Pharmacokinetics, pharmacodynamics, safety, and tolerability of nebulized sodium nitrite (AIR001) following repeat-dose inhalation in healthy subjects

Peter J Rix, Andrew Vick, Neil J Attkins, Geoffrey E Barker, Adrian W Bott, Harry Alcorn Jr, Mark T Gladwin, Sruti Shiva, Stephen Bradley, Azra Hussaini, William L Hoye, Ed L Parsley, Hiroko Masamune, Peter J Rix, Andrew Vick, Neil J Attkins, Geoffrey E Barker, Adrian W Bott, Harry Alcorn Jr, Mark T Gladwin, Sruti Shiva, Stephen Bradley, Azra Hussaini, William L Hoye, Ed L Parsley, Hiroko Masamune

Abstract

Introduction: The efficacy of nebulized sodium nitrite (AIR001) has been demonstrated in animal models of pulmonary arterial hypertension (PAH), but it was not known if inhaled nitrite would be well tolerated in human subjects at exposure levels associated with efficacy in these models.

Methods: Inhaled nebulized sodium nitrite was assessed in three independent studies in a total of 82 healthy male and female subjects. Study objectives included determination of the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) under normal and mildly hypoxic conditions, and following co-administration with steady-state sildenafil, assessment of nitrite pharmacokinetics, and evaluation of the fraction exhaled nitric oxide (FENO) and concentrations of iron-nitrosyl hemoglobin (Hb(Fe)-NO) and S-nitrosothiols (R-SNO) as biomarkers of local and systemic NO exposure, respectively.

Results: Nebulized sodium nitrite was well tolerated following 6 days of every 8 h administration up to 90 mg, producing significant increases in circulating Hb(Fe)-NO, R-SNO, and FENO. Pulmonary absorption of nitrite was rapid and complete, and plasma exposure dose was proportional through the MTD dosage level of 90 mg, without accumulation following repeated inhalation. At higher dosage levels, DLTs were orthostasis (observed at 120 mg) and hypotension with tachycardia (at 176 mg), but venous methemoglobin did not exceed 3.0 % at any time in any subject. Neither the tolerability nor pharmacokinetics of nitrite was impacted by conditions of mild hypoxia, or co-administration with sildenafil, supporting the safe use of inhaled nitrite in the clinical setting of PAH.

Conclusion: On the basis of these results, nebulized sodium nitrite (AIR001) has been advanced into randomized trials in PAH patients.

Figures

Fig. 1
Fig. 1
Concentration–time profiles of plasma nitrite relative to the start of a 10-min sodium nitrite inhalation in healthy male and female subjects following a single dose (left) or on day 6 (right) following a total of 16 (every 8 h) administrations in Part A of study CS04. Values represent mean (n = 3 or 6), and error bars represent standard deviation. Dosage levels represent the mass of sodium nitrite loaded into the nebulizer medication chamber. 120-mg dosage level discontinued after day 1 due to DLT in two of three subjects
Fig. 2
Fig. 2
Inter-study comparison of dose response for plasma nitrite Cmax and AUCinf (or AUC0–t for cohorts where AUCinf could not be calculated) following a single sodium nitrite inhalation in healthy male and female subjects. Nitrite concentrations in subjects from studies CS01 and CS02 were individually background subtracted prior to calculation of Cmax and AUC, while no correction was used for study CS04 because pre-dose nitrite was below the limit of detection in all subjects (cf Online Resource 1; Table 1). Values represent mean (n = 3–6), and error bars represent standard deviation. AUCinf area under the plasma concentration–time curve from time zero to infinity, AUC0–t area under the plasma concentration-time curve from time zero to last quantifiable, Cmax maximum plasma concentration
Fig. 3
Fig. 3
Dose response of Cmax and AUC0–8 h for nitrate metabolite in plasma following single (day 1) or repeated (day 6, after 16 total every 8 h doses) inhalation of sodium nitrite in healthy male and female subjects from Part A of study CS04. Nitrate concentrations in subjects were individually background subtracted prior to calculation of Cmax and AUC. Values represent mean (n = 3 or 6), and error bars represent standard deviation. AUC area under the plasma concentration–time curve, AUC0–8 h area under the plasma concentration–time curve from time zero to 8 h, Cmax maximum plasma concentration
Fig. 4
Fig. 4
Fraction exhaled nitric oxide (FENO) in parts per billion (ppb), pre-dose and 5 min after the end of inhalation following single dose (left) or after 16 total every 8 h doses (right) in all sodium nitrite-treated subjects at well-tolerated dosage levels of 15, 45, and 90 mg from Part A of study CS04 (n = 18). p values from paired Student’s t tests of pre- vs. post-dose values on each study day. Mean FENO increased from 16.1 to 68.7 ppb on day 1 (4.3-fold increase) and from 14.8 to 84.3 ppb on day 6 (5.7-fold increase)
Fig. 5
Fig. 5
Concentration–time profiles (n = 3 or 6) of S-nitrosothiol (R-SNO) species (upper) and iron-nitrosyl-hemoglobin (lower) in whole blood following a single 10-min inhalation of sodium nitrite in healthy male and female subjects in Part A of study CS04. Values represent mean (n = 3–6), and error bars represent standard deviation. Dosage levels represent the mass of sodium nitrite loaded into the nebulizer medication chamber. FENO fraction exhaled nitric oxide

References

    1. Rubin LJ. Pulmonary arterial hypertension. Proc Am Thorac Soc. 2006;3(1):111–115. doi: 10.1513/pats.200510-112JH.
    1. Arnold WP, Mittal CK, Katsuki S, Murad F. Nitric oxide activates guanylate cyclase and increases guanosine 3′:5′-cyclic monophosphate levels in various tissue preparations. Proc Natl Acad Sci. 1977;74:3203–3207. doi: 10.1073/pnas.74.8.3203.
    1. Moncada S, Palmer RM, Higgs EA. The discovery of nitric oxide as the endogenous nitrovasodilator. Hypertension. 1988;12:365–372. doi: 10.1161/01.HYP.12.4.365.
    1. Gladwin MT, Shelhamer JH, Schechter AN, Pease-Fye ME, Waclawiw MA, Panza JA, et al. Role of circulating nitrite and S-nitrosohemoglobin in the regulation of regional blood flow in humans. Proc Natl Acad Sci. 2000;97:11482–11487. doi: 10.1073/pnas.97.21.11482.
    1. Doyle MP, Pickering RA, DeWeert TM, Hoekstra JW, Pater D. Kinetics and mechanism of the oxidation of human deoxyhemoglobin by nitrites. J Biol Chem. 1981;256:12393–12398.
    1. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, Martyr S, et al. Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med. 2003;9:1498–1505. doi: 10.1038/nm954.
    1. Shiva S, Huang Z, Grubina R, Sun J, Ringwood LA, MacArthur PH, et al. Deoxymyoglobin is a nitrite reductase that generates nitric oxide and regulates mitochondrial respiration. Circ Res. 2007;100:654–661. doi: 10.1161/01.RES.0000260171.52224.6b.
    1. Rassaf T, Flögel U, Drexhage C, Hendgen-Cotta U, Kelm M, Schrader J. Nitrite reductase function of deoxymyoglobin: oxygen sensor and regulator of cardiac energetics and function. Circ Res. 2007;100:1749–1754. doi: 10.1161/CIRCRESAHA.107.152488.
    1. Hendgen-Cotta UB, Merx MW, Shiva S, Schmitz J, Becher S, Klare JP, et al. Nitrite reductase activity of myoglobin regulates respiration and cellular viability in myocardial ischemia-reperfusion injury. Proc Natl Acad Sci. 2008;105:10256–10261. doi: 10.1073/pnas.0801336105.
    1. Godber BL, Doel JJ, Sapkota GP, Blake DR, Stevens CR, Eisenthal R, et al. Reduction of nitrite to nitric oxide catalyzed by xanthine oxidoreductase. J Biol Chem. 2000;275:7757–7763. doi: 10.1074/jbc.275.11.7757.
    1. Li H, Samouilov A, Liu X, Zweier JL. Characterization of the magnitude and kinetics of xanthine oxidase-catalyzed nitrite reduction: evaluation of its role in nitric oxide generation in anoxic tissues. J Biol Chem. 2001;276:24482–24489. doi: 10.1074/jbc.M011648200.
    1. Zuckerbraun BS, Shiva S, Ifedigbo E, Mathier MA, Mollen KP, Rao J, et al. Nitrite potently inhibits hypoxic and inflammatory pulmonary arterial hypertension and smooth muscle proliferation via xanthine oxidoreductase-dependent nitric oxide generation. Circulation. 2010;121:98–109. doi: 10.1161/CIRCULATIONAHA.109.891077.
    1. Li H, Kundu TK, Zweier JL. Characterization of the magnitude and mechanism of aldehyde oxidase-mediated nitric oxide production from nitrite. J Biol Chem. 2009;284:33850–33858. doi: 10.1074/jbc.M109.019125.
    1. Pinder AG, Pittaway E, Morri K, James PE. Nitrite directly vasodilates hypoxic vasculature via nitric oxide-dependent and -independent pathways. Br J Pharmacol. 2009;157:1523–1530. doi: 10.1111/j.1476-5381.2009.00340.x.
    1. Castello PR, David PS, McClure T, Crook Z, Poyton RO. Mitochondrial cytochrome oxidase produces nitric oxide under hypoxic conditions: implications for oxygen sensing and hypoxic signaling in eukaryotes. Cell Metab. 2006;3:277–287. doi: 10.1016/j.cmet.2006.02.011.
    1. Gautier C, van Faassen E, Mikula I, Martasek P, Slama-Schwok A. Endothelial nitric oxide synthase reduces nitrite anions to NO under anoxia. Biochem Biophys Res Commun. 2006;341:816–821. doi: 10.1016/j.bbrc.2006.01.031.
    1. Vanin AF, Bevers LM, Slama-Schwok A, van Faassen EE. Nitric oxide synthase reduces nitrite to NO under anoxia. Cell Mol Life Sci. 2007;64:96–103. doi: 10.1007/s00018-006-6374-2.
    1. Qiu Y, Sutton L, Riggs AF. Identification of myoglobin in human smooth muscle. J Biol Chem. 1998;273:23426–23432. doi: 10.1074/jbc.273.36.23426.
    1. Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde oxidase: an enzyme of emerging importance in drug discovery. J Med Chem. 2010;53:8441–8460. doi: 10.1021/jm100888d.
    1. Bueno M, Wang J, Mora AL, Gladwin MT. Nitrite signaling in pulmonary hypertension: mechanisms of bioactivation, signaling, and therapeutics. Antioxid Redox Signal. 2013;18:1797–1809. doi: 10.1089/ars.2012.4833.
    1. Hendgen-Cotta UB, Kelm M, Rassaf T. Myoglobin’s novel role in nitrite-induced hypoxic vasodilation. Trends Cardiovasc Med. 2014;24:69–74. doi: 10.1016/j.tcm.2013.06.006.
    1. Cannon RO, 3rd, Schechter AN, Panza JA, Ognibene FP, Pease-Fye ME, Waclawiw MA, et al. Effects of inhaled nitric oxide on regional blood flow are consistent with intravascular nitric oxide delivery. J Clin Invest. 2001;108:279–287. doi: 10.1172/JCI200112761.
    1. Curtis E, Hsu LL, Noguchi AC, Geary L, Shiva S. Oxygen regulates tissue nitrite metabolism. Antioxid Redox Signal. 2012;17:951–961. doi: 10.1089/ars.2011.4242.
    1. Dejam A, Hunter CJ, Tremonti C, Pluta RM, Hon YY, Grimes G, et al. Nitrite infusion in humans and nonhuman primates: endocrine effects, pharmacokinetics, and tolerance formation. Circulation. 2007;116:1821–1831. doi: 10.1161/CIRCULATIONAHA.107.712133.
    1. Pluta RM, Oldfield EH, Bakhtian KD, Fathi AR, Smith RK, Devroom HL, et al. Safety and feasibility of long-term intravenous sodium nitrite infusion in healthy volunteers. PLoS One. 2011;6(1):e14504. doi: 10.1371/journal.pone.0014504.
    1. Maher AR, Arif S, Madhani M, Abozguia K, Ahmed I, Fernandez BO, et al. Impact of chronic congestive heart failure on pharmacokinetics and vasomotor effects of infused nitrite. Br J Pharmacol. 2013;169:659–670. doi: 10.1111/bph.12152.
    1. Hunter CJ, Dejam A, Blood AB, Shields H, Kim-Shapiro DB, Machado RF, et al. Inhaled nebulized nitrite is a hypoxia-sensitive NO-dependent selective pulmonary vasodilator. Nat Med. 2004;10:1122–1127. doi: 10.1038/nm1109.
    1. Ingram TE, Pinder AG, Bailey DM, Fraser AG, James PE. Low-dose sodium nitrite vasodilates hypoxic human pulmonary vasculature by a means that is not dependent on a simultaneous elevation in plasma nitrite. Am J Physiol Heart Circ Physiol. 2010;298:H331–H339. doi: 10.1152/ajpheart.00583.2009.
    1. Hunault CC, van Velzen AG, Sips AJ, Schothorst RC, Meulenbelt J. Bioavailability of sodium nitrite from an aqueous solution in healthy adults. Toxicol Lett. 2009;190:48–53. doi: 10.1016/j.toxlet.2009.06.865.
    1. Oldfield EH, Loomba JJ, Monteith SJ, Crowley RW, Medel R, Gress DR, et al. Safety and pharmacokinetics of sodium nitrite in patients with subarachnoid hemorrhage: a phase IIa study. J Neurosurg. 2013;119:634–641. doi: 10.3171/2013.3.JNS13266.
    1. Mack AK, McGowan Ii VR, Tremonti CK, Ackah D, Barnett C, Machado RF, et al. Sodium nitrite promotes regional blood flow in patients with sickle cell disease: a phase I/II study. Br J Haematol. 2008;142:971–978. doi: 10.1111/j.1365-2141.2008.07259.x.
    1. Maher AR, Milsom AB, Gunaruwan P, Abozguia K, Ahmed I, Weaver RA, et al. Hypoxic modulation of exogenous nitrite-induced vasodilation in humans. Circulation. 2008;117:670–677. doi: 10.1161/CIRCULATIONAHA.107.719591.
    1. International Conference on Harmonisation (ICH) guideline for good clinical practice: E6(R1). 1996. . Accessed 15 Dec 2011.
    1. FDA/CDER/CVM Guidance for industry. Bioanalytical method validation. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Veterinary Medicine (CVM). 2001.
    1. MacArthur PH, Shiva S, Gladwin MT. Measurement of circulating nitrite and S-nitrosothiols by reductive chemiluminescence. J Chromatogr B. 2007;851:93–105. doi: 10.1016/j.jchromb.2006.12.012.
    1. Yang BK, Vivas EX, Reiter CD, Gladwin MT. Methodologies for the sensitive and specific measurement of S-nitrosothiols, iron-nitrosyls, and nitrite in biological samples. Free Radic Res. 2003;37:1–10. doi: 10.1080/1071576021000033112.
    1. Boxenbaum H, Battle M. Effective half-life in clinical pharmacology. J Clin Pharmacol. 1995;35:763–766. doi: 10.1002/j.1552-4604.1995.tb04117.x.
    1. Wright RO, Lewander WJ, Woolf AD. Methemoglobinemia: etiology, pharmacology, and clinical management. Ann Emerg Med. 1999;34:646–656. doi: 10.1016/S0196-0644(99)70167-8.
    1. Dejam A, Hunter CJ, Pelletier MM, Hsu LL, Machado RF, Shiva S, et al. Erythrocytes are the major intravascular storage sites of nitrite in human blood. Blood. 2005;106:734–739. doi: 10.1182/blood-2005-02-0567.
    1. Davies B, Morris T. Physiological parameters in laboratory animals and humans. Pharm Res. 1993;10:1093–1095. doi: 10.1023/A:1018943613122.
    1. Wagner DA, Schultz DS, Deen WM, Young VR, Tannenbaum SR. Metabolic fate of an oral dose of 15N-labeled nitrate in humans: effect of diet supplementation with ascorbic acid. Cancer Res. 1983;43:1921–1925.

Source: PubMed

3
Abonnieren