Melatonin as a potential therapy for sepsis: a phase I dose escalation study and an ex vivo whole blood model under conditions of sepsis

Helen F Galley, Damon A Lowes, Lee Allen, Gary Cameron, Lorna S Aucott, Nigel R Webster, Helen F Galley, Damon A Lowes, Lee Allen, Gary Cameron, Lorna S Aucott, Nigel R Webster

Abstract

Sepsis is a massive inflammatory response mediated by infection, characterized by oxidative stress, release of cytokines, and mitochondrial dysfunction. Melatonin accumulates in mitochondria, and both it and its metabolites have potent antioxidant and anti-inflammatory activities and may be useful in sepsis. We undertook a phase I dose escalation study in healthy volunteers to assess the tolerability and pharmacokinetics of 20, 30, 50, and 100 mg oral doses of melatonin. In addition, we developed an ex vivo whole blood model under conditions mimicking sepsis to determine the bioactivity of melatonin and the major metabolite 6-hydroxymelatonin at relevant concentrations. For the phase I trial, oral melatonin was given to five subjects in each dose cohort (n = 20). Blood and urine were collected for measurement of melatonin and 6-hydroxymelatonin, and symptoms and physiological measures were assessed. Validated sleep scales were completed. No adverse effects after oral melatonin, other than mild transient drowsiness with no effects on sleeping patterns, were seen, and no symptoms were reported. Melatonin was rapidly cleared at all doses with a median [range] elimination half-life of 51.7 [29.5-63.2] min across all doses. There was considerable variability in maximum melatonin levels within each dose cohort, but 6-hydoxymelatonin sulfate levels were less variable and remained stable for several hours. For the ex vivo study, blood from 20 volunteers was treated with lipopolysaccharide and peptidoglycan plus a range of concentrations of melatonin/6-hydroxymelatonin. Both melatonin and 6-hydroxymelatonin had beneficial effects on sepsis-induced mitochondrial dysfunction, oxidative stress, and cytokine responses at concentrations similar to those achieved in vivo.

Keywords: 6-hydroxymelatonin; cytokines; melatonin; phase I clinical trial; sepsis.

© 2014 The Authors. Journal of Pineal Research Published by John Wiley & Sons Ltd.

Figures

Fig 1
Fig 1
Physiological measures in healthy subjects over 6 hr after receiving an oral dose of melatonin. (A) Heart rate, (B) oxygen saturation, (C) systolic blood pressure, and (D) diastolic blood pressure. Median values shown for clarity, n = 5 subjects per dose cohort. Multilevel linear modeling showed that oxygen saturation and diastolic blood pressure decreased over time, unrelated to dose (P = 0.017 and P < 0.001, respectively), and systolic blood pressure increased with melatonin dose (P < 0.005).
Fig 2
Fig 2
Seum 6-hydroxymelatonin sulfate levels in healthy subjects over 6 hr after an oral dose of melatonin. Median and full range shown, n = 5 per dose cohort. There was a significant effect of dose (P = 0.028), and levels after 30, 50, or 100 mg were significantly higher than after 20 mg (all < 0.001).
Fig 3
Fig 3
Serum melatonin (red) and serum 6-hydroxymelatonin (blue) in individual subjects following an oral dose of 50 mg melatonin (n = 5 per dose cohort). Lines show median values.
Fig 4
Fig 4
(A) Plasma interleukin-6 (IL-6), (B) IL-10, and (C) lipid hydroperoxide (LPO) from whole blood treated with solvent control (green), lipopolysaccharide and peptidoglycan G (LPS/PepG) plus melatonin (red) or 6-hydroxymelatonin (blue). Median and interquartile range, n = 20 subjects. P-value is Page's trend test. and * = significantly lower than with LPS/PepG alone (Wilcoxon Signed Ranks, P < 0.0001)
Fig 5
Fig 5
Respiratory burst in whole blood treated with solvent control (green), lipopolysaccharide and peptidoglycan G (LPS/PepG) plus melatonin (red) or 6-hydroxymelatonin (blue). Median and interquartile range, n = 20 subjects. P-value is Page's trend test. and * = significantly lower than with LPS/PepG alone (Wilcoxon Signed Ranks, P < 0.0001)

References

    1. Marshall JC, Vincent JL, Guyatt G, et al. Outcome measures for clinical research in sepsis: a report of the 2nd Cambridge Colloquium of the international sepsis forum. Crit Care Med. 2005;33:1708–1716.
    1. Rinaldi S, Landucci F, de Gaudio AR. Antioxidant therapy in critically ill septic patients. Curr Drug Targets. 2009;10:872–880.
    1. Crouser ED. Mitochondrial dysfunction in septic shock and multiple organ dysfunction syndrome. Mitochondrion. 2004;4:729–741.
    1. Galley HF. Bench to bedside review: Targeting antioxidants to mitochondria in sepsis. Crit Care. 2010;14:230.
    1. Víctor VM, Espulgues JV, Hernández-Mijares A, et al. Oxidative stress and mitochondrial dysfunction in sepsis: a potential therapy with mitochondria-targeted antioxidants. Infect Disord Drug Targets. 2009;9:376–389.
    1. Lowes DA, Thottakam BMVJ, Webster NR, et al. The mitochondria-targeted antioxidant MitoQ protects against organ damage in a lipopolysaccharide-peptidoglycan model of sepsis. Free Radic Biol Med. 2008;45:1559–1565.
    1. Lowes DA, Webster NR, Murphy MP, et al. Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis. Br J Anaesth. 2013;110:472–480.
    1. Galano A, Tan DX, Reiter RJ. Melatonin as a natural ally against oxidative stress: a physicochemical examination. J Pineal Res. 2011;51:1–16.
    1. Reiter RJ, Tan DX, Rosales-Corral S, et al. The universal nature, unequal distribution and antioxidant functions of melatonin and its derivatives. Mini Rev Med Chem. 2013;13:373–384.
    1. Venegas C, García JA, Escames G, et al. Extrapineal melatonin: analysis of its subcellular distribution and daily fluctuations. J Pineal Res. 2012;52:217–227.
    1. Lowes DA, Almawash AM, Webster NR, et al. Melatonin and structurally similar compounds have differing effects on inflammation and mitochondrial function in endothelial cells under conditions mimicking sepsis. Br J Anaesth. 2011;107:193–201.
    1. Galano A, Tan DX, Reiter RJ. On the free radical scavenging activities of melatonin's metabolites. AFMK and AMK. J Pineal Res. 2013;54:245–257.
    1. Li Volti G, Musumeci T, Pignatello R, et al. Antioxidant potential of different melatonin-loaded nanomedicines in an experimental model of sepsis. Exp Biol Med. 2012;237:670–677.
    1. Shang Y, Xu SP, Wu Y, et al. Melatonin reduces acute lung injury in endotoxemic rats. Chin Med J (Engl) 2009;122:1388–1393.
    1. Wu JY, Tsou MY, Chen TH, et al. Therapeutic effects of melatonin on peritonitis-induced septic shock with multiple organ dysfunction syndrome in rats. J Pineal Res. 2008;45:106–116.
    1. Snyder-Halpern R, Verran JA. Instrumentation to describe subjective sleep characteristics in healthy subjects. Res Nurs Health. 1987;10:155–163.
    1. Kumar A, Ray P, Kanwar M, et al. A comparative analysis of antibody repertoire against Staphylococcus aureus antigens in patients with deep-seated versus superficial staphylococcal infections. Int J Med Sci. 2005;2:129–136.
    1. Mukhopadhyay P, Rajesh M, Yoshihiro K, et al. Simple quantitative detection of mitochondrial superoxide production in live cells. Biochem Biophys Res Commun. 2007;358:203–208.
    1. Collesa S, Chisolm GM. Lysophosphatidylcholine-induced cellular injury in cultured fibroblasts involves oxidative events. J Lipid Res. 2000;41:1188–1198.
    1. Smiley ST, Reers M, Mottola-Hartshorn C, et al. Intracellular heterogeneity in mitochondrial membrane potentials revealed by a J-aggregate-forming lipophilic cation JC-1. Proc Natl Acad Sci USA. 1991;88:3671–3675.
    1. Petita C, Tri-Rouxela FP, Lesneb A, et al. Oxygen consumption by cultured human cells is impaired by a nucleoside analogue cocktail that inhibits mitochondrial DNA synthesis. Mitochondrion. 2005;5:154–161.
    1. Demuro RL, Nafziger AN, Blask DE, et al. The absolute bioavailability of oral melatonin. J Clin Pharmacol. 2000;40:781–784.
    1. Waldhauser F, Waldhauser M, Lieberman HR, et al. Bioavailability of oral melatonin in humans. Neuroendocrinology. 1984;39:307–313.
    1. Fourtillan JB, Brisson AM, Gobin P, et al. Bioavailability of melatonin in humans after day-time administration of D(7) melatonin. Biopharm Drug Dispos. 2000;21:15–22.
    1. Di WL, Kadva A, Johnston A, et al. Variable bioavailability of oral melatonin. N Engl J Med. 1997;336:1028–1029.
    1. Lane EA, Moss HB. Pharmacokinetics of melatonin in man: first pass hepatic metabolism. J Clin Endocrinol Metab. 1985;61:1214–1216.
    1. Ma X, Idle JR, Krausz KW, et al. Metabolism of melatonin by human cytochromes p450. Drug Metab Dispos. 2005;33:489–494.
    1. Facciolá G, Hidestrand M, von Bahr C, et al. Cytochrome P450 isoforms involved in melatonin metabolism in human liver microsomes. Eur J Clin Pharmacol. 2001;56:881–888.
    1. Young IM, Leone RM, Francis P, et al. Melatonin is metabolized to n-acetyl serotonin and 6-hydroxymelatonin in man. J Clin Endocrinol. 1985;60:114–119.
    1. Ortiz F, García JA, Acuña-Castroviejo D, et al. The beneficial effects of melatonin against heart mitochondrial impairment during sepsis: inhibition of iNOS and preservation of nNOS. J Pineal Res. 2014;56:71–81.
    1. Maharaj DS, Walker RB, Glass BD, et al. 6-Hydroxymelatonin protects against cyanide induced oxidative stress in rat brain homogenates. J Chem Neuroanat. 2003;26:103–107.
    1. Duan Q, Wang Z, Lu T, et al. Comparison of 6-hydroxylmelatonin or melatonin in protecting neurons against ischemia/reperfusion-mediated injury. J Pineal Res. 2006;41:351–357.
    1. Calvo JR, Reiter RJ, García JJ, et al. Characterization of the protective effects of melatonin and related indoles against alpha-naphthylisothiocyanate-induced liver injury in rats. J Cell Biochem. 2001;80:461–470.
    1. Lopez-Burillo S, Tan DX, Rodriguez-Gallego V, et al. Melatonin and its derivatives cyclic 3-hydroxymelatonin, N1-acetyl-N2-formyl-5-methoxykynuramine and 6-hydroxymelatonin reduce oxidative DNA damage induced by Fenton reagents. J Pineal Res. 2003;34:178–184.
    1. Benloucif S, Burgess HJ, Klerman EB, et al. Measuring melatonin in humans. J Clin Sleep Med. 2008;4:66–69.
    1. Harthe C, Claustrat B, Brun J, et al. Direct radioimmunoassay of 6-sulfatoxymelatonin in plasma with use of an iodinated tracer. Clin Chem. 1991;37:536–539.
    1. Härtter S, Morita S, Bodin K, et al. Determination of exogenous melatonin and its 6-hydroxy metabolite in human plasma by liquid chromatography-mass spectrometry. Ther Drug Monit. 2001;23:282–286.
    1. Griefahn B, Remer T, Blaszkewicz M, et al. Long-term stability of 6-hydroxy melatonin sulfate in 24-h urine samples stored at -20 degrees C. Endocrine. 2001;15:199–202.
    1. Guardiola-Lemaître B. Toxicology of melatonin. J Biol Rhythms. 1997;12:697–706.
    1. Harris AS, Burgess HJ, Dawson D. The effects of day-time exogenous melatonin administration on cardiac autonomic activity. J Pineal Res. 2001;31:199–205.
    1. Sletten T, Burgess H, Savic N, et al. The effects of bright light and nighttime melatonin administration on cardiac activity. J Hum Ergol (Tokyo) 2001;30:273–278.
    1. Markantonis SL, Tsakalozou E, Paraskeva A, et al. Melatonin pharmaco- kinetics in premenopausal and postmenopausal healthy female volunteers. J Clin Pharmacol. 2008;48:240–245.
    1. Seabra ML, Bignotto M, Pinto LRJR, et al. Randomized, double-blind clinical trial, controlled with placebo, of the toxicology of chronic melatonin treatment. J Pineal Res. 2000;29:193–200.
    1. Hughes RJ, Badia P. Sleep-promoting and hypothermic effects of daytime melatonin administration in humans. Sleep. 1997;20:124–131.
    1. Zhdanova I, Wurtman R. Efficacy of melatonin as a sleep-promoting agent. J Biol Rhythms. 1997;12:644–650.
    1. Herxheimer A, Petrie K. Melatonin for the prevention and treatment of jet lag. Cochrane Database Syst Rev. 2002;2:CD00152.
    1. Mishima K, Satoh K, Shimizu T, et al. Hypnotic and hypothermic action of daytime administered melatonin. Psychopharmacol. 1997;133:168–171.
    1. James SP, Mendelson WB, Sack DA, et al. The effect of melatonin on normal sleep. Neuropsychopharmacology. 1987;1:41–44.
    1. Waterhouse J, Reilly T, Atkinson G, et al. Jet lag: trends and coping strategies. Lancet. 2007;369:1117–1129.
    1. Mistraletti G, Sabbatini G, Taverna M, et al. Pharmacokinetics of orally administered melatonin in critically ill patients. J Pineal Res. 2010;48:142–147.
    1. Bourne RS, Mills GH, Minelli C. Melatonin therapy to improve nocturnal sleep in critically ill patients: encouraging results from a small randomised controlled trial. Crit Care. 2008;12:R52.
    1. van der Poll T. Experimental human sepsis models. Drug Discov Today Dis Models. 2012;9:e3–e9.
    1. Calvano SE, Coyle SM. Experimental human endotoxemia: a model of the systemic inflammatory response syndrome? Surg Infect. 2012;13:293–299.
    1. Chen HM, Hsu JT, Chen JC, et al. Delayed neutrophil apoptosis attenuated by melatonin in human acute pancreatitis. Pancreas. 2005;31:360–364.
    1. Miletic AV, Graham DB, Montgrain V, et al. Vav proteins control MyD88-dependent oxidative burst. Blood. 2007;109:3360–3368.
    1. Remer KA, Brcic M, Jungi TW. Toll-like receptor-4 is involved in eliciting an LPS-induced oxidative burst in neutrophils. Immunol Lett. 2003;85:75–80.
    1. Mauriz JL, Collado PS, Veneroso C, et al. A review of the molecular aspects of melatonin's anti-inflammatory actions: recent insights and new perspectives. J Pineal Res. 2013;54:1–14.
    1. Zhou J, Zhang S, Zhao X, et al. Melatonin impairs NADPH oxidase assembly and decreases superoxide anion production in microglia exposed to amyloid-beta1-42. J Pineal Res. 2008;45:157–165.
    1. Xia MZ, Liang YL, Wang H, et al. Melatonin modulates TLR4-mediated inflammatory genes through MyD88- and TRIF-dependent signaling pathways in lipopolysaccharide-stimulated RAW264.7 cells. J Pineal Res. 2012;53:325–334.
    1. Shi D, Xiao X, Wang J, et al. Melatonin suppresses proinflammatory mediators in lipopolysaccharide-stimulated CRL1999 cells via targeting MAPK, NF-kB, c/EBPB, and p300 signaling. J Pineal Res. 2012;53:154–165.
    1. Kizaki T, Suzuki K, Hitomi Y, et al. Uncoupling protein 2 plays an important role in nitric oxide production of lipopolysaccharide-stimulated macrophages. Proc Natl Acad Sci USA. 2002;99:9392–9397.
    1. Emre Y, Hurtaud C, Nübel T, et al. Mitochondria contribute to LPS-induced MAPK activation via uncoupling protein UCP2 in macrophages. Biochem J. 2007;402:271–278.
    1. Bai Y, Onuma H, Bai X, et al. Persistent nuclear factor-κB activation in Ucp2–/– mice leads to enhanced nitric oxide and inflammatory cytokine production. J Biol Chem. 2005;280:19062–19069.
    1. Nishio K, Qiao S, Yamashita H. Characterization of the differential expression of uncoupling protein 2 and ROS production in differentiated mouse macrophage-cells (Mm1) and the progenitor cells (M1) J Mol Histol. 2005;36:35–44.
    1. Alamili M, Bendtzen K, Lykkesfeldt J, et al. Melatonin suppresses markers of inflammation and oxidative damage in a human daytime endotoxemia model. J Crit Care. 2014;29:184.

Source: PubMed

3
Iratkozz fel