Vous êtes sur la page 1sur 10

s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610

available at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/steroids

Development of a highly sensitive and selective


UPLC/MS/MS method for the simultaneous determination
of testosterone and 5␣-dihydrotestosterone in human
serum to support testosterone replacement therapy
for hypogonadism

Hermes Licea-Perez ∗ , Sherry Wang, Matthew E. Szapacs, Eric Yang


Worldwide Bioanalysis, Drug Metabolism and Pharmacokinetics, GlaxoSmithkline Pharmaceuticals,
709 Swedeland Road, King of Prussia, PA 19406, USA

a r t i c l e i n f o a b s t r a c t

Article history: A highly sensitive and selective quantitative method to accurately determine testos-
Received 28 November 2007 terone (Te) and 5␣-dihydrotestosterone (DHT) in human serum is crucial to the success
Received in revised form of Te replacement therapy for hypogonadism. To this end we have developed and vali-
14 January 2008 dated a semi-automated and relatively high-throughput method in a 96-well plate format
Accepted 15 January 2008 using ultra-performance liquid chromatography coupled with tandem mass spectrometry
Published on line 2 February 2008 (UPLC/MS/MS) for the simultaneous determination of Te and DHT in human serum. Te and
DHT along with the internal standards [2 H3 ]-Te and [2 H3 ]-DHT were extracted from 300 ␮L of
Keywords: human serum by liquid–liquid extraction using methyl tertiary-butyl ether (MTBE), followed
Testosterone by derivatization with 2,3-pyridinedicarboxylic anhydride and solid-phase extraction for
Dihydrotestosterone sample clean up. A novel chemical derivatization approach using 2,3-pyridinedicarboxylic
Derivatization, anhydride was employed to achieve the MS sensitivity and selectivity required for DHT.
2,3-Pyridinedicarboxilic anhydride Baseline separation of Te and DHT derivatives from endogenous steroid derivatives was
UPLC, Surrogate Matrix achieved using UPLC technology on a C18 stationary-phase column with 1.7 ␮m particle size.
The validity of using double charcoal-stripped female human serum as surrogate matrix for
preparation of calibration standards was demonstrated through standard addition exper-
iments. The method was validated over the concentration ranges of 0.2–40 ng/mL for Te
and 0.01–2 ng/mL for DHT. The validation and study sample analysis results show that the
method is rugged, precise, accurate, and well suited to support pharmacokinetic studies
where approximately 300 samples can be extracted and analyzed in 1 day.
© 2008 Elsevier Inc. All rights reserved.

1. Introduction tration of Te in serum is lower than 3 ng/mL [1]. Men with


hypogonadism experience symptoms such as a decrease in
Testosterone (Te), the major androgen in males, has been libido, lean body mass, muscle strength, bone density, and sex-
widely used in androgen replacement therapy in hypogonadal ual function [1]. Although Te replacement therapy alleviates
elderly men and is often recommended when the concen- these symptoms, there are some concerns about the impact


Corresponding author. Tel.: +1 610 270 5116; fax: +1 610 270 5005.
E-mail address: hermes.2.licea perez@gsk.com (H. Licea-Perez).
0039-128X/$ – see front matter © 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.steroids.2008.01.018
602 s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610

of Te therapy on prostate health in elderly men [2]. Te is exten- [14,15,18–23]. Due to the presence of an ␣,␤-unsaturated
sively metabolized by 5␣-reductase enzymes (type 1 and type carbonyl and the subsequent charge delocalization on its pro-
2) to 5␣-dihydrotestosterone (DHT) which in turn binds to the tonated form (conjugate acid) [18,19], it was possible to reach a
androgen receptor [2]. 5␣-Reductase enzyme type 1 is predom- LOQ in the low pg/mL range for Te without the need for chem-
inantly present in the skin and liver while type 2 is expressed ical derivatization. DHT, on the other hand, is a neutral steroid
in the prostate [2,3]. DHT is a more potent androgen than and ionizes poorly using conventional ionization techniques
Te with an anabolic/androgenic ratio of approximately 4/1, such as electrospray ionization (ESI), atmosphere chemical
thus the concentrations of DHT in serum must be kept within ionization (APCI), and atmosphere photon ionization (APPI)
the normal range for a patient undergoing Te replacement [18,19]. In an effort to validate a high-throughput method
therapy, since high concentrations of DHT are considered to with a LOQ of 0.01 ng/mL, DHT must first be derivatized to
be the major pathogenesis of benign prostatic hyperplasia enhance MS detection sensitivity. A number of derivatization
(BPH) in adult males [4,5]. A considerable volume of data from reagents such as 2-fluoro-1-methylpyridine (FMP), hydrox-
both pre-clinical and clinical studies show that agents such ylamine, nitrobenzylhydroxylamine, methyl hydroxylamine,
as dutasteride, an inhibitor of both type 1 and type 2 5␣- Girard reagent P hydrazone, 2-hydrazino-1-methylpyridine,
reductase, and finasteride, an inhibitor of type 2 5␣-reductase, nitrobenzoyl chloride (NBC), have been reported in the lit-
block the production of DHT preventing prostate enlargement erature for derivatization of carbonyl or hydroxyl functional
[6–9]. Gleave et al. demonstrated that a ≥0.5 mg/day dose of groups of steroids (neutral or oxosteroids) [17–25]. A recent
dutasteride led to a ≥90% decrease in serum DHT levels as well paper using hydroxylamine to derivatize Te and DHT reports
as a decrease in total prostate and tumor volumes in men with a LOQ of 0.029 ng/mL using 100 ␮L of human serum; however,
localized prostate cancer. Therefore, Te therapy is often com- this method uses a large amount of solvent for liquid–liquid
bined with dutasteride or finasteride with the DHT/Te ratio extraction (LLE) that is incompatible with a 96-well plate for-
being closely monitored as an indicator of 5␣-reductase activ- mat which is required for a high-throughput method [25].
ity. In this paper, we describe a relatively high-throughput
A sensitive and accurate bioanalytical method for the anal- UPLC/MS/MS method for the simultaneous determination of
ysis of Te and DHT is required for the diagnosis and treatment Te and DHT from human serum in a 96-well plate format.
of androgen deficiency in men. The development of a method A three-step process consisting of LLE, derivatization with
to simultaneously quantify Te and DHT led to a number of 2,3-pyridinedicarboxylic anhydride and solid-phase extrac-
challenges. First, Te and DHT are both endogenous steroids, tion was employed before sample analysis. Several additional
so a surrogate matrix must be used for the preparation of experiments, including standard addition and stability with
calibration standards and the method accuracy using the sur- incurred samples, were included in the method validation to
rogate matrix needs to be proven. Second, the levels of Te confirm the accuracy and the integrity of the results.
and DHT in serum from hypogonadal patients are expected
to be lower than in normal human serum (≤2.5 ng/mL for
Te and ≤0.29 ng/mL for DHT [9]); therefore, a very sensitive 2. Experimental
method with the lower limit of quantification (LOQ) in the
pg/mL range is required. Finally, there are a number of endoge- 2.1. Chemicals and reagents
nous steroids with similar chemical structure which could
interfere with Te/DHT quantification, so the method needs to Te, DHT, androsterone, trans-androsterone, dehydroepia-
be selective to prevent the overestimation of Te/DHT levels in ndrosterone, 2,3-pyridine dicarboxylic anhydride, 3,4-
human serum. Traditionally, radioimmunoassay (RIA)-based pyridine dicarboxylic anhydride, pyridine, and N,N-dime-
methods have been used for analysis of Te in biological matri- thylformamide were purchased from Sigma–Aldrich (St.
ces [10–13]. However, these methods often involve complex Louis, MO, USA). Internal standards, Te-[2 H3 ] and DHT-[2 H3 ]
and tedious sample preparation procedures and also require were obtained from CDN Isotopes (Quebec, Canada). HPLC
handling of radioactive materials. Furthermore, RIA methods grade acetonitrile was purchased from Burdick and Jackson
are known to suffer from the lack of sensitivity and selectiv- (Muskegon, MI, USA). ␤-DHT was obtained from Steraloids
ity due to cross reactivity from polar metabolites of Te, other (Newport, RI, USA). Methyl tertiary-butyl ether (MTBE) and
endogenous steroids, and non-specific binding, particularly formic acid were purchased from EMD Scientific (Gibbstown,
at low Te concentrations [12–15]. GC/MS methods employ- NJ, USA). Double charcoal-stripped female (DCSF) human
ing derivatization of steroids to mono- or tri-methylsilyl and serum was obtained from Bioreclamation Inc. (East Meadow,
heptafluorobutyric ether derivatives have also been developed NY, USA).
[16,17]. Although these GC/MS methods are selective and sen-
sitive, sample preparation is usually very time-consuming and 2.2. Equipment
analysis run times may exceed 20 min per sample, making
them less suitable for high-throughput analysis. In addition, An Eppendorf 5810R centrifuge with a rotor capacity for four
these methods often suffer from problems associated with the 96-well plates (Brinkmann Instrument, Westbury, NY, USA)
instability of the derivatives and potential thermal decompo- and a Mettler UMX2 balance (Hightown, NJ, USA) were used.
sition during analysis [17]. A Hamilton Mircrolab STAR liquid handler (Reno, NV, USA)
Recently, high-performance liquid chromatography cou- was used for serum transfer, while a TomTec Quadra 3 SPE
pled with tandem mass spectrometry (HPLC/MS/MS) has (Hamden, CT, USA) was used for liquid transfer. Arctic White
proven to be a useful tool for analysis of Te in serum LLC 96-well round 2-mL plates with silicone and PTFE film
s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610 603

seal mats (Bethlehem, PA, USA) were used to extract ana- to all wells to hydrolyze the excess 2,3-pyridinedicarboxylic
lytes and their internal standards from serum. One milliliter anhydride. The samples were then loaded onto a 30-mg HLB
deactivated (silanized) glass vials along with 96-well plate SPE 96-well plate, the SPE plate was washed with 2× 1000 ␮L of
covers (Blue CapMat with Pre-Cut T/S Septa) from MicroLiter 40% methanol in water (v/v) and eluted with 2× 400 ␮L of 80%
Analytical Supplies (Suwanee, GA, USA) were used for sam- methanol in water into a 2-mL 96-well plate containing 1 mL
ple introduction to the UPLC. The SPE sample clean up after deactivated (silanized) glass-inserts. These sample extracts
derivatization was carried out on the Oasis 30-mg HLB 96-well were evaporated at 45 ◦ C under a stream of nitrogen, recon-
plates from Waters (Milford, MA, USA). stituted with 100 ␮L of 30/70 acetonitrile/water, capped and
vortex mixed before analysis.
2.3. Preparation of standards and quality control (QC)
samples 2.5. Chromatographic conditions

Stock solutions of Te, DHT, Te-[2 H3 ] and DHT-[2 H3 ] were An ACQUITYTM UPLC integrated system from Waters (Milford,
individually prepared in dimethylformide at concentrations MA, USA), consisting of an autosampler combined with a sam-
of 1.0 mg/mL and stored at room temperature. The stock ple organizer capable of holding ten 96-deep well plates, binary
solutions of Te and DHT were combined and diluted with solvent manager, and an ACQUITY UPLCTM BEH C18 column
50/50 acetonitrile/water to make the working solution WS1 (100 mm × 2.1 mm, 1.7 ␮m particle size) was used. The column
containing Te at 80 ␮g/mL and DHT at 4 ␮g/mL. WS1 was temperature was maintained at 50 ◦ C and the sample com-
diluted to make working solution WS2 at a concentration partment was maintained at 10 ◦ C. Mobile phase A was water
of 800/40 ng/mL of Te/DHT in 50/50 acetonitrile/water. The and mobile phase B consisted of 2 mM ammonium acetate
WS2 was used to make calibration standards at 40/2, 20/1, (native pH) in acetonitrile/water 98/2. The LC system was held
10/0.5, 4/0.2, 2/0.1, 1/0.05, 0.4/0.02, and 0.2/0.01 ng/mL of at 0% B for 0.1 min followed by a non-linear gradient (convex
Te/DHT using a serial dilution procedure. Working solutions with steep initial gradient, curve 9) profile from 0% B to 30% B
(WQ1–WQ4) for the QC samples were prepared at concen- for 1.0 min and then from 30% B at 1.0 min to 32% B at 2.0 min.
trations of 80,000/4000, 4000/200, 400/20, and 40/2 ng/mL of The system then was held at 32% B until 3.4 min. The column
Te/DHT in 50/50 acetonitrile/water. The WQ1–WQ4 were used then was washed with 90% B from 3.4 to 4.0 min and the LC was
to make QC samples at 200/10, 40/2, 32/1.6, 4/0.2, 0.8/0.04, equilibrated at 0% B from 4.01 to 4.5 min. The total run time
and 0.2/0.01 ng/mL of Te/DHT. QC samples were divided into including the sample load was 5 min and the flow rate was
2.5-mL aliquots and frozen at −80 ◦ C or extracted immedi- maintained constant at 0.5 mL/min throughout the run. A typ-
ately. In the first validation run, freshly prepared QC samples ical injection volume was 5 ␮L using the partial loop injection
were analyzed against freshly prepared calibration stan- mode. To achieve better assay performance we recommend fil-
dards. For each subsequent validation run, frozen replicate tering the mobile phase before use and preparing it fresh every
aliquots of the QC samples were thawed at room tem- 2 days because of a shift in the retention times observed when
perature and analyzed against a freshly prepared standard using unfiltered or old mobile phases. Also, the authors advise
curve. to wash the analytical column with methanol:water (50:50) for
1 h before starting the analysis.
2.4. Sample preparation
2.6. Mass spectrometric conditions
MTBE (1 mL) was added to each well of the 2-mL ArcticWhite
96-well polypropylene plate. The plate was sealed with the A triple quadrupole mass spectrometer API 5000 (Applied
ArctiSeal mat and vortex mixed in an inverted position for Biosystems/MDS Sciex, Concord, Ontario, Canada) with an
3 min. Subsequently, the MTBE was discarded and the plate electrospray ionization interface (ESI), operated in the nega-
was allowed to dry in the chemical hood. This wash step tive ionization mode was used. The instrument was optimized
was used to remove any plastic residue from the plates and for the 2,3-pyridinedicarboxilic anhydride derivatives of Te,
plate seals. Serum samples (300 ␮L) were then transferred to Te-[2 H3 ], DHT, and DHT-[2 H3 ] by infusing a 10 ng/mL solu-
the washed 96-well plate using a Hamilton STAR liquid han- tion of SPE purified derivatives in acetonitrile:water (50:50,
dler. A 25-␮L aliquot of internal standard solution (20 ng/mL v/v) at 500 ␮L/min through an Agilent pump 1100 series (Palo
for Te-[2 H3 ], and 4 ng/mL for DHT–[2 H3 ]) was added to all Alto, CA, USA) directly connected to the mass spectrome-
tubes with the exception of the blanks, which received 25 ␮L ter. The MRM transitions of m/z 437–286, m/z 440–288, m/z
of 50/50 acetonitrile/water, followed by recapping and vortex 438–287, and m/z 441–288 were chosen for Te, Te-[2 H3 ], DHT,
mixing for 1 min. Following addition of 1 mL of MTBE, the wells and DHT-[2 H3 ] derivatives, respectively. The m/z 437–286 and
were capped with the ArctiSeal mat; vortex mixed for 3 min, m/z 440–288 transitions for Te and Te-[2 H3 ], respectively, were
and centrifuged at 3220 × g for 5 min. The MTBE layer was chosen instead of m/z 436–285 and m/z 439–287 to maintain MS
transferred to a 2-mL polypropylene 96-well plate with 1-mL response linearity and avoid MS detection saturation at high
glass-inserts using a TomTec liquid handler and the MTBE was concentrations of Te.
evaporated under a stream of nitrogen at 45 ◦ C. After evapora- The optimized mass spectrometric conditions were: ion
tion, 100 ␮L of 100 mg/mL 2,3-pyridine dicarboxylic anhydride source temperature, 700 ◦ C; ion spray voltage, −4500 V; curtain
in anhydrous pyridine was added to all wells. The plate was gas, 30 psi (nitrogen); nebulizing gas (GS1), 60 psi (zero air); TIS
sealed and vortex mixed for 3 min, followed by incubation at gas (GS2), 80 psi (zero air); collision energy, −50 eV for Te/Te-
60 ◦ C for 60 min. After incubation, 900 ␮L of water was added [2 H3 ] (detuned) and −35 eV for DHT and DHT-[2 H3 ] derivatives.
604 s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610

The dwell times were 150 and 100 ms for the analytes and the 3.3. Derivatization of Te and DHT
internal standards, respectively. The longer dwell time for the
analyte channel was used to obtain a consistent peak shape A number of derivatization reagents were tested to achieve
at the LOQ. the LOQ of 0.01 ng/mL for DHT. Initially we aimed to derivatize
the carbonyl moiety of Te and DHT. A variety of reagents such
2.7. Data analysis as hydroxylamine, nitrobenzylhydroxylamine, and methyl
hydroxylamine were tested but did not provide the sensitiv-
MS data were acquired and processed (integrated) using the ity needed for the desired LOQ for DHT using a 96-well plate
proprietary software application AnalystTM (Version 1.4.1, format. Then derivatization of the hydroxyl moiety of Te and
Applied Biosystems/MDS Sciex, Canada). Calibration plots of DHT was tested. Since the hydroxyl group is a poor leaving
analyte/internal standard peak area ratio versus Te and DHT group it is difficult for Te and DHT to undergo SN 2 substitu-
concentrations were constructed and a weighted 1/x2 linear tion reactions, thus the most effective way to derivatize the
regression was applied to the data. Concentrations of Te and hydroxyl moiety is to use oxygen as a nucleophile. Alcohols in
DHT in validation samples were determined from the appro- general are weak nucleophiles and often difficult to derivatize.
priate calibration line, and used to calculate the bias and Normally high concentrations of the reagent are required with
precision of the method with an in-house LIMS (Study Man- long incubation times. A number of derivatization reagents
agement System, SMS2000, Version 1.6, GlaxoSmithKline). such as 2-fluoro-1-methylpyridine, nitrobenzoyl chloride,
N-methyl isotonic anhydride, 3,4-pyridine dicarboxylic anhy-
dride, 2,3-pyridine dicarboxylic anhydride, phthalic anhy-
dride, 3-nitrophthalic anhydride, 2,3-pyrazinedicarboxylic
3. Results and discussion
anhydride were evaluated. The biggest challenge using high
concentrations of the reagent is the removal of the excess
3.1. Method development
reagent after the reaction is complete to avoid MS detec-
tion interference. In most cases the excess reagent can be
Numerous challenges were faced during method development
hydrolyzed by the addition of water to the reaction mixture.
including: (1) the low limit of quantification of 0.01 ng/mL for
In some cases, the reagent was ruled out because of the large
DHT requiring chemical derivatization to enhance MS sensi-
amount of precipitation observed after hydrolysis (e.g. NBC),
tivity, (2) chromatographic separation of Te/DHT derivatives
while other reagents were ruled out due to the lack of sensi-
from other endogenous steroid derivatives, (3) only a two mass
tivity they provided for DHT detection (e.g. N-methylisatonic
unit difference between Te and DHT requiring baseline sepa-
anhydride, 3-nitrophthalic anhydride, phthalic anhydride, 2,3-
ration between analytes, (4) difficulty finding a native serum
pyrazinedicarboxylic anhydride, and FMP).
matrix with acceptable levels of Te and DHT for preparation
The reaction of alcohols with cyclic anhydrides has
of QC and calibration standard samples, and (5) a three mass
been reported previously in the literature [26]. The reac-
unit difference between the analytes and their respective
tion of methanol with 3,4-pyridinedicarboxylic anhydride
internal standards leading to contribution from the internal
results in a mixture of two amphoteric derivatives where
standards to the analyte signals requiring special attention
4-methoxycarbonyl-3-carboxylic acid predominates (84%)
to the amount of the internal standard that was used. The
[27]. These amphoteric derivatives can be detected in both
approaches used to resolve these challenges will be discussed
positive and negative ionization modes. Derivatization of
in detail below.
Te and DHT with 3,4-pyridinedicarboxylic anhydride was
tested to determine if this derivatization reagent would allow
3.2. Underivatized Te and DHT the quantitation of DHT at the desired LOQ in serum. The
excess reagent was hydrolyzed with water and the precipitate
Since the expected ranges for Te were relatively high was separated from the supernatant by centrifugation. Two
(0.2–40 ng/mL) it was not difficult to develop a method for positional isomers were observed for Te and DHT derivatives.
Te. Moreover, the presence of an ␣,␤-unsaturated carbonyl This derivatization method provided excellent MS sensi-
group in the structure of Te facilitates its ionization and sta- tivity allowing DHT detection in DCSF at the desired LOQ
bilizes these ions in the mass spectrometer. A method for (0.01 ng/mL). However, chromatographic separation between
analysis of Te alone was validated in our laboratory over the the DHT derivative and other endogenous steroid derivatives
range of 0.05–10 ng/mL without derivatization using just 50 ␮L such as androsterone, trans-androsterone, and ␤-DHT present
of serum (data not shown). Briefly, Te was extracted from in incurred samples was not achieved, leading to an overes-
serum using liquid–liquid extraction with 1 mL of MTBE. Te timation of DHT concentrations in the incurred samples. The
was analyzed using isocratic conditions with a 50:50 mix- complexity of the DHT chromatogram is partially related to
ture of acetonitrile/water on an ACQUITY UPLC equipped the fact that two peaks are expected for each interference
with a BEH C18 column (50 mm × 2 mm, 1.7 ␮m particle size) compound leading to a higher chance for peak overlap when
and detected with an API-5000 in the positive ionization running short chromatographic separations. A symmetrical
mode. Unfortunately, this method could not be applied for derivatization reagent like 2,3-pyrazinedicarboxylic anhydride
analysis of DHT due to insufficient sensitivity. The low- generates only one peak for each compound and would make
est quantifiable level for DHT using the above method was the LC separation simpler, but the MS sensitivity of the DHT
0.2 ng/mL, a 20-fold difference from the desired LOQ of derivative was insufficient to reach a LOQ of 0.01 ng/mL for
0.01 ng/mL. DHT.
s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610 605

Finally, derivatization of Te and DHT with 2,3- in the control human serum significantly exceeded the tar-
pyridinedicarboxylic anhydride was tested. This reagent geted LOQ of the method. The testes in healthy human males
generated two positional isomeric products (Te-1,2 and DHT- produce about 0.24 ␮mol of Te daily [1] leading to serum con-
1,2; see Fig. 1) with the amphoteric derivatives being detected centrations between 2.5 and 9.5 ng/mL [9]. For this reason
in both positive and negative ionization mode. The intensity DCSF human serum was chosen as the surrogate matrix for
of peaks Te-1 and DHT-1 was the greatest with the second preparation of calibration standards and quality control sam-
peak (Te-2 and DHT-2) eluting as a hump (see Fig. 2A and B). ples. Multiple lots of DCSF human serum, were screened for
The analysis was performed in the negative ion mode due trace amounts of Te and DHT before being used for preparation
to a lower base line and less interference. The derivatization of QC and calibration standards. From 50 lots of DCSF human
reaction efficiency of the 2,3-pyridinedicarboxylic anhydride serum screened only three lots were found to contain Te/DHT
reaction was evaluated by monitoring the disappearance levels low enough (less than 20% of the LOQ) to be acceptable
of underivatized Te after extraction and derivatization. as a matrix blank.
This was done because pure standards for derivatized Te
and DHT were not available. The presence of underiva-
3.6. Recovery
tized Te observed was <1% of the original concentration
of Te after a 1-h reaction time indicating a high-reaction
The recovery of Te from DCSF human serum using LLE was
efficiency. This reagent allowed the quantitation of DHT
assessed using the validated method for underivatized Te
at the desired LOQ of 0.01 ng/mL and intensive LC opti-
described in Section 3.2. The recovery was calculated as the
mization (see below) using incurred samples allowed the
difference in the peak area ratios (analyte/internal standard)
separation of Te/DHT derivatives from endogenous steroid
for Te spiked in DCSF human serum either prior to LLE or
derivatives.
in DCSF human serum extracts post-LLE at 0.2 and 4 ng/mL.
The experiment was carried out in replicates of six and the
3.4. LC optimization
recovery was found to be 92.1% and 87.7% at 0.2 and 4 ng/mL,
respectively.
Our objective was to develop an UPLC method with the short-
est run time, highest sensitivity, but without compromising
the selectivity of the assay. A baseline separation between 3.7. Selectivity and linearity
the DHT derivative and other endogenous steroid derivatives
with the same MS/MS transition as DHT is required. UPLC The characteristic precursor [M−H]− to product ion transi-
parameters such as pH, flow rate, column type, and buffer tions, m/z 436–285, m/z 439–287, and m/z 438–287, 441–289 are
concentration were optimized to achieve the best sensitiv- consistent with the structures of the derivatives of Te, [2 H3 ]-Te,
ity, peak shape and selectivity. The presence of ammonium DHT, and [2 H3 ]-DHT, respectively. However, less sensitive pre-
acetate in the mobile phase was crucial for the separa- cursor to product ion transitions of m/z 437–286 and 440–288
tion of all components of the mixture as the lack of the were chosen for the Te and Te-[2 H3 ] derivatives, respectively, to
ammonium acetate resulted in much less retention of the maintain MS response linearity and to avoid MS detector satu-
peaks and a poor separation. The higher the concentra- ration at high concentrations. Since all human serum samples
tion of ammonium acetate, the better the peak resolution contain detectable levels of Te and DHT, it is a challenge
and the longer the peak retention for the analytes. Unfor- to demonstrate that the method is selective. The selectivity
tunately, the higher concentration of ammonium acetate of this method was established by the analysis of incurred
also led to decreased sensitivity. Changing the mobile phase samples with a variety of LC conditions (isocratic and long gra-
from pH 3 to pH 7 increased the MS sensitivity in the dients). UPLC/MS/MS chromatograms were visually examined
negative ion mode for the DHT derivative, but the selectiv- and compared for chromatographic integrity and potential
ity and peak shape got worse. Two millimolar ammonium interferences. The retention times of Te and DHT derivatives
acetate in a mixture of acetonitrile/water 98/2 was found to were the same as that of their respective internal standard
offer the optimal balance between sensitivity and selectiv- derivatives for all LC programs used. Moreover, the area ratios
ity. between the analyte and internal standard were also con-
The column selection was also important because of the stant. Potential interference from endogenous steroids such as
complexity of the separation. A 100 mm × 2.1 mm UPLC col- androsterone, trans-androsterone, ␤-DHT, androstanediol, and
umn with 1.7 ␮m particle size showed the highest resolving dehydroepiandrosterone was also investigated. This was done
power which allowed for the use of a high-linear velocity thus, by spiking DCSF human serum with the above-mentioned
shortening run time to 5 min without sacrificing peak resolu- reference standards and running the corresponding deriva-
tion. In addition, we tested conventional analytical columns tives with the validated LC method. The retention times of
with larger particle size, however, we were unable to sepa- the derivatives from all investigated compounds differ from
rate all peaks in this complex mixture without significantly that of the derivatives of Te and DHT and their internal stan-
extending the run time. dards.
The linearity of the method was evaluated by analyzing
3.5. Surrogate matrix eight calibration standards in duplicate over the nominal con-
centration range of 0.2–40 ng/mL for Te and 0.01–2 ng/mL for
Non-stripped human serum cannot be used for preparation DHT. The correlation coefficients obtained using 1/x2 weighted
of calibration standards as endogenous levels of Te and DHT linear regressions were better than 0.9983 for both Te and DHT.
606 s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610

Fig. 1 – Reaction scheme for the reaction of Te and DHT with 2,3-pyridinedicarboxilic anhydride in the presence of pyridine.
The LC elution order of Te-1/Te-2 and DHT-1/DHT-2 is unknown.

3.8. Bias and precision 5.5% for Te and 10.1% for DHT. The maximum between-run
precision values observed were 9.4% for Te and 5.7% for DHT.
At all QC concentrations examined, the bias and precision val- As defined by the lower and upper validation sample concen-
ues were less than 15% over three analytical runs (see Table 1). trations possessing acceptable accuracy and precision, the val-
The maximum bias observed was −10.8% for Te and −8.2% for idated range of this method based on 300 ␮L of EDTA human
DHT. The maximum within-run precision value observed was serum is 0.01–2.0 ng/mL for DHT and 0.2–40.0 ng/mL for Te.

Table 1 – Bias, precision and mean validation sample concentrations for Te and DHT in DCSF human serum
Concentration (ng/mL) Te DHT

0.20 0.80 4.00 32.00 40.00 0.01 0.04 0.200 1.60 2.00

Run 1 (n = 6)
Mean 0.21 0.84 4.11 31.04 38.41 0.010 0.042 0.210 1.565 2.002
%CV 4.9 2.8 2.7 2.3 1.7 10.1 5.4 3.8 3.1 3.6
Bias% 7.0 4.8 2.6 −3.0 −4.0 4.2 4.2 5.1 −2.2 0.1

Run 2 (n = 6)
Mean 0.18 0.82 4.04 30.07 38.00 0.092 0.039 0.197 1.575 1.920
%CV 5.4 5.5 4.9 3.4 4.6 3.7 4.0 3.2 3.4 4.7
Bias% −10.8 2.8 1.0 −6.0 −5.0 −8.2 −2.2 −1.3 −1.6 −4.0

Run 3 (n = 6)
Mean 0.19 0.79 3.96 30.41 37.48 0.099 0.041 0.202 1.544 1.922
%CV 3.4 1.9 3.0 2.6 2.5 3.2 4.1 3.8 3.0 1.7
Bias% −6.0 −1.4 −1.1 −5.0 −6.3 −1.0 1.7 1.2 −3.5 −3.9

Overall totals
Mean 0.19 0.82 4.03 30.50 37.96 0.098 0.041 0.203 1.561 1.948
Average bias% −3.3 2.1 0.9 −4.7 −5.1 −1.7 1.2 1.7 −2.4 −2.6

Between-run %CV (n = 18) 9.4 2.7 1.1 1.1 Negligible 5.7 2.6 2.8 Negligible 1.9
s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610 607

Fig. 2 – (A) Chromatograms for Te after derivatization with 2,3-pyridine dicarboxilic anhydride from blank DCSF (A), DCSF
spiked with 0.2 ng/mL of Te (B), 40 ng/mL of Te (C) and incurred sample determined at a concentration of 33.4 ng/mL (D). (B)
Chromatograms for DHT after derivatization with 2,3-pyridine dicarboxilic anhydride from blank DCSF (A), DCSF spiked
with 0.01 ng/mL (B), 2 ng/mL (C) and an incurred sample determined at a concentration of 0.130 ng/mL (D).

3.9. Stability of Te and DHT during freeze–thaw cycles freshly spiked serum or pooled incurred serum samples had a
percent difference and CV (%) between replicates of less than
The stability of Te and DHT in spiked DCSF samples dur- 15% which indicates that Te and DHT are stable in human
ing freeze–thaw cycles from −80 ◦ C to room temperature was serum after at least three freeze–thaw cycles from −80 ◦ C to
assessed. This was done by subjecting QC samples at 0.8/0.04 room temperature.
and 32/1.6 ng/mL Te/DHT to three freeze–thaw cycles from
−80 ◦ C to room temperature and then comparing the mean 3.10. Matrix dilution
concentrations (in replicates of 6) against those of the freshly
prepared spiked QC samples. This experiment gave a percent The ability to dilute samples containing Te and DHT at con-
difference of −4.4 and −2.0 for Te at 0.8 and 32 ng/mL and −1.7 centrations above the high limit of quantitation (HOQ) was
and −3.8 for DHT at 0.04 and 1.6 ng/mL DHT, respectively. The demonstrated by performing a 10-fold dilution of DCSF human
maximum CV (%) observed in this experiment was 3.4 for Te serum validation sample spiked at 200/10 (Te/DHT) ng/mL (in
and 5.3 for DHT. In addition, since the stability in DCSF human replicates of 6). Since it was difficult to obtain clean DCSF
serum may not reflect stability in native serum, the stability human serum, both serum and water were tested as viable
of Te/DHT in pooled incurred human serum also was assessed diluents. Concentrations of Te and DHT in these matrix dilu-
after three freeze–thaw cycles from −80 ◦ C to room temper- tion samples were determined and corrected for dilution
ature at two concentration levels (in replicates of 6). These factor. The bias and within-run precision values using DCSF
pooled samples were prepared from samples collected from a human serum as the diluent were less than 15% (0.1% and
study where normal volunteers received Lupron to simulate 1.1%, respectively for Te, and 3.4% and 3.8%, respectively for
hypogonadism (low level of Te). This experiment gave a per- DHT) indicating that a 10-fold dilution of human serum sam-
cent difference of −3.7 and −2.2 for Te at 0.818 and 31.4 ng/mL ples containing Te and DHT above the HOQ is valid. Similarly,
and −8.0 and −0.5 for DHT at 0.015 and 0.118 ng/mL, respec- the bias and within-run precision values using water as the
tively. The maximum CV (%) observed in this experiment was diluent were also less than 15% (actually −1.0% and 2.0%,
4.2 for Te and 7.3 for DHT. The difference between samples respectively for Te, and −1.5% and 2.8%, respectively for DHT)
that underwent three freeze–thaw cycles compared to either indicating that water can be used for dilution of human serum
608 s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610

samples containing Te and DHT above the HOQ. In addition,


a pooled incurred sample with a concentration above HOQ for
Te and 0.03 ng/mL for DHT was subjected to a 10-fold dilution
using either DCSF or water as the diluent (in replicates of 6).
Again the within-run precision values using DCSF or water as
the diluent were less than 15% (4.0% and 2.7%) for Te. This
indicates that a 10-fold dilution of the pooled incurred human
serum samples containing Te above the HOQ is valid. Data
for DHT was unavailable in this experiment due to its low
concentration in the pooled sample.

3.11. Stability in processed samples

The stability of Te and DHT in processed samples of human


serum was assessed by re-injecting extracted validation sam-
ples from a previous run after storage at 10 ◦ C for 48 h along
with a freshly prepared standard curve. The accuracy and
precision of these samples were found to be acceptable (less
than 15%) and no changes in the sensitivity for Te and DHT
derivatives were observed on re-injection, indicating that pro-
cessed samples are stable when stored at 10 ◦ C for at least
48 h.

3.12. Accuracy determination using standard addition


method

There are concerns about the accuracy of the results in


incurred samples since the standards and QC may not ade-
quately mimic the samples from dosed subjects. Potential
interference from metabolites or endogenous steroids, pro-
Fig. 3 – Determination of the accuracy of the concentration
tein binding, and matrix effects, is unknown and needs to
of Te and DHT in a pooled incurred sample using the plots
be evaluated. In addition, a surrogate matrix is used for
of analyte/internal standard area ratios against the
preparation of calibration standards and quality control sam-
additional amount of Te (A) and DHT (B) added.
ples in our method. Therefore, it is necessary to confirm
the accuracy of the data for Te and DHT in incurred sam-
ple using another method. To test this issue a standard
addition method was used [28]. The concentrations of Te
(0.800 ng/mL, n = 4, %CV = 1.8) and DHT (0.149 ng/mL, n = 4, the extrapolated and actual concentrations shows that the val-
%CV = 2.6) in a pooled incurred sample were determined idated method using DCSF as the surrogate matrix is accurate
using the validated method (refer to as ‘actual concentra- for determination Te/DHT concentrations in incurred sam-
tion’). Then known quantities of Te/DHT were added to this ples.
pooled sample to create a “calibration line” with Te/DHT con-
centrations of 0.4/0.2, 0.8/0.4, 1.2/0.6, 1.6/0.8, 2.0/1.0, 2.4/1.2, 3.13. Application to pharmacokinetic study
2.8/1.4, and 3.2/1.6 ng/mL in addition to the actual concentra-
tions. These spiked “standards” were extracted and analyzed Following validation the method was used to analyze samples
using the validated method. The analyte/internal standard from a pharmacokinetic efficacy and safety evaluation study
peak area ratios were plotted against the added concentra- of Te combined with dutasteride. It was an open-label study
tions of Te/DHT using GraphPad Prisma 5 software and linear with twice daily oral doses of Te (150–400 mg) co-administered
regression was performed with 1/x2 weighting (see Fig. 3). with 0.25 mg dutasteride compared with 400 mg Te alone and
The standard addition lines then were extrapolated to the 0.25 mg dutasteride alone to the hypogonadal patients. The
x-intercept, with the concentration of Te and DHT in the serum concentrations of Te and DHT were determined as
pooled incurred sample being equal to the absolute value described in the method. Blood samples were drawn pre-dose
of the x-intercept. The x-intercept for Te was −0.863 indi- and at intervals from 0.5 to 24 h post-dose. The pharmacoki-
cating that the extrapolated Te concentration of this pooled netic profiles of two subjects are shown (Fig. 4). Subject 1
incurred sample was 0.863 ng/mL. Comparing the extrapo- was dosed twice with 400 mg Te only while subject 2 was
lated value with the actual concentration (0.800 ng/mL) gives dosed twice with 400 mg Te combined with 0.25 mg dutas-
a percent difference of 7.9%. Similarly, the extrapolated DHT teride. The maximum concentrations (Cmax ) of Te were 26.4
concentration in the pooled incurred sample was 0.155 ng/mL and 34.7 ng/mL for subjects 1 and 2, respectively, with a Tmax at
corresponding to a 4.0% difference from the actual concentra- 0.5 h for both patients. The maximum concentrations (Cmax ) of
tion of DHT (0.149 ng/mL). The excellent agreement between DHT were 4 ng/mL at 0.5 h (Tmax ) and 0.13 ng/mL at 2 h for sub-
s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610 609

references

[1] Basaria S, Dobs AS. Hypogonadism and androgen


replacement therapy in elderly men. Am J Med
2001;110:563–72.
[2] Qoubaitary A, Swerdloff RS, Wang C. Advances in male
hormone substitution therapy. Expert Opin Pharmacother
2005;6:1493–6.
[3] Steers WD. 5Alpha-reductase activity in the prostate.
Urology 2001;58:17–24.
[4] Heracek J, Richard H, Martin H, Luboslav S, Jana S, Jitka K, et
al. Tissue and serum levels of principal androgens in benign
prostatic hyperplasia and prostate cancer. Steroids
2007;72:375–80.
[5] Bartsch G, Rittmaster RS, Klocker H. Dihydrotestosterone
and the concept of 5alpha-reductase inhibition in human
benign prostatic hyperplasia. World J Urol 2002;19:413–25.
[6] Page ST, Amory JK, Bowman FD, Anawalt BD, Matsumoto
AM, Bremner WJ, et al. Exogenous testosterone (T) alone or
with finasteride increases physical performance, grip
strength, and lean body mass in older men with low serum
T. J Clin Endocrinol Metab 2005;90:1502–10.
[7] Thompson IM. Chemoprevention of prostate cancer: agents
and study designs. J Urol 2007;178:9–13.
[8] Gleave M, Qian J, Andreou C, Pommerville P, Chin J, Casey R,
et al. The effects of the dual 5alpha-reductase inhibitor
dutasteride on localized prostate cancer—results from a
4-month pre-radical prostatectomy study. Prostate
Fig. 4 – Pharmacokinetic profiles for Te (A) and DHT (B) from
2006;66:1674–85.
the hypogonadal patients after daily administration of [9] Amory JK, Page ST, Bremner WJ. Oral testosterone in oil:
400 mg Te Alone or 400 mg Te co-administered with pharmacokinetic effects of 5alpha reduction by finasteride
0.25 mg dutasteride. or dutasteride and food intake in men. J Androl 2006;27:72–8.
[10] Shrivastav TG, Kanaujia PK. Direct radioimmunoassay for
the measurement of serum testosterone using 3H as label. J
Immunoassay Immmunochem 2007;28:127–36.
[11] Funderburgh LJ, Zipf WB, Sotos JF. Direct measurement of
jects 1 and 2, respectively. As expected, dutasteride effectively testosterone in a pediatric center, with use of a
suppressed DHT formation. To date, approximately 1300 clin- radioimmunoassay kit and unextracted serum. Clin Chem
ical samples have been analyzed using the method described 1983;29:1796–8.
[12] Warner MH, Kane JW, Atkin SL, Kilpatrick ES.
here.
Dehydroepiandrosterone sulphate interferes with the
Abbott Architect direct immunoassay for testosterone. Ann
Clin Biochem 2006;43:196–9.
4. Conclusion [13] Middle JG. Dehydroepiandrostenedione sulphate interferes
in many direct immunoassays for testosterone. Ann Clin
A semi-automated sample preparation method in 96-well Biochem 2007;44:173–7.
plate format for the determination of Te and DHT concentra- [14] Cawood ML, Field HP, Ford CG, Gillingwater S, Kicman A,
Cowan D, et al. Testosterone measurement by
tions in human serum was developed and validated over the
isotope-dilution liquid chromatography–tandem mass
range of 0.2–40 and 0.01–2 ng/mL, respectively. The method spectrometry: validation of a method for routine clinical
was proven to be highly sensitive, accurate and selective, practice. Clin Chem 2005;51:1472–9.
making it suitable for high-throughput quantitative analy- [15] Turpeinen U, Linko S, Itkonen O, Hämäläinen E.
sis of Te and DHT in clinical studies. The LOQ for analysis Determination of testosterone in serum by liquid
of Te can be modified to 0.01 ng/mL if greater sensitivity is chromatography–tandem mass spectrometry. Scand J Clin
Lab Invest; in press.
needed. Also, the method can be adapted for the simultaneous
[16] Shackleton CL. Mass spectrometry: application to steroid
determination of Te, DHT, androsterone, trans-androsterone,
and peptide research. Endocr Rev 1985;6:441–86.
dehydroepiandrosterone, and ␤-DHT from the same aliquot. [17] Salermo R, Moneti G, Forti G, Magini A, Natali A, Saltutti C,
et al. Simultaneous determination of testosterone,
dihydrotestosterone and 5␣-androstan-3␣,-17␤-diol by
Acknowledgements isotopic dilution mass spectrometry in plasma and prostatic
tissue of patients affected by benign prostatic hyperplasia
effects of 3-month treatment with a GnRH analog. J Androl
The authors would like to acknowledge Matt Cyronak, 1988;9:234–40.
Jacob Dunbar, Michael Leonard, and Russ Yeager from the [18] Griffiths WJ, Liu S, Alvelius G, Sjovall J. Derivatisation for the
Department of Drug Metabolism and Pharmacokinetics at characterisation of neutral oxosteroids by electrospray and
GlaxoSmithKline for their assistance with this project. matrix-assisted laser desorption/ionisation tandem mass
610 s t e r o i d s 7 3 ( 2 0 0 8 ) 601–610

spectrometry: the Girard P derivative. J Rapid Commun Mass [24] Mao L, Sun C, Zhang H, Li Y, Wu D. Determination of
Spectrom 2003;17:924–35. environmental estrogens in human urine by high
[19] Nishio T, Higashi T, Funaishi A, Tanaka J, Shimada K. performance liquid chromatography after fluorescent
Development and application of electrospray-active derivatization with p-nitrobenzoyl chloride. Anal Chim Acta
derivatization reagents for hydroxysteroids. J Pharma 2004;522:241–6.
Biomed Anal 2007;44:786–95. [25] Kalhorn TF, Page ST, Howald WN, Mostaghel EA, Nelson PS.
[20] Higashi T, Shimada K. Derivatization of neutral steroids to Analysis of testosterone and dihydrotestosterone from
enhance their detection characteristics in liquid biological fluids as the oxime derivatives using
chromatography–mass spectrometry. Anal Bioanal Chem high-performance liquid chromatography/tandem mass
2004;378:875–82. spectrometry. Rapid Commun Mass Spectrom 2007;21:
[21] Higashi T, Yamauchi A, Shimada K, Koh E, Mizokami A, 3200–6.
Namiki M. Determination of prostatic androgens in 10 mg of [26] Quirke JME, Adams CL, Van Berkel GJ. Chemical
tissue using liquid chromatography–tandem mass derivatization for electrospray ionization mass
spectrometry with charged derivatization. Anal Bioanal spectrometry. 1. Alkyl halides, alcohols, phenols, thiols, and
Chem 2005;382:1035–43. amines. Anal Chem 1994;66:1302–
[22] Kushnir MM, Rockwood AL, Roberts WL, Pattison EG, Bunker 15.
AM, Fitzgerald RL, et al. Performance characteristics of a [27] Ashcroft WR, Beal MG, Joule JA. Synthesis of the pyridine
novel tandem mass spectrometry assay for serum analogues of phthalide. J Chem Soc Perkin Trans
testosterone. Clin Chem 2006;52:120–8. 1981;1:3012–
[23] Kirk JM, Tarbin J, Keely BJ. Analysis of androgenic steroid 5.
Girard P hydrazones using multistage tandem mass [28] Robison JW. Undergraduate instrumental analysis. New
spectrometry. Rapid Commun Mass Spectrom York: Marcel Dekker;
2006;20:1247–52. 2004.

Vous aimerez peut-être aussi