Pre-treatment whole blood gene expression is associated with 14-week response assessed by dynamic contrast enhanced magnetic resonance imaging in infliximab-treated rheumatoid arthritis patients

Kenzie D MacIsaac, Richard Baumgartner, Jia Kang, Andrey Loboda, Charles Peterfy, Julie DiCarlo, Jonathan Riek, Chan Beals, Kenzie D MacIsaac, Richard Baumgartner, Jia Kang, Andrey Loboda, Charles Peterfy, Julie DiCarlo, Jonathan Riek, Chan Beals

Abstract

Approximately 30% of rheumatoid arthritis patients achieve inadequate response to anti-TNF biologics. Attempts to identify molecular biomarkers predicting response have met with mixed success. This may be attributable, in part, to the variable and subjective disease assessment endpoints with large placebo effects typically used to classify patient response. Sixty-one patients with active RA despite methotrexate treatment, and with MRI-documented synovitis, were randomized to receive infliximab or placebo. Blood was collected at baseline and genome-wide transcription in whole blood was measured using microarrays. The primary endpoint in this study was determined by measuring the transfer rate constant (Ktrans) of a gadolinium-based contrast agent from plasma to synovium using MRI. Secondary endpoints included repeated clinical assessments with DAS28(CRP), and assessments of osteitis and synovitis by the RAMRIS method. Infliximab showed greater decrease from baseline in DCE-MRI Ktrans of wrist and MCP at all visits compared with placebo (P<0.001). Statistical analysis was performed to identify genes associated with treatment-specific 14-week change in Ktrans. The 256 genes identified were used to derive a gene signature score by averaging their log expression within each patient. The resulting score correlated with improvement of Ktrans in infliximab-treated patients and with deterioration of Ktrans in placebo-treated subjects. Poor responders showed high expression of activated B-cell genes whereas good responders exhibited a gene expression pattern consistent with mobilization of neutrophils and monocytes and high levels of reticulated platelets. This gene signature was significantly associated with clinical response in two previously published whole blood gene expression studies using anti-TNF therapies. These data provide support for the hypothesis that anti-TNF inadequate responders comprise a distinct molecular subtype of RA characterized by differences in pre-treatment blood mRNA expression. They also highlight the importance of placebo controls and robust, objective endpoints in biomarker discovery.

Trial registration: ClinicalTrials.gov NCT01313520.

Conflict of interest statement

Competing Interests: The authors of this manuscript have the following competing interests: K. MacIsaac, J. Kang, A. Loboda, C. Beals, R. Baumgartner, are currently (or were at the time the study was conducted) employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Whitehouse Station, NJ, who may own stock and/or hold stock options in Merck. C. Peterfy and J. DiCarlo are employees of Spire Sciences LLC, employed by the study sponsor Merck to analyze study results; J. Riek is an employee of VirtualScopics, Inc, employed by the study sponsor Merck to analyze study results. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Gene expression is associated with…
Figure 1. Gene expression is associated with change in disease activity measured by DCE-MRI.
(A) High signature score correlates with Ktrans improvement in the treatment arm and Ktrans deterioration in the placebo arm. Scatter plots show baseline and treatment adjusted 14-week change in log Ktrans vs. signature score in both the treatment and placebo arms at weeks 2, 4, and 14. Linear models including terms for baseline Ktrans, treatment allocation, signature score, and the interaction between signature and treatment were fit to log Ktrans change from baseline at each week. At both week 4 and 14, the signature score main effect and interaction with treatment were significant at p<0.05. (B) Whole blood gene expression improves prediction of week 14 change in Ktrans. In ten repeated rounds of random subsampling, 40 patients were selected and their whole blood gene expression data was used to identify genes associated with treatment response measured by Ktrans, DAS28(CRP), and RAMRIS. A linear model including terms for baseline disease activity, treatment allocation, signature score, and the interaction between signature and treatment was fit to week 14 data and used to predict week 14 changes for held out subjects. The distribution of mean squared prediction errors (MSE) minus the MSE achieved by a model excluding signature score terms is plotted for each endpoint. For Ktrans, but not DAS28(CRP), or RAMRIS, incorporation of baseline blood gene expression consistently improved prediction performance (p = 0.015, t-test).
Figure 2. Gene signature score is higher…
Figure 2. Gene signature score is higher in EULAR responders than non-responders in two independent studies.
The predictive signature is associated with clinical response in two additional whole blood gene expression studies. The distribution of predictive signature scores was compared in responder and non-responder groups in the studies of Julia et al. and Toonen et al. In both cases, a one-tailed t-test identified statistically significant (p

References

    1. Hyrich KL, Watson KD, Silman AJ, Symmons DPM (2006) Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Rheumatology (Oxford) 45:1558–1565.
    1. Bienkowska JR, Dalgin GS, Batliwalla F, Allaire N, Roubenoff R, et al. (2009) Convergent Random Forest predictor: methodology for predicting drug response from genome-scale data applied to anti-TNF response. Genomics 94:423–432.
    1. Julià A, Erra A, Palacio C, Tomas C, Sans X, et al. (2009) An eight-gene blood expression profile predicts the response to infliximab in rheumatoid arthritis. PLoS One 4.
    1. Koczan D, Drynda S, Hecker M, Drynda A, Guthke R, et al. (2008) Molecular discrimination of responders and nonresponders to anti-TNF alpha therapy in rheumatoid arthritis by etanercept. Arthritis Res Ther 10.
    1. Lequerre T, Gauthier-Jauneau A-C, Bansard C, Derambure C, Hiron M, et al. (2006) Gene profiling in white blood cells predicts infliximab responsiveness in rheumatoid arthritis. Arthritis Res Ther 8.
    1. Sekiguchi N, Kawauchi S, Furuya T, Inaba N, Matsuda K, et al. (2008) Messenger ribonucleic acid expression profile in peripheral blood cells from RA patients following treatment with an anti-TNF-alpha monoclonal antibody, infliximab. Rheumatology (Oxford) 47:780–788.
    1. Stuhlmuller B, Haupl T, Hernandez MM, Grutzkau A, Kuban RJ, et al. (2010) CD11c as a transcriptional biomarker to predict response to anti-TNF monotherapy with adalimumab in patients with rheumatoid arthritis. Clin Pharmacol Ther 87:311–321.
    1. Tanino M, Matoba R, Nakamura S, Kameda H, Amano K, et al. (2009) Prediction of efficacy of anti-TNF biologic agent, infliximab, for rheumatoid arthritis patients using a comprehensive transcriptome analysis of white blood cells. Biochem Biophys Res Commun 387:261–265.
    1. van Baarsen LG, Wijbrandts CA, Rustenburg F, Cantaert T, van der Pouw Kraan TC, et al. (2010) Regulation of IFN response gene activity during infliximab treatment in rheumatoid arthritis is associated with clinical response to treatment. Arthritis Res Ther 12.
    1. Toonen EJM, Gilissen C, Franke B, Kievit W, Eijsbouts AM, et al. (2012) Validation study of existing gene expression signatures for anti-TNF treatment in patients with rheumatoid arthritis. PLoS One 7.
    1. Strand V, Cohen S, Crawford B, Smolen JS, Scott DL (2004) Patient-reported outcomes better discriminate active treatment from placebo in randomized controlled trials in rheumatoid arthritis. Rheumatology (Oxford) 43:640–647.
    1. Ostergaard M, Stoltenberg M, Lovgreen-Nielsen P, Volck B, Sonne-Holm S, et al. (1998) Quantification of synovistis by MRI: correlation between dynamic and static gadolinium-enhanced magnetic resonance imaging and microscopic and macroscopic signs of synovial inflammation. Magn Reson Imaging 16:743–754.
    1. Workie DW, Dardzinski BJ (2005) Quantifying dynamic contrast-enhanced MRI of the knee in children with juvenile rheumatoid arthritis using an arterial input function (AIF) extracted from popliteal artery enhancement, and the effect of the choice of the AIF on the kinetic parameters. Magn Reson Med 54:560–568.
    1. Hodgson RJ, O'Connor P, Moots R (2008) MRI of rheumatoid arthritis image quantitation for the assessment of disease activity, progression and response to therapy. Rheumatology (Oxford) 47:13–21.
    1. Prevoo ML, van 't Hof MA, Kuper HH, van Leeuwen MA, van de Putte LB, et al. (1995) Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum 38:44–48.
    1. McQueen F, Lassere M, Edmonds J, Conaghan P, Peterfy C, et al. (2003) OMERACT Rheumatoid Arthritis Magnetic Resonance Imaging Studies. Summary of OMERACT 6 MR Imaging Module. J Rheumatol 30:1387–1392.
    1. Liang KY ZS (2000) Longitudinal data analysis ofcontinuous and discrete responses for pre-post designs. Indian Journal of Statistics 62:134–148.
    1. Fransen J, van Riel PLCM (2005) The Disease Activity Score and the EULAR response criteria. Clin Exp Rheumatol 23:93–99.
    1. Okada Y, Wu D, Trynka G, Raj T, Terao C, et al. (2014) Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506:376–381.
    1. Van Steendam K, Tilleman K, Deforce D (2011) The relevance of citrullinated vimentin in the production of antibodies against citrullinated proteins and the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford) 50:830–837.
    1. Parker A, Izmailova ES, Narang J, Badola S, Le T, et al. (2007) Peripheral blood expression of nuclear factor-kappab-regulated genes is associated with rheumatoid arthritis disease activity and responds differentially to anti-tumor necrosis factor-alpha versus methotrexate. J Rheumatol 34:1817–1822.
    1. Villalvilla A, Gomez R, Roman-Blas JA, Largo R, Herrero-Beaumont G (2014) SDF-1 signaling: a promising target in rheumatic diseases. Expert Opin Ther Targets 18:1077–1087.
    1. Trocme C, Marotte H, Baillet A, Pallot-Prades B, Garin J, et al. (2009) Apolipoprotein A-I and platelet factor 4 are biomarkers for infliximab response in rheumatoid arthritis. Ann Rheum Dis 68:1328–1333.
    1. Chaussabel D, Quinn C, Shen J, Patel P, Glaser C, et al. (2008) A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity 29:150–164.
    1. Prajapati R, Plant D, Barton A (2011) Genetic and genomic predictors of anti-TNF response. Pharmacogenomics 12:1571–1585.
    1. Greenwood MC, Rathi J, Hakim AJ, Scott DL, Doyle DV (2007) Regression to the mean using the disease activity score in eligibility and response criteria for prescribing TNF-alpha inhibitors in adults with rheumatoid arthritis. Rheumatology (Oxford) 46:1165–1167.
    1. American College of Rheumatology Rheumatoid Arthritis Clinical Trial Investigators Ad Hoc Task F (2011) American College of Rheumatology Clinical Trial Priorities and Design Conference, July 22-23, 2010. Arthritis Rheum 63:2151–2156.
    1. Peterfy C, Ostergaard M, Conaghan PG (2013) MRI comes of age in RA clinical trials. Ann Rheum Dis 72:794–796.
    1. American College of Rheumatology Rheumatoid Arthritis Clinical Trials Task Force Imaging G, Outcome Measures in Rheumatology Magnetic Resonance Imaging Inflammatory Arthritis Working G (2013) Review: the utility of magnetic resonance imaging for assessing structural damage in randomized controlled trials in rheumatoid arthritis. Arthritis Rheum 65:2513–2523.
    1. Axelsen MB, Poggenborg RP, Stoltenberg M, Kubassova O, Boesen M, et al. (2013) Reliability and responsiveness of dynamic contrast-enhanced magnetic resonance imaging in rheumatoid arthritis. Scand J Rheumatol 42:115–122.
    1. Hodgson R, Grainger A, O'Connor P, Barnes T, Connolly S, et al. (2008) Dynamic contrast enhanced MRI of bone marrow oedema in rheumatoid arthritis. Ann Rheum Dis 67:270–272.
    1. Orguc S, Tikiz C, Aslanalp Z, Erbay PD (2013) Comparison of OMERACT-RAMRIS scores and computer-aided dynamic magnetic resonance imaging findings of hand and wrist as a measure of activity in rheumatoid arthritis. Rheumatol Int 33:1837–1844.
    1. Coulthard LR, Geiler J, Mathews RJ, Church LD, Dickie LJ, et al. (2012) Differential effects of infliximab on absolute circulating blood leucocyte counts of innate immune cells in early and late rheumatoid arthritis patients. Clin Exp Immunol 170:36–46.
    1. Oduro KA, Liu F, Tan Q, Kim C-K, Lubman O, et al. (2012) Myeloid skewing in murine autoimmune arthritis occurs in hematopoietic stem and primitive progenitor cells. Blood 120:2203–2213.
    1. Leizer T, Cebon J, Layton JE, Hamilton JA (1990) Cytokine regulation of colony-stimulating factor production in cultured human synovial fibroblasts: I. Induction of GM-CSF and G-CSF production by interleukin-1 and tumor necrosis factor. Blood 76:1989–1996.
    1. Kaushansky K, Broudy VC, Harlan JM, Adamson JW (1988) Tumor necrosis factor-alpha and tumor necrosis factor-beta (lymphotoxin) stimulate the production of granulocyte-macrophage colony-stimulating factor, macrophage colony-stimulating factor, and IL-1 in vivo. J Immunol 141:3410–3415.
    1. Boilard E, Blanco P, Nigrovic PA (2012) Platelets: active players in the pathogenesis of arthritis and SLE. Nat Rev Rheumatol 8:534–542.
    1. Pignatelli P, De Biase L, Lenti L, Tocci G, Brunelli A, et al. (2005) Tumor necrosis factor-alpha as trigger of platelet activation in patients with heart failure. Blood 106:1992–1994.
    1. Dan K, Gomi S, Inokuchi K, Ogata K, Yamada T, et al. (1995) Effects of interleukin-1 and tumor necrosis factor on megakaryocytopoiesis: mechanism of reactive thrombocytosis. Acta Haematol 93:67–72.
    1. Meany HJ, Seibel NL, Sun J, Finklestein JZ, Sato J, et al. (2008) Phase 2 trial of recombinant tumor necrosis factor-alpha in combination with dactinomycin in children with recurrent Wilms tumor. J Immunother 31:679–683.
    1. Piguet PF, Vesin C, Da Kan C (2002) Activation of platelet caspases by TNF and its consequences for kinetics. Cytokine 18:222–230.
    1. Tacchini-Cottier F, Vesin C, Redard M, Buurman W, Piguet PF (1998) Role of TNFR1 and TNFR2 in TNF-induced platelet consumption in mice. J Immunol 160:6182–6186.
    1. Ueda Y, Yang K, Foster SJ, Kondo M, Kelsoe G (2004) Inflammation controls B lymphopoiesis by regulating chemokine CXCL12 expression. J Exp Med 199:47–58.
    1. Kekow J, Mueller-Ladner U, Schulze-Koops H (2012) Rituximab is more effective than second anti-TNF therapy in rheumatoid arthritis patients and previous TNFα blocker failure. Biologics 6:191–199.

Source: PubMed

3
S'abonner