory environment dominated by fibroblasts. First, we addressed whether untreated co‐cultures of FLSs with PBMCs were different from monocultures measured as the production of MCP‐1. In co‐cultures with FLSs and PBMCs, there was much higher production of MCP‐1 (3890.0 pg/ml mean) compared with monocultures of either FLSs (7.8 pg/ml mean [cutoff]) or PBMCs (124.0 pg/ml mean) measured after 48 hours. However, none of these results were statistically significant because of the small number of cultures (n ≤ 2) (Supplementary Figure 3A).
Cytokine production in FLS co‐cultures with autologous PBMCs and FLS co‐cultures with allogenic PBMCs was not statistically different
We then investigated whether co‐culture of FLSs and PBMCs was influenced by the PBMCs being either autologous or allogenic. This difference was evaluated by measuring IFN‐γ, TNFα, IL‐1β, IL‐2, IL‐4, IL‐6, IL‐8, IL‐10, IL‐12p70, IL‐13, and MCP‐1 in the co‐culture supernatants. Overall, production from co‐cultures of FLSs with allogenic PBMCs was 1.8 (1.8 mean [CI 1.5‐2.1]) times higher than the production by co‐cultures of FLSs with autologous PBMCs. The amount of IL‐12p70, IL‐1β, IL‐2, and TNFα on average more than doubled in co‐cultures with allogenic PBMCs compared with autologous PBMCs. None of these differences reached statistical significance (Supplementary Figure 3B).
Tocilizumab considerably reduced the production of MCP‐1 from co‐cultures with FLSs and autologous PBMCs
Next, the effects of different DMARDs were evaluated in the co‐cultures of FLSs and autologous PBMCs. Tocilizumab (0.449 mean [CI, 0.30‐0.59; P < 0.001]) and the two Janus kinase (JAK) inhibitors tofacitinib (0.49 mean [CI, 0.34‐64; P < 0.05]) and baricitinib, (0.38 mean [CI, 0.27‐49; P < 0.05]) were exclusive in lowering the production of MCP‐1 by roughly 50%. A significant decrease was also observed in cultures with ustekinumab (0.82 mean [CI, 0.70‐0.94; P < 0.05]), secukinumab (0.78 mean [CI, 0.58‐0.97; P < 0.05]), and methotrexate (0.68 mean [CI, 0.58‐79; P < 0.001]), though to a much lesser extent. Furthermore, the level of MCP‐1 was significantly lower in tocilizumab‐treated co‐cultures compared with co‐cultures treated with either of the TNFα inhibitors adalimumab (0.80 mean [CI, 0.51‐1.1; P < 0.05]) or etanercept (0.91 mean [CI, 0.62‐1.2; P < 0.01]) (Figure 3). The response to the different DMARDs was again analyzed separately for RA, SpA, and PsA without obvious differences (Supplementary Figure 4).
Figure 3.

Secretion of monocyte chemoattractant protein (MCP‐1; n = 6) by co‐cultures of fibroblast‐like synovial cells (FLSs) and autologous peripheral blood mononuclear cells (PBMCs) cultured for 48 hours in culture medium, treated with dimethyl sulfoxide (DMSO) control, or treated with the panel of different disease‐modifying antirheumatic drugs (DMARDs) in Table 2. A, Data are expressed as concentrations. B, Data are expressed as ratios of the concentration of MCP‐1 in DMARD‐treated cultures divided by the concentration in untreated cultures (biological DMARDs and methotrexate) or DMSO‐treated cultures (tofacitinib and baricitinib). Differences were analyzed using the paired t tests to compare two groups. Bars indicate mean and SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Discussion
One of the big challenges in the management and treatment of IMIA is to choose the right treatment to the right patient 34, 35. Recent findings indicate that it might be possible to stratify patients based on synovial pathobiology and cellular subsets found in the peripheral blood 7, 16, 36, 37. Subtyping these diseases based on phenotyping and pathobiology may provide important insights into the heterogeneity of RA, SpA, and PsA, opening the possibility of treatment matched to the dominating pathogenic pathways or immunological mechanism 38. This approach is already being implemented in oncology and hematology.
Our study reveals a fairly diverse response signature between various DMARDs in different ex vivo models. First, SFMC 48‐hour cultures were used. This model has been extensively used following the identification of the in vitro effect of TNFα inhibition 3 decades ago 29. In the SFMC 48‐hour cultures, all DMARDs except anakinra effectively decreased MCP‐1 production. This is in line with the literature showing that IL‐1 receptor blockade is less effective in the treatment of RA compared with other biological DMARDs such as IL‐6 or TNFα inhibition 39. Anti‐TNFα treatment was found to most effectively decrease the MCP‐1 production. The decrease in MCP‐1 could be caused by either a decrease in the production of MCP‐1 per monocyte or a change in the number or subsets of monocytes.
Next, SFMC 21‐day cultures were studied. This is a simplistic model of inflammatory osteoclastogenesis without the addition of macrophage colony‐stimulating factor or receptor activator of nuclear factor kappa‐Β ligand 22. In this model, only anti‐TNFα treatment reduced the production of TRAP and was thus superior to all the other DMARDs we tested. This is in line with the finding that anti‐TNFα treatment reduces erosive joint damage in RA 15 even though the literature on this matter is contradictory. Our SFMC 21‐day culture model has specific limitations because the osteoclast formation in our model is only modest and potentially not effective enough to detect small treatment effects. This implies that some of the DMARDs could potentially have an effect in models with more pronounced osteoclastogenesis.
Finally, co‐cultures of FLSs and PBMCs were used. The differences observed between the co‐cultures with FLSs and autologous PBMCs compared with allogenic PBMCs points to an increased activation in cultures with allogenic PBMCs in which T cells encounter foreign FLSs. This could be due to a degree of graft‐versus‐host response. However, of note, the autologous and allogenic responses were comparable without statistically significant differences. IL‐6 receptor inhibition with tocilizumab showed a selective effect on co‐cultures of FLSs and autologous PBMCs, whereas anti‐TNFα treatment had no or very limited effect. These findings are supported by a recent study showing that IL‐6 is central for the transformation of FLSs in early RA from an immunosuppressive to an inflammatory phenotype 40. The IL‐6 pathway has been further shown to be important in FLSs, evaluated by joint‐specific DNA methylation in RA 41. Furthermore, IL‐6 knockout mice have been shown to be protected from developing arthritis 42. IL‐6 is primarily produced by FLSs and mediates the recruitment and activation of T cells at sites of chronic inflammation 43, 44. Taken together, this study underlines the central role of IL‐6 in synovial microenvironments dominated by FLSs.
Methotrexate induced a significant reduction of MCP‐1 production by both SFMC 48‐hour cultures and co‐cultures of FLSs and PBMCs. This finding is in line with the actions of methotrexate in arthritis being both immunosuppressive and anti‐proliferative. Methotrexate is known to lower both IL‐6 and TNFα production 45.
It is interesting that anti‐TNFα had the most pronounced effect in the SFMC 21‐day cultures, whereas tocilizumab showed the most robust decrease in MCP‐1 production in the FLS and PBMC co‐cultures. This finding indicates that differences in cellular subsets in the inflammatory microenvironment are likely to determine the effectiveness of DMARDs with differences in mode of action. This is in line with studies showing that targeting the IL‐6 receptor is beneficial in RA anti‐TNFα nonresponders 46. The different response signatures between anti‐TNFα and tocilizumab observed in this study are also supported by the fact that the primary downstream targets are different. Tocilizumab affects the JAK/signal transducer and activator of transcription proteins pathway. In contrast, anti‐TNFα primarily affects the NF‐κB pathway 8. The two JAK inhibitors tofacitinib and baricitinib were also found to profoundly decrease the production of MCP‐1 in the FLS and PBMC co‐cultures. These JAK inhibitors modulate IL‐6 production by inhibition of JAK1/JAK2 47. This could be the mechanism for the treatment effect in the FLS‐dominated ex vivo model. However, JAK inhibition also modulated signaling through many other cytokine receptors, preventing signaling from IFN‐γ, IL‐2, IL‐7, and IL‐23. Taken together, the present study supports the hypothesis that different pathotypes of IMIA respond differently to different DMARD treatments. Specifically, this study substantiates that patients with the fibroid pathotype could probably benefit from a treatment targeting the IL‐6 axis rather than anti‐TNFα treatment, as has been suggested previously 41.
Our study has some notable limitations. First, the ex vivo study findings cannot be translated directly into an in vivo setting. It is unlikely that these models truly reflect disease pathotypes of IMIA. Furthermore, the cellular composition and activation state of specific signaling pathways could be very different in comparing the patient samples. This might explain some of the variation in the treatment response for each of the DMARDs in each of the ex vivo models. In line with this, the patients received different treatments at the time of sampling (Table 1). This could also influence the results. However, patients were primarily receiving methotrexate or a TNFα inhibitor, and these compounds still showed significant treatment effects in the SFMC 48‐hour cultures (even though the patients could be classified as at least partial nonresponders to these drugs). Drug concentrations were chosen in order to simulate in vivo plasma concentrations as well as possible (Table 2). Our in vitro models also have some strengths. First, the in vitro design allowed paralleled assessment of different DMARDs on cells activated in vivo with no further ex vivo stimulation. Furthermore, the in vitro experimental design offers us the ability to isolate the different components of arthritis pathobiology and evaluate them separately.
This study reveals a fairly sharp distinction between the efficacy profiles of the different DMARDs on different cellular subsets derived from the synovium of patients with IMIA. Tocilizumab showed a clear reduction in an FLS‐dominated environment, whereas anti‐TNFα treatment had the most pronounced effects in the SFMC 21‐day culture. This study herby points to a different efficacy profile of the different DMARDs, depending on the cellular subsets present in the in vitro culture. This could be of great interest in the future when synovial biopsies or other stratification tools are further established in the clinic and holds promises for future studies on treatments based on the composition of the synovial environment.
Author Contributions
Drs. Nielsen, Buckley, and Kragstrup drafted the manuscript. All authors were involved in revising the manuscript and read and approved the final manuscript.
Study conception and design
Nielsen, Kragstrup.
Acquisition of data
Nielsen, Lomholt, Mellemkjær, Andersen, Kragstrup.
Analysis and interpretation of data
Nielsen, Buckley, Kragstrup.
Supporting information
Acknowledgments
We thank Karin Skovgård Sørensen (Department of Biomedicine, Aarhus University) for technical assistance concerning the ELISA data. We thank Professor Bent Deleuran (Department of Biomedicine, Aarhus University) for the use of laboratory equipment and for supervision of the project. We thank medical doctors and nurses at the Department of Rheumatology, Aarhus University Hospital for helping to collect the patient samples. Additionally, please contact corresponding author for data requests.
This work was supported by Independent Research Fund Denmark (9039‐00015B), the Faculty of Health at Aarhus University, The Danish Rheumatism Association, Aage Bang Foundation, Augustinus Foundation, Nyegaard Foundation, Bjarne Jensen Foundation, A. P. Møller Foundation, and Axel Muusfeldt Foundation.
Dr. Kragstrup has engaged in educational activities talking about immunology in rheumatic diseases receiving speaking fees from Pfizer (<$10,000), speaking fees from Bristol‐Myers Squibb (<$10,000), speaking fees from Eli Lilly (<$10,000), speaking fees from Novartis (<$10,000), and speaking fees from UCB (<$10,000) and has received a consultancy fee from Bristol‐Myers Squibb (<$10,000). No other disclosures relevant to this article were reported.
References
- 1. Taylor PC. Developing anti‐TNF and biologic agents. Rheumatology (Oxford) 2011;50:1351–3. [DOI] [PubMed] [Google Scholar]
- 2. Atzeni F, Sarzi‐Puttini P, Gorla R, Marchesoni A, Caporali R. Switching rheumatoid arthritis treatments: an update. Autoimmun Rev 2011;10:397–403. [DOI] [PubMed] [Google Scholar]
- 3. Baeten D, Baraliakos X, Braun J, Sieper J, Emery P, van der Heijde D, et al. Anti‐interleukin‐17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double‐blind, placebo‐controlled trial. Lancet. 2013;382:1705–13. [DOI] [PubMed] [Google Scholar]
- 4. Schett G, Elewaut D, McInnes IB, Dayer JM, Neurath MF. How cytokine networks fuel inflammation: Toward a cytokine‐based disease taxonomy. Nat Med 2013;19:822–4. [DOI] [PubMed] [Google Scholar]
- 5. Nam JL, Takase‐Minegishi K, Ramiro S, Chatzidionysiou K, Smolen JS, van der Heijde D, et al. Efficacy of biological disease‐modifying antirheumatic drugs: a systematic literature review informing the 2016 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2017;76:1113–36. [DOI] [PubMed] [Google Scholar]
- 6. Navarro‐Compán V, Plasencia‐Rodríguez C, de Miguel E, Diaz Del Campo P, Balsa A, Gratacós J. Switching biological disease‐modifying antirheumatic drugs in patients with axial spondyloarthritis: results from a systematic literature review. RMD Open 2017;3:e000524–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Pitzalis C, Kelly S, Humby F. New learnings on the pathophysiology of RA from synovial biopsies. Curr Opin Rheumatol 2013;25:334–44. [DOI] [PubMed] [Google Scholar]
- 8. Dennis G Jr, Holweg CT, Kummerfeld SK, Choy DF, Setiadi AF, Hackney JA, et al. Synovial phenotypes in rheumatoid arthritis correlate with response to biologic therapeutics. Arthritis Res Ther 2014;16:R90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Townsend MJ. Molecular and cellular heterogeneity in the Rheumatoid Arthritis synovium: clinical correlates of synovitis. Best Pract Res Clin Rheumatol 2014;28:539–49. [DOI] [PubMed] [Google Scholar]
- 10. Dawidowicz K, Allanore Y, Guedj M, Pierlot C, Bombardieri S, Balsa A, et al. The interferon regulatory factor 5 gene confers susceptibility to rheumatoid arthritis and influences its erosive phenotype. Ann Rheum Dis 2011;70:117–21. [DOI] [PubMed] [Google Scholar]
- 11. Queiro‐Silva R, Torre‐Alonso JC, Tinturé‐Eguren T, López‐Lagunas I. A polyarticular onset predicts erosive and deforming disease in psoriatic arthritis. Ann Rheum Dis 2003;62:68–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Razawy W, van Driel M, Lubberts E. The role of IL‐23 receptor signaling in inflammation‐mediated erosive autoimmune arthritis and bone remodeling. Eur J Immunol 2018;48:220–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Forsblad‐d'Elia H, Bengtsson K, Kristensen LE, Jacobsson LT. Drug adherence, response and predictors thereof for tocilizumab in patients with rheumatoid arthritis: results from the Swedish biologics register. Rheumatology (Oxford) 2015;54:1186–93. [DOI] [PubMed] [Google Scholar]
- 14. Humby FC, Balushi Al F, Lliso G, Cauli A, Pitzalis C. Can synovial pathobiology integrate with current clinical and imaging prediction models to achieve personalized health care in rheumatoid arthritis? [Review]. Front Med (Lausanne) 2017;4:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Lipsky PE, van der Heijde DMFM, Clair EWS, Furst DE, Breedveld FC, Kalden JR, et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti‐Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med 2000;343:1594–602. [DOI] [PubMed] [Google Scholar]
- 16. Miyagawa I, Nakayamada S, Nakano K, Kubo S, Iwata S, Miyazaki Y, et al. Precision medicine using different biological DMARDs based on characteristic phenotypes of peripheral T helper cells in psoriatic arthritis. Rheumatology (Oxford) 2019;58:336–44. [DOI] [PubMed] [Google Scholar]
- 17. Takasugi T, Hollingsworth JW. Morphologic studies of mononuclear cells of human synovial fluid. Arthritis Rheum 1967;10:495–501. [DOI] [PubMed] [Google Scholar]
- 18. Petersen J, Andersen V, Bendixen G, Bendtzen K, Halkjaer‐Kristensen J, Ingemann‐Hansen T, et al. Functional characteristics of synovial fluid and blood mononuclear cells in rheumatoid arthritis and traumatic synovitis. Scand J Rheumatol 1982;11:75–80. [DOI] [PubMed] [Google Scholar]
- 19. Kerkman PF, Kempers AC, van der Voort EI, van Oosterhout M, Huizinga TW, Toes RE, et al. Synovial fluid mononuclear cells provide an environment for long‐term survival of antibody‐secreting cells and promote the spontaneous production of anti‐citrullinated protein antibodies. Ann Rheum Dis 2016;75:2201–7. [DOI] [PubMed] [Google Scholar]
- 20. Kragstrup TW, Jalilian B, Keller KK, Zhang X, Laustsen JK, Stengaard‐Pedersen K, et al. Changes in soluble CD18 in murine autoimmune arthritis and rheumatoid arthritis reflect disease establishment and treatment response. PLoS One 2016;11:e0148486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Greisen SR, Einarsson HB, Hvid M, Hauge EM, Deleuran B, Kragstrup TW. Spontaneous generation of functional osteoclasts from synovial fluid mononuclear cells as a model of inflammatory osteoclastogenesis. APMIS 2015;123:779–86. [DOI] [PubMed] [Google Scholar]
- 22. Feng W, Guo J, Li M. RANKL‐independent modulation of osteoclastogenesis. J Oral Biosci 2019;61:16–21. [DOI] [PubMed] [Google Scholar]
- 23. Tang Y, Wang B, Sun X, Li H, Ouyang X, Wei J, et al. Rheumatoid arthritis fibroblast‐like synoviocytes co‐cultured with PBMC increased peripheral CD4+ CXCR23+ ICOS+ T cell numbers. Clin Exp Immunol 2017;190:384–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Dankers W, González‐Leal C, Davelaar N, Asmawidjaja PS, Mus AM, Hazes JM, et al. 1,25(OH)2D3 and dexamethasone additively suppress synovial fibroblast activation by CCR24+ T helper memory cells and enhance the effect of tumor necrosis factor α blockade. Arthritis Res Ther 2018;20:212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Noack M, Ndongo‐Thiam N, Miossec P. Evaluation of anti‐inflammatory effects of steroids and arthritis‐related biotherapies in an in vitro coculture model with immune cells and synoviocytes. Front Immunol 2016;7:e509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Koch AE, Kunkel SL, Harlow LA, Johnson B, Evanoff HL, Haines GK, et al. Enhanced production of monocyte chemoattractant protein‐1 in rheumatoid arthritis. J Clin Invest 1992;90:772–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Gu J, Rihl M, Märker‐Hermann E, Baeten D, Kuipers JG, Song YW, et al. Clues to pathogenesis of spondyloarthropathy derived from synovial fluid mononuclear cell gene expression profiles. J Rheumatol 2002;29:2159–64. [PubMed] [Google Scholar]
- 28. Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein‐1 (MCP‐1): an overview. J Interferon Cytokine Res 2009;29:313–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Brennan FM, Chantry D, Jackson A, Maini R, Feldmann M. Inhibitory effect of TNF α antibodies on synovial cell interleukin‐1 production in rheumatoid arthritis. Lancet 1989;2:244–7. [DOI] [PubMed] [Google Scholar]
- 30. Nielsen MA, Andersen T, Etzerodt A, Kragstrup TW, Rasmussen TK, Stengaard‐Pedersen K, et al. A disintegrin and metalloprotease‐17 and galectin‐9 are important regulators of local 4‐1BB activity and disease outcome in rheumatoid arthritis. Rheumatology (Oxford) 2016;55:1871–9. [DOI] [PubMed] [Google Scholar]
- 31. Kragstrup TW, Greisen SR, Nielsen MA, Rhodes C, Stengaard‐Pedersen K, Hetland ML, et al. The interleukin‐20 receptor axis in early rheumatoid arthritis: novel links between disease‐associated autoantibodies and radiographic progression. Arthritis Res Ther 2016;18:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Stebulis JA, Rossetti RG, Atez FJ, Zurier RB. Fibroblast‐like synovial cells derived from synovial fluid. J Rheumatol 2005;32:301–6. [PubMed] [Google Scholar]
- 33. Kragstrup TW, Andersen MN, Schiøttz‐Christensen B, Jurik AG, Hvid M, Deleuran B. Increased interleukin (IL)‐20 and IL‐24 target osteoblasts and synovial monocytes in spondyloarthritis. Clin Exp Immunol 2017;189:342–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Emery P, Keystone E, Tony HP, Cantagrel A, van Vollenhoven R, Sanchez A, et al. IL‐6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti‐tumour necrosis factor biologicals: results from a 24‐week multicentre randomised placebo‐controlled trial. Ann Rheum Dis 2008;67:1516–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Nam JL, Ramiro S, Gaujoux‐Viala C, Takase K, Leon‐Garcia M, Emery P, et al. Efficacy of biological disease‐modifying antirheumatic drugs: a systematic literature review informing the 2013 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2014;73:516–28. [DOI] [PubMed] [Google Scholar]
- 36. Van de Sande MG, Gerlag DM, Lodde BM, van Baarsen LG, Alivernini S, Codullo V, et al. Evaluating antirheumatic treatments using synovial biopsy: a recommendation for standardisation to be used in clinical trials. Ann Rheum Dis 2011;70:423–7. [DOI] [PubMed] [Google Scholar]
- 37. Orr C, Vieira‐Sousa E, Boyle DL, Buch MH, Buckley CD, Cañete JD, et al. Synovial tissue research: a state‐of‐the‐art review. Nat Rev Rheumatol 2017;13:463–75. [DOI] [PubMed] [Google Scholar]
- 38. McGonagle D, Watad A, Savic S. Mechanistic immunological based classification of rheumatoid arthritis. Autoimmun Rev 2018;17:1115–23. [DOI] [PubMed] [Google Scholar]
- 39. Tarp S, Furst DE, Dossing A, Østergaard M, Lorenzen T, Hansen MS, et al. Defining the optimal biological monotherapy in rheumatoid arthritis: a systematic review and meta‐analysis of randomised trials. Semin Arthritis Rheum 2017;46:699–708. [DOI] [PubMed] [Google Scholar]
- 40. Filer A, Ward LSC, Kemble S, Davies CS, Munir H, Rogers R, et al. Identification of a transitional fibroblast function in very early rheumatoid arthritis. Ann Rheum Dis 2017;76:2105–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ai R, Hammaker D, Boyle DL, Morgan R, Walsh AM, Fan S, et al. Joint‐specific DNA methylation and transcriptome signatures in rheumatoid arthritis identify distinct pathogenic processes. Nat Commun 2016;7:11849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Alonzi T, Fattori E, Lazzaro D, Costa P, Probert L, Kollias G, et al. Interleukin 6 is required for the development of collagen‐induced arthritis. J Exp Med 1998;187:461–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. McGettrick HM, Smith E, Filer A, Kissane S, Salmon M, Buckley CD, et al. Fibroblasts from different sites may promote or inhibit recruitment of flowing lymphocytes by endothelial cells. Eur J Immunol 2009;39:113–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Patel R, Filer A, Barone F, Buckley CD. Stroma: fertile soil for inflammation. Best Pract Res Clin Rheumatol 2014;28:565–76. [DOI] [PubMed] [Google Scholar]
- 45. Wessels JA, Huizinga TW, Guchelaar HJ. Recent insights in the pharmacological actions of methotrexate in the treatment of rheumatoid arthritis. Rheumatology (Oxford) 2008;47:249–55. [DOI] [PubMed] [Google Scholar]
- 46. Kim HL, Lee MY, Park SY, Park SK, Byun JH, Kwon S, et al. Comparative effectiveness of cycling of tumor necrosis factor‐α (TNF‐α) inhibitors versus switching to non‐TNF biologics in rheumatoid arthritis patients with inadequate response to TNF‐α inhibitor using a Bayesian approach. Arch Pharm Res 2014;37:662–70. [DOI] [PubMed] [Google Scholar]
- 47. O'Shea JJ, Kontzias A, Yamaoka K, Tanaka Y, Laurence A. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis 2013;72 Suppl 2:ii111–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
