A randomized double-blind, placebo-controlled clinical phase IIa trial on safety, immunomodulatory effects and pharmacokinetics of EA-230 during experimental human endotoxaemia

Roger van Groenendael, Matthijs Kox, Guus Leijte, Bouke Koeneman, Jelle Gerretsen, Lucas van Eijk, Peter Pickkers, Roger van Groenendael, Matthijs Kox, Guus Leijte, Bouke Koeneman, Jelle Gerretsen, Lucas van Eijk, Peter Pickkers

Abstract

Aims: EA-230 is a human chorionic gonadotropin hormone-derived linear tetrapeptide, developed for the treatment of systemic inflammation-related disorders. EA-230 has shown promising immunomodulatory and tissue-protective effects in animals and an excellent safety profile in human phase I studies that we performed. The present phase IIa study follows-up on these results by investigating the safety, efficacy and pharmacokinetics of EA-230 under systemic inflammatory conditions induced by experimental human endotoxaemia.

Methods: In this randomized, double blind, placebo-controlled phase IIa study, systemic inflammation was induced by intravenous administration of Escherichia coli-derived lipopolysaccharide (LPS). At t = 0 hours, 36 healthy male volunteers received 2 ng/kg LPS, followed by a 2-hour continuous infusion of EA-230 (15, 45 and 90 mg/kg/h, n = 8 per group) or placebo (n = 12).

Results: EA-230 was well tolerated and showed a favourable safety profile. Treatment with the highest dose of EA-230 resulted in a significant attenuation of the LPS-induced increase in plasma levels of inflammatory mediators interleukin (IL)-6, IL-8, IL-1 receptor antagonist, monocyte chemoattractant protein-1, macrophage inflammatory proteins-1α and -1β, and vascular cell adhesion protein-1 (% reduction of 48, 28, 33, 28, 14, 16 and 19 respectively, p < .01), and reduced fever (peak decrease from 1.8 ± 0.1°C to 1.3 ± 0.2°C, P < .05) and symptom scores (peak decrease from 7.4 ± 1.0 to 4.0 ± 1.2 points, P < .05). EA-230 exhibited a very short elimination half-life and a large volume of distribution in the highest dosage group (geometric mean and 95% confidence interval: 0.17 [0.12-0.24] hours and 2.2 [1.3-3.8] L/kg, respectively).

Conclusion: Administration of EA-230 is safe and results in attenuation of the systemic inflammatory response in humans.

Trial registration: ClinicalTrials.gov NCT02629874.

Keywords: cytokines; drug safety; immunotherapy; inflammation; pharmacokinetics.

Conflict of interest statement

P.P. received consulting fees and travel reimbursements from EBI. All other authors have no competing interests to declare.

All protocols, including amendments, were approved by the local ethics committee (CMO Arnhem‐Nijmegen, NL56102.091.15; 2015–2231). The study was conducted in accordance with the principles of the Declaration of Helsinki, and in compliance with the International Conference on Harmonisation E6 Guideline for Good Clinical Practice (CPMP/ICH/135/95). All healthy volunteers who participated in the study provided written informed consent before the start of any study‐related procedures.

© 2019 The Authors. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

Figure 1
Figure 1
Schematic overview of study procedures. Time points on the horizontal axes are in hours relative to LPS administration unless specified otherwise. ICU: intensive care unit; LPS: lipopolysaccharide; (S)AEs: (serious) adverse events; h: hours; d: days
Figure 2
Figure 2
Plasma levels of cytokines during endotoxaemia. A, Interleukin (IL)‐6, B, IL‐1 receptor antagonist (RA), C, tumour necrosis factor (TNF)‐α, D, IL‐10. Data are represented as means with standard error of the mean of n = 7 in the EA‐230 90 mg/kg/h group and n = 12 in the placebo group. Grey box indicates the period in which the active group received EA‐230. P‐values between groups were calculated using repeated measures 2‐way analysis of variance (ANOVA, interaction term)
Figure 3
Figure 3
Plasma levels of chemokines during endotoxaemia. A, Interleukin (IL)‐8, B, monocyte chemoattractant protein (MCP)‐1, C, macrophage inflammatory protein (MIP)‐1α D, MIP‐1β. Data are represented as means with standard error of the mean of n = 7 in the EA‐230 90 mg/kg/h group and n = 12 in the placebo group. Grey box indicates the period in which the active group received EA‐230. P‐values between groups were calculated using repeated measures 2‐way analysis of variance (ANOVA, interaction term)
Figure 4
Figure 4
Plasma levels of endothelial cell adhesion molecules during endotoxaemia. A, Vascular cell adhesion molecule (vCAM)‐1; B, intercellular adhesion molecule (iCAM)‐1. Data are represented as means with standard error of the mean of n = 7 in the EA‐230 90 mg/kg/h group and n = 12 in the placebo group. Grey box indicates the period in which the active group received EA‐230. P‐values between groups were calculated using repeated measures 2‐way analysis of variance (ANOVA, interaction term)
Figure 5
Figure 5
Circulating leucocyte numbers during endotoxaemia. A, Leucocytes, B, neutrophils, C, lymphocytes, D, monocytes. Data are represented as means with standard error of the mean of n = 7 in the EA‐230 90 mg/kg/h group and n = 12 in the placebo group. Grey box indicates the period in which the active group received EA‐230. P‐values between groups were calculated using repeated measures 2‐way analysis of variance (ANOVA, interaction term)
Figure 6
Figure 6
Clinical variables during endotoxaemia. A, Increase in body temperature B, symptom scores, C, mean arterial pressure, D, heart rate. Data are represented as means with standard error of the mean of n = 7 in the EA‐230 90 mg/kg/h group and n = 12 in the placebo group. Grey box indicates the period in which the active group received EA‐230. P‐values between groups were calculated using repeated measures 2‐way analysis of variance (ANOVA, interaction term). AU: arbitrary unit; Bpm: beats/min
Figure 7
Figure 7
Pharmacokinetics of EA‐230. A, Plasma concentration‐time profiles of EA‐230. The grey area indicates the study drug administration period. B, C, Dose proportionality of dose‐normalized, log‐transformed exposure parameters Cmax (B), and AUC0‐last (C). Linear regression lines are shown, dotted lines indicate the 95% confidence interval, a P‐value of <.05 would have indicated nonproportionality. Data are expressed as geometric means and 95% confidence interval

References

    1. Balk RA. Systemic inflammatory response syndrome (SIRS): where did it come from and is it still relevant today? Virulence. 2014;5(1):20‐26.
    1. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis‐3). JAMA. 2016;315(8):801‐810.
    1. Vincent JL, Sakr Y, Sprung CL, et al. Sepsis in European intensive care units: results of the SOAP study. Crit Care Med. 2006;34(2):344‐353.
    1. Dulhunty JM, Lipman J, Finfer S. Does severe non‐infectious SIRS differ from severe sepsis? Results from a multi‐centre Australian and New Zealand intensive care unit study. Intensive Care Med. 2008;34(9):1654‐1661.
    1. Thiele RH, Isbell JM, Rosner MH. AKI associated with cardiac surgery. Clin J Am Soc Nephrol. 2015;10(3):500‐514.
    1. Hoste EA, Bagshaw SM, Bellomo R, et al. Epidemiology of acute kidney injury in critically ill patients: the multinational AKI‐EPI study. Intensive Care Med. 2015;41(8):1411‐1423.
    1. Rabb H, Griffin MD, McKay DB, et al. Inflammation in AKI: current understanding, key questions, and knowledge gaps. J Am Soc Nephrol. 2016;27(2):371‐379.
    1. Payen D, Lukaszewicz AC, Legrand M, et al. A multicentre study of acute kidney injury in severe sepsis and septic shock: association with inflammatory phenotype and HLA genotype. PLoS One. 2012;7(6):e35838.
    1. Cohen J, Vincent JL, Adhikari NK, et al. Sepsis: a roadmap for future research. Lancet Infect Dis. 2015;15(5):581‐614.
    1. Lord JM, Midwinter MJ, Chen YF, et al. The systemic immune response to trauma: an overview of pathophysiology and treatment. Lancet. 2014;384(9952):1455‐1465.
    1. Landis RC, Brown JR, Fitzgerald D, et al. Attenuating the systemic inflammatory response to adult cardiopulmonary bypass: a critical review of the evidence base. J Extra Corpor Technol. 2014;46(3):197‐211.
    1. Munoz‐Suano A, Hamilton AB, Betz AG. Gimme shelter: the immune system during pregnancy. Immunol Rev. 2011;241(1):20‐38.
    1. Racicot K, Kwon JY, Aldo P, Silasi M, Mor G. Understanding the complexity of the immune system during pregnancy. Am J Reprod Immunol. 2014;72(2):107‐116.
    1. Vukusic S, Hutchinson M, Hours M, et al. Pregnancy and multiple sclerosis (the PRIMS study): clinical predictors of post‐partum relapse. Brain. 2004;127(Pt 6):1353‐1360.
    1. Kaaja RJ, Greer IA. Manifestations of chronic disease during pregnancy. JAMA. 2005;294(21):2751‐2757.
    1. Hazes JM, Coulie PG, Geenen V, et al. Rheumatoid arthritis and pregnancy: evolution of disease activity and pathophysiological considerations for drug use. Rheumatology (Oxford). 2011;50(11):1955‐1968.
    1. Murase JE, Chan KK, Garite TJ, Cooper DM, Weinstein GD. Hormonal effect on psoriasis in pregnancy and post partum. Arch Dermatol. 2005;141(5):601‐606.
    1. Adcock EW 3rd, Teasdale T, August CS, et al. Human chorionic gonadotropin: its possible role in maternal lymphocyte suppression. Science. 1973;181(4102):845‐847.
    1. Wan H, Versnel MA, Leijten LM, et al. Chorionic gonadotropin induces dendritic cells to express a tolerogenic phenotype. J Leukoc Biol. 2008;83(4):894‐901.
    1. Bansal AS, Bora SA, Saso S, Smith JR, Johnson MR, Thum MY. Mechanism of human chorionic gonadotrophin‐mediated immunomodulation in pregnancy. Expert Rev Clin Immunol. 2012;8(8):747‐753.
    1. Schumacher A, Brachwitz N, Sohr S, et al. Human chorionic gonadotropin attracts regulatory T cells into the fetal‐maternal interface during early human pregnancy. J Immunol. 2009;182(9):5488‐5497.
    1. Khan NA, Benner R. Human chorionic gonadotropin: a model molecule for oligopeptide‐based drug discovery. Endocr Metab Immune Disord Drug Targets. 2011;11(1):32‐53.
    1. Khan NA, Khan A, Savelkoul HF, Benner R. Inhibition of septic shock in mice by an oligopeptide from the beta‐chain of human chorionic gonadotrophin hormone. Hum Immunol. 2002;63(1):1‐7.
    1. Khan NA, Susa D, van den Berg JW, et al. Amelioration of renal ischaemia‐reperfusion injury by synthetic oligopeptides related to human chorionic gonadotropin. Nephrol Dial Transplant. 2009;24(9):2701‐2708.
    1. Gueler F, Shushakova N, Mengel M, et al. A novel therapy to attenuate acute kidney injury and ischemic allograft damage after allogenic kidney transplantation in mice. PLoS One. 2015;10(1):e0115709.
    1. Khan NA, Vierboom MP, van Holten‐Neelen C, et al. Mitigation of septic shock in mice and rhesus monkeys by human chorionic gonadotrophin‐related oligopeptides. Clin Exp Immunol. 2010;160(3):466‐478.
    1. van der Zee M, Dik WA, Kap YS, et al. Synthetic human chorionic gonadotropin‐related oligopeptides impair early innate immune responses to Listeria monocytogenes in mice. J Infect Dis. 2010;201(7):1072‐1080.
    1. van den Berg HR, Khan NA, van der Zee M, et al. Synthetic oligopeptides related to the [beta]‐subunit of human chorionic gonadotropin attenuate inflammation and liver damage after (trauma) hemorrhagic shock and resuscitation. Shock. 2009;31(3):285‐291.
    1. van der Zee M, van den Berg JW, van Holten‐Neelen C, Dik WA. The beta‐human chorionic gonadotropin‐related peptide LQGV exerts anti‐inflammatory effects through activation of the adrenal gland and glucocorticoid receptor in C57BL/6 mice. J Immunol. 2010;185(9):5066‐5073.
    1. van den Berg JW, Dik WA, van der Zee M, et al. The beta‐human chorionic gonadotropin‐related peptide LQGV reduces mortality and inflammation in a murine polymicrobial sepsis model. Crit Care Med. 2011;39(1):126‐134.
    1. Zamorina SA, Shirshev SV. Oligopeptides of chorionic gonadotropin beta‐subunit in induction of T cell differentiation into Treg and Th17. Bull Exp Biol Med. 2015;160(1):72‐75.
    1. van Groenendael R, Aarnoutse R, Kox M, van Eijk LT, Pickkers P. Pharmacokinetics, safety and tolerability of the novel β‐hCG derived immunomodulatory compound, EA‐230. Br J Clin Pharmacol. 2019;85(7):1572‐1584.
    1. Dorresteijn MJ, van Eijk LT, Netea MG, Smits P, van der Hoeven JG, Pickkers P. Iso‐osmolar prehydration shifts the cytokine response towards a more anti‐inflammatory balance in human endotoxemia. J Endotoxin Res. 2005;11(5):287‐293.
    1. van Eijk LT, Pickkers P, Smits P, Bouw MP, van der Hoeven JG. Severe vagal response after endotoxin administration in humans. Intensive Care Med. 2004;30(12):2279‐2281.
    1. van Lier D, Geven C, Leijte GP, Pickkers P. Experimental human endotoxemia as a model of systemic inflammation. Biochimie. 2019; 159:99‐106.
    1. NIH . Common Terminology Criteria for Adverse Events (CTCAE) v4.0. 2009. (May 28 (v4.03: June 14 2010)).
    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(1–2):124‐135.
    1. Watt DG, Horgan PG, McMillan DC. Routine clinical markers of the magnitude of the systemic inflammatory response after elective operation: a systematic review. Surgery. 2015;157(2):362‐380.
    1. Kellum JA, Kong L, Fink MP, et al. Understanding the inflammatory cytokine response in pneumonia and sepsis: results of the Genetic and Inflammatory Markers of Sepsis (GenIMS) Study. Arch Intern Med. 2007;167(15):1655‐1663.
    1. Cuschieri J, Bulger E, Schaeffer V, et al. Early elevation in random plasma IL‐6 after severe injury is associated with development of organ failure. Shock. 2010;34(4):346‐351.
    1. Zhang WR, Garg AX, Coca SG, et al. Plasma IL‐6 and IL‐10 concentrations predict AKI and long‐term mortality in adults after cardiac surgery. J Am Soc Nephrol. 2015;26(12):3123‐3132.
    1. Vaahersalo J, Skrifvars MB, Pulkki K, et al. Admission interleukin‐6 is associated with post resuscitation organ dysfunction and predicts long‐term neurological outcome after out‐of‐hospital ventricular fibrillation. Resuscitation. 2014;85(11):1573‐1579.
    1. Bustamante A, Sobrino T, Giralt D, et al. Prognostic value of blood interleukin‐6 in the prediction of functional outcome after stroke: a systematic review and meta‐analysis. J Neuroimmunol. 2014;274(1–2):215‐224.
    1. Remick DG, Bolgos GR, Siddiqui J, Shin J, Nemzek JA. Six at six: interleukin‐6 measured 6 h after the initiation of sepsis predicts mortality over 3 days. Shock. 2002;17(6):463‐467.
    1. Nechemia‐Arbely Y, Barkan D, Pizov G, et al. IL‐6/IL‐6R axis plays a critical role in acute kidney injury. J Am Soc Nephrol. 2008;19(6):1106‐1115.
    1. Pecoits‐Filho R, Barany P, Lindholm B, Heimburger O, Stenvinkel P. Interleukin‐6 is an independent predictor of mortality in patients starting dialysis treatment. Nephrol Dial Transplant. 2002;17(9):1684‐1688.
    1. Howell KW, Cleveland JC Jr, Meng X, et al. Interleukin 6 production during cardiac surgery correlates with increasing age. J Surg Res. 2016;201(1):76‐81.
    1. van Eijk LT, Dorresteijn MJ, Smits P, van der Hoeven JG, Netea MG, Pickkers P. Gender differences in the innate immune response and vascular reactivity following the administration of endotoxin to human volunteers. Crit Care Med. 2007;35(6):1464‐1469.
    1. van Groenendael R, Beunders R, Hofland J, et al. The safety, tolerability, and effects on the systemic inflammatory response and renal function of the human chorionic gonadotropin hormone‐derivative EA‐230 following on‐pump cardiac surgery (the EASI Study): protocol for a randomized, double‐blind, placebo‐controlled phase 2 study. JMIR Res Protoc. 2019;8(2):e11441.

Source: PubMed

3
Suscribir