LC-MS in Drug Analysis - Methods and Protocols-Humana Press
LC-MS in Drug Analysis - Methods and Protocols-Humana Press
Loralie J. Langman
Christine L.H. Snozek
Editors
LC-MS in Drug
Analysis
Methods and Protocols
Second Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Second Edition
Edited by
Loralie J. Langman
Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC, part of
Springer Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
The second edition of this book is again intended to provide detailed LC-MS(/MS)
procedures for the analysis of compounds of clinical significance. The main focus points
for this edition were new developments including novel drugs (both therapeutic and
recreational) and updated methodologies, as well as discussing alternate matrices not
addressed in the first edition.
We thank our colleagues who contributed to the contents of the book for the countless
hours of work that these chapters represent. We hope that you, the reader, find this book
useful.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 An Introduction to Drug Testing: The Expanding Role
of Mass Spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Christine L.H. Snozek, Loralie J. Langman, and Steven W. Cotten
2 Quantification of Eight Cannabinoids Including Cannabidiol
in Human Urine Via Liquid Chromatography Tandem Mass Spectrometry . . . . 11
Karl B. Scheidweiler and Allan J. Barnes
3 Analysis of Benzodiazepines for Drug-Facilitated Assaults
and Abuse Settings (Urine) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Olaf H. Drummer, Matthew Di Rago, and Dimitri Gerostamoulos
4 Targeted Opioid Screening Assay for Pain Management Using
High-Resolution Mass Spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
Darlington Danso, Loralie J. Langman, and Paul J. Jannetto
5 Measurement of Buprenorphine and Norbuprenorphine in Urine. . . . . . . . . . . . . 51
Andrea R. Terrell, Vipin Adhlakha, and Poluru Reddy
6 Quantitation of Tapentadol by Liquid Chromatography: Tandem
Mass Spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
Graham R. Jones and Russell P. Handy
7 Therapeutic Drug Monitoring of Lacosamide by LC-MS/MS . . . . . . . . . . . . . . . . 67
He Sarina Yang and Leslie Edinboro
8 LC-MS/MS Method for the Quantification of the Leflunomide
Metabolite, Teriflunomide, in Human Serum/Plasma . . . . . . . . . . . . . . . . . . . . . . . 75
Geoffrey S. Rule, Alan L. Rockwood, and Kamisha L. Johnson-Davis
9 Analysis of Tryptic Peptides from Therapeutic Monoclonal Antibodies
Using LC-MS/MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Maria Alice V. Willrich
10 Quantification of Methotrexate in Human Serum and Plasma
by Liquid Chromatography Tandem Mass Spectrometry. . . . . . . . . . . . . . . . . . . . . 101
Gabrielle N. Winston-McPherson, Michael Schmeling,
and Andrew N. Hoofnagle
11 Simultaneous Determination of Tacrolimus and Cyclosporine A
in Whole Blood by Ultrafast LC-MS/MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
Matthew W. Bjergum, Paul J. Jannetto, and Loralie J. Langman
12 Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)
Method to Quantify Gabapentin and Pregabalin in Urine. . . . . . . . . . . . . . . . . . . . 119
Stephen Merrigan and Kamisha L. Johnson-Davis
13 The Evolving Landscape of Designer Drugs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Sherri L. Kacinko and Donna M. Papsun
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 273
Contributors
ix
x Contributors
Abstract
Measurement of drugs and their metabolites in biological fluids is the foundation of both therapeutic drug
monitoring (TDM) and toxicology. The introduction of methods based on mass spectrometry (MS),
coupled with gas or liquid chromatography, has revolutionized these areas. This chapter will introduce
the reader to the application of MS to TDM and toxicology, the steps that should be considered during
implementation and the processes that should be implemented to assure continued quality. Points of
emphasis include advances and recent trends since the publication of the first edition of this book, such
as high-resolution mass spectrometry and increased interest in alternate matrices.
Key words Mass spectrometry, Gas chromatography, Liquid chromatography, Therapeutic drug
monitoring, Toxicology, Drug testing
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_1,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
1
2 Christine L.H. Snozek et al.
Gas Chromatography
Liquid Chromatography
Mass Library
Sample Preparation Separation Ionization Detection
Analyzer(s) Scanning/Identification
3 Toxicology
For many years, GC-MS was considered the gold standard for
confirmation of the presence of abused drugs in urine, but as
methods and libraries have developed, many laboratories have
turned to LC-MS/MS for these analyses. In the instance of pain
management, screening and confirmatory drug testing using
LC-MS/MS allow for detection of both morphine-based and syn-
thetic opioids. The technique provides superior sensitivity and
specificity compared to immunoassays which are usually targeted
to morphine and may thus fail to detect synthetic opioids and
oxycodone. The ability to detect multiple parent drugs and meta-
bolites in a relatively short run makes LC-MS/MS a powerful tool
in support of pain management testing. The use of this technique
also provides an extended analytical measuring range of approxi-
mately 105 ng/mL that is most useful in this setting [13].
Matrices other than blood and urine are an increasingly impor-
tant component of clinical and forensic toxicology. Meconium and
umbilical cord tissue can reveal in utero exposure to illicit drugs.
Oral fluid, hair, and dried blood spots all provide certain advantages
over traditional analytical matrices. For example, oral fluid testing
can allow for observed collection of a specimen during a roadside
stop from a seemingly intoxicated driver, while dried blood spots
can facilitate testing collected in remote locations far from a refer-
ence laboratory.
4 Evaluation of Methods
Exogenous Endogenous
immunosuppressants hormones
neurological agents vitamin D
antiviral agents organic acids
Therapeutic acylcarnitines
Drug
Monitoring
Mass
Spectrometry
D.O.A Forensics poisons
opiates
Confirmatory and toxins
steroids Drug Testing Toxicological D.O.A
Analysis
5 Quality Assurance
Table 1
CLSI documents relevant to mass spectrometry analysis
6 Conclusions
References
1. CLSI (2010) Gas chromatography/mass spec- 8. Salm P, Taylor PJ, Rooney F (2008) A high-
trometry confirmation of drugs; approved performance liquid chromatography-mass
guideline, 2nd edn. CLSI Document C43-A2. spectrometry method using a novel atmo-
Clinical and Laboratory Standards Institute, spheric pressure chemical ionization approach
Wayne, PA for the rapid simultaneous measurement of
2. CLSI (2007) Mass spectrometry in the clinical tacrolimus and cyclosporin in whole blood.
laboratory: general principles and guidance; Ther Drug Monit 30:292–300
approved guideline. CLSI Document C50-A. 9. Koster RA, Dijkers ECF, Uges DRA (2009)
Clinical and Laboratory Standards Institute, Robust, high-throughput LC-MS/MS
Wayne, PA method for therapeutic drug monitoring of
3. Kirchherr H, Kühn-Velten WN (2006) Quan- cyclosporine, tacrolimus, everolimus, and siro-
titative determination of forty-eight antide- limus in whole blood. Ther Drug Monit
pressants and antipsychotics in human serum 31:116–125
by HPLC tandem mass spectrometry: a multi- 10. Bogusz MJ et al (2007) Simultaneous LC-MS-
level, single-sample approach. J Chromatogr B MS determination of cyclosporine a, tacroli-
Analyt Technol Biomed Life Sci 843:100–113 mus, and sirolimus in whole blood as well as
4. Zhang Y, Mehrotra N, Budha NR, Christensen mycophenolic acid in plasma using common
ML, Meibohm B (2008) A tandem mass spec- pretreatment procedure. J Chromatogr B Ana-
trometry assay for the simultaneous determina- lyt Technol Biomed Life Sci 850:471–480
tion of acetaminophen, caffeine, phenytoin, 11. Vande Casteele N et al (2014) Therapeutic
ranitidine, and theophylline in small volume drug monitoring in inflammatory bowel dis-
pediatric plasma specimens. Clin Chim Acta ease: current state and future perspectives.
398:105–112 Curr Gastroenterol Rep 16:378
5. Sørensen LK (2010) Determination of acidic 12. Wu AH, Gerona R, Armenian P, French D,
and neutral therapeutic drugs in human blood Petrie M, Lynch KL (2012) Role of liquid
by liquid chromatography-electrospray tandem chromatography-high-resolution mass spec-
mass spectrometry. Forensic Sci Int. https:// trometry (LC-HR/MS) in clinical toxicology.
doi.org/10.1016/j.forsciint.2010.07.016 Clin Toxicol (Phila) 50:733–742
6. Subramanian M, Birnbaum AK, Remmel RP 13. Mikel C, Almazan P, West R, Crews B et al
(2008) High-speed simultaneous determina- (2009) LC-MS/MS extends the range of
tion of nine antiepileptic drugs using liquid drug analysis in pain patients. Ther Drug
chromatography-mass spectrometry. Ther Monit 31:746–748
Drug Monit 30:347–356 14. Centers for Medicare & Medicaid Services
7. Nair H, Lawrence L, Hoofnagle AN (2012) (2003) Interpretive guidelines for laboratories.
Liquid chromatography-tandem mass spec- http://www.cms.gov/CLIA/03_Interpretive_
trometry work flow for parallel quantification Guidelines_for_Laboratories.asp. Accessed
of methotrexate and other immunosuppres- 20 Nov 2017
sants. Clin Chem 58:943–945
10 Christine L.H. Snozek et al.
15. Committee on Identifying the Needs of the quality management system in drug testing
Forensic Sciences Community, National laboratories. http://www.unodc.org/
Research Council (2009) Strengthening foren- documents/scientific/QMS_Ebook.pdf.
sic science in the United States: a path forward. Accessed 20 Nov 2017
The National Academies Press, Washington, 18. United States Department of Justice Drug
D.C. Enforcement Administration (2010) Scientific
16. Kaye DH (2010) The good, the bad, the ugly: working group for the analysis of seized drugs
the NAS report on strengthening forensic sci- recommendations. http://www.swgdrug.org/
ence in America. Sci Justice 50:8–11 Documents/SWGDRUG%
17. United Nations Office on Drugs and Crime 20Recommendations%20Version%207-1.pdf.
(2009) Guidance for the implementation of a Accessed 20 Nov 2017
Chapter 2
Abstract
Medical and recreational cannabis legalization has highlighted the importance of being able to identify
recent cannabis use and impairment. Monitoring minor plant cannabinoids has been proposed to assist in
identifying recent cannabis use. Additionally, cannabidiol (CBD) has been proposed for epilepsy, pain,
inflammatory disorder, anxiety, and addiction treatment; therefore, monitoring CBD is of increasing
clinical importance. However, few methods exist capable of monitoring extensive panels of traditional
cannabinoid analytes and minor cannabinoids (including CBD). This chapter details a liquid chromatogra-
phy tandem mass spectrometry method capable of measuring Δ9-tetrahydrocannabinol (THC),
11-hydroxy-THC, 11-nor-9-carboxy-THC, cannabinol, cannabigerol, tetrahydrocannabivarin (THCV),
and its metabolite, 11-nor-9-carboxy-THCV, in urine.
Key words Cannabinoids, Cannabidiol, Urine, Liquid chromatography, Mass spectrometry, Pipette
tip extraction
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_2,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
11
12 Karl B. Scheidweiler and Allan J. Barnes
2 Materials
Table 1
Final concentrations (μg/L) for calibrators in urine
Calibrator 1 2 3 4 5 6 7 8 9
THC 1 2 5 10 20 50 100 250 –
CBG 1 2 5 10 20 50 100 250 –
11-OH-THC – 2 5 10 20 50 100 250 –
CBD – 2 5 10 20 50 100 250 –
CBN – 2 5 10 20 50 100 250 –
THCV – 2 5 10 20 50 100 250 –
THCVCOOH – 2 5 10 20 50 100 250 –
THCCOOH 1 2 5 10 20 50 100 250 500
3 Methods
4 Notes
Table 2
LC-MS/MS parameters for cannabinoids, their metabolites, and internal standards in human
hydrolyzed urine
Analyte Q1 (amu) Q3 (amu) DP (V) CE (eV) CXP (V) MRM period Dwell time (msec)
THC 315.0 193.1 91 31 20 4 58
315.0 123.1 91 43 14 4 58
11-OH-THC 331.0 201.0 31 33 18 2 58
331.0 313.2 31 19 30 2 58
THCCOOH 345.0 193.0 61 35 14 2 58
345.0 327.2 61 21 28 2 58
CBD 315.0 193.0 51 29 18 3 58
315.0 123.0 51 41 14 3 58
CBN 311.0 223.0 51 29 18 4 58
311.0 241.1 51 25 24 4 58
CBG 317.0 193.2 36 21 14 3 58
317.0 122.8 36 41 18 3 58
THCV 287.1 165.0 26 29 14 3 58
287.1 123.0 26 41 10 3 58
THCVCOOH 317.0 165.1 31 35 14 1 120
317.0 271.1 31 25 20 1 120
d3-THC 318.1 196.1 76 31 20 4 58
318.1 123.0 76 43 14 4 58
d3-11-OH-THC 334.1 201.0 26 35 16 2 58
334.1 316.0 26 19 30 2 58
d9-THCCOOH 354.1 196.1 56 35 18 2 58
354.1 335.8 56 23 28 2 58
d3-CBD 318.1 195.9 26 27 16 3 58
318.1 135.2 26 25 10 3 58
d3-CBN 314.0 223.1 51 29 18 4 58
314.0 241.0 51 25 22 4 58
THC delta9-tetrahydrocannabinol, 11-OH-THC 11-hydroxy-THC, THCCOOH 11-nor-9-carboxy-THC, CBD canna-
bidiol, CBN cannabinol, CBG cannabigerol, THCV delta9-tetrahydrocannabivarin, THCVCOOH 11-nor-9-carboxy-
THCV
Bold masses depicted quantifier transitions. DP declustering potential, CE collision energy, CXP collision cell exit
potential, MRM scheduled multiple reaction monitoring
20 Karl B. Scheidweiler and Allan J. Barnes
Fig. 1 Extracted ion chromatogram showing quantifier MRM transitions after injecting an extract prepared
from 200 μL fortified urine containing each analyte at 3 μg/L. Δ9-Tetrahydrocannabinol (THC: m/z 315–193),
11-hydroxy-THC (11-OH-THC: m/z 331–201), 11-nor-9-carboxy-THC (THCCOOH: m/z 345–193), cannabinol
(CBN: m/z 311–223), cannabigerol (CBG: m/z 317–193), tetrahydrocannabivarin (THCV: m/z 287–165), and its
metabolite, 11-nor-9-carboxy-THCV (THCVCOOH: m/z 317–165)
Acknowledgments
References
1. Hall W, Kozlowski LT (2017) The diverging 7. Hidvegi E, Somogyi GP (2010) Detection of
trajectories of cannabis and tobacco policies in cannabigerol and its presumptive metabolite in
the United States: reasons and possible impli- human urine after Cannabis consumption.
cations. Addiction. https://doi.org/10.1111/ Pharmazie 65(6):408–411
add.13845 8. Kemp PM, Abukhalaf IK, Manno JE, Manno
2. Smith A (2017) 10 things to know about legal BR, Alford DD, Abusada GA (1995) Cannabi-
pot. http://money.cnn.com/2017/04/19/ noids in humans. I. Analysis of delta
news/legal-marijuana-420/index.html. 9-tetrahydrocannabinol and six metabolites in
Accessed 1 Aug 2017 plasma and urine using GC-MS. J Anal Toxicol
3. Bergamaschi MM, Karschner EL, Goodwin 19(5):285–291
RS, Scheidweiler KB, Hirvonen J, Queiroz 9. Campbell CT, Phillips MS, Manasco K (2017)
RH, Huestis MA (2013) Impact of prolonged Cannabinoids in pediatrics. J Pediatr Pharma-
cannabinoid excretion in chronic daily cannabis col Ther 22(3):176–185. https://doi.org/10.
smokers’ blood on per se drugged driving laws. 5863/1551-6776-22.3.176
Clin Chem 59(3):519–526. https://doi.org/ 10. Blessing EM, Steenkamp MM, Manzanares J,
10.1373/clinchem.2012.195503 Marmar CR (2015) Cannabidiol as a potential
4. Aizpurua-Olaizola O, Zarandona I, Ortiz L, treatment for anxiety disorders. Neurothera-
Navarro P, Etxebarria N, Usobiaga A (2017) peutics 12(4):825–836. https://doi.org/10.
Simultaneous quantification of major cannabi- 1007/s13311-015-0387-1
noids and metabolites in human urine and 11. Sloan ME, Gowin JL, Ramchandani VA, Hurd
plasma by HPLC-MS/MS and enzyme-alkaline YL, Le Foll B (2017) The endocannabinoid
hydrolysis. Drug Test Anal 9(4):626–633. system as a target for addiction treatment: trials
https://doi.org/10.1002/dta.1998 and tribulations. Neuropharmacology doi:
5. Bergamaschi MM, Barnes A, Queiroz RH, https://doi.org/10.1016/j.neuropharm.
Hurd YL, Huestis MA (2013) Impact of enzy- 2017.05.031
matic and alkaline hydrolysis on CBD concen- 12. Abraham TT, Lowe RH, Pirnay SO, Darwin
tration in urine. Anal Bioanal Chem 405 WD, Huestis MA (2007) Simultaneous GC-
(14):4679–4689. https://doi.org/10.1007/ EI-MS determination of Delta9-
s00216-013-6837-x tetrahydrocannabinol, 11-hydroxy-Delta9-
6. ElSohly MA, deWit H, Wachtel SR, Feng S, tetrahydrocannabinol, and 11-nor-9-carboxy-
Murphy TP (2001) Delta9- Delta9-tetrahydrocannabinol in human urine
tetrahydrocannabivarin as a marker for the following tandem enzyme-alkaline hydrolysis.
ingestion of marijuana versus Marinol: results J Anal Toxicol 31(8):477–485
of a clinical study. J Anal Toxicol 25 13. Andersson M, Scheidweiler KB, Sempio C,
(7):565–571 Barnes AJ, Huestis MA (2016) Simultaneous
22 Karl B. Scheidweiler and Allan J. Barnes
Abstract
An overview of the detection of benzodiazepines and their respective metabolites and target analytes in
urine by LC-MS/MS is described. This overview shows substantial differences in the approach to detection
using this technique including optional use of β-glucuronidase to hydrolyze conjugates present in urine.
There are also significant variations in the extraction method employed from the use of direct injection,
liquid-liquid extraction to solid-phase extraction options, with little apparent difference in limits of
detection. Chromatography was largely based on the use of C18-bonded columns; however both C8-
and phenyl-bonded columns were used to affect separation. Modern-day tandem mass spectrometers are
capable of exceptional sensitivity enabling detection of sub-nanogram per milliliter amounts in urine, which
provide for longer detection times in the urine of suspected drug-facilitated assaults. A method employed in
the laboratory of the authors is provided by way of an example for readers wishing to establish a method in
their own laboratory.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_3,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
23
24 Olaf H. Drummer et al.
1.2 Overview There have been numerous methods published using LC-MS/MS
of Published Methods to determine the presence of benzodiazepines in urine, particularly
over the last 10 years. A selection of those specializing in drugs used
in assault cases, and in particular benzodiazepines and/or their
Z-drug analogs, and relevant metabolite analytes (n ¼ 20) are
summarized in Table 2 [3–22]. The list is restricted to references
published over the last 10 years.
The methods vary substantially in their approach; most (15 of
20) use some form of hydrolysis to liberate conjugated benzodia-
zepines from their respective glucuronides, although there is sig-
nificant variation in the source of enzyme used and the hydrolysis
conditions. The optimization of these hydrolysis conditions is
rarely reported, let alone for all of the known conjugates.
LC-MS/MS used in these publications represented all of the
major instrument manufacturers and will vary substantially in sen-
sitivity due to the configuration and design of the mass analyzers.
Many of the more recent analyzers show much higher sensitivity
than the older LC-MS/MS instruments. Almost all LC-MS/MS
Benzodiazepines in Urine 25
Table 1
Selection of benzodiazepines and related drugs, their key metabolites, and target analytes in urine
Table 2
Analysis parameters for selected publications describing tandem LC-MS detection of benzodiazepines in urine
ElSohly et al. β-Glucuronidase/sulfatase LLE ChCl3/isopropanol (9:1) Luna C8 (4.6 100 mm, 3 μm), Agilent 1100 SL TOF-MS ESI
[3] (4 h, 37 C) 0.1% acetic acid/ACN 22 BZ/metabolites
LOD 0.5–3 ng/mL
Glover and β-Glucuronidase LLE DCM/DCE/heptane/ HyPURITY C8 (3 150 mm), 1% API 3000 TQ API
Allen [4] (PV) (0.5 h, 56 C) isopropanol HCOOH, 1% isopropanol ACN 6 BZ
gradient LOD 2.5–5 ng/mL
Guale et al. None SPE Strata-X-Drug B Eclipse Plus C18 (3 100 mm, Agilent 6230 TOF-MS,
[5] 1.8 μm), 5 mM NH4COO, 5–60% 15 BZ/metabolites,
MeOH gradient LOD not given
Hegsted β-Glucuronidase (HP-2) Direct injection ACQUITY HSS T3 (2.1 100 mm, Waters Xevo TQ-S ESI
et al. [22] (1 h, 65 C) and without 1.8 μm), 0.1% HCOOH/MeOH, 5 BZ
50 C LOD 2–6 ng/mL
Jagerdeo and β-Glucuronidase SPE Isolute SLE, Cortex C18 (2.1 50 mm, 1.6 μm), Waters Orbitrap
Schaff [6] (RA) (0.5 h, 68 C) DCM/isopropanol (95:5) 0.1% HCOOH/ACN 22 BZ/metabolites,
LOD 1 ng/mL
Jeong et al. None Direct injection Zorbax SB-C18 (2.1 100 mm, API 3200 TQ ESI
[7] 3.5 μm), 2 mM trifluoroacetate/ 18 BZ
0.2% acetic acid ACN gradient LOD 0.3–3 ng/mL
Karampela None Direct injection XTerra MS C8 (2.1 250 mm, Shimadzu 2010EV ESI LC-MS,
et al. [8] 5 μm), 0.05% HCOOH/ACN 10 BZ
gradient LOD >10 ng/mL
Lee et al. [9] None Ethyl acetate LLE ACE5 C18 (4.6 250 mm, 5 μm), API 4000 Q-Trap ESI
0.1% HCOOH/ACN gradient 21 BZ and others
LOD >0.5 ng/mL
Mata et al. β-Glucuronidase (HP-2) DPX WAX tips ACQUITY BEH C18 Waters Xevo TQ-S, ESI
[18] (3 h, 55 C) (2.1 100 mm, 1.7 μm), water 17 BZ,
0.1% HCOOH/ACN 0.1% LOD 0.5–12.5 ng/mL
HCOOH
Marin et al. β-Glucuronidase SPE Trace-B, ethyl acetate: NH3 XTerra MS C18 (2.1 150 mm, Waters, Micromass, Alliance,
[10] (BL) (2 h, 60 C) (98:2) 3.5 μm), 100 mM HCOOH/ Quattro ESI
ACN gradient 13 BZ/metabolites
LOD <10 ng/mL
Ming and β-Glucuronidase Direct injection ACQUITY BEH C18 Waters TQD ESI,
Heathcote (EC) (1.5 h 60 C) (2.1 50 mm, 1.7 μm), 0.2% 13 BZ/metabolites LOD
[11] HCOOH/MeOH gradient, 45 C 0.5–2 ng/mL
Perez et al. β-Glucuronidase (HP-2) SPE UCT Clean Screen XCEL I, ACQUITY BEH C18 (1.7 μm, Waters Quattro micro, ESI
[19] (1 h, 55 C) ethyl acetate: NH4OH (100:2) 2.1 50 mm), (A) 0.1% HCOOH 5 BZ, LOD 2–16 ng/mL
and (B) acetonitrile
Petterson β-Glucuronidase (10 min, Direct injection ACQUITY BEH phenyl column Waters Xevo TQ ESI
Bergstrand 25 C) (1.0 50 mm, 1.7 μm), 11 designer BZ
et al. [12] ACN/0.1% HCOOH gradient LOD 1–10 ng/mL
Remane et al. β-Glucuronidase Direct injection Zorbax Eclipse XDB C18 API 4000 Q-Trap ESI
[13] (EC) (0.5 h, 55 C) and (4.6 100 mm, 5 μm), 50 mM 24 BZ/metabolites plus others
without HCOOH LOD meet SOFT criteria
Rosano et al. β-Glucuronidase (1 h, Direct injection ACQUITY BEH Phenyl Waters Xevo TQD, ESI
[20] 55 C) (2.1 50 mm, 1.7 μm), 2 mM 16 BZ,
NH4COO/2 mM NH4COO, LOD 25 ng/mL
0.1% HCOOH in ACN
Salomone β-Glucuronidase LLE pH 7.5, DCM/propan-2-ol Eclipse XDB C18 (4.6 50 mm, API 3200 triple Q-Trap,
Benzodiazepines in Urine
et al. [14] (HP) (1 h, 55 C) (85:15) 1.8 μm), MeOH/water gradient 23 BZ/analogs, LOD
0.5–30 ng/mL
(continued)
27
28
Table 2
(continued)
Tang et al. β-Glucuronidase (A) (1 h, SPE Waters Oasis MCX (combined Eclipse C8 (3 100 mm, 1.8 μm), Agilent TQ ESI
[21] 55 C) acidic, basic) 5 mM NH4COO, 0.1% HCOOH, 14 BZ
MeOH LOD 2–100 ng/mL
Verplaetse β-Glucuronidase Bond Elut Plexa PCX SPE, elution ACQUITY C18 (2.1 50 mm, API 3200 Q-Trap, ESI,
et al. [16] (PV) (3 h, 65 C) acetone/CHCl3 (1:1), and 1.7 μm), 10 mM NH4HCO3 29 BZ/metabolites
ammoniated ethyl acetate MeOH gradient LOD >0.02 ng/mL
Xiong et al. None Online SPE (Oasis HLB) with XTerra MS C18 (2.1 150 mm, Waters Quattro Premier XE ESI,
[17] formic acid 3.5 μm), ACN/MeOH/water 8 BZ and glucuronidated
metabolites
LOD from 0.2 ng/mL
Benzodiazepines in Urine 29
2 Materials
2.1 Solutions 1. Borate buffer, pH 9.8: Dissolve 0.775 g of boric acid and 4.1 g
and Reagents of sodium tetraborate in 250 mL of water. Adjust to pH 9.8
(See Note 5) using 1 M sodium hydroxide.
2. 1.1 M sodium acetate buffer, pH 4.5: Dissolve 22.56 g of
anhydrous sodium acetate in 250 mL of water. Adjust to pH
4.5 with glacial acetic acid.
3. 1 M sodium hydroxide (NaOH): Add 4 g of sodium hydroxide
to 100 mL of water. Mix well.
4. β-Glucuronidase: Commercial enzyme sourced from red aba-
lone (Haliotis rufescens) 7000 units (U) per mL or 14,000 U
per sample.
5. Extraction solvent: Dichloromethane/isopropanol/ethyl ace-
tate, 1:1:3 v/v/v. Add 100 mL each of dichloromethane and
isopropanol to 300 mL of ethyl acetate, and mix gently.
6. Stock standard: 1 mg/mL each drug or metabolite. Dilute
2.00 mg of each benzodiazepine or metabolite in 2.00 mL
methanol. Stock standards can be kept for at least 3 months
stored at 20 C.
7. Working standard 1 (WS1): Dilute 0.1 mL of each stock stan-
dard to 1 mL with methanol (see Note 6).
8. Working standard 2 (WS2): Dilute 0.05 mL of each stock
standard to 5 mL with water.
9. Working standard 3 (WS3): Dilute 0.05 mL of each stock
standard to 50 mL with water.
10. Mobile phase A: 50 mM ammonium formate pH 3.5. Add
3.15 g of ammonium formate to 1 L of water. Adjust pH to
3.5 with concentrated formic acid.
11. Mobile phase B: 0.1% formic acid in acetonitrile. Add 1 mL of
formic acid to 1 L of acetonitrile.
3 Methods
3.1 Sample Analysis 1. Obtain drug-free urine (with consent) from a volunteer, and
use this to prepare a series of calibration standards for one or
more of the benzodiazepine and/or metabolites (see Note 7).
Typically a range of concentrations are prepared at or above the
limit of detection to a concentration within the range of the
spectrometer and often seen in samples. The typical calibration
ranges for some of the more common analytes are listed in
Table 3 (see Note 8).
Table 3
Multiple-reaction mode transitions and typical calibration range for selected benzodiazepines,
metabolites, and related analogs
(continued)
32 Olaf H. Drummer et al.
Table 3
(continued)
Shake on
2mL Urine
reciprocating Centrifuge 3000
Sample, QC or
shaker 5 min, rpm, 5 min
Calibrator
5000 rpm
Add 14000 IU
Abalone Add 6mL Transfer solvent
β-glucuronidase extraction solvent layer to glass vial
enzyme
Adjust sample
Add 0.5 mL 1.1M Evaporate to Inject 3uL into
pH to 8.5 with
sodium acetate dryness under N2 LC-MS/MS
1M sodium
buffer gas @ 40˚C system
hydroxide
Add 1mL 1M
Incubate Reconstitute with Transfer to
borate buffer pH
overnight @ 55˚C 0.05mL methanol autosampler vial
9.8
4 Notes
Fig. 2 (a) Calibration standard in urine containing benzodiazepines and some metabolites at 0.25 mg/L, except
for flurazepam, nordiazepam, and temazepam which are at 0.1 mg/L. (b) Chromatogram of a urine from a
DFSA case containing alprazolam (0.01 mg/L) and its 1-hydroxy metabolite (0.03 mg/L) and diazepam with
three of its metabolites, nordiazepam (0.2 mg/L), oxazepam (0.02 mg/L), and temazepam (0.2 mg/L)
36 Olaf H. Drummer et al.
Acknowledgments
References
15. Schaefer N, Peters B, Schmidt P, Ewald AH 93 conventional and emerging drugs of abuse
Development and validation of two LC-MS/ and their metabolites in urine by UHPLC-
MS methods for the detection and quantifica- MS/MS. J Chromatogr B Analyt Technol
tion of amphetamines, designer amphetamines, Biomed Life Sci 969:272–284
benzoylecgonine, benzodiazepines, opiates, 22. Hegstad S, Hermansson S, Betner I, Spigset O,
and opioids in urine using turbulent flow chro- Falch BM (2014) Screening and quantitative
matography. Anal Bioanal Chem 405 determination of drugs of abuse in diluted
(1):247–258 urine by UPLC-MS/MS. J Chromatogr B
16. Verplaetse R, Cuypers E, Tytgat J (2012) The Analyt Technol Biomed Life Sci
evaluation of the applicability of a high pH 947-948:83–95
mobile phase in ultrahigh performance liquid 23. Hoiseth G, Tuv SS, Karinen R (2016) Blood
chromatography tandem mass spectrometry concentrations of new designer benzodiaze-
analysis of benzodiazepines and pines in forensic cases. Forensic Sci Int
benzodiazepine-like hypnotics in urine and 268:35–38
blood. J Chromatogr A 1249:147–154 24. Al-Saffar Y, Stephanson NN, Beck O (2013)
17. Xiong L, Wang R, Liang C et al (2015) Deter- Multicomponent LC-MS/MS screening
mination of co-administrated opioids and ben- method for detection of new psychoactive
zodiazepines in urine using column-switching drugs, legal highs, in urine-experience from
solid-phase extraction and liquid chromatogra- the Swedish population. J Chromatogr B Ana-
phy–tandem mass spectrometry. J Chromatogr lyt Technol Biomed Life Sci 930:112–120
A 1395:99–108 25. Sturm S, Hammann F, Drewe J, Maurer HH,
18. Mata DC, Davis JF, Figueroa AK, Stanford MJ Scholer A (2010) An automated screening
(2016) Ultra performance liquid chromatogra- method for drugs and toxic compounds in
phy with tandem mass spectrometry for the human serum and urine using liquid
quantitation of seventeen sedative hypnotics chromatography-tandem mass spectrometry. J
in six common toxicological matrices. J Anal Chromatogr B Analyt Technol Biomed Life Sci
Toxicol 40(1):58–63 878(28):2726–2732
19. Perez ER, Knapp JA, Horn CK, Stillman SL, 26. Sundström M, Pelander A, Ojanpera I (2017)
Evans JE, Arfsten DP (2016) Comparison of Comparison of post-targeted and pre-targeted
LC-MS-MS and GC-MS analysis of benzodiaz- urine drug screening by UHPLC-HR-
epine compounds included in the drug demand QTOFMS. J Anal Toxicol
reduction urinalysis program. J Anal Toxicol 40 27. Sargent M (ed) (2013) Guide to achieving
(3):201–207 relaible quantitative LC-MS measurements,
20. Rosano TG, Ohouo PY, Wood M (2017) 1st edn. RSC Analytical Standards Committee,
Screening with quantification for 64 drugs Cambridge
and metabolites in human urine using UPLC- 28. SWGTOX 2013 Scientific working group for
MS-MS analysis and a threshold accurate cali- forensic toxicology (SWGTOX) standard prac-
bration. J Anal Toxicol:1–11 tices for method validation in forensic toxicol-
21. Tang MH, Ching CK, Lee CY, Lam YH, Mak ogy 20 May
TW (2014) Simultaneous detection of
Chapter 4
Abstract
The use and adherence monitoring of opioids in pain management is recommended by numerous clinical
practice guidelines. Many physicians use urine immunoassay screening tests, which suffer from a lack of
sensitivity and specificity, to verify compliance to pain medications. However, several immunoassay tests are
required to comprehensively detect the synthetic, semisynthetic, and natural opioids due to the limited
cross-reactivity of each assay. Superior testing strategies are required to specifically identify low concentra-
tions of opioids found in adherent pain management patients. Therefore we present a method for the
qualitative identification of 33 opioids and metabolites (codeine, codeine-6-β-glucuronide, morphine,
morphine-6-β-glucuronide, 6-acetylmorphine, hydrocodone, norhydrocodone, dihydrocodeine, hydro-
morphone, hydromorphone-3-β-glucuronide, oxycodone, noroxycodone, oxymorphone, oxymorphone-
3-β-glucuronide, noroxymorphone, meperidine, normeperidine, methadone, EDDP, propoxyphene, nor-
propoxyphene, tramadol, O-desmethyltramadol, tapentadol, tapentadol-β-glucuronide, N-desmethylta-
pentadol, buprenorphine, norbuprenorphine, norbuprenorphine glucuronide, naloxone, naloxone
glucuronide, fentanyl, and norfentanyl) in unhydrolyzed urine using a liquid chromatography tandem
mass spectrometry (LC-MS/MS) with high-resolution, accurate-mass Orbitrap detection.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_4,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
41
42 Darlington Danso et al.
for abuse and diversion which is one of the main reasons many
professional practice guidelines recommend compliance testing in
patients prescribed these medications.
Opioids are readily absorbed from the gastrointestinal tract,
nasal mucosa, and lungs and after subcutaneous or intramuscular
injection. Opioids are primarily excreted from the kidney in both
free and conjugated forms. This assay does not hydrolyze the urine
sample and therefore allows detection of both parent drugs and
metabolites (including glucuronide forms). The detection window
for most opioids in urine is approximately 1–3 days with longer
detection times for some compounds (e.g., methadone). We
describe a liquid chromatography tandem mass spectrometry
(LC-MS/MS) method with high-resolution, accurate-mass Orbi-
trap detection to qualitatively (present vs. not detected) identify
33 opioid compounds (parent drugs and metabolites) in urine to
help determine compliance and/or identify illicit opioid drug use.
The method is a simple dilute-and-shoot in which urine samples are
diluted (1:10) with internal standard (IS) in clinical laboratory
reagent water (CLRW) and then subjected to chromatographic
separation on a Restek Ultra Biphenyl column and analyzed on
the Orbitrap with heated electrospray ionization in positive mode.
The mass spectrometry method is full MS followed by data-
dependent MS2. This data-dependent experiment collects full
scan data and MS/MS spectra for all compounds defined in the
inclusion list. A step gradient elution of the Ultra Biphenyl column
achieves a complete chromatographic separation of isobaric com-
pounds with a total run time of 8 min. Compound identification
combines exact mass (<5 ppm), delta retention time, and MS2
spectral library match.
2 Materials
2.1 Samples and 1. Collect urine samples under proper supervision to ensure spec-
Reagents imen integrity and to avoid specimen manipulation that might
cause false negative results (see Note 1).
2. 0.5% formic acid in CLRW. Add 5 mL of formic acid to a 1 L
reagent bottle. Bring to volume with CLRW and mix thor-
oughly. Stable for 1 month at 20–27 C.
3. Negative (drug-free) urine. Obtain drug-free urine commer-
cially (e.g., UTAK) or from volunteers. Verify as drug-free by
assaying prior to use in this method.
4. Wash solution # 1: 2% acetonitrile/1% formic acid in water.
Add 20 mL of acetonitrile and 10 mL of formic acid to a 1 L
reagent bottle. Bring to volume with CLRW and mix thor-
oughly. Stable for 1 month at 20–27 C.
Opioids by HRMS 43
2.2 Standards and Targeted drugs and stock concentrations are shown in Table 1.
Controls
1. Intermediate stock internal standards: 10 μg/mL of each deut-
erated compound. To separate 10 mL volumetric flasks, add
1 mL of each 100 μg/mL stock solution (dihydrocodeine-d6,
buprenorphine-d4, norbuprenorphine-d3, fentanyl-d5,
norfentanyl-d5) or 100 μL of each 1 mg/mL stock solution
(oxycodone-d6 or methadone-d9). Bring to volume with
methanol. Stable 10 C in amber glass vials with Teflon
caps for 2 years.
2. Working internal standard: 7.5 ng/mL of oxycodone-d6, dihy-
drocodeine-d6, and methadone-d9, 1.0 ng/mL of fentanyl-d5
and norfentanyl-d5, and 2.5 ng/mL of buprenorphine-d4 and
norbuprenorphine-d5. Add 75 μL of the oxycodone-d6 inter-
mediate, 75 μL of the dihydrocodeine-d6 intermediate, 75 μL
of the methadone-d9 intermediate, 25 μL of the
buprenoprhine-d4 intermediate, 25 μL of the
norbuprenorphine-d5 intermediate, 10 μL of the fentanyl-d5
intermediate, and 10 μL of the norfentanyl-d4 intermediate
stock solution to a 100 mL volumetric flask. Bring to volume
with 0.5% formic acid in CLRW. Mix well for 20–30 min.
Stable for 2 months at 2–8 C.
3. Intermediate opioid standard: 10 μg/mL of each drug. Add
100 μL of each 1.0 mg/mL stock of methadone, EDDP,
meperidine, cis-tramadol, tapentadol, o-desmethyltramadol,
dihydrocodeine, n-desmethyltramadol, norhydrocodone, oxy-
codone, noroxycodone, hydromorphone, oxymorphone, nor-
meperidine, morphine, noroxymorphone, hydrocodone,
codeine, and naloxone to a 10 mL volumetric flask. Bring to
volume with methanol, and mix well. Stable at or below
10 C in amber glass vials with Teflon caps for 2 years or
until manufacturer expiration of stock I, whichever comes first.
44 Darlington Danso et al.
Table 1
Standard concentrations (stock and intermediate calibration solution), negative control, cutoff, and
positive control values for the targeted opioid screen
Positive
Stock Intermediate Negative Cutoff control
concentration concentration control value value value
Drug (mg/mL) (μg/mL) (ng/mL) (ng/mL) (ng/mL)
Codeine 1.0 10.0 10.0 25.0 50.0
Codeine-6-β-glucuronide 0.1 10.0 50.0 100.0 200.0
Morphine 1.0 10.0 10.0 25.0 50.0
Morphine- 1.0 10.0 50.0 100.0 200.0
6-β-glucuronide
6-Acetylmorphine 1.0 10.0 10.0 25.0 50.0
Hydrocodone 1.0 10.0 10.0 25.0 50.0
Norhydrocodone 1.0 10.0 10.0 25.0 50.0
Dihydrocodeine 1.0 10.0 10.0 25.0 50.0
Hydromorphone 1.0 10.0 10.0 25.0 50.0
Hydromorphone- 0.1 10.0 50.0 100.0 200.0
3-β-glucuronide
Oxycodone 1.0 10.0 10.0 25.0 50.0
Noroxycodone 1.0 10.0 10.0 25.0 50.0
Oxymorphone 1.0 10.0 10.0 25.0 50.0
Oxymorphone- 0.1 10.0 50.0 100.0 100.0
3-β-glucuronide
Noroxymorphone 1.0 10.0 10.0 25.0 50.0
Fentanyl 1.0 1.0 1.0 2.0 5.0
Norfentanyl 1.0 1.0 1.0 2.0 5.0
Meperidine 1.0 10.0 10.0 25.0 50.0
Normeperidine 1.0 10.0 10.0 25.0 50.0
Naloxone 1.0 10.0 10.0 25.0 50.0
Naloxone- 1.0 10.0 50.0 100.0 200.0
3-β-glucuronide
Methadone 1.0 10.0 10.0 25.0 50.0
EDDP 1.0 10.0 10.0 25.0 50.0
Propoxyphene 1.0 10.0 10.0 25.0 50.0
Norpropoxyphene 1.0 10.0 10.0 25.0 50.0
Tramadol 1.0 10.0 10.0 25.0 50.0
(continued)
Opioids by HRMS 45
Table 1
(continued)
Positive
Stock Intermediate Negative Cutoff control
concentration concentration control value value value
Drug (mg/mL) (μg/mL) (ng/mL) (ng/mL) (ng/mL)
O-Desmethyltramadol 1.0 10.0 10.0 25.0 50.0
Tapentadol 1.0 10.0 10.0 25.0 50.0
N-Desmethyltapentadol 1.0 20.0 20.0 50.0 100.0
Tapentadol-β-glucuronide 0.1 10.0 50.0 100.0 200.0
Buprenorphine 1.0 2.0 2.0 5.0 10.0
Norbuprenorphine 1.0 2.0 2.0 5.0 10.0
Norbuprenorphine 1.0 10.0 10.0 20.0 50.0
glucuronide
3 Methods
3.1 Extraction 1. Mix all patient samples briefly, aliquot into labelled
Procedure (see Note 3) 12 75 mm disposable borosilicate glass test tubes, and cen-
trifuge for 10 min at 2850 g.
2. Aliquot 100 μL of each blank, standard, control, and patient
sample into its own sample well of a 96-well plate. This assay
uses a one point calibration (the cutoff calibrator), a negative
control, a positive control, and a blank (drug-free urine) with
each batch (see Note 4).
3. Add 900 μL of working internal standard to each well, and then
mix by vortexing.
4. Cover the plate with an adhesive seal for analysis.
Table 2
Gradient conditions
Table 3
Mass spec parameters (source conditions)
4 Notes
Table 4
Transitions and retention times for drug identification
Rt Rt
Drug m/z (min) Drug m/z (min)
Codeine 300.15918 3.10 Meperidine 248.16428 3.86
Codeine-6-β-glucuronide 476.19151 2.62 Normeperidine 234.14871 3.85
Morphine 286.14337 1.97 Naloxone 328.15433 2.90
Morphine-6-β-glucuronide 462.17586 1.77 Naloxone-3-β-glucuronide 504.18642 1.82
6-Acetylmorphine 328.15359 3.37 Methadone 310.21628 4.10
Hydrocodone 300.15912 3.71 EDDP 278.19012 4.11
Norhydrocodone 286.14328 3.50 Propoxyphene 340.22687 4.02
Dihydrocodeine 302.17435 2.83 Norpropoxyphene 308.20056 4.11
Hydromorphone 286.14346 2.42 Tramadol 264.19547 3.83
Hydromorphone-3-β- 462.17586 1.43 O-Desmethyltramadol 250.17993 3.12
glucuronide
Oxycodone 316.15387 3.48 Tapentadol 222.18501 3.83
Noroxycodone 302.13846 3.28 N-desmethyltapentadol 208.16959 3.83
Oxymorphone 302.13837 2.12 Tapentadol-β-glucuronide 398.21713 3.29
Oxymorphone-3-β- 478.17080 1.08 Buprenorphine 468.31061 3.97
glucuronide
Noroxymorphone 288.12303 1.63 Norbuprenorphine 414.26343 3.85
Fentanyl 337.22711 3.97 Norbuprenorphine glucuronide 590.29597 3.51
Norfentanyl 233.16461 3.76
Rt retention time
References
Abstract
Buprenorphine, a synthetic opioid possessing both analgesic and opioid receptor antagonist properties, has
proven to be an effective therapeutic aid for opioid dependency and chronic pain management. The
downside, as with all opioids, natural or synthetic, is its potential for misuse and abuse. The euphoria
induced by buprenorphine leads to abuse. Additionally, individuals with an active addiction to short-acting
opioids such as heroin may use buprenorphine between doses of their drug of choice to stave off withdrawal
symptoms. As such, buprenorphine monitoring is utilized in medication-assisted therapy programs for
opioid dependency, as well as chronic pain management settings. Buprenorphine may also be included in
drug testing programs for law enforcement purposes. The assay described here was designed to detect and
quantify both buprenorphine and its metabolite norbuprenorphine.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_5,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
51
52 Andrea R. Terrell et al.
2 Materials
2.1 Prepared 1. Mobile phase A: 0.1% formic acid in water: Break 1 ampule of
Reagents (See Note 1) high-grade formic acid and transfer the entire contents (1 mL)
using a glass pipette (see Note 2) into 1 L of HPLC grade
water. Store at room temperature for up to 1 month (see
Note 3).
2. Mobile phase B: 0.1% formic acid in methanol: Break 1 ampule
of high-grade formic acid and transfer the entire contents
(1 mL) using a glass pipette into 1 L of HPLC grade methanol.
Store at room temperature for up to 1 month.
3. Needle rinse: 60/20/20 isopropanol/methanol/acetonitrile:
Combine 200 mL of methanol, 200 mL of acetonitrile, and
600 mL of isopropanol. Store at room temperature for up to
1 month.
4. 200 mM phosphate buffer, pH 6.8: Weigh out 8.0 g of
sodium phosphate monobasic dihydrate and 8.7 g of sodium
phosphate dibasic dihydrate and mix to dissolve in 500 mL of
water. Using pH paper or a pH meter, verify pH is 6.8 (0.2).
Store at room temperature for up to 2 months.
5. 20 mM ammonium formate buffer in 80/20 water/meth-
anol, pH 3.7: Weigh 250 mg of ammonium formate and
dissolve in 160 mL of mobile phase A. Add 40 mL of mobile
phase B. Using pH paper or a pH meter, verify pH is 3.7
(0.2). Store at room temperature for up to 1 month.
6. Internal Standard (IS) spiking solution: 50 ng/mL each
IS. Combine 10 μL of 100 μg/mL buprenorphine-d4 stock
solution, 10 μL of 100 μg/mL norbuprenorphine-d3 stock
solution, 5 mL of acetonitrile, and 15 mL of water in a glass
or polypropylene tube. Mix well. Store refrigerated for up to
1 month.
7. Composite mix solution (CMS BUP): 10,000 ng/mL
buprenorphine and norbuprenorphine. Combine 100 μL
of 100 μg/mL buprenorphine standard, 100 μL of 100 μg/
mL norbuprenorphine standard, and 800 μL of acetonitrile in a
glass screw cap vial. Mix well.
8. Composite spiking solution (CSS) 1: 2000 ng/mL bupre-
norphine and norbuprenorphine. Combine 200 μL of CMS
BUP with 800 μL of acetonitrile in a glass screw cap vial. Store
at 20 C for up to 2 months.
9. CSS 2: 400 ng/mL buprenorphine and norbuprenor-
phine. Mix 200 μL of CSS 1 with 800 μL of acetonitrile in a
glass screw cap vial. Store at 20 C for up to 2 months.
54 Andrea R. Terrell et al.
2.2 Preparation of 1. Cal 1: 2 ng/mL both analytes. Combine 100 μL of CSS 4 and
Urine Calibrators and 900 μL of urine in a glass screw cap vial. Store at 20 C for up
Controls to 2 months (see Note 4).
2. Cal 2: 5 ng/mL both analytes. Combine 50 μL of CSS 3 and
950 μL of urine in a glass screw cap vial. Store at 20 C for up
to 2 months.
3. Cal 3: 10 ng/mL both analytes. Combine 100 μL of CSS 3 and
900 μL of urine in a glass screw cap vial. Store at 20 C for up
to 2 months.
4. Cal 4: 20 ng/mL both analytes. Combine 50 μL of CSS 2 and
950 μL of urine in a glass screw cap vial. Store at 20 C for up
to 2 months.
5. Cal 5: 50 ng/mL both analytes. Combine 125 μL of CSS 2 and
875 μL of urine in a glass screw cap vial. Store at 20 C for up
to 2 months.
6. Cal 6: 100 ng/mL both analytes. Combine 50 μL of CSS 1 and
950 μL of urine in a glass screw cap vial. Store at 20 C for up
to 2 months.
Buprenorphine and Norbuprenorphine 55
2.3 Supplies and 1. Sciex 4500 system with Turbo V Source and diverter valve.
Analytical Equipment 2. MultiQuant software v3.0 or later.
3. Shimadzu Nexera XR HPLC System.
4. LC-20AD XR binary pumps.
5. SIL-20AC Autosampler.
6. CTO-20AC column oven.
7. CBM-20A System Controller.
7. Phenomenex HPLC column, 2.6 μm, Kinetex Biphenyl,
50 3.0 mm.
8. Phenomenex SecurityGuard ULTRA UHPLC Biphenyl Car-
tridges, 3.0 mm.
9. Phenomenex SecurityGuard ULTRA Holder.
10. Screw cap glass vials capable of holding 2 mL.
11. Glass autosampler vials with inserts.
12. β-glucuronidase (IMCSzyme [recombinant], >50,000 U/
mL, or similar).
13. 1.5 mL or 2.0 mL microcentrifuge tubes.
14. Polypropylene tubes capable of holding >5 mL.
15. pH meter or pH paper.
3 Method
3.1 Sample 1. Aliquot 50 μL of urine from each calibrator, QC, and sample
Preparation into a microcentrifuge tube.
2. For each double blank and negative control, aliquot 50 μL of
blank urine.
56 Andrea R. Terrell et al.
3.2 Analysis 1. Inject 100 μL of the system suitability standard before each
batch.
2. Injection volume: 5 μL.
3. Needle rinse mode: before and after aspiration.
4. Autosampler temperature: 8 C.
5. Flow rate: 0.7 mL/min.
6. Column oven temperature: 40 C.
7. Gradient conditions are shown in Table 1.
8. Divert flow to waste for the first 1 min.
9. Mass spectrometer conditions are shown in Table 2.
10. Analyte-specific parameters are shown in Table 3.
11. Process the data using the first transition for each analyte as the
quantifier, and the second transition as the ion ratio qualifier.
12. Use linear regression model with 1/x2 weighting for all
analytes.
Table 1
Chromatography gradient
Table 2
Mass spectrometer parameters
Parameter Setting
Polarity ESI positive/negative switching
Curtain gas 30 psi
CAD gas 10
Ionspray voltage (Pos) 2500 V
Ionspray voltage (Neg) 4500 V
Temperature 550 C
Ion source gas 1 60 psi
Ion source gas 2 60 psi
Acquisition time 5.9 min
Q1 resolution Unit
Q3 resolution Unit
MRM pause time 3 ms
Settling time 50 ms
MRM window (Pos) 40 s
Target scan time (Pos) 0.3 s
MRM window (Neg) 20 s
Target scan time (Neg) 0.05 s
EP 10 V
Table 3
Analyte-specific parameters
Fig. 2 A sample that has been adulterated by “pill scraping” where a small amount of pill or film is directly
added to the urine sample. In this scenario, samples will have an elevated concentration of parent drug and
little to no metabolite
13. Confirm that batch meets acceptance criteria (see Note 5).
14. Report patient samples using a cutoff of 10 ng/mL. Results
below this are reported as negative or not detected (see Note
6). Figures 1, 2, and 3 show examples of patient results using
this method.
4 Notes
References
1. Samhsa.gov/medication-assisted-treatment/ 4. Baselt RC (2017) Disposition of toxic drugs and
buprenorphine chemicals in man, 11th edn. Biomedical Publi-
2. Drug Enforcement Administration Office of cations, Seal Beach, CA
Diversion Control, Drug and Chemical Evaluation 5. Yokell MA et al (2011) Buprenorphine and
Section, July 2013 publication “Buprenorphine” buprenorphine/naloxone diversion, misuse,
3. Crane EH (2015) Emergency department visits and illicit use: an international review. Curr
involving narcotic pain relievers. The CBHSQ Drug Abuse Rev 4(1):28–41
report, November 5
Chapter 6
Abstract
Tapentadol is an orally active analgesic with a similar structure to tramadol. Its primary mechanism of action
is agonist action on the mu-opioid receptor. The method described here quantitates tapentadol in the
whole blood using a matching deuterated internal standard, extraction via a protein “crash” with acetoni-
trile, followed by analysis using liquid chromatography-tandem mass spectrometry.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_6,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
61
62 Graham R. Jones and Russell P. Handy
2 Materials
3 Methods
Table 1
Spiking scheme for calibration standards
Final conc. tapentadol (mg/L) 0.025 0.05 0.1 0.25 0.5 1 2.5 5
Working standard A (μL) – – – – 10 20 50 100
Working standard B (μL) 5 10 20 50 – – – –
3.2 Analysis 1. Place the extracts on the autosampler in the following order:
blank, calibrators in order of lowest to highest concentrations,
blank (can reinject from same vial), QC sample, unknown
samples, and 0.5 mg/L calibrator (reinject from same vial)
(see Note 4).
2. Autosampler parameters cool to constant 4 C.
3. Injection volume 0.5 μL.
4. Column temperature 45 C.
5. Flow rate 0.5 mL/min.
6. Mobile phase program (see Note 5):
(a) 0.0 min: mobile B ratio 10%
(b) 4.0 min: mobile B ratio 50%
(c) 6.0 min: mobile B ratio 50%
(d) 6.01 min: mobile B ratio 95%
(e) 8.5 min: mobile B ratio 95%
(f) 8.51 min: mobile B ratio 10%
(g) 14.0 min: mobile B ratio 10%
7. Ion source parameters: gas temp ¼ 350 C, gas flow 11 L/min;
nebulizer ¼ 35 psi, capillary voltage 4000 V.
64 Graham R. Jones and Russell P. Handy
Table 2
Analytical and detection conditions for tapentadol and the deuterated internal standard
Qualifier
Qualifier Qualifier Qualifier #2
Ret. Quantifier Quantifier #1 #1 #2 collision
time Precursor product collision product collision product energy
Compound (min) ion (m/z) ion (m/z) energy (V) ion (m/z) energy (V) ion (m/z) (V)
Tapentadol 3.6 222.2 107 24 121 18 77 57
Tapentadol- 3.6 225.2 107 24 121 18 77 57
D3
Fig. 1 MRM chromatograms of tapentadol and tapentadol-D3, showing the quantifying ion transitions 222.2 to
107 and 225.2 to 107, respectively, and the corresponding qualifying ions of 121 and 77
4 Notes
References
1. Baselt RC (2017) Disposition of toxic drugs and 2. Ortho-McNeil-Janssen (2010) Nucynta (tapen-
chemicals in man, 11th edn. Biomedical Publi- tadol) prescribing information. PriCara Divi-
cations, Seal Beach, CA sion, Raritan, NJ
Chapter 7
Abstract
High-performance liquid chromatography tandem mass spectrometry (LC-MS/MS) has become a primary
analytical methodology in therapeutic drug monitoring of antiepileptic drugs (AEDs). To demonstrate the
utility of LC-MS/MS in measuring drug concentrations in serum or plasma, analysis of lacosamide
(Vimpat™) is discussed in this chapter. Lacosamide is an example of the newer-generation AEDs. The
drug is extracted by protein precipitation and dilution of the serum specimen. A small volume of the
extracted specimen is injected into a reversed-phase chromatography column, and lacosamide is identified
by positive electrospray ionization (ESI) mass spectrometry in the multiple reaction monitoring (MRM)
mode, which provides selectivity for quantitative analysis. A deuterated internal standard is used to correct
for any loss of analyte during the process of extraction and analysis. A seven-point calibration curve and two
levels of quality controls are included in each batch.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_7,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
67
68 He Sarina Yang and Leslie Edinboro
2 Materials
2.2 Supplies 1. Agilent 1200 series pump system operating in laminar flow
and Analytic mode (or equivalent).
Equipment 2. Applied Biosystems API 3200 mass spectrometer
(or equivalent).
3. Sciex Analyst® software.
4. Restek guard column.
5. Restek Pinnacle DB Biphenyl 5 μm 50 x 2.1 mm column.
6. Allegra X-15R benchtop centrifuge, Beckman Coulter.
7. Autosampler Vials-Robo Type 1, Class A (or equivalent).
8. Flat-bottom glass inserts (250 μL).
9. Vials with caps and septa.
10. Vortex mixer.
11. Eppendorf microcentrifuge or equivalent.
12. Tubes, Eppendorf 1.5 mL.
13. 2 mL and 1 mL 96-well collection plates.
3 Methods
Table 1
Gradient program
Step Total time (min) Flow rate (mL/min) A (%) [aqueous] B (%) [organic]
0 0.10 0.75 90 10
1 0.20 0.75 30 70
2 1.90 0.75 10 90
3 2.00 0.75 90 10
Table 2
Mass transitions for lacosamide and internal standard
Fig. 1 Example of chromatography for lacosamide (254.1/91.0 and 254.1/108.0) and internal standard
lacosamide-13C, D3 (255.4/91.0)
3.3 Data Analysis 1. Generate calibration curves using a weighted (1/x) linear
regression curve.
2. Quantitate analyte peaks by normalizing the peak area of the
quantifier ion of each compound to the internal standard.
3. Calculate lacosamide concentration from the calibration curve
(see Note 8).
4. Analytical measure range (AMR) of this assay is 0.5 to 20 μg/mL.
4 Notes
References
1. Krasowski MD (2010) Therapeutic drug moni- retigabine and eslicarbazepine acetate. Expert
toring of the newer anti-epilepsy medications. Opin Pharmacother 13(5):699–715
Pharmaceuticals 3:1909–1905 5. Jacob S, Nair AB (2016) An updated overview
2. Anderson GD (2008) Pharmacokinetic, phar- on therapeutic drug monitoring of recent anti-
macodynamics, and pharmacogenetic targeted epileptic drugs. Drug R D 16:303–316
therapy of antiepileptic drugs. Ther Drug 6. Ben-Menachem E (2008) Lacosamide: an inves-
Monit 30(2):173–180 tigational drug for adjunctive treatment of
3. Deeb S, McKeown DA, Torrance HJ, Wylie FM, partial-onset seizures. Drugs Today 44
Logan BK, Scott KS (2014) Simultaneous anal- (1):35–40
ysis of 22 antiepileptic drugs in postmortem 7. Eadie MJ (1998) Therapeutic drug monitor-
blood, serum and plasma using LC-MS-MS ing—antiepileptic drugs. Br J Clin Pharmacol
with a focus on their role in forensic cases. J 46:185–193
Anal Toxicol 38(8):485–494 8. Krasowski MD 2013 Antiepileptic drugs: thera-
4. Patsalo PN, Berry DJ (2012) Pharmacotherapy peutic drug monitoring of the newer generation
of the third-generation AEDs: lacosamide, drugs. Clinical Laboratory News
Chapter 8
Abstract
Leflunomide is a prodrug that is metabolized to the active metabolite, teriflunomide (A77 1726), to inhibit
the enzyme dihydroorotate dehydrogenase and decrease the synthesis of pyrimidine nucleotides for DNA
and RNA synthesis. Teriflunomide is primarily used for the treatment of rheumatoid arthritis and multiple
sclerosis.
A liquid chromatography tandem mass spectrometry (LC-MS/MS) method was developed and validated
to quantify the drug teriflunomide over a concentration range of 5 ng/mL–200 μg/mL in serum or plasma.
The calibration curve was divided into two separate overlapping regions of the analytical measurement
range, with a high curve and a low curve range. Samples are first analyzed using the high-range calibration
curve after a 100-fold dilution of the sample extract. Samples falling below the upper curve region are
evaluated again without dilution and quantified, if possible, against the low curve calibration standards.
This method can be used to support therapeutic drug monitoring of patients that are administered with
leflunomide therapy.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_8,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
75
76 Geoffrey S. Rule et al.
2 Materials
2.2 Standards The high calibration curve consists of standards 6–10; the low
and Controls calibration curve consists of standards 1–5. Controls for the high
curve are D, E, and F; controls for the low curve are A, B, and C.
1. Stock solution: 10 mg/mL teriflunomide in DMSO. Add
10 mg of teriflunomide powder to 1 mL of dimethyl sulfoxide
(DMSO). Mix until completely dissolved.
2. Standard 10: 200 μg/mL teriflunomide. Add 5 μL of stock
solution to 245 μL of drug-free serum. Vortex to mix. Make
fresh daily.
3. Standard 9: 50 μg/mL teriflunomide. Add 40 μL of standard
10–120 μL of drug-free serum. Vortex to mix. Make fresh daily.
4. Standard 8: 10 μg/mL teriflunomide. Add 10 μL of standard
10–190 μL of drug-free serum. Vortex to mix. Make fresh daily.
5. Standard 7: 2 μg/mL teriflunomide. Add 10 μL of standard
10–990 μL of drug-free serum. Vortex to mix. Make fresh daily.
6. Standard 6: 0.8 μg/mL teriflunomide. Add 100 μL of standard
7–150 μL of drug-free serum. Vortex to mix. Make fresh daily.
7. Standard 5: 1 μg/mL teriflunomide. Add 200 μL of standard
7–200 μL of drug-free serum. Vortex to mix. Make fresh daily.
8. Standard 4: 0.5 μg/mL teriflunomide. Add 100 μL of standard
5–100 μL of drug-free serum. Vortex to mix. Make fresh daily.
9. Standard 3: 0.1 μg/mL teriflunomide. Add 25 μL of standard
4–100 μL of drug-free serum. Vortex to mix. Make fresh daily.
78 Geoffrey S. Rule et al.
2.3 Solutions 1. Mobile phase a: 0.1% formic acid in deionized water. Add 1 mL
of formic acid to 999 mL of deionized water in a graduated
cylinder. Mix.
2. Mobile phase B: 0.1% formic acid in 1:1:18 water/methanol/
acetonitrile. Add 1 mL of formic acid, 50 mL of deionized
water, and 50 mL of methanol to a 1 L graduated cylinder. Fill
to volume with acetonitrile and mix.
Teriflunomide LC-MS/MS Method 79
3 Methods
3.1 Sample 1. Label a microcentrifuge tube for each standard [1–10], control
Preparation (A–F), patient sample, blank, and double blank (see Note 1).
2. Add 100 μL of each blank serum, standard, control, and patient
serum/plasma to the appropriate microcentrifuge tubes.
3. Add 300 μL of “double blank solution” to the tubes designated
double blanks.
4. Add 300 μL of “working internal standard” to all other blanks,
calibrators, controls, and patient samples (see Note 2).
5. Cap tubes and vortex mix for 2 min.
6. Centrifuge for 10 min at 20783 rcf.
7. Prepare samples for injection:
(a) High curve: Transfer 2 μL of each patient, calibrator 6–10,
control D-F, and blank supernatant into autosampler vials
containing 200 μL of dilution solution. Cap and mix.
(b) Low curve: Transfer 100 μL of each calibrator 1–5, control
A–C, blank, and low patient (see Note 3) supernatant into
autosampler vials and cap.
80 Geoffrey S. Rule et al.
Table 1
Chromatography conditions
Table 2
MS/MS operating parameters
Table 3
List of precursor and product ions for teriflunomide and internal standard
Collision cell
Precursor ion Product ion Dwell (msec) Collision energy (eV) exit potential
Teriflunomide 269.1 82.0 50 17 5
269.1 160.0 50 26 2
d4-teriflunomide 273.1 82.0 50 27 5
273.1 164.0 50 34 2
XIC of -MRM (4 pairs): 269.000/160.000 Da from Sample 1 (Test inject std) of Run14.w if... Max. 3.1e5 cps.
1.06e6
1.00e6
N
9.00e5
H F F
8.00e5 N
F O
F
7.00e5 O OH
F N
N
F H
Intensity, cps
6.00e5 O
MW = 270
teriflunomide
5.00e5 MW = 270
4.00e5 leflunomide
2.18
3.00e5
2.49
2.00e5
1.00e5
0.00
0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0
Time, min
Fig. 1 Extracted ion current chromatograms of teriflunomide in standard 1 (5 ng/mL), and internal standard (d4
teriflunomide), showing two transitions for each
4 Notes
1. For each curve range, both a blank and double blank sample are
prepared. A blank is analyzed as the first and last injection of
each sequence and consists of a known negative serum sample.
A double blank is analyzed after each high standard. A “double
blank” sample is one prepared in identical fashion to the ordi-
nary blank but in the absence of internal standard. Placing this
sample after the high calibration standard allows one to distin-
guish between autosampler carryover and unlabeled analyte
that may be contributed through the internal standard addition
as an impurity.
2. Prepare the low and high calibration curve standards, all quality
control samples and patient samples in the same batch. Prepare
each patient sample with 100 dilution, as described, and
evaluate against the high curve calibration standards. If within
the high calibration curve region, report the determined value.
Samples above 200 μg/mL must be extracted a second time
with appropriate matrix dilution. Patient samples below
0.8 μg/mL (the lower limit of quantitation of the high curve)
can be prepared according to instructions for the low curve,
then analyzed along with the low curve calibrators and quality
control samples. Report the resulting value as appropriate.
3. The concentration of internal standard utilized is equivalent to
1 μg/mL in the plasma/serum sample. This concentration is
near the low end of the upper curve and at the high end of the
lower curve. It is possible to use this same solution (concentra-
tion) for both curves since there is a lack of a significant analyte
isotope interference with the IS and because only a small
amount of unlabeled analyte is present in the IS. We discuss
this topic elsewhere in detail [16].
4. A test injection is made with a solution of teriflunomide and
leflunomide to verify instrument performance (sensitivity and
retention time) and chromatographic separation (resolution)
prior to each batch of samples. Results are compared with
historical values, and a judgment is made regarding system
suitability for patient sample analysis.
5. Although the parent drug, leflunomide, is generally found only
at very low concentrations, if at all, it is separated chromato-
graphically from the metabolite, teriflunomide, due to the fact
that the two have both the same precursor and product ion
masses.
Teriflunomide LC-MS/MS Method 83
Acknowledgments
The authors would like to thank the ARUP Institute for Clinical
and Experimental Pathology for making this work possible.
References
Abstract
Immunotherapies are a hot topic, with the potential to impact our understanding of the immune system
and treat a diverse array of conditions. Therapeutic monoclonal antibodies (mAbs) are part of this revolu-
tion, and clinical chemists are aware of the success of the biologic drugs. Antibodies are not just immuno-
assay reagents anymore but are also present in clinical serum samples from more and more patients each day.
The clinical laboratory will have many roles as mAb therapies expand, including the development of new
assays to differentiate a mAb from an endogenous, disease-causing clone and monitoring therapeutic drugs
for better patient outcomes and assessing for the loss of response to therapy.
Therapeutic mAbs use has expanded significantly in the last 5 years, and depending on their target or
their concentration, they may impact routine clinical testing for patients. Optimizing therapy during the
induction phase to keep the mAb concentrations above certain thresholds has proven to be associated with
improved responses and better outcomes in chronic conditions such as inflammatory bowel disease. This
chapter will describe a LC-MS/MS protocol for analysis of tryptic peptides unique to infliximab (clonotypic
peptides) for quantitation of the mAb. The protocol can be adapted to other mAbs with similar outcomes
and is a useful, relatively simple strategy for measurement of mAbs.
Key words LC-MS/MS, Infliximab, Therapeutic monoclonal antibodies, Trypsin, Tryptic peptides,
Method development
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_9,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
85
86 Maria Alice V. Willrich
Fragment
antigen
binding (Fab)
Disulfide bonds
hinge
CH2 CH2
Crystallizable
fraction (Fc)
CH3 CH3
1.1 Tryptic Peptide In order to quantify mAbs using mass spectrometry, the mAb of
Methods interest must first be differentiated from the very similar polyclonal
background of over 1 g/dL of endogenous human Igs in serum.
Historically, proteins have been quantitated by LC-MS/MS using
specific tryptic fragments (referred to as “proteotypic” peptides)
[14]. Multiple tryptic peptides can be quantitated at the same time,
as shown for the quantitation of IgG subclasses in serum by
LC-MS/MS [15]. For chimeric mAbs, whose entire variable region
is of animal origin, such as infliximab and rituximab, trypsin diges-
tion is possible as the nonhuman variable region is large (>250
amino acids). This increases the likelihood of finding unique signa-
ture peptides on the light chain and/or heavy chain which is specific
to that mAb and not found in the human polyclonal background.
Not every mAb will be amiable to the digest method especially
as newer mAbs are humanized (only small portions of the variable
region are of animal source, and they are engrafted onto a human
framework, such as eculizumab) or even fully human (genetically
engineered antibodies generated by phage display libraries, such as
adalimumab). Experiments to find a peptide specific for adalimu-
mab, for instance, failed to detect a tryptic peptide not found in the
polyclonal serum background (data not published). For other
mAbs, sensitivity of the method will be the limiting factor. For
88 Maria Alice V. Willrich
1) Digest mAb to
generate clonotypic
peptides
mixture containing H
2
H
2
non-specific peptides
using LC-MS/MS C
H
C
H
3 3
Intensity (cps)
2 Materials
2.1 Reagents and 1. 2,2,2-Trifluoroethanol (TFE): When handling it, wear gloves
Mobile Phases and pipette under a hood. Store at room temperature.
2. 50 mM ammonium bicarbonate: Weigh out 791 mg of ammo-
nium bicarbonate and dissolve in 200 mL of water, mixing well
at room temperature. Store at room temperature.
3. Saturated ammonium sulfate solution: Weigh out 150 g of
ammonium sulfate. Add 200 mL of water. Keep solution at
room temperature. Mix periodically over 8 h or more to ensure
solution is saturated and crystals are still visible (see Note 1).
Immediately before use, mix solution for 30 min using a stir
plate. Store at room temperature.
4. 200 mM dithiothreitol (DTT). Weigh out 30.85 mg of DTT.
Add 1 mL of 50 mM ammonium bicarbonate to dissolve. This
solution can be scaled up for larger batches; each sample requires
0.01 mL of DTT. This reagent should be prepared fresh daily;
do not store (see Note 2).
5. 200 mM iodoacetamide (IAA). Weigh out 37 mg of IAA. Add
1 mL of 50 mM ammonium bicarbonate to dissolve the powder.
This solution can be scaled up for larger batches; each sample
requires 0.02 mL of IAA. This reagent should be prepared daily.
Once ready, wrap the solution in aluminum foil (see Note 3) and
keep it at room temperature.
6. 1 mg/mL trypsin from bovine pancreas. Weigh out 5 mg of
trypsin. Dissolve in 5 mL of 50 mM ammonium bicarbonate.
The solution should be prepared fresh daily (see Note 4).
7. 1% formic acid. To a 25 mL volumetric flask, add 25 μL of
formic acid. Bring up to volume with water, mixing well. The
solution should be stored at room temperature.
8. Mobile phase A: 0.1% formic acid in water. In a 2 L reagent
bottle, add 2 L of water. Then, add 2 mL of formic acid, mixing
well. The solution should be stored at room temperature and
discarded after 1 week of use.
90 Maria Alice V. Willrich
2.2 Standards and Calibrators (standards) and controls are prepared by spiking inflix-
Controls imab into normal pooled human serum (see Note 5). Prepare
standards using volumetric flasks and good laboratory pipetting
practices.
1. 10 mg/mL Infliximab stock solution: Obtain the pharmaceu-
tical preparation (trade name Remicade, Janssen Biotech) from
a pharmacy. Keep refrigerated at 4 C until the day of reconsti-
tution. The infliximab vial contains 100 mg lyophilized powder
of infliximab. Add 10 mL of water using a volumetric pipette to
the entire vial contents to obtain a concentration of 10 mg/mL
(see Notes 6 and 7).
2. 100 μg/mL infliximab standard. Add 1 mL of 10 mg/mL
infliximab stock solution to a 100 mL volumetric flask. Bring
to volume with normal (drug-free) human serum. Aliquot
350 μL into microcentrifuge tubes; stable frozen for 2 years
at < 60 C.
3. 50 μg/mL infliximab standard. Add 25 mL of the 100 μg/mL
infliximab standard to a 50 mL volumetric flask. Bring to
volume with normal (drug-free) human serum. Aliquot
350 μL into microcentrifuge tubes; stable frozen for 2 years
at < 60 C.
4. 20 μg/mL infliximab standard. Add 10 mL of the 100 μg/mL
infliximab standard to a 50 mL volumetric flask. Bring to
volume with normal (drug-free) human serum. Aliquot
350 μL into microcentrifuge tubes; stable frozen for 2 years
at < 60 C.
5. 10 μg/mL infliximab standard. Add 5 mL of the 100 μg/mL
infliximab standard to a 50 mL volumetric flask. Bring to
volume with normal (drug-free) human serum. Aliquot
350 μL into microcentrifuge tubes; stable frozen for 2 years
at < 60 C.
6. 5 μg/mL infliximab standard. Add 2.5 mL of the 100 μg/mL
infliximab standard to a 50 mL volumetric flask. Bring to
volume with normal (drug-free) human serum. Aliquot
350 μL into microcentrifuge tubes; stable frozen for 2 years
at < 60 C.
7. 2 μg/mL infliximab standard. Add 1 mL of the 100 μg/mL
infliximab standard to a 50 mL volumetric flask. Bring to
volume with normal (drug-free) human serum. Aliquot
350 μL into microcentrifuge tubes; stable frozen for 2 years
at < 60 C.
LC-MS/MS for Therapeutic Monoclonal Antibodies 91
Table 1
Tryptic peptides and transitions used to quantify infliximab
3 Methods
3.2 Trypsin Digestion 1. Add 10 μL of 200 mM DTT and 100 μL of TFE to every
sample in the analytical run. The reducing agents will break up
the disulfide bonds which hold the light and heavy chains of the
immunoglobulins together. This will expose the cleavage sites
of the light and heavy chains to trypsin.
94 Maria Alice V. Willrich
3.3 LC-MS/MS Peptides (Table 1) are unique to infliximab heavy and light chains,
Set-Up and and two transitions are monitored per peptide for added specificity.
Quantitation of The primary transition for quantitation is the light chain y6 transi-
Infliximab tion (LC-y6), with the y10 monitored as a qualitative ion. The
heavy chain y4 ion (HC-y4) is used as a second quantification ion;
the HC-y5 ion was also added as a qualitative ion.
1. Add 5 μL of the 5 μg/mL isotopically labeled standard working
solutions (retention time standards) to each well. Vortex the
plate for 10 s. These are used to monitor the retention time of
each desired peptide (clonotypic peptides from the light and
heavy chains of infliximab).
2. Place the plate in the Cohesive LX4 autosampler. Make sure
mobile phases A and B are in place, columns are equilibrated,
and the chromatography method is set up on your preferred
software (see Note 11).
3. Inject 20 μL of each digest onto a Phenomenex C8 Security-
Guard column (4 2.0 mm ID) followed by a Waters XBridge
C8 column (3.0 30 mm; 3.5 μm) with a flow of 400 μL/min.
4. Separate peptides using a 5.5 min gradient from 95% aqueous
(A: water + 0.1% formic acid) to 35% organic (B: acetoni-
trile + 0.1% formic acid) (Fig. 3).
5. Set instrument Turbo V ion source conditions as IS, 5500;
TEM, 600; CAD, 12; CUR, 40; GS1, 35; GS2, 30; and EP,
8. The DP, CE, and CXP values for the SRM transitions were
LC-MS/MS for Therapeutic Monoclonal Antibodies 95
1.8e4
Heavy chain-y4
1.3e5
Heavy chain-y4 (IS)
8.0e4
Horse IgG Light Chain-y4
1 2 3 4 5
Time (min)
Fig. 3 Chromatograms for heavy chain, light chain, horse IgG, and labeled internal standard peptides
transitions. The chromatograms illustrated above show a sample containing 10 μg/mL of infliximab. Horse
IgG was added at 50 μg/mL; isotopically labeled internal standards were added at 5 μg/mL
4 Notes
Acknowledgments
References
1. Ladwig PM, Barnidge DR, Willrich MAV 3. Lazar-Molnar E, Delgado JC (2016) Immuno-
(2017) Mass spectrometry approaches for genicity assessment of tumor necrosis factor
identification and quantitation of therapeutic antagonists in the clinical laboratory. Clin
monoclonal antibodies in the clinical labora- Chem 62(9):1186–1198. https://doi.org/10.
tory. Clin Vaccine Immunol 24(5). https:// 1373/clinchem.2015.242875
doi.org/10.1128/CVI.00545-16 4. Pavlov IY, Carper J, Lazar-Molnar E, Delgado
2. Ordas I, Mould DR, Feagan BG, Sandborn WJ JC (2016) Clinical laboratory application of a
(2012) Anti-TNF monoclonal antibodies in reporter-gene assay for measurement of func-
inflammatory bowel disease: tional activity and neutralizing antibody
pharmacokinetics-based dosing paradigms. response to infliximab. Clin Chim Acta
Clin Pharmacol Ther 91(4):635–646. 453:147–153. https://doi.org/10.1016/j.
https://doi.org/10.1038/clpt.2011.328 cca.2015.12.015
LC-MS/MS for Therapeutic Monoclonal Antibodies 99
Abstract
Mass spectrometry (MS) is a highly specific and sensitive technique that is used for the detection of many
different analytes with diverse chemical characteristics. It has been adopted by clinical laboratories for the
quantification of small molecules and, by extension, has been widely used for therapeutic drug monitoring.
It is an attractive alternative to immunoassay methods, because it is not subject to the same interferences. A
limitation of MS (relative to immunoassays) is the turnaround time. However, this can be addressed by
workflow parallelization with other assays. Herein we describe a tandem LC-MS/MS method for the
detection and quantification of methotrexate in human plasma with a lower limit of quantification of
0.01 μM and within-assay and between-assay coefficients of variation of less than 15%. This method lacks
interference from high-abundance metabolites and utilizes kindred chromatography to improve turn-
around time in the therapeutic drug monitoring laboratory.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_10,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
101
102 Gabrielle N. Winston-McPherson et al.
Fig. 1 The structure of folate, methotrexate, and methotrexate metabolites. Key structural differences are
highlighted
2 Materials
3 Methods
3.1 Extraction 1. Bring samples and supplies to room temp. Mix all standards,
controls, blanks, and samples well before pipetting.
2. Label a 1.6 mL microcentrifuge tube for each blank, standard,
control, and sample. The suggested sample order is given
below (see Note 6):
(a) Blank
(b) Standard 6
(c) Standard 5
(d) Standard 4
(e) Standard 3
(f) Standard 2
(g) Standard 1
(h) Blank
106 Gabrielle N. Winston-McPherson et al.
Fig. 2 Protein precipitation with organic solvent. The reaction mixture is shown
prior to addition of sample (a), after addition of sample and mixing (b), and after
centrifugation (c)
Table 1
Chromatographic program for the assay
Time (min) Flow (min/mL) Mobile phase A (%) Mobile phase B (%)
0 0.6 90 10
0.6 0.6 0 100
1.2 0.6 90 10
1.8 End N/A N/A
4 Notes
References
1. Jannetto PJ, Fitzgerald RL (2016) Effective 12. Aumente D, Buelga DS, Lukas JC et al (2006)
use of mass spectrometry in the clinical labora- Population pharmacokinetics of high-dose
tory. Clin Chem 62:92–98. https://doi.org/ methotrexate in children with acute lympho-
10.1373/clinchem.2015.248146 blastic leukaemia. Clin Pharmacokinet
2. Strathmann FG, Hoofnagle AN (2011) Cur- 45:1227–1238. https://doi.org/10.2165/
rent and future applications of mass spectrom- 00003088-200645120-00007
etry to the clinical laboratory. Am J Clin Pathol 13. Przybylski M, Preiss J, Dennebaum R, Fischer J
136:609–616. https://doi.org/10.1309/ (1982) Identification and quantitation of
AJCPW0TA8OBBNGCK methotrexate and methotrexate metabolites in
3. Garg U, Zhang YV (2016) Mass spectrometry clinical high-dose therapy by high pressure liq-
in clinical laboratory: applications in therapeu- uid chromatography and field desorption mass
tic drug monitoring and toxicology. In: Clini- spectrometry. Biomed Mass Spectrom
cal applications of mass spectrometry in drug 9:22–32. https://doi.org/10.1002/bms.
analysis. Springer, New York, pp 1–10 1200090106
4. Al-Turkmani MR, Law T, Narla A, Kellogg 14. Wu D, Wang Y, Sun Y et al (2015) A simple,
MD (2010) Difficulty measuring methotrexate rapid and reliable liquid chromatography-mass
in a patient with high-dose methotrexate- spectrometry method for determination of
induced nephrotoxicity. Clin Chem 56:1792. methotrexate in human plasma and its applica-
https://doi.org/10.1373/clinchem.2010. tion to therapeutic drug monitoring. Biomed
144824 Chromatogr BMC 29:1197–1202. https://
5. Albertioni F, Rask C, Eksborg S et al (1996) doi.org/10.1002/bmc.3408
Evaluation of clinical assays for measuring 15. Schofield RC, Ramanathan LV, Murata K et al
high-dose methotrexate in plasma. Clin Chem (2015) Development and validation of a turbu-
42(39):39–44 lent flow chromatography and tandem mass
6. Bath RK, Brar NK, Forouhar FA, Wu GY spectrometry method for the quantitation of
(2014) A review of methotrexate-associated methotrexate and its metabolites. J Chroma-
hepatotoxicity. J Dig Dis 15:517–524. togr B Analyt Technol Biomed Life Sci
https://doi.org/10.1111/1751-2980.12184 1002:169–175. https://doi.org/10.1016/j.
jchromb.2015.08.025
7. Widemann BC, Adamson PC (2006) Under-
standing and managing methotrexate nephro- 16. Gunther V, Mueller D, von Eckardstein A,
toxicity. Oncologist 11:694–703. https://doi. Saleh L (2016) Head to head evaluation of
org/10.1634/theoncologist.11-6-694 the analytical performance of two commercial
methotrexate immunoassays and comparison
8. Cavone JL, Yang D, Wang A (2014) Glucarpi- with liquid chromatography-mass spectrome-
dase intervention for delayed methotrexate try and the former fluorescence polarization
clearance. Ann Pharmacother 48:897–907. immunoassay. Clin Chem Lab Med
https://doi.org/10.1177/ 54:823–831. https://doi.org/10.1515/cclm-
1060028014526159 2015-0578
9. Green JM (2012) Glucarpidase to combat 17. Steinborner S, Henion J (1999) Liquid-liquid
toxic levels of methotrexate in patients. Ther extraction in the 96-well plate format with
Clin Risk Manag 8:403–413. https://doi.org/ SRM LC/MS quantitative determination of
10.2147/TCRM.S30135 methotrexate and its major metabolite in
10. Ahmed YAAR, Hasan Y (2013) Prevention and human plasma. Anal Chem 71:2340–2345
management of high dose methotrexate toxic- 18. Nair H, Lawrence L, Hoofnagle AN (2012)
ity. J Cancer Sci Ther 5:106–112. https://doi. Liquid chromatography–tandem mass spec-
org/10.4172/1948-5956.1000193 trometry work flow for parallel quantification
11. Evans WE, Crom WR, Abromowitch M et al of methotrexate and other immunosuppres-
(1986) Clinical pharmacodynamics of high- sants. Clin Chem 58:943. https://doi.org/
dose methotrexate in acute lymphocytic leuke- 10.1373/clinchem.2011.175067
mia. N Engl J Med 314:471–477. https://doi.
org/10.1056/NEJM198602203140803
Chapter 11
Abstract
Numerous methods for the measurement of tacrolimus and cyclosporine A involving traditional liquid
chromatography tandem mass spectrometry (LC-MS/MS) have previously been described. The majority of
these methods use solid-phase extraction, liquid-liquid extraction, or protein precipitation extraction with
instrument run times greater than 15 s per sample. Continued demands in clinical labs for greater efficiency
and throughput have put increased stress on traditional technologies such as high-performance liquid
chromatography-ultraviolet detection (HPLC-UV) and traditional LC-MS/MS. As an improvement to the
existing methods, we describe a sensitive ultrafast LC-MS/MS with run times of less than 15 s per sample.
Key words Prograf, FK506, Neoral, CSA, Gengraf, Mass spectrometry, Rapid fire
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_11,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
111
112 Matthew W. Bjergum et al.
2 Materials
Bring to volume with bovine whole blood and mix well. Stable
up to 6 months at or below at 60 C.
11. Working Standard 5: 40.0 ng/mL tacrolimus/1000 ng/mL
cyclosporine A. Add 20 mL of water to a 1000 mL volumetric
flask, then add 4000 μL of tacrolimus Stock II standard and
1000 μL of 1.0 mg/mL cyclosporine A reference standard.
Bring to volume with bovine whole blood and mix well. Stable
up to 6 months at or below at 60 C.
12. Working Internal Standard: 2.0 ng/mL ascomycin/50 ng/mL
cyclosporine A-13C2,d4. Add 20 mL of water to a 2000 mL
volumetric flask, add 56 g of zinc sulfate heptahydrate, and mix
until completely dissolved. Add 1800 μL of methanol, 1000 μL
of 100 μg/mL Cyclosporine A-13C2,d4, and 50 μL of 100 μg/
mL ascomycin; mix well. Stable up to 14 days at or below at
10 C.
13. Quality control 1: 5.0 ng/mL tacrolimus/100 ng/mL cyclos-
porine A, in drug-free whole blood. Obtained commercially.
14. Quality control 2: 15.0 ng/mL tacrolimus/300 ng/mL
cyclosporine A, in drug-free whole blood. Obtained
commercially.
15. Quality control 3: 30.0 ng/mL tacrolimus/500 ng/mL
cyclosporine A, in drug-free whole blood. Obtained
commercially.
3 Methods
3.2 Analysis 1. Place the sample extracts in the 96-well plate in the following
order (see Note 4):
Table 1
Analytical and detection conditions for tacrolimus, cyclosporine A, and internal standards
Mean
Retention Quantifier Quant. Qualifier Qual. #1 ratio
time Precursor product collision product ion collision qualifier
Drug (Min.) ion (m/z) ion (m/z) energy (V) #1 (m/z) energy (V) MRM #1
Tacrolimus 0.145 821.8 768.5 17 786.5 13 35
Ascomycin 0.146 809.6 756.6 17 774.6 14 41
Cyclosporine 0.144 1219.8 1202.6 14 1184.5 30 9
A
Cyclosporine 0.144 1225.8 1208.6 15 – – –
A- 13C2,d4
Fig. 1 Chromatography of a working standard containing tacrolimus and internal standard (ascomycin)
4 Notes
References
1. Gabardi S, Halloran PF, Friedewald J (2011) 5. Scott LJ, McKeage K, Keam SJ et al (2003)
Managing risk in developing transplant Drugs 63:1247–1297. https://doi.org/10.
immunosuppressive agents: the new regulatory 2165/00003495-200363120-00006
environment. Am J Transplant 11:1803–1809. 6. Dunn CJ, Wagstaff AJ, Perry CM, Plosker GL,
https://doi.org/10.1111/j.1600-6143.2011. Goa KL (2001) Cyclosporin: an updated review
03653.x of the pharmacokinetic properties, clinical effi-
2. Burdmann EA, Andoh TF, Yu L, Bennett WM cacy and tolerability of a microemulsion-based
(2003) Cyclosporine nephrotoxicity. Semin formulation (neoral)1 in organ transplantation.
Nephrol 23(5):465–476 Drugs 61:1957–2016
3. Naesens M, Kuypers DR, Sarwal M (2009) Cal- 7. Moyer TP, Post GR, Sterioff S et al (1988)
cineurin inhibitor nephrotoxicity. Clin J Am Soc Cyclosporine nephrotoxicity is minimized by
Nephrol 4:481–508 adjusting dosage on the basis of drug concen-
4. Kahan BD, Keown P, Levy GA, Johnston A tration in blood. Mayo Clin Proc 63
(2002) Therapeutic drug monitoring of immu- (3):241–247
nosuppressant drugs in clinical practice. Clin
Ther 24:330–350
Chapter 12
Abstract
Gabapentin and pregabalin are anticonvulsant drugs that are also utilized for pain management. A mass
spectrometry method was developed and validated to quantify gabapentin and pregabalin in urine to
support testing for adherence.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_12,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
119
120 Stephen Merrigan and Kamisha L. Johnson-Davis
2 Materials
2.1 Supplies 1. Cellulose acetate filter for plasma preparation, 0.45 μm.
and Equipment 2. Volumetric glassware and pipettes.
3. 10 L container with a spigot or tap.
4. Transfer pipettes with tips.
5. 96 deep-well plate.
Gabapentin and Pregabalin in Urine 121
3 Methods
3.2 Analysis This assay employs electrospray ionization in positive ion mode and
multiple reaction monitoring (MRM) of mass transitions.
1. Set autosampler parameters according to Table 1. The injec-
tion volume is 10 μL.
2. Set mobile phase gradient according to Table 2. The flow rate
is 0.4 mL/min (see Note 7).
3. Set integrated switching valve parameters according to
Table 3.
4. Set mass spectrometer conditions according to Table 4.
5. Run samples. Retention times and MRM transitions are
shown in Tables 5 and 6, respectively.
6. Analyze data. The calibration curve is linear with 1/x weight-
ing (see Note 8).
4 Notes
Table 1
Autosampler parameters
Table 2
Mobile phase gradient
Table 3
Integrated valve parameters
Table 4
Mass spectrometer parameters
Table 5
Approximate retention times
Table 6
MRM transitions
Acknowledgments
The authors would like to thank the ARUP Institute for Clinical
and Experimental Pathology for support.
References
aminobutyric acid analogue in the treatment of 14. Hamandi K, Sander JW (2006) Pregabalin: a
neuropathic pain, partial-onset seizures, and new antiepileptic drug for refractory epilepsy.
anxiety disorders. Clin Ther 29:26–48 Seizure 15:73–78
11. Zareba G (2005) Pregabalin: a new agent for 15. Blommel ML, Blommel AL (2007) Pregabalin:
the treatment of neuropathic pain. Drugs an antiepileptic agent useful for neuropathic
Today 41:509–516 pain. Am J Health Syst Pharm 64:1475–1482
12. Guglielmo R, Martinotti G, Clerici M, Janiri L 16. Schulze-Bonhage A (2013) Pharmacokinetic
(2012) Pregabalin for alcohol dependence: a and pharmacodynamic profile of pregabalin
critical review of the literature. Adv Ther and its role in the treatment of epilepsy. Expert
29:947–957 Opin Drug Metab Toxicol 9:105–115
13. Buoli M, Caldiroli A, Serati M (2017) Pharma- 17. Heltsley R, Depriest A, Black DL, Robert T,
cokinetic evaluation of pregabalin for the treat- Caplan YH, Cone EJ (2011) Urine drug test-
ment of generalized anxiety disorder. Expert ing of chronic pain patients. IV. Prevalence of
Opin Drug Metab Toxicol 13:351–359 gabapentin and pregabalin. J Anal Toxicol
35:357–359
Chapter 13
Abstract
Since 2008 there has been an onslaught of new drugs in the illicit marketplace. Often referred to as
“research chemicals,” “designer drugs,” or “novel psychoactive substances” (NPS), these substances are
used for their pharmacological effects which are often similar to more widely known drugs such as ecstasy or
heroin. In some cases users specifically seek out these new chemicals, in other cases they are simply
purchasing what they believe to be their normal drug of choice from a dealer, but the product is not
what it is purported to be. Implementation of national and international systems to monitor the appearance
of new compounds enables laboratories to be prepared with validated tests to detect them in biological
specimens. The most common classes of NPS are synthetic cannabinoids, novel opioids, novel benzodia-
zepines, stimulants, and hallucinogens. Within these groups the compounds may be drugs that were
originally synthesized for research purposes during the pursuit of new therapeutic agents such as the
synthetic cannabinoid JWH-018 and the designer opioid U47700. Others like etizolam are compounds
used in other countries but not commonly seen in the USA. Some are drugs synthesized specifically to
circumvent legal controls. In all cases, these compounds present a unique challenge to forensic toxicology
laboratories which must quickly develop and validate analytical methods for the identification and quantifi-
cation in biological matrices.
This chapter is a condensed and updated version of an article originally published in Clinical and Forensic
Toxicology News.
Key words Research chemicals, Novel psychoactive substances, Synthetic drugs, Synthetic cannabi-
noids, Analogs
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_13,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
129
130 Sherri L. Kacinko and Donna M. Papsun
2 Monitoring NPS
3 Classes of NPS
3.1 Synthetic The first incidence of synthetic cannabinoids being detected in the
Cannabinoids USA occurred in late 2008 when US Customs and Border Patrol
verified the presence of HU-210 in packets of an herbal incense
product called “spice.” HU-210 was already a controlled sub-
stance, but shortly thereafter another uncontrolled drug,
JWH-018, was detected in similar products. This compound was
one of the 400 substances synthesized by Dr. Huffman. Since early
2009 the components of these herbal incenses have changed rap-
idly. As soon as the DEA adds a substance to the list of scheduled
compounds, chemists make slight modifications to the structure to
create a compound that is not listed.
A thorough review on the pharmacology and toxicology of
synthetic cannabinoids was published in 2014 and updated, along
with other classes of synthetic cannabinoids, in 2017 [6, 7]. Recrea-
tional synthetic cannabinoids target the CB1 receptor though many
also bind to and have an effect at the CB2 receptor. It is possible
that CB2 receptor activity modulates some of the undesirable
effects seen with the pure CB1 receptor agonists. The most com-
mon in vivo test for cannabinoids is the mouse tetrad test which
evaluates the effect of a compound on several different mouse
behaviors. This series of tests determines the dose of drug required
to change specific behaviors by 50% (ED50). By comparing the
ED50 of one drug with the ED50 of another drug, the potency can
be estimated. Unfortunately, this information is only available for a
handful of synthetic cannabinoids being used; the mouse tetrad has
not been performed on the vast majority of substances.
132 Sherri L. Kacinko and Donna M. Papsun
3.2 Opioids Novel opioid compounds have a longer history than synthetic
cannabinoids, but after a long period of inactivity with respect to
new compounds, acetyl fentanyl burst onto the recreational drug
scene in 2013, and then in late 2015, numerous other opioid-
related compounds began to appear. These primarily consist of
substances synthesized by pharmaceutical companies looking for
new analgesic drugs for pain treatment which were abandoned at
some point in the development process. These represent a chemi-
cally diverse class of substances, most of which do not have struc-
tural similarity to opiates.
There is evidence that these drugs are both sought after by
drug users for recreational use and by drug manufacturers as adul-
terants to more commonly abused opioids such as heroin. While
novel opioids may be structurally dissimilar, in general, these com-
pounds target the μ-opioid receptor. Activation of this receptor
causes analgesia, sedation, and euphoria which are the desired
effects for users. Adverse effects include itching, nausea, constipa-
tion, and decreased respiration. Users develop tolerance to both the
desired and undesired effects of opioid compounds, thus requiring
larger and larger doses. The development of tolerance along with
unknown identity of street drugs is a dangerous combination. The
potency of these compounds varies greatly. Individuals who believe
they are using heroin may use a dose consistent with previous doses
that resulted in the desired effect, but the presence of another,
more potent, opioid could make this dose deadly. Also of concern
is the increased toxicity of opioids when combined with benzodia-
zepines. Taken alone prescription benzodiazepines rarely result in
death, but the combination of benzodiazepines and opioids can
have a synergistic effect on respiratory depression.
Overdose with novel opioids is assumed to be similar to that
which is observed with any other μ-opioid receptor agonist and, like
overdoses with heroin or prescription opiates such as oxycodone or
oxymorphone, can be treated with naloxone. Naloxone acts as
antagonist at the μ-opioid receptor. Since naloxone has a must
stronger binding affinity for the receptor than opioid agonists, it
can replace them at the receptor and block additional receptors,
quickly and effectively reversing the central nervous system (CNS)
Designer Drugs 133
3.3 Benzodiazepines Benzodiazepines are one of the most commonly prescribed psycho-
tropic drug classes in the USA. There are 14 benzodiazepines
available by prescription used to treat a variety of physical and
psychological conditions. Compounds such as midazolam and tria-
zolam have half-lives of 2–3 h and are often used to induce anes-
thesia or as sleep medication. Alprazolam, lorazepam, and
clonazepam are common medications used to treat anxiety, panic
disorders, and seizures with effects lasting for 6–10 h. Chlordiaz-
epoxide which is used as a muscle relaxant and diazepam which is an
anti-anxiolytic are two examples of long-acting benzodiazepines.
These compounds typically have half-lives exceeding 24 h. The
gamma-aminobutyric acid A receptor is the target receptor for
benzodiazepines. This receptor has multiple binding sites which
may explain the differences in degree and type of reactions observed
from different benzodiazepines. All benzodiazepines have sedating
effects making them a target for abuse. In general, the drugs are
safe, but when combined with other CNS depressants, such as
alcohol or opioids, the danger increases.
Novel benzodiazepines are often referred to as “research ben-
zos” by drug users. Medications available in other countries but not
legal prescription medications in the USA, and compounds which
are not used in any country comprise this group. Phenazepam and
etizolam were the first two benzodiazepines to appear in the illicit
drug market in the USA. The DEA reports that between 2008 and
2013, there were 284 reports of phenazepam, and the drug was
found in 31 states [9]. Etizolam appeared in 2012 with a total of
140 reports between its first appearance and June 2014 according
to the DEA (14). Other compounds that have been reported
include bromazepam, flubromazepam, flubromazelam deloraze-
pam, diclazepam, and clonazelam.
Some signs and symptoms of benzodiazepine overdose include
anxiety, agitation, dizziness, confusion, nystagmus, slurred speech,
altered mental state, amnesia, hypotension, and impaired cogni-
tion. Respiratory depression may occur with benzodiazepine use,
the degree of which will be significantly increased if used in combi-
nation with other central nervous system depressants. Flumazenil is
a specific antidote for benzodiazepine overdose, but the risk of
flumazenil may outweigh potential benefits, so it is ideally only
used to treat acute overdose in the benzodiazepine naı̈ve individual.
Designer benzodiazepines are increasingly being detected in hospi-
talizations, impaired driving cases, and overdose deaths.
134 Sherri L. Kacinko and Donna M. Papsun
3.4 Other Classes At the same time synthetic cannabinoids were making an appear-
ance in the USA, synthetic cathinones and sympathomimetic
amines – derivatives of cathinone and methamphetamine – were
gaining in popularity. Commonly called “bath salts” or “plant
food,” the first generation of these products contained compounds
such as MDPV, mephedrone, and methylone. Although the pro-
ducts were all classified under the same street name of “bath salts”
and later “party pills” or “party powders,” the types and activities of
the compounds or combinations of compounds they contained
varied greatly including stimulants, hallucinogens, entactogens,
and dissociative anesthetics.
These substances are chemically similar to cathinone, a natural
stimulant found in the khat plant, as well as amphetamine and
methamphetamine, which are two stimulants that are routinely
found as drugs of abuse. Small chemical changes to the structure
of cathinone and/or amphetamine result in several new drugs that
pharmacologically behave very similarly, producing the same spec-
trum of stimulant effects such as increased energy, tachycardia, and
hypertension.
Like synthetic cannabinoids, trends in the popularity of these
compounds can be traced to legislation controlling them. Follow-
ing passage of the Synthetic Drug Abuse Prevention Act of 2012,
the stimulant MDPV was replaced by alpha-PVP; reported “flakka”
use soared between 2014 and 2015; however, after China was
pressured by the USA to ban sale of number of chemicals, including
alpha-PVP, the chemical has disappeared. Methylone was a popular
entactogen among attendees at electronic music festivals but over
time was replaced by ethylone followed by butylone and
dibutylone.
2C-B and 5-MeO-DiPT are just two of the many psychedelic
substances described by Alexander Shulgin in his drug tomes. Of
the novel psychoactive substances that would be considered syn-
thetic hallucinogens, most only have appeared in a handful of cases,
with no one particular substance gaining significant popularity
[10, 11].
Dissociative anesthetics such as methoxetamine and 3-meth-
oxy-phencyclidine are structurally related to ketamine and phency-
clidine and are reported to have similar effects. Though the
mechanism of action is not completely understood, these com-
pounds are antagonists at the NMDA receptor. Antagonism of
this receptor results in hallucinations and dissociation.
4 Challenges of NPS
References
1. Kacinko SL, Papsun D (June 2016) Designer 6. Gurney S (2014) Pharmacology, toxicology,
drugs keep evolving: novel psychoactive sub- and adverse effects of synthetic cannabinoid
stances challenge laboratories and laws. Clinical drugs. Forensic Sci Rev 26(1):53–78
and Forensic Toxicology News (Quarterly, 7. Logan BK, Mohr ALA, Friscia M et al (2017)
AACC/CAP), 1-8. Available from: https:// Reports of adverse events associated with use of
www.aacc.org/publications/clinical-and-foren novel psychoactive substances, 2013–2016: a
sic-toxicology-news review. J Anal Toxicol:1–38
2. Cambridge University Press (1913) The sec- 8. Murphy TD, Weidenbach KN, Van Houten C
ond international opium conference. Am J Int (2013) Acute kidney injury associated with
Law 7:838–847 synthetic cannabinoid use—multiple states,
3. Hoffmann, A. (1996). LSD: Completely Per- 2012. MMWR Morb Mortal Wkly Rep
sonal. Presentation, 1996 Worlds of Con- 62:93–98
sciousness Conference. Available at http:// 9. Drug Enforcement Agency, Office of Diversion
www.maps.org/news-letters/v06n3/ Control, Drug & Chemical Identifcation Sec-
06346hof.html. tion. (2014). Phenazepam. Retrieved from
4. EMCDDA–Europol (2009) Annual Report on http://www.deadiversion.usdoj.gov/drug_
the implementation of Council Decision chem_info/phenazepam.pdf.
2005/387/JHA, European Monitoring Cen- 10. Araújo AM, Carvalho F, Bastos M de L et al
tre for Drugs and Drug Addiction and Euro- (2015) The hallucinogenic world of trypta-
pol, Publications Office of the European mines: an updated review. Arch Toxicol
Union, Luxembourg 89:1151–1173
5. National Drug Early Warning System 11. Dean BV, Stellpflug SJ, Burnett AM et al
(NDEWS) https://www.drugabuse.gov/ (2013) 2C or not 2C: phenethylamine
related-topics/trends-statistics/national-drug- designer drug review. J Med Toxicol
early-warning-system-ndews 9:172–178
Chapter 14
Abstract
The analysis of synthetic cannabinoid compounds in a urine sample is currently one of the more complex
tasks facing toxicologists. The list of prevalent compounds in circulation at any given time is constantly in
flux, changing at a rapid rate to avoid legal control and to a lesser extent to avoid detection. Even with
knowledge of the chemical entities, their detection in urine is complicated by the fact that they are present
exclusively as both phase I metabolites and phase II conjugates. With proper knowledge of the correct
analytical targets, relatively simple procedures are capable of extracting and analyzing synthetic cannabi-
noids. Following enzymatic hydrolysis, compounds can be extracted through liquid partitioning proce-
dures, and the extracts are analyzed via LC-MS/MS.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_14,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
137
138 Gregory C. Janis
2 Materials
JWH-018
JWH-018 Petanoic acid
2.2 Supplies 1. β-Glucuronidase enzyme (E. coli with approximately 140 U/mg
and Equipment of activity at 37 C).
2. Multi-tube vortex (VWR VX-2500 or similar).
3. Heat block for extraction tubes.
4. Nitrogen evaporator, e.g., TurboVap or similar.
142 Gregory C. Janis
3 Methods
3.2 Extraction 1. Pipette 0.5 mL of each sample into labeled 15 mL glass tubes.
2. Add 20 μL of the internal standard spiking solution into each
calibrator, control, and sample tube.
3. Add 500 μL of 50 mM potassium phosphate buffer, pH 6 to
each tube.
4. Add 20 μL of β-glucuronidase enzyme to each tube (see Note 2).
5. Vortex tubes at medium speed for 1 min.
6. Incubate samples at approximately 55 C for 20 min.
7. Remove samples promptly and place at room temperature for
approximately 10 min.
8. Add 1.0 mL of 0.1 N HCl to each tube and vortex mix samples
for 1 min.
Synthetic Cannabinoids 143
Table 1
UPLC gradient
Quantitative
Precursor fragment Collision
Analyte/IS Parent species (m/z) (m/z) RT (min) energy Paired internal standard
UR-144 N-pentanoic acid UR-144/XLR-11 342.2 144.1 6.42 48 UR-144 N-pentanoic acid-D5
Gregory C. Janis
JWH-073 N-butanoic acid JWH-073 358.0 155.0 4.06 30 JWH 073 N-butanoic acid-D5
AB-CHMINACA butanoic acid AB-CHMINACA 358.3 241.1 7.05 25 UR-144 N-pentanoic acid-D5
ADBICA N-(4-hydroxypentyl) ADBICA 360.1 156.1 4.06 30 AM2201 N-(4-hydroxypentyl)-
D5
AB-PINACA pentanoic acid AB-PINACA and 361.1 217.0 1.37 43 RCS-4 N-pentanoic acid-D5
F-AB-PINACA
JWH-018 N-pentanoic acid JWH-018 and AM2201 372.0 155.0 5.39 27 JWH 018 N-pentanoic acid-D4
ADBICA pentanoic acid ADBICA 374.2 244.1 2.01 30 RCS-4 N-pentanoic acid-D5
ADB-PINACA pentanoic acid ADB-PINACA 375.3 245.2 2.01 32 RCS-4 N-pentanoic acid-D5
MAM-2201 N-pentanoic acid MAM-2201 386.2 141.1 6.36 54 JWH 398 N-pentanoic acid-D5
AKB-48 N-pentanoic acid AKB-48 and 5F-AKB-48 396.3 107.1 7.11 62 UR-144 N-pentanoic acid-D5
AB-FUBINACA oxobutanoic acid AB-FUBINACA 399.3 253.3 1.87 28 RCS-4 N-pentanoic acid-D5
MAB-CHMINACA butanoic acid MAB-CHMINACA 372.6 145.0 7.70 52 UR-144 N-pentanoic acid-D5
JWH 073 N-butanoic acid-D5 – 363.2 155.1 3.96 33
RCS-4 N-pentanoic acid-D5 – 357.2 135.1 2.85 45
UR-144 N-pentanoic acid-D5 – 347.3 125.0 6.41 30
AM2201 N-(4-hydroxypentyl)-D5 – 381.3 155.1 4.57 45
JWH 018 N-pentanoic acid-D4 – 376.3 155.1 5.35 30
Intensity, cps
0.00
5.00e5
1.00e6
1.50e6
2.00e6
2.50e6
3.00e6
3.50e6
4.00e6
4.50e6
5.00e6
5.50e6
6.00e6
6.50e6
7.00e6
7.50e6
8.00e6
8.50e6
9.00e6
9.50e6
1.00e7
1.05e7
1.10e7
1.15e7
0.0
ADBICA N-(4-hydroxypentyl)
4.0
4.5
Time, min
JWH-073 N-Butanoic Acid
5.0
JWH-018 N-Pentanoic Acid
5.5
MAM-2201 N-Pentanoic Acid
6.0
6.25
6.5
6.49
UR-144 N-Pentanoic Acid AB-CHMINACA Butanoic Acid
7.0
7.5
AKB-48 N-Pentanoic Acid
8.0
AB-CHMINACA Butanoic Acid
8.5
9.0
145 Synthetic Cannabinoids
146 Gregory C. Janis
4 Notes
References
1. Huffman JW, Dai D, Martin BR, Compton DR the cannabimimetic JWH-018 using chemically
(1994) Design, synthesis and pharmacology of synthesised reference material for the support of
cannabimimetic indoles. Bioorg Med Chem Lett LC-MS/MS-based drug testing. Anal Bioanal
4(4):563–566 Chem 401(2):493–505
2. Beuck S, Möller I, Thomas A, Klose A, 3. Chimalakonda KC, Bratton SM, Le VH, Yiew
Schlörer N, Sch€anzer W, Thevis M (2011) Struc- KH, Dineva A, Moran CL, James LP, Moran JH,
ture characterisation of urinary metabolites of Radominska-Pandya A (2011) Conjugation of
Synthetic Cannabinoids 147
Abstract
Opioids including heroin and commonly prescribed drugs such as oxycodone and fentanyl are among the
most commonly abused drugs. In recent years, the abuse of opioids has spread beyond these commonly
encountered analytes and now includes novel psychoactive drugs such as AH-7921 and U47700 and a
variety of fentanyl-related compounds such as acetyl fentanyl and furanyl fentanyl. The assay described is for
the quantitative determination of 19 designer opioids in serum, plasma, and whole blood. Also included is a
discussion on the challenges of keeping an analytical method current as new analytes appear on the illicit
drug market.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_15,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
149
150 Sherri L. Kacinko and Joseph W. Homan
2 Materials
Table 1
Designer opioid analytes
2.2 Calibrators See Table 1 for information on certified reference materials (CRM)
used to prepare calibrators, internal standard (IS), and controls.
1. 100 ng/μL individual stock standards: Transfer 1.0 mg of
4-ANPP and U-47700, 1.2 mg of U-50488, and 1.1 mg of
the remaining powdered CRM drugs, each to their own 10 mL
volumetric flasks (see Note 1). Dilute to volume with methanol
(MeOH). Store in amber glass vials with Teflon-lined caps.
Unless otherwise noted, all stock solutions are stable for
1 year when stored in appropriate container at < 10 C.
2. 100 ng/μL MT-45 and carfentanil substock standards: Com-
bine 100 μL of each 1000 ng/μL stock of MT-45 or carfentanil
with 900 μL of MeOH. Discard immediately after use.
3. Mixed bulk standard 1: 100 ng/mL Group A drugs, 200 ng/
mL Group B drugs, and 500 ng/mL Group C drug. Transfer
50 μL of each 100 ng/μL Group A drug stock, 10 μL of the
1000 ng/mL AH-7921 stock, 100 μL of each remaining
100 ng/μL Group B drug stock, and 250 μL of the 100 ng/
μL beta-hydroxythiofentanyl stock to a plastic 50 mL volumet-
ric flask, and dilute to volume with drug-free human serum.
Transfer 0.3 mL aliquots to labeled 2.0 mL plastic snap-cap
conical tubes for storage in freezer (see Note 2).
4. Intermediate 10 calibrator 1: Thaw one tube of mixed bulk
standard 1, and vortex briefly to mix. The undiluted mixed
bulk standard is used as 10 calibrator 1.
5. Intermediate 10 calibrator 2: Transfer 200 μL of intermediate
10 calibrator 1 and 200 μL of drug-free serum to a
12 75 mm test tube. Mix well. Discard immediately after
use. Concentrations of the 10 calibrators are shown in
Table 2.
6. Intermediate 10 calibrator 3: Transfer 200 μL of intermediate
10 calibrator 2 and 300 μL of drug-free serum to a
12 75 mm test tube. Mix well. Discard immediately after use.
7. Intermediate 10 calibrator 4: Transfer 200 μL of intermediate
10 calibrator 3 and 600 μL of drug-free serum to a
12 75 mm test tube. Mix well. Discard immediately after use.
8. Intermediate 10 calibrator 5: Transfer 200 μL of intermediate
10 calibrator 4 and 300 μL of drug-free serum to a
12 75 mm test tube. Mix well. Discard immediately after use.
9. Intermediate 10 calibrator 6: Transfer 200 μL of intermediate
10 calibrator 5 and 200 μL of drug-free serum to a
12 75 mm test tube. Mix well. Discard immediately after use.
Designer Opioids in Blood 153
Table 2
Intermediate 10 and working calibrators
Table 3
Preparation of working internal standard solution
Table 4
Concentration of bulk 10 and working low and high controls
Concentration (ng/mL)
Analyte Bulk 10 high Bulk 10 low Working low Working high
4-ANPP 100 3.5 0.35 7.0
4-Methoxybutyryl fentanyl 100 3.5 0.35 7.0
4-Methylphenethyl acetyl fentanyl 100 3.5 0.35 7.0
Acryl fentanyl 100 3.5 0.35 7.0
Alpha-methyl fentanyl 100 3.5 0.35 7.0
Butyryl fentanyl/isobutyryl fentanyl 100 3.5 0.35 7.0
Carfentanil 100 3.5 0.35 7.0
Furanyl fentanyl 100 3.5 0.35 7.0
MT-45 100 3.5 0.35 7.0
Ortho-fluorofentanyl 100 3.5 0.35 7.0
Para-fluorobutyryl fentanyl/FIBF 100 3.5 0.35 7.0
Para-fluorofentanyl 100 3.5 0.35 7.0
Valeryl fentanyl 100 3.5 0.35 7.0
AH-7921 200 7.0 0.7 14
U-47700 200 7.0 0.7 14
U-50488 200 7.0 0.7 14
Beta-hydroxythiofentanyl 500 17.5 1.75 35
3 Methods
3.2 Solid-Phase 1. Place one SPE column for each sample to be extracted in a
Extraction vacuum manifold rack.
2. Condition columns with 2.0 mL of HPLC-grade ACN; aspi-
rate slowly through column.
3. Equilibrate columns with 2.0 mL of DI water; aspirate through
column. Do not allow columns to dry before application of
samples.
4. Transfer prepared samples to columns; aspirate slowly.
5. Rinse columns with 2.0 mL of 0.1 N HCl. Follow by rinsing
with 2.0 mL of ACN; aspirate slowly.
6. Place new, labeled glass tubes beneath each SPE column. Add
2.0 mL of 5% ammonium hydroxide in ACN to elute by
gravitational flow.
Designer Opioids in Blood 157
Table 5
Data acquisition parameters
Time (min) Channel Reaction Dwell (secs) Cone volt. Col. energy
1.400–2.400 1 359.20 > 111.00 0.007 35 34
2 359.20 > 192.10 0.007 35 22
3 364.20 > 111.00 0.007 35 34
4 364.20 > 192.10 0.007 35 22
1.500–2.500 1 395.30 > 113.00 0.007 40 34
2 395.30 > 335.20 0.007 40 18
1.700–2.700 1 365.20 > 105.00 0.006 45 36
2 365.20 > 188.20 0.006 45 24
1 369.30 > 105.10 0.006 50 38
2 369.30 > 188.10 0.006 50 24
1 375.10 > 105.00 0.006 45 40
2 375.10 > 188.10 0.006 45 24
1.800–2.800 1 355.30 > 105.10 0.006 48 38
2 355.30 > 188.10 0.006 48 24
3 360.30 > 105.10 0.006 48 38
4 360.30 > 188.10 0.006 48 24
1 281.20 > 105.00 0.006 35 30
2 281.20 > 188.20 0.006 35 16
1.900–2.900 1 335.20 > 105.00 0.006 45 32
2 335.20 > 188.20 0.006 45 22
1 351.10 > 105.00 0.006 45 36
2 351.10 > 188.00 0.006 45 22
2.000–3.000 1 381.20 > 105.00 0.006 45 38
2 381.20 > 188.20 0.006 45 24
2.100–3.100 1 349.10 > 169.00 0.006 45 18
2 349.10 > 181.00 0.006 45 22
2.200–3.200 1 351.20 > 91.10 0.006 45 38
2 351.20 > 202.10 0.006 45 22
3 354.20 > 91.10 0.006 45 38
4 354.20 > 202.10 0.006 45 22
(continued)
Designer Opioids in Blood 159
Table 5
(continued)
Time (min) Channel Reaction Dwell (secs) Cone volt. Col. energy
2.500–3.500 1 337.20 > 119.00 0.006 45 34
2 337.20 > 202.20 0.006 45 22
3 342.20 > 119.00 0.006 45 34
4 342.20 > 202.20 0.006 45 22
1 323.10 > 105.00 0.006 45 36
2 323.10 > 188.00 0.006 45 22
3 329.10 > 105.00 0.006 45 36
4 329.10 > 188.00 0.006 45 22
3.200–5.400 1 329.20 > 204.10 0.03 45 22
2 329.20 > 284.10 0.03 45 18
3 335.20 > 173.00 0.03 45 28
4 335.20 > 284.10 0.03 45 18
3.600–5.400 1 329.10 > 173.00 0.03 45 26
2 329.10 > 284.00 0.03 45 16
3 334.10 > 178.00 0.03 45 26
4 334.10 > 289.00 0.03 45 16
4.100–5.400 1 369.20 > 112.00 0.03 45 30
2 369.20 > 298.10 0.03 45 20
4 Notes
Figure 2
100
e
%
a c
b
g
f
h i
0
1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 Time
Fig. 1 Multiple reaction monitoring (MRM) chromatogram of extracted whole blood fortified with 10–50 ng/mL
of each compound (a) beta-hydroxythiofentanyl, (b) carfentanil, (c) MT-45, (d) alpha-methyl fentanyl, (e)
4-methylphenethyl acetyl fentanyl, (f) acetyl fentanyl, (g) U-47700, (h) AH-7921, (i) U-50488
100
c f
i
% g
a e
b
h
0
2.20 2.25 2.30 2.35 2.40 2.45 2.50 2.55 2.60 2.65 Time
Fig. 2 Expanded view of Fig. 2 across 2.20–2.70 min (a) valeryl fentanyl, (b)para-fluorobutyryl fentanyl/FIBF,
(c) 2-furanyl fentanyl, (d) ortho-fluorofentanyl, (e) 4-ANPP, (f) butyryl fentanyl/isobutyryl fentanyl, (g) acryl
fentanyl, (h) 4-methoxybutyryl fentanyl, (i) para-fluorofentanyl
Designer Opioids in Blood 161
Table 6
Analyte retention times and monitored MRM transitionsa
Quantifier Qualifier
Analyte Retention time (min) transition transition
Beta-hydroxythiofentanyl 1.92 359.2 > 192.1 359.2 > 111
D5-Beta-hydroxythiofentanyl 1.93 364.2 > 192.1 364.2 > 111.0
Carfentanil 1.94 395.3 > 335.2 395.2 > 113.0
Valeryl fentanyl 2.17 365.2 > 188.2 365.2 > 105.0
Para-fluorobutyryl fentanyl 2.19 369.3 > 188.1 369.3 > 105.1
Furanyl fentanyl 2.22 375.1 > 188.1 375.1 > 105.0
Ortho-fluorofentanyl 2.23 355.3 > 105.1 355.3 > 188.1
4-ANPP 2.28 281.2 > 105.0 281.2 > 188.2
Butyryl fentanyl/isobutyryl fentanyl 2.33 351.1 > 105.0 351.1 > 188.0
4-Methoxybutyryl fentanyl 2.36 381.2 > 105.0 381.2 > 188.2
Para-fluorofentanyl 2.37 355.3 > 105.1 355.3 > 188.1
Acryl fentanyl 2.37 335.2 > 105.0 335.2 > 188.2
D5-Para-fluorofentanyl 2.39 360.3 > 105.1 360.3 > 188.1
MT-45 2.55 349.1 > 181.0 349.1 > 169.0
Alpha-methyl fentanyl 2.62 351.2 > 91.1 351.2 > 202.1
D3-Alpha-methyl fentanyl 2.63 354.2 > 91.1 354.2 > 202.1
4-Methylphenethyl acetyl fentanyl 2.88 337.2 > 119.0 337.2 > 202.2
D5-4-Methylphenethyl acetyl fentanyl 2.90 342.2 > 119.0 342.2 > 202.2
13C6-Acetyl fentanyl 2.96 329.1 > 188.0 329.1 > 105.0
U-47700 3.61 329.2 > 284.1 329.2 > 204.1
D6-U-47700 3.95 335.2 > 284.1 335.2 > 173.0
AH-7921 3.95 329.1 > 284.0 329.1 > 173.0
D3-AH-7921 3.96 334.1 > 289.0 334.1 > 178.0
U-50488 4.63 369.2 > 298.1 369.2 > 112.0
a
Analytes listed below IS used for quantification
References
1. McIntyre IM, Trochta A, Gary RD et al (2016) 5. Kronstrand R, Thelander G, Lindstedt D et al
An acute Butyr-fentanyl fatality: a case report (2014) Fatal intoxications associated with the
with postmortem concentrations. J Anal Tox- designer opioid AH-7921. J Anal Toxicol
icol 40:162–166 38:599–604
2. McIntyre IM, Trochta A, Gary RD et al (2015) 6. Fels H, Krueger J, Sachs H et al (2017) Two
An acute acetyl fentanyl fatality: a case report fatalities associated with synthetic opioids:
with postmortem concentrations. J Anal Tox- AH-7921 and MT-45. Forensic Sci Int 277:
icol 39:490–494 e30–e35
3. McIntyre IM, Gary RD, Joseph S et al (2017) 7. Vorce SP, Knittel JL, Holler JM et al (2014) A
A fatality related to the synthetic opioid fatality involving AH-7921. J Anal Toxicol
U-47700: postmortem concentration distribu- 38:226–230
tion. J Anal Toxicol 41(2):158–160 8. Poklis J, Poklis A, Wolf C et al (2016) Two fatal
4. Fort C, Curtis B, Nichols C et al (2016) Acetyl intoxications involving Butyryl fentanyl. J Anal
fentanyl toxicity: two case reports. J Anal Tox- Toxicol 40:703–708
icol 40:754–757
Designer Opioids in Blood 163
9. Mohr ALA, Friscia M, Papsun D et al (2016) 12. Sofalvi S, Schueler HE, Lavins ES et al (2017)
Analysis of novel synthetic opioids U-47700, An LC-MS-MS method for the analysis of Car-
U-50488 and Furanyl fentanyl by LC–MS/MS fentanil, 3-Methyl fentanyl, 2-Furanyl fentanyl,
in postmortem casework. J Anal Toxicol acetyl fentanyl, fentanyl and Norfentanyl in
40:709–717 postmortem and impaired-driving cases. J
10. Fleming SW, Cooley JC, Johnson L et al Anal Toxicol 41:473–483
(2017) Analysis of U-47700, a novel synthetic 13. Shanks KG, Behonick GS (2017) Detection of
opioid, in human urine by LC-MS-MS and Carfentanil by LC-MS-MS and reports of asso-
LC-QToF. J Anal Toxicol 41:173–180 ciated fatalities in the USA. J Anal Toxicol
11. Papsun D, Krywanczyk A, Vose JC et al (2016) 41:466–472
Analysis of MT-45, a novel synthetic opioid, in 14. Seither J, Reidy L (2017) Confirmation of Car-
human whole blood by LC–MS-MS and its fentanil, U-47700 and other synthetic opioids
identification in a drug-related death. J Anal in a human performance case by LC-MS-MS. J
Toxicol 40:313–317 Anal Toxicol 41:493–497
Chapter 16
Abstract
The increase in the number of new substances appearing on the drug market has been observed in the
1980s and 1990s of the last century, when many phenethylamine and tryptamine derivatives entered the
market. However, the phenomenon of mass marketing of new designer stimulants (being the components
of so-called “legal highs”) began to develop since 2006 in Europe, and it was something new. Since then,
the number of stimulants introduced on the drug market is growing regularly, rapidly, and intensively. Such
a situation creates a need for comprehensive screening methods for detection of these drugs in biological
specimens. The fast and simple liquid chromatography-tandem mass spectrometry qualitative screening
procedure presented here is designed to detect and identify a wide range of designer stimulants in the
blood. The assay has wide applicability for rapid screening of new stimulants in forensic or clinical samples.
The procedure can be easily modified for additional novel psychoactive substances.
Key words Designer stimulants, New psychoactive substances (NPS), Legal highs, Drug screening,
Blood analysis, LC-MS/MS
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_16,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
165
166 Piotr Adamowicz and Bogdan Tokarczyk
2 Materials
2.1 Laboratory 1. Liquid chromatograph coupled with mass detector (e.g., Agi-
Equipment lent Technologies 1200 Series liquid chromatograph with
6460 Triple Quad mass spectrometer).
2. Agilent Technologies SB-C18 chromatography column
(2.1 50 mm, 1.8 μm).
3. Thermoblock with evaporator.
4. Minicentrifuge for vials.
5. Vortexer.
6. Single-channel automatic pipettes.
7. Freezer for temperatures below 15 C.
8. Refrigerator providing a temperature below 4 C.
2.2 Reagents All solvents should be gradient grade or higher (see Note 1).
and Solutions
1. Mobile phase A: 0.1% formic acid in water (v/v). Add 1 mL of
formic acid to 1 L of distilled water (or proportionately smaller
volumes) (see Note 2).
2. Mobile phase B: 0.1% formic acid in acetonitrile (v/v). Add
1 mL of formic acid to 1 L of acetonitrile (or proportionately
smaller volumes).
Designer Stimulants 167
3 Methods
4 Notes
Retention Relative
Precursor Product Fragmentor Collision time retention
Name ion ion voltage [V] energy [V] RT [min] time RRT
2-AI (2-aminoindane) 134.1 117 87 12 1.2 0.48
115 24
91.1 32
2-DPMP (desoxypipradrol) 252.2 167 120 16 5.4 2.16
91.1 24
65.1 60
2-FA/4-FA (2/4-fluoroamphetamine) 154.1 137 85 4 1.6 0.64
109 16
83 44
2,3-DMEC (2,3-dimethylethcathinone) 206.2 188.2 56 8 4.8 1.92
159.2 16
158.2 32
2,4-DMEC (2,4-dimethylethcathinone) 206.2 115.1 68 60 5.1 2.04
91.2 60
72.2 12
3,4-DMMC (3,4-dimethylmethcathinone) 192.1 174.1 85 8 5.03 2.01
159.1 20
3-CMC (3-chloromethcathinone) 198.1 180.1 25 8 2.54 1.02
145.1 16
144.1 32
4-CMC (4-chloromethcathinone) 198.1 180 83 8 2.54 1.02
Designer Stimulants
145.1 16
144.1 36
(continued)
169
170
Table 1
(continued)
Retention Relative
Precursor Product Fragmentor Collision time retention
Name ion ion voltage [V] energy [V] RT [min] time RRT
3-EMC (3-ethylmethcathinone) 192.1 91.2 58 40 4.8 1.92
77.1 52
65.2 60
3-FMC (3-fluoromethcathinone) 182.1 164.1 93 8 1.3 0.52
149 20
148 36
3-FPM (3-fluorophenmetrazine) 196.1 135.1 25 20 2.2 0.88
115.1 32
Piotr Adamowicz and Bogdan Tokarczyk
109.1 24
3-MEC (3-methylethcathinone) 192.1 174.2 62 8 3.4 1.36
145.2 16
144.2 28
4-BMC (brephedrone, 4-bromomethcathinone) 242 145.1 85 12 3.6 1.44
144 36
77.1 60
4-CEC (4-chloroethcathinone) 212.1 194.1 86 8 4.23 1.69
144.1 28
77.1 60
4Cl-α-PVP (4-chloro-α-pyrrolidinovalerophenone) 266.1 125 102 24 5.47 2.19
111 48
74.1 132
4-EEC (4-ethylethcathinone) 206.2 188.2 25 8 4.98 1.99
159.1 16
144.1 28
4-EMC (4-ethylmethcathinone) 192.1 174.1 68 8 4.68 1.87
145.1 20
144.1 36
4-FMA (4-fluoromethamphetamine) 168.1 137.1 62 8 1.85 0.74
109 20
83.1 44
4-FMC (flephedrone, 4-fluoromethcathinone) 182.1 164.1 83 8 1.29 0.52
149 20
148 36
4-MBC (benzedrone) 254.2 236.1 87 8 5.55 2.22
91.1 24
65.1 60
4-MDMC (4-methyldimethcathinone) 192.1 91.2 54 40 2.9 1.16
77.2 60
72.2 28
4-MEAP (NEMNP, 220.2 202.1 78 8 5.25 2.10
4-methyl-α-ethylaminopentiophenone) 144.1 32
91.1 48
4-MEC (4-methylethcathinone) 192.1 174.1 143 8 3.34 1.34
145.1 16
91.1 36
4-MeMABP (4-methylbuphedrone) 192.1 174.2 56 8 4.4 1.76
145.2 20
144.2 32
4-MPD (4-methylpentedrone) 206.2 188.1 80 8 5.11 2.04
144.1 36
77.1 60
4-MPHP (PV-4, 40 -methyl-α-pyrrolidinohexiophenone) 260.2 140.1 104 24 5.72 2.29
Designer Stimulants
105.1 20
91.1 48
171
(continued)
172
Table 1
(continued)
Retention Relative
Precursor Product Fragmentor Collision time retention
Name ion ion voltage [V] energy [V] RT [min] time RRT
4-MTA (4-methylthioamphetamine) 182.1 165 62 4 4.46 1.78
137 20
117 16
5-APB (5-(2-aminopropyl)benzofuran) 176.1 159.1 70 4 3.17 1.27
131.1 16
77 44
5-MAPB (5-(2-methylaminopropyl)benzofuran) 190.1 159.1 68 8 4.0 1.60
131 20
Piotr Adamowicz and Bogdan Tokarczyk
91.1 36
6-APB (6-(2-aminopropyl)benzofuran) 176.1 159.1 64 4 4.04 1.62
131 20
91.1 36
6-EAPB (1-(benzofuran-6-yl)-N-ethylpropan-2-amine) 204.1 159.2 25 8 4.7 1.88
131.1 20
91.1 36
6-IT (6-(2-aminopropyl)indole) 175.1 158.1 60 4 2.53 1.01
130 20
117 24
6-MAPB (6-(2-methylaminopropyl)benzofuran) 190.1 159.2 25 8 4.2 1.68
131.1 16
91.1 36
α-MT (α-methyltryptamine) 175.1 158.1 58 4 2.35 0.94
130 24
117 28
α-PVP (α-pyrrolidinopentiophenone) 232.2 126.1 85 24 4.56 1.82
91 24
77 48
α-PBP (α-pyrrolidinobutiophenone) 218.2 112.1 83 24 2.57 1.03
91.1 20
77.1 48
α-PPP (α-pyrrolidinopropiophenone) 204.1 133 120 16 1.65 0.66
105.1 24
98.1 28
α-PVT (α-pyrrolidinopentiothiophenone) 238.1 126.1 41 20 3.18 1.27
111 36
97 20
BDB (1,3-benzodioxolylbutanamine) 194.1 179.1 143 12 5.1 2.04
164.1 20
77.1 52
bk-DMBDB (dibutylone) 236.1 161 145 16 2.38 0.95
86.1 24
65.1 56
bk-MDDMA (dimethylone) 222.1 147 83 16 1.5 0.60
91.1 36
72.1 16
BMDP (4-methylenedioxy-N-benzylcathinone) 284.1 266.1 87 8 5.18 2.07
91 24
65.1 60
Buphedrone 178.1 160.1 85 4 1.83 0.73
91.1 16
77.1 48
Bupropion 240.1 184 93 4 5.05 2.02
Designer Stimulants
166 12
131.1 24
173
(continued)
174
Table 1
(continued)
Retention Relative
Precursor Product Fragmentor Collision time retention
Name ion ion voltage [V] energy [V] RT [min] time RRT
Butylone 222.1 204.1 91 4 2.14 0.86
174.1 12
131 32
BZP (benzylpiperazine) 177.1 91.1 101 20 0.5 0.20
85.1 12
65.1 52
D2PM (diphenylprolinol) 254.2 236.1 83 8 4.91 1.96
165 56
Piotr Adamowicz and Bogdan Tokarczyk
130 28
Diethylpropion 206.2 105.1 112 16 1.88 0.75
100.1 20
77.1 48
Ephedrone 164.1 146 85 8 1.05 0.42
131 16
77 52
Ethylone 222.1 204.1 89 4 1.72 0.69
174.1 12
146.1 24
Ethylphenidate 248.2 84.1 85 20 5.1 2.04
56.1 56
55.1 56
Ethcathinone 178.1 160.1 89 8 1.31 0.52
131 16
117 28
Eutylone 236.1 218.1 89 8 2.97 1.19
188.1 16
174 32
MDPBP (30 ,40 -methylenedioxy- 262.2 161.1 118 16 3.6 1.44
α-pyrrolidinobutyrophenone) 112.1 24
65.1 60
MDPPP (30 ,40 -methylenedioxy- 248.1 147 91 20 2.08 0.83
α-pyrrolidinopropiophenone) 98.1 24
91.1 48
MDPV (methylenedioxypyrovalerone) 276.2 175.1 124 16 4.9 1.96
135 20
126.1 24
MeBP (methylbuphedrone) 191.2 105.1 99 20 0.74 0.30
79.1 40
77.1 52
MeOPP (para-methoxyphenylpiperazine) 193.1 150 116 16 2.1 0.84
120 36
65.1 60
Mephedrone/2-MMC/3-MMC 178.1 160.1 87 8 2.5 1.00
145.1 20
77.1 56
MePPP (40 -methyl-α-pyrrolidinopropiophenone) 218.2 119.2 54 24 4.1 1.64
98.2 28
91.2 40
Metamfepramone (dimethylcathinone) 178.1 133 85 12 1.2 0.48
105.1 20
72.1 24
Methedrone (4-methoxymethcathinone) 194.1 176.1 85 8 1.8 0.72
Designer Stimulants
161 16
146 28
175
(continued)
176
Table 1
(continued)
Retention Relative
Precursor Product Fragmentor Collision time retention
Name ion ion voltage [V] energy [V] RT [min] time RRT
Mexedrone 208.1 158.2 56 8 3.3 1.32
119.1 20
91.2 40
MDMC (methylone) 208.1 190 91 4 1.56 0.62
160 12
132 24
MPA (methiopropamine) 156.1 125 58 8 1.0 0.40
97 20
Piotr Adamowicz and Bogdan Tokarczyk
58.2 4
MPBP (40 -methyl-α-pyrrolidinobutiophenone) 232.2 161.1 85 12 4.79 1.92
105.1 24
91.1 48
MPHP (40 -methyl-α-pyrrolidinohexiophenone) 260.2 140.2 25 28 5.9 2.36
105.1 24
91.1 52
MPPP (desmethylprodine) 248.2 174.1 97 8 4.61 1.84
44.2 20
42.2 60
Naphyrone 282.2 211.1 126 12 5.76 2.30
155 24
141 20
NEB (N-ethylbuphedrone) 192.1 174.1 68 8 2.25 0.90
130 32
91.1 28
NEH (N-ethylhexedrone) 220.2 130.1 84 36 5.2 2.08
91.1 32
77.1 60
N-ethylpentylone (ephylone) 250.1 232.1 84 8 4.7 1.88
202.2 16
174.1 32
Pentedrone (α-methylaminovalerophenone) 192.1 174.1 89 8 3.91 1.56
132.1 16
91.1 24
Pentylone (methylenedioxypentedrone) 236.1 218.1 62 8 4.4 1.76
188.1 16
175.1 20
PV-7 (α-PHP, α-pyrrolidinohexiophenone) 246.2 140.2 54 24 5.3 2.12
91.2 24
77.2 60
PV-8 (α-PHPP) 260.2 154.2 5 28 5.62 2.25
91.1 24
77.1 60
TFMPP (trifluoromethylphenylpiperazine) 231.1 188 122 16 5.08 2.03
145 44
44.1 20
Mephedrone-D3 (internal standard) 181.1 163.1 87 8 2.5 1.00
Designer Stimulants
177
178 Piotr Adamowicz and Bogdan Tokarczyk
Fig. 1 Example of chromatograms obtained for blood from real forensic cases in which 3-methylethcathinone
(a), 4-chloroethcathinone (b), and N-ethylpentylone (c) were revealed. Subsequent analysis by other quanti-
tative methods determined drug concentrations to be, respectively, 3-MEC, 69 ng/mL; 4-CEC, 49 ng/mL; and
N-ethylpentylone, 196 ng/mL
Designer Stimulants 179
References
1. European Monitoring Centre for Drugs and chromatographic and spectrometric methods.
Drug Addiction (2017) European Drug Report Aust J Forensic Sci 49:637. https://doi.org/
2017: Trends and Developments, Publications 10.1080/00450618.2016.1167240
Office of the European Union, Luxembourg. 3. Ellefsen KN, Concheiro M, Huestis MA (2016)
http://www.emcdda.europa.eu/system/files/ Synthetic cathinone pharmacokinetics, analytical
publications/4541/TDAT17001ENN.pdf. methods, and toxicological findings from
Accessed 11 July 2017 human performance and postmortem cases.
2. Zuba D, Adamowicz P (2016) Distinction of Drug Metab Rev 48(2):237–265. https://doi.
constitutional isomers of mephedrone by org/10.1080/03602532.2016.1188937
180 Piotr Adamowicz and Bogdan Tokarczyk
4. Namera A, Kawamura M, Nakamoto A, Saito T, tandem mass spectrometry. Drug Test Anal 8
Nagao M (2015) Comprehensive review of the (7):652–667
detection methods for synthetic cannabinoids 6. Stone P, Glauner T, Kuhlmann F, Schlabach T,
and cathinones. Forensic Toxicol 33 Miller K (2009) New dynamic MRM mode
(2):175–194 improves data quality and triple quad quantifica-
5. Adamowicz P, Tokarczyk B (2016) Simple and tion in complex analyses. Technical overview.
rapid screening procedure for 143 new psycho- Agilent technologies. https://www.agilent.
active substances by liquid chromatography- com/cs/library/technicaloverviews/Public/
5990-3595en_lo%20CMS.pdf.
Chapter 17
Abstract
Drug screening using high-resolution mass spectrometers, including quadrupole time-of-flight mass ana-
lyzers (QqTOFs), is becoming increasingly popular due to the additional flexibility that these instruments
offer laboratories. Liquid chromatography (LC) coupled to TOF, as in an LC-QqTOF, offers comparable
sensitivity and a shortened method development time relative to triple quadrupole-based mass spectrome-
try. In addition, LC-QqTOF data that are collected in untargeted mode can be analyzed retrospectively to
detect additional compounds that were not predefined targets. Much of the power of LC-QqTOF lies in
data processing, and the data analysis workflow that a lab uses must be adequately validated.
Key words High-resolution mass spectrometry, Time of flight, Drug screen, Toxicology
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_17,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
181
182 Jennifer M. Colby and Kara L. Lynch
2 Materials
Table 1
Example of drug mixes
3 Methods
6. Pipet 100 μL of internal standard into each vial, except for the
double blank.
7. Pipet 200 μL of negative control, positive control, and patient
urine to the correspondingly labeled vials.
8. Cap the vials and vortex to mix.
9. Place the vials in the autosampler tray for testing.
3.2 Analysis 1. Aliquot 250 μL of each serum or sodium heparin plasma speci-
of Serum/Plasma men or control sample using an air displacement pipette with
disposable tips into a 1.5 mL microcentrifuge tube. Add
750 μL of protein dump solution. Pipette up and down to
mix. Cap the tube. Vortex for at least 30 s.
2. Centrifuge specimens for 10 min at 8500 g in a benchtop
centrifuge.
3. Carefully remove samples from the centrifuge, ensuring that
the pellet at the bottom of the tube is not disturbed.
4. Open the tube, and transfer 750 μL of supernatant into a
13 100 mm glass tube. Some supernatant should remain in
the tube.
5. Dry supernatant under nitrogen flow at 37 C for 20 min or
until dry.
6. Label an amber autosampler vial for the negative control,
positive control, each patient specimen, and double blank sam-
ples. Due to the possibility of carryover, a double blank
(no drug, no internal standard) sample must be injected
between each patient sample.
7. Transfer 1000 μL of sample preparation buffer to the double
blank vial. Cap and place in the autosampler tray.
8. Pipet 100 μL of sample preparation buffer into the tubes con-
taining dried control and patient samples, ensuring that the
dried sample is resuspended.
9. Pipet the resuspended patient and control samples into the
appropriate autosampler vials.
10. Cap the vials and place in the autosampler tray for testing.
3.4 Data Analysis 1. Load the data file(s) and premade targeted analysis extraction
ion chromatogram (XIC, Fig. 1) list into MasterView (see
Notes 8 and 9).
5.0e5
4.5e5
4.0e5
3.5e5
3.0e5
Intensity
2.5e5
2.0e5
1.5e5
1.0e5
5.0e4
0.0e0
1 2 3 4 5 6 7 8 9
Time, min
Fig. 1 Extracted-ion chromatogram of a mix of 40 drugs and metabolites. Drug (retention time in minutes):
morphine (2.59), oxymorphone (2.81), hydromorphone (3.01), codeine (3.49), amphetamine (3.69), oxycodone
(3.73), 6-monoacetylmorphine (3.88), hydrocodone (3.88), MDA (3.89), methamphetamine (3.90), desmethyl-
tramadol (3.94), MDMA (4.05), benzoylecgonine (4.5), norfentanyl (4.56), tramadol (4.85),
7-aminoclonazepam (4.96), cocaine (5.11), norbuprenorphine (5.71), fentanyl (6.12), fentanyl-D5 (6.12),
flurazepam (6.33), midazolam (6.38), EDDP (6.49), buprenorphine (6.51), clonazepam (6.73), alpha-
hydroxymidazolam (6.80), alpha-hydroxytriazolam (7.12), nitrazepam (7.15), methadone (7.21), alpha-
hydroxyalprazolam (7.22), oxazepam (7.33), lorazepam (7.43), flunitrazepam (7.44),
2-hydroxyethylflurazepam (7.45), alprazolam (7.57), triazolam (7.58), desalkylflurazepam (7.66), temazepam
(7.77), nordiazepam (7.88), diazepam (8.33)
188 Jennifer M. Colby and Kara L. Lynch
4 Notes
References
1. Thoren KL, Colby JM, Shugarts SB, Wu AHB, Screening new psychoactive substances in urban
Lynch KL (2016) Comparison of information- wastewater using high resolution mass spec-
dependent acquisition on a tandem quadrupole trometry. Anal Bioanal Chem 408:4297
TOF vs a triple quadrupole linear ion trap mass 4. Moschet C, Piazzoli A, Singer H, Hollender J
spectrometer for broad-spectrum drug screen- (2013) Alleviating the Reference standard
ing. Clin Chem 62:170–178 dilemma using a systematic exact mass suspect
2. Bade R, Rousis NI, Bijlsma L, Gracia-Lor E, screening approach with liquid
Castiglioni S, Sancho JV et al (2015) Screening chromatography-high resolution mass spec-
of pharmaceuticals and illicit drugs in wastewater trometry. Anal Chem 85:10312
and surface waters of Spain and Italy by high 5. Colby JM, Thoren KL, Lynch KL (2016) Opti-
resolution mass spectrometry using UHPLC- mization and validation of high-resolution mass
QTOF MS and LC-LTQ-Orbitrap MS. Anal spectrometry data analysis parameters. J Anal
Bioanal Chem 407:8979 Toxicol 1:1–5
3. González-Mariño I, Gracia-Lor E, Bagnati R,
Martins CPB, Zuccato E, Castiglioni S (2016)
Chapter 18
Abstract
Drug testing commonly involves serum, blood, or urine. More recently, alternative specimens for drug
testing have been increasingly used for clinical and forensic toxicology. Examples include oral fluid (saliva),
hair, meconium, and umbilical cord tissue. Each of these matrices has unique properties that provide
advantages for certain applications. Oral fluid has easier and less invasive collection requirements than
urine, the most common specimen for drug screening. Oral fluid drug testing is common in Europe and
steadily gaining popularity in the United States. Hair accumulates drugs and drug metabolites and provides
a much longer window of detection than blood or urine. Meconium and umbilical cord tissue each allow for
assessment of prenatal drug exposure over the course of months. Limitations of these alternative matrices
include need for laboratory-developed tests (exception being some oral fluid immunoassays), challenges
with the specimen matrix, and incomplete understanding of drug incorporation and kinetics. This chapter
briefly describes each of the above alternative specimens in terms of their utility, advantages, and limitations.
Key words Drug screening, Neonatal testing, In utero exposure, Saliva testing, Detection window
1 Introduction
Historically, blood and urine were the mainstay specimens for drug
testing; however, the use of alternative specimens in drug testing
has become increasingly important in clinical and forensic toxicol-
ogy. Some commonly used alternative specimens include oral fluid,
hair, meconium, and umbilical cord tissue. Each of these matrices
has unique qualities which provide advantages in various settings.
This chapter will briefly describe each of the above alternative
specimens in terms of their utility, advantages, and limitations.
Detailed testing methods will be presented in subsequent chapters.
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_18,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
191
192 Kendra L. Palmer and Matthew D. Krasowski
2 Oral Fluid
Oral fluid is the liquid present in the oral cavity and consists
primarily of saliva which is an aqueous solution produced by the
salivary glands. Other minor constituents include oral mucosal
transudate, bacteria, and epithelial cells. Oral fluid, as an alternative
to blood and/or urine, is becoming a more widely used matrix.
While oral fluid is commonly used in Europe, its use in the United
States is still limited, and oral fluid testing is not widely available on
routine chemistry analyzers. In areas where it is utilized, oral fluid is
predominantly used in workplace testing and drug monitoring in
substance abuse programs [1]. Oral fluid has some advantages over
urine, which is another specimen often used in these settings. The
primary advantage to oral fluid testing is that collection is simple
and noninvasive, which eliminates the need for restroom facilities
and makes it possible to collect in remote settings. Oral fluid
collection can be more closely observed than urine collection,
which limits the opportunity for specimen adulteration and also
the need for same-sex collectors. An interesting feature of oral fluid
as a toxicology specimen is that the parent drug is frequently the
predominant substance [2]. For example, following use of heroin,
oral fluid may contain the parent drug itself in addition to the
metabolite 6-monoacetylmorphine (6-MAM). In contrast, heroin
itself is rarely detected in blood or urine; the metabolites 6-MAM
and morphine are much more common in these specimens.
There are some limitations to oral fluid testing. Drug concen-
trations are typically lower than those found in urine. Also, collect-
ing sufficient sample volume to allow for confirmatory testing,
should it be necessary, can be difficult. This is especially problematic
as some drugs, including anti-adrenergic and anticholinergic drugs,
will result in reduced salivation [3]. The oral fluid can be contami-
nated from ingested food or beverages, and drug concentrations
will be higher for drugs that are smoked, inhaled, insufflated, or
taken orally. When testing in a quantitative manner, this may be a
limitation as concentrations will not correlate well with blood con-
centrations. However, when performing qualitative testing, this
may present as an advantage.
Due to the relative newness of this specimen type, analyte
stability has not been well characterized. Similarly, collection device
designs are still being perfected. Some early sample collection
devices caused adsorption of the drug to the device, resulting in
false-negative test results.
Alternate Matrices 193
3 Hair
4 Meconium
6 Summary
References
1. Bosker WM, Huestis MA (2009) Oral fluid (SPME) with GC/MS. Forensic Sci Int 218
testing for drugs of abuse. Clin Chem 55 (1–3):31–36. https://doi.org/10.1016/j.
(11):1910–1931. https://doi.org/10.1373/ forsciint.2011.10.002
clinchem.2008.108670 6. Bar-Oz B, Klein J, Karaskov T, Koren G (2003)
2. Drummer OH (2005) Review: Pharmacoki- Comparison of meconium and neonatal hair
netics of illicit drugs in oral fluid. Forensic Sci analysis for detection of gestational exposure
Int 150(2–3):133–142. https://doi.org/10. to drugs of abuse. Arch Dis Child Fetal Neo-
1016/j.forsciint.2004.11.022 natal Ed 88(2):F98–F100
3. Aps JK, Martens LC (2005) Review: the physi- 7. Cooper GA (2011) Hair testing is taking root.
ology of saliva and transfer of drugs into saliva. Ann Clin Biochem 48(Pt 6):516–530. https://
Forensic Sci Int 150(2–3):119–131. https:// doi.org/10.1258/acb.2011.011112
doi.org/10.1016/j.forsciint.2004.10.026 8. Nakahara Y (1999) Hair analysis for abused
4. Cooper GA, Kronstrand R, Kintz P, Society of and therapeutic drugs. J Chromatogr B
Hair Testing (2012) Society of Hair Testing Biomed Sci Appl 733(1–2):161–180
guidelines for drug testing in hair. Forensic 9. Henderson GL (1993) Mechanisms of drug
Sci Int 218(1–3):20–24. https://doi.org/10. incorporation into hair. Forensic Sci Int 63
1016/j.forsciint.2011.10.024 (1–3):19–29
5. Aleksa K, Walasek P, Fulga N, Kapur B, 10. Stauffer SL, Wood SM, Krasowski MD (2015)
Gareri J, Koren G (2012) Simultaneous detec- Diagnostic yield of hair and urine toxicology
tion of seventeen drugs of abuse and metabo- testing in potential child abuse cases. J Forensic
lites in hair using solid phase micro extraction
Alternate Matrices 197
Abstract
Meconium, the first stool of a newborn, can be analyzed to identify prenatal exposure to drugs of abuse.
Meconium accumulates in a fetus during the second and third trimesters of pregnancy providing a wide
window of exposure. Identification of in utero drug exposure is essential for the diagnosis and treatment of
infants for dependency/withdrawal caused from the exposure. However, testing of meconium samples is
often cumbersome and time-consuming. Unlike liquid samples, meconium is a viscous, semisolid, tar-like
substance that needs to be individually weighed prior to extraction. Additionally, the meconium matrix is
not homogeneous and not easily mixed or extracted. A method for analyzing cocaine and metabolites as
well as amphetamines in meconium utilizing ceramic homogenizers prior to salt-assisted liquid-liquid
extraction and liquid chromatography tandem-mass spectrometry (LC-MS/MS) is presented.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_19,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
199
200 Melissa M. Goggin and Gregory C. Janis
2 Materials
2.1 Solutions and 1. 50:50 methanol/water (v/v): Add 100 mL of methanol and
Standards 100 mL of water to a glass container, cap tightly, and mix. Store
at room temperature for up to 1 year.
2. 1 μg/mL COC-d3 and CE-d3, 3 μg/mL AMP-d8, MAMP-
d14, MDA-d5, and MDMA-d5 internal standard spiking solu-
tion: Combine 100 μL of 1 mg/mL COC-d3 stock and 1 mL
of 0.1 mg/mL CE-d3 stock, plus 300 μL of each 1 mg/mL
stock of AMP-d8, MAMP-d14, MDA-d5, and MDMA-d5 in a
100 mL volumetric flask, and bring to volume with 50:50
methanol/water. Stable up to 1 year stored at 4 C in glass
screw-cap tubes with Teflon septa.
3. 30 μg/mL standard substock and control substock.
(a) Standard substock: Combine 0.3 mL of each 1 mg/mL
stock of AMP, MAMP, MDA, MDMA, BE, and m-OHBE
in a 10 mL volumetric flask. Fill to volume with 50:50
methanol/water. Stable up to 1 year stored at 4 C in glass
screw-cap tubes with Teflon septa.
(b) Control substock: Repeat step 3a with independent pre-
parations of the standard solution stocks.
4. 25 μg/mL AMP, MAMP, MDA, MDMA, BE, and m-OHBE;
12.5 μg/mL COC and CE spiking standard and spiking
control.
202 Melissa M. Goggin and Gregory C. Janis
3 Methods
3.1 Calibrator and 1. Weigh 0.5 g of negative meconium into a labeled 15 mL tube
Control Preparation for each calibrator and control (see Note 1).
2. Spike the tube labeled Standard 1 with 10 μL of the 0.25/
0.05 μg/mL spiking standard (see Note 2). Final concentration
is 5 ng/g AMP, MAMP, BDA, MDMA, BE, m-OHBE, and
1 ng/g COC, CE.
3. Spike the tube labeled Standard 2 with 20 μL of the 0.25/
0.05 μg/mL spiking standard. Final concentration is 10 ng/g
AMP, MAMP, BDA, MDMA, BE, m-OHBE, and 2 ng/g
COC, CE.
4. Spike the tube labeled Standard 3 with 10 μL of the 10/2 μg/
mL spiking standard. Final concentration is 200 ng/g AMP,
MAMP, BDA, MDMA, BE, m-OHBE, and 40 ng/g
COC, CE.
5. Spike the tube labeled Standard 4 with 20 μL of the 10/2 μg/
mL spiking standard. Final concentration is 400 ng/g AMP,
MAMP, BDA, MDMA, BE, m-OHBE, and 80 ng/g
COC, CE.
204 Melissa M. Goggin and Gregory C. Janis
Table 1
Chromatographic gradient parameters
4 Notes
Table 2
Monitored transitions
Fig. 1 (continued)
References
1. SAMHSA (2016) Results from the 2015 isolation of cocaine/crack biomarkers in meco-
national survey on drug use and health: nium. J Chromatogr B 957:14–23
detailed tables. Rockville, MD: Substance 7. Gray TR, Kelly T, LaGasse LL et al (2009)
Abuse and Mental Health Services Administra- Novel biomarkers of prenatal methamphet-
tion. https://www.samhsa.gov/data/sites/ amine exposure in human meconium. Ther
default/files/NSDUH-DetTabs-2015/ Drug Monit 31:70–75
NSDUH-DetTabs-2015/NSDUH-DetTabs- 8. Bordin D, Alves M, Cabices O et al (2014) A
2015.pdf. Accessed 21 June 2017 rapid assay for the simultaneous determination
2. Huestis MA, Choo RE (2002) Drug abuse’s of nicotine, cocaine and metabolites in meco-
smallest victims: in utero drug exposure. nium using disposable pipette extraction and
Forensic Sci Int 128:20–30 gas chromatography-mass spectrometry. J Anal
3. Płotka J, Narkowicz S, Polkowska Ż, Biziuk M, Toxicol 38:31–38
Namieśnik J (2014) Effects of addictive sub- 9. Cabarcos P, Tabernero J Alvarez I et al (2012)
stances during pregnancy and infancy and their A new method for quantifying prenatal expo-
analysis in biological materials. In: Whitacre D sure to ethanol by microwave-assisted extrac-
(ed) Reviews of environmental contamination tion (MAE) of meconium followed by gas
and toxicology, vol 227. Springer International chromatography—mass spectrometry
Publishing, Basel, Switzerland (GC-MS). Anal Bioanal Chem 404:147–155
4. Narkowicz S, Płotka J, Polkowska Ż, Biziuk M, 10. Valente IM, Gonçalves LM, Rodrigues JA
Namieśnik J (2013) Prenatal exposure to sub- (2013) Another glimpse over the salting—out
stance of abuse: a worldwide problem. Environ assisted liquid—liquid extraction in acetoni-
Int 54:141–163 trile/water mixtures. J Chromatogr A
5. Lozano J, Garcia-Algar O, Vall O, Torre R, 1308:58–62
Scaravelli G, Pichini S (2007) Biological matri- 11. Miller AM, Goggin MM, Nguyen A, Gozum
ces for the evaluation of in utero exposure to SD, Janis GC (2017) Profiting from probabil-
drugs of abuse. Ther Drug Monit 29:711–734 ity; combining low and high probability iso-
6. Mantovani Cde C, Lima MB, Oliveira CD, topes as a tool extending the dynamic range
Menck Rde A, Diniz EM, Yonamine M of an assay measuring AMPHETAMINE and
(2014) Development and practical application methamphetamine in urine. J Anal Toxicol
of accelerated solvent extraction for the 41:355–359
Chapter 20
Abstract
In utero exposure to cannabis may cause various short- and long-term health problems in newborns, such as
low birth weight and neonatal withdrawal syndrome. Drug testing with umbilical cord tissue can be used to
identify in utero exposure to cannabis. Here, we described a liquid chromatography-tandem mass spec-
trometry (LC-MS/MS) method that simultaneously quantifies four cannabinoids in umbilical cord tissue,
including tetrahydrocannabinol (THC), 11-nor-Δ9-carboxy-THC (THC-COOH), cannabinol (CBN),
and 11-hydroxy-THC (11-OH-THC). Umbilical cord specimens are weighed and homogenized, and
cannabinoids are extracted using anion exchange solid-phase extraction columns (AX-SPE). Liquid chro-
matography separation is performed, and quantitative results are obtained using LC-MS/MS.
Key words Cannabis exposure, THC, THC-COOH, 11-OH-THC, CBN, Umbilical cord,
LC-MS/MS
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_20,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
211
212 Fang Wu et al.
2 Materials
2.6 Drug-Free 1. Evaluate residual umbilical cord tissue samples using the pre-
Umbilical Cord Pool sented LC-MS/MS method to ensure absence of detectable
cannabinoids. De-identify residual umbilical cord tissue sam-
ples. Pool approximately 80 residual blank umbilical cord
tissue samples. Homogenize drug-free umbilical cord pool
using a mini food chopper (approximately 1/2 cm cubes).
Fill individual 50 mL polypropylene conical centrifuge tubes
(~30 g per tube). Sixty grams of blank umbilical cord tissue is
able to accommodate approximately 1 week of testing if one
run is performed per day. Store in freezer (65 to 75 C);
blank umbilical cord tissue is stable at these temperatures for
at least 1 year and up to two freeze-thaw cycles.
3 Methods
3.1 Sample, 1. Cut umbilical cord tissue using a clean razor blade for each
Calibrator, and Control sample. Discard used razor blades in the sharp container.
Weigh Out Razors should never be reused.
2. Weigh 1.0 0.1 g of each patient umbilical cord tissue (see
Note 3) into individual 5 mL conical polypropylene tubes
with screw top caps.
Fig. 1 Schematic workflow for processing and analyzing umbilical cord tissue
216 Fang Wu et al.
3.2 Calibrator and A calibration curve is generated with the following calibrator con-
Control Spiking centrations: 0.2, 0.5, 1, 3, and 5 ng/g in drug-free umbilical cord
tissue. Three controls are included at 0, 0.25, and 4 ng/g (i.e.,
negative, low, and high) (see Note 5).
1. Spike tube 1 with 20 μL of stock 3 to create calibrator
1 (0.2 ng/g).
2. Spike tube 2 with 50 μL of stock 3 to create calibrator
2 (0.5 ng/g).
3. Spike tube 3 with 10 μL of stock 2 to create calibrator
3 (1 ng/g).
4. Spike tube 4 with 30 μL of stock 2 to create calibrator
4 (3 ng/g).
5. Spike tube 5 with 50 μL of stock 2 to create calibrator
5 (5 ng/g).
6. Spike tube 6 with 25 μL of stock 6 to create a low QC
(0.25 ng/g).
7. Spike tube 7 with 40 μL of stock 5 to create a high QC
(4 ng/g).
8. Spike tube 8 with stock 7 (internal standard) only. Tube
8 without non-deuterated standard solution is used as a
negative QC.
3.3 Homogenization 1. Add six stainless steel beads into each tube.
and Base Hydrolysis 2. Add 2.5 mL of methanol into each tube.
3. Store samples in freezer (65 to 75 C) for 10 min.
4. Remove samples from freezer and immediately place them in a
homogenizer.
5. Homogenize samples for 4 min at 5 m/s in 30 s intervals with
30 s rest periods to minimize heat exposure to the tissue.
6. Centrifuge homogenates at 20,598 g (14,000 rpm) and
4 C for 10 min in a microcentrifuge.
7. Transfer supernatants to individual pre-labeled 10 mL culture
tubes.
8. Add 2.5 mL of 0.5 M NaOH into each supernatant.
Detection of In Utero Exposure to Cannabis 217
Table 1
Liquid chromatography separation gradienta
Time (min) %A %B
0 40 60
0.5 40 60
1.2 20 80
2.3 20 80
2.7 10 90
3.2 10 90
3.21 5 95
4 5 95
4.01 40 60
4.7 40 60
a
A, 5 mM ammonium bicarbonate in water, pH 9.5; B, methanol; flow rate used is
0.5 mL/min
Table 2
Mass spectrometry parameters
3.6 Data Analysis 1. Construct calibration curves via linear least-squares regression
with 1/x weighting factor.
2. Review calibrators (see Note 10). Review calibrators for any
missing internal standards. Review retention times (RTs) for
Detection of In Utero Exposure to Cannabis 219
Table 3
Drug testing components, multiple reaction monitoring transitions, retention time (RT), and internal
standardsa
analytes and internal standards for any flags, and review inte-
gration and chromatogram quality.
3. Review QCs. QC values should be within 20% of the target
concentration (see Note 11).
4. Review patient samples (see Note 12).
4 Notes
Fig. 2 Representative LC separation (a) and extracted ion chromatograms with ion transitions noted for (b) THC
(1.3 ng/g), (c) THC-COOH (17.5 ng/g), (d) 11-OH-THC (0.9 ng/g), and (e) CBN (0.3 ng/g) from an authentic
clinical sample
Acknowledgments
This work was supported by the ARUP Institute for Clinical and
Experimental Pathology.
Disclosure: The authors have no potential conflicts of interest.
References
1. Huestis MA, Choo RE (2002) Drug abuse’s 4. Jutras-Aswad D, DiNieri JA, Harkany T, Hurd
smallest victims: in utero drug exposure. YL (2009) Neurobiological consequences of
Forensic Sci Int 128(1–2):20–30 maternal cannabis on human fetal development
2. Substance AbuseCenter for Behavioral Health and its neuropsychiatric outcome. Eur Arch Psy-
Statistics and Quality. Results from the 2015 chiatry Clin Neurosci 259(7):395–412. https://
National Survey on Drug Use and Health: doi.org/10.1007/s00406-009-0027-z
Detailed Tables SAMHSA. https://www. 5. Calvigioni D, Hurd YL, Harkany T, Keimpema
samhsa.gov/data/sites/default/files/ E (2014) Neuronal substrates and functional
NSDUH-DetTabs-2015/NSDUH-DetTabs- consequences of prenatal cannabis exposure.
2015/NSDUH-DetTabs-2015pdf. Published Eur Child Adolesc Psychiatry 23(10):931–941.
September 8, 2016. Accessed 18 Jan 2017 https://doi.org/10.1007/s00787-014-0550-y
3. Sithisarn T, Granger DT, Bada HS (2012) 6. Behnke M, Smith VC, Committee on
Consequences of prenatal substance use. Int J Substance A, Committee on F, Newborn
Adolesc Med Health 24(2):105–112. https:// (2013) Prenatal substance abuse: short- and
doi.org/10.1515/ijamh.2012.016 long-term effects on the exposed fetus.
222 Fang Wu et al.
Abstract
In utero exposure to alcohol may adversely affect the development of the embryo or fetus and result in
adverse outcomes known as fetal alcohol spectrum disorders (FASD) which encompass a range of physical,
behavioral, and cognitive impairments in the newborn. Since maternal self-reports are often unreliable,
biomarkers of gestational alcohol consumption are necessary for accurate identification of exposed
newborns at risk. Ethyl-β-D-glucuronide (EtG) is a minor phase II metabolite of ethyl alcohol (ethanol),
formed by enzymatic conjugation of ethanol with glucuronic acid in the liver. As a direct biomarker for
alcohol, detection of EtG in neonatal biological matrices provides accurate identification of maternal
alcohol consumption and fetal alcohol exposure during pregnancy. This chapter describes the quantitation
of EtG in human umbilical cord tissue by liquid chromatography tandem mass spectrometry (LC-MS/MS).
Key words Ethyl glucuronide (EtG), LC-MS/MS, Prenatal alcohol exposure, Umbilical cord, New-
born drug testing
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_21,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
223
224 Simuli L. Wabuyele and Gwendolyn A. McMillin
2 Materials
Label all reagents, calibrators, and controls with contents, lot num-
ber, date prepared, expiration dates, and initials of preparer. Use LC
grade or LC-MS grade reagents and follow guidelines/regulations
for proper handling, storage, and disposal of hazardous waste. Note
that similar equipment and supplies to those listed here can be used
as available.
10. Synergi™ Hydro RP, C18 column, 50 mm 3.0 mm, 2.5 μm.
11. Ultra HPLC In-Line Filter, 0.5 μm depth filter 0.00400 ID.
12. 5.6 mm stainless steel UFO beads for Biotage® Bead Ruptor.
13. Mini food chopper/processor, 1.5-cup capacity.
14. Standard glassware, pipette tips, and other supplies.
2.3 Samples (see 1. Patient specimen: Collect at least 6 in. of umbilical cord for
Note 2) testing. Excess cord blood should be drained and exterior of
the cord segment rinsed with saline solution or water and
patted dry.
2. Pooled drug-free (blank) umbilical cord: De-identify residual
umbilical cord tissue specimens and screen for the presence of
EtG using the LC-MS/MS method described here. Pool about
80–100 drug-free human umbilical cord samples. Using the
mini food chopper/processor, cut the umbilical cords into
approximately 0.5 cm cubes. Aliquot the pooled umbilical
cord tissue into 50 mL polypropylene tubes (~30 g per tube).
Sixty grams of blank cord is able to accommodate approxi-
mately 1 week of testing if one run is performed per day. Pooled
drug-free (blank) umbilical cord is stable at freezer (65 to
80 C) temperatures for at least 1 year and up to two freeze-
thaw cycles.
2.4 Standards and 1. Standard stock solution: 50 μg/mL EtG in methanol. Add
Calibrators 500 μL of 1.0 mg/mL EtG to 5 mL of methanol in a 10 mL
Class A volumetric flask. Fill to volume with methanol and
mix well.
Detection of In Utero Alcohol Exposure 227
Table 1
Concentration of calibrators (Cal 1–6) and quality controls (QCs)
2.5 Quality Controls 1. QC standard stock solution: 50 μg/mL EtG in methanol. Add
(QCs) and Internal 500 μL of 1.0 mg/mL EtG (different lot number) to 5 mL of
Standard methanol in a 10 mL Class A volumetric flask. Fill to volume
with methanol and mix well (see Note 4).
2. QC working solution 4: 3.30 μg/mL EtG. Add 660 μL of QC
standard stock solution to 5 mL of methanol in a 10 mL Class
A volumetric flask. Fill to volume with methanol and mix well
(see Note 5 and Table 1).
3. QC working solution 3: 0.600 μg/mL EtG. Add 1.0 mL of
QC working solution 4 to 4.5 mL of methanol and mix well.
228 Simuli L. Wabuyele and Gwendolyn A. McMillin
3 Methods
3.1 Sample 1. Label a series of 7.0 mL polypropylene screw cap tubes, a series
Preparation of 16 100 mm, 14 mL culture tubes, and a series of auto-
sampler vials with matching identifiers for calibrators, controls,
and patient specimens (i.e., tubes 1–6 for calibrators, tubes
7–10 for the controls, and the other tubes for patient samples).
2. Using a clean razor blade, cut approximately 0.5 cm cubes of
patient cord specimen (see Note 9).
3. Accurately weigh 1 0.025 g of each patient cord specimen
(0.975–1.025 g). Transfer to the appropriate pre-labeled
7.0 mL polypropylene screw cap tubes. Aliquot each patient
cord specimen using a new, clean wooden stir stick for each
specimen.
4. Accurately weigh 1 0.025 g of drug-free umbilical cord
(0.975–1.025 g) to pre-labeled 7.0 mL polypropylene screw
cap tubes 1–10 for calibrators and controls (see Note 10).
5. Add 50 μL of the internal standard working solution to each
tube (see Note 11). It is acceptable to use a repeating pipette.
Detection of In Utero Alcohol Exposure 229
3.2 Solid-Phase 1. Place the SPE columns on a positive pressure manifold in the
Extraction (SPE) proper sequence in which they are labeled. SPE is by gravity
flow only. Do not apply any pressure to the manifold.
2. Condition the SPE columns with 4 mL of methanol using
gravity flow.
230 Simuli L. Wabuyele and Gwendolyn A. McMillin
Table 2
LC gradient conditions
Table 3
Autosampler parameters
Parameter Value
Loop volume 1 (μL) 20
Loop volume 2 (μL) 20
Syringe type 100 μL DLW
Syringe volume (μL) 20
Injection volume (μL) 10.0
Cycle name Analyst LC-Inj DLW Fast_Rev05
Airgap volume (μL) 3
Front volume (μL) 5
Rear volume (μL) 5
Filling speed (μL/s) 5
Pullup delay (ms) 3
Inject to LC Vlv1
Injection speed (μL/s) 5
Pre inject delay (ms) 500
Post inject delay (ms) 500
Needle gap valve clean (mm) 3
Valve clean time solvent 2 (s) 3
Valve clean time solvent 1 (s) 3
Post clean time solvent 1 (s) 2
232 Simuli L. Wabuyele and Gwendolyn A. McMillin
Table 4
Diverter valve parameters
Table 5
MS source parameters
Parameter Value
CUR (Curtain gas, psi) 35
CAD (Collisions gas, psi) 10
TEM (Temperature, C) 600
GS1 (Nebulizer gas, psi) 50
GS2 (Heater gas, psi) 50
IS (IonSpray voltage, V) 3500
Declustering potential (V) 90
Dwell time (ms) 80
Resolution Unit mass (Q1 and Q3)
Table 6
MRM transitions and MS compound-dependent parameters
Fig. 2 The representative mass chromatograms for EtG and internal standard in (a) calibrator 1 (5 ng/g) and (b)
internal standard (25 ng/g); (c) positive patient specimen (EtG ¼ 62.9 ng/g) and (d) internal standard (25 ng/g)
ion mass ratios for EtG and IS for any flags. Verify the linear
calibration range for the method (see Note 15).
11. Review patient data. For the patient data to be reported, all
qualitative and quantitative criteria for the analyte and internal
standard established must be met.
4 Notes
Acknowledgments
This work was supported by the ARUP Institute for Clinical and
Experimental Pathology.
Disclosure: The authors have no potential conflict of interest.
References
1. Forray A (2016) Substance use during preg- 7. Dinis-Oliveira RJ (2016) Oxidative and
nancy. F1000Res 5:F1000 Faculty non-oxidative metabolomics of ethanol. Curr
Rev–F1000 Faculty1887 Drug Metab 17:327–335
2. Center for Behavioral Health Statistics and 8. Walsham NE, Sherwood RA (2012) Ethyl glu-
Quality (2016). 2015 National Survey on drug curonide. Ann Clin Biochem 49:110–117
use and health: detailed tables. Substance Abuse 9. Foti RS, Fisher MB (2005) Assessment of
and Mental Health Services Administration, UDP-glucuronosyltransferase catalyzed forma-
Rockville, MD. https://www.samhsa.gov/ tion of ethyl glucuronide in human liver micro-
data/sites/default/files/NSDUH-DetTabs- somes and recombinant UGTs. Forensic Sci Int
2015/NSDUH-DetTabs-2015/NSDUH- 153:109–116
DetTabs-2015.pdf. Accessed July, 13 10. Bager H, Christensen LP, Husby S et al (2017)
3. Wilhelm CJ, Guizzetti M (2015) Fetal alcohol Biomarkers for the detection of prenatal alco-
Spectrum disorders: an overview from the glia hol exposure: a review. Alcohol Clin Exp Res
perspective. Front Integr Neurosci 9:65 41:251–261
4. Svetlana P, Shannon L, Larry B, et al (2016) 11. Dahl H, Hammarberg A, Franck J et al (2011)
Burden and social cost of fetal alcohol spec- Urinary ethyl glucuronide and ethyl
trum disorders. In: Oxford Handbooks Online sulfate testing for recent drinking in alcohol-
(ed). Oxford, Oxford University Press. http:// dependent outpatients treated with
www.oxfordhandbooks.com/view/10.1093/ acamprosate or placebo. Alcohol Alcohol
oxfordhb/9780199935291.001.0001/ 46:553–557
oxfordhb-9780199935291-e-78. Accessed 12. Morini L, Marchei E, Tarani L et al (2013)
15 Jul 2017 Testing ethylglucuronide in maternal hair and
5. Paley B, O’Connor MJ (2009) Intervention nails for the assessment of fetal exposure to
for individuals with fetal alcohol spectrum dis- alcohol: comparison with meconium testing.
orders: treatment approaches and case manage- Ther Drug Monit 35:402–407
ment. Dev Disabil Res Rev 15:258–267 13. Pichini S, Morini L, Pacifici R et al (2014)
6. Lange S, Shield K, Koren G et al (2014) A Development of a new immunoassay for the
comparison of the prevalence of prenatal alco- detection of ethyl glucuronide (EtG) in meco-
hol exposure obtained via maternal self-reports nium: validation with authentic specimens ana-
versus meconium testing: a systematic litera- lyzed using LC-MS/MS. Preliminary results.
ture review and meta-analysis. BMC Pregnancy Clin Chem Lab Med 52:1179–1185
Childbirth 14:127–127
236 Simuli L. Wabuyele and Gwendolyn A. McMillin
14. Himes SK, Dukes KA, Tripp T et al (2015) 17. Joya X, Friguls B, Ortigosa S et al (2012)
Clinical sensitivity and specificity of meconium Determination of maternal-fetal biomarkers of
fatty acid ethyl ester, ethyl glucuronide, and prenatal exposure to ethanol: a review. J Pharm
ethyl sulfate for detecting maternal drinking Biomed Anal 69:209–222
during pregnancy. Clin Chem 61:523–532 18. Montgomery D, Plate C, Alder SC et al (2006)
15. Morini L, Falcon M, Pichini S et al (2011) Testing for fetal exposure to illicit drugs using
Ethyl-glucuronide and ethyl-sulfate in placen- umbilical cord tissue vs meconium. J Perinatol
tal and fetal tissues by liquid chromatography 26:11–14
coupled with tandem mass spectrometry. Anal 19. Montgomery DP, Plate CA, Jones M et al
Biochem 418:30–36 (2008) Using umbilical cord tissue to detect
16. Jones J, Jones M, Plate C, Lewis D (2012) The fetal exposure to illicit drugs: a multicentered
detection of 1-palmitoyl-2-oleoyl-sn-glycero- study in Utah and New Jersey. J Perinatol
3-phosphoethanol and ethyl glucuronide in 28:750–753
human umbilical cord. Am J Analyt Chem
3:800–810
Chapter 22
Abstract
Oral fluid analysis for drugs is increasingly used in a variety of testing areas: pain management and
medication monitoring, parole and probation situations, driving under the influence of drugs (DUID),
therapeutic drug monitoring, and testing for drugs in the workplace. The sample collection itself is
straightforward, rapid, observable, and noninvasive, requiring no special facilities (compared to urine) or
medical personnel (compared to blood). The pH of saliva is slightly acidic relative to blood; therefore, drugs
which are more basic tend to be present in higher concentration in oral fluid than in blood: cocaine,
amphetamines, oxycodone, tramadol, buprenorphine, methadone, and fentanyl. Conversely, acidic drugs
and drugs which are strongly protein bound have lower concentrations in oral fluid than in blood: examples
include benzodiazepines, barbiturates, and carisoprodol. Because of the low volume of specimen available
for analysis and the drug concentrations present (generally much lower than those in urine), efficient
extraction methods and sensitive confirmation procedures are necessary for routine analysis of drugs in oral
fluid. In this chapter, solid-phase extraction methods are described for a variety of drugs with liquid
chromatography—tandem mass spectrometry detection.
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_22,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
237
238 Cynthia A. Coulter and Christine M. Moore
2 Materials
3 Methods
3.1 Extraction Basic drugs (e.g., amphetamines, cocaine, etc.) diffuse into saliva
and Analysis of Basic from the blood relatively easily. A procedure for the extraction of
Drugs cocaine and its metabolites from oral fluid using a dilution factor of
four is described below. The general method can be applied to
other basic drug classes for extraction.
3.1.1 Preparation 1. Label borosilicate glass test tubes for Calibrators 1–5, each
of Calibrators, Controls, quality control, and specimens.
and Samples 2. Mix 2 mL of synthetic oral fluid with 6 mL of Quantisal®
extraction buffer (or other buffers depending on collection
device).
3. Pipette 1 mL of this mixture into a borosilicate glass test tube
for each calibrator and control (unless using external QC) to
be used.
4. Pipette 1 mL of each sample to be tested into its own borosili-
cate glass test tube.
5. Add 40 μL of each 100 ng/mL internal standard (BZE-d3,
COC-d3, and CE-d8) to each calibrator, control, and
specimen.
6. Do not spike any drug standards into the tube labeled Calibra-
tor 1. This is the negative standard.
7. Spike 10 μL of each 100 ng/mL drug standard (BZE and
COC/CE) into the tube labeled Calibrator 2, to give a final
concentration of 4 ng/mL.
8. Spike 20 μL of each 100 ng/mL drug standard (BZE and
COC/CE) into the tube labeled Calibrator 3, to give a final
concentration of 8 ng/mL.
9. Spike 40 μL of each 100 ng/mL drug standard (BZE and
COC/CE) into the tube labeled Calibrator 4, to give a final
concentration of 16 ng/mL.
10. Spike 80 μL of each 100 ng/mL drug standard (BZE and
COC/CE) into the tube labeled Calibrator 5, to give a final
concentration of 32 ng/mL (see Note 5).
11. Add 1 mL of 0.1 M potassium phosphate pH 6.0 to each tube
as a buffer, and mix well (see Note 6).
3.1.2 Extraction 1. Use one 3 mL, 35 mg CEREX Clin II extraction column per
calibrator, control, and sample. Place each column in the posi-
tive pressure manifold with the pressure setting no higher than
10 psi.
2. Condition the columns by adding 2 mL of methanol to each.
Allow the methanol to flow through the column bed at a rate
no greater than 1 mL/min or one drop at a time. Do not allow
246 Cynthia A. Coulter and Christine M. Moore
Table 1
MRM transitions for BZE, COC, CE, and their deuterated analogs
Compound Precursor ion (m/z) Product ion (m/z) Fragmentor voltage (V) Collision energy (V)
BZE-d3 293.3 171.2 120 20
BZE 290.3 168.1 120 15
BZE 290.3 105.1 120 15
COC-d3 307.3 185.3 120 20
COC 304.3 182.3 120 20
COC 304.3 82.3 120 25
CE-d8 326.3 204.4 160 20
CE 318.3 196.4 120 25
CE 318.3 82.2 120 25
3.2.1 Preparation 1. Label borosilicate glass test tubes for Calibrators 1–5, each
of Calibrators, Controls, quality control, and specimens.
and Samples 2. Mix 2 mL of synthetic oral fluid with 6 mL of Quantisal®
extraction buffer (or other buffers depending on collection
device).
248 Cynthia A. Coulter and Christine M. Moore
3.2.2 Extraction 1. Use one 3 mL, 15 mg Trace-N extraction column per calibra-
tor, control, and sample. Place each column in the positive
pressure manifold with the pressure setting no higher than
10 psi.
2. Condition the columns by adding 500 μL of methanol to each
one. Allow the methanol to flow through the column bed at a
rate no greater than 1 mL/min or one drop at a time. Do not
allow the column bed to become dry once the conditioning has
begun (see Note 7).
3. Add 100 μL of 0.1 M acetic acid to each column. Allow the
0.1 M acetic acid to flow through the column bed at the same
rate as the methanol. Do not allow the column to become dry
before loading the sample.
4. Pour each calibrator, control, and sample onto the appropriate
column. Allow the sample to load on the column bed without
the use of positive pressure.
5. Once the samples have completely loaded, wash the columns
with 1 mL of Step 1 wash solution. Positive pressure with
nitrogen can be used up to a pressure of 10 psi.
Drugs in Oral Fluid 249
Table 2
MRM transitions for THC, CBN, and CBD and their deuterated analogs
Compound Precursor ion (m/z) Product ion (m/z) Fragmentor voltage (V) Collision energy (V)
THC-d3 318.3 196.3 150 20
THC 315 193 120 20
THC 315 123 120 30
CBN 311.2 223.1 120 20
CBN 311.2 195.1 120 25
CBD 315 193 120 20
CBD 315 123 120 30
3.3.2 Extraction 1. Use one 3 mL, 15 mg Trace-N extraction column per calibra-
of THC-COOH tor, control, and sample. Place each column in the positive
pressure manifold with the pressure setting no higher than
10 psi.
2. Condition the columns by adding 500 μL of methanol to each.
Allow the methanol to flow through the column bed at a rate
no greater than 1 mL/min or one drop at a time. Do not allow
the column bed to become dry once the conditioning has
begun (see Note 7).
3. Add 100 μL of 0.1 M acetic acid to each column. Allow the
0.1 M acetic acid to flow through the column bed at the same
rate as the methanol. Do not allow the column to become dry
before loading the sample.
4. Pour each calibrator, control, or sample onto the appropriate
column. Allow the sample to load on the column bed without
the use of positive pressure.
5. Once the samples have completely loaded, wash the columns
with 1 mL of Step 1 wash solution, allowing the wash to flow at
a rate of 1 mL/min. Positive pressure with nitrogen can be
used up to a pressure of 10 psi.
252 Cynthia A. Coulter and Christine M. Moore
Table 3
MRM transitions for THC-COOH, THC, CBN, and CBD and their deuterated analogs
3.3.5 Extraction 1. Use one 3 mL, 35 mg CEREX Clin II extraction column per
for Chiral Separation calibrator, control, and sample. Place each column in the posi-
of AMP and METH tive pressure manifold with the pressure setting no higher than
10 psi.
2. Condition the columns by adding 2 mL methanol to each.
Allow the methanol to flow through the column bed at a rate
no greater than 1 mL/min or one drop a time. Do not allow
the column bed to become dry once the conditioning has
begun (see Note 7).
3. Add 2 mL of 0.1 M sodium phosphate buffer pH 6.0 to each
column. Allow the buffer to flow through the column bed at
the same rate as the methanol. Do not allow the column to
become dry before loading the sample.
4. Pour each calibrator, control, or sample onto the appropriate
column. Allow the sample to load on the column bed without
the use of positive pressure.
5. Once the samples have completely loaded, wash the columns
with 2 mL of DI water allowing the water to flow at a rate of
1 mL/min. Positive pressure with nitrogen can be used up to a
pressure of 10 psi.
6. Add 2 mL of 0.1 M hydrochloric acid to each column, allowing
to flow at the same rate as DI water.
7. Dry the columns under nitrogen using positive pressure at
30 psi for 1 min.
Drugs in Oral Fluid 255
Table 4
MRM transitions for AMP and METH and their deuterated analogs
Compound Precursor ion (m/z) Product ion (m/z) Fragmentor voltage (V) Collision energy (V)
AMP-d5 393.5 96 80 10
AMP 388.5 119 80 10
AMP 388.5 91 80 35
METH-d5 407.5 92 80 40
METH 402.4 119 80 10
METH 402.4 91 80 30
4 Notes
References
1. Gallardo E, Barroso M, Queiroz JA (2009) zolpidem in oral fluid and its application to
Current technologies and considerations for authentic samples from regular drug users. J
drugs bioanalysis in oral fluid. Bioanalysis 1 Pharm Biomed Anal 74:213–222
(3):637–667 8. Langel K, Gunnar T, Ariniemi K, Rajam€aki O,
2. Vindenes V, Yttredal B, Oiestad EL, Waal H, Lillsunde P (2011) A validated method for the
Bernard JP, Mørland JG, Christophersen AS detection and quantitation of 50 drugs of
(2011) Oral fluid is a viable alternative for abuse and medicinal drugs in oral fluid by gas
monitoring drug abuse: detection of drugs in chromatography-mass spectrometry. J Chro-
oral fluid by liquid chromatography-tandem matogr B Analyt Technol Biomed Life Sci
mass spectrometry and comparison to the 879(13–14):859–870
results from urine samples from patients trea- 9. Coulter C, Garnier M, Moore C (2012) Anal-
ted with methadone or buprenorphine. J Anal ysis of tetrahydrocannabinol and its metabolite,
Toxicol 35(1):32–39 11-nor-d9-tetrahydrocannabinol-9-carboxylic
3. Moore C (2015) Drug testing and adherence acid, in oral fluid using liquid chromatography
monitoring in pain management: Oral fluid with tandem mass spectrometry. J Anal Toxicol
testing. J Opioid Manag 11(1):69–75 36(6):413–417
4. Crouch DJ (2005) Oral fluid collection: the 10. Newmeyer MN, Concheiro M, da Costa JL,
neglected variable in oral fluid testing. Forensic Flegel R, Gorelick DA, Huestis MA (2015)
Sci Int 150(2–3):165–173 Oral fluid with three modes of collection and
5. Coucke LD, De Smet L, Verstraete AG (2016) plasma methamphetamine and amphetamine
Influence of sampling procedure on codeine enantiomer concentrations after controlled
concentrations in oral fluid. J Anal Toxicol 40 intranasal l-methamphetamine administration.
(2):148–152 Drug Test Anal 7(10):877–883
6. Valen A, Leere Øiestad ÅM, Strand DH, 11. Hughes J, Tuyay J, Coulter C, Moore C
Skari R, Berg T (2016) Determination of (2013) Drugs and metabolites in oral fluid:
21 drugs in oral fluid using fully automated Immunoassay screening and LC/MS-MS con-
supported liquid extraction and UHPLC- firmation and quantification. www.agilent.com
MS/MS. Drug Test Anal. https://doi.org/ (Agilent Technologies)
10.1002/dta.2045 12. Tuyay J, Coulter C, Rodrigues W, Moore C
7. Jang M, Chang H, Yang W, Choi H, Kim E, Yu (2012) Disposition of opioids in oral fluid:
BH, Oh Y, Chung H (2013) Development of Importance of chromatography and mass spec-
an LC-MS/MS method for the simultaneous tral transitions in LC-MS/MS. Drug Test Anal
determination of 25 benzodiazepines and 4(6):395–401
Chapter 23
Abstract
Cocaine is one of the most common illegal drugs in Europe and other parts of the world. It is mainly
metabolized to benzoylecgonine and ecgonine methyl ester but also to minor metabolites such as norco-
caine and meta-hydroxy-benzoylecgonine. If ethanol is consumed simultaneously, cocaethylene is formed.
Dried blood spots (DBS) are a minimally invasive sampling technique producing easy-to-ship specimens
and have garnered increasing attention in forensic and clinical contexts in recent years. We hereby present a
liquid chromatography/tandem mass spectrometry-based (LC-MS/MS) method for the quantification of
cocaine, benzoylecgonine, ecgonine methyl ester, norcocaine, meta-hydroxy-benzoylecgonine, and
cocaethylene in DBS.
Key words Dried blood spots, Cocaine, Benzoylecgonine, Ecgonine methyl ester, Norcocaine,
Cocaethylene, Meta-hydroxy-benzoylecgonine, HPLC-MS/MS
1 Introduction
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5_23,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
261
262 Lars Ambach and Christophe Stove
O O O
H 3C CH3 H3C H3C CH3
N O N OH N O
O O OH
O O
cocaine benzoylecgonine ecgonine methyl ester
O O O
CH3 H3 C H3 C
CH3
HN O N OH N O
OH
O O O
O O O
norcocaine meta-hydroxy-benzoylecgonine cocaethylene
2 Materials
2.1 Prepared All solvents and reagents are of LC-MS or analytical grade.
Reagents
1. Stock solutions: 1 mg/mL COC, nor-COC, COC-Et, m-OH-
BE; 0.1 mg/mL BE, EME, and internal standards (IS) co-
caine-D3, benzoylecgonine-D8, ecgonine methyl ester-D3,
norcocaine-D3, cocaethylene-D8 reference standards in sealed
glass ampoules. Open, and transfer the contents of the
ampoules into glass vials with a plastic screw cap and rubber/
Teflon septum, and wrap in parafilm. Store vials at 20 C.
2. Blank blood for calibration and quality control (QC) samples:
Obtain blood from healthy volunteers using collection tubes
with sodium fluoride and potassium oxalate (NaF/K2Ox).
Simultaneously collect one ethylenediaminetetraacetic acid
(EDTA) tube of blood and determine the HCT value on a
standard clinical analyzer. Based on the measured HCT, adjust
the HCT of the NaF/K2Ox-stabilized blood to 40% by adding
or removing plasma to or from the blood (see Notes 1 and 2).
3. Extraction solvent: 6 ng/mL of each IS in 2 mM ammonium
acetate. Dissolve 15.42 mg of ammonium acetate in 100 mL of
ultrapure water (resistivity >18 MΩ cm). Add 3 μL of each IS
stock solution to 50 mL of 2 mM ammonium acetate solution.
Store at 20 C (see Note 3).
4. Mobile phase A: 10 mM ammonium formate with 0.1% formic
acid. Dissolve 630.6 mg of ammonium formate in 1 L of
ultrapure water. Add 1000 μL of formic acid. Store at room
temperature. Prepare freshly every week.
5. Mobile phase B: Methanol (MeOH) with 0.1% formic acid.
Add 1000 μL of formic acid to 1 L of MeOH.
6. Stock blood for calibration samples: 1000 ng/mL of each
analyte. Mix 2928 μL of blank blood with 3 μL of each of the
1.0 mg/mL reference solutions of COC, nor-COC, COC-Et,
and m-OH-BE and 30 μL of each of the 0.1 mg/mL reference
solutions of BE and EME. Stock blood also serves as
Calibrator 7.
7. Calibration samples 1–6: Dilute stock blood with blank blood
according to the scheme laid out in Table 1.
8. Stock blood for quality control (QC) samples: 1000 ng/mL of
each analyte. Mix 2928 μL of blank blood with 3 μL of each of
the 1.0 mg/mL reference solutions of COC, nor-COC,
COC-Et, and m-OH-BE and 30 μL of each of the 0.1 mg/
mL reference solutions of BE and EME.
9. QC samples 1–4: Dilute stock blood with blank blood accord-
ing to the scheme laid out in Table 2.
264 Lars Ambach and Christophe Stove
Table 1
Dilution scheme for calibration samples
Calibrator C7 C6 C5 C4 C3 C2 C1
Volume blank blood added (μL) – 300 630 570 630 570 540
Volume stock blood added (μL) 600 300 70 30 – – –
Volume C5 added (μL) – – – – 70 30 –
Volume C3 added (μL) – – – – – – 60
Final concentration (ng/mL) 1000 500 100 50 10 5.0 1.0
Table 2
Dilution scheme for quality control (QC) samples
QC sample QC IV QC III QC II QC I
Volume blank blood added (μL) 150 555 540 580
Volume stock blood added (μL) 450 45 – –
Volume QCIII added (μL) – – 60 20
Final concentration (ng/mL) 750 75 7.5 2.5
3 Methods
3.2 Extraction 1. Take a 6 mm punch from the center of each DBS (see Note 9).
2. Transfer each punch into its own 2 mL Eppendorf cup.
3. Add 200 μL of extraction solvent to each tube.
4. Cap and shake for 15 min at 1000 rpm/1.12 g at room
temperature.
5. Add 1000 μL of ice-cold acetonitrile (MeCN) (see Note 10).
6. Cap and shake for 10 min at 1000 rpm at room temperature.
7. Centrifuge the samples for 20 min at 14,000 g, 4 C.
8. Transfer the supernatants into labelled glass tubes or vials.
9. Evaporate the supernatants under a stream of nitrogen at 40 C
(see Note 11).
10. Reconstitute in 50 μL of mobile phase A (see Note 12).
11. Transfer to a labelled autosampler vial with a 100 μL insert.
Table 3
Lower limits of quantification (LLOQ) and MRM transitions and parameters
Dwell
LLOQ m/z time DP EP CXP
Analyte (ng/mL) Q1 m/z Q3 (ms) (V) (V) CE (V) (V)
Cocaine 2.5 304.3 82.0/150.0 30 46 10 39/33 12
Benzoylecgonine 1.0 290.1 82.0/168.1/150.1 30 70 10 37/27/32 10
Ecgonine methyl ester 5.0 200.1 82.0/182.1 30 45 10 32/23 9
Norcocaine 2.5 290.2 136.1/168.1/108.0 30 70 10 30/22/44 11
Cocaethylene 2.5 318.2 82.0/150.1 30 70 10 37/33 10
m-OH-benzoylecgonine 2.5 306.3 121.1/82.0 30 70 10 37/36 7
Cocaine-D3 n/a 307.2 185.2 30 70 9 28 10
Benzoylecgonine-D8 n/a 298.3 171.1 30 60 9 27 10
Ecgonine methyl n/a 203.3 185.2 30 70 10 24 10
ester-D3
Norcocaine-D3 n/a 293.2 136.1 30 57 10 30 10
Cocaethylene-D8 n/a 326.3 204.2 30 70 10 28 12
Q1 quadrupole 1, Q3 quadrupole 3, the first transition is used as quantifier, the following transitions as qualifiers, DP
declustering potential, EP entrance potential, CE collision energy, CXP collision cell exit potential
Cocaine and Metabolites in DBS 267
4 Notes
Fig. 2 Extracted ion chromatograms for all analytes and internal standards at the lower limit of quantitation of
EME (5.0 ng/mL). Blue ¼ qualifier transition, red/green ¼ quantifier transitions
268 Lars Ambach and Christophe Stove
Fig. 2 (continued)
References
1. EMCDDA (2017) European drug report 3. Baselt RC (2011) Cocaine. Disposition of toxic
2017: trends and developments. luxembourg: drugs and chemicals in man, 9th edn. Biomed-
european monitoring centre for drugs and ical Publications, Seal Beach, CA, pp 390–394
drug addiction (EMCDDA); June 2017. 4. Moore C, Negrusz A, Lewis D (1998) Deter-
http://www.emcdda.europa.eu/publications/ mination of drugs of abuse in meconium. J
edr/trends-developments/2017 Chromatogr B 713(1):137–146
2. UNODC (2017) World Drug Report 2017. 5. Lester BM, ElSohly M, Wright LL, Smeriglio
Vienna: United Nations Office on Drugs and VL, Verter J, Bauer CR et al (2001) The mater-
Crime (UNODC). Report No.: ISBN 978-92- nal lifestyle study: drug use by meconium toxi-
1-148291-1. http://www.unodc.org/ cology and maternal self-report. Pediatrics 107
wdr2017/ (2):309–317
Cocaine and Metabolites in DBS 271
6. Klette KL, Poch GK, Czarny R, Lau CO 6-acetylmorphine in blood with use of dried
(2000) Simultaneous GC-MS analysis of blood spots. Ther Drug Monit 30
meta- and para-hydroxybenzoylecgonine and (6):733–739. https://doi.org/10.1097/
norbenzoylecgonine: a secondary method to FTD.0b013e31818d9fdb
corroborate cocaine ingestion using nonhydro- 16. Ingels A-S, De Paepe P, Anseeuw K, Van
lytic metabolites. J Anal Toxicol 24 Sassenbroeck D, Neels H, Lambert WE et al
(7):482–488 (2011) Dried blood spot punches for confir-
7. Guthrie R, Susi A (1963) A simple phenylala- mation of suspected gamma-hydroxybutyric
nine method for detecting phenylketonuria in acid intoxications: validation of an optimized
large populations of newborn infants. Pediat- GC-MS procedure. Bioanalysis 3
rics 32:338–343 (20):2271–2281. https://doi.org/10.4155/
8. Edelbroek PM, van der Heijden J, Stolk LML bio.11.204
(2009) Dried blood spot methods in therapeu- 17. Hernández Redondo A, Schroeck A,
tic drug monitoring: methods, assays, and pit- Kneubuehl B, Weinmann W (2013) Determi-
falls. Ther Drug Monit 31(3):327–336. nation of ethyl glucuronide and ethyl sulfate
https://doi.org/10.1097/FTD. from dried blood spots. Int J Legal Med 127
0b013e31819e91ce (4):769–775. https://doi.org/10.1007/
9. McDade TW, Williams S, Snodgrass JJ (2007) s00414-012-0815-2
What a drop can do: dried blood spots as a 18. De Kesel PMM, Sadones N, Capiau S, Lambert
minimally invasive method for integrating bio- WE, Stove CP (2013) Hemato-critical issues in
markers into population-based research. quantitative analysis of dried blood spots: chal-
Demography 44(4):899–925 lenges and solutions. Bioanalysis 5
10. Ambach L, Hernández Redondo A, König S, (16):2023–2041. https://doi.org/10.4155/
Weinmann W (2014) Rapid and simple bio.13.156
LC-MS/MS screening of 64 novel psychoac- 19. Ellefsen KN, da Costa JL, Concheiro M,
tive substances using dried blood spots. Drug Anizan S, Barnes AJ, Pirard S et al (2015)
Test Anal 6(4):367–375. https://doi.org/10. Cocaine and metabolite concentrations in
1002/dta.1505 DBS and venous blood after controlled intra-
11. Odoardi S, Anzillotti L, Strano-Rossi S (2014) venous cocaine administration. Bioanalysis 7
Simplifying sample pretreatment: application (16):2041–2056. https://doi.org/10.4155/
of dried blood spot (DBS) method to blood bio.15.127
samples, including postmortem, for UHPLC- 20. Saussereau E, Lacroix C, Gaulier JM, Goulle JP
MS/MS analysis of drugs of abuse. Forensic Sci (2012) On-line liquid chromatography/tan-
Int 243:61–67. https://doi.org/10.1016/j. dem mass spectrometry simultaneous determi-
forsciint.2014.04.015 nation of opiates, cocainics and amphetamines
12. den Burger JCG, Wilhelm AJ, Chahbouni A, in dried blood spots. J Chromatogr B
Vos RM, Sinjewel A, Swart EL (2012) Analysis 885–886:1–7. https://doi.org/10.1016/j.
of cyclosporin A, tacrolimus, sirolimus, and jchromb.2011.11.035
everolimus in dried blood spot samples using 21. Mercolini L, Mandrioli R, Gerra G, Raggi MA
liquid chromatography tandem mass spectrom- (2010) Analysis of cocaine and two metabolites
etry. Anal Bioanal Chem 404 in dried blood spots by liquid chromatography
(6–7):1803–1811. https://doi.org/10.1007/ with fluorescence detection: a novel test for
s00216-012-6317-8 cocaine and alcohol intake. J Chromatogr A
13. Sadones N, Capiau S, De Kesel PMM, Lambert 1217(46):7242–7248. https://doi.org/10.
WE, Stove CP (2014) Spot them in the spot: 1016/j.chroma.2010.09.037
analysis of abused substances using dried blood 22. Déglon J, Thomas A, Mangin P, Staub C
spots. Bioanalysis 6(17):2211–2227. https:// (2012) Direct analysis of dried blood spots
doi.org/10.4155/bio.14.156 coupled with mass spectrometry: concepts and
14. Déglon J, Versace F, Lauer E, Widmer C, biomedical applications. Anal Bioanal Chem
Mangin P, Thomas A et al (2012) Rapid 402(8):2485–2498. https://doi.org/10.
LC-MS/MS quantification of the major ben- 1007/s00216-011-5161-6
zodiazepines and their metabolites on dried 23. Antelo-Domı́nguez Á, Cocho JÁ, Tabernero
blood spots using a simple and cost-effective MJ, Bermejo AM, Bermejo-Barrera P, Mor-
sample pretreatment. Bioanalysis 4 eda-Piñeiro A (2013) Simultaneous determina-
(11):1337–1350. https://doi.org/10.4155/ tion of cocaine and opiates in dried blood spots
bio.12.42 by electrospray ionization tandem mass spec-
15. Garcia Boy R, Henseler J, Mattern R, Skopp G trometry. Talanta 117:235–241. https://doi.
(2008) Determination of morphine and org/10.1016/j.talanta.2013.09.010
272 Lars Ambach and Christophe Stove
24. Capiau S, Veenhof H, Alffenaar J-W, Stove CP cocaine interactions in humans. J Pharmacol
(2018) A guideline on bioanalytical method Exp Ther 266(3):1364–1373
validation for dried blood sample analysis. 27. Guidance document on analytical quality con-
Ther Drug Monit (submitted for publication) trol and validation procedures for pesticide
25. Inaba T, Stewart DJ, Kalow W (1978) Metab- residues analysis in food and feed.: European
olism of cocaine in man. Clin Pharmacol Ther Commission, EU Reference Laboratories for
23(5):547–552 Residues of Pesticides; 2014. Report No.:
26. Farré M, de la Torre R, Llorente M, Lamas X, SANCO/12571/2013. http://ec.europa.eu/
Ugena B, Segura J et al (1993) Alcohol and food/plant/plant_protection_products/guid
ance_documents/docs/qualcontrol_en.pdf
INDEX
A G
Accurate mass .................................................42, 181, 189 Gabapentin ............................................ 67, 119–126, 184
Alcohol biomarkers ....................................................... 224 Glucuronidase ...................................................12, 26, 27,
Anticonvulsants .....................................68, 112, 119, 120 29, 30, 32, 55, 140, 142, 146
Antiepileptic drugs ......................................................4, 67
H
B
High-resolution mass spectrometry (HRMS)................ 1,
Biologics .................................................................. 1, 3, 5, 4, 7, 8, 41–50, 181, 182, 189
12, 68, 135, 149, 165, 166, 179, 181, 258
Buprenorphine ........................................... 41, 43, 45, 46, I
48, 49, 51–59, 187, 256 Immunosuppressant...................................................3, 76,
102, 111, 112
C
Cannabidiol (CBD) .................................................11–21, L
184, 241
Lacosamide ................................................................67–73
Cannabinoids..................................11–20, 130–132, 134, Leflunomide ......................................................... 8, 75–82
137–146, 212, 215, 241–243, 256, 258 Legal highs ........................................................... 129, 165
Cocaine (COC) .......................................... 187, 199–209,
Liquid chromatography (LC)..............................v, 1, 3, 5,
240, 245, 256, 261–270 6, 11–20, 24, 26, 29, 30, 34, 36, 37, 42, 52, 55,
Cyclosporine.................................................103, 111–117 58, 61–65, 67–73, 75–82, 85–98, 101–109,
111–117, 119–126, 137–146, 149–162,
D
165–179, 181–190, 199–209, 211–221,
Designer drugs ........................................... 129–135, 137, 223–235, 237–259, 261–270
165, 179, 182
Designer stimulants ............................................. 165–179 M
Disease-modifying antirheumatic drug Mass spectrometry (MS).............................................. 1–9,
(DMARD) ........................................................... 75
11–20, 24, 28–30, 34, 36, 37, 42, 52, 61–65,
Dried blood spots ............................................ 5, 261–270 67–73, 75–82, 85–98, 101–109, 111–117,
Drug-facilitated assaults............................................23–38 119–126, 137–146, 149–162, 165–179,
181–190, 199–209, 211–221, 223–235,
E
237–259, 261–270
Ethanol biomarkers....................................................... 224 Meconium .......................................................5, 191–196,
Ethyl-glucuronide (EtG) ..................................... 223–235 199–209, 211, 212, 225
Extraction Methotrexate ................................................3, 8, 101–109
liquid-liquid ................................................. 5, 26, 149, Monoclonal antibody (mAb) therapies........... 3, 8, 85–98
199–209, 238, 268
pipette tip .................................................................. 12 N
salt-assisted liquid-liquid ............................... 199–209 Novel benzodiazepines ........................................ 133, 135
solid-phase ..................................................... 6, 12, 26,
Novel opioids ....................................................... 132, 133
28, 113, 146, 149, 150, 154, 156, 200, 212–214, Novel psychoactive drugs .........................................23, 29
217, 220, 225, 229, 230, 238, 251, 258, 268
Loralie J. Langman and Christine L.H. Snozek (eds.), LC-MS in Drug Analysis: Methods and Protocols,
Methods in Molecular Biology, vol. 1872, https://doi.org/10.1007/978-1-4939-8823-5,
© Springer Science+Business Media, LLC, part of Springer Nature 2019
273
LC-MS IN DRUG ANALYSIS: METHODS AND PROTOCOLS
274 Index
O T
Opioids ..........................................................5, 12, 41–52, Tacrolimus ....................................................103, 111–118
131–133, 149–162, 195, 256, 258 Tapentadol.........................................................41, 43, 45,
Orbitrap ............................................................26, 42, 181 49, 61–65, 256
Teriflunomide ................................................... 75–82, 103
P Therapeutic drug monitoring (TDM)........................ 1–3,
Pain management.................................. 5, 41–50, 52, 120 8, 67–73, 101–103, 109, 111, 120, 262
Time-of-flight (TOF).................................................... 181
Pregabalin .......................................................67, 119–126
Toxicology ..............................................1, 4, 5, 8, 34, 69,
R 70, 131, 137, 182, 190–194