
Association between the response to TNF-antagonists and plasma type I interferon activity and interferon-beta/alpha ratios in rheumatoid arthritis patients: a post-hoc analysis of a predominantly Hispanic Cohort
Clio P Mavragani,MD
Dan T La,MD
William Stohl,MD, PhD
Mary K Crow,MD
Corresponding address: Mary K. Crow, MD, Hospital for Special Surgery, 535 East 70th St., New York, NY 10021, Tel: 212-606-1397, Fax: 212-774-2337,crowm@hss.edu
Abstract
Objective
Despite substantial clinical efficacy of tumor necrosis factor-α (TNF)-antagonist therapy in patients with rheumatoid arthritis (RA), some patients respond poorly to such agents. Since an interferon (IFN) signature is variably expressed among RA patients, we investigated whether plasma type I IFN activity might predict response to TNF-antagonist therapy.
Methods
RA patients (n=35), the majority Hispanic, from a single center were evaluated prior to and following initiation of TNF-antagonist therapy. As controls, 12 RA patients from the same center who were not treated with a TNF antagonist were studied. Plasma type I IFN activity was measured using a reporter cell assay and disease status assessed using the Disease Activity Score (DAS28). IL-1 receptor antagonist (IL-1ra) levels were determined in baseline samples using a commercial ELISA. The clinical response was classified according to the European League against Rheumatism (EULAR) RA improvement criteria.
Results
Plasma type I IFN activity at baseline was significantly associated with clinical response (OR=1.36, CI: 1.05-1.76, p=0.02), with high baseline IFN activity associated with a good response. Changes in DAS28 scores were greater among patients with a baseline plasma IFNβ/α ratio >0.8 (indicating elevated plasma IFNβ levels). Consistent with the capacity of IFNβ to induce IL-1ra, elevated baseline IL-1ra levels were associated with better therapeutic outcomes (OR=1.82, CI: 1.1-3.29, p=0.027).
Conclusion
Plasma type I IFN activity, IFNβ/α ratio, and IL-1ra levels were predictive of therapeutic response in TNF-antagonist-treated RA patients, indicating that those parameters might define clinically meaningful subgroups of RA patients with distinct responses to therapeutic agents.
INTRODUCTION
Treatment of rheumatoid arthritis (RA) patients with biologic agents that antagonize tumor necrosis factor-α (TNF) has resulted in decreased morbidity and mortality as well as clinically meaningful improvement in quality of life. Nevertheless, a considerable proportion of RA patients, ranging from 20 to 50% in clinical trials, has failed to mount adequate clinical or radiographic responses to these agents (1-5). Given the high cost and potential serious toxicities associated with TNF antagonists, identification of predictors of response to TNF-antagonist therapy should help optimize the clinical management of RA patients.
Previous analyses have pointed to several demographic, clinical, serologic, or gene expression parameters as being independently associated with poor response to TNF-antagonist therapy, and high levels of inflammation and TNF expression in the synovial membrane of RA patients prior to treatment may be characteristics associated with favorable response to TNF-antagonist therapy (6-13). We have previously reported that plasma levels of B lymphocyte stimulator (BLyS) are higher in TNF-antagonist responder RA patients than in TNF-antagonist poor responders, although the difference did not achieve statistical significance (14). Of great importance, none of the features studied to date has demonstrated a predictive power and practical utility sufficient to guide routine clinical practice. The clinician caring for the RA patient still faces a 30-50% likelihood that his/her patient will not adequately respond to the costly and potentially toxic TNF antagonist.
The differential responsiveness among RA patients to TNF antagonists might reflect genetic differences among individuals or heterogeneity in disease pathogenesis per se, as illustrated by variability in clinical features, autoantibody profiles, or activation of distinct immunologic mechanisms or molecular pathways. A recent genome-wide association scan in 89 RA patients treated with TNF antagonists identified significant associations between response to therapy and 16 single nucleotide polymorphisms, but none of those associations has as yet been replicated (15). While recent progress in genome-wide association studies may ultimately lead to diagnostics helpful in patient management, the currently available data do not suggest that genetic variations will be useful in predicting response to biologic agents in the near or intermediate term.
Recent studies of gene expression in peripheral blood mononuclear cells (PBMC) and synovial membranes from RA patients suggest that certain patients may harbor distinct gene expression patterns. Of interest, a pathogen-response gene expression program, characterized by increased expression of type I interferon (IFN)-inducible genes, has been identified in a subgroup of RA patients who also expressed high circulating anti-CCP levels (16-17). The potential relevance of type I IFN, and more specifically IFNβ, in RA is suggested by in vitro studies of RA synovial membrane and experiments in murine models of inflammatory arthritis. IFNβ is a type I IFN with pleiotropic immunomodulatory actions, including decreased expression of the proinflammatory cytokines IL-1β and TNF-α and enhancement of the anti-inflammatory cytokines IL-1 receptor antagonist (IL-1ra), IL-10, and transforming growth factor β (18-21). It has also been shown to mediate inhibition of MHC class II expression on activated PBMC (21), inhibition of T-cell activation (22) and decreased expression of adhesion molecules (23). In collagen-induced and adjuvant arthritis models, intraperitoneal or intraarticular injection of IFNβ resulted in reduction of disease activity and inhibition of cartilage and bone destruction through a significant decrease of TNF and IL-6 expression and enhancement of IL-10 responses at the site of inflammation (24-26). IFNβ is present in RA synovial membranes and reduces synoviocyte proliferation in vitro, observations that have led to the suggestion that IFNβ is an anti-inflammatory mediator that serves a protective role in RA (27-29).
In view of the high cost of treatment and the potentially serious adverse effects, identification of predictors of response to TNF antagonists would be highly useful in clinical practice. With the availability of additional new therapeutic options, patients predicted to be TNF-antagonist non-responders might be more successfully treated with other agents, leading to earlier therapeutic responses. Inasmuch as type I IFNs, particularly IFNβ, have been associated with anti-inflammatory activities in the setting of RA and in view of the variable expression of an IFN signature among RA patients (16-17), we postulated that expression of type I IFN might represent a positive predictor of response to TNF-antagonist therapy in RA patients, while low levels of type I IFN might identify RA patients who would be candidates for alternative therapeutic options. To pursue this hypothesis, we took advantage of the availability of plasma samples from a previously described RA cohort (14) to determine type I IFN activity prior to TNF-antagonist therapy and perform a post-hoc analysis in relation to clinical response.
PATIENTS AND METHODS
Study Subjects
The study population included three subject cohorts: 1) RA patients receiving their care at the Los Angeles County + University of Southern California Medical Center (LAC+USC MC) Rheumatology clinics (n = 38) who, based on the clinical judgment of their attending physicians, required the addition of a TNF antagonist (etanercept, n = 16; infliximab, n = 9; adalimumab, n = 13) to their therapeutic regimens and who agreed to such treatment; 2) RA patients receiving medical care at the same clinics (total n = 12) who either refused (n = 2) or were felt not to be candidates for TNF-antagonist therapy (n=10); and 3) healthy volunteers (n = 50) of age and sex distribution similar to that of the RA cohorts. Two of the 38 TNF-antagonist-treated patients were excluded from analysis based on less than 8 weeks monitoring on therapy, and one patient was excluded due to pregnancy during the course of the study, possibly confounding interpretation of disease activity. Each of the 3 excluded RA patients was on a distinct TNF-antagonist agent. Baseline characteristics of the RA cohorts have been previously described (14). Consistent with the LAC+USC MC patient population at-large, the majority of the study subjects were Hispanic (33 of 35). Healthy control subjects selected for comparison included a comparable proportion of Hispanic individuals.
Clinical Outcomes
Outcome was evaluated during a window of therapy greater than 3 months and less than 9 months, allowing for sufficient time for clinical response, and was categorized by the Disease Activity Score (DAS28), as defined by the EULAR RA improvement criteria (30,31), into three groups: non-response, moderate response, and good response, based on the absolute DAS28 at follow-up (mean 5.6±1.4 months after initiation of TNF-antagonist therapy for TNF-antagonist-treated patients; 8.8±2.9 months for non-treated control patients) and the change in DAS28 from baseline. To be considered a good responder, the patient must have experienced an improvement of ≥1.2 in DAS28 with an absolute DAS28 of ≤3.2. To be considered a non-responder, the patient must have experienced an improvement of <0.6 in DAS28 with an absolute DAS28 of >5.1. If the patient met criteria for neither good responder nor non-responder he/she was classified as a moderate responder.
Plasma type I IFN activity
Type I IFN activity was detected using a reporter cell assay (32-33). In brief, cells of the WISH epithelial cell line (ATCC #CCL-25, Manassas, VA) express the type I IFN receptor and are highly responsive to type I IFN. WISH cells were plated at a density of 5×105 cells/ml in 96 well plates in Minimum Essential Media (Cellgro, Herndon, VA) with 10% fetal calf serum (FCS) and then were cultured with 50% patient plasma for 6 hours. Recombinant human IFNα (IFNaA, Biosource International, Camarillo, CA) and media only were used as positive and negative controls, respectively.
Total cellular mRNA was purified from stimulated cells at the end of the culture period using the Qiagen Turbocapture oligo-dT coated 96 well plate system as per the manufacturer’s protocol (Qiagen, Valencia, CA). Total cellular mRNA was reverse-transcribed to cDNA immediately following purification using the TaqMan® Reverse Transcription Reagents (Applied Biosystems, Foster City, CA).
Quantitative real-time polymerase chain reaction (PCR) was used to quantify specific cDNAs using the Bio-Rad SYBR Green intercalating fluorophore system with a Bio-Rad I-cycler thermocycler and fluorescence detector (Bio-Rad, Hercules, CA). Primers for genes that are highly induced by type I IFN signaling were used in the PCR reaction on the WISH cell-derived cDNAs (Operon, Huntsville, AL): interferon induced with tetratricopeptide repeats 1 (IFIT-1, Forward CTCCTTGGGTTCGTCTATAAATTG; Reverse AGTCAGCAGCCAGTCTCAG), double-stranded RNA-dependent protein kinase (PKR) (Forward CTTCCATCTGACTCAGGTTT; Reverse TGCTTCTGACGGTATGTATTA) and myxovirus resistance 1 (MX-1) (Forward TACCAGGACTACGAGATTG; Reverse TGCCAGGAAGGTCTATTAG). The housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH, Forward CAACGGATTTGGTCGTATT; Reverse GATGGCAACAATATCCACTT) was also quantified in the cDNA samples to control for background gene expression. Threshold values were recorded for each sample at the logarithmic portion of the amplification. Melt curve analysis was used to ensure the specificity of the PCR product. The type I IFN-induced genes were compared with housekeeping gene expression to determine relative expression. The relative expression was then normalized to the relative expression of the respective genes in unstimulated cells from the same cell preparation. RA samples were compared to the mean and SD of a pool of healthy donors in the same assay, and the sum of the number of SD above healthy donors for each of the three genes was calculated for each sample (34).
Determination of IFNβ/α ratio
To determine whether the capacity of RA plasma to stimulate expression of type I IFN-inducible genes is predominantly due to the presence of IFNα or IFNβ, samples from TNF-antagonist-treated RA patients with detectable type I IFN activity at baseline were incubated with monoclonal anti-IFNα or anti-IFNβ antibody (10μg/ml) (Chemicon, Temecula, CA) prior to addition to WISH cells. These antibodies have previously been shown to be specific for their designated IFNs (data not shown). An IFNβ/α ratio was calculated based on the ratio of % inhibition of type I IFN activity by anti-IFNβ to the % inhibition of type I IFN activity by anti-IFNα antibody.
IL-1 receptor antagonist (IL-1ra) ELISA
Determination of IL-1ra levels was performed by a solid-phase ELISA according to manufacturer’s instructions (Quantikine HS, R&D Systems, Minneapolis, US). This assay employs the quantitative “sandwich” enzyme immunoassay technique.
Statistical Analysis
Comparison between continuous variables was performed by the paired t-test for matched data and by the Mann-Whitney test for non-matched data. Comparisons between categorical variables were performed by Fisher’s exact two sided tests. Comparisons between three groups were performed using the one-way ANOVA test and Kruskal-Wallis test. Correlations between continuous variables were determined using the nonparametric Spearman’s test. Type I IFN and IL-1ra baseline levels were analyzed using both univariate and multivariate ordinal logistic regression to model the probability of achieving a higher response category in the presence of each predictor variable. A logistic multivariate model was constructed to identify independent predictors that could be associated with different therapeutic outcomes. Age, which was shown to be statistically significantly different between non-, moderate, and good responder groups (p<0.05) at baseline, was included in a multivariate model. Results have been presented as the odds ratio (OR) with corresponding 95% confidence intervals (CI).
RESULTS
Demographics
The baseline characteristics of the TNF-antagonist-treated RA patients according to response to therapy are presented inTable 1. While no statistically significant differences were revealed among the three groups in sex, race, medications, disease activity, disease duration, or prevalence of RF or anti-CCP positivity, the moderate responders were significantly older than the good responders (p<0.05).
Table 1. Baseline characteristics of anti-TNF treated RA patients classified as good, moderate and non-responders according to EULAR response criteria.
| Non-Response (n=11) | Moderate Response (n=17) | Good Response (n=7) | |
|---|---|---|---|
| Agea | 49 (33-64) | 49 (32-64)* | 39 (25-51) |
| Sex (F/M)b | 11/0 (100) | 16/1 (94) | 7/0 (100) |
| Race (H/A/C)c | 9/1/1 | 17/0/0 | 7/0/0 |
| Disease durationd | 10.01 (0.4-30.4) | 10.6 (0.4-30.7) | 7.5 (2.7- 13.1) |
| DAS28e | 5.9 (4.29-8) | 6.5 (4.9-8.0) | 5.7 (4.5-7.4) |
| Prednisonef | 3/11 (27; 2.1mg/d) | 6/17 (35; 2.2mg/d) | 4/7 (57; 4.5mg/d) |
| DMARDg | |||
| MTX | 11/11 (100) | 14/17 (82) | 5/7 (71) |
| HCQ | 7/11 (64) | 7/17 (41) | 6/7 (86) |
| LEF | 1/11 (9) | 3/17 (18) | 1/7 (14) |
| SSA | 3/11 (27) | 5/17 (29) | 2/7 (29) |
| RF positivityh | 7/9 (78) | 16/16 (100) | 6/7 (86) |
| anti-CCP positivityh,i | 5/5 (100) | 8/10 (80) | 3/4 (75) |
Mean (range) in years
F=Female; M=Male; % female indicated in parentheses.
H=Hispanic, A=Asian, C=Caucasian
Mean (range) in years
Mean (range)
Number of patients taking prednisone; % taking prednisone and mean daily dose among patients taking the medication indicated in parentheses.
Number of patients taking the indicated DMARD. MTX=Methotrexate; HCQ=hydroxychloroquine; LEF=Leflunomide; SSA=sulfasalazine; % taking each drug indicated in parentheses.
percentages of patients with positive RF or anti-CCP titers (>20IU/ml); % RF or anti-CCP positive in parentheses.
data available for 20 patients
p<0.05 (Comparison between good and moderate response group). All other comparisons non- significant
Increased plasma type I IFN activity in some RA patients
The WISH epithelial cell assay was used to determine the capacity of plasma to induce expression of IFN-regulated genes. As shown inFigure 1A, high type I IFN levels (defined as levels above the mean + 2 SD of a pool of healthy controls tested in the same assay) were detected in 29.7% (14/47) of all RA patients at baseline compared to 6.3% (3/47) of healthy controls (p<0.0001) of comparable age, gender, and ethnicity. These results represent a lower proportion of patients and a lower mean IFN score compared to results observed in SLE patients (47%; >10 mean IFN activity score among SLE patients meeting 4 classification criteria) (34). No baseline differences in type I IFN activity were detected between anti-TNF and non anti-TNF treated RA groups (p=0.82) (Figure 1B).
Figure 1. Type I interferon (IFN) plasma activity in patients with rheumatoid arthritis (RA) and healthy control donors (HD).
Plasma type I IFN activity was quantified using the WISH epithelial cell line assay as described in Patients and Methods. The dotted line shows the cut-off point between high and low levels, defined as the type I IFN level above the mean + 2SD of a pool of healthy controls tested in the same assay.
Association between baseline plasma type I IFN activity and clinical response among RA patients treated with TNF antagonists
To investigate whether type I IFN plasma activity levels prior to initiation of TNF-antagonist therapy might be associated with response to therapy, we compared baseline plasma activities in RA patients who were categorized by non-, moderate or good response at follow-up, based on the EULAR response criteria. As shown inFigure 2A, there was a graded relationship between type I IFN activity at baseline and response to TNF-antagonist therapy (mean ± SD: 0.4±1.9 versus 1.7±2.0 versus 3.4±4.9 for RA patients with non-, moderate, and good response, respectively; p=0.08). When the moderate and good responders were clustered together and compared to the non-response group, a statistically significant difference in baseline type I IFN levels emerged (p=0.04,Figure 2B). Regression analysis demonstrated that patients with higher baseline type I IFN scores were more likely to respond [OR 1.36 (95% CI 1.05– 1.76), p=0.02] per unit increase in type I IFN activity (Table 2). The association remained significant when the OR was adjusted for age (adjusted OR=1.35, CI: 1.04-1.77, p=0.027,Table 2). No statistically significant differences in baseline type I IFN activity between non-, moderate, and good responders were observed in the non-TNF-antagonist-treated group, although the number of subjects is likely too small to draw definite conclusions (data not shown).
Figure 2. Baseline plasma type I IFN activity in TNF-antagonist-treated RA patients as a function of clinical response.

A. Plasma type I IFN activity was determined at baseline (prior to initiation of therapy) in RA patients with non-response (n = 11), moderate response (n = 17), or good response (n = 7) after 5.6±1.4 months of TNF-antagonist therapy. Each symbol indicates an individual subject. B. Plasma type I IFN activity was compared between non responders and the group comprising moderate and good responders. C. Plasma type I IFN activity was compared between good responders and the group comprising non and moderate responders.
Table 2. Association of type I IFN and IL-1ra baseline levels with disease outcome according to the EULAR criteria by ordinal univariate and multivariate logistic regression analysis.
| OR (95% CI) | p-value | |
|---|---|---|
| Type I IFN (baseline) | ||
| Univariate analysis | 1.36 (1.05-1.76) | 0.020 |
| Multivariate analysis* | 1.35 (1.04 – 1.77) | 0.027 |
| IL-1ra (baseline) | ||
| Univariate analysis | 1.82 (1.1-3.29) | 0.027 |
| Multivariate analysis* | 1.79 (1.02 – 3.16) | 0.044 |
Age included in the multivariate model
Association of increased IFNβ/α ratio with favorable response to TNF-antagonist treatment
The assay for type I IFN activity does not distinguish between IFNα and IFNβ. Although type I IFN activity in SLE is predominantly mediated by IFNα (34), there may be a greater contribution from IFNβ in RA, given the documented expression of IFNβ in synovial membranes of RA patients and in animal models of inflammatory arthritis (24-29). Accordingly, the percent inhibition of plasma type I IFN activity after addition of neutralizing anti-IFNα or anti-IFNβ antibody to the plasma of those patients with elevated type I IFN activity was determined, and the results were converted to IFNβ/α ratios. In contrast to our results in SLE, plasma type I IFN activity from most RA patients was partially neutralized by anti-IFNβ as well as anti-IFNα antibodies (Figure 3A), demonstrating that IFNβ and IFNα each contribute to the type I IFN activity in RA plasma.
Figure 3. Association of increased plasma IFNβ/α ratio with response to TNF antagonists.
A. Antibody specific for IFNα or antibody specific for IFNβ was added to RA plasma prior to establishing cultures with WISH cells and IFN activity measured as described in the Patients and Methods section. B. Changes of DAS28 levels between baseline and follow-up in TNF-antagonist-treated patients are demonstrated according to the baseline IFNβ/α ratio (the median value, 0.8, of the distribution was used to differentiate two groups). Results are presented as inFigure 2.
Indeed, when RA patients with elevated baseline plasma type I IFN activity were subdivided into two groups according to their baseline IFNβ/α ratio (patients were categorized as having ratios greater than or less than 0.8, the median value of the distribution), those patients with higher IFNβ/α ratios (≥0.8) had greater changes in DAS28 scores at follow-up in comparison to those with lower IFNβ/α ratio (<0.8) [(2.8±1.2 vs 1.5±1.4, p=0.04);Figure 3B], suggesting that IFNβ might represent an important contributor to the increased control of inflammation and therapeutic responses among TNF-antagonist-treated RA patients. No other differences in demographic variables or treatment modalities were detected between the two groups (data not shown).
Association between baseline plasma IL-1ra levels and clinical response among RA patients treated with TNF antagonists
Since IL-1ra can be induced in PBMC and synoviocytes by IFNβ (18,19), we postulated that IL-1ra might represent an IFNβ-induced mediator that contributes to the superior therapeutic response to TNF-antagonist therapy. We observed significantly higher baseline IL-1ra levels in plasmas from good responders compared to non-/moderate responders (Figure 4). The magnitude of the association between higher IL-1ra levels and response to anti-TNF treatment was further determined by ordinal logistic regression (OR=1.82, CI: 1.1-3.29, p=0.027,Table 2). The association remained significant when OR was adjusted for age (adjusted OR=1.79, CI: 1.02-3.16, p=0.044,Table 2).
Figure 4. Plasma IL-1ra levels are increased in TNF-antagonist-treated RA patients with a good clinical response.
A. Baseline IL-1ra levels (pg/100ml) were determined at baseline (prior to initiation of therapy) in RA patients with non-response (n=11), moderate response (n = 17), or good response (n = 6). B. Baseline IL-1ra levels (pg/100ml) were compared between non responders and the group comprising moderate and good responders. C. Baseline IL-1ra levels (pg/100ml) were compared between good responders and the group comprising non and moderate responders. Results are presented as inFigure 2.
DISCUSSION
In recent years, TNF antagonists have revolutionized the treatment of RA and other inflammatory conditions. However, the responsiveness to treatment is highly variable among individuals. Of interest, this heterogeneity in responsiveness seems to be a stable trait over time. Repeated infusions add very limited benefit to primary non-responders, suggesting that stable factors, such as genetic constitution of the host, affect either the pharmacokinetics or the pharmacodynamics of the TNF antagonists and might account for the differential responses among individuals (35). Another possible contributor to the highly heterogeneous response is the diversity and complexity of pathogenic mechanisms (36).
Recent gene expression data from synovial tissues and PBMC from RA patients highlight the expression of type I IFN or IFN-induced genes in a subset of patients (16,17,27). While the factors that contribute to IFN pathway activation in the setting of RA have not been adequately elucidated, IFNβ is present in the rheumatoid synovium of some patients and is associated with control of inflammatory responses (27-29). Studies of several disease-associated genes common to SLE and RA, specifically IRF5, PTPN22 and STAT4, have shown increased plasma type I IFN activity or response in SLE patients with the risk alleles (37-42). Taken together, the association of RA-associated genes with type I IFN pathway activation, the expression of IFNβ and IFN-induced genes in some RA patients, and the functional data in animal models of inflammatory arthritis support type I IFN as a relevant mediator in RA for further study.
Acknowledging the high cost of treatment and the potentially serious adverse effects, investigators and clinicians recognize that identification of predictors of response to TNF antagonists would contribute to improved patient management. With the availability of additional new therapeutic options, patients predicted to be TNF-antagonist non-responders might be more successfully treated with other agents, leading to earlier (and, ultimately, less costly and toxic) therapeutic responses. Inasmuch as the IFN pathway is activated in some RA patients and bearing in mind a plausible protective role for IFNβ in RA, we hypothesized that type I IFN activity in RA could contribute to better inflammatory control and serve as a predictor of response to therapy.
In the present report, we document that RA patients collectively express increased plasma type I IFN activity relative to levels in healthy controls. While our RA patients were highly enriched in individuals of Hispanic ethnicity, the healthy control group was similarly composed. We have not defined any significant differences in plasma type I IFN activity among healthy individuals from different ethnic groups (data not shown). Importantly, higher levels of type I IFN activity at baseline are associated with better outcomes as defined by EULAR RA response criteria.
In contrast to SLE, in which IFNα appears to be the major contributor to plasma type I IFN activity (34), inhibition experiments using monoclonal anti-IFNα and anti-IFNβ antibodies revealed that both IFNα and IFNβ contribute to type I IFN activity in RA plasma. Moreover, a higher IFNβ/α ratio prior to initiation of TNF-antagonist therapy was found to be associated with a better clinical response, pointing to IFNβ, rather than IFNα, as a key contributor to control of inflammation and predictor for a better response to TNF-antagonist therapy.
Given that IL1-ra, an anti-inflammatory cytokine, can be induced by IFNβ, we measured IL-1ra levels in RA patient samples. A statistically significant association was detected between baseline IL-1ra levels and therapeutic outcome, pointing to elevated plasma IL-1ra level as an additional predictor of good response in TNF-antagonist-treated patients. Measurement of expression of additional gene targets induced by IFNβ might provide improved power to the predictive capacity of type I IFN and IL-1ra.
Categorization of the TNF-antagonist-treated patients into non-, moderate and good responders allowed analysis of other previously implicated parameters in relation to clinical response (7-9). However, neither presence of rheumatoid factor nor anti-CCP antibodies was associated with response to TNF antagonists in our patients. In addition, treatment with hydroxychloroquine, a putative inhibitor of induction of type I IFN through Toll-like receptor pathways (43), did not affect the predictive value of baseline type I IFN, which remained significant when hydroxychloroquine therapy was included in the statistical model [adjusted OR 1.34 (1.03 – 1.75, p=0.027)].
In conclusion, we have demonstrated that some RA patients express elevated levels of plasma type I IFN activity, which are associated with favorable responses to TNF-antagonist treatment. Our findings point to baseline elevated IFNβ activity, rather than IFNα activity, as the relevant factor associated with the superior clinical responses. Plasma IL-1ra levels, known to be induced by IFNβ, may serve as an additional predictor of response to TNF-antagonist therapy. We acknowledge that our patient cohort is relatively small and predominantly Hispanic. The statistically significant association of type I IFN activity with therapeutic response could be a function of statistical error and will have to be replicated in additional cohorts that include patients of other ethnicities. A prospective study of the parameters quantified in this study in relation to clinical response to therapy will be needed to validate our post-hoc analysis. The requirement for appropriate replication of our data and those of other investigators points to the urgent need for a coordinated approach to the generation of clinical datasets that include RA patients with well-defined clinical outcomes linked to biologic samples.
While the anti-proliferative immunomodulatory properties of IFNβ provided good rationale for efforts to test that cytokine as a potential therapy for RA, controlled clinical trials have failed to demonstrate efficacy (44,45). Nonetheless, increased production of IFNβ in some patients with RA, whether based on genetic diversity or other explanations for individual variability, might represent an anti-proliferative factor that complements the anti-inflammatory effects of TNF inhibition, resulting in improved efficacy. Our data provide the rationale for further studies to define a panel of factors, including type I IFN and IFNβ-induced gene products, that best identifies RA patient subgroups based on therapeutic response to TNF-antagonists and other agents, along with prospective studies of larger numbers and more racially and ethnically diverse RA patients.
Acknowledgments
Supported by:
A Stavros Niarchos Fellowship from the New York Chapter of the Arthritis Foundation to C.P.M., and grants from the NIH (AI059893), the Alliance for Lupus Research, the Lupus Research Institute, and the Mary Kirkland Center for Lupus Research to M.K.C.
REFERENCES
- 1.Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, Macfarlane JD, et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum. 1998;41(9):1552–63. doi: 10.1002/1529-0131(199809)41:9<1552::AID-ART5>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
- 2.Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld FC, Kalden JR, et al. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group Infliximab and methotrexate in the treatment of rheumatoid arthritis. N Engl J Med. 2000;343(22):1594–602. doi: 10.1056/NEJM200011303432202. [DOI] [PubMed] [Google Scholar]
- 3.Weinblatt ME, Kremer JM, Bankhurst AD, Bulpitt KJ, Fleischmann RM, Fox RI, et al. A trial of etanercept, a recombinant tumor necrosis factor receptor:Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate. N Engl J Med. 1999;340(4):253–9. doi: 10.1056/NEJM199901283400401. [DOI] [PubMed] [Google Scholar]
- 4.Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA, et al. Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum. 2003;48(1):35–45. doi: 10.1002/art.10697. [DOI] [PubMed] [Google Scholar]
- 5.Keystone EC, Kavanaugh AF, Sharp JT, Tannenbaum H, Hua Y, Teoh LS, et al. Radiographic, clinical, and functional outcomes of treatment with adalimumab (a human anti-tumor necrosis factor monoclonal antibody) in patients with active rheumatoid arthritis receiving concomitant methotrexate therapy: a randomized, placebo-controlled, 52-week trial. Arthritis Rheum. 2004;50(5):1400–11. doi: 10.1002/art.20217. [DOI] [PubMed] [Google Scholar]
- 6.Hyrich KL, Watson KD, Silman AJ, Symmons DP. Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Rheumatology (Oxford) 2006;45(12):1558–65. doi: 10.1093/rheumatology/kel149. [DOI] [PubMed] [Google Scholar]
- 7.Mancarella L, Bobbio-Pallavicini F, Ceccarelli F, Falappone PC, Ferrante A, Malesci D, et al. Good clinical response, remission, and predictors of remission in rheumatoid arthritis patients treated with tumor necrosis factor-alpha blockers: the GISEA study. J Rheumatol. 2007;34(8):1670–3. [PubMed] [Google Scholar]
- 8.Braun-Moscovici Y, Markovits D, Zinder O, Schapira D, Rozin A, Ehrenburg M, et al. Anti-cyclic citrullinated protein antibodies as a predictor of response to anti-tumor necrosis factor-alpha therapy in patients with rheumatoid arthritis. J Rheumatol. 2006;33(3):497–500. [PubMed] [Google Scholar]
- 9.Potter C, Hyrich KL, Tracey A, Lunt M, Plant D, Symmons DP, et al. Association of RF and anti-CCP positivity, but not carriage of shared epitope or PTPN22 susceptibility variants, with anti-TNF response in RA. Ann Rheum Dis. 2009;68(1):69–74. doi: 10.1136/ard.2007.084715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Rustenburg F, Baggen JM, Verweij CL, et al. Responsiveness to anti-tumor necrosis factor alpha therapy is related to pre-treatment tissue inflammation levels in rheumatoid arthritis patients. Ann Rheum Dis. 2008;67(4):563–6. doi: 10.1136/ard.2007.081950. [DOI] [PubMed] [Google Scholar]
- 11.Wijbrandts CA, Dijkgraaf MG, Kraan MC, Vinkenoog M, Smeets TJ, Dinant H, et al. The clinical response to infliximab in rheumatoid arthritis is in part dependent on pre-treatment tumor necrosis factor alpha expression in the synovium. Ann Rheum Dis. 2007;67(8):1139–44. doi: 10.1136/ard.2007.080440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lequerre T, Gauthier-Jauneau AC, Bansard C, Derambure C, Hiron M, Vittecoq O, et al. Gene profiling in white blood cells predicts infliximab responsiveness in rheumatoid arthritis. Arthritis Res Ther. 2006;8(4):R105. doi: 10.1186/ar1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Koczan D, Drynda S, Hecker M, Drynda A, Guthke R, Kekow J, et al. Molecular discrimination of responders and nonresponders to anti-TNF alpha therapy in rheumatoid arthritis by etanercept. Arthritis Res Ther. 2008;10(3):R50. doi: 10.1186/ar2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.La DT, Collins CE, Yang HT, Migone TS, Stohl W. B lymphocyte stimulator expression in patients with rheumatoid arthritis treated with tumour necrosis factor alpha antagonists: differential effects between good and poor clinical responders. Ann Rheum Dis. 2008;67(8):1132–8. doi: 10.1136/ard.2007.079954. [DOI] [PubMed] [Google Scholar]
- 15.Liu C, Batliwalla F, Li W, Lee A, Roubenoff R, Beckman E, et al. Genome-wide association scan identifies candidate polymorphisms associated with differential response to anti-TNF treatment in rheumatoid arthritis. Mol Med. 2008;14(910):575–81. doi: 10.2119/2008-00056.Liu. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Voskuyl AE, Rustenburg F, Baggen JM, et al. Rheumatoid arthritis subtypes identified by genomic profiling of peripheral blood cells: assignment of a type I interferon signature in a subpopulation of patients. Ann Rheum Dis. 2007;66(8):1008–14. doi: 10.1136/ard.2006.063412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.van der Pouw Kraan TC, van Baarsen LG, Wijbrandts CA, Voskuyl AE, Rustenburg F, Baggen JM, et al. Expression of a pathogen-response program in peripheral blood cells defines a subgroup of rheumatoid arthritis patients. Genes Immun. 2008;9(1):16–22. doi: 10.1038/sj.gene.6364438. [DOI] [PubMed] [Google Scholar]
- 18.Coclet-Ninin J, Dayer JM, Burger D. Interferon-beta not only inhibits interleukin-1beta and tumor necrosis factor-alpha but stimulates interleukin-1 receptor antagonist production in human peripheral blood mononuclear cells. Eur Cytokine Netw. 1997;8(4):345–9. [PubMed] [Google Scholar]
- 19.Palmer G, Mezin F, Juge-Aubry CE, Plater-Zyberk C, Gabay C, Guerne PA. Interferon beta stimulates interleukin 1 receptor antagonist production in human articular chondrocytes and synovial fibroblasts. Ann Rheum Dis. 2004;63(1):43–9. doi: 10.1136/ard.2002.005546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.van Holten J, Plater-Zyberk C, Tak PP. Interferon-beta for treatment of rheumatoid arthritis? Arthritis Res. 2002;4(6):346–52. doi: 10.1186/ar598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yong VW, Chabot S, Stuve O, Williams G. Interferon beta in the treatment of multiple sclerosis: mechanisms of action. Neurology. 1998;51(3):682–9. doi: 10.1212/wnl.51.3.682. [DOI] [PubMed] [Google Scholar]
- 22.Noronha A, Toscas A, Jensen MA. Interferon beta decreases T cell activation and interferon gamma production in multiple sclerosis. J Neuroimmunol. 1993;46(12):145–53. doi: 10.1016/0165-5728(93)90244-s. [DOI] [PubMed] [Google Scholar]
- 23.Calabresi PA, Pelfrey CM, Tranquill LR, Maloni H, McFarland HF. VLA-4 expression on peripheral blood lymphocytes is downregulated after treatment of multiple sclerosis with interferon beta. Neurology. 1997;49(4):1111–6. doi: 10.1212/wnl.49.4.1111. [DOI] [PubMed] [Google Scholar]
- 24.van Holten J, Reedquist K, Sattonet-Roche P, Smeets TJ, Plater-Zyberk C, Vervoordeldonk MJ, et al. Treatment with recombinant interferon-beta reduces inflammation and slows cartilage destruction in the collagen-induced arthritis model of rheumatoid arthritis. Arthritis Res Ther. 2004;6(3):R239–49. doi: 10.1186/ar1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Treschow AP, Teige I, Nandakumar KS, Holmdahl R, Issazadeh-Navikas S. Stromal cells and osteoclasts are responsible for exacerbated collagen-induced arthritis in interferon-beta-deficient mice. Arthritis Rheum. 2005;52(12):3739–48. doi: 10.1002/art.21496. [DOI] [PubMed] [Google Scholar]
- 26.Adriaansen J, Kuhlman RR, van Holten J, Kaynor C, Vervoordeldonk MJ, Tak PP. Intraarticular interferon-beta gene therapy ameliorates adjuvant arthritis in rats. Hum Gene Ther. 2006;17(10):985–96. doi: 10.1089/hum.2006.17.985. [DOI] [PubMed] [Google Scholar]
- 27.van Holten J, Smeets TJ, Blankert P, Tak PP. Expression of interferon beta in synovial tissue from patients with rheumatoid arthritis: comparison with patients with osteoarthritis and reactive arthritis. Ann Rheum Dis. 2005;64(12):1780–2. doi: 10.1136/ard.2005.040477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Smeets TJ, Dayer JM, Kraan MC, Versendaal J, Chicheportiche R, Breedveld FC, et al. The effects of interferon-beta treatment of synovial inflammation and expression of metalloproteinases in patients with rheumatoid arthritis. Arthritis Rheum. 2000;43(2):270–4. doi: 10.1002/1529-0131(200002)43:2<270::AID-ANR5>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
- 29.Tak PP. IFN-beta in rheumatoid arthritis. Front Biosci. 2004;9:3242–7. doi: 10.2741/1475. [DOI] [PubMed] [Google Scholar]
- 30.Van Gestel AM, Prevoo MLL, Van ‘T Hof MA, Van Rijswijk MH, Van de Putte LBA, Van Riel PLCM. Development and validation of the European League Against Rheumatism Response Criteria for rheumatoid arthritis. Arthritis Rheum. 1996;39(1):34–40. doi: 10.1002/art.1780390105. [DOI] [PubMed] [Google Scholar]
- 31.van Gestel AM, Haagsma CJ, van Riel PL. Validation of rheumatoid arthritis improvement criteria that include simplified joint counts. Arthritis Rheum. 1998;41(10):1845–50. doi: 10.1002/1529-0131(199810)41:10<1845::AID-ART17>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
- 32.Hua J, Kirou K, Lee C, Crow MK. Functional assay of type I interferon in systemic lupus erythematosus plasma and association with anti-RNA binding protein autoantibodies. Arthritis Rheum. 2006;54(6):1906–16. doi: 10.1002/art.21890. [DOI] [PubMed] [Google Scholar]
- 33.Mavragani CP, Niewold TB, Moutsopoulos NM, Pillemer SR, Wahl SM, Crow MK. Augmented interferon-alpha pathway activation in patients with Sjogren’s syndrome treated with etanercept. Arthritis Rheum. 2007;56(12):3995–4004. doi: 10.1002/art.23062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Niewold TB, Hua J, Lehman TJ, Harley JB, Crow MK. High serum IFN-alpha activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun. 2007;8(6):492–502. doi: 10.1038/sj.gene.6364408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mascheretti S, Schreiber S. The role of pharmacogenomics in the prediction of efficacy of anti-TNF therapy in patients with Crohn’s disease. Pharmacogenomics. 2004;5(5):479–86. doi: 10.1517/14622416.5.5.479. [DOI] [PubMed] [Google Scholar]
- 36.van der Pouw Kraan TC, van Gaalen FA, Kasperkovitz PV, Verbeet NL, Smeets TJ, Kraan MC, et al. Rheumatoid arthritis is a heterogeneous disease: evidence for differences in the activation of the STAT-1 pathway between rheumatoid tissues. Arthritis Rheum. 2003;48(8):2132–45. doi: 10.1002/art.11096. [DOI] [PubMed] [Google Scholar]
- 37.Sigurdsson S, Padyukov L, Kurreeman FA, Liljedahl U, Wiman AC, Alfredsson L, et al. Association of a haplotype in the promoter region of the interferon regulatory factor 5 gene with rheumatoid arthritis. Arthritis Rheum. 2007;56(7):2202–10. doi: 10.1002/art.22704. [DOI] [PubMed] [Google Scholar]
- 38.Begovich AB, Carlton VE, Honigberg LA, Schrodi SJ, Chokkalingam AP, Alexander HC, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet. 2004;75(2):330–7. doi: 10.1086/422827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens TW, et al. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med. 2007;357(10):977–86. doi: 10.1056/NEJMoa073003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Niewold TB, Kelly JA, Flesch MH, Espinoza LR, Harley JB, Crow MK. Association of the IRF5 risk haplotype with high serum interferon-alpha activity in systemic lupus erythematosus patients. Arthritis Rheum. 2008;58(8):2481–7. doi: 10.1002/art.23613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kariuki SN, Crow MK, Niewold TB. The PTPN22 C1858T polymorphism is associated with skewing of cytokine profiles toward high IFN-alpha activity and low tumor necrosis factor-alpha levels in patients with lupus. Arthritis Rheum. 2008;58(9):2818–23. doi: 10.1002/art.23728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kariuki SN, Kirou KA, MacDermott EJ, Barillas-Arias L, Crow MK, Niewold TB. Cutting edge: Autoimmune disease risk variant of STAT4 confers increased sensitivity to IFN-alpha in lupus patients in vivo. J Immunol. 2009;182(1):34–8. doi: 10.4049/jimmunol.182.1.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.van Holten J, Pavelka K, Vencovsky J, Stahl H, Rozman B, Genovese M, et al. A multicentre, randomised, double blind, placebo controlled phase II study of subcutaneous interferon beta-1a in the treatment of patients with active rheumatoid arthritis. Ann Rheum Dis. 2005;64(1):64–9. doi: 10.1136/ard.2003.020347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Genovese MC, Chakravarty EF, Krishnan E, Moreland LW. A randomized, controlled trial of interferon-beta-1a (Avonex(R)) in patients with rheumatoid arthritis: a pilot study [ISRCTN03626626] Arthritis Res Ther. 2004;6(1):R73–R77. doi: 10.1186/ar1026. [DOI] [PMC free article] [PubMed] [Google Scholar]


