<<

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

Journal of Chromatography A, xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Journal of Chromatography A

j ournal homepage: www.elsevier.com/locate/chroma

Review article

Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human perturbations

a,b,c a,b,c c,d e

Fabienne Jeanneret , David Tonoli , Michel F. Rossier , Martial Saugy ,

a a,c,∗

Julien Boccard , Serge Rudaz

a

School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva 4, Switzerland

b

Human Protein Sciences Department, University of Geneva, 1211 Geneva 4, Switzerland

c

Swiss Centre for Applied Human Toxicology, Geneva, Switzerland

d

Institut Central (ICHV), Hôpital du Valais, Sion, Switzerland

e

Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Epalinges, Switzerland

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

Article history: This review presents the evolution of steroid analytical techniques, including gas chromatography

Received 29 April 2015

coupled to mass spectrometry (GC–MS), immunoassay (IA) and targeted liquid chromatography cou-

Received in revised form 22 June 2015

pled to mass spectrometry (LC–MS), and it evaluates the potential of extended steroid profiles by a

Accepted 1 July 2015

metabolomics-based approach, namely steroidomics. regulate essential biological functions

Available online xxx

including growth and reproduction, and perturbations of the steroid homeostasis can generate serious

physiological issues; therefore, specific and sensitive methods have been developed to measure steroid

Keywords:

concentrations. GC–MS measuring several steroids simultaneously was considered the first historical

Steroid analysis

Review standard method for analysis. Steroids were then quantified by immunoassay, allowing a higher through-

Chromatography put; however, major drawbacks included the measurement of a single compound instead of a panel and

Mass spectrometry cross-reactivity reactions. Targeted LC–MS methods with selected reaction monitoring (SRM) were then

Human disease introduced for quantifying a small steroid subset without the problems of cross-reactivity. The next step

Metabolomics was the integration of metabolomic approaches in the context of steroid analyses. As metabolomics tends

Steroidomics

to identify and quantify all the metabolites (i.e., the metabolome) in a specific system, appropriate strate-

gies were proposed for discovering new biomarkers. Steroidomics, defined as the untargeted analysis of

the steroid content in a sample, was implemented in several fields, including doping analysis, clinical

studies, in vivo or in vitro toxicology assays, and more. This review discusses the current analytical meth-

ods for assessing steroid changes and compares them to steroidomics. Steroids, their pathways, their

implications in diseases and the biological matrices in which they are analysed will first be described.

Then, the different analytical strategies will be presented with a focus on their ability to obtain relevant

information on the steroid pattern. The future technical requirements for improving steroid analysis will

also be presented.

© 2015 Elsevier B.V. All rights reserved.

Contents

1. Introduction ...... 00

2. Steroidogenesis and steroid metabolism ...... 00

3. Situations of steroid dysregulation ...... 00

3.1. Disorders of synthesis and metabolism of steroids ...... 00

3.2. Steroid disruption associated with “global public health problems”: diseases such as diabetes, cancer or male infertility,

as well as toxic environmental exposure such as endocrine disrupting chemicals ...... 00

Corresponding author at: School of Pharmaceutical Sciences, University of Geneva, 20 Bd d’Yvoy, 1211 Geneva 4, Switzerland. Tel.: +41 22 379 63 36; fax: +41 22 379 68 08.

E-mail address: [email protected] (S. Rudaz).

http://dx.doi.org/10.1016/j.chroma.2015.07.008

0021-9673/© 2015 Elsevier B.V. All rights reserved.

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

2 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

4. Biological matrices and models for steroid studies ...... 00

4.1. Human matrices: urine, blood, saliva and semen analysis ...... 00

4.2. Tissue and cell models ...... 00

5. Targeted steroid analysis: current techniques and future directions ...... 00

6. Untargeted steroid analysis: steroidomics, an “omics”-based approach ...... 00

6.1. Survey of current steroidomic applications...... 00

6.1.1. Steroidomics in human matrices ...... 00

6.1.2. Steroidomics in animals and cell models ...... 00

6.2. Proposed steroidomic workflow: reduction of data dimensionality and identification assisted by database filtering ...... 00

7. Conclusion ...... 00

Acknowledgments ...... 00

References ...... 00

1. Introduction of diseases or one of their consequences; therefore, other path-

ways than steroidogenesis may be dysregulated, and information

Steroid analysis was first developed for the diagnosis of

about those pathways can provide valuable indications regarding

endocrine diseases. Initial gas chromatographic (GC) methods

biological mechanisms. In the context of metabolomics, which

developed in the 1950s were substituted by high-throughput

aims at the identification and the quantification of all metabo-

immunoassays (IA) since 1970 [1]. However, IA techniques often

lites (m/z < 1000 Da) in a biological system, steroidomics represents

reveal problems of cross-reactivity, particularly in the case of

a sub field aiming to analyse the steroid content of a system.

steroids. For example, screening for congenital adrenal hyperpla-

Steroidomics was first described in 2004 as the “characterisa-

sia (CAH) based on the measurement of 17␣-hydroxyprogesterone

tion and quantification of metabolic profiles of steroids” [21].

unfortunately resulted in many false positive cases [2,3]. It was

Steroid analysis represents a vast range of investigation as already

therefore recommended to perform both screening and confir-

867 “Steroid and Steroids Derivatives” have been reported in The

mation of CAH by a liquid chromatography method coupled to

Human Metabolome Database (HDMB, version 3.6) [22]. Therefore,

mass spectrometry (LC–MS) and to measure a panel of steroids

steroids represent an important part of the metabolomic content,

instead of a unique metabolite. This approach represents the cur-

particularly in humans, where steroidogenesis and pathologies

rent trend for steroid analysis. The increasing levels of endocrine

related to steroid disturbances are of the utmost interest. The topic

disrupting chemicals (EDCs) found in the environment have clearly

of this review is to compare the current technologies in the context

contributed to evaluating overall steroid perturbations as a pub-

of the evolution of steroid analysis in the direction of steroidomics.

lic health problem [4–6]. Today, steroid dysregulations are not

After an overview of steroid structures, the established ana-

only related to endocrine gland dysfunction but also to numerous

lytical methods such as IA, GC–MS and LC–MS, including the

important diseases, such as cancer, diabetes, cardiovascular dis-

hyphenation of LC with high-resolution MS will be described.

orders and environment-linked male and female infertility. The

Steroidomics will be critically discussed to evaluate whether this

increasing number of publications on steroid analysis may also

new approach could improve the current analytical methodologies

have arisen from the establishment of mass spectrometry (MS),

and contribute to additional pertinent information.

especially in combination with liquid chromatography (LC), as a

routine analysis in clinical laboratories. LC–MS is indeed taking

an increasing part in clinical laboratories principally due to the 2. Steroidogenesis and steroid metabolism

improved robustness and user-friendliness of the LC–MS systems in

the last few years for the analysis of small molecules, either endoge- Steroids contain a structure formed by four fused rings. The rings

nous or exogenous [7–11]. Hence, the growing interest in steroid of the steroid nucleus (called gonane) are designated by letters from

analysis is directly related to the development and publication A to D, and the carbon atoms are sequentially numbered (Fig. 1A).

of new analytical methodologies, from GC to IA, and for approxi- The synthesis of steroids from their unique precursor, cholesterol,

mately the past 10 years, to LC–MS. Triple quadrupole instruments is called steroidogenesis. Cholesterol contains 27 carbon atoms

(QqQ) still represent the gold standard MS platform for quan- (Fig. 1B), and the five classes composing the steroid family present a

tification, but instruments offering simultaneous qualitative and core structure of 21, 19 or 18 carbon atoms. Progestogens, glucocor-

quantitative (Qual/Quant) possibilities using high resolution MS ticoids and mineralocorticoids have a core structure of 21 carbon

such as Orbitrap or Time-of-Flight (TOF) are now entering clinical atoms, whereas estrogens and androgens have 18 and 19 carbon

or production laboratories [12]. These high-resolution instruments atoms, respectively (Fig. 1C).

now present performances in terms of quantification approaching Steroids are synthesised in numerous organs, including the

QqQ instruments [13,14]. In chromatography, the introduction of adrenal glands, testis, ovaries, brain, placenta, and adipose tis-

ultra high-pressure liquid chromatography (UHPLC) and core–shell sue [23,24]. Two major sites of synthesis are the adrenal glands,

particles permitted improvements in terms of analysis time and where mineralocorticoids, glucocorticoids and adrenal androgens

chromatographic resolution [15,16]. are produced, and the gonads for the production of progestogens,

System biology studies are now investigated by different omics androgens and estrogens [23]. An overview of steroidogenesis is

strategies, and scientists must then address the huge amounts of reported (Fig. 2), but slightly different pathways are observed in

data generated, for example, from metabolomics, transcriptomics, each steroidogenic tissue, resulting from distinct enzymatic distri-

and proteomics. Advances in chemometrics with in particular butions among tissues [25]. Two classes of enzymes are involved

multi-block models allow to process the data as a whole [17,18], in steroidogenesis, namely the cytochrome P450 family (CYP) and

and one of the ultimate steps remains the mechanistic eluci- the two dehydrogenases ((HSDs) (i) the short-chain

dation of the perturbation, such as discovering the biochemical alcohol dehydrogenase/reductase and (ii) the aldo-keto reductase)

pathways affected by a disease or a toxic exposure. Recent tools [25,26]. The CYP enzymes catalyse reactions of hydroxylation and

import results from different platforms and annotate the altered cleavage, and the HSDs reduce or oxidise steroids, often by con-

pathways [19,20]. Steroid perturbations can be either the cause verting secondary alcohol groups to ketones (or vice versa). Other

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 3

A) B) 26

27 25 21 24 23 18 22 20

12 12 17 17 11 13 19 11 13 C D 16 16 14 14 15 15 1 9 1 9 2 10 8 10 A B 2 8 3 5 7 3 5 7 6 4 6 4 HO

C) 21

18 18 18 20

12 17 12 12 19 11 13 17 17 19 11 13 11 16 13 16 14 16 15 14 14 1 9 15 15 9 2 10 8 1 1 9 2 10 8 2 10 8 3 5 7 6 3 5 7 3 5 7 4 6 6 4 4 C-21 C-19 C-18

Progestogens Androgens Estrogens

Fig. 1. Steroid structures. Molecular representation of (A) steroid nucleus core, (B) cholesterol, and (C) steroid classes.

Fig. 2. Steroidogenesis. Major steroids from the , gluco- and mineralo-corticoid, androgen, and estrogen classes are depicted in grey, green, blue, and red,

respectively. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

4 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

reactions are observed such as a dehydrogenation accompanied by breast cancer therapy [35,36]. Another important issue concerns

the isomerisation of the double bond from carbons 5–6 to carbons male infertility, which is associated with poor semen quality.

4–5 by specific HSDs or the reduction of double bonds, such as the Testicular Dysgenesis Syndrome (TDS), which includes impaired

formation of 5␣-dihydrotestosterone (DHT) from testosterone by spermatogenesis in addition to urogenital malformations, such as

the 5␣-reductases (this latter class of enzymes is often grouped hypospadias, cryptorchidism, and testicular cancer, is recognised

with HSDs). as a rising problem in industrialised countries for the last 50–70

Steroids are extensively metabolised by phase II reactions such years [37]. As demonstrated by many studies, TDS and androgen

as sulphation or glucuronidation, which mainly occur in the liver insufficiency [38] may result from perturbations during foetal life

and kidneys. Conjugated steroids are more easily excreted in but also from important changes in environmental factors and

urine compared to their parent forms. Steroid sulphation has been chemical exposure. In 2005, Wild et al. proposed, as additional

described as a way to excrete steroids, but several sulphated information to the genome, the concept of exposome, which cor-

steroids also serve as sources of steroid precursors and may have responds to the “life-course environmental exposures (including

important functions, such as DHEAS in the adrenal glands or pre- lifestyle factors), from the prenatal period onwards” [39]. Among

gnenolone sulphate in brain tissue [27]. From the 17 functionally environmental contaminants, the U.S. Environmental Protection

important steroids presented in Fig. 2, more than 60 additional Agency (EPA) has defined an EDC as “the exogenous agent that

metabolites have been observed in plasma and urine [28,29]. interferes with synthesis, secretion, transport, metabolism, binding

Metabolites present different isomeric forms, according to the action, or elimination of natural blood-borne hormones that are

position of the polar moieties and conjugated groups. The main present in the body and are responsible for homeostasis, reproduc-

metabolites associated with each class of steroids are cited in Fig. 3. tion, and developmental process” [40]. EDCs are both natural and

This list is far from being exhaustive, as 867 steroids are described man-made substances such as phytoestrogens, pesticides or indus-

in HMDB; moreover, other databases, such as LIPID MAPS [30] also trial chemicals; food seems to be the main contamination route

have extended lists of steroids. In summary, steroids present very for humans, but exposure occurs also from the air and through the

similar backbone structures, and differences come mainly from the skin [4,6]. It is now recognised that EDCs affect hormonal function

presence of additional functional groups, their localisation, their by interfering with steroidogenic enzymes [23] and altering gene

relative position (␣- or ␤-) and finally the ring saturation. These expression by triggering epigenetic modifications [41,42]. EDCs

structural similarities and their low concentrations represent an targets are not restricted to nuclear hormone receptors such as AR

analytical challenge to obtaining specific and sensitive measure- or ER; interactions with neurotransmitter and orphan receptors

ments. (such as the aryl hydro-carbon receptor, AhR) are also described

[6]. The multiple EDC mechanisms of action lead to a wide range

3. Situations of steroid dysregulation of effects, and therefore EDCs are cited as factors in many diseases

including not only the already mentioned cancer, diabetes and TDS

Diseases with direct defects in steroidogenesis or steroid but also female infertility [43,44], obesity and metabolic disorders

metabolism enzymes were initially reported, but numerous other [45], and pathologies linked to the immune system (for reviews

disorders associated with concomitant disruptions of steroid see [5,6,46]). Therefore, numerous public health diseases can be

homeostasis or toxic exposures to EDCs have also recently been directly related with steroid perturbations even if the direct link

described. with an EDC exposure remains difficult to prove. As an example,

structured exposure studies with anti-androgenic substances or

3.1. Disorders of synthesis and metabolism of steroids estrogens were insufficient to draw a complete scheme inducing

TDS [47,48]. In fact, because humans are not massively exposed

Several diseases associated with direct steroid perturbations, to a single compound but to low doses of mixtures of different

due to dysfunctions in steroidogenesis or steroid metabolism EDC classes, effects can be additive or synergic, and moreover, low

enzymes, are shown in Table 1. In some cases, the similarities of doses show non-traditional dose-response [49,50].

syndromes of endocrine diseases and the benefits of appropriate Finally, the use of steroids as therapeutic drugs is another

treatment have led to the mandatory measurements of a set of important domain to be cited, such as glucocorticoids, which

steroids to establish the right diagnosis (e.g., in case of CAH). Defi- have anti-inflammatory and immunosuppressive properties and

ciencies in different enzymes drive various forms of pathologies, are therefore used for the treatment of many diseases. Their major

and the final diagnosis is only possible by the measurement of sev- drawback is the development of diabetes or steroid-induced hyper-

eral steroids. For instance, the differential diagnosis of hirsutism glycaemia [51]. Anabolic androgenic steroids are often used in

(excessive facial and body hair), which is a common symptom of sports as performance enhancers, but these compounds have also

CAH, Cushing’s disease and polycystic ovary syndrome (PCOS), is been used as therapeutic agents since the 1940s for the treatment

solely obtained by the determination of a restricted number of of trauma and burns and are now very useful in the treatment

specific steroids. of cachexia associated with chronic disease states [52,53]. How-

ever, various adverse effects are associated with their use, such

3.2. Steroid disruption associated with “global public health as the development of gynecomastia, depression, hepato- and

problems”: diseases such as diabetes, cancer or male infertility, as cardiotoxicity, and cancer [54]. The benefits of combined hor-

well as toxic environmental exposure such as endocrine disrupting monal contraception (aside from the prevention of unwanted

chemicals pregnancy), such as a protective effect against epithelial ovarian

or endometrial cancer, counterbalance the risks linked to venous

In recent years, various pathologies have also been associated thromboembolism [55]. Menopausal hormone therapy relieves

with steroid alterations. As an example, patients with prostate unpleasant symptoms of menopause and diminishes osteoporosis

cancer have a worse prognosis if they display an elevated con- risks, but higher incidences of ovarian and endometrial can-

centration of testosterone and are therefore treated by chemical cers were observed. However, controversial results are obtained

or surgical androgen depletion [33]. In the case of castration- depending on the type of cancer, composition and duration of hor-

resistant prostate cancer, other therapies can be applied, such as mone therapy [56,57].

targeting the androgen receptor (AR) signalling [34]. In a parallel Hence, these various pathologies associated with steroid pertur-

manner, action on the estrogen receptor (ER) must be blocked for bations need to be better understood and require various analytical

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 5

11α-Hydroxyprogesterone

21-Hydroxypregnenolone Corcoids 20α-Hydroxyprogesterone

7α-Hydroxypregnenolone 11β-Hydroxyprogesterone Corcoids

Pregnenolone Progesterone 5β--3,20-dione 3α-Hydroxy-5 β-pregnane-20-one Pregnenolone 3β-sulfate 5α-Pregnane-3,20-dione 3α -Hydroxy-5α-pregnane-20-one

α α 5α-Pregnan-3α,20α-diol

17α-Hydroxypregnenolone 17α-Hydroxyprogesterone Corcoids 5 -Pregnane-20 -ol-3-one α α 17α,21-Dihydroxypregnenolone 17 ,20 -Dihydroxypregn-4-en-3-one

11β,17α,21-Trihydroxypregnenolone Corcoids

11-Deoxycorcosterone 5α-Dihydrodeoxycorcosterone Allotetrahydrodeoxycorcosterone

Aldosterone hemiacetal

Aldosterone 11β,21-Dihydroxy-3,20-dioxo-5 β-pregnane-18-al 3α ,11β,21-Trihydroxy-20-oxo-5β-pregnane-18-al

18-Hydroxycorcosterone 11-Dehydroxycorcosterone 21-Hydroxy-5β-pregnane-3,11,20-trione

Corcosterone 11β,21-Dihydroxy-5 β-pregnane-3,20-dione Tetrahydrocorcosterone 3 α,21-Dihydroxy-5β-pregnane-11,20-dione 11β-Hydroxyprogesterone 3 α,20α,21-Trihydroxy-5β-pregnane-11-one

21-Deoxycorsol

Androgens Corsol 11β,17α,21-Trihydroxy-5 β-pregnane-3,20-dione Urocorsol Cortol

Corsone 17α,21-Dihydroxy-5β-pregnane-3,11,20-trione Urocorsone Cortolone

11-Deoxycorsol

17α-Hydroxypregnenolone α

DHEAS DHEAG 17 -Hydroxyprogesterone 11β-Hydroxyandrostanedione Adrenosterone DHEA 7α-HydroxyDHEA Eocholanolone 3α-glucuronide 5β- Eocholanolone

α

16α-HydroxyDHEA 16α-Hydroxyandrostenedione Eocholanolone 3 -sulfate Androsterone 3α-glucuronide

Androstenedione 5α-Androstanedione Androsterone

5- Androsterone 3α-sulfate 7α-Hydroxyandrostenedione 19-Hydroxyandrostenedione 19-Oxoandrostenedione Estrogens Testolactone 7α-Hydroxytestosterone Testosterone 19-Hydroxytestosterone 19-Oxotestosterone Estrogens

5α-Dihydrotestosterone Androstanediol 5β-Dihydrotestosterone Testosterone 17β-glucuronide Testosterone 17β-sulfate

Estrone 3-sulfate 3-glucuronide 17α- 2-Methoxyestrone 3-glucuronide 2-Hydroxyestrone 2-Methoxyestrone 2-Methoxyestrone 3-sulfate Estrone 16α-Hydroxyestrone

17β-Estradiol Estriol 16α-glucuronide 2-Methoxyestradiol 3-glucuronide 2-Hydroxyestradiol 2-Methoxyestradiol 2-Methoxyestradiol 3-sulfate 6β-Hydroxyestradiol 17β-Estradiol 3-glucuronide

17β-Estradiol 3-sulfate

Fig. 3. Steroidogenesis and steroid metabolism pathways.

Adapted from Kegg pathways [31].

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

6 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

Table 1

Examples of disorders associated with dysregulation of steroid synthesis or metabolism. Table is modified from [32].

Disease Aetiology Affected steroids

Congenital adrenal hyperplasia Several mutations (mainly in CYP21A1) affecting the synthesis of Glucocorticoids (−),

(CAH) glucocorticoids (−) and therefore activating the hypothalamic pituitary mineralo-corticoids (−/+),

adrenal axis androgens (+)

Congenital lipoid adrenal Mutation affecting the gene coding for StAR and preventing the transport All steroids (−)

hyperplasia of cholesterol into mitochondria, the common initial step of

steroidogenesis

Polycystic ovary syndrome (PCOS) Genetic mutations and environmental factors Androgens (+)

Cushing’s syndrome/Cushing’s Tumour, dysregulation of cellular signalling or illegitimate expression of Cortisol (+)

disease GPCR in adrenal fasciculata cells/Dysregulation of ACTH secretion

Primary aldosteronism Mutation of various regulatory genes (KCNJ5, ATP1A1, ATP2B3, CACNA1D) Aldosterone (+)

leading to overexpression or stimulation of CYP11B2

Apparent mineralocorticoid excess Genetic mutation or environmental factors affecting the 11b-HSD type 2 Aldosterone (−)

(AME) enzyme and therefore preventing the metabolism of cortisol into cortisone

5␣-reductase deficiency Mutation of SRD5A2 gene Testosterone (+), DHT (−)

Aromatase deficiency Mutation of CYP19A1 gene Androgens (+), estrogens (−)

developments for measuring steroid concentrations in the biolog- blood sampling is more convenient for many patients than 24-h

ical matrix where the perturbations occur. urine sampling, which is more prone to pre-analytical errors such

as false sampling time or missing micturition in the 24 h. Measure-

4. Biological matrices and models for steroid studies ments are described in serum and plasma as well as dried blood

spots (DBS) that reduce greatly the invasiveness of the whole pro-

Steroids are measured in various human matrices, such as tis- cedure [66]. Blood has become the matrix of choice for a “restricted”

sue, blood, urine, semen, hair and saliva. The results can be affected panel of steroid analysis and for a first screening for steroid disor-

by technical factors related to the analytical method (e.g., cross- ders. It is to be noted that steroid blood concentrations vary from

reactivity of IA) and by biological factors such as age, sex, diurnal pg/ml (estrogens) to ng/ml (testosterone, cortisol, etc.) and until

rhythm, and for women, pregnancy and phase of menstruation. The higher values such as g/ml for DHEAS.

numerous biological factors make the establishment of reference Saliva is described as an alternative non-invasive matrix to

values rather difficult. A very detailed review of these preclinical replace serum but is currently dedicated to specific diagnosis and

challenges for steroid analysis was published in 2010 by Ceglarek not to obtain a panel of steroids. Various saliva collection devices

et al. [58], describing the biological factors that can affect the and different sampling techniques such as direct spitting with

steroid content. Steroids are also measured in numerous differ- or without stimulation are used; the results can vary greatly for

ent in vitro matrices (cell lines, different tissues such as brain, steroids and other compounds [67]. Cushing’s diagnosis is the main

skin, etc.) to support mechanistic investigations of steroid pertur- application reported for saliva. Cortisol has a circadian rhythm, with

bations in which the analytical challenges may differ from in vivo a lower secretion rate near midnight [68]. Patients with Cushing’s

matrices. syndrome lack a circadian rhythm, and the diagnosis can be made

based on 24-h urine or midnight saliva [69]. Testosterone is also

measured in saliva and was proposed to replace the measurement

4.1. Human matrices: urine, blood, saliva and semen analysis

of free testosterone in serum [70]. Salivary steroids correspond to

the fraction of steroids that are unbound to proteins and there-

Urine was described as the gold standard matrix to establish

fore their concentrations are approximately 1% of the blood values

accurate diagnoses for diseases associated with steroid synthe-

[58,71].

sis and metabolism [29], including newborn screening for inborn

Finally, the rising male infertility observed in recent years has

errors of steroidogenesis [59]. These methods involved the hydrol-

led to the study of biomarkers of infertility [72] in semen, where

ysis of steroid conjugates [60]. Because urinary steroid excretion

steroids were directly measured [73,74]. To the best of our knowl-

presents a circadian rhythm, 24-h urine collections are often

edge, there is currently no established reference values for this

recommended [61]. Doping analysis is one of the other major

matrix. Because human steroid perturbation studies cannot always

applications of steroid analysis in urine, but for this domain spot

be addressed due to the structure of clinical studies, in vitro analysis

urines are often used. Screening of anabolic androgenic steroids

is of the utmost importance and represents an important emerging

are performed following lists of banned compounds coming from

topic.

the World Anti-Doping Agency (WADA); screened compounds can

be exogenous or endogenous substances [62]. As example, testos-

terone is used in sports to maximise performances and suspicion 4.2. Tissue and cell models

of its misuse is first based on the measurement of a testos-

terone/epitestosterone ratio higher than 4 and then confirmed by In addition to the measurements in the aforementioned biologi-

profiling different steroids such as androsterone, etiocholanolone, cal matrices, studies of steroidogenic tissue remain fundamental to

␣ ␣ ␤

5 --3 ,17 -diol, and 5␤-androstane-3␣,17␤-diol [63]. obtaining a deeper understanding of steroid perturbation mech-

The most recent methodologies in doping analysis deal directly anisms. In vitro models were therefore developed for studying

with conjugated steroids without the hydrolysis step [64]. Urinary tissues, such as the adrenal glands, the gonads, the brain, and the

steroid concentrations are often indicated by ratios of concentra- skin.

tion (moles or gram) on creatinine or 24 h. Range order is from low Human adrenal steroidogenesis can be studied in primary

to high ␮g/24 h for urinary steroids analysed in clinical chemistry cultures of adrenocortical cells, but the requirements of fresh

[29]. tissues and important variability between donors led to the

Although blood sampling is invasive and represents a unique development of cell lines from adrenocortical carcinomas [75].

time point and not a picture over a long time span as it is H295R is the most used cell line for which the Organisation

the case for urine, blood-based matrices have become more fre- for Economic Co-operation and Development (OECD) has estab-

quently used for steroid analysis [65]. Despite its invasive character, lished guidelines for the methodological screening of H295R

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 7

steroidogenesis disruption by putative EDCs. According to the The mass spectrometry fragmentation of steroids has been stud-

guidelines, results were based on the determination of testosterone ied since 1950, but the first publication of a GC–MS study occurred

and 17␤-estradiol [76,77]. Because the H295R cell lines contain in 1964 [106]. The advantages of GC–MS are the moderate instru-

most of the steroidogenic enzymes, recent studies analysed a higher ment costs and the possibility to analyse an extended panel of

number of steroids rather than only testosterone and 17␤-estradiol non-conjugated steroids (e.g., 30–70 steroids). However, the large

[78–81]. number of steroids measured has the drawback of lacking a simple

Human male gonad steroidogenesis is studied in testes and tes- and interpretable presentation of the data; recent visualisation in

ticular fragments from donors [82]. There is currently no human heat maps or other forms by separating the steroids into blocks

testicular cell line similar to the rodent Leydig cells, which pro- have ameliorated these issues [28,29,107]. An important draw-

duce androgens such as testosterone, or DHEA back regarding GC–MS concerns the need to apply at least one

[83]. In vitro cultured testes are mainly used for screening EDCs derivatisation procedure, which reduces the throughput. However,

and then understanding their effects on fertility by measuring for GC–MS remains the method of choice for the diagnosis of particular

instance the testosterone production [84,85]. Studies on female diseases, where ratios of specific steroids are required [29]. Further

gonad steroidogenesis are principally made on ovarian granulosa details on the technical developments of GC–MS for steroids were

primary cells or derived cell lines [86,87]. Steroid diseases such as reviewed in 2010 [108] and therefore are not further discussed

polycystic ovary syndrome are investigated in these cell lines [88], here.

and, as in male gonads, the putative toxic effects of exogenous com- The hyphenation of LC with MS to analyse steroids had already

pounds are evaluated, such as steroidogenesis impairment caused been proposed 30 years ago [109] and was widely applied in the

by chemotherapy drugs [89]. 1990s due to the implementation of atmospheric ionisation sources

The brain is a particularly interesting steroidogenic tissue such as electrospray (ESI) [110]. In contrast to GC–MS, LC–MS does

because unlike the adrenal glands, steroidogenic enzymes are not require a derivatisation step, although it can be used to improve

present in different cell types, such as the neuron and the glia sensitivity [111–113]. From historical analytical methods measur-

[90,91]. Therefore, neurosteroids are only produced when various ing one or a few steroids in blood, determinations of up to about

cell types are present. Cellular models for studying neurosteroido- 15 steroids in one run are now reported [114–117]. Furthermore,

genesis have to contain different cells, such as in the organotypic commercially available kits comprising reagents, standards, qual-

culture of brain slides [92]. Neurosteroids were shown to have a ity controls and solid phase-extraction procedures are available for

protective effect on the central nervous system (CNS) [93], but the analysis of steroid panels (comprising from 10 to 16 steroids),

TM

many studies were performed in rodents. New cellular models including the CHS MSMS Steroids Tool Box from Perkin-Elmer

®

are regularly proposed, and a greater number of human studies [118] and the SteroIDQ Kit from Biocrates [119]. Diagnoses such

aiming at toxicological and fundamental research purposes can be as CAH, previously possible only by GC–MS or IA, are now made by

expected in the near future [94,95]. LC–MS [120]. It must be noted that while the most used ionisation

Skin, as a barrier against the external environment, is an impor- source in LC–MS remains ESI, some steroids present better response

tant organ in which local steroidogenic activity can be found. The factors using atmospheric pressure chemical ionisation (APCI) or

production of steroids is dependent on the cutaneous compart- atmospheric pressure photoionisation (APPI) [121]. A study has

ment that can produce glucocorticoids, androgens or estrogens compared these three ionisation sources (ESI, APPI and APCI) for

[96]. Modulation of this activity is proposed as a treatment for a panel of 12 main steroids. Results showed that estrogens and

inflammatory skin disorders or wound repair [97,98]. Primary kera- aldosterone are poorly ionised by ESI, and in several cases APPI

tinocyte cells or 3D in vitro epidermal models of human skin are has a better sensitivity than APCI but presents a lower coverage in

now produced and are proposed for investigating the mechanisms terms of the number of steroids that can be detected [122]. Semi-

of defects in the epidermal barrier [99]. automation, sensitivity, specificity, and multiplexing capabilities

to analyse several steroids in a small volume of sample are the

main advantages of LC–MS. Initially developed for blood analysis,

5. Targeted steroid analysis: current techniques and future

directions including DBS [123], LC–MS was then applied to other matrices,

including urine and saliva. Different sample preparation strategies

Steroid analyses were initially performed by GC, first with an have been developed for LC–MS, including protein precipitation

argon ionisation detector [100] and, since the mid-1960s, using (PP) and solid or liquid phase extraction (SPE and LLE). Recently, an

MS detection. Then, analyses were performed with IAs and, more extension of LC–MS methodologies was achieved in the direction of

recently, with LC–MS. These approaches are mainly done in a the simultaneous analysis of conjugated steroids in addition to the

targeted manner, meaning that only previously specified and/or unconjugated ones [124]. Targeted analyses in LC–MS are generally

identified compounds are measured. Immunoassays have the achieved by a highly specific acquisition mode called selected reac-

advantages of a reduced sample preparation, automation, and high tion monitoring (SRM), allowing very good quantification results:

throughput capabilities, but the major problems concern cross- each compound is analysed by targeting a specific transition that

reactivity between steroids. IA reaction is often performed with corresponds to a collision-induced dissociation of a precursor ion

a specific antibody targeting one antigen. Therefore, when many into a specific fragment. New MS technologies such as ion mobil-

compounds must be analysed for a diagnosis, different tests are ity mass spectrometry [125] could lead to an improvement in

required. Various IA kits are commercially available, but users must steroid analysis by adding an orthogonal degree of separation to the

test parameters such as sample spiking, cross-reactivity and dilu- LC–MS/MS strategies. This was demonstrated as a valuable tool for

tions if they do not use the exact same sample and species for distinguishing isomers with similar retention time (tR) or MS/MS

which the test was validated. Different studies have compared spectra and could also remove interfering peaks that co-elute with

results from various IAs for steroids such as 17␤-estradiol [101], steroids of interest [126,127]. Nevertheless other studies needed an

DHEAS [102] or testosterone [103]. Kit performances were found additional derivatisation step to obtain a separation of steroid iso-

to be heterogeneous in terms of recovery, linearity or specificity. mers [128,129]. Ion mobility mass spectrometry was both applied

Therefore, numerous societies and articles recommend a standard- for free and conjugated steroid forms and will be probably more

isation of steroid tests and the use of separation-based techniques, investigated in the next years. Even if a clear tendency could be

such as chromatographic methods coupled to mass spectrometry observed in the direction of MS-based detection methods, some

[103–105]. key points still need to be established, such as the interlaboratory

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

8 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

standardisation of steroid assays, which remains an issue [130]. classification could be summarised according to the acquisition

Comparison of testosterone measurements of identical samples by mode of data, which could be targeted metabolomics (or metabolic

different MS facilities showed slight differences but lower than with profiling, i.e., detection of a pre-determined number of metabo-

IA [131]. Standardisation improvement is now in progress with the lites) or untargeted metabolomics (metabolic fingerprinting, global

establishment of reference materials for dedicated hormones and metabolomic or global metabolic profile, i.e., analysis based on the

steroids to use as certified calibrators or controls [132]; for example, untargeted acquisition of a non-predetermined set of molecules

a validation of testosterone measurement by LC–MS was performed resulting in an extended view of the metabolites present in a

using a certified standard of the National Institute of Standards and sample). Briefly, the metabolomic workflow begins with a sam-

Technology (NIST) [133]. Recent reviews have compiled the litera- ple preparation, ranging from short to extended procedures. This

ture concerning steroid analysis by LC–MS, and readers are invited step is generally followed by a separation technique combined with

to consult the following reviews [9,10,65,108,134–136] for further a high-resolution MS. In the context of this review, nuclear mag-

details. netic resonance (NMR), another important tool in the field, will

not be discussed [139]. After data acquisition, signals are extracted

from the raw data by different processing strategies and then ana-

6. Untargeted steroid analysis: steroidomics, an

lysed by multivariate statistical analysis (MVA). A general MS-based

“omics”-based approach

metabolomic workflow with the different steps is illustrated in

Fig. 4. These steps, especially metabolite identification, which rep-

As previously indicated, steroidogenesis is complex, and, due to

resents the current bottleneck in metabolomics, will be discussed

its “cascade” structure, perturbations usually affect many steroids.

in greater details in our proposed steroidomic workflow after an

Therefore, analysis for diagnostic purposes and for understand-

overview of the current steroidomic applications.

ing the processes of homeostasis disruption requires not only

the simultaneous determination of the largest number of steroids

but also specificity, sensitivity, and standardisation. Unfortunately, 6.1. Survey of current steroidomic applications

for most of the current methods, the targeted approach is pri-

oritised, meaning that only a few steroids previously validated As the analyses of sugars and lipids are called glycomics and

for the assay are generally determined. The use of untargeted lipidomics, respectively, the analysis of the steroid content of

methods, based on holistic methodologies for studying steroid a sample was called steroidomics [21]. It was defined in the

dysregulations should now be prioritised and more deeply evalu- same publication as “the characterisation and quantification of

ated. Steroidomics could be considered as a metabolomics-based metabolic profiles of steroids”. Other appellations were used such

approach, focusing on a subset of the metabolome. Additional as “steroid metabolome”, “steroidome”, “steroidomic footprint-

definitions were proposed to specify metabolomic approaches, ing” and/or “extended steroid profiling”. The confusion of terms

such as metabolic profiling, metabolic fingerprinting or metabolic between targeted and untargeted approaches is also present in the

footprinting, and more recently targeted, post-targeted or untar- case of steroid analysis. Numerous steroidomic studies were asso-

geted metabolomics [137,138]. For the sake of simplicity, the ciated to targeted analysis, even if these scientific papers generally

MS metabo lomic workflow

Samples

Targeted approach Untargeted approach

ver y specific to targeted compounds specific or generic for a maximum metabolite coverage

at ory UHPLC/ LC, GC or CE

t labo r MS

we Analyzer Triple quadrupole (SRM) QTOF, Orbitrap, FT-ICR

Noise filterin g, peak picking, ion an-

Data processing targeted metabolites

at ory Data a nalysis y labo r

dr

Fig. 4. MS-based metabolomic workflow. Sample preparation. Specific sample preparations are used in targeted metabolomics, such as SPE or LLE in contrary to untargeted

metabolomics where non-selective techniques such as dilution or protein precipitation are used to obtain an extensive metabolite coverage. Separation technique. Capillary

electrophoresis (CE) and GC present an important separation power but stability issues of the coupling CE–MS and the derivatisation procedures needed for GC contributed

to the establishment of LC techniques as the gold standard for metabolomics. MS part. The use of ESI and APCI or APPI ionisation sources and the choice of polarity mode

depend of the metabolite physico-chemical properties. QqQ instruments are used for targeted approaches and high-resolution instruments such as QTOF, Orbitrap or FT-ICR

are particularly well adapted for untargeted metabolomics because of their sensitivity and mass accuracy capabilities. Data processing. Dedicated open-source or commercial

software programmes manage all the steps of data processing (for a detailed review on software for untargeted metabolomics, see [140]). Data analysis. Main multivariate

analyses used are principal component analysis, partial least squares, orthogonal partial least squares discriminant analysis, t-test, S-Plot, hierarchical cluster analysis, and

so on (for details, see [141,142]).

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 9

described the analysis of a relatively high number of steroids and/or dedicated SPE for conjugated androgens, a separation on a UHPLC

E

bile acids. Investigations mainly concerned diagnosis of disorders column and an untargeted QTOF-MS acquisition. A steroid

of synthesis and metabolism, but other topics were also explored; database for signal filtering and chemometric tools were then

these studies were performed in different matrices such as urine, applied. From our perspective, database filtering is one of the key

blood, tissue, and cell culture supernatant. points for the future of steroidomics. In the continuance of this

work, a similar strategy was applied by Jeanneret et al. to search

6.1.1. Steroidomics in human matrices biomarkers of dioxin exposure in humans [162]. Here, data were

Numerous targeted GC–MS studies were developed for the acquired by untargeted UHPLC–QTOF and then filtered by m/z val-

screening of CAH and other steroidogenesis dysfunctions. For most ues of steroid masses extracted from the lipids class of LIPID

of these applications and for the following cited GC–MS methods (if MAPS. In this study, the use of an extreme phenotype (poisoning

no other protocol is described), the sample preparation is based on of the former President of Ukraine, Victor Yushchenko) highlighted

three steps, namely (1) a conjugates hydrolysis, (2) an extraction a panel of 24 biomarkers associated with steroid structures able

protocol, and (3) a derivatisation procedure (for a complete review to discriminate workers with occupational dioxin exposure from

see [29]). LC–MS was also applied for studying steroid metabolism a control group. Unfortunately, only few biomarkers were unam-

disorders; a recent untargeted UHPLC–QTOF investigation of serum biguously identified.

from women with polycystic ovary syndrome led to a panel of

biomarkers, including dihydrotestosterone sulphate (DHTS), for the 6.1.2. Steroidomics in animals and cell models

diagnosis of this pathology [143]. The latter approach was done As previously indicated, animal models and in vitro cell cul-

with a shorter sample preparation, based only on protein precipi- tures have been developed to aid understanding of the effects

tation. and mechanisms of toxic exposure, which cannot be achieved in

Follow-up of pregnancy and foetus development were studied humans. For example, Kumar et al. analysed rat urine after toxic

in different matrices by explorating the steroidome by GC– and exposures to carbon tetrachloride, acetaminophen, methotrexate,

LC–MS strategies. The steroid metabolome in the blood of pregnant and atorvastatin by combining targeted and untargeted approaches

women and foetuses were obtained by targeted GC–MS for approx- on CE–MS, GC–MS and LC–TOF following a single step of urine

imately 70 steroids: the numerous isomers were resolved by the filtration as sample preparation procedure; steroid, and

high-resolution separation of GC [144,145]. Free steroids and glu- amino acid biomarkers were found to be altered [163,164]. How-

curonide conjugates were analysed in the urine of gravid women ever, the prediction of toxicity or drug treatment success in humans

using both targeted (GC–MS and LC–MS) and untargeted (LC–MS) based on animal models is poor [165]. In addition to the limited

strategies after a SPE procedure for studying the course of preg- prediction, there are other difficulties related to animal testing:

nancy [146]. high costs and ethical problems. Organisations such as the “Euro-

Discovery of steroid as disease biomarkers is one another impor- pean Union Reference Laboratory for alternatives to animal testing”

tant topic and especially for cancer applications. The detection of (EURL ECVAM) or the “National Centre for the Replacement, Refine-

steroid biomarkers for adrenal carcinoma was studied in human ment and Reduction of Animals in Research” (NC3Rs) promote

urine by a GC–MS targeted metabolomic approach (32 quantified the establishment of the 3Rs framework for animal experiments:

steroids), and appropriate data mining was able to achieve dis- replacement, reduction and refinement of animal use. As a replace-

crimination between benign and malign tumours [147]. Another ment tool, the use of in vitro human models is promoted [166].

study of urinary steroid biomarkers of urogenital tract cancer was Steroid disturbances were measured by IA, targeted GC–MS and

performed by a SPE followed by targeted LC–MS determination LC–MS but also recently by a combination of targeted GC–MS and

[148]. An untargeted approach to discriminate cirrhosis from early untargeted UHPLC–QTOF acquisition in H295R cell cultures used for

stage of hepatocellular carcinoma was also done in urine by an screening EDCs [80,81]. In these studies, a SPE step was done prior

untargeted LC–MS/MS approach after SPE and a derivatisation step, LC–MS analysis and then the results were represented by corre-

that enhanced the detection of the urinary steroids [149,150]. The lation analyses and/or heatmap visualisations, highlighting which

steroid metabolome was determined in human breast cancer tis- steroid classes were modified.

sue by LC–MS in SRM mode. Breast cancer risk is hypothesised to be In summary, various steroidomic studies have already been

higher after extended estrogen therapy, and the author developed published, but most of them are targeted approaches. The few

a method for analysing approximately hundred steroids extracted untargeted strategies described many metabolites, but only a small

from breast tissue by a simple tissue homogenisation in a water- number could be identified. Mechanistic interpretations of the

methanol mixture [151]. results therefore remain quite complex. Identification is undoubt-

Biomarkers of neurodegenerative diseases are also studied, such edly the challenging step, as observed in our above cited study

as Griffiths et al. that published several steroidomic studies on the concerning the investigation of human dioxin exposure, and it will

brain. Analysis of (bile acids and steroids) was first per- be discussed hereafter.

formed by targeted GC–MS, then by targeted LC–MS and later with

untargeted LC–MS approaches [152–158]. The sample preparations 6.2. Proposed steroidomic workflow: reduction of data

of the latter GC– and LC–MS cited studies rely mainly on conjugates dimensionality and identification assisted by database filtering

hydrolysis, extraction by SPE or LLE and different derivatisation

procedures. The prior knowledge on sterol fragmentation rules Our vision of steroidomics concerns the biomarker discovery

obtained through GC–MS and LC–MS was very profitable for untar- process of steroidogenesis perturbations and their identification in

3

geted studies where multiple-stage mass spectrometry (MS ) were a generic analytical workflow (Fig. 5). This process should allow

performed to refine the structure and then identify a metabolite analyses of different biological matrices to understand the mech-

of cholesterol (bile acid) with a putative role in the aetiology of anisms of the dysregulation of homeostasis due to the maximum

Alzheimer’s disease. level of steroidome coverage. First, steroidomics should be acquired

Doping screening is another topic investigated by steroidomic by an untargeted MS acquisition and be performed in a generic way,

approaches. Additional steroid biomarkers of exogenous testos- with complementary determinations on the same sample such as

terone delivery as the ones cited by the WADA (see [63]) different ionisation processes, different sample preparation proce-

were highlighted in an untargeted steroidomic approach by dures or different liquid chromatography stationary phases. Steroid

UHPLC–QTOF [64,159–161]. This approach was made by a structures are similar, but differences can be observed due to

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

10 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

Steroidomic or extended steroid profile

Samples

Untargeted or Post-Targeted approach

Combining at ory Combining UHPLC modes, GC and CE strategies for extending the steroidome coverage

t labo r MS

Analyzer QTOF, Orbitrap, FT-ICR we

based on exact masses retrieved

Data processing from open-source databases

compleme nted by experimental at ory comparisons R

y labo r Combining results from

dr Data analysis orthogonal

including

Fig. 5. Proposed steroidomic workflow.

chromatographic behaviour and/or ionisation efficiency with ESI, reduction is mandatory because both statistical and biological

APCI or APPI (in addition to the MS polarity). For example, samples interpretations are easier with a reduced number of compounds.

can be treated by a simple procedure (e.g., dilution) to observe in Empirical data will be obtained by specific studies and authentic

priority highly concentrated metabolites and by different extrac- chemical standards. Each new steroid (identified or commercially

tion procedures (e.g., SPE or LLE) to observe compounds present in available) has to be integrated in a database by also importing the

lower concentration. structure (e.g., .mol or SMILES files). The format of the database

Each one of these experiments will generate large amounts of should also be applicable to metabolomic softwares, such as XLS

data. Each run contains many thousands peaks to be summarised (excel), CSV (comma separated values) or SDF (Simple Data For-

as a list of features defined by an m/z value eluting at a tR with mat) files. In contrast to GC–MS with electron impact ionisation (EI),

an associated intensity: m/z@tR@intensity. The information con- where spectra are similar between different instrument manufac-

tained in the data is contaminated by chemical and random noise, turers, LC–MS spectra can be quite different (for ESI, APPI or APCI

mainly originating from molecules of solvents and from the detec- acquisitions), and the most abundant peak might be not identical

tor, respectively. Furthermore, a single analyte in the raw data between two instruments (e.g., because of the presence of adducts

may correspond to several features, such as isotopic peaks, adducts or variations resulting from different source geometries). Because

or in-source fragments; for example, testosterone can produce relative intensities could be different for the same injected com-

many ions (Fig. 6). The grouping of ions belonging to the same pound either in MS or MS/MS, a database should have for each

molecule is called ion annotation [167]. Various strategies have compound (i) the MS spectrum and (ii) MS/MS spectra with incre-

been developed to filter the raw data into meaningful information, mented collision energies. A consensus spectrum (experimental or

which could be highly important, as a significant data reduction is compiled) can hence be used as a reference in which the precur-

obtained, with approximately 60 to more than 90% of signals being sor ion and all fragments can be observed on the same spectrum.

removed [168]. It must be noted that the inclusion of non-pertinent This MS data acquisition strategy is actually started to be imple-

features dramatically affects the data processing speed and the data mented in open-source databases such as HMDB and Metlin. We

interpretation. Thus, for metabolomics and especially steroidomics, also believe that information regarding retention time, especially

in which many steroids can be converted and observed as other in steroidomics analyses, should be integrated into the identifica-

steroids by chemical group losses in the ion source of the MS (gen- tion process, even if some challenges still need to be overcome.

erally loss of water molecule(s)), the processing steps of denoising First, small variations in retention times could be observed from

and ion annotation are crucial. one instrument to another, including difficulties regarding gradient

Once proper and relevant annotated features are obtained, the transfer or column batch reproducibility. Minor differences could

next step is the signal separation of the steroidome from the also be observed with the same stationary phase with the same

metabolome in the acquired data. Hence our steroidomic approach, gradient due to the instrument configuration, such as dead volume

as an investigation of a sub-data set of an untargeted acquisition, from tubing lengths and valves [169]. Therefore, after system quali-

requires filtering and identification tools to retrieve the informa- fication by determining dead and delay volume, the use of a generic

tion coming from the steroid compartment. The use of a steroid linear gradient (e.g., 5–90%) will contribute to easily transposing

database gathering entries from steroids comprised in HMDB or the method to different systems and columns (e.g., of transfer

LIPID MAPS (as seen in the cited doping and dioxin biomarker tool from HPLC to UHPLC by http://www.unige.ch/sciences/pharm/

studies) combined with empirical data including exact mass, MS, fanal/lcap/telechargement.html). The database should fully inte-

MS/MS spectra, and tR should serve as a basis for (1) filtering fea- grate information from chromatographic separation, which often

tures and reducing the initial data dimensionality (post-targeted allows the separation of positional isomers and, in certain case,

approach) and (2) biomarker identification. The dimensionality even diastereoisomers, prior to MS detection.

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 11

A) Intens ity + 6 [M + H] 1x10 289.218 6 0.8x10 289.218 6 0.6x10 [2M + H]+ 6 290.220 0.4x10 577.427 291.223

6 +

0.2x10 288 290 292 m/z [M + Na] + 311.199 [2M + Na] 599.408 0.0

m/z 100 200 300 400 500 600 700 800

B) Intensity 4 1x10 157.084 4 1.5 x10 [M + H-H O]+ 171.099 2 4 149.023 279.159 1.0 x10 271.190 4 205.086 0.5 255.159 x10 193.084 217.106 237.147 123.080 137.096 181.122 263.162

97.064 113.059

0.0

m/z

100 120 140 160 180 200 220 240 260 280

+ + +

Fig. 6. Example of ion annotation for testosterone. (A) In addition to the proton adduct [M+H] , the sodium adduct [M+Na] , the dimeric form of testosterone [2M+H] and the

+

dimeric form with sodium adduct [2M+Na] are also present. The proton adduct is displayed on a larger scale to show the isotopic pattern. (B) Water loss from testosterone

+

[M+H−H2O] and interfering signals from other co-eluting peaks.

The next step is the matching of annotated features to this only if steroid “candidates” can be bought or synthesised. It is to be

steroid database. Once features are matched to candidates (first noted that only a few MS/MS spectra are available in open-source

criterion is the exact mass, followed by the tR and then the databases and therefore features identified with situation (3) are

MS/MS if available), different levels of identification are obtained. more frequent than with situations (2).

As indicated in Table 2, the ultimate identification is achieved by For steroids and numerous other lipids, authentic standards are

comparison with an authentic chemical standard, which repre- not always available. Therefore, prior time-consuming syntheses

sents the higher level of identification for metabolomic studies or identification strategies such as MS fragmentation studies or

[170]. Results of the steroidomic filtering will be the following: in silico explorations (mentioned later in the text) to identify the

(1) features matching a steroid already identified by an authen- biomarkers, a further reduction of the relevant features has to be

tic chemical standard by comparison of exact mass, MS/MS and performed by MVA analysis. Specific features of a disease or a toxic

tR (level 1); (2) features matching steroids having the same exact exposure are for example obtained by S-plot or correlation analysis

mass and MS/MS spectra (level 2 or 3); and (3) features match- by comparison of sick/exposed patients with controls. If multiple

ing steroids having the same exact mass (level 2 or 3). In the first investigations (e.g., ESI and APCI) were done on samples, a further

situation (1), the identification is achieved, further certainty could challenge is then to gather the data and to analyse them using

be achieved by spiking the certified standard in the biological sam- an appropriate MVA tools, such as multiblock models, in order to

ple and/or the use of orthogonal separation techniques. For the extract the most correct information from the data set that can con-

second and third situations, level 1 identification could be obtained tain redundant information. This dimensionality reduction spares

time by selecting the important features to be further investigated

Table 2 and as already mentioned, biological interpretations are easier with

Classification of different confidence levels for metabolite identification in a reduced number of biomarkers.

metabolomics. Adapted from [170].

If authentic chemical standards are missing, the first strat-

Level Confidence of identity Level of evidence egy is to produce them. As different enzymes are involved in

steroidogenesis, dedicated enzymatic metabolic reactions could

1 Confidently identified Comparison of two or more orthogonal

be used to bio-synthesise steroid standards. For this purpose,

compounds properties with an authentic chemical

standard analysed under identical numerous recombinant enzymes and different liver fractions are

analytical conditions commercially available. In vitro metabolic reactions of hydroxy-

2 Putatively annotated Based upon physicochemical properties

lations, glucuronidations and sulphations are obtained by adding

compounds and/or spectral similarity with 

the appropriate cofactors, such as NADPH for CYPs, uridine 5 -

public/commercial spectral libraries,

without reference to authentic chemical diphospho-glucuronic acid for the uridine glucuronide transferases

 

standards and 3 -phosphoadenosine-5 -phosphosulphate for the sulpho-

3 Putatively annotated Based upon characteristic physicochemical

transferases. Nevertheless, chemical syntheses are also well

compound classes properties of a chemical class of

described for small molecules, such as for glucuronides [171,172]

compounds or upon spectral similarity to

and sulphate conjugates [173,174]. The WADA supports projects

known compounds of a chemical class

4 Unknown compounds Although unidentified and unclassified, regarding the synthesis of phase II metabolites for their inclusion

these metabolites can still be differentiated in routine doping control analysis. These syntheses strategies are

and quantified based upon spectral data

not restricted to doping analysis. Different classes of endogenous

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

12 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

metabolites coming from LC–MS analysis of urine were identified be better described and putatively explained using untargeted

by the help of human liver microsomes (HLM) [175]. Another exam- approaches. Clinical sample classification according to the numer-

ple is the identification of urinary steroid biomarkers of dioxin ous steroid measurements in laboratories is a complex process, but

exposure by applying HLM incubations to produce glucuronide steroidomics can be used to quickly stratify samples and help to

metabolites and cytosol fractions for sulphated metabolites. This establish a more accurate diagnosis. Mathematical statistics can

work currently in progress in our laboratory was shown to be suc- even assist in the follow-up of disease by comparing the steroid

cessful to identify steroid biomarkers. profiles of a patient over a long period of time, similar to the WADA

In the absence of authentic standards or synthesised com- promotion of the use of a biological passport. Endogenous steroid

pounds, the identification process requires supplementary steps to profiles made at different time points in the patient’s life could

study MS/MS spectra and other physicochemical properties of the provide information on the recovery after a treatment or on dis-

biomarkers. Although steroid mass fragmentation has been sub- ease progressions or regressions. This information is particularly

ject of detailed reviews [60,153], the MS/MS spectra were mainly relevant in the context of a general increase in life expectancy in

acquired with low resolution MS instruments. Indications based humans and in light of the current tendency to accumulate large

on fragment ratios to identify different testosterone and hydrox- amounts of personal biomedical data in electronic medical file.

ytestosterone isomers [176] can though be helpful to discriminate In our steroidomic approach, data are acquired in an untargeted

several putative biomarkers. Recent publications investigated high- way and then filtered to reduce the metabolome to the steroidome.

resolution MS fragmentations of specific steroids such as estradiol It must be noted, however, that all the information about other

[177] or sulphated steroids obtained by chemical sulphation [178]; metabolites is preserved. The data can be analysed in a later step for

these studies are still scarce and restricted to a few compounds. other compound classes, given evidence of additional metabolite

The steroidomic experimental spectra have then to be compared perturbations. This strategy is clearly comparable to the approach

to published results or to computational predicted fragmentation proposed in the field of genomics, where the complete genome

spectra based either on fragmentation rules derived from the lit- of a patient can be sequenced in a first single step and relevant

erature or from combinatorial fragmentation [179]. Fragmentation genes examined later “on demand”. Because steroid perturbations

produced independently from prior empirical experiments gener- are linked to many other diseases, assembling data from different

ates rich in silico MS/MS spectra based on molecular decomposition sources such as anamnesis information, blood and urine analysis,

into substructures [180]. Manually interpretation of mass spectra metabolomics, and transcriptomics should be of utmost interest.

remains a time-consuming process and inherently difficult to be Data treatment by advanced chemometric tools and pathway anal-

applied to steroid identification due to the number of fragments ysis are relatively new topics, and their use remain reserved to

generated. Some software packages such as MolFind, an open- research groups; their translation into the clinical domain will need

source java software, allow the complete computational workflow easier interfaces that have not yet been developed, but they rep-

[181] and results are then given by ranking scores based on the resent the future of investigations of steroid perturbations and

number of common peaks and the relative intensities. Other in other diseases. Future steroid analysis will be investigated by

silico approaches based on computational modelling could help to combining targeted and untargeted analytical methods, advanced

further identify biomarkers [182]. Quantitative structure property chemometric analyses and knowledge from chemical analysts,

relationship (QSPR) models are used to predict physicochemical chemometricians, and physicians. For untargeted approaches, the

properties on the basis of molecular descriptors. In the case of main obstacle to direct translation into the clinic is the currently

LC–MS, the main QSPR models are based on retention and MS low rate of identification, which hopefully will soon be improved.

parameters, such as retention index (RI, retention time relative to

a homologous series of compounds), ECOM50 (energy for decom- Acknowledgments

posing 50% of a precursor in the collision cell of the MS [183]) or

fragmentation spectra as already described. F.J., D.T., and S.R. would like to acknowledge the Swiss Centre

In vitro metabolic synthesis is accessible for main laboratories, for Applied Human Toxicology (SCAHT, Switzerland) for supporting

but the main in silico tools are still in development and require this work.

a programming knowledge (e.g., java, R or Matlab). Identification

is still a challenging point for steroidomics, and further progress References

is needed to yield a high rate of identified steroids. Following the

[1] F. Fanelli, I. Belluomo, V.D. Di Lallo, G. Cuomo, R. De Iasio, M. Baccini, E. Casa-

identification, the next challenging points will be the interpreta-

dio, B. Casetta, V. Vicennati, A. Gambineri, G. Grossi, R. Pasquali, U. Pagotto,

tion of the results and the confirmation of fundamental biological

Serum steroid profiling by isotopic dilution–liquid chromatography–mass

mechanisms implicated in the steroid changes. spectrometry: comparison with current immunoassays and reference inter-

vals in healthy adults, Steroids 76 (2011) 244–253.

[2] J.P. Holst, O.P. Soldin, T.D. Guo, S.J. Soldin, Steroid hormones: relevance and

measurement in the clinical laboratory, Clin. Lab. Med. 24 (2004) 105–118.

7. Conclusion [3] C.Z. Minutti, J.M. Lacey, M.J. Magera, S.H. Hahn, M. McCann, A. Schulze, D.

Cheillan, C. Dorche, D.H. Chace, J.F. Lymp, D. Zimmerman, P. Rinaldo, D.

Matern, Steroid profiling by tandem mass spectrometry improves the positive

Steroid perturbation is a fundamental public health problem

predictive value of newborn screening for congenital adrenal hyperplasia, J.

that requires dedicated analytical strategies. Human dysregu-

Clin. Endocr. Metab. 89 (2004) 3687–3693.

lations due to known diseases can be diagnosed by targeted [4] C.L. Acerini, I.A. Hughes, Endocrine disrupting chemicals: a new and emerging

public health problem? Arch. Dis. Child. 91 (2006) 633–638.

approaches. For a restricted number of situations, these methods

[5] S. De Coster, N. van Larebeke, Endocrine-disrupting chemicals: associated

fulfil expectations, but unfortunately they determine only pre-

disorders and mechanisms of action, J. Environ. Public Health (2012) 1–52,

defined steroids and check solely for anticipated modifications id713696.

[6] T.T. Schug, A. Janesick, B. Blumberg, J.J. Heindel, Endocrine disrupting chem-

that could be observed. Therefore, both multi-analyte targeted and

icals and disease susceptibility, J. Steroid Biochem. Mol. Biol. 127 (2011)

untargeted methods should be used to achieve different purposes. 204–215.

Steroidomics, as an untargeted acquisition-based strategy, will cer- [7] K.S. Leung, B.M. Fong, LC–MS/MS in the routine clinical laboratory: has its

time come? Anal. Bioanal. Chem. 406 (2014) 2289–2301.

tainly have a place in the future of steroid analysis for many disease

[8] A.H.B. Wu, D. French, Implementation of liquid chromatography/mass spec-

and toxicology diagnoses. Steroidomics can also provide, in addi-

trometry into the clinical laboratory, Clin. Chim. Acta 420 (2013) 4–10.

tion to diagnoses, indications regarding disease evolution, aetiology [9] M. Rauh, Steroid measurement with LC–MS/MS. Application examples in

pediatrics, J. Steroid Biochem. 121 (2010) 520–527.

and unexplained cases. Furthermore, mechanistic information can

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 13

[10] S.J. Soldin, O.P. Soldin, analysis by tandem mass spectrom- [40] A.K. Harding, G.P. Daston, G.R. Boyd, G.W. Lucier, S.H. Safe, J. Stewart, D.E.

etry, Clin. Chem. 55 (2009) 1061–1066. Tillitt, G. Van der Kraak, Endocrine disrupting chemicals research program of

[11] M. Vogeser, F. Kirchhoff, Progress in automation of LC–MS in laboratory the US Environmental Protection Agency: summary of a peer-review report,

medicine, Clin. Biochem. 44 (2011) 4–13. Environ. Health Persp. 114 (2006) 1276–1282.

[12] A.H. Wu, R. Gerona, P. Armenian, D. French, M. Petrie, K.L. Lynch, Role of liquid [41] M.D. Anway, A.S. Cupp, M. Uzumcu, M.K. Skinner, Epigenetic transgenera-

chromatography–high-resolution mass spectrometry (LC–HR/MS) in clinical tional actions of endocrine disruptors and male fertility, Science 308 (2005)

toxicology, Clin. Toxicol. 50 (2012) 733–742. 1466–1469.

[13] R. Ramanathan, M. Jemal, S. Ramagiri, Y.Q. Xia, W.G. Humpreys, T. Olah, W.A. [42] M.D. Anway, M.K. Skinner, Epigenetic transgenerational actions of endocrine

Korfmacher, It is time for a paradigm shift in drug discovery bioanalysis: from disruptors, Endocrinology 147 (2006) S43–S49.

SRM to HRMS, J. Mass Spectrom. 46 (2011) 595–601. [43] G.M.B. Louis, M.A. Cooney, C.M. Peterson, The ovarian dysgenesis syndrome,

[14] H. Henry, H.R. Sobhi, O. Scheibner, M. Bromirski, S.B. Nimkar, B. Rochat, J. Dev. Orig. Health Dis. 2 (2011) 25–35.

Comparison between a high-resolution single-stage Orbitrap and a triple [44] P.A. Fowler, M. Bellingham, K.D. Sinclair, N.P. Evans, P. Pocar, B. Fischer,

quadrupole mass spectrometer for quantitative analyses of drugs, Rapid Com- K. Schaedlich, J.S. Schmidt, M.R. Amezaga, S. Bhattacharya, S.M. Rhind, P.J.

mun. Mass Spectrom. 26 (2012) 499–509. O’Shaughnessy, Impact of endocrine-disrupting compounds (EDCs) on female

[15] R. Plumb, J. Castro-Perez, J. Granger, I. Beattie, K. Joncour, A. Wright, Ultra- reproductive health, Mol. Cell. Endocrinol. 355 (2012) 231–239.

performance liquid chromatography coupled to quadrupole-orthogonal [45] M.A. Elobeid, D.B. Allison, Putative environmental-endocrine disruptors and

time-of-flight mass spectrometry, Rapid Commun. Mass Spectrom. 18 (2004) obesity: a review, Curr. Opin. Endocrinol. Diabetes Obes. 15 (2008) 403–408.

2331–2337. [46] R. Hampl, J. Kubatova, L. Starka, Steroids and endocrine disruptors – history,

[16] R. Hayes, A. Ahmed, T. Edge, H. Zhang, Core–shell particles: preparation, fun- recent state of art and open questions, J. Steroid Biochem. Mol. Biol. (2014),

damentals and applications in high performance liquid chromatography, J. http://dx.doi.org/10.1016/j.jsbmb.2014.1004.1013

Chromatogr. A 1357 (2014) 36–52. [47] A. Kortenkamp, M. Scholze, S. Ermler, Mind the gap: can we explain declin-

[17] J. Boccard, D.N. Rutledge, A consensus orthogonal partial least squares dis- ing male reproductive health with known antiandrogens? Reproduction 147

criminant analysis (OPLS-DA) strategy for multiblock Omics data fusion, Anal. (2014) 515–527.

Chim. Acta 769 (2013) 30–39. [48] O.V. Martin, T. Shialis, J.N. Lester, M.D. Scrimshaw, A.R. Boobis, N. Voulvoulis,

[18] J. Boccard, S. Rudaz, Harnessing the complexity of metabolomic data with Testicular dysgenesis syndrome and the estrogen hypothesis: a quantitative

chemometrics, J. Chemometrics 28 (2014) 1–9. meta-analysis, Environ. Health Persp. 116 (2008) 149–157.

[19] J. Eichner, L. Rosenbaum, C. Wrzodek, H.U. Haring, A. Zell, R. Lehmann, [49] G.J. Nohynek, C.J. Borgert, D. Dietrich, K.K. Rozman, Endocrine disruption: fact

Integrated enrichment analysis and pathway-centered visualization of or urban legend? Toxicol. Lett. 223 (2013) 295–305.

metabolomics, proteomics, transcriptomics, and genomics data by using the [50] E. Diamanti-Kandarakis, J.P. Bourguignon, L.C. Giudice, R. Hauser, G.S.

InCroMAP software, J. Chromatogr. B 966 (2014) 77–82. Prins, A.M. Soto, R.T. Zoeller, A.C. Gore, Endocrine-disrupting chemicals: an

[20] T.C. Kuo, T.F. Tian, Y.J. Tseng, 3Omics: a web-based systems biology tool for endocrine society scientific statement, Endocr. Rev. 30 (2009) 293–342.

analysis, integration and visualization of human transcriptomic, proteomic [51] S. Kwon, K.L. Hermayer, Glucocorticoid-induced hyperglycemia, Am. J. Med.

and metabolomic data, BMC Syst. Biol. 7 (2013) 64. Sci. 345 (2013) 274–277.

[21] J. Sjovall, Fifty years with bile acids and steroids in health and disease, Lipids [52] N.T. Shahidi, A review of the chemistry, biological action, and clinical appli-

39 (2004) 703–722. cations of anabolic-androgenic steroids, Clin. Ther. 23 (2001) 1355–1390.

[22] D.S. Wishart, T. Jewison, A.C. Guo, M. Wilson, C. Knox, Y. Liu, Y. Djoumbou, [53] S. Basaria, J.T. Wahlstrom, A.S. Dobs, Clinical review 138: anabolic-androgenic

R. Mandal, F. Aziat, E. Dong, S. Bouatra, I. Sinelnikov, D. Arndt, J. Xia, P. Liu, F. steroid therapy in the treatment of chronic diseases, J. Clin. Endocrinol. Metab.

Yallou, T. Bjorndahl, R. Perez-Pineiro, R. Eisner, F. Allen, V. Neveu, R. Greiner, 86 (2001) 5108–5117.

A. Scalbert, HMDB 3.0 – the human metabolome database in 2013, Nucleic [54] A.T. Kicman, Pharmacology of anabolic steroids, Brit. J. Pharmacol. 154 (2008)

Acids Res. 41 (2013) D801–D807. 502–521.

[23] J.T. Sanderson, The steroid hormone biosynthesis pathway as a target for [55] J. Brynhildsen, Combined hormonal contraceptives: prescribing patterns,

endocrine-disrupting chemicals, Toxicol. Sci. 94 (2006) 3–21. compliance, and benefits versus risks, Ther. Adv. Drug Saf. 5 (2014) 201–213.

[24] A. Arukwe, Steroidogenic acute regulatory (StAR) protein and cholesterol [56] G.K. Reeves, V. Beral, J. Green, T. Gathani, D. Bull, Hormonal therapy for

side-chain cleavage (P450scc)-regulated steroidogenesis as an organ-specific menopause and breast-cancer risk by histological type: a cohort study and

molecular and cellular target for endocrine disrupting chemicals in fish, Cell meta-analysis, Lancet Oncol. 7 (2006) 910–918.

Biol. Toxicol. 24 (2008) 527–540. [57] A. Fournier, S. Mesrine, M.C. Boutron-Ruault, F. Clavel-Chapelon,

[25] W.L. Miller, R.J. Auchus, The molecular biology, biochemistry, and physiology Estrogen–progestagen menopausal hormone therapy and breast can-

of human steroidogenesis and its disorders, Endocr. Rev. 32 (2011) 81–151. cer: does delay from menopause onset to treatment initiation influence

[26] A.H. Payne, D.B. Hales, Overview of steroidogenic enzymes in the pathway risks? J. Clin. Oncol. 27 (2009) 5138–5143.

from cholesterol to active steroid hormones, Endocr. Rev. 25 (2004) 947–970. [58] U. Ceglarek, M. Werner, L. Kortz, A. Korner, W. Kiess, J. Thiery, J. Kratzsch, Pre-

[27] C.A. Strott, Sulfonation and molecular action, Endocr. Rev. 23 (2002) 703–732. clinical challenges in steroid analysis of human samples, J. Steroid Biochem.

[28] Y.W. Ha, J.Y. Moon, H.J. Jung, B.C. Chung, M.H. Choi, Evaluation of plasma Mol. Biol. 121 (2010) 505–512.

enzyme activities using gas chromatography–mass spectrometry based [59] M.I. New, Inborn errors of adrenal steroidogenesis, Mol. Cell. Endocrinol. 211

steroid signatures, J. Chromatogr. B 877 (2009) 4125–4132. (2003) 75–83.

[29] N. Krone, B.A. Hughes, G.G. Lavery, P.M. Stewart, W. Arlt, C.H.L. Shackleton, [60] H.L.J. Makin, D.B. Gower, Steroid Analysis, 2nd ed., Springer, Netherlands,

Gas chromatography/mass spectrometry (GC/MS) remains a pre-eminent dis- Dordrecht, 2010.

covery tool in clinical steroid investigations even in the era of fast liquid [61] W.K. Jerjes, A.J. Cleare, T.J. Peters, N.F. Taylor, Circadian rhythm of urinary

chromatography tandem mass spectrometry (LC/MS/MS), J. Steroid Biochem. steroid metabolites, Ann. Clin. Biochem. 43 (2006) 287–294.

Mol. Biol. 121 (2010) 496–504. [62] F. Badoud, D. Guillarme, J. Boccard, E. Grata, M. Saugy, S. Rudaz, J.L. Veuthey,

[30] E. Fahy, S. Subramaniam, R.C. Murphy, M. Nishijima, C.R.H. Raetz, T. Shimizu, Analytical aspects in doping control: challenges and perspectives, Forensic

F. Spener, G. van Meer, M.J.O. Wakelam, E.A. Dennis, Update of the LIPID MAPS Sci. Int. 213 (2011) 49–61.

comprehensive classification system for lipids, J. Lipid Res. 50 (2009) S9–S14. [63] W.A.-D.A. (W.A.D.A.), Endogenous Anabolic Androgenic Steroids, Measure-

[31] M. Kanehisa, S. Goto, Y. Sato, M. Kawashima, M. Furumichi, M. Tanabe, Data, ment and Reporting, TD2014EAAS, 2014.

information, knowledge and principle: back to metabolism in KEGG, Nucleic [64] F. Badoud, E. Grata, J. Boccard, D. Guillarme, J.L. Veuthey, S. Rudaz, M. Saugy,

Acids Res. 42 (2014) D199–D205. Quantification of glucuronidated and sulfated steroids in human urine by

[32] C.H.L. Shackleton, Role of a disordered steroid metabolome in the elucidation ultra-high pressure liquid chromatography quadrupole time-of-flight mass

of sterol and steroid biosynthesis, Lipids 47 (2012) 1–12. spectrometry, Anal. Bioanal. Chem. 400 (2011) 503–516.

[33] R. Ferraldeschi, N. Sharifi, R.J. Auchus, G. Attard, Molecular pathways: inhib- [65] M.M. Kushnir, A.L. Rockwood, W.L. Roberts, B.F. Yue, J. Bergquist, A.W. Meikle,

iting steroid biosynthesis in prostate cancer, Clin. Cancer Res. 19 (2013) Liquid chromatography tandem mass spectrometry for analysis of steroids in

3353–3359. clinical laboratories, Clin. Biochem. 44 (2011) 77–88.

[34] G.R. MacVicar, M.H. Hussain, Emerging therapies in metastatic castration- [66] M. Wagner, D. Tonoli, E. Varesio, G. Hopfgartner, The use of mass spectrometry

sensitive and castration-resistant prostate cancer, Curr. Opin. Oncol. 25 to analyze dried blood spots, Mass Spectrom. Rev. (2014), http://dx.doi.org/

(2013) 252–260. 10.1002/mas.21441

[35] I.E. Obiorah, P. Fan, S. Sengupta, V.C. Jordan, Selective estrogen-induced apo- [67] M. Groschl, H. Kohler, H.G. Topf, T. Rupprecht, M. Rauh, Evaluation of saliva

ptosis in breast cancer, Steroids 90 (2014) 60–70. collection devices for the analysis of steroids, peptides and therapeutic drugs,

[36] V.C. Jordan, A century of deciphering the control mechanisms of sex steroid J. Pharm. Biomed. Anal. 47 (2008) 478–486.

action in breast and prostate cancer: the origins of targeted therapy and [68] L. de Lacerda, A. Kowarski, C.J. Migeon, Integrated concentration of plasma

chemoprevention, Cancer Res. 69 (2009) 1243–1254. cortisol in normal subjects, J. Clin. Endocrinol. Metab. 36 (1973) 227–238.

[37] E. Carlsen, A. Giwercman, N. Keiding, N.E. Skakkebaek, Evidence for decreasing [69] L. Manetti, G. Rossi, L. Grasso, V. Raffaelli, I. Scattina, S. Del Sarto, M. Cosottini,

quality of semen during past 50 years, Brit. Med. J. 305 (1992) 609–613. A. Iannelli, M. Gasperi, F. Bogazzi, E. Martino, Usefulness of salivary cortisol

[38] N.E. Skakkebaek, E. Rajpert-De Meyts, K.M. Main, Testicular dysgenesis syn- in the diagnosis of hypercortisolism: comparison with serum and urinary

drome: an increasingly common developmental disorder with environmental cortisol, Eur. J. Endocrinol. 168 (2013) 315–321.

aspects, Hum. Reprod. 16 (2001) 972–978. [70] B.G. Keevil, P. MacDonald, W. Macdowall, D.M. Lee, F.C. Wu, N. Team, Sali-

[39] C.P. Wild, Complementing the genome with an “exposome”: the outstanding vary testosterone measurement by liquid chromatography tandem mass

challenge of environmental exposure measurement in molecular epidemiol- spectrometry in adult males and females, Ann. Clin. Biochem. 51 (2014)

ogy, Cancer Epidemiol. Biomarkers Prev. 14 (2005) 1847–1850. 368–378.

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

14 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

[71] E. Aardal, A.C. Holm, Cortisol in saliva – reference ranges and relation to [96] A. Slominski, B. Zbytek, G. Nikolakis, P.R. Manna, C. Skobowiat, M. Zmijewski,

cortisol in serum, Eur. J. Clin. Chem. Clin. 33 (1995) 927–932. W. Li, Z. Janjetovic, A. Postlethwaite, C.C. Zouboulis, R.C. Tuckey, Steroidogen-

[72] K.R. Loughlin, Changes in male fertility in the last two decades, Urol. Clin. esis in the skin: implications for local immune functions, J. Steroid Biochem.

North Am. 39 (2012) 33–36. Mol. Biol. 137 (2013) 107–123.

[73] R. Hampl, M. Pohanka, M. Hill, L. Starka, The content of four immunomodula- [97] A. Tiganescu, M. Hupe, Y. Uchida, T. Mauro, P.M. Elias, W.M. Holleran,

tory steroids and major androgens in human semen, J. Steroid. Biochem. Mol. Increased glucocorticoid activation during mouse skin wound healing, J.

Biol. 84 (2003) 307–316. Endocrinol. 221 (2014) 51–61.

[74] M. Pohanka, R. Hampl, I. Sterzl, L. Starka, Steroid hormones in human semen [98] R.F. Hannen, A.E. Michael, A. Jaulim, R. Bhogal, J.M. Burrin, M.P. Philpott,

with particular respect to dehydroepiandrosterone and its immunomodula- Steroid synthesis by primary human keratinocytes; implications for skin dis-

tory metabolites, Endocr. Regul. 36 (2002) 79–86. ease, Biochem. Bioph. Res. Commun. 404 (2011) 62–67.

[75] T. Wang, W.E. Rainey, Human adrenocortical carcinoma cell lines, Mol. Cell. [99] A. Petrova, A. Celli, L. Jacquet, D. Dafou, D. Crumrine, M. Hupe, M. Arno, C.

Endocrinol. 351 (2012) 58–65. Hobbs, A. Cvoro, P. Karagiannis, L. Devito, R. Sun, L.C. Adame, R. Vaughan, J.A.

[76] M. Hecker, H. Hollert, R. Cooper, A.M. Vinggaard, Y. Akahori, M. Murphy, McGrath, T.M. Mauro, D. Ilic, 3D in vitro model of a functional epidermal per-

C. Nellemann, E. Higley, J. Newsted, R. Wu, P. Lam, J. Laskey, A. Buckalew, meability barrier from human embryonic stem cells and induced pluripotent

S. Grund, M. Nakai, G. Timm, J. Giesy, The OECD validation program of the stem cells, Stem Cell Rep. 2 (2014) 675–689.

H295R steroidogenesis assay for the identification of in vitro inhibitors and [100] C.C. Sweeley, E.C. Horning, Microanalytical separation of steroids by gas chro-

inducers of testosterone and estradiol production. Phase 2: Inter-laboratory matography, Nature 187 (1960) 144–145.

pre-validation studies, Environ. Sci. Pollut. Res. 14 (2007) 23–30. [101] D.J. Haisenleder, A.H. Schoenfelder, E.S. Marcinko, L.M. Geddis, J.C. Marshall,

[77] M. Hecker, H. Hollert, R. Cooper, A.M. Vinggaard, Y. Akahori, M. Murphy, C. Estimation of estradiol in mouse serum samples: evaluation of commercial

Nellemann, E. Higley, J. Newsted, J. Laskey, A. Buckalew, S. Grund, S. Maletz, J. estradiol immunoassays, Endocrinology 152 (2011) 4443–4447.

Giesy, G. Timm, The OECD validation program of the H295R steroidogenesis [102] R.M. Buttler, A. Kruit, M.A. Blankenstein, A.C. Heijboer, Measurement of dehy-

assay: Phase 3. Final inter-laboratory validation study, Environ. Sci. Pollut. droepiandrosterone sulphate (DHEAS): a comparison of Isotope-Dilution

Res. 18 (2011) 503–515. Liquid Chromatography Tandem Mass Spectrometry (ID-LC–MS/MS) and

[78] F.K. Nielsen, C.H. Hansen, J.A. Fey, M. Hansen, N.W. Jacobsen, B. Halling- seven currently available immunoassays, Clin. Chim. Acta 424 (2013)

Sorensen, E. Bjorklund, B. Styrishave, H295R cells as a model for steroidogenic 22–26.

disruption: a broader perspective using simultaneous chemical analysis of 7 [103] W. Rosner, R.J. Auchus, R. Azziz, P.M. Sluss, H. Raff, Position statement: util-

key steroid hormones, Toxicol. In Vitro 26 (2012) 343–350. ity, limitations, and pitfalls in measuring testosterone: an endocrine society

[79] S. Kang, S. Park, M.J. Kim, S.M. Oh, K.H. Chung, S. Lee, A sensitive and selective position statement, J. Clin. Endocr. Metab. 92 (2007) 405–413.

LC–MS/MS analysis coupled with an online sample enrichment technique [104] D.J. Handelsman, L. Wartofsky, Requirement for mass spectrometry sex

for H295R steroidogenesis assay and its application in the investigation of steroid assays in the journal of clinical endocrinology and metabolism, J. Clin.

the effect of sildenafil on steroidogenesis, Anal. Bioanal. Chem. 405 (2013) Endocr. Metab. 98 (2013) 3971–3973.

9489–9496. [105] L. Wartofsky, D.J. Handelsman, Standardization of hormonal assays for the

[80] J.C.W. Rijk, A.A.C.M. Peijnenburg, M.H. Blokland, A. Lommen, R.L.A.P. Hoogen- 21st century, J. Clin. Endocr. Metab. 95 (2010) 5141–5143.

boom, T.F.H. Bovee, Screening for modulatory effects on steroidogenesis using [106] P. Eneroth, K. Hellstroem, R. Ryhage, Identification and quantification of neu-

the human H295R adrenocortical cell line: a metabolomics approach, Chem. tral fecal steroids by gas–liquid chromatography and mass spectrometry:

Res. Toxicol. 25 (2012) 1720–1731. studies of human excretion during two dietary regimens, J. Lipid Res. 5 (1964)

[81] D. Tonoli, C. Furstenberger, J. Boccard, D. Hochstrasser, F. Jeanneret, 245–262.

A. Odermatt, S. Rudaz, Steroidomic footprinting based on ultra-high [107] J.Y. Moon, H.J. Jung, M.H. Moon, B.C. Chung, M.H. Choi, Heat-map visualization

performance liquid chromatography coupled with qualitative and quanti- of gas chromatography–mass spectrometry based quantitative signatures on

tative high-resolution mass spectrometry for the evaluation of endocrine steroid metabolism, J. Am. Soc. Mass Spectr. 20 (2009) 1626–1637.

disrupting chemicals in H295R cells, Chem. Res. Toxicol. 28 (2015) [108] C. Shackleton, Clinical steroid mass spectrometry: a 45-year history culmi-

955–966. nating in HPLC–MS/MS becoming an essential tool for patient diagnosis, J.

[82] V. Roulet, H. Denis, C. Staub, A. Le Tortorec, B. Delaleu, A.P. Satie, J.J. Steroid Biochem. Mol. Biol. 121 (2010) 481–490.

Patard, B. Jegou, N. Dejucq-Rainsford, Human testis in organotypic cul- [109] D.J. Liberato, A.L. Yergey, N. Esteban, C.E. Gomezsanchez, C.H.L. Shackleton,

ture: application for basic or clinical research, Hum. Reprod. 21 (2006) Thermospray Hplc/Ms – a new mass-spectrometric technique for the profiling

1564–1575. of steroids, J. Steroid Biochem. Mol. Biol. 27 (1987) 61–70.

[83] A.L. Forgacs, Q. Ding, R.G. Jaremba, I.T. Huhtaniemi, N.A. Rahman, T.R. [110] J.B. Fenn, M. Mann, C.K. Meng, S.F. Wong, C.M. Whitehouse, Electrospray

Zacharewski, BLTK1 murine leydig cells: a novel steroidogenic model for eval- ionization for mass-spectrometry of large biomolecules, Science 246 (1989)

uating the effects of reproductive and developmental toxicants, Toxicol. Sci. 64–71.

127 (2012) 391–402. [111] D. Tamae, M. Byrns, B. Marck, E.A. Mostaghel, P.S. Nelson, P. Lange, D. Lin,

[84] C. Desdoits-Lethimonier, O. Albert, B. Le Bizec, E. Perdu, D. Zalko, F. Courant, L. M.E. Taplin, S. Balk, W. Ellis, L. True, R. Vessella, B. Montgomery, I.A. Blair, T.M.

Lesne, F. Guille, N. Dejucq-Rainsford, B. Jegou, Human testis steroidogenesis Penning, Development, validation and application of a stable isotope dilution

is inhibited by phthalates, Hum. Reprod. 27 (2012) 1451–1459. liquid chromatography electrospray ionization/selected reaction monitor-

[85] O. Albert, C. Desdoits-Lethimonier, L. Lesne, A. Legrand, F. Guille, K. Bensalah, ing/mass spectrometry (SID-LC/ESI/SRM/MS) method for quantification of

N. Dejucq-Rainsford, B. Jegou, Paracetamol, aspirin and indomethacin dis- keto-androgens in human serum, J. Steroid Biochem. Mol. Biol. 138 (2013)

play endocrine disrupting properties in the adult human testis in vitro, Hum. 281–289.

Reprod. 28 (2013) 1890–1898. [112] K. Yamashita, M. Okuyama, R. Nakagawa, S. Honma, F. Satoh, R. Morimoto,

[86] J.C. Havelock, W.E. Rainey, B.R. Carr, Ovarian granulosa cell lines, Mol. Cell. S. Ito, M. Takahashi, M. Numazawa, Development of sensitive derivatization

Endocrinol. 228 (2004) 67–78. method for aldosterone in liquid chromatography–electrospray ionization

[87] L. Ophir, Y. Yung, E. Maman, N. Rubinstein, G.M. Yerushalmi, J. Haas, E. tandem mass spectrometry of corticosteroids, J. Chromatogr. A 1200 (2008)

Barzilay, A. Hourvitz, Establishment and validation of a model for non- 114–121.

luteinized human mural granulosa cell culture, Mol. Cell. Endocrinol. 384 [113] K. Rigdova, Y. Wang, M. Ward, W.J. Griffiths, A new derivative for oxosteroid

(2014) 165–174. analysis by mass spectrometry, Biochem. Biophys. Res. Commun. 446 (2014)

[88] D.H. Choi, W.S. Lee, M. Won, M. Park, H.O. Park, E. Kim, K.A. Lee, J. Bae, The 762–767.

apolipoprotein A-I level is downregulated in the granulosa cells of patients [114] T. Guo, R.L. Taylor, R.J. Singh, S.J. Soldin, Simultaneous determination of 12

with polycystic ovary syndrome and affects steroidogenesis, J. Proteome Res. steroids by isotope dilution liquid chromatography–photospray ionization

9 (2010) 4329–4336. tandem mass spectrometry, Clin. Chim. Acta 372 (2006) 76–82.

[89] R. Chatterjee, M. Helal, M. Mobberley, T. Ryder, R. Bajoria, Impaired steroido- [115] T.D. Guo, M. Chan, S.J. Soldin, Steroid profiles using liquid

genesis and apoptosis of granulosa-luteal cells in primary culture induced by chromatography–tandem mass spectrometry with atmospheric pressure

cis-platinum, Am. J. Obstet. Gynecol. 210 (252) (2014) e1–e7. photoionization source, Arch. Pathol. Lab. Med. 128 (2004) 469–475.

[90] S.H. Mellon, L.D. Griffin, Neurosteroids: biochemistry and clinical significance, [116] T.D. Guo, J.H. Gu, O.P. Soldin, R.J. Singh, S.J. Soldin, Rapid measure-

Trends Endocrin. Met. 13 (2002) 35–43. ment of estrogens and their metabolites in human serum by liquid

[91] G. Pelletier, Steroidogenic enzymes in the brain: morphological aspects, Prog. chromatography–tandem mass spectrometry without derivatization, Clin.

Brain Res. 181 (2010) 193–207. Biochem. 41 (2008) 736–741.

[92] N. Daviaud, E. Garbayo, P.C. Schiller, M. Perez-Pinzon, C.N. Montero-Menei, [117] P.C. Kao, D.A. Machacek, M.J. Magera, J.M. Lacey, P. Rinaldo, Diagnosis of

Organotypic cultures as tools for optimizing central nervous system cell ther- adrenal cortical dysfunction by liquid chromatography–tandem mass spec-

apies, Exp. Neurol. 248 (2013) 429–440. trometry, Ann. Clin. Lab. Sci. 31 (2001) 199–204.

[93] R.C. Melcangi, G. Panzica, L.M. Garcia-Segura, Neuroactive steroids: focus on [118] A. Taylor, C. Shackleton, S. Taylor, U.G. Jensen, M. Ojala, W. Arlt, Concurrent

human brain, Neuroscience 191 (2011) 1–5. analysis of 10 serum steroids by mass spectrometry: investigation of the via-

TM

[94] H.T. Hogberg, J. Bressler, K.M. Christian, G. Harris, G. Makri, C. O’Driscoll, bility of the Perkin-Elmer CHS MSMS steroids kit on a waters xevo mass

D. Pamies, L. Smirnova, Z. Wen, T. Hartung, Toward a 3D model of human spectrometer with acquity UPLC system, Endocr. Abstr. 25 (2011) P300.

brain development for studying gene/environment interactions, Stem Cell [119] T. Koal, D. Schmiederer, H. Pham-Tuan, C. Rohring, M. Rauh, Standardized

Res. Ther. 4 (2013), S4:1–7. LC–MS/MS based steroid hormone profile-analysis, J. Steroid Biochem. Mol.

[95] M.A. Lancaster, M. Renner, C.A. Martin, D. Wenzel, L.S. Bicknell, M.E. Hurles, T. Biol. 129 (2012) 129–138.

Homfray, J.M. Penninger, A.P. Jackson, J.A. Knoblich, Cerebral organoids model [120] C. Rossi, L. Calton, G. Hammond, H.A. Brown, A.M. Wallace, P. Sacchetta,

human brain development and microcephaly, Nature 501 (2013) 373–379. M. Morris, Serum steroid profiling for congenital adrenal hyperplasia using

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx 15

liquid chromatography–tandem mass spectrometry, Clin. Chim. Acta 411 determined by liquid chromatography–electrospray ionization-tandem mass

(2010) 222–228. spectrometry, Anal. Chim. Acta 802 (2013) 56–66.

[121] F.B. Lih, M.A. Titus, J.L. Mohler, K.B. Tomer, Atmospheric pressure photoion- [147] W. Arlt, M. Biehl, A.E. Taylor, S. Hahner, R. Libe, B.A. Hughes, P. Schneider,

ization tandem mass spectrometry of androgens in prostate cancer, Anal. D.J. Smith, H. Stiekema, N. Krone, E. Porfiri, G. Opocher, J. Bertherat, F. Man-

Chem. 82 (2010) 6000–6007. tero, B. Allolio, M. Terzolo, P. Nightingale, C.H.L. Shackleton, X. Bertagna, M.

[122] U. Ceglarek, L. Kortz, A. Leichtle, G.M. Fiedler, J. Kratzsch, J. Thiery, Rapid Fassnacht, P.M. Stewart, Urine steroid metabolomics as a biomarker tool for

quantification of steroid patterns in human serum by on-line solid phase detecting malignancy in adrenal tumors, J. Clin. Endocr. Metab. 96 (2011)

extraction combined with liquid chromatography–triple quadrupole linear 3775–3784.

ion trap mass spectrometry, Clin. Chim. Acta 401 (2009) 114–118. [148] L. Konieczna, T. Baczek, M. Belka, A. Fel, M. Markuszewski, W. Struck, M.

[123] K. Dhillon, T. Ho, P. Rich, D.D. Xu, F. Lorey, J.W. She, A. Bhandal, An auto- Markuszewski, R. Kaliszan, Steroid profiles as potential biomarkers in patients

mated method on analysis of blood steroids using liquid chromatography with urogenital tract cancer for diagnostic investigations analyzed by liquid

tandem mass spectrometry: application to population screening for congeni- chromatography coupled to mass spectrometry, J. Pharm. Biomed. Anal. 73

tal adrenal hyperplasia in newborns, Clin. Chim. Acta 412 (2011) 2076–2084. (2013) 108–115.

[124] C.E. Galuska, M.F. Hartmann, A. Sanchez-Guijo, K. Bakhaus, J. Geyer, G. Schuler, [149] W.D. Dai, Q. Huang, P.Y. Yin, J. Li, J. Zhou, H.W. Kong, C.X. Zhao, X. Lu, G.W.

K.P. Zimmer, S.A. Wudy, Profiling intact steroid sulfates and unconjugated Xu, Comprehensive and highly sensitive urinary steroid hormone profiling

steroids in biological fluids by liquid chromatography–tandem mass spec- method based on stable isotope-labeling liquid chromatography mass spec-

trometry (LC–MS-MS), Analyst 138 (2013) 3792–3801. trometry, Anal. Chem. 84 (2012) 10245–10251.

[125] C. Lapthorn, F. Pullen, B.Z. Chowdhry, Ion mobility spectrometry–mass spec- [150] W.D. Dai, P.Y. Yin, P. Chen, H.W. Kong, P. Luo, Z.L. Xu, X. Lu, G.W. Xu,

trometry (IMS–MS) of small molecules: separating and assigning structures Study of urinary steroid hormone disorders: difference between hepatocel-

to ions, Mass Spectrom. Rev. 32 (2013) 43–71. lular carcinoma in early stage and cirrhosis, Anal. Bioanal. Chem. 406 (2014)

[126] J.A. Ray, M.M. Kushnir, R.A. Yost, A.L. Rockwood, A. Wayne Meikle, Per- 4325–4335.

formance enhancement in the measurement of 5 endogenous steroids by [151] N.W. Gaikwad, Ultra performance liquid chromatography–tandem mass

LC–MS/MS combined with differential ion mobility spectrometry, Clin. Chim. spectrometry method for profiling of steroid metabolome in human tissue,

Acta 438 (2015) 330–336. Anal. Chem. 85 (2013) 4951–4960.

[127] G. Kaur-Atwal, J.C. Reynolds, C. Mussell, E. Champarnaud, T.W. Knapman, [152] W. Griffiths, Why steroidomics in brain? Eur. J. Lipid Sci. Technol. 108 (2006)

A.E. Ashcroft, G. O’Connor, S.D.R. Christie, C.S. Creaser, Determination of 707–708.

testosterone and epitestosterone glucuronides in urine by ultra performance [153] W.J. Griffiths, Tandem mass spectrometry in the study of fatty acids, bile acids,

liquid chromatography–ion mobility-mass spectrometry, Analyst 136 (2011) and steroids, Mass Spectrom. Rev. 22 (2003) 81–152.

3911–3916. [154] W.J. Griffiths, K. Karua, M. Hornshaw, G. Woffendin, Y. Wanga, Metabolomics

[128] L. Ahonen, M. Fasciotti, G.B. af Gennas, T. Kotiaho, R.J. Daroda, M. Eberlin, R. and metabolite profiling: past heroes and future developments, Eur. J. Mass

Kostiainen, Separation of steroid isomers by ion mobility mass spectrometry, Spectrom. 13 (2007) 45–50.

J. Chromatogr. A 1310 (2013) 133–137. [155] W.J. Griffiths, J. Sjovall, Bile acids: analysis in biological fluids and tissues, J.

[129] W. Jin, M. Jarvis, M. Star-Weinstock, M. Altemus, A sensitive and selective LC- Lipid Res. 51 (2010) 23–41.

differential mobility-mass spectrometric analysis of allopregnanolone and [156] W.J. Griffiths, Y.Q. Wang, Mass spectrometry: from proteomics to

pregnanolone in human plasma, Anal. Bioanal. Chem. 405 (2013) 9497–9508. metabolomics and lipidomics, Chem. Soc. Rev. 38 (2009) 1882–1896.

[130] F.Z. Stanczyk, J.S. Lee, R.J. Santen, Standardization of steroid hormone assays: [157] W.J. Griffiths, Y.Q. Wang, The importance of steroidomics in the study of neu-

why, how, and when? Cancer Epidem. Biomarkers Prev. 16 (2007) 1713–1719. rodegenerative disease and ageing, Comb. Chem. High Throughput Screen. 12

[131] H.W. Vesper, S. Bhasin, C. Wang, S.S. Tai, L.A. Dodge, R.J. Singh, J. Nel- (2009) 212–228.

son, S. Ohorodnik, N.J. Clarke, W.A. Salameh, C.R. Parker Jr., R. Razdan, E.A. [158] M. Ogundare, S. Theofilopoulos, A. Lockhart, L.J. Hall, E. Arenas, J. Sjovall,

Monsell, G.L. Myers, Interlaboratory comparison study of serum total testos- A.G. Brenton, Y. Wang, W.J. Griffiths, Cerebrospinal fluid steroidomics: are

terone [corrected] measurements performed by mass spectrometry methods, bioactive bile acids present in brain? J. Biol. Chem. 285 (2010) 4666–4679.

Steroids 74 (2009) 498–503. [159] F. Badoud, J. Boccard, C. Schweizer, F. Pralong, M. Saugy, N. Baume, Profiling

[132] H.W. Vesper, J.C. Botelho, C. Shacklady, A. Smith, G.L. Myers, CDC project of steroid metabolites after transdermal and oral administration of testos-

on standardizing steroid hormone measurements, Steroids 73 (2008) terone by ultra-high pressure liquid chromatography coupled to quadrupole

1286–1292. time-of-flight mass spectrometry, J. Steroid Biochem. Mol. Biol. 138 (2013)

[133] D. French, Development and validation of a serum total testosterone liquid 222–235.

chromatography–tandem mass spectrometry (LC–MS/MS) assay calibrated [160] J. Boccard, F. Badoud, E. Grata, S. Ouertani, M. Hanafi, G. Mazerolles, P. Lanteri,

to NIST SRM 971, Clin. Chim. Acta 415 (2013) 109–117. J.L. Veuthey, M. Saugy, S. Rudaz, A steroidomic approach for biomarkers dis-

[134] M.M. Kushnir, A.L. Rockwood, J. Bergquist, Liquid chromatography–tandem covery in doping control, Forensic Sci. Int. 213 (2011) 85–94.

mass spectrometry applications in endocrinology, Mass Spectrom. Rev. 29 [161] J. Boccard, F. Badoud, N. Jan, R. Nicoli, C. Schweizer, F. Pralong, J.L. Veuthey, N.

(2010) 480–502. Baume, S. Rudaz, M. Saugy, Untargeted profiling of urinary steroid metabolites

[135] M. Rauh, Steroid measurement with LC–MS/MS in pediatric endocrinology, after testosterone ingestion: opening new perspectives for antidoping testing,

Mol. Cell. Endocrinol. 301 (2009) 272–281. Bioanalysis 6 (2014) 2523–2536.

[136] J. Abdel-Khalik, E. Bjorklund, M. Hansen, Simultaneous determination of [162] F. Jeanneret, J. Boccard, F. Badoud, O. Sorg, D. Tonoli, D. Pelclova, S. Vlck-

endogenous steroid hormones in human and animal plasma and serum by ova, D.N. Rutledge, C.F. Samer, D. Hochstrasser, J.H. Saurat, S. Rudaz, Human

liquid or gas chromatography coupled to tandem mass spectrometry, J. Chro- urinary biomarkers of dioxin exposure: analysis by metabolomics and biologi-

matogr. B 928 (2013) 58–77. cally driven data dimensionality reduction, Toxicol. Lett. 230 (2014) 234–243.

[137] R. Goodacre, S. Vaidyanathan, W.B. Dunn, G.G. Harrigan, D.B. Kell, [163] B.S. Kumar, B.C. Chung, O.S. Kwon, B.H. Jung, Discovery of common

Metabolomics by numbers: acquiring and understanding global metabolite urinary biomarkers for hepatotoxicity induced by carbon tetrachlo-

data, Trends Biotechnol. 22 (2004) 245–252. ride, acetaminophen and methotrexate by mass spectrometry-based

[138] K. Dettmer, P.A. Aronov, B.D. Hammock, Mass spectrometry-based metabolomics, J. Appl. Toxicol. 32 (2012) 505–520.

metabolomics, Mass Spectrom. Rev. 26 (2007) 51–78. [164] B.S. Kumar, Y.J. Lee, H.J. Yi, B.C. Chung, B.H. Jung, Discovery of safety biomark-

[139] G.A. Theodoridis, H.G. Gika, E.J. Want, I.D. Wilson, Liquid ers for atorvastatin in rat urine using mass spectrometry based metabolomics

chromatography–mass spectrometry based global metabolite profiling: a combined with global and targeted approach, Anal. Chim. Acta 661 (2010)

review, Anal. Chim. Acta 711 (2012) 7–16. 47–59.

[140] S. Castillo, P. Gopalacharyulu, L. Yetukuri, M. Oresic, Algorithms and tools for [165] A. Knight, Systematic reviews of animal experiments demonstrate poor con-

the preprocessing of LC–MS metabolomics data, Chemometr. Intell. Lab. 108 tributions toward human healthcare, Rev. Recent Clin. Trials 3 (2008) 89–96.

(2011) 23–32. [166] M.D. O’Connor, The 3R principle: advancing clinical application of human

[141] J. Boccard, J.L. Veuthey, S. Rudaz, Knowledge discovery in metabolomics: an pluripotent stem cells, Stem Cell Res. Ther. 4 (21) (2013) 1–7.

overview of MS data handling, J. Sep. Sci. 33 (2010) 290–304. [167] R.A. Scheltema, S. Decuypere, J.C. Dujardin, D.G. Watson, R.C. Jansen, R.

[142] M. Katajamaa, M. Oresic, Data processing for mass spectrometry-based Breitling, Simple data-reduction method for high-resolution LC–MS data in

metabolomics, J. Chromatogr. A 1158 (2007) 318–328. metabolomics, Bioanalysis 1 (2009) 1551–1557.

[143] X. Zhao, F. Xu, B. Qi, S. Hao, Y. Li, Y. Li, L. Zou, C. Lu, G. Xu, L. Hou, [168] H. Takahashi, T. Morimoto, N. Ogasawara, S. Kanaya, AMDORAP: non-targeted

Serum metabolomics study of polycystic ovary syndrome based on liquid metabolic profiling based on high-resolution LC–MS, BMC Bioinformatics 12

chromatography–mass spectrometry, J. Proteome Res. 13 (2014) 1101–1111. (259) (2011) 1–8.

[144] M. Hill, A. Parizek, R. Kancheva, M. Duskova, M. Velikova, L. Kriz, M. Klimkova, [169] D. Spaggiari, S. Fekete, P.J. Eugster, J.L. Veuthey, L. Geiser, S. Rudaz, D.

A. Paskova, Z. Zizka, P. Matucha, M. Meloun, L. Starka, Steroid metabolome in Guillarme, Contribution of various types of liquid chromatography–mass

plasma from the umbilical artery, umbilical vein, maternal cubital vein and spectrometry instruments to band broadening in fast analysis, J. Chromatogr.

in amniotic fluid in normal and preterm labor, J. Steroid Biochem. Mol. Biol. A 1310 (2013) 45–55.

121 (2010) 594–610. [170] W.B. Dunn, A. Erban, R.J.M. Weber, D.J. Creek, M. Brown, R. Breitling,

[145] M. Hill, A. Pasková,ˇ R. Kanceva,ˇ M. Velíková, J. Kubátová, L. Kancheva, K. Adam- T. Hankemeier, R. Goodacre, S. Neumann, J. Kopka, M.R. Viant, Mass

cová, M. Mikesová,ˇ Z. Ziˇ zka,ˇ M. Koucky,´ H. Sarapatková,ˇ V. Kacer,ˇ P. Matucha, appeal: metabolite identification in mass spectrometry-focused untargeted

M. Meloun, A. Parízek,ˇ Steroid profiling in pregnancy: a focus on the human metabolomics, Metabolomics 9 (2013) S44–S66.

fetus, J. Steroid Biochem. Mol. Biol. 139 (2014) 201–222. [171] A.V. Stachulski, X.L. Meng, Glucuronides from metabolites to medicines: a

[146] S.E. Jantti, M. Hartonen, M. Hilvo, H. Nygren, T. Hyotylainen, R.A. Ketola, R. survey of the in vivo generation, chemical synthesis and properties of glu-

Kostiainen, Steroid and steroid glucuronide profiles in urine during pregnancy curonides, Nat. Prod. Rep. 30 (2013) 806–848.

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

G Model

CHROMA-356635; No. of Pages 16 ARTICLE IN PRESS

16 F. Jeanneret et al. / J. Chromatogr. A xxx (2015) xxx–xxx

[172] A.V. Stachulski, J.R. Harding, J.C. Lindon, J.L. Maggs, B.K. Park, I.D. Wilson, Acyl [178] Y. Yan, D.L. Rempel, T.E. Holy, M.L. Gross, Mass spectrometry combinations for

glucuronides: biological activity, chemical reactivity, and chemical synthesis, structural characterization of sulfated-steroid metabolites, J. Am. Soc. Mass

J. Med. Chem. 49 (2006) 6931–6945. Spectrom. 25 (2014) 869–879.

[173] C.C. Waller, M.D. McLeod, A simple method for the small scale synthesis [179] D.W. Hill, T.M. Kertesz, D. Fontaine, R. Friedman, D.F. Grant, Mass spec-

and solid-phase extraction purification of steroid sulfates, Steroids 92 (2014) tral metabonomics beyond elemental formula: chemical database querying

74–80. by matching experimental with computational fragmentation spectra, Anal.

[174] R.A. Al-Horani, U.R. Desai, Chemical sulfation of small molecules-advances Chem. 80 (2008) 5574–5582.

and challenges, Tetrahedron 66 (2010) 2907–2918. [180] S. Wolf, S. Schmidt, M. Muller-Hannemann, S. Neumann, In silico fragmen-

[175] M. Clements, L.A. Li, Strategy of using microsome-based metabolite pro- tation for computer assisted identification of metabolite mass spectra, BMC

duction to facilitate the identification of endogenous metabolites by liquid Bioinformatics 11 (148) (2010) 1–12.

chromatography mass spectrometry, Anal. Chim. Acta 685 (2011) 36–44. [181] L.C. Menikarachchi, S. Cawley, D.W. Hill, L.M. Hall, L. Hall, S. Lai, J. Wilder, D.F.

[176] T.M. Williams, A.J. Kind, E. Houghton, D.W. Hill, Electrospray collision-induced Grant, MolFind: a software package enabling HPLC/MS-based identification

dissociation of testosterone and testosterone hydroxy analogs, J. Mass Spec- of unknown chemical structures, Anal. Chem. 84 (2012) 9388–9394.

trom. 34 (1999) 206–216. [182] T.M. Kertesz, D.W. Hill, D.R. Albaugh, L.H. Hall, L.M. Hall, D.F. Grant, Database

[177] K.M. Wooding, R.M. Barkley, J.A. Hankin, C.A. Johnson, A.P. Bradford, N. searching for structural identification of metabolites in complex biofluids for

Santoro, R.C. Murphy, Mechanism of formation of the major estradiol mass spectrometry-based metabonomics, Bioanalysis 1 (2009) 1627–1643.

product ions following collisional activation of the molecular anion in a [183] T.M. Kertesz, L.H. Hall, D.W. Hill, D.F. Grant, CE50: quantifying collision

tandem quadrupole mass spectrometer, J. Am. Soc. Mass Spectr. 24 (2013) induced dissociation energy for small molecule characterization and iden-

1451–1455. tification, J. Am. Soc. Mass Spectr. 20 (2009) 1759–1767.

Please cite this article in press as: F. Jeanneret, et al., Evaluation of steroidomics by liquid chromatography hyphenated to

mass spectrometry as a powerful analytical strategy for measuring human steroid perturbations, J. Chromatogr. A (2015), http://dx.doi.org/10.1016/j.chroma.2015.07.008

View publication stats