Elsevier

Journal of Chromatography B

Volumes 949–950, 15 February 2014, Pages 94-98
Journal of Chromatography B

Determination of 3-mercaptopyruvate in rabbit plasma by high performance liquid chromatography tandem mass spectrometry

https://doi.org/10.1016/j.jchromb.2014.01.006Get rights and content

Highlights

  • A method for the analysis of a cyanide antidote, 3-MP, was developed.

  • The method produced a linear range of 0.5–100 μM 3-MP in rabbit plasma.

  • The method was successful at detecting 3-MP from treated rabbits.

Abstract

Accidental or intentional cyanide poisoning is a serious health risk. The current suite of FDA approved antidotes, including hydroxocobalamin, sodium nitrite, and sodium thiosulfate is effective, but each antidote has specific major limitations, such as large effective dosage or delayed onset of action. Therefore, next generation cyanide antidotes are being investigated to mitigate these limitations. One such antidote, 3-mercaptopyruvate (3-MP), detoxifies cyanide by acting as a sulfur donor to convert cyanide into thiocyanate, a relatively nontoxic cyanide metabolite. An analytical method capable of detecting 3-MP in biological fluids is essential for the development of 3-MP as a potential antidote. Therefore, a high performance liquid chromatography tandem mass spectrometry (HPLC-MS-MS) method was established to analyze 3-MP from rabbit plasma. Sample preparation consisted of spiking the plasma with an internal standard (13C3-3-MP), precipitation of plasma proteins, and reaction with monobromobimane to inhibit the characteristic dimerization of 3-MP. The method produced a limit of detection of 0.1 μM, a linear dynamic range of 0.5–100 μM, along with excellent linearity (R2  0.999), accuracy (±9% of the nominal concentration) and precision (<7% relative standard deviation). The optimized HPLC-MS-MS method was capable of detecting 3-MP in rabbits that were administered sulfanegen, a prodrug of 3-MP, following cyanide exposure. Considering the excellent performance of this method, it will be utilized for further investigations of this promising cyanide antidote.

Introduction

Humans are exposed to cyanide (LD50, human = 1.1 mg/kg) [1], [2] in a variety of ways, such as ingestion of some edible plants (spinach or cassava), industrial operations, smoke inhalation from fires and/or cigarettes, and terrorist activities [3], [4]. Once cyanide is absorbed, it inhibits the enzyme cytochrome c oxidase in the electron transport system, thereby disrupting aerobic metabolism. There are currently three U.S. Food and Drug Administration (FDA) approved cyanide treatments: hydroxocobalamin, sodium nitrite, and sodium thiosulfate [2], [5], [6], [7].

Hydroxocobalamin (vitamin B12a) is a large molecular-weight cyanide antidote that detoxifies cyanide by sequestration. It forms a very strong bond with cyanide because of the high affinity of cyanide for the central cobalt atom (KA  1012 M−1) [8]. Cyanide binds to cobalt to produce cyanocobalamin (vitamin B12) [9], [10], [11], which resides in the plasma and is excreted in urine. The potential adverse effects of hydroxocobalamin are generally mild and include elevated blood pressure, decreased heart rate, rashes, and red coloring of the skin, tears, urine and sweat [12], [13]. The recommended dose of hydroxocobalamin is 5 g (administered over 15 min). Because of the high dose needed for optimum therapeutic effect, hydroxocobalamin must be administered intravenously [2], [14], limiting the applicability of hydroxocobalamin in mass casualty situations.

Similar to hydroxocobalamin, the mechanism of action of sodium nitrite is to sequester cyanide from cytochrome c oxidase. However, the sequestration of cyanide is indirect. Sodium nitrite causes the conversion of hemoglobin to methemoglobin, which has a high affinity towards cyanide [14], [15]. Recently, another mechanism of action of sodium nitrite was proposed as the prominent method of detoxification in which nitrite is converted to nitric oxide, which subsequently displaces cyanide bound to the active site of cytochrome c oxidase [16], [17]. Although sodium nitrite works well to detoxify cyanide, it is toxic at large concentrations [5], [18] and has a small therapeutic window. Sodium nitrite is especially toxic when smoke inhalation has occurred, due to the conversion of hemoglobin to methemoglobin, which reduces the oxygen carrying capacity of the blood [18]. Due to its limited therapeutic efficacy, sodium nitrite is typically administered in tandem with sodium thiosulfate.

Sodium thiosulfate detoxifies cyanide by donating a sulfur to convert cyanide to the much less toxic thiocyanate [18], [19], [20]. Therefore, sodium thiosulfate belongs to a class of cyanide therapeutics known as sulfur donors, which utilize sulfurtransferase enzymes as catalysts. Sodium thiosulfate utilizes rhodanese, which is mainly found in the liver and kidneys [14], [19], leaving the heart and central nervous system less protected and the main locations of cyanide toxicity [14]. It also has a slow onset of action, attributed to slow entry into cells and the mitochondria [5]. This necessitates its use in combination with faster acting therapeutics, typically sodium nitrite.

Considering that current cyanide antidotes each have major limitations, alternative cyanide antidotes are being investigated [5], [8], [21]. One such alternate antidote is 3-mercaptopyruvate (3-MP). Similar to sodium thiosulfate, 3-MP acts as a sulfur donor to produce thiocyanate but is instead catalyzed by 3-mercaptopyruvate sulfurtransferase (3-MST) [19], [21], [22]. Sulfanegen, a prodrug of 3-MP (i.e., sulfanegen converts to 3-MP upon administration), has been found to be highly effective in reversing cyanide toxicity [14], [20], [21], [23], [24]. Although a method for the detection of 3-MP by HPLC from mouse tissue has been proposed [25], a multistep, lengthy (>60 min), and high temperature (95 °C) modification of 3-MP is necessary. Therefore, the objective of this study was to develop a simple and sensitive analytical method for the analysis of 3-MP from rabbit plasma to facilitate further development of 3-MP as a cyanide antidote.

Section snippets

Reagents and standards

All solvents were LC–MS grade unless otherwise noted. Ammonium formate and 3-mercaptopyruvate (3-MP; HSCH2COCOOH) were purchased from Sigma–Aldrich (St. Louis, MO, USA). Acetone (HPLC grade, 99.5%) was purchased from Alfa Aesar (Ward Hill, MA, USA). Isotopically-labeled 3-MP (HS13CH213CO13COOH) was synthesized and provided by the Center for Drug Design, University of Minnesota (Minneapolis, MN, USA) [21]. Millex tetrafluoropolyethylene syringe filters (0.22 μm, 4 mm, Billerica, MA, USA) were

HPLC-MS-MS analysis of 3-MP from rabbit plasma

Under biological conditions, 3-MP is in rapid equilibrium with its dimer [30]. This equilibrium is difficult to control and results in poor chromatographic behavior. Because MBB reacts with the thiol group of 3-MP [26], [27], which is essential for dimerization, a single 3-MPB complex is created (Fig. 1), which produced excellent chromatographic behavior. Fig. 2 shows representative chromatograms of spiked and nonspiked 3-MPB in plasma, with 3-MPB eluting at approximately 2.75 min. The method

Conclusion

An HPLC-MS-MS method for the detection of 3-MP was developed which features simple sample preparation, excellent accuracy and precision, an excellent detection limit, and has a linear range of over 2 orders of magnitude. While Ogasawara et al. [25] reported an HPLC-fluorescence method for the analysis of 3-MP in mouse tissue, the method presented here featured simple and low-temperature sample preparation (i.e., 3-MP is highly unstable at high temperatures), rapid analysis, a reduced lower

Acknowledgements

We gratefully acknowledge the support from a U.S. Dept. of Education, Graduate Assistance in Areas of National Need (GAANN) award to the Department of Chemistry & Biochemistry (P200A100103). We thank the National Science Foundation Major Research Instrumentation Program (Grant Number CHE-0922816) for funding the AB SCIEX QTRAP 5500 LC-MS-MS. We also would like to acknowledge the support by the CounterACT Program, National Institutes of Health Office of the Director (NIH OD), and the National

References (31)

  • A.H. Hall et al.

    J. Emerg. Med.

    (1987)
  • H.A. Schwertner et al.

    J. Chromatogr. B

    (2012)
  • C. Brunel et al.

    Forensic Sci. Int.

    (2012)
  • M. Brenner et al.

    Toxicol. Appl. Pharm.

    (2010)
  • G.E. Isom et al.
  • A. Spallarossa et al.

    J. Mol. Biol.

    (2004)
  • Y. Ogasawara et al.

    J. Chromatogr. B

    (2013)
  • N.S. Kosower et al.
  • R.C. Fahey et al.
  • A.J.L. Cooper et al.

    J. Biol. Chem.

    (1982)
  • C.D. Barnes et al.

    Drug Dosages in Laboratory Animals–A Handbook

    (1973)
  • T.F. Cummings

    Occ. Med.

    (2004)
  • R.K. Bhandari et al.

    Anal. Bioanal. Chem.

    (2012)
  • B.A. Logue et al.

    Crit. Rev. Anal. Chem.

    (2010)
  • A.H. Hall et al.

    Crit. Rev. Toxicol.

    (2009)
  • Cited by (14)

    • Determination of free cyano-cobinamide in swine and rabbit plasma by liquid chromatography tandem mass spectrometry

      2019, Journal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences
    • Methionine restriction leads to hyperhomocysteinemia and alters hepatic H<inf>2</inf>S production capacity in Fischer-344 rats

      2018, Mechanisms of Ageing and Development
      Citation Excerpt :

      This enzyme produces H2S from 3-mercaptopyruvate (3-MP). Several studies reported that the endogenous level of 3-MP is very low and it is often below the quantitation level or not found at all (Ogasawara et al., 2013; Stutelberg et al., 2016, 2014). Therefore, we measured the activity of MPST in the liver to test for possible compensatory changes in response to MR. The activity of MPST was elevated in MR animals compared to controls at both 4 and 8 weeks (Fig. 6).

    View all citing articles on Scopus
    View full text