Prerequisites for cytokine measurements in clinical trials with multiplex immunoassays

Wilco de Jager, Katarzyna Bourcier, Ger T Rijkers, Berent J Prakken, Vicki Seyfert-Margolis, Wilco de Jager, Katarzyna Bourcier, Ger T Rijkers, Berent J Prakken, Vicki Seyfert-Margolis

Abstract

Background: Growing knowledge about cellular interactions in the immune system, including the central role of cytokine networks, has lead to new treatments using monoclonal antibodies that block specific components of the immune system. Systemic cytokine concentrations can serve as surrogate outcome parameters of these interventions to study inflammatory pathways operative in patients in vivo. This is now possible due to novel technologies such as multiplex immunoassays (MIA) that allows detection of multiple cytokines in a single sample. However, apparently trivial underappreciated processes, (sample handling and storage, interference of endogenous plasma proteins) can greatly impact the reliability and reproducibility of cytokine detection.Therefore we set out to investigate several processes that might impact cytokine profiles such as blood collecting tubes, duration of storage, and number of freeze thawing cycles.

Results: Since under physiological conditions cytokine concentrations normally are low or undetectable we spiked cytokines in the various plasma and serum samples. Overall recoveries ranged between 80-120%. Long time storage showed cytokines are stable for a period up to 2 years of storage at -80 degrees C. After 4 years several cytokines (IL-1alpha, IL-1beta, IL-10, IL-15 and CXCL8) degraded up to 75% or less of baseline values. Furthermore we show that only 2 out of 15 cytokines remained stable after several freeze-thawing cycles. We also demonstrate implementation of an internal control for multiplex cytokine immunoassays.

Conclusion: All together we show parameters which are essential for measurement of cytokines in the context of clinical trials.

Figures

Figure 1
Figure 1
Cytokine profiles in different blood collection tubes. Blood samples were obtained from 4 healthy volunteers using various blood collection tubes after centrifugation cell free plasma (sodium heparin plasma, EDTA plasma and sodium citrate plasma) and serum cytokine levels were measured using a multiplex assay. Color profiles were generated using geometric mean values and plotted using a semi log scale as previously described [14].
Figure 2
Figure 2
Recovery of spiked cytokines in various types of plasma and serum. Cytokines (1000 pg/ml) were spiked in various types of plasma and serum. Recoveries were measured using a multiplex immunoassay. Se serum, NH sodium heparin plasma, E EDTA plasma, CP sodium citrate plasma. Shown are means ± SD.
Figure 3
Figure 3
Long time stability of induced cytokine profiles. Sodium heparin whole blood from 3 individuals was stimulated with a combination of LPS and PHA to induce cytokine profiles. Samples were measured at baseline (BL) and at various time points. Baseline value was set at 100%. Shown are means ± SD.
Figure 4
Figure 4
Recovery of cytokines after multiple freeze - thawing cycle. Aliquots of the same LPS/PHA stimulated samples were freshly thawed or underwent multiple freeze - thawing cycles and cytokines were measured simultaneously. Shown are means ± SD.
Figure 5
Figure 5
Longitudinal assay performance using an internal control sample. Left panel shows variability in time of observed cytokine values of the internal control sample for IL-1β (A), IL-4 (B), I-L6 (C) and TNFα (D) in 50 consecutive runs covering a period of 12 months. Right-hand panel shows overlays of 4 separate standard curves of the corresponding cytokines.

References

    1. Chatenoud L. Immune therapies of autoimmune diseases: are we approaching a real cure? Curr Opin Immunol. 2006;18:710–717. doi: 10.1016/j.coi.2006.09.004.
    1. Holgate ST, Polosa R. The mechanisms, diagnosis, and management of severe asthma in adults. Lancet. 2006;368:780–793. doi: 10.1016/S0140-6736(06)69288-X.
    1. Pascual M, Theruvath T, Kawai T, Tolkoff-Rubin N, Cosimi AB. Strategies to improve long-term outcomes after renal transplantation. N Engl J Med. 2002;346:580–590. doi: 10.1056/NEJMra011295.
    1. Magliocca JF, Knechtle SJ. The evolving role of alemtuzumab (Campath-1H) for immunosuppressive therapy in organ transplantation. Transpl Int. 2006;19:705–714. doi: 10.1111/j.1432-2277.2006.00343.x.
    1. Morris PJ, Russell NK. Alemtuzumab (Campath-1H): a systematic review in organ transplantation. Transplantation. 2006;81:1361–1367. doi: 10.1097/01.tp.0000219235.97036.9c.
    1. Chatenoud L, Bluestone JA. CD3-specific antibodies: a portal to the treatment of autoimmunity. Nat Rev Immunol. 2007;7:622–632. doi: 10.1038/nri2134.
    1. Goodyear M. Learning from the TGN1412 trial. BMJ. 2006;332:677–678. doi: 10.1136/bmj.38797.635012.47.
    1. Mayor S. Severe adverse reactions prompt call for trial design changes. BMJ. 2006;332:683. doi: 10.1136/bmj.332.7543.683.
    1. O'Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Rev Immunol. 2002;2:37–45. doi: 10.1038/nri702.
    1. Mosmann TR, Fong TA. Specific assays for cytokine production by T cells. J Immunol Methods. 1989;116:151–158. doi: 10.1016/0022-1759(89)90198-1.
    1. de Jager W, Rijkers GT. Solid-phase and bead-based cytokine immunoassay: a comparison. Methods. 2006;38:294–303. doi: 10.1016/j.ymeth.2005.11.008.
    1. Fulton RJ, McDade RL, Smith PL, Kienker LJ, Kettman JR., Jr Advanced multiplexed analysis with the FlowMetrix system. Clin Chem. 1997;43:1749–1756.
    1. Selby C. Interference in immunoassay. Ann Clin Biochem. 1999;36:704–721.
    1. de Jager W, Prakken BJ, Bijlsma JW, Kuis W, Rijkers GT. Improved multiplex immunoassay performance in human plasma and synovial fluid following removal of interfering heterophilic antibodies. J Immunol Methods. 2005;300:124–135. doi: 10.1016/j.jim.2005.03.009.
    1. de Jager W, Te Velthuis H, Prakken BJ, Kuis W, Rijkers GT. Simultaneous detection of 15 human cytokines in a single sample of stimulated peripheral blood mononuclear cells. Clin Diagn Lab Immunol. 2003;10:133–139.
    1. Kamphuis S, Kuis W, de Jager W, Teklenburg G, Massa M, Gordon G, et al. Tolerogenic immune responses to novel T-cell epitopes from heat-shock protein 60 in juvenile idiopathic arthritis. Lancet. 2005;366:50–56. doi: 10.1016/S0140-6736(05)66827-4.
    1. de Jager W, Hoppenreijs EP, Wulffraat NM, Wedderburn LR, Kuis W, Prakken BJ. Blood and synovial fluid cytokine signatures in patients with juvenile idiopathic arthritis: a cross-sectional study. Ann Rheum Dis. 2007;66:589–598. doi: 10.1136/ard.2006.061853.
    1. Ham HJ van den, de JW, Bijlsma JW, Prakken BJ, de Boer RJ. Differential cytokine profiles in juvenile idiopathic arthritis subtypes revealed by cluster analysis. Rheumatology (Oxford) 2009;48:899–905. doi: 10.1093/rheumatology/kep125.
    1. Skurkovich SV, Klinova EG, Eremkina EI, Levina NV. Immunosuppressive effect of an anti-interferon serum. Nature. 1974;247:551–552. doi: 10.1038/247551a0.
    1. Blanco P, Palucka AK, Gill M, Pascual V, Banchereau J. Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science. 2001;294:1540–1543. doi: 10.1126/science.1064890.
    1. Shakoor N, Michalska M, Harris CA, Block JA. Drug-induced systemic lupus erythematosus associated with etanercept therapy. Lancet. 2002;359:579–580. doi: 10.1016/S0140-6736(02)07714-0.
    1. Kellar KL, Kalwar RR, Dubois KA, Crouse D, Chafin WD, Kane BE. Multiplexed fluorescent bead-based immunoassays for quantitation of human cytokines in serum and culture supernatants. Cytometry. 2001;45:27–36. doi: 10.1002/1097-0320(20010901)45:1<27::AID-CYTO1141>;2-I.
    1. Vignali DA. Multiplexed particle-based flow cytometric assays. J Immunol Methods. 2000;243:243–255. doi: 10.1016/S0022-1759(00)00238-6.
    1. Petrovsky N, McNair P, Harrison LC. Diurnal rhythms of pro-inflammatory cytokines: regulation by plasma cortisol and therapeutic implications. Cytokine. 1998;10:307–312. doi: 10.1006/cyto.1997.0289.
    1. Petrovsky N, Harrison LC. The chronobiology of human cytokine production. Int Rev Immunol. 1998;16:635–649. doi: 10.3109/08830189809043012.
    1. Lissoni P, Rovelli F, Brivio F, Brivio O, Fumagalli L. Circadian secretions of IL-2, IL-12, IL-6 and IL-10 in relation to the light/dark rhythm of the pineal hormone melatonin in healthy humans. Nat Immun. 1998;16:1–5. doi: 10.1159/000069464.
    1. Arvidson NG, Gudbjornsson B, Elfman L, Ryden AC, Totterman TH, Hallgren R. Circadian rhythm of serum interleukin-6 in rheumatoid arthritis. Ann Rheum Dis. 1994;53:521–524. doi: 10.1136/ard.53.8.521.
    1. Cutolo M, Seriolo B, Craviotto C, Pizzorni C, Sulli A. Circadian rhythms in RA. Ann Rheum Dis. 2003;62:593–596. doi: 10.1136/ard.62.7.593.
    1. Suzuki K, Nakaji S, Yamada M, Totsuka M, Sato K, Sugawara K. Systemic inflammatory response to exhaustive exercise. Cytokine kinetics. Exerc Immunol Rev. 2002;8:6–48.
    1. Woods JA, Vieira VJ, Keylock KT. Exercise, inflammation, and innate immunity. Neurol Clin. 2006;24:585–599. doi: 10.1016/j.ncl.2006.03.008.
    1. Duvigneau JC, Hartl RT, Teinfalt M, Gemeiner M. Delay in processing porcine whole blood affects cytokine expression. J Immunol Methods. 2003;272:11–21. doi: 10.1016/S0022-1759(02)00372-1.
    1. Stack G, Snyder EL. Cytokine generation in stored platelet concentrates. Transfusion. 1994;34:20–25. doi: 10.1046/j.1537-2995.1994.34194098597.x.
    1. Kim SH, Eisenstein M, Reznikov L, Fantuzzi G, Novick D, Rubinstein M, et al. Structural requirements of six naturally occurring isoforms of the IL-18 binding protein to inhibit IL-18. Proc Natl Acad Sci USA. 2000;97:1190–1195. doi: 10.1073/pnas.97.3.1190.
    1. Svenson M, Hansen MB, Heegaard P, Abell K, Bendtzen K. Specific binding of interleukin 1 (IL-1) beta and IL-1 receptor antagonist (IL-1ra) to human serum. High-affinity binding of IL-1ra to soluble IL-1 receptor type I. Cytokine. 1993;5:427–435. doi: 10.1016/1043-4666(93)90032-Z.
    1. Aziz N, Nishanian P, Mitsuyasu R, Detels R, Fahey JL. Variables that affect assays for plasma cytokines and soluble activation markers. Clin Diagn Lab Immunol. 1999;6:89–95.
    1. Martins TB. Development of internal controls for the Luminex instrument as part of a multiplex seven-analyte viral respiratory antibody profile. Clin Diagn Lab Immunol. 2002;9:41–45.
    1. Hulse RE, Kunkler PE, Fedynyshyn JP, Kraig RP. Optimization of multiplexed bead-based cytokine immunoassays for rat serum and brain tissue. J Neurosci Methods. 2004;136:87–98. doi: 10.1016/j.jneumeth.2003.12.023.
    1. Phillips DJ, League SC, Weinstein P, Hooper WC. Interference in microsphere flow cytometric multiplexed immunoassays for human cytokine estimation. Cytokine. 2006;36:180–188. doi: 10.1016/j.cyto.2006.12.002.
    1. Khan SS, Smith MS, Reda D, Suffredini AF, McCoy JP., Jr Multiplex bead array assays for detection of soluble cytokines: comparisons of sensitivity and quantitative values among kits from multiple manufacturers. Cytometry. 2004;61B:35–39. doi: 10.1002/cyto.b.20021.
    1. Djoba Siawaya JF, Roberts T, Babb C, Black G, Golakai HJ, Stanley K, et al. An evaluation of commercial fluorescent bead-based luminex cytokine assays. PLoS ONE. 2008;3:e2535. doi: 10.1371/journal.pone.0002535.
    1. Kricka LJ. Human anti-animal antibody interferences in immunological assays. Clin Chem. 1999;45:942–956.
    1. Martins TB, Pasi BM, Litwin CM, Hill HR. Heterophile antibody interference in a multiplexed fluorescent microsphere immunoassay for quantitation of cytokines in human serum. Clin Diagn Lab Immunol. 2004;11:325–329.
    1. Pfleger C, Schloot N, ter VF. Effect of serum content and diluent selection on assay sensitivity and signal intensity in multiplex bead-based immunoassays. J Immunol Methods. 2008;329:214–218. doi: 10.1016/j.jim.2007.09.011.
    1. Svenson M, Hansen MB, Kayser L, Rasmussen AK, Reimert CM, Bendtzen K. Effects of human anti-IL-1 alpha autoantibodies on receptor binding and biological activities of IL-1. Cytokine. 1992;4:125–133. doi: 10.1016/1043-4666(92)90047-U.

Source: PubMed

3
S'abonner