Synovial phenotypes in rheumatoid arthritis correlate with response to biologic therapeutics

Glynn Dennis Jr, Cécile T J Holweg, Sarah K Kummerfeld, David F Choy, A Francesca Setiadi, Jason A Hackney, Peter M Haverty, Houston Gilbert, Wei Yu Lin, Lauri Diehl, S Fischer, An Song, David Musselman, Micki Klearman, Cem Gabay, Arthur Kavanaugh, Judith Endres, David A Fox, Flavius Martin, Michael J Townsend, Glynn Dennis Jr, Cécile T J Holweg, Sarah K Kummerfeld, David F Choy, A Francesca Setiadi, Jason A Hackney, Peter M Haverty, Houston Gilbert, Wei Yu Lin, Lauri Diehl, S Fischer, An Song, David Musselman, Micki Klearman, Cem Gabay, Arthur Kavanaugh, Judith Endres, David A Fox, Flavius Martin, Michael J Townsend

Abstract

Introduction: Rheumatoid arthritis (RA) is a complex and clinically heterogeneous autoimmune disease. Currently, the relationship between pathogenic molecular drivers of disease in RA and therapeutic response is poorly understood.

Methods: We analyzed synovial tissue samples from two RA cohorts of 49 and 20 patients using a combination of global gene expression, histologic and cellular analyses, and analysis of gene expression data from two further publicly available RA cohorts. To identify candidate serum biomarkers that correspond to differential synovial biology and clinical response to targeted therapies, we performed pre-treatment biomarker analysis compared with therapeutic outcome at week 24 in serum samples from 198 patients from the ADACTA (ADalimumab ACTemrA) phase 4 trial of tocilizumab (anti-IL-6R) monotherapy versus adalimumab (anti-TNFα) monotherapy.

Results: We documented evidence for four major phenotypes of RA synovium - lymphoid, myeloid, low inflammatory, and fibroid - each with distinct underlying gene expression signatures. We observed that baseline synovial myeloid, but not lymphoid, gene signature expression was higher in patients with good compared with poor European league against rheumatism (EULAR) clinical response to anti-TNFα therapy at week 16 (P =0.011). We observed that high baseline serum soluble intercellular adhesion molecule 1 (sICAM1), associated with the myeloid phenotype, and high serum C-X-C motif chemokine 13 (CXCL13), associated with the lymphoid phenotype, had differential relationships with clinical response to anti-TNFα compared with anti-IL6R treatment. sICAM1-high/CXCL13-low patients showed the highest week 24 American College of Rheumatology (ACR) 50 response rate to anti-TNFα treatment as compared with sICAM1-low/CXCL13-high patients (42% versus 13%, respectively, P =0.05) while anti-IL-6R patients showed the opposite relationship with these biomarker subgroups (ACR50 20% versus 69%, P =0.004).

Conclusions: These data demonstrate that underlying molecular and cellular heterogeneity in RA impacts clinical outcome to therapies targeting different biological pathways, with patients with the myeloid phenotype exhibiting the most robust response to anti-TNFα. These data suggest a path to identify and validate serum biomarkers that predict response to targeted therapies in rheumatoid arthritis and possibly other autoimmune diseases.

Trial registration: ClinicalTrials.gov NCT01119859

Figures

Figure 1
Figure 1
Stratification of rheumatoid arthritis (RA) transcriptional heterogeneity into homogeneous molecular phenotypes.(A) Two-dimensional hierarchical clustering of approximately 7,000 probes (rows), representing quantile-normalized and scaled expression values of the top 40% most variable probe sets (variability assessed using SD), in 49 RA patients (columns) inferring five molecular subgroups of synovial tissues. Patient-sample ordering and dendrogram based on agglomerative hierarchical clustering (Ward method): resulting tree used to select patient subgroups; number of patient subgroups selected to maximize mean silhouette width and k-nearest neighbor distances (k = 5 considered optimal). z-score-based color intensity scale for each probe in each sample is shown. Patient samples clustering into five main branches are color-coded left to right (bottom of the heatmap): C1 = red (n = 8), C2 = purple (n = 14), C3 = gray (n = 16), C4 = green (n = 8), C5 = light blue (n = 3). (B) Heatmap depicting over-represented Database for Annotation, Visualization and Integrated Discovery biological process categories for genes upregulated in the four largest synovial clusters. Each column represents one cluster (C1 to C4), color-coordinated as in panel A. Each row corresponds to a biological process category. Heatmap colors reflect log10 (adjusted P-value) from modified Fisher exact test for categorical over-representation. Annotation for each cluster based on the key biological processes is indicated. BMP, bone morphogenetic protein; TGF, transforming growth factor; SMAD, Sma Mothers Against Decapentaplegic; NOD, nucleotide-binding oligomerization domain; JAK-STAT, Janus kinase-signal transducer and activator of transcription.
Figure 2
Figure 2
Rheumatoid arthritis (RA) molecular phenotypes reflect cellular and biological differences. (A) Immunohistochemical detection of T cells (CD3) and B cells (CD20) in synovial tissue sections. Columns correspond to representative sections for each of the RA molecular phenotypes designated by color-coordinated bars on top. Scales on images refer to a length of 500 microns. (B) Fluorescence activated cell-sorting analysis of fresh synovial tissue samples. Cells were stained with CD3- and CD20- gated by forward and side-scatter lymphocyte parameters and fluorescent intensities plotted in a scatter-plot with T cells (CD3) on the y-axis and B cells (CD20) on the x-axis (top panel). Contour-plots from the same patients above showing macrophages (CD45+, lymphocyte-gate exclusion) along the y-axis and fibroblasts (CD90) along the x-axis (bottom panel). Samples are arranged left to right according to their phenotype membership as in panel A. (C) Bar plots of the percentages of patient synovial tissues that contained non-aggregated (Agg-) or aggregated (Agg+) cellular infiltration as determined by immunohistological assessment of CD3- and CD20-positive cells.
Figure 3
Figure 3
Distribution of biological process genes and gene sets across the synovial tissue phenotypes. (A) Heatmap of expression of selected genes in lymphoid (red), myeloid (purple) and fibroid (green) patient subgroups. Patient-sample clusters are supervised by prior phenotype assignment, and genes are distributed by unsupervised clustering. (B-G) Distribution of biological processes for each synovial phenotype (L = lymphoid, M = myeloid, X = low inflammatory, F = fibroid) was assessed using predefined gene sets to interrogate the respective microarray datasets. Gene sets reflecting B cells (B), T cells (C), M1 classically activated monocytes (D), genes induced by TNFα (E), M2 alternatively activated monocytes (F) and angiogenesis (G). Each subgroup was compared to all other groups using the f-test, and significant Benjamini-Hochberg-corrected P-values for a group compared with all other groups are indicated (*P ≤0.05, **P ≤0.01, ***P ≤0.001) for subgroups with positive t-statistic values.
Figure 4
Figure 4
Pretreatment magnitude of gene sets derived from the synovial myeloid phenotype and classically activated monocytes correlates with clinical response to anti-TNFα (infliximab) therapy. Analysis of synovial tissue microarray data from 62 rheumatoid arthritis patients in GSE21537 prior to initiation of infliximab (anti-TNFα therapy). Scores for gene sets for phenotypes, defined from the Michigan cohort training data, as well as gene sets derived from purified immune cell lineages (see Methods), were calculated from the GSE21537 data and compared against anti-TNFα clinical outcome at 16 weeks as defined by European League Against Rheumatism (EULAR) response criteria as assigned in GSE21537. Scores versus EULAR response are plotted for the synovial myeloid phenotype (A), lymphoid phenotype (B), fibroid phenotype (C), as well as classically activated M1 monocytes (D), B cells (E) and T cells (F). Statistical significance for good compared with poor EULAR response for the level of each gene-set module was calculated based upon the t-statistic (* = P ≤0.05, **P ≤0.01).
Figure 5
Figure 5
Assessment of serum biomarkers extrapolated from lymphoid and myeloid synovial phenotype gene expression in the synovial transcriptome training dataset. Intercellular adhesion molecule 1 (ICAM1) (A) and C-X-C motif chemokine 13 (CXCL13) (B) genes are expressed at highest levels in the myeloid (M) and lymphoid (L) phenotypes respectively. Array probes for each transcript were compared across all groups using the f-test, and in both cases Benjamini-Hochberg-corrected, *P < 0.001. X = low inflammatory phenotype and F = fibroid phenotype. Soluble (s)ICAM1 (C) and CXCL13 (D) are elevated in serum samples from rheumatoid arthritis (RA) patients (ADACTA trial) as compared with normal control (NC) serum. P-values derived from the Wilcoxon test are indicated. (E) Serum sICAM1 and CXCL13 levels were only weakly correlated in RA (ρ < 0.33, Spearman rank correlation coefficient).
Figure 6
Figure 6
Lymphoid (C-X-C motif chemokine 13 (CXCL13)) and myeloid (soluble intercellular adhesion molecule 1 (sICAM1)) serum biomarkers define rheumatoid arthritis patient subgroups with differential clinical response to anti-TNFα (adalimumab) compared with anti-IL-6R (tocilizumab) in the ADACTA trial. Relative treatment effectiveness (week-24 American College of Rheumatology (ACR)50 response) of adalimumab compared with tocilizumab was assessed by logistic regression for (A) each individual biomarker and (B) biomarker combination-defined subgroups using their respective medians as cutoffs (see Methods). Relative treatment effectiveness for adalimumab versus tocilizumab is represented by odds ratio and 95% CI for ACR50 response. Week-24 ACR20 (gray), ACR50 (green), and ACR70 (purple) response rates (%) per biomarker-defined subgroup are represented by radial plot for adalimumab (C) and tocilizumab (D) treatment arms. The direction of each radial line corresponds to a biomarker subgroup as follows: sICAM1 low (bottom) and high (top), CXCL13 low (left) and high (right). Low and high designations refer to biomarker values above and below their respective medians. Distance from radial plot center indicates response rate. Summary of week-24 ACR50 response rates for sICAM1-high/CXCL13-low, sICAM1-high/CXCL13-high, sICAM1-low/CXCL13-low and sICAM1-low/CXCL13-high ADACTA RA patients (E). The treatment-effect deltas between sICAM1-high/CXCL13-low and sICAM1-low/CXCL13-high patient groups are indicated for both adalimumab and tocilizumab.

References

    1. Goronzy JJ, Weyand CM. Rheumatoid arthritis. Immunol Rev. 2005;204:55–73. doi: 10.1111/j.0105-2896.2005.00245.x.
    1. Lee DM, Weinblatt ME. Rheumatoid arthritis. Lancet. 2001;358:903–911. doi: 10.1016/S0140-6736(01)06075-5.
    1. Tak PP, Bresnihan B. The pathogenesis and prevention of joint damage in rheumatoid arthritis: advances from synovial biopsy and tissue analysis. Arthritis Rheum. 2000;43:2619–2633. doi: 10.1002/1529-0131(200012)43:12<2619::AID-ANR1>;2-V.
    1. Lindstrom TM, Robinson WH. Biomarkers for rheumatoid arthritis: making it personal. Scand J Clin Lab Invest Suppl. 2010;242:79–84.
    1. Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet. 2010;376:1094–1108. doi: 10.1016/S0140-6736(10)60826-4.
    1. Weyand CM, Goronzy JJ. Ectopic germinal center formation in rheumatoid synovitis. Ann NY Acad Sci. 2003;987:140–149. doi: 10.1111/j.1749-6632.2003.tb06042.x.
    1. Chan AC, Behrens TW. Personalizing medicine for autoimmune and inflammatory diseases. Nat Immunol. 2013;14:106–109. doi: 10.1038/ni.2473.
    1. van der Pouw Kraan TC, van Gaalen FA, Huizinga TW, Pieterman E, Breedveld FC, Verweij CL. Discovery of distinctive gene expression profiles in rheumatoid synovium using cDNA microarray technology: evidence for the existence of multiple pathways of tissue destruction and repair. Genes Immun. 2003;4:187–196. doi: 10.1038/sj.gene.6363975.
    1. van der Pouw Kraan TC, van Gaalen FA, Kasperkovitz PV, Verbeet NL, Smeets TJ, Kraan MC, Fero M, Tak PP, Huizinga TW, Pieterman E, Breedveld FC, Alizadeh AA, Verweij CL. Rheumatoid arthritis is a heterogeneous disease: evidence for differences in the activation of the STAT-1 pathway between rheumatoid tissues. Arthritis Rheum. 2003;48:2132–2145. doi: 10.1002/art.11096.
    1. van Baarsen LG, Bos CL, van der Pouw Kraan TC, Verweij CL. Transcription profiling of rheumatic diseases. Arthritis Res Ther. 2009;11:207. doi: 10.1186/ar2557.
    1. Timmer TC, Baltus B, Vondenhoff M, Huizinga TW, Tak PP, Verweij CL, Mebius RE, van der Pouw Kraan TC. Inflammation and ectopic lymphoid structures in rheumatoid arthritis synovial tissues dissected by genomics technology: identification of the interleukin-7 signaling pathway in tissues with lymphoid neogenesis. Arthritis Rheum. 2007;56:2492–2502. doi: 10.1002/art.22748.
    1. van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Rustenburg F, Baggen JM, Verweij CL, Tak PP. Responsiveness to anti-tumour necrosis factor alpha therapy is related to pre-treatment tissue inflammation levels in rheumatoid arthritis patients. Ann Rheum Dis. 2008;67:563–566.
    1. Wijbrandts CA, Dijkgraaf MG, Kraan MC, Vinkenoog M, Smeets TJ, Dinant H, Vos K, Lems WF, Wolbink GJ, Sijpkens D, Dijkmans BA, Tak PP. The clinical response to infliximab in rheumatoid arthritis is in part dependent on pretreatment tumour necrosis factor alpha expression in the synovium. Ann Rheum Dis. 2008;67:1139–1144. doi: 10.1136/ard.2007.080440.
    1. Badot V, Galant C, Nzeusseu Toukap A, Theate I, Maudoux AL, Van den Eynde BJ, Durez P, Houssiau FA, Lauwerys BR. Gene expression profiling in the synovium identifies a predictive signature of absence of response to adalimumab therapy in rheumatoid arthritis. Arthritis Res Ther. 2009;11:R57. doi: 10.1186/ar2678.
    1. Lindberg J, Wijbrandts CA, van Baarsen LG, Nader G, Klareskog L, Catrina A, Thurlings R, Vervoordeldonk M, Lundeberg J, Tak PP. The gene expression profile in the synovium as a predictor of the clinical response to infliximab treatment in rheumatoid arthritis. PLoS One. 2010;5:e11310. doi: 10.1371/journal.pone.0011310.
    1. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH, Luthra HS, Medsger TA Jr, Mitchell DM, Neustadt DH, Pinals RS, Schaller JG, Sharp JT, Wilder RL, Hunder GG. The American rheumatism association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–324. doi: 10.1002/art.1780310302.
    1. Barrett T, Troup DB, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Muertter RN, Holko M, Ayanbule O, Yefanov A, Soboleva A. NCBI GEO: archive for functional genomics data sets–10 years on. Nucleic Acids Res. 2011;39:D1005–D1010. doi: 10.1093/nar/gkq1184.
    1. Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–210. doi: 10.1093/nar/30.1.207.
    1. R Development Core Team. R Foundation for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2011. R: a language and environment for statistical computing. . ISBN 3-900051-07-0.
    1. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, Hornik K, Hothorn T, Huber W, Iacus S, Irizarry R, Leisch F, Li C, Maechler M, Rossini AJ, Sawitzki G, Smith C, Smyth G, Tierney L, Yang JY, Zhang J. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:R80. doi: 10.1186/gb-2004-5-10-r80.
    1. Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19:185–193. doi: 10.1093/bioinformatics/19.2.185.
    1. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–264. doi: 10.1093/biostatistics/4.2.249.
    1. Hackstadt AJ, Hess AM. Filtering for increased power for microarray data analysis. BMC Bioinformatics. 2009;10:11. doi: 10.1186/1471-2105-10-11.
    1. Bourgon R, Gentleman R, Huber W. Independent filtering increases detection power for high-throughput experiments. Proc Natl Acad Sci USA. 2010;107:9546–9551. doi: 10.1073/pnas.0914005107.
    1. Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA. DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 2003;4:3. doi: 10.1186/gb-2003-4-5-p3.
    1. Oron AP, Jiang Z, Gentleman R. Gene set enrichment analysis using linear models and diagnostics. Bioinformatics. 2008;24:2586–2591. doi: 10.1093/bioinformatics/btn465.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Abbas AR, Baldwin D, Ma Y, Ouyang W, Gurney A, Martin F, Fong S, van Lookeren CM, Godowski P, Williams PM, Chan AC, Clark HF. Immune response in silico (IRIS): immune-specific genes identified from a compendium of microarray expression data. Genes Immun. 2005;6:319–331. doi: 10.1038/sj.gene.6364173.
    1. Hochberg Y, Benjamini Y. More powerful procedures for multiple significance testing. Stat Med. 1990;9:811–818. doi: 10.1002/sim.4780090710.
    1. Gabay C, Emery P, van Vollenhoven R, Dikranian A, Alten R, Pavelka K, Klearman M, Musselman D, Agarwal S, Green J, Kavanaugh A. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. Lancet. 2013;381:1541–1550. doi: 10.1016/S0140-6736(13)60250-0.
    1. Lazar AA, Cole BF, Bonetti M, Gelber RD. Evaluation of treatment-effect heterogeneity using biomarkers measured on a continuous scale: subpopulation treatment effect pattern plot. J. Clin Oncol. 2010;28:4539–4544. doi: 10.1200/JCO.2009.27.9182.
    1. Kishimoto T. Interleukin-6: from basic science to medicine–40 years in immunology. Annu Rev Immunol. 2005;23:1–21. doi: 10.1146/annurev.immunol.23.021704.115806.
    1. Weyand CM, Kang YM, Kurtin PJ, Goronzy JJ. The power of the third dimension: tissue architecture and autoimmunity in rheumatoid arthritis. Curr Opin Rheumatol. 2003;15:259–266. doi: 10.1097/00002281-200305000-00013.
    1. van Baarsen LG, Wijbrandts CA, Timmer TC, van der Pouw Kraan TC, Tak PP, Verweij CL. Synovial tissue heterogeneity in rheumatoid arthritis in relation to disease activity and biomarkers in peripheral blood. Arthritis Rheum. 2010;62:1602–1607.
    1. van Oosterhout M, Bajema I, Levarht EW, Toes RE, Huizinga TW, van Laar JM. Differences in synovial tissue infiltrates between anti-cyclic citrullinated peptide-positive rheumatoid arthritis and anti-cyclic citrullinated peptide-negative rheumatoid arthritis. Arthritis Rheum. 2008;58:53–60. doi: 10.1002/art.23148.
    1. Hogan VE, Holweg CT, Choy DF, Kummerfeld SK, Hackney JA, Teng YK, Townsend MJ, van Laar JM. Pretreatment synovial transcriptional profile is associated with early and late clinical response in rheumatoid arthritis patients treated with rituximab. Ann Rheum Dis. 1888–1894;2012:71.
    1. Hueber W, Tomooka BH, Batliwalla F, Li W, Monach PA, Tibshirani RJ, Van Vollenhoven RF, Lampa J, Saito K, Tanaka Y, Genovese MC, Klareskog L, Gregersen PK, Robinson WH. Blood autoantibody and cytokine profiles predict response to anti-tumor necrosis factor therapy in rheumatoid arthritis. Arthritis Res Ther. 2009;11:R76. doi: 10.1186/ar2706.
    1. Lal P, Su Z, Holweg CT, Silverman GJ, Schwartzman S, Kelman A, Read S, Spaniolo G, Monroe JG, Behrens TW, Townsend MJ. Inflammation and autoantibody markers identify rheumatoid arthritis patients with enhanced clinical benefit following rituximab treatment. Arthritis Rheum. 2011;63:3681–3691. doi: 10.1002/art.30596.
    1. Klaasen R, Thurlings RM, Wijbrandts CA, van Kuijk AW, Baeten D, Gerlag DM, Tak PP. The relationship between synovial lymphocyte aggregates and the clinical response to infliximab in rheumatoid arthritis: a prospective study. Arthritis Rheum. 2009;60:3217–3224. doi: 10.1002/art.24913.
    1. Canete JD, Celis R, Moll C, Izquierdo E, Marsal S, Sanmarti R, Palacin A, Lora D, de la Cruz J, Pablos JL. Clinical significance of synovial lymphoid neogenesis and its reversal after anti-tumour necrosis factor alpha therapy in rheumatoid arthritis. Ann Rheum Dis. 2009;68:751–756. doi: 10.1136/ard.2008.089284.
    1. Krenn V, Schedel J, Doring A, Huppertz HI, Gohlke F, Tony HP, Vollmers HP, Muller-Hermelink HK. Endothelial cells are the major source of sICAM-1 in rheumatoid synovial tissue. Rheumatol Int. 1997;17:17–27. doi: 10.1007/PL00006846.
    1. Witkowska AM, Borawska MH. Soluble intercellular adhesion molecule-1 (sICAM-1): an overview. Eur Cytokine Netw. 2004;15:91–98.
    1. Corsiero E, Bombardieri M, Manzo A, Bugatti S, Uguccioni M, Pitzalis C. Role of lymphoid chemokines in the development of functional ectopic lymphoid structures in rheumatic autoimmune diseases. Immunol Lett. 2012;145:62–67. doi: 10.1016/j.imlet.2012.04.013.
    1. Rosengren S, Wei N, Kalunian KC, Kavanaugh A, Boyle DL. CXCL13: a novel biomarker of B-cell return following rituximab treatment and synovitis in patients with rheumatoid arthritis. Rheumatol (Oxford) 2011;50:603–610. doi: 10.1093/rheumatology/keq337.
    1. Meeuwisse CM, van der Linden MP, Rullmann TA, Allaart CF, Nelissen R, Huizinga TW, Garritsen A, Toes RE, van Schaik R, van der Helm-van Mil AH. Identification of CXCL13 as a marker for rheumatoid arthritis outcome using an in silico model of the rheumatic joint. Arthritis Rheum. 2011;63:1265–1273. doi: 10.1002/art.30273.
    1. Choy E. Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatol (Oxford) 2012;51:v3–v11. doi: 10.1093/rheumatology/kes113.
    1. Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science. 2002;296:1634–1635. doi: 10.1126/science.1071924.
    1. Emery P, Keystone E, Tony HP, Cantagrel A, van Vollenhoven R, Sanchez A, Alecock E, Lee J, Kremer J. 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;67:1516–1523. doi: 10.1136/ard.2008.092932.

Source: PubMed

3
Subscribe