Blood autoantibody and cytokine profiles predict response to anti-tumor necrosis factor therapy in rheumatoid arthritis

Wolfgang Hueber, Beren H Tomooka, Franak Batliwalla, Wentian Li, Paul A Monach, Robert J Tibshirani, Ronald F Van Vollenhoven, Jon Lampa, Kazuyoshi Saito, Yoshiya Tanaka, Mark C Genovese, Lars Klareskog, Peter K Gregersen, William H Robinson, Wolfgang Hueber, Beren H Tomooka, Franak Batliwalla, Wentian Li, Paul A Monach, Robert J Tibshirani, Ronald F Van Vollenhoven, Jon Lampa, Kazuyoshi Saito, Yoshiya Tanaka, Mark C Genovese, Lars Klareskog, Peter K Gregersen, William H Robinson

Abstract

Introduction: Anti-TNF therapies have revolutionized the treatment of rheumatoid arthritis (RA), a common systemic autoimmune disease involving destruction of the synovial joints. However, in the practice of rheumatology approximately one-third of patients demonstrate no clinical improvement in response to treatment with anti-TNF therapies, while another third demonstrate a partial response, and one-third an excellent and sustained response. Since no clinical or laboratory tests are available to predict response to anti-TNF therapies, great need exists for predictive biomarkers.

Methods: Here we present a multi-step proteomics approach using arthritis antigen arrays, a multiplex cytokine assay, and conventional ELISA, with the objective to identify a biomarker signature in three ethnically diverse cohorts of RA patients treated with the anti-TNF therapy etanercept.

Results: We identified a 24-biomarker signature that enabled prediction of a positive clinical response to etanercept in all three cohorts (positive predictive values 58 to 72%; negative predictive values 63 to 78%).

Conclusions: We identified a multi-parameter protein biomarker that enables pretreatment classification and prediction of etanercept responders, and tested this biomarker using three independent cohorts of RA patients. Although further validation in prospective and larger cohorts is needed, our observations demonstrate that multiplex characterization of autoantibodies and cytokines provides clinical utility for predicting response to the anti-TNF therapy etanercept in RA patients.

Figures

Figure 1
Figure 1
Workflow of experiments and types of analysis. Upper panel: in the discovery steps, synovial antigen microarrays and multiplex cytokine assays were employed to determine candidate molecules that are differentially expressed in pretreatment sera of etanercept responders (≥ ACR50) and nonresponders (

Figure 2

Rheumatoid arthritis antigen microarrays. Rheumatoid…

Figure 2

Rheumatoid arthritis antigen microarrays. Rheumatoid arthritis (RA) antigen microarrays were used for autoantibody…

Figure 2
Rheumatoid arthritis antigen microarrays. Rheumatoid arthritis (RA) antigen microarrays were used for autoantibody profiling of sera derived from patients with RA prior to initiation of etanercept therapy. (a), (b) Array results from two representative RA patients. Yellow features are false-colored features utilized for array orientation, while green features represent autoantibody reactivities. Selected autoantibody reactivities are highlighted in colored boxes. (c) Quantification of the highlighted features. ApoE, apolipoprotein E; COMP, cartilage oligomeric matrix protein.

Figure 3

Elevated pretreatment autoantibody profiles in…

Figure 3

Elevated pretreatment autoantibody profiles in etanercept responders compared with nonresponders in the ABCoN…

Figure 3
Elevated pretreatment autoantibody profiles in etanercept responders compared with nonresponders in the ABCoN cohort. Significance analysis of microarrays (SAM) and hierarchical clustering were applied to identify and display autoantibody profiles that differentiate etanercept responders from nonresponders; results from one of several representative experiments are presented. SAM was utilized to identify antigens with statistical differences in antibody reactivity between etanercept responders (≥ ACR50) and nonresponders (q < 4.3%). The SAM-identified variables and individual patients were then hierarchically clustered, and results presented in tree dendrograms that represent the relationships in reactivities between patients as well as between antigens. Red font, citrullinated antigens; black font, native antigens. Patients are listed across the top of the heatmap image, and the ACR response rate for each patient is indicated. Red bar, responder cluster; green bar, nonresponder cluster. Numbers of misclassified samples are shown for each cluster. Array fluorescence units are color coded and indicated in the bar in the right upper corner of the image. ACR, American College of Rheumatology response; CCP, cyclic citrullinated peptide.

Figure 4

Elevated pretreatment blood cytokines are…

Figure 4

Elevated pretreatment blood cytokines are associated with response to etanercept therapy. Logistic regression…

Figure 4
Elevated pretreatment blood cytokines are associated with response to etanercept therapy. Logistic regression analysis was applied to cytokine measurements in 93 samples derived from three cohorts of etanercept new-start rheumatoid arthritis patients. Green circles, samples from the Japanese cohort; red circles, samples from the ABCoN cohort; blue circles, samples from the Swedish cohort; grey circles, the best-fit logistic regression curve; x values, actual cytokine concentrations; y values, an artificial noise value was added to achieve better visual separation of the actual cytokine values. P values are shown for each cytokine. The grey bar links the actual responder or nonresponder label of a sample with the logistic regression result of the same sample (probability of being a responder). For better readability, only six cytokines are shown. MCP-1, monocyte chemoattractant protein-1.

Figure 5

Identification of a 24-antibody and…

Figure 5

Identification of a 24-antibody and cytokine biomarker that differentiates pretreatment etanercept responders from…

Figure 5
Identification of a 24-antibody and cytokine biomarker that differentiates pretreatment etanercept responders from nonresponders. Prediction analysis of microarrays (PAM) was applied to establish a rank list of the variables, by training PAM on all 93 samples. An overlay of error plots derived from PAM analysis of the 93 samples is displayed. First, PAM was trained on the multi-parameter biomarker; blue, training error graph. Second, internal cross-validation of the dataset was performed; red, overall error of the cross-validation. For better readability, error bars are shown for the cross-validation graph only. The number of markers is shown in ascending order from right to left across the top of the panel, and the selected PAM-derived threshold is indicated.

Figure 6

Prediction of responders and nonresponders.…

Figure 6

Prediction of responders and nonresponders. Calculations of classification errors for (a) the PAM…

Figure 6
Prediction of responders and nonresponders. Calculations of classification errors for (a) the PAM training/cross-validation step on all cohorts combined, and (c) the PAM prediction steps for the three cohorts individually (top panel, ABCoN cohort; middle panel, Swedish cohort; bottom panel, Japanese cohort). R, responder; NR, nonresponder; NPV, negative predictive value; PPV, positive predictive value. (b) Complete biomarker of 24 discriminators listed according to rank order, with associated scores for nonresponders and responders in the right and far-right columns, respectively. ApoE, apolipoprotein E; COMP, cartilage oligomeric matrix protein; FGF-2, fibroblast growth factor-2; GM-CSF, granulocyte- macrophage colony-stimulating factor; IP-10, IFNγ-inducible protein 10; MCP-1, monocyte chemoattractant protein-1.
Figure 2
Figure 2
Rheumatoid arthritis antigen microarrays. Rheumatoid arthritis (RA) antigen microarrays were used for autoantibody profiling of sera derived from patients with RA prior to initiation of etanercept therapy. (a), (b) Array results from two representative RA patients. Yellow features are false-colored features utilized for array orientation, while green features represent autoantibody reactivities. Selected autoantibody reactivities are highlighted in colored boxes. (c) Quantification of the highlighted features. ApoE, apolipoprotein E; COMP, cartilage oligomeric matrix protein.
Figure 3
Figure 3
Elevated pretreatment autoantibody profiles in etanercept responders compared with nonresponders in the ABCoN cohort. Significance analysis of microarrays (SAM) and hierarchical clustering were applied to identify and display autoantibody profiles that differentiate etanercept responders from nonresponders; results from one of several representative experiments are presented. SAM was utilized to identify antigens with statistical differences in antibody reactivity between etanercept responders (≥ ACR50) and nonresponders (q < 4.3%). The SAM-identified variables and individual patients were then hierarchically clustered, and results presented in tree dendrograms that represent the relationships in reactivities between patients as well as between antigens. Red font, citrullinated antigens; black font, native antigens. Patients are listed across the top of the heatmap image, and the ACR response rate for each patient is indicated. Red bar, responder cluster; green bar, nonresponder cluster. Numbers of misclassified samples are shown for each cluster. Array fluorescence units are color coded and indicated in the bar in the right upper corner of the image. ACR, American College of Rheumatology response; CCP, cyclic citrullinated peptide.
Figure 4
Figure 4
Elevated pretreatment blood cytokines are associated with response to etanercept therapy. Logistic regression analysis was applied to cytokine measurements in 93 samples derived from three cohorts of etanercept new-start rheumatoid arthritis patients. Green circles, samples from the Japanese cohort; red circles, samples from the ABCoN cohort; blue circles, samples from the Swedish cohort; grey circles, the best-fit logistic regression curve; x values, actual cytokine concentrations; y values, an artificial noise value was added to achieve better visual separation of the actual cytokine values. P values are shown for each cytokine. The grey bar links the actual responder or nonresponder label of a sample with the logistic regression result of the same sample (probability of being a responder). For better readability, only six cytokines are shown. MCP-1, monocyte chemoattractant protein-1.
Figure 5
Figure 5
Identification of a 24-antibody and cytokine biomarker that differentiates pretreatment etanercept responders from nonresponders. Prediction analysis of microarrays (PAM) was applied to establish a rank list of the variables, by training PAM on all 93 samples. An overlay of error plots derived from PAM analysis of the 93 samples is displayed. First, PAM was trained on the multi-parameter biomarker; blue, training error graph. Second, internal cross-validation of the dataset was performed; red, overall error of the cross-validation. For better readability, error bars are shown for the cross-validation graph only. The number of markers is shown in ascending order from right to left across the top of the panel, and the selected PAM-derived threshold is indicated.
Figure 6
Figure 6
Prediction of responders and nonresponders. Calculations of classification errors for (a) the PAM training/cross-validation step on all cohorts combined, and (c) the PAM prediction steps for the three cohorts individually (top panel, ABCoN cohort; middle panel, Swedish cohort; bottom panel, Japanese cohort). R, responder; NR, nonresponder; NPV, negative predictive value; PPV, positive predictive value. (b) Complete biomarker of 24 discriminators listed according to rank order, with associated scores for nonresponders and responders in the right and far-right columns, respectively. ApoE, apolipoprotein E; COMP, cartilage oligomeric matrix protein; FGF-2, fibroblast growth factor-2; GM-CSF, granulocyte- macrophage colony-stimulating factor; IP-10, IFNγ-inducible protein 10; MCP-1, monocyte chemoattractant protein-1.

References

    1. Feldmann M, Maini R. Role of cytokines in rheumatoid arthritis: an education in pathophysiology and therapeutics. Immunol Rev. 2008;223:7–19. doi: 10.1111/j.1600-065X.2008.00626.x.
    1. Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith C, Katz LM, Lightfoot R, Jr, Paulus H, Strand V, Tugwell P, Weinblatt M, Williams HJ, Wolfe W, Kieszak American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum. 1995;38:727–735. doi: 10.1002/art.1780380602.
    1. Maini R, St Clair E, Breedveld F, Furst D, Kalden J, Weisman M, Smolen J, Emery P, Harriman G, Feldmann M. Infliximab (chimeric anti-tumour necrosis factor α monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. Lancet. 1999;354:1932–1939. doi: 10.1016/S0140-6736(99)05246-0.
    1. Weinblatt M, Keystone E, Furst D, Moreland L, Weisman M, Birbara C, Teoh L, Fischkoff S, Chartash E. 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;48:35–45. doi: 10.1002/art.10697.
    1. Weinblatt ME, Kremer JM, Bankhurst AD, Bulpitt KJ, Fleischmann RM, Fox RI, Jackson CG, Lange M, Burge DJ. 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;340:253–259. doi: 10.1056/NEJM199901283400401.
    1. Felson D, Anderson J, Lange M, Wells G, LaValley M. Should improvement in rheumatoid arthritis clinical trials be defined as fifty percent or seventy percent improvement in core set measures, rather than twenty percent? Arthritis Rheum. 1998;41:1564–1570.
    1. Kievit W, Fransen J, Oerlemans AJ, Kuper HH, Laar MA van der, de Rooij DJ, De Gendt CM, Ronday KH, Jansen TL, van Oijen PC, Brus HL, Adang EM, van Riel PL. The efficacy of anti-TNF in rheumatoid arthritis, a comparison between randomised controlled trials and clinical practice. Ann Rheum Dis. 2007;66:1473–1478. doi: 10.1136/ard.2007.072447.
    1. Weaver AL, Lautzenheiser RL, Schiff MH, Gibofsky A, Perruquet JL, Luetkemeyer J, Paulus HE, Xia HA, Leff JA, Investigators TR. Real-world effectiveness of select biologic and DMARD monotherapy and combination therapy in the treatment of rheumatoid arthritis: results from the RADIUS observational registry. Curr Med Res Opin. 2006;22:185–198. doi: 10.1185/030079905X65510.
    1. Chen Y, Jobanputra P, Barton P, Jowett S, Bryan S, Clark W, Fry-Smith A, Burls A. A systematic review of the effectiveness of adalimumab, etanercept and infliximab for the treatment of rheumatoid arthritis in adults and an economic evaluation of their cost-effectiveness. Health Technol Assess. 2006;10:1–229.
    1. Zink A, Strangfeld A, Schneider M, Herzer P, F H, Stoyanova-Scholz M, Wassenberg S, Kapelle A, Listing J. Effectiveness of tumor necrosis factor inhibitors in rheumatoid arthritis in an observational cohort study: comparison of patients according to their eligibility for major randomized clinical trials. Arthritis Rheum. 2006;54:3399–3407. doi: 10.1002/art.22193.
    1. Ranganathan P. Pharmacogenomics of tumor necrosis factor antagonists in rheumatoid arthritis. Pharmacogenomics. 2005;6:481–490. doi: 10.2217/14622416.6.5.481.
    1. Hueber W, Tomooka BH, Zhao X, Kidd BA, Drijfhout JW, Fries JF, van Venrooij WJ, Metzger AL, Genovese MC, Robinson WH. Proteomic analysis of secreted proteins in early rheumatoid arthritis: anti-citrulline autoreactivity is associated with up regulation of proinflammatory cytokines. Ann Rheum Dis. 2007;66:712–719. doi: 10.1136/ard.2006.054924.
    1. Robinson WH, DiGennaro C, Hueber W, Haab BB, Kamachi M, Dean EJ, Fournel S, Fong D, Genovese MC, de Vegvar HE, Skriner K, Hirschberg DL, Morris RI, Muller S, Pruijn GJ, van Venrooij WJ, Smolen JS, Brown PO, Steinman L, Utz PJ. Autoantigen microarrays for multiplex characterization of autoantibody responses. Nat Med. 2002;8:295–301. doi: 10.1038/nm0302-295.
    1. Hueber W, Kidd BA, Tomooka BH, Lee BJ, Bruce B, Fries JF, Sonderstrup G, Monach P, Drijfhout JW, van Venrooij WJ, Utz PJ, Genovese MC, Robinson WH. Antigen microarray analysis defines subtypes of rheumatoid arthritis. Arthritis Rheum. 2005;52:2645–2655. doi: 10.1002/art.21269.
    1. Arnett F, Edworthy S, Bloch D, McShane D, Fries J, Cooper N, Healey L, Kaplan S, Liang M, Luthra H. 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. Eisen Lab
    1. Tibshirani R, Hastie T, Narasimhan B, Chu G. Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci USA. 2002;99:6567–6572. doi: 10.1073/pnas.082099299.
    1. PAM: Prediction Analysis for Microarrays
    1. The R Project for Statistical Computing
    1. Van Vollenhoven R, Klareskog L. Clinical responses to tumor necrosis factor alpha antagonists do not show a bimodal distribution: data from the Stockholm Tumor Necrosis Factor alpha Followup Registry. Arthritis Rheum. 2003;48:1500–1503. doi: 10.1002/art.11027.
    1. Buch M, Seto Y, Bingham S, Bejarano V, Bryer D, White J, Emery P. C-reactive protein as a predictor of infliximab treatment outcome in patients with rheumatoid arthritis: defining subtypes of nonresponse and subsequent response to etanercept. Arthritis Rheum. 2005;52:42–48. doi: 10.1002/art.20711.
    1. Liu C, Batliwalla F, Li W, Lee A, Roubenoff R, Beckman E, Khalili H, Damle A, Kern M, Plenge RM, Coenen MJ, Behrens TW, Carulli JP, Gregersen PK. Genome-wide association scan identifies candidate polymorphisms associated with differential response to anti-TNF treatment in rheumatoid arthritis. Mol Med. 2008;14:575–581.
    1. Tutuncu Z, Kavanaugh A, Zvaifler N, Corr M, Deutsch R, Boyle D. Fcγ receptor type IIIA polymorphisms influence treatment outcomes in patients with inflammatory arthritis treated with tumor necrosis factor alpha-blocking agents. Arthritis Rheum. 2005;52:2693–2696. doi: 10.1002/art.21266.
    1. Seitz M, Wirthmuller U, Moller B, Villiger PM. The -308 tumour necrosis factor-alpha gene polymorphism predicts therapeutic response to TNF alpha-blockers in rheumatoid arthritis and spondyloarthritis patients. Rheumatology. 2007;46:93–96. doi: 10.1093/rheumatology/kel175.
    1. Potter C, Hyrich KL, Tracey A, Lunt M, Plant D, Symmons DP, Thomson W, Worthington J, Emery P, Morgan AW, Wilson AG, Isaacs J, Barton A, BRAGGSS Association of rheumatoid factor and anti-cyclic citrullinated peptide positivity, but not carriage of shared epitope or PTPN22 susceptibility variants, with anti-tumour necrosis factor response in rheumatoid arthritis. Ann Rheum Dis. 2009;68:69–74. doi: 10.1136/ard.2007.084715.
    1. Rifai N, Gillette M, Carr S. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat Biotechnol. 2006;24:971–983. doi: 10.1038/nbt1235.
    1. O'Dell JR. Therapeutic strategies for rheumatoid arthritis. N Engl J Med. 2004;350:2591–2602. doi: 10.1056/NEJMra040226.
    1. Fabre S, D AM, Dossat N, Guisset C, Cohen J, Cristol J, Daures J, Jorgensen C. Protein biochip array technology for cytokine profiling predicts etanercept responsiveness in rheumatoid arthritis. Clin Exp Immunol. 2008
    1. Braun-Moscovici Y, Markovits D, Zinder O, Schapira D, Rozin A, Ehrenburg M, Dain L, Hoffer E, Nahir A, Balbir-Gurman A. 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;33:497–500.
    1. Vis M, Bos W, Wolbink G, Voskuyl A, Twisk J, Stadt R Van de, Hamann D, Dijkmans B, Lems W. IgM-rheumatoid factor, anti-cyclic citrullinated peptide, and anti-citrullinated human fibrinogen antibodies decrease during treatment with the tumor necrosis factor blocker infliximab in patients with rheumatoid arthritis. J Rheumatol. 2008;35:425–428.
    1. Smolen J, Aletaha D, Grisar J, Redlich K, Steiner G, Wagner O. The need for prognosticators in rheumatoid arthritis. Biological and clinical markers: where are we now? Arthritis Res Ther. 2008;10:208. doi: 10.1186/ar2418.

Source: PubMed

3
Předplatit