Open access peer-reviewed chapter

Testosterone Misuse

Written By

Zied Kaabia

Submitted: 17 September 2022 Reviewed: 22 November 2022 Published: 30 August 2023

DOI: 10.5772/intechopen.109110

From the Edited Volume

Testosterone - Functions, Uses, Deficiencies, and Substitution

Edited by Hirokazu Doi

Chapter metrics overview

57 Chapter Downloads

View Full Metrics

Abstract

Testosterone is a key compound of the anabolic androgenic steroids (AAS) family. It has largely been misused in human and animal doping targeting a muscle tissue growth and an enhancement of performances. Such practices constitute a violation against ethical values, food safety, and animal welfare. Consequently, the use of such substance is regulated by WADA and International committees for some animal species such as equine and bovine. Although efficient, the detection of testosterone misuse remains challenging in some cases due to its endogenous origin and its inter- and intra-individual level fluctuation in biological fluids. Novel analytical strategies have been developed and are continuously evolving in order to tackle this issue and to provide a better control of testosterone misuse.

Keywords

  • anti-doping control
  • analytical chemistry
  • mass spectrometry
  • regulation
  • anabolic androgenic steroids

1. Introduction

“Doping” as a term has usually been correlated to fraudulent practices undertaken by dishonest athletes to improve their sporting results. However, the use of this term overpasses the boundaries of human sport field to cover the field of sports and food-producing animals such as horses and bovine. Androgenic anabolic steroids (AAS) misuse, in particular, represents one of the hottest and most challenging topics in this context.

Testosterone belongs to androgenic anabolic steroids (AAS) family, which is a group of derivatives originating from the cholesterol biosynthesis in mammals or issued from synthetic chemical reactions. These compounds are reported to be masculinizing agents (androgenic) and promoting skeletal muscle building (anabolic) [1, 2]. The interesting anabolic properties were demonstrated for the first time by Brown-Sequard [3], who reported an improvement in strength and force upon an injection of an extract of dog and guinea pig testicles. Based on its beneficial effects, testosterone has originally been used as therapeutical agent for clinical treatments such as the treatment of hypogonadism or anemia. Consequently, the availability of testosterone increased, and its use was diverted from their original purpose.

In addition to their unethical use to enhance performances and produce higher volume of muscle carcasses, these doping practices pop out as a major concern and a threat for human and animal health and welfare.

In food-producing animals, testosterone is often used by unscrupulous cattle breeders in order to stimulate bovine muscle growth and make a higher profit with regard to the consequent gain in weight over a short period of time [4]. The worldwide debate about the use of such practices has existed since the discovery of their misuse. Several international projects such as the “Transatlantic Trade and Investment Partnership” tend to harmonize the laws regarding the use of AAS in the world. These projects are mainly bringing together for negotiating process the European Union countries, which are imposing a strict regulation system regarding the AAS misuse and the USA, regulations of which offer some permissions for the use of some AAS such as estradiol and stanozolol.

The strict regulation toward AAS misuse in food-producing animals adopted by the European Union countries originates from the various scandals that stroke the region due to the misuse of these substances. One of the biggest high-profile scandals occurred in the 1980s in Italy with the discovery of diethylstilbestrol, a growth hormone promoter, in baby food. Consequently, several measures have been taken by the European Union [5, 6]. According to these directives, adequate analytical methods had to be developed and implemented in the different accredited control laboratories in order to guarantee the consumer’s security toward these active chemicals [7]. The analytical methods are mainly based on chromatography coupled to mass spectrometry techniques permitting the detection of residues in biological matrices. The system revealed to be efficient for detecting a large spectrum of anabolic strategies. However, some particular cases remain challenging with regard to the current methodologies adopted by reference laboratories. The use of “hormone cocktails” is among these particular issues [8] and consists of the administration of different anabolic agents such as testosterone, each occurring at low concentration levels. These low quantities of anabolic agents cannot be detected with regard to the detectability limitations of the analytical methods. The use of “Designer Drugs” constitutes another issue for control laboratories. Indeed, “Designers” are exclusively xenobiotic substances not listed among prohibited substances. Considering the lack of knowledge on their exact chemical structure, they cannot be detected by the classical targeted analytical strategies. Finally, the use of natural steroids such as testosterone constitutes a challenge for control laboratories with regard to the similar structures exhibited by these administered substances and their corresponding endogenous analogs.

In the equestrian world, “doping” has been in existence since the ancient Roman era where horses were fed with a “magic potion” called “Hydromel,” a mixture of milk and honey, in order to improve their physical performances in the Arena. In the twentieth century, administering alkaloids such as cocaine or strytchine to horses emerged as a new doping treatment permitting to minimize the feeling of physical tiredness. The fraudulent use of these exciting substances increased simultaneously with the appearance of horserace bets, which generated important financial gains. In addition, the use of AAS spread considering their power to enhance physical performances. In order to tackle this issue threatening the equestrian world as a whole in terms of ethical values and animal welfare and with the hope to avoid a tarnished image of horse racing competitions, the International Federation of Horseracing Authorities (IFHA) put into practice a strict control system similar to the one regulating athletes doping implemented by the World Anti-Doping Agency (WADA). This control system, “the racing code,” forbids the use of any medication during horseracing competitions and bans in particular the use of AAS in horses during training sessions, pre-competition or competition periods. Targeting an efficient application of this control system, IFHA reference laboratories are committed to developing analytical methods mainly based on chromatography coupled to mass spectrometry techniques allowing the identification and quantification of residues in biological matrices. Although being highly efficient, these methods highlighted, similarly to the official analytical methods adopted in food-producing animal field, some limitations for detecting naturally occurring steroids misuse.

Advertisement

2. Structure and pharmacology of steroid hormones

Anabolic-androgenic steroid hormones share the same substructure based on a sterane nucleus (Figure 1). The different families of steroid hormones result from the existence of minor modifications occurring at the sterane nucleus. Progestagens such as progesterone, corticosteroids such as cortisone and aldosterone, estrogens such as estradiol, and androgens such as testosterone are some example of these families.

Figure 1.

Structure of a cyclopentanoperhydrophenantrene nucleus or sterane.

All of these steroids are derived from cholesterol, which is mainly biosynthesized after dietary intake. This steroid biosynthetic pathway begins with an oxidation of the side chain of the cholesterol to form the androgens prior to the formation of estrogens. This oxidation reaction, catalyzed by the Cytochrome P450, generates pregnenolone, which whether can be biotransformed into progesterone through the enzyme 3β-HSD initiating the creation of the progestagen family or into 17α-hydroxypregnenolone through an oxidation by CP17α. The latter compound is subsequently metabolized into DHEA and then to 4-androstenedione, precursor of testosterone.

Estrogens are formed during the reaction of aromatization occurring under the action of CP450. The aromatization consists of the oxidation of the methyl group at position C19 and the consecutive elimination of this newly formed to form estrogens such as estrone and estradiol. This reaction results in the creation of secondary products such as nandrolone. All these transformations are summarized in Figure 2.

Figure 2.

Global overview of the metabolization products deriving from cholesterol and the occurring biotransformations generating the different steroid families.

The secretion of naturally occurring hormones is enhanced by the Luteinizing Hormone (LH), which activates the biotransformation of cholesterol into pregnenolone and the Follicle-Stimulating Hormone (FSH), which participates in the aromatization of testosterone into estradiols. The secretion of two latter hormones is regulated by the Gonadotropin-Releasing Hormone (GnRH) according to the occurring plasmatic concentration levels of steroid hormones.

The steroid hormones are subsequently submitted to phase I biotransformations (oxidations, reductions, epimerizations, etc.) and phase II biostransformations (mainly sulfo and glucuruno conjugation) in order to be eliminated predominantly through urine.

The nature of these biotransformations is essentially correlated to the steroid nature and the occurring enzymatic system in the organism. Therefore, the steroids biotransformations present some differences depending on the species. For instance, phase II steroid biotransformations consist mainly of sulfoconjugation reactions in equine species while they consist of glucurunoconjugation reactions in humans and bovine species.

While the previously described steroid hormones such as testosterone occur naturally in the organism, some other steroids are exclusively synthetic. They usually exhibit minor modifications to their endogenous analogues such as methyltestosterone. They can also be part of a new category of steroids called “Designers.” These steroids are unmarketed compounds exhibiting a chemical structure based on marketed substances presenting minor modifications. They are usually synthesized as part of the pharmaceutical industry for medical use. With regard to their inexistence in the list of WADA banned substances, their detection using classical targeted analytical methods becomes problematic. Estra-4,9-diene-3,17-dione [9] and tetrahydrogestrinone [10] are some examples of this category of synthetic compounds.

Advertisement

3. Use of steroid hormones

The use of anabolic steroids in food-producing and sport animals has been flourishing with regard to their capacity to increase zootechnic performances [1112]. Testosterone has been one of the main steroids used in this context. Typical quantities of administered steroids to human, bovine, and equine for doping purposes range between hundreds of micrograms and few milligrams per kilogram. The administration of these steroids may be performed orally, through parenteral or alternative pathways:

Oral pathway: minor chemical modifications to the structure of natural anabolic steroids have to be performed prior to an oral administration. These modifications protect the steroid from a submission to a quick metabolization through the hepatic pathway. These modifications consist mainly of an alkylation or an esterification. One example of such steroids is the methyl testosterone, efficiency of which is four times higher than testosterone when administered orally.

Parenteral pathway: this relates to any administration occurring on a different way than the digestive one. When the steroid is administered under its free form through this pathway, it undergoes a quick metabolization leading to its degradation and elimination. Therefore, a step of esterification of the steroid is necessary to provide the desired effect on an extended period of time. The intramuscular administration of the steroid ester is immediately followed by an enzymatic hydrolysis in blood [13]. Prior to hydrolysis, the steroid ester migrates gradually from its lipophilic excipient toward blood stream [14]. This gradual liberation results in a longer effect of the doping substance. The effect is even longer when the steroid ester presents a longer lipophilic side chain [15]. Once the active steroid is formed, it is transferred through the blood stream to the corresponding hormonal receptor.

The administration of the anabolic steroid could be also performed through an intravenous pathway or through solid implants.

Alternative pathways: the transdermal application of steroids is also a possible way for administration with regard to the lipophilic nature of the skin. Anabolic steroids are usually administered through the application of spray or “pour-on” [16]. These pathways have the advantage not to submit the steroids to the first hepatic transition caused by the liver. However, a frequent application is necessary with regard to the low biodisponibility of the steroids using this pathway.

3.1 Transport of steroid hormones

Hormone steroids are rarely present in the blood under their free form. Instead, they are carried in the bloodstream bound to serum carrier proteins [17]. There are two categories of binding proteins:

Non-specific binding carrier proteins: albumin is one example of these binding proteins, and it is characterized by a high capacity of transport but a low affinity with steroids. It is found at a relatively high concentration in blood. Its main function is to ensure the transport of steroids to their corresponding hormonal receptors.

Specific binding carrier proteins: SHBG (Sex Hormones Binding Globulin) is an example of this class of proteins [18]. They are characterized by a high affinity with steroids but a low capacity of transport.

The bioavailability of the endogenous steroids is ensured by these proteins. These proteins play indeed the role of a reservoir of steroids regulating the concentration levels of steroid hormones in the metabolism. Moreover, these proteins protect the transported steroids by preventing their biotransformation and subsequent diffusion and elimination [19].

3.2 Protein production activation

When the steroids reach the target cell, they penetrate through the phospholipidic membrane of the cell by passive diffusion. This passage is guaranteed with regard to the fat-soluble nature of the hormonal steroids. Once inside the cell, the steroids bind to their specific androgenic receptor located in cytoplasm [20]. This binding results in the formation of a steroid-receptor complex, which infiltrates the nucleus to bind to a specific receptor located on chromatine. Consequently, the production of RNA messenger (mRNA) is initiated [21]. These mRNAs are then transported to the cytoplasm for further proteins synthesis.

3.3 Biological effects

The AAS are well-known to enhance muscle growth (anabolic effect) and to promote masculinization (androgenic effect). In addition, the use of these substances may lead to harmful effects such as hypogonadism and cardiovascular risks [22]. These effects are mainly caused by the disturbance occurring through the administration of exogenous steroids, which results in a negative feedback on the secretion of natural steroid hormones. Furthermore, the increase in steroids concentration leads to a higher synthesis of proteins and subsequent higher amount of muscular tissue, which needs, as a consequence, a higher volume of blood irrigating it and finally causing cardiovascular problems. High blood pressure could be observed also due to a regular administration of testosterone. In fact, such an administration causes a reduction in the secretion of HDL (High-Density Lipoproteins) resulting in an imbalance between HDL and LDL amounts leading to hypertension troubles. Other effects could be observed for female subjects submitted to a regular intake of AAS on hormonal cycle (ovulation and menstruation). The abuse affects also the psychological aspect and could lead to an aggressive attitude and even to depression.

Advertisement

4. Regulation

4.1 Bovine

The use of anabolic steroids as growth promoters in food-producing animals is prohibited in the European Union according to the EU Directive [5]. Different EU Directives have been issued to date in order to avoid any misuse of these substances in food-producing animals. The main purpose of these directives is to guarantee the security of the consumer toward these active chemicals [7].

The harmonization of the regulation concerning the use of these growth promoters in bovine, mainly between the European Union and the United States, has been problematic. The problem became even greater with the increase in the trade flows of bovine meat all over the world. While the use of anabolic steroids in the European Union is strictly banned in food-producing animals, the use of implants of 17β-testosterone, la 17β-estradiol, progesterone, trenbolone, or zeranol in cattle is still legal outside the European Union. As a consequence of the important position of the European Union in the bovine meat trade market, the bovine field in the United States witnessed a gradual decrease in the use of growth promoters. Although several arrangements have recently been taken in order to meet the different expectations of the different parties (MOU) (WT/DS26/28, September 30, 2009, 74 Federal Register), the problem is still persisting. In June 2013, talks between the European Union and the United States have been taken over in order to find a solution to this recurring issue. In July 2013, the TTIP “Transatlantic Trade and Investment Partnership,” a project aiming to set a transatlantic market based on a liberal model ruled by a free circulation of goods and investments, was created. This project will also cover the agriculture field and would affect the regulation put into practice for the control of agricultural products such as bovine meat quality.

The recognized reference laboratories are submitted to the identification criteria established by the European Decisions 2002/657, which defines mainly the tolerated relative retention time and abundances percentual errors depending on the performed analytical technique.

4.2 Equine

The use of anabolic steroids is strictly banned in equestrian sports. Unlike WADA, where the list of prohibited substances for athletes is classified according to their respective pharmacological families, the IFHA has classified the doping substances for horses with regard to their respective target system of the organism. For instance, article 6 of IFHA “International Agreement on Breeding, Racing and Wagering” highlights the ban of substances that affect the nervous, the cardiovascular, the respiratory, the digestive, the urinary, the reproductive, the musculoskeletal, the blood, the immune, or the endocrine system.

The control of the use of these substances protects the integrity of horse races. This control can be conducted at any time of the year on a large panel of biological matrices.

With regard to the natural occurrence of some anabolic steroids such as nandrolone, the International Equestrian Federation put into practice in March 1988 an advisory committee formed of analysts and veterinarians, whose duty is to establish concentration or ratio thresholds of certain prohibited substances when it is necessary. Consequently, different confirmation thresholds have been approved by IFHA for natural occurring anabolic steroids misuse. For instance, boldenone urinary concentration should not overpass 15 ng/mL in entire male horses, testosterone urinary concentration should be inferior to 20 ng/mL in gelded horses and less than 55 ng/mL in mares and fillies. Finally, 5α-estrane-3β,17α-diol, one of the main recognized biomarkers of nandrolone administration, urinary concentration should be lower than 45 ng/mL in entire male horses [23]. In addition to the latter criterion, a complementary threshold consisting of a ratio of the urinary concentration of 5α-estrane-3β,17α-diol and 5(10)-estrene-3β,17α-diol of 1 has been supplemented [23].

The reference IFHA laboratories are submitted to the identification criteria established by AORC (Association of Official Racing Chemists), which defines mainly the tolerated retention time and relative abundances percentual errors depending on the analytical technique adopted [24].

Advertisement

5. The detection of steroid abuse in bovine and equine species

With regard to the high number of analysis performed by control laboratories, high-throughput screening analytical methods are required in order to offer a first discrimination between negative and suspicious samples. Once the sample is considered suspicious, it is subsequently submitted to confirmatory methods.

5.1 Screening

Screening methods require a dual compromising aspect by being high throughput and multi-residue but at the same time showing a high efficiency and low costs. These methods allow the detection or the concentration level estimation of a certain group of prohibited doping agents. Consequently, they affect a suspicious or a compliant status to the biological sample. These methods should present an as low as possible of “false positive” results and especially minimize the rate of “false negative” results (Decision 2002/657/EEC). These analytical techniques can either be immunological or based on chromatography coupled to mass spectrometry. The following synthesis review will highlight additionally novel analytical techniques, “omics” techniques, aiming at an indirect screening of prohibited substances, in particular natural occurring steroids.

5.1.1 Immunochemical techniques

Immunochemical techniques are based on an antigen-antibody interaction. There are two main techniques used: Radioimmunoassay (RIA), which is based on a radio element, and enzyme-linked immunosorbent assay (ELISA), which is based on an enzymatic system. These techniques permit the quantification of steroid hormones through the bindings established between their corresponding tracers, an analog of antigen, and the antibodies occurring in the biological sample. They classify hence the sample as compliant or suspicious. RIA needs a prior purification of the biological sample and offers several advantages, mainly the possibility to analyze different substances at once and to denature the sex hormone binding globulin (SHBG), which is transporting and bound to these substances. However, this tool requires large sample volumes especially when the substance is present at low concentration levels. It could also lead to erroneous results due to the reactivity of the antibody [25]. As for ELISA, it permits to measure the antigen through an enzymatic pathway. ELISA represented the reference tool for screening steroids abuse before the emergence of mass spectrometry tools. The main advantage of ELISA, in comparison with RIA, lies in the absence of a radio element tracer. However, ELISA exhibits lower specificity as compared with what can offer mass spectrometry.

5.1.2 Chromatography coupled to mass spectrometry

The analytical methods based on this technique allow identifying the administered substances or the markers resulting from such an administration. As far as endogenous AAS are concerned, this technique permits to give an idea about their concentration levels in the biological matrix in order to compare it with the settled thresholds. Two main branches of chromatography have been widely used for screening purposes: Gas Chromatography and Liquid Chromatography coupled to mass spectrometry.

5.1.2.1 Gas Chromatography-Mass Spectrometry (GC-MS)

GC-MS remains the most widespread technique used for screening analysis of steroid hormones in the anti-doping control laboratories [26, 27]. A sample preparation step consisting of a hydrolysis, extraction, concentration, and purification of the analytes is necessary in order to detect trace levels of substances of interest in the biological matrix. This technique usually requires a prior derivatization of the compounds with N-methyl-N-trimethylsilyl-trifluoroacetamide (MSTFA), PentaFluoroBenzaldehyde (PFB), or PentaFluoroPropionic Anhydride (PFPA) to enable their analysis on GC-MS. 5MS columns (5% phenyl and 95% of methylpolysiloxane) are usually used for chromatographic separation. Negative Chemical Ionization (NCI) and Electron Impact (EI) ionization modes are frequently used for analyses. Different analyzers have been used: ion trap [28], simple and triple quadripole [27, 29]. High-Resolution Mass spectrometry (HRMS) analyzers have also been used such as Time of Flight and Electromagnetic sector [29, 30].

The GC-MS techniques permit to obtain a better chromatographic separation than when operating with a Liquid Chromatography-Mass Spectrometry (LC-MS), even with the emergence of Ultra Performance Liquid Chromatography (UPLC). The main advantage of GC-MS, in comparison with LC-MS, is the absence of ion suppression effects.

GC-MS was in particular used for testosterone screening in equine species [31]. In bovine, GC-MS was used to figure out some potential biomarkers and thresholds for natural occurring steroids misuse. For instance, 5α-pregnane-3β,17α-diol was suggested as a biomarker for progesterone misuse, 5β-androstane-3α,17β-diol for testosterone misuse, 17β-androst-1-ene-3-one for boldenone misuse, and 17α-estradiol for estradiol misuse [32, 33].

New bidimensional separative techniques (GC × GC) have recently been reported in the anti-doping field [34]. The chromatographic separation on these systems is performed through two capillary columns placed in series permitting to obtain a higher separation power in comparison with unidimensional GC technique. In addition, the bidimensional gas chromatography results in a simplification of the sample preparation step by eliminating the purification process [35].

5.1.2.2 Liquid Chromatography-Mass Spectrometry (LC-MS)

LC-MS use in the anti-doping field as a screening technique has greatly increased with regard to the performance allowed [36]. LC-MS process results in a drastic gain of time in comparison with GC-MS technique regarding the lack of a derivatization step. Moreover, it enables the study of the conjugated form of the steroid in addition to its free form and therefore requires no hydrolysis step. This capacity allows a better understanding of the global metabolism of the steroids.

The ionization modes frequently used when dealing with AAS analyses in LC-MS are Atmospheric Pressure Chemical Ionization (APCI), Atmospheric Pressure Photo-Ionization (APPI), and Electrospray Ionization (ESI) [37]. The ionization type and mode depend strongly on the nature of the steroid. For instance, conjugated steroids are usually ionized in ESI+ or ESI modes [38] and free steroid uniquely in ESI+ mode.

In addition to the mass analyzers described previously for GC-MS analyses, LC offers the possibility to be coupled with other types of high-resolution mass analyzers such as Orbitrap or Fourier Transform Ion Cyclotron Resonance (FT-ICR) [39]. All of these analyzers offer the possibility to work with a high throughput, which is extremely important criterion for screening analyses [40, 41].

LC-MS was performed for screening purposes of more than 50 doping agents including testosterone, estradiol, boldenone, and norandrostenedione in porcine and bovine species [42]. It also detects fraudulent use of boldenone in bovine through the detection of the sulfoconjugated form [43]. In addition, it was performed in equine to detect testosterone, nandrolone, and boldenone misuse in entire male horses through the quantification of sulfoconjugated forms [44].

The emergence of UPLC permitted to improve the separation power of steroids and considerably reduced the analysis time. Different studies used this technique for AAS analysis and screening [45].

5.1.3 “Omic” techniques

“Omic” strategies have been applied in different scientific research fields mainly for biological applications. The “omic” techniques aim to study the recorded variations at different biological levels ranging from gene sequencing to metabolites expression [46]. It generates consequently a large amount of data permitting to understand the biological functioning of the organism as a whole. In the present context, the main objective of these approaches would be to determine biomarkers capable of characterizing the administration of a particular anabolic agent or a particular family of those agents [47]. These techniques have already shown their relevance in the anti-doping field, particularly, dealing with the issue of natural occurring steroids misuse. “Genomics” and “transcriptomics,” “proteomics,” and “metabolomics” have in particular been investigated to reveal candidate biomarkers that would serve screening purposes. Most of reported transcriptomic studies in the anti-doping field dealt with the influence of peptide hormones such as human Growth Hormone (hGH), Erythropoietin EPO and Insulin-like growth factor (IGF), and anabolic steroid hormones on the Deoxyribonucleic Acid (DNA) transcription [48, 49, 50]. These studies permitted to highlight some gene biomarkers.

The second strategy of interest in the cascade of “omics” (Figure 3) is “proteomics,” which sheds the light on the proteome investigation. This approach is of high importance in the anti-doping field since permitting a profiling of the proteins concentration variations following the administration of anabolic substances [51]. Proteomics study may either be performed in an untargeted way or more targeted, focusing on a limited number of proteins of interest, a-priori selected. The untargeted proteomic study is usually performed in three main steps: first, the proteins are separated based on two-dimensional electrophoresis (2-DE) or multidimensional chromatography, then the analysis of the separated proteins is made by Matrix-Assisted Laser Desorption Ionization-Time Of Flight (MALDI-TOF) or MALDI-TOF/TOF, and finally, the proteins are identified. Very few untargeted proteomics works are reported in the field. However, several studies reported the use of targeted proteomics. For instance, Mooney confirmed the decrease of binding capacity of SHBG globulins in bovine following an administration of AAS [52]. In another study, Cacciatore demonstrated the variations occurring in concentration levels of IGF1, inhibins, and osteocalcin after administration of estradiol and nandrolone [53]. In equine, Nagata confirmed a decrease in Luteinizing Hormone (LH) and inhibins concentration levels after an administration of nandrolone in stallions [54].

Figure 3.

“ Omics ” cascade.

“Metabolomics” is the third family of interest in this “omics” family cascade. It focuses on the metabolome that is formed by the whole group of low-molecular-weight molecules. Sugar, organic acids, amino acids, vitamins, and steroids are part of the molecules included in metabolomic studies. Metabolomics consists of the study of the disturbance resulting from a genetic tampering of the organism or from a toxicological exposition or a treatment. The main objective of metabolomics is to shed light on compounds of interest potentially used to detect biological disturbance encountered by the organism. Many studies in the anti-doping field have evaluated this strategy, namely in human [55, 56], equine [57], or bovine [58, 59] species. These studies confirmed the feasibility and the efficiency of such an approach in this field. The different steps encountered for a metabolomic study are represented in Figure 4.

Figure 4.

The different steps of a metabolomic study.

“Steroidomics,” a subfamily of “Metabolomics,” focuses on the study of the steroidome, the whole group of steroids in the organism. Steroidomics exhibits a similar analytical workflow as the one performed in metabolomics except that it focuses, through sample preparation and acquisition mode, on steroids monitoring [60]. It may be of interest in the anti-doping field, to study the variations of steroid concentration. This monitoring enables a better understanding of the consequences of anabolic steroids administration on AAS secretions in the organism. Consequently, a more accurate identification of doping cases with natural occurring steroids and an identification of potential candidate biomarkers are realized. This multidimensional approach mixing analytical and statistical tools requires an adequate sample preparation step and a specific detection method. Once the steroid profiles are obtained, they are submitted to a multivariate statistical processing to offer a first general view of the data distribution and the generated discrimination. Principal Components Analysis (PCA) and Orthogonal Partial Least Squares (OPLS) are two examples of, respectively, non-supervised and supervised statistical multivariate analysis tools. While PCA models permit to obtain a first overview of the sample distribution without any a priori instruction regarding their status, OPLS models are characterized by an incremented Y variable indicating the status of the sample and affecting a supervised nature to the model. Before proceeding to the statistical process of the data, the latter is normalized in order to decrease the statistical dominance of very intense peaks over less intense but interesting peaks. Finally, quality control samples, consisting of pools of the different samples of the study, should be injected all along the analyzed batches to assess the analytical robustness of the whole process and guarantee accurate analytical findings. Steroidomics can be divided in three main categories: non-targeted steroidomics, semi-targeted steroidomics, and targeted steroidomics. Non-targeted steroidomics requires a minimal sample preparation to avoid the loss of any steroid of interest. Rijk tested this technique to evidence many phase I and II steroid hormones biomarkers following testosterone precursors, DeHydroEpiAndrosterone (DHEA) and pregnenolone, administration in bovine [61]. Semi-targeted steroidomics consist of the assessment and monitoring of a specific fragment characterizing a group or a family of steroids. This approach has been used by Thevis, who discovered new synthetic steroids based on fragments at m/z 77, 91, and 105 corresponding respectively to fragments of steroids A ring and phenol group of estrogens [62]. In another study, Anizan selected fragment ions at m/z 97 and 113 to identify new sulfo and glucuruno-conjugated steroids attesting of an illegal administration of testosterone precursor, androstenedione, to bovine [63]. Finally, targeted steroidomics consists of the quantitative profiling of a selected set of steroids in order to study their concentration levels occurring variations following an anabolic treatment. Such a strategy was for example performed in order to establish suspicion thresholds and identify new biomarkers for DHEA, DiHydroTestosterone (DHT), and testosterone administration in men [64], to discriminate estradiol-treated bovine from the control group [65] and nandrolone-treated equine from control group [66] and served to evidence the evolution of steroid concentrations with regard to the seasonality [67].

5.2 Confirmatory strategies

The confirmation step is performed once the sample is declared suspicious following the screening process. The following part will review two main analytical approaches implemented to confirm natural AAS misuse.

5.2.1 Steroid esters detection

Steroid esters are synthetic compounds composed of a steroid moiety (the active part of the substance) and a side chain, which can be located on carbon 3 or more often on carbon 17 of the steroid. Since steroid esters are widely used in bovine and equine, several analytical strategies have already been developed to detect their misuse.

With regard to their lipophilic nature, the steroid esters cannot be detected in hydrophilic matrices such as urine. However, they can be detected in tissues [68], fat [69], hair [70], and blood [71].

While tissues and fat can only be collected at the slaughterhouse, hair and blood may be collected unambiguously from bovine and equine. Regarding hair, substances are actually transmitted to the hair through the hair follicle from blood stream or from sweat to the hair shaft. Hair can consequently provide a historical background on the steroid esters misuse. However, hair is a relatively complex matrix, and the analytical process adopted to extract substances from it is quite challenging. In fact, using harsh analytical conditions such as strong acidic or basic digestion of the hair can lead to the degradation of the ester. Instead, the use of soft analytical conditions such as methanolic extraction will result in a very low extraction recovery of the ester.

Taking into account these drawbacks, blood appears to be an interesting matrix with regard to its containment in steroid esters and the relatively easy analytical process to perform for extraction. The esters are transported through this matrix to the target cells and are usually bound to some blood proteins such as albumin [72] and SHBG [73]. These esters are gradually hydrolyzed in the blood stream by enzymatic esterase and are consequently found at trace levels (dozens to hundreds of pg/mL) [74]. Therefore, very sensitive analytical methods have been developed in order to detect them in blood [75] using some specific derivatization reactions in order to optimize the sensitivity [76, 77].

5.2.2 Isotopic approach

This approach consists of calculating isotopic ratios of different chemical elements such as 13C/12C. This ratio can be expressed as an isotopic deviation following the application of the equation below:

δ13C[]=((13C/12C)sample(13C/12C)standard(13C/12C)standard)×103E1

This ratio is measured based on a reference compound, the isotopic deviation of which is fixed at zero may be used for the purpose (e.g., Vienna Pee Dee Belemite (VPDB)).

In the anti-doping field, the purpose of this technique is to differentiate endogenous and exogenous anabolic steroids according to their enrichment in 13C. The first applications involving the use of a GC-C-IRMS permitted to different anti-doping laboratories to identify exogenous testosterone in human [78, 79, 80]. This technique covered thereafter the animal anti-doping field and different studies using GC-C-IRMS focused on problematic issues related to the administration of natural steroids to equine [81] and bovine [82].

The small differences between endogenous and exogenous anabolic steroids isotopic deviations are one of the main difficulties encountered when applying the GC-Isotope Ratio Mass Spectrometry (IRMS) technique. In fact, the endogenous isotopic deviation of anabolic steroids depends mainly on the diet [83, 84]. Depending on the type of food consumed, the isotopic deviation of ingested cholesterol, and hence deriving anabolic steroids, will not be the same. Some plants called “C3 plants” such as wheat, rice, and soya exhibit low endogenous isotopic deviations in comparable ranges with those exhibited by synthetic steroids, mainly synthesized starting from C3 plants. However, the “C4 plants” such as maize and sugar cane generate a high endogenous isotopic deviation, which in this case of food-based diet permits to identify unambiguously exogenous administration of anabolic steroids.

Advertisement

6. Conclusion and perspectives

While the classical analytical methods, targeting a single or a large panel of anabolic substances, as currently implemented in the anti-doping and control laboratories are efficient to detect abuse with a large set of prohibited compounds, they have been showing some limits to detect abuse in some specific cases related to androgenic anabolic steroids misuse in racing and food producing animals. The difficulty has become even greater with the application of new doping methodologies aiming at bypassing the current analytical methods detection limits. The administrations of anabolic steroids cocktails, designer steroids, or natural occurring steroids are some examples of these new doping methodologies, which permit to escape control.

The use of natural occurring steroids such as testosterone appears to be one of the most challenging and problematic issues for doping control laboratories. With regard to their natural occurrence in the metabolism, the differentiation between their endogenous and exogenous origin becomes problematic. Moreover, the concentration levels of these steroid hormones vary between the different subjects part of the same species introducing inter-variability factors. Furthermore, these levels vary in the same subject at different time points due to different parameters such as seasonality, mating, and diet, which define the intra-variability factors. These two factors render the settling of a decision limit for a natural occurring steroid misuse difficult.

Testosterone in particular is among these steroids a problematic one for control laboratories since it has been shown to occur naturally in equine and bovine species while its administration under synthetic form is known to be very potent. Therefore, analytical strategies allowing to point out the exogenous administration are more than expected by the scientific community.

While confirmatory strategies seem to enable tackling the natural hormones issue and fulfill corresponding requirements, an additional effort appears necessary to validate the developed screening approach based on steroidomic profiling.

With regard to the general issue of natural steroids misuse in equine, more research work is needed in order to investigate minor metabolites of such administrations, which can reinforce the current international criteria. A stronger collaboration between the different reference laboratories worldwide is essential in order to settle population reference ranges of endogenous AAS and hence obtain a repository for future steroid doping challenges. A deeper investigation of the biosynthetic and physiological system of horses is also very important in order to have a better understanding of the biosynthetic pathway of endogenous steroids and hence generating a higher efficiency in tackling the issue.

Furthermore, other “upstream” omics techniques such as “genomic” and “transcriptomic” have been reported to be efficient for detecting natural steroids misuse and collaboration between these different fields would offer a mass global approach. For instance, Cannizzo developed a method based on DNA microarray data analysis [85] showing a large set of differentially expressed genes between controls and prednisolone treated bovine. Moreover, genomic instability and DNA damage were proven after nandrolone decanoate administration to rats [86]. However, such genotyping is costly in addition to the uneasiness of its application in the context of doping control. Many studies reported the possibility of generating information about the genotype starting from the phenotype and specifically steroid profiles [87, 88, 89]. These reported findings would offer a general overview of different organism levels and would be of a great help to evidence several challenging doping cases.

References

  1. 1. Heitzman RJ. The effectiveness of anabolic agents in increasing rate of growth in farm animals; report on experiments in cattle. Environmental Quality and Safety. 1976;5:89-98
  2. 2. Barceloux DG, Palmer RB. Anabolic—Androgenic steroids. Disease-a-Month. 2013;59:226-248
  3. 3. Brown S. Note on the effects produced on man by subcutaneous injections of a liquid obtained from the testicles of animals. The Lancet. 1889;134:105-107
  4. 4. Moran C, Quirke JF, Prendiville DJ, Bourke S, Roche JF. The effect of estradiol, trenbolone acetate, or zeranol on growth rate, mammary development, carcass traits, and plasma estradiol concentrations of beef heifers. Journal of Animal Science. 1991;69:4249-4258
  5. 5. Council Directive 96/22/EC of 29 April 1996 concerning the prohibition on the use in stockfarming of certain substances having a hormonal or thyrostatic action and of ß-agonists, and repealing Directives 81/602/EEC, 88/146/EEC and 88/299/EEC
  6. 6. Council Directive 96/23/EC of 29 April 1996 on measures to monitor certain substances and residues thereof in live animals and animal products and repealing Directives 89/187/EEC and 91/664/EEC
  7. 7. Decision 2002/657/EEC CD, 2002. The Official Journal of European Communities
  8. 8. Cantiello M, Carletti M, Cannizzo FT, Nebbia C, Bellino C, Pié S, et al. Effects of an illicit cocktail on serum immunoglobulins, lymphocyte proliferation and cytokine gene expression in the veal calf. Toxicology. 2007;242:39-51
  9. 9. Scarth JP, Clarke AD, Teale P, Pearce CM. Comparative in vitro metabolism of the ‘designer’ steroid estra-4,9-diene-3,17-dione between the equine, canine and human: Identification of target metabolites for use in sports doping control. Steroids. 2010;75:643-652
  10. 10. Catlin DH, Sekera MH, Ahrens BD, Starcevic B, Chang Y-C, Hatton CK. Tetrahydrogestrinone: Discovery, synthesis, and detection in urine. Rapid Communications in Mass Spectrometry. 2004;18:1245-1049
  11. 11. Pitts SC, Davis M. An evidence-based analysis of anabolic steroids as performance enhancers in horses. Equine Veterinary Education. 2007;19:388-391
  12. 12. Fajt VR, McCook C. An evidence-based analysis of anabolic steroids as therapeutic agents in horses. Equine Veterinary Education. 2008;20:542-544
  13. 13. Fujioka M, Shinohara Y, Baba S, Irie M, Inoue K. Pharmacokinetic properties of testosterone propionate in norman men. Journal of Clinical Endocrinology & Metabolism. 1986;63:1361-1364
  14. 14. Enever RP, Fotherby K, Naderi S, Lewis GA. Long-acting contraceptive agents: The influence of physicochemical properties of some esters of norethisterone upon the plasma levels of free norethisterone. Steroids. 1983;41:381-396
  15. 15. Van der Vies J. Implications of basic pharmacology in the therapy with esters of nandrolone. Acta Edocrinologica Supplementum. 1985;271:38-44
  16. 16. Stolker A, Groot M, Lasaroms J, Nijrolder A, Blokland M, Riedmaier I, et al. Detectability of testosterone esters and estradiol benzoate in bovine hair and plasma following pour-on treatment. Analytical and Bioanalytical Chemistry. 2009;395:1075-1087
  17. 17. Tikkanen MJ, Vihma V, Jauhiainen M, Höckerstedt A, Helisten H, Kaamanen M. Lipoprotein-associated estrogens. Cardiovascular Research. 2002;56:184-188
  18. 18. Hammond GL, Robinson PA, Sugino H, Ward DN, Finne J. Physicochemical characteristics of human sex hormone binding globulin: Evidence for two identical subunits. Journal of Steroid Biochemistry. 1986;24:815-824
  19. 19. Lee IR, Greed LC, Hahnel R. Comparative measurements of plasma binding capacity and concentration of human sex hormone binding globulin. Clinica Chimica Acta. 1984;137:131-139
  20. 20. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, et al. The nuclear receptor superfamily: The second decade. Cell. 1995;83:835-839
  21. 21. Lehmann S. Etude de DAX-1 et de sa perte de fonction dans l'hypoplasie congénitale des glandes surrénales [PhD]. Strasbourg, France: Université Louis Pasteur Strasbourg I; 2005
  22. 22. Van Amsterdam J, Opperhuizen A, Hartgens F. Adverse health effects of anabolic-androgenic steroids. Regulatory Toxicology and Pharmacology. 2010;57:117-123
  23. 23. IFHA. International Agreement on Breeding, Racing and Wagering. Article 6. Paris, France: International Federation of Horseracing Authorities; 2022
  24. 24. AORC. AORC Guidelines for the Minimum Criteria for Identification by Chromatography and Mass Spectrometry. Connecticut, USA: Association of Racing Chemists; 2016
  25. 25. Stanczyk FZ, Clarke NJ. Advantages and challenges of mass spectrometry assays for steroid hormones. The Journal of Steroid Biochemistry and Molecular Biology. 2010;121:491-495
  26. 26. Marcos J, Pascual JA, de la Torre X, Segura J. Fast screening of anabolic steroids and other banned doping substances in human urine by gas chromatography/tandem mass spectrometry. Journal of Mass Spectrometry. 2002;37:1059-1073
  27. 27. De Brabander H, Van Gansbeke W, Geldof L, Van Eenoo P. An improved gas chromatography screening method for doping substances using triple quadrupole mass spectrometry, with an emphasis on quality assurance. Biomedical Chromatography. 2012;26:1416-1435
  28. 28. Deventer K, Eenoo PV, Delbeke FT. Screening for anabolic steroids in doping analysis by liquid chromatography/electrospray ion trap mass spectrometry. Biomedical Chromatography. 2006;20:429-433
  29. 29. Van Eenoo P, Van Gansbeke W, De Brabanter N, Deventer K, Delbeke FT. A fast, comprehensive screening method for doping agents in urine by gas chromatography–triple quadrupole mass spectrometry. Journal of Chromatography A. 2011;1218:3306-3316
  30. 30. Revelsky AI, Samokhin AS, Virus ED, Rodchenkov GM, Revelsky IA. High sensitive analysis of steroids in doping control using gas chromatography/time-of-flight mass-spectrometry. Drug Testing and Analysis. 2011;3:263-267
  31. 31. Yamada M, Aramaki S, Kurosawa M, Kijima-Suda I, Saito K, Nakazawa H. Simultaneous doping analysis of main urinary metabolites of anabolic steroids in horse by ion-trap gas chromatography-tandem mass spectrometry. Analytical Sciences. 2008;24:1199-1204
  32. 32. Scarth JP, Clarke A, Teale P, Mill A, Macarthur R, Kay J. Detection of endogenous steroid abuse in cattle: Results from population studies in the UK. Food Additives & Contaminants: Part A. 2011;28:44-61
  33. 33. Scarth JP, Kay J, Teale P, Akre C, Le Bizec B, De Brabander HF, et al. A review of analytical strategies for the detection of ‘endogenous’ steroid abuse in food production. Drug Testing and Analysis. 2012;4:40-49
  34. 34. Mitrevski BS, Wilairat P, Marriott PJ. Comprehensive two-dimensional gas chromatography improves separation and identification of anabolic agents in doping control. Journal of Chromatography A. 2010;1217:127-135
  35. 35. Zhang Y, Tobias HJ, Auchus RJ, Brenna JT. Comprehensive 2-dimensional gas chromatography fast quadrupole mass spectrometry (GC × GC-qMS) for urinary steroid profiling: Mass spectral characteristics with chemical ionization. Drug Testing and Analysis. 2011;3:857-867
  36. 36. Noppe H, Le Bizec B, Verheyden K, De Brabander HF. Novel analytical methods for the determination of steroid hormones in edible matrices. Analytica Chimica Acta. 2008;611:1-16
  37. 37. Jin Z, Daiya S, Kenttämaa HI. Characterization of nonpolar lipids and selected steroids by using laser-induced acoustic desorption/chemical ionization, atmospheric pressure chemical ionization, and electrospray ionization mass spectrometry. International Journal of Mass Spectrometry. 2011;301:234-239
  38. 38. Kuuranne T, Vahermo M, Leinonen A, Kostiainen R. Electrospray and atmospheric pressure chemical ionization tandem mass spectrometric behavior of eight anabolic steroid glucuronides. Journal of the American Society for Mass Spectrometry. 2000;11:722-730
  39. 39. Greig MJ, Bolaños B, Quenzer T, Bylund JMR. Fourier transform ion cyclotron resonance mass spectrometry using atmospheric pressure photoionization for high-resolution analyses of corticosteroids. Rapid Communications in Mass Spectrometry. 2003;17:2763-2768
  40. 40. Liu Y, Uboh CE, Soma LR, Li X, Guan F, You Y, et al. Detection and confirmation of 60 anabolic and androgenic steroids in equine plasma by liquid chromatography-tandem mass spectrometry with instant library searching. Drug Testing and Analysis. 2011;3:54-67
  41. 41. Wong CHF, Leung DKK, Tang FPW, Wong JKY, Yu NH, Wan TSM. Rapid screening of anabolic steroids in horse urine with ultra-high-performance liquid chromatography/tandem mass spectrometry after chemical derivatisation. Journal of Chromatography A. 2012;1232:257-265
  42. 42. Yang Y, Shao B, Zhang J, Wu Y, Duan H. Determination of the residues of 50 anabolic hormones in muscle, milk and liver by very-high-pressure liquid chromatography-electrospray ionization tandem mass spectrometry. Journal of Chromatography B. 2009;877:489-496
  43. 43. Destrez B, Bichon E, Rambaud L, Courant F, Monteau F, Pinel G, et al. Criteria to distinguish between natural situations and illegal use of boldenone, boldenone esters and boldione in cattle: 2. Direct measurement of 17β-boldenone sulpho-conjugate in calf urine by liquid chromatography–high resolution and tandem mass spectrometry. Steroids. 2008;74:803-808
  44. 44. Grace PB, Drake EC, Teale P, Houghton E. Quantification of 19-nortestosterone sulphate and boldenone sulphate in urine from male horses using liquid chromatography/tandem mass spectrometry. Rapid Communications in Mass Spectrometry. 2008;22:2999-3007
  45. 45. Mazzarino M, Botrè F. A fast liquid chromatographic/mass spectrometric screening method for the simultaneous detection of synthetic glucocorticoids, some stimulants, anti-oestrogen drugs and synthetic anabolic steroids. Rapid Communications in Mass Spectrometry. 2006;20:3465-3476
  46. 46. Nicholson JK, Holmes E, Elliott P. The metabolome-wide association study: A new look at human disease risk factors. Journal of Proteome Research. 2008;7:3637-3638
  47. 47. Riedmaier I, Becker C, Pfaffl MW, Meyer HHD. The use of omic technologies for biomarker development to trace functions of anabolic agents. Journal of Chromatography A. 2009;1216:8192-8199
  48. 48. Bailly-Chouriberry L, Pinel G, Garcia P, Popot M-A, Le Bizec B, Bonnaire Y. Identification of recombinant equine growth hormone in horse plasma by LC−MS/MS: A confirmatory analysis in doping control. Analytical Chemistry. 2008;80:8340-8347
  49. 49. Riedmaier I, Tichopad A, Reiter M, Pfaffl MW, Meyer HHD. Identification of potential gene expression biomarkers for the surveillance of anabolic agents in bovine blood cells. Analytica Chimica Acta. 2009;638:106-113
  50. 50. Riedmaier I, Pfaffl MW. Transcriptional biomarkers—High throughput screening, quantitative verification, and bioinformatical validation methods. Methods. 2013;59:3-9
  51. 51. Cunningham MJ. Genomics and proteomics: The new millennium of drug discovery and development. Journal of Pharmacological end Toxicological Methods. 2000;44:291-300
  52. 52. Mooney MH, Bergwerff AA, van Meeuwen JA, Luppa PB, Elliott CT. Biosensor-based detection of reduced sex hormone-binding globulin binding capacities in response to growth-promoter administrations. Analytica Chimica Acta. 2009;637:235-240
  53. 53. Cacciatore G, Eisenberg SWF, Situ C, Mooney MH, Delahaut P, Klarenbeek S, et al. Effect of growth-promoting 17β-estradiol, 19-nortestosterone and dexamethasone on circulating levels of nine potential biomarker candidates in veal calves. Analytica Chimica Acta. 2009;637:351-359
  54. 54. Nagata S, Kurosawa M, Mima K, Nambo Y, Fujii Y, Watanabe G, et al. Effects of anabolic steroid (19-nortestosterone) on the secretion of testicular hormones in the stallion. Journal of Reproduction and Fertility. 1999;115:373-379
  55. 55. Fiehn O. Metabolomics—The link between genotypes and phenotypes. Plant Molecular Biology. 2002;48:155-171
  56. 56. Fiehn O. Extending the breadth of metabolite profiling by gas chromatography coupled to mass spectrometry. TrAC Trends in Analytical Chemistry. 2008;27:261-269
  57. 57. Cloteau C, Dervilly G, Kaabia Z, Bagilet F, Delcourt V, Loup B, et al. From a non-targeted metabolomics approach to a targeted biomarkers strategy to highlight testosterone abuse in equine. Illustration of a methodological transfer between platforms and laboratories. Drug Testing and Analysis. 2022;14(5):864-878
  58. 58. Courant F, Pinel G, Bichon E, Monteau F, Antignac J-P, Le Bizec B. Development of a metabolomic approach based on liquid chromatography-high resolution mass spectrometry to screen for clenbuterol abuse in calves. Analyst. 2009;134:1637-1646
  59. 59. Anizan S, Bichon E, Duval T, Monteau F, Cesbron N, Antignac J-P, et al. Gas chromatography coupled to mass spectrometry-based metabolomic to screen for anabolic practices in cattle: Identification of 5α-androst-2-en-17-one as new biomarker of 4-androstenedione misuse. Journal of Mass Spectrometry. 2012;47:131-140
  60. 60. Kaabia Z, Laparre J, Dervilly-Pinel G, Mooney M, Buckley T, Sherry M, et al. Comprehensive steroid profiling by liquid chromatography coupled to high resolution mass spectrometry. The Journal of Steroid Biochemistry and Molecular Biology. 2018;183:106-115
  61. 61. Rijk JCW, Lommen A, Essers ML, Groot MJ, Van Hende JM, Doeswijk TG, et al. Metabolomics approach to anabolic steroid urine profiling of bovines treated with prohormones. Analytical Chemistry. 2009;81:6879-6888
  62. 62. Thevis M, Bommerich U, Opfermann G, Schänzer W. Characterization of chemically modified steroids for doping control purposes by electrospray ionization tandem mass spectrometry. Journal of Mass Spectrometry. 2005;40:494-502
  63. 63. Anizan S, Di Nardo D, Bichon E, Monteau F, Cesbron N, Antignac J-P, et al. Targeted phase II metabolites profiling as new screening strategy to investigate natural steroid abuse in animal breeding. Analytica Chimica Acta. 2011;700:105-113
  64. 64. Van Renterghem P, Van Eenoo P, Sottas P-E, Saugy M, Delbeke F. Subject-based steroid profiling and the determination of novel biomarkers for DHT and DHEA misuse in sports. Drug Testing and Analysis. 2010;2:582-588
  65. 65. Regal P, Nebot C, Díaz-Bao M, Barreiro R, Cepeda A, Fente C. Disturbance in sex-steroid serum profiles of cattle in response to exogenous estradiol: A screening approach to detect forbidden treatments. Steroids. 2011;76:365-375
  66. 66. Kaabia Z, Dervilly-Pinel G, Popot MA, Bailly-Chouriberry L, Plou P, Bonnaire Y, et al. Monitoring the endogenous steroid profile disruption in urine and blood upon nandrolone administration: An efficient and innovative strategy to screen for nandrolone abuse in entire male horses. Drug Testing and Analysis. 2014;6:376-388
  67. 67. Moeller B, Brown S, Pierce S, Van Balen C, Stanley S. Endogenous steroid profiling of young thoroughbred horses in athletic training. In: Proceedings of the 55th Annual Convention of the American Association of Equine Practitioners; December 5-9, 2009; Las Vegas, Nevada. Vol. 55. Nevada, USA: American Association of Equine Practitioners; 2009. p. 317
  68. 68. Hochberg RB. Biological esterification of steroids. Endocrinology Reviews. 1998;19:331-348
  69. 69. Badeau M, Vihma V, Mikkola TS, Tiitinen A, Tikkanen MJ. Estradiol fatty acid esters in adipose tissue and serum of pregnant and pre- and postmenopausal women. The Journal of Clinical Endocrinology and Metabolism. 2007;92:4327-4331
  70. 70. Nielen MWF, Lasaroms JJP, Mulder PPJ, Van Hende J, van Rhijn JA, Groot MJ. Multi residue screening of intact testosterone esters and boldenone undecylenate in bovine hair using liquid chromatography electrospray tandem mass spectrometry. Journal of Chromatography B. 2006;830:126-134
  71. 71. Kaabia Z, Dervilly-Pinel G, Hanganu F, Cesbron N, Bichon E, Popot MA, et al. Ultra high performance liquid chromatography/tandem mass spectrometry based identification of steroid esters in serum and plasma: An efficient strategy to detect natural steroids abuse in breeding and racing animals. Journal of Chromatography A. 2013;1284:126-140
  72. 72. Burton R, Westphal U. Steroid hormone-binding proteins in blood plasma. Metabolism. 1972;21:253-276
  73. 73. Dunn J, Nisula B, Rodbard D. Transport of steroid hormones: Binding of 21 endogenous steroids to both testosterone-binding globulin and corticosteroid-binding globulin in human plasma. The Journal of Clinical Endocrinology and Metabolism. 1981;53:58-68
  74. 74. Bischoff F, Yee YS, Moran JJ, Katherman RE. Enzymatic hydrolysis of steroid hormone esters by blood serum. The Journal of Biological Chemistry. 1951;189:729-738
  75. 75. Shackleton CHL, Chuang H, Kim J, de la Torre X, Segura J. Electrospray mass spectrometry of testosterone esters: Potential for use in doping control. Steroids. 1997;62:523-529
  76. 76. Kirk JM, Tarbin J, Keely BJ. Analysis of androgenic steroid Girard P hydrazones using multistage tandem mass spectrometry. Rapid Communications in Mass Spectrometry. 2006;20:1247-1252
  77. 77. Gray BP, Teale P, Pearce CM. Analysis of methyloxime derivatives of intact esters of testosterone and boldenone in equine plasma using ultra high performance liquid chromatography tandem mass spectrometry. Drug Testing and Analysis. 2011;3:206-213
  78. 78. Southan G, Mallat A, Jumeau J, Craig S, Poojara N, Mitchell D. Programme and Abstracts of the Second International Symposium on Applied Mass Spectrometry in the Health Sciences. Barcelona: Biomedical & Environmental Mass Spectrometry; 1990. p. 360
  79. 79. Becchi M, Aguilera R, Farizon Y, Flament MM, Casabianca H, James P. Gas chromatography/combustion/isotope-ratio mass spectrometry analysis of urinary steroids to detect misuse of testosterone in sport. Rapid Communication in Mass Spectrometry. 1994;8:304-308
  80. 80. Aguilera R, Becchi M, Casabianca H, Hatton CK, Catlin DH, Starcevic B, et al. Improved method of detection of testosterone abuse by gas hromatography/combustion/isotope ratio mass spectrometry analysis of urinary steroids. Journal of Mass Spectrometry. 1996;31:169-176
  81. 81. Aguilera R, Becchi M, Mateus L, Popot MA, Bonnaire Y, Casabianca H, et al. Detection of exogenous hydrocortisone in horse urine by gas chromatography–combustion–carbon isotope ratio mass spectrometry. Journal of Chromatography B: Biomedical Sciences and Applications. 1997;702:85-91
  82. 82. Ferchaud V, Le Bizec B, Monteau F, Andre F. Determination of the exogenous character of testosterone in bovine urine by gas chromatography-combustion-isotope ratio mass spectrometry[dagger]. Analyst. 1998;123:2617-2620
  83. 83. Ferchaud V, Le Bizec B, Monteau F, Andre F. Characterization of exogenous testosterone in livestock by gas chromatography/combustion/isotope ratio mass spectrometry: Influence of feeding and age. Rapid Communications in Mass Spectrometry. 2000;14:652-656
  84. 84. Saudan C, Kamber M, Barbati G, Robinson N, Desmarchelier A, Mangin P, et al. Longitudinal profiling of urinary steroids by gas chromatography/combustion/isotope ratio mass spectrometry: Diet change may result in carbon isotopic variations. Journal of Chromatography B. 2006;831:324-327
  85. 85. Cannizzo FT, Pegolo S, Starvaggi Cucuzza L, Bargelloni L, Divari S, Franch R, et al. Gene expression profiling of thymus in beef cattle treated with prednisolone. Research in Veterinary Science. 2013;95:540-547
  86. 86. Pozzi R, Fernandes K, Moura C, Ferrari R, Fernandes K, Renno A, et al. Nandrolone decanoate induces genetic damage in multiple organs of rats. Archives of Environmental Contamination and Toxicology. 2013;64:514-518
  87. 87. Malin B, Sweeney L. Inferring genotype from clinical phenotype through a knowledge based algorithm. Pacific Symposium on Biocomputing. 2002:41-52
  88. 88. Prank K, Schulze E, Eckert O, Nattkemper TW, Bettendorf M, Maser-Gluth C, et al. Machine learning approaches for phenotype–genotype mapping: Predicting heterozygous mutations in the CYP21B gene from steroid profiles. European Journal of Endocrinology. 2005;153:301-305
  89. 89. Schulze J, Rane A, Ekstrom L. Genetic variation in androgen disposition: Implications in clinical medicine including testosterone abuse. Expert Opinion on Drug Metabolism & Toxicology. 2009;5:731-744

Written By

Zied Kaabia

Submitted: 17 September 2022 Reviewed: 22 November 2022 Published: 30 August 2023