The Vis 2014

Embed Size (px)

Citation preview

  • 8/18/2019 The Vis 2014

    1/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

     Journal of Pharmaceutical and Biomedical Analysis xxx (2014) xxx–xxx

    Contents lists available at ScienceDirect

     Journal of Pharmaceutical and Biomedical Analysis

     journa l homepage: www.elsevier .com/ locate / jpba

    Review

    Analytical approaches for the detection of emerging therapeutics andnon-approved drugs in human doping controls

    Mario Thevis a,∗, Wilhelm Schänzerb

    a Center for Preventive DopingResearch – Institute of Biochemistry, GermanSport University Cologne, Am SportparkMüngersdorf 6,

    50933 Cologne, Germanyb EuropeanMonitoringCenter for Emerging Doping Agents, Cologne/Bonn, Germany

    a r t i c l e i n f o

     Article history:Received 3 March 2014

    Received in revised form 5 May 2014

    Accepted 6 May 2014

    Available online xxx

    Keywords:

    Doping

    Sport

    Mass spectrometry

    Emerging drugs

    Stamulumab

    Anti-myostatin antibody

    a b s t r a c t

    The number and diversity of  potentially performance-enhancing substances is continuously growing,

    fueled by new pharmaceutical developments but also by the inventiveness and, at the same time,

    unscrupulousness of black-market (designer) drug producers and providers. In terms of sports drug test-

    ing, this situation necessitates reactive as well as proactive research and expansion of  the analytical

    armamentarium to ensure timely, adequate, and comprehensive doping controls. This review summa-

    rizes literature published over the past 5 years on new drug entities, discontinued therapeutics, and

    ‘tailored’ compounds classified as doping agents according to the regulations of the World Anti-Doping

    Agency, with particular attention to analytical strategies enabling their detection in human blood or urine.

    Among these compounds, low- and high-molecular mass substances of peptidic (e.g.modified insulin-like

    growth factor-1, TB-500, hematide/peginesatide, growth hormone releasing peptides, AOD-9604, etc.)

    and non-peptidic(selective androgen receptor modulators, hypoxia-inducible factor stabilizers, siRNA, S-

    107and ARM036/aladorian,etc.) as well as inorganic (cobalt) nature are considered and discussed in terms

    of specific requirements originating from physicochemical properties, concentration levels, metabolism,

    and their amenability for chromatographic-mass spectrometric or alternative detection methods.

    © 2014 Elsevier B.V. All rights reserved.

    1. Introduction

    With the constantly increasing knowledge about biochemi-

    cal mechanisms at cellular and molecular levels, more and more

    optionsfor pharmacologicalinterventions havebeen identified that

    suggest newpaths to desired therapiespotentiallyallowing curefor

    severe if not fatal diseases. The flipside of such research is the mis-

    use potentialoffered by a subsetof newdrug candidates, especially

    those that promote muscle growth, stimulate erythrocyte produc-

    tion, or enhance physical stamina and athletic performance via

    other routes [1]. Such drug candidates have been offered and soldvia Internet-based providers for years, despite the lack of clinical

    approval and, in some cases, discontinuation of their development

    dueto severeside effects. Thetargeted ‘clientele’ of such offerings is

    composed of recreational as well as professional athletes, with the

    latter ones being at risk of violating regulations established by the

    ∗ Corresponding author at: Institute of Biochemistry – Center for Preventive

    Doping Research, German Sport University Cologne, Am Sportpark Müngersdorf 6,

    50933 Cologne, Germany. Tel.: +49 221 4982 7070; fax: +49 221 4982 7071.

    E-mail address: [email protected] (M. Thevis).

    World Anti-Doping Agency (WADA) [2] These regulations as pre-

    sented in WADA’s Prohibited List include a category of substances

    dedicated to particularly such compounds, i.e. ‘non-approved for

    human use’/discontinued drug candidates, referred to as S0. In

    order to enable comprehensive doping controls, accredited labo-

    ratories update, expand, and improve their portfolio of analytical

    assays, mostof which relyon chromatographic-mass spectrometric

    approaches[3,4]; however,the implementation of newcompounds

    into sports drug testing protocols requires a substantial amount

    of information including therapeutic dosage, pharmacokinetics,

    metabolism, and elimination. Moreover, specific physicochemi-

    cal properties might necessitate dedicated sample collection and

    transport conditions, sample preparation or analytical procedures

    to ensure the required sensitivity and specificity to detect the tar-

    get analyte with appropriate limits of detection (LODs) [5] With

    the constraints in budget, time, sample volume(s), laboratory staff 

    and instrumentation, sports drug testing laboratories however are

    urged to combine as many detection assays as possible without

    compromising the necessary analytical requirements, preferably

    by using and expanding existing analytical approaches. Hence, test

    menus need to be rationally arranged and their fitness-for-purpose

    as appropriate initial testing procedure has to be demonstrated.

    http://dx.doi.org/10.1016/j.jpba.2014.05.020

    0731-7085/© 2014 Elsevier B.V. All rightsreserved.

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://www.sciencedirect.com/science/journal/07317085http://www.elsevier.com/locate/jpbamailto:[email protected]://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020mailto:[email protected]://www.elsevier.com/locate/jpbahttp://www.sciencedirect.com/science/journal/07317085http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    2/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    2 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

    While formerly drug classes dictated the composition of analyti-

    cal assays, nowadaysthe available analytical equipmentcommonly

    governs the employed test strategies [3]. To date, routine doping

    control matrices are urine, serum and blood, occasionally comple-

    mented by alternative matrices such as hair potentially providing

    supporting evidence. The collection protocols follow stringent reg-

    ulationsand require trained doping control officers/phlebotomists;

    transport times and conditions have to be controlled and docu-

    mentedespeciallyincaseofbloodsamplesfortheAthleteBiological

    Passport (ABP), where also time limits for transport and analy-

    sis apply. In addition, sample storage (urine and serum) has to be

    ensured for up to 10 years to allow for re-testing if requested.

    In the present review, literature published between 2009 and

    2013 concerning emerging, ‘designer’, and discontinued drugs is

    discussed in the context of human doping controls. Challenges

    arising from structural feature of substances are presented and

    metabolite identification and detection strategies are outlined for

    a representative selection of compounds covering low- and high

    molecular mass analytes of non-peptidic, peptidic, and ribonucleic

    acid composition.

    2. Compounds affecting skeletal muscle performance

    Due to the substantial number of compounds with evident or

    presumed impact on skeletal muscle physiology and/or perfor-

    mance, the substances considered in the followingare divided into

    the categories of low and high molecular mass products.

     2.1. Lowmolecular mass substances

     2.1.1. Ryanodine receptor-calstabin-complex stabilizers (Rycals)

    Studies on cardiac arrhythmia as well as sarcopenia (as defined

    as the age-related loss of muscle mass, force, and exercise capac-

    ity) and muscular dystrophy have revealed the relevance of 

    the ryanodine receptor 1 (RyR1) and its Ca2+-channel complex

    building partner molecule calstabin-1 (FK506 binding protein 12,

    FKBP12) with regard to normal skeletal and cardiac muscle func-tion. Substantial research on mechanisms of post-translational

    modifications has been conducted in animal models and, more

    recently, also in humans indicating particularly S-nitrosylation and

    (hyper)phosphorylation of RyR1 as main factors of the aging-,

    disease-, or exercise-induced functional impairment of myocytes

    [6–8]. A potential therapy is based on benzothiazepine-derived

    drug candidates such as the first- and second-generation thera-

    peutics JTV-519 and S107 (Fig. 1a, 1 and 2) [9], which have been

    shown to reduce muscle fatigue and improve exercise capacity in

    laboratory rodents by restoring the RyR1-FKBP12 complex. Conse-

    quently, the relevance of such compounds for sports drug testing

    was recognized and detection assays for the intact drugs and/or in

    vitro generated metabolites in blood and urine were established.

    The mass spectrometric behavior of JTV-519 and S-107 wasstudied in extensousing electrospray ionization (ESI) and collision-

    induced dissociation (CID) [10] as well as electron ionization (EI)

    [11] employing high resolution/high accuracy mass spectrome-

    try, stable isotope labeling and, in case of ESI-CID, H/D-exchange

    experiments. By means of the obtained information, test meth-

    ods for urine [10,11] and plasma [12] were developed enabling

    the detection of the intact molecules at LODs of 0.1–6ng/ml. In

    case of blood plasma, peak concentrations of the drug candidates

    after therapeutic dosing were expected at approximately 40 ng/ml,

    whichwas well withinthe detection windowof the developed test

    method. In the absence of data on the metabolism and(renal)elim-

    ination of the benzothiazepines, urine samples were subjected to

    enzymatic hydrolysis followed by liquid–liquid extraction (LLE) of 

    the target analytes and subsequent detection by means of liquid

    chromatography–(tandem) mass spectrometry (LC–MS/MS) or gas

    chromatography–mass spectrometry (GC–MS). In order to further

    complement the analytical approach with putative metabolites,

    phase-I and phase-II metabolic reactions were simulated for S-

    107 in vitro, yielding predominantly N- and S-oxygenated species

    as well as N- and O-demethylated metabolites. Moreover, glu-

    curonic acid conjugates of the intact drug and its O-demethylated

    phase-I metabolic product were identified representing viable tar-

    gets for future doping controls [13]. Moreover, the development of 

    next-generation benzothiazepine-derived compounds needs fur-

    ther consideration, e.g . with regardto thephase-II clinical trial drug

    candidate referred to as ARM036 (Aladorian, Fig. 1a, 3) [14], the

    product ion mass spectrum of which is presented in Fig. 1b.

     2.1.2. Selective androgen receptor modulators (SARMs)

    Non-steroidal selective androgen receptor modulators (SARMs)

    have been subject of extensive preclinical and clinical trials since

    the first-in-class compounds were identified in 1998, predom-

    inantly aiming at the treatment and prevention of sarcopenia,

    osteoporosis, and disease-related losses of skeletal muscle mass,

    strength, and function [15,16]. Moreover, the potential utility of 

    SARMs in cardiology has been discussed [17], and the substan-

    tial interest in new drug entities with SARM-like properties is

    still on the incline according to recent reviews [16,18] and pub-lications on advances in SARM-related research [19–21]. With

    the increasing amount of possible non-steroidal and steroidal

    SARM drug candidates, examples of which are illustrated in Fig. 2

    (4–13), the portfolio of compounds potentially misused in sports

    is expanded accordingly [22,23] and detection assays plus ample

    information on metabolism and elimination are vital for appro-

    priate doping controls. Consequently, studies focusing on the

    metabolism of SARMs and possibilities to detect intact as well as

    metabolized SARMs have been initiated and continued, and the

    relevance and necessity of adequate test methods was demon-

    strated with the first adverse analytical findings (AAFs) for SARMs

    in 2010 and the following years [24,25]. The analytical assays

    for SARMs have been established for plasma [12,26], dried blood

    spots (DBS) [27], and urine targeting either the intact substances(plasma and DBS) or main metabolites (urine) as identified and

    characterized in in vitro [28] and in vivo studies [29–31]. Despite

    modest structural similarities between some SARMs comprising

    e.g. a 4-substituted aniline moiety, a substantial heterogeneity of 

    pharmacophores is present in currentlyinvestigated SARMsinclud-

    ing (amongst others) arylpropionamide, quinolinone, tropanol,

    tetrahydroquinoline, hydantoin, thiophene, phenyl-oxadiazol, and

    steroid derivatives (Fig. 2, Table 1). Hence, various projects have

    been required providing insights into main metabolic pathways

    and the mass spectrometric behavior of identified and character-

    ized target compounds.

    All SARMs recently studied in a doping control context demon-

    strated good or excellent ionization properties using electrospray,

    thus supporting the sensitive detection of these compounds andrelated metabolic products in sports drug test samples employ-

    ing LC–MS/MS-based strategies [3,32]. Arylpropionamide-derived

    SARMs were among the first category of emerging anabolic

    agents investigated with ESI-MS/MS, EI-MS(/MS), and under

    in vitro and in vivo metabolism conditions. Substantial agreement

    between results of in vitro and in vivo studies was observed, and

    post-administration study urine samples of the arylpropionamide-

    derived SARMs S-4 and S-22 (Fig. 2, 4 and 5, respectively)

    predominantly yielded the glucuronic acid conjugates of the intact

    drugs andcorrespondingmono-hydroxylated metabolites as viable

    analytesfor routine doping controls[29,30] withLODs forthe intact

    drug candidates found below 1 ng/ml. Complementary, LODs of 

    0.05–20ng/ml [27,33] and 10ng/ml [26] were determined in DBS

    and plasma, respectively, for the intact therapeutics. Substance

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    3/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx 3

    Fig. 1. (a) Structures of JTV-519 (1), S -107 (2), and Aladorian (ARM036, 3); (b) product ion mass spectrum of the protonated molecule [M+H]+ at m/z 268 of Aladorian,

    recorded at a collisionenergy of 25eV.

    characterization by mass spectrometric techniques, particularly

    LC–MS/MS employing high resolution/high accuracy mass spec-

    trometry, was further conducted for the related arylpropionamides

    S-1, S-9, S-23, and S-24 [34], all of which were also subjected

    to systems simulating metabolic reactions such as human liver

    microsomal [28] or fungal [35] preparations to provide reference

    material for (provisional) targets for sports drug testing. Simi-

    larly, investigations into the mass spectrometry of SARMs and

    their detection in human urine were conducted for quinolinone-

    (e.g. LGD-2226, Fig. 2, 6), tetrahydroquinoline- (e.g. S-40503, Fig. 2,

    8), and hydantoin-derived substances (e.g. BMS564929, Fig. 2, 10)

    [34], complemented by more recent studies on phenyl-oxadiazol-

    (RAD140, Fig. 2, 11) and tropanol-based SARMs (ACP-105, Fig. 2,

    12) [36]. The elimination of ACP-105 was further studied in

    a rat model, demonstrating the production of various different

    mono- and bishydroxylated metabolites serving as preferred target

    Fig. 2. Structures of S-4 (Andarine, 4), S-22 (Enobosarm, 5), LGD-2226 (6) , LG 121071 (7), S-40503 (8), S-101479 (9), BMS-564929 (10), RAD140 (11), ACP-105 (12), and

    LGD-4033 (13).

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    4/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    4 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

     Table 1

    Structure characteristics of selected SARMs.

    No. (Fig. 2) SARM Pharmacophore Elemental composition Molecular mass (Da) Refs.

    S-1 Arylpropionamide C17 H14F4N2O5   402.0839 [165]

    4 S-4 (Andarine) Arylpropionamide C19 H18F3N3O6   441.1148 [18]

    S-9 Arylpropionamide C17 H14ClF3N2 O5   418.0543 [165]

    5 S-22 (Ostarine) Arylpropionamide C19 H14F3N3O3   389.0987 [18]

    S-23 Arylpropionamide C18 H13ClF4N2 O3   416.0551 [165]

    S-24 Arylpropionamide C18 H14F4N2O3   382.0941 [165]

    6 LGD-2226 Quinolinone C14 H9F9N2O 392.0571 [18]LGD-2941 Quinolinone C17 H16F6N2O2   394.1116 [18]

    LGD-3303 Quinolinone C16 H14ClF3N2 O 342.0747 [18]

    7 LG-121071 Quinolinone C15 H15F3N2O 296.1136 [166]

    8 S-40503 Tetrahydroquinoline C15 H23N3 O3   293.1739 [18]

    S-49288 Tetrahydroquinoline C25 H26N4 O 398.2107 [20]

    9 S-101479 Tetrahydroquinoline C26 H24F2N4O3   478.1816 [16]

     JNJ-28330835 Phenyl-pyrazol-carboxyamide C14 H10F6N4O 364.0759 [18]

    10 BMS-564929 Hydantoin C14 H12ClN3O3   305.0567 [18]

     JNJ-37654032 Benzoimidazole C11 H7Cl2 F3N2O 309.9888 [18]

    11 RAD140 Phenyl-oxadiazole C20H16ClN5O2   393.0993 [18]

    12 ACP-105 Tropanol C16 H19ClN2O 290.1186 [18]

    AC-262536 Tropanol C18 H18N2 O 278.1419 [18]

    13 LGD-4033 Pyrrolidinyl-benzonitrilea C14 H12F6N2O 338.0854

    RAD35010 Indole C13 H11ClF3NO 289.0481 [18]

    Ly2452473 Indole C23 H23N3 O2   373.1790 [16]

    FTBU-1 Benzoimidazole C19 H16FN5OS 381.1060 [18]

    2-FPA Pyridinylmethanamide C17 H19FN2O 286.1481 [18]

    GLPG0634 Diarylimidazolidinedione C19 H14F3N3O3   389.0987 [16]

    MK-3984 Phenylmethanamide C17 H12F7NO2   395.0756 [16]

    NEP28 Thiophene C10H10BrF3N2S 325.9700 [21]

    MK-0773 Steroidal C27 H34FN5O2   479.2697 [18]

    Cl-4-AS-1 Steroidal C26 H33ClN2O2   440.2231 [18]

    TFM-4AS-1 Steroidal C27 H33F3N2O2   474.2494 [18]

    YK11 Steroidal C25 H34O6   430.2355 [18]

    S-42 Steroidal C21 H28O 296.2140 [18]

    a Unconfirmed.

    analytes as the administered compound was detected intact only

    up to 24 h [31]. In contrast, human in vitro and in vivo studies

    with RAD140 suggested the use of the administered SARM as uri-

    nary target compound for doping control purposes as only modest

    metabolic reactions to a monohydroxylated analog were observedand RAD140 was detected in post-administration study urine sam-

    ples up to 8 days [37]. While recent reviews on SARMs in clinical

    development have referred to the structure of the drug candidate

    LGD-4033 as undisclosed [16], Internet-based suppliers of SARMs

    have assigned 4-(2-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-

    1-yl)-2-(trifluoromethyl)-benzonitrile (Fig. 2, 13) to it. Despite the

    absence of confirmation, the drug entity is part of LIGAND PHAR-

    MACEUTICAL’s patents on compounds with SARM-like properties

    [38] and thus also a candidate for doping controls. Hence, detec-tion assays are required particular in the light of its arguably illicit

    availability and information on the mass spectrometric properties

    of the substance and its metabolite(s) are needed to complement

    routine sports drug testing (Fig. 3).

    Fig. 3. Production mass spectrum of thedeprotonated molecule [M−H]−

    at m/z 337 of LGD-4033, recorded at a collision energy of 15eV.

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    5/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx 5

     2.1.3. Sirtuin-1 activators

    Sirtuins (1–7) represent a family of NAD+-dependent histone

    deacetylase enzymes, with sirtuin-1 (SIRT1) being a key regulator

    in the deacetylation of metabolism-modulating protein substrates

    suchas forkhead box proteins (FOXO) and peroxisomeproliferator-

    activated receptor coactivator 1 (PGC-1) [39]. Since thediscovery of resveratrol’s SIRT1-activating effect with significant

    impact on skeletal muscle mitochondrial content, extended life

    span and anti-aging as well as antidiabetogenic properties in ani-

    mal models, much effort was invested into research concerning

    synthetic SIRT1-activating drug entities (STACs) [40,41]. Despite an

    extensive debate concerning the underlying mechanism(s) respon-

    sible for the observed beneficial effects [42–46], a series of STACs

    lead drug candidates such as SRT1720, SRT1460, and SRT2104

    (Fig. 4, 14 and 15) was produced and has been subject of advanced

    clinical trials since. The arguably comparable outcome of STAC

    administration with other metabolic modulators such as e.g . AICAR 

    (vide infra) in terms of pharmacologically enhanced endurance has

    triggered concerns as to the misuse potential of these substances.

    Consequently, analytical assays have been established for first-

    generation STACs and additional model compounds in blood and

    urine. Moreover, invitroand invivobiotransformation studies were

    conducted to provide information on viable targets for extended

    detection windows in routine sports drug testing.Commercially available and synthesized STAC reference sub-

    stances including SRT1720 and 4 structurally related compounds

    were studied concerning their ESI-MS and MS/MS behavior and

    measured from human plasma [47]. A total of 100l of specimenwas enriched with an eight-fold deuterated analog of SRT1720 and

    deuterated resveratrol (internal standards), and plasma proteins

    were precipitated by means of acetonitrile. The supernatant was

    analyzed by LC–ESI-MS/MS in multiple reaction monitoring (MRM)

    mode, allowing for detection limits between 0.1 and 1ng/ml.

    Considering the reported plasma concentrations of STACsin clinical

    trials reaching up to 390ng/ml, the accomplished LODs demon-

    strate the fitness-for-purpose of the presented approach. In order

    to complement the approach with analyses conducted from urine,

    target analytes were generated using in vitro biotransformationsystems [48]. Main metabolic reactions included N -oxidation and

    hydroxylation, and the resulting products were further observed in

    subsequent rat administration studies [49]. By means of the intact

    STACs, the test method was characterized indicating detection lim-

    its for the target analytes in urinary matrix of 0.5 ng/ml. It remains

    to be clarified whether the intact drugs or selected metabolites

    will be the most appropriate compounds for routine doping control

    analyses; however, the principle option to detect these compounds

    in blood/plasma and urine is established and serves the purpose of 

    proactive and preventive anti-doping research.

     2.1.4. AICAR (5-amino-4-imidazolecarboxamide ribonucleoside)

    Adenosine monophosphate (AMP)-activated protein kinase

    (AMPK) is an essential factorof mitochondrial biogenesis andfunc-tion [50]. Due to the key role of skeletal muscle mitochondria

    in health and disease as well as in exercise, numerous studies

    concerning the at least partially unclear mechanisms of mitochon-

    drial biogenesis have been conducted with the goal of identifying

    potential pharmacological means to cure genetically caused (e.g .

    Duchenne muscular dystrophy, DMD, or amyotrophic lateral scle-

    rosis, ALS) and non-genetic (e.g . obesity or metabolic syndrome)

    disorders [51]. The so-called master regulator of mitochondrial

    numbers is the earlier mentioned PGC-1, controlling the cell’s‘powerhouse’, whichis affected by stressors such as caloric restric-

    tion and exercise [52]. The activation of PGC-1 can be triggeredthroughgene expressionas wellas post-translationalmodifications

    including phosphorylation, which (among other factors) depends

    on the activity of AMPK, since only the phosphorylated PGC-1 is

    believed to be subsequently primed by SIRT1via deacetylation and

    capable of inducinggene expressionrequiredfor mitochondriogen-

    esis. This interconnection, the relevance of AMPK andits activation

    through potent agonists suchas 5-amino-4-imidazolecarboxamide

    ribonucleoside (AICAR, Fig. 4, 16) [53] have been the rationale to

    include this compound and related substances to WADA’s Pro-

    hibited List in 2009, first categorized as gene doping substance

    and later as metabolic modulator [2]. The administration of the

    natural and cell-permeable AMPK activator AICAR to laboratory

    rodents at 500mg/kg/day was shown to effectively activate the

    AMPK signaling pathway resulting in an improved endurance of 

    untrained mice by 23–44% [54]. This supportedthe necessityof the

    compound’s inclusion in doping control programs as well as the

    fact that the substance has been readily available as chemical and

    via Internet-based suppliers [23] and hassupposedly found its way

    into the world of sport meanwhile [55,56].

    Due to the natural occurrence of AICAR in human urine, pro-

    viding evidence for the illicit use of the non-approved drug

    candidate has required sophisticated analytical strategies similar

    to approaches employed for the detection of natural/endogenous

    steroid misuse. By means of liquid chromatography/isotope-

    dilution tandem mass spectrometry (LC-IDMS/MS), naturally

    occurring urinary concentrations of AICAR werequantified in a pop-

    ulation of 499 athletes, demonstrating a significant correlation of urinary AICAR levels and gender, type of sport (e.g. endurance and

    strength sport) and time point of sample collection (i.e. in/out of 

    competition) [57]. A mean value of approximately 2200 ng/ml was

    reported, and under consideration of another set of 500 urine sam-

    ples collected fromrecreationalathletes,a tentative thresholdlevel

    of 3500 ng/ml was suggested (unpublished results). However, the

    mere exceeding of urinary AICAR concentrations above reference

    values will not allow proving the abuse of synthetic AICAR, par-

    ticularly due to substantial intra- and inter-individual variations;

    such information will nevertheless enable and trigger further anal-

    yses supporting or disproving a possible AICAR administration and

    eventually revealing the source of the compound (endogenous or

    exogenous).

    An alternative matrix presumably conserving administeredAICAR for a prolonged period of time is the erythrocyte. Upon

    introduction into the blood stream, AICAR was shown to cross

    the red blood cell (RBC) membrane followed by its conversion

    into the 5-monophosphate derivative, which does not allow its

    efflux from the RBC and causes an accumulation of the produced

    AICAR-ribotide, arguably for the lifespan of the erythrocyte. Hence,

    measuring intra-erythrocytic AICAR-ribotide concentrations could

    provide complementary information as to whether unnaturally

    high levels prevail or not. A quantitative analytical assay also based

    on LC-IDMS/MS was established for AICAR-ribotide in RBCs, and

    99 recreational athletes’ samples were measured to obtain ranges

    of normal physiological values (10–500ng/ml) of the analyte [58].

    By means of  in vitro incubation reactions, the administration of 

    pharmacological doses of AICAR were simulated, demonstratingan increase of intra-erythrocytic AICAR-ribotide concentrations of 

    1–10g/ml providing proof-of-concept for this approach. Moredata for reference ranges and authentic administration study

    samples will be required to assess the significance of these num-

    bers; however, the intra-individual profile of AICAR concentrations

    might be a sensible contribution to the Athlete Biological Passport

    (ABP) to allow flagging an unusual blood parameter finding.

    Conclusive test results concerning the endogenous or exoge-

    nousorigin of a naturallyoccurring substance in an athlete’sdoping

    control sample is commonly obtained by isotope-ratio mass spec-

    trometry (IRMS) [59]. Thistechnical approachhas beensuccessfully

    employed in steroid analyses for over a decade in doping con-

    trols, and its utility for tackling the AICAR issue was suggested

    as early as 2010. Two major obstacles had to be managed for the

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    6/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    6 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

    Fig. 4. Structures of SRT1720 (14) , SRT1460 (15), AICAR (16), Rolipram (17), Roflumilast (18), Cilomilast (19), L-739943 (20), MK-0677 (Ibutamoren, 21), CP-424391

    (Capromorelin, 22), and SM-130686 (23).

    successful IRMS analysis of AICAR, namely the low volatility of 

    the analyte, which necessitates derivatization prior to introduc-

    ing the substance into the gas chromatography-combustion-IRMS

    (GC/C/IRMS) system, andthe isolationof AICAR from urine without

    isotopic fractionation. Both challenges have recently been solved,

    providing a validated assay allowing to differentiate synthetic and

    natural AICAR by means of their carbon isotope signature [60].

    The test method requires a volume of 3ml of urine and currently

    urine samples with AICAR levels higher than 1500ng/ml are rec-

    ommended to be subjected to GC/C/IRMS. Due to the considerable

    amounts of AICAR in urine, also LC-IRMS, which is commonly

    inferior to GC/C/IRMS in terms of analytical sensitivity, has been

    considered as a viable means of measuring the carbon isotope sig-

    nature; to date however, no methodology has been established or

    reported.

     2.1.5. Phosphodiesterase-4 (PDE4) inhibitors

    In continuation of the quest for therapeutics supporting

    the therapy of mitochondria-related disorders, the impact of 

    phosphodiesterase-4 (PDE4) inhibitors has recently been revis-

    ited. Phosphodiesterases comprise a family of 11 currently known

    members with specific characteristics and stimulating or inhibi-

    ting agents. PDE4, a major target in chronic obstructive pulmonary

    disease (COPD) treatment [61–63], catalyzes the conversion of 

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    7/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx 7

    3-5-cyclic adenosine monophosphate (cAMP) to 5-AMP. Its inhi-

    bition was shown to result in a cascade of consequences allowing

    to explain the beneficial effects of the unspecific PDE inhibitor

    resveratrol in animal test models [50,64] including, amongst oth-

    ers, increased mitochondrial biogenesis and function associated

    with improved fatutilizationand,last butnot least,enhanced exer-

    cise performance. The administration of the archetypical synthetic

    PDE4-inhibitor rolipram (Fig. 4, 17) to laboratory rodents yielded

    similar results, indicating that another master regulator of skeletal

    muscle mitochondriogenesis upstream of the above reported key

    factors (e.g . AMPK and PGC-1) has been identified. Consequently,approved drugs and drug candidates of this category inevitably

    move into the focus of sports drug testing and preventive doping

    research organizations, revealing a considerable number of at least

    50 candidates that have been mentioned as potential therapeu-

    tic agents with PDE4-inhibiting properties [65]; however, to date

    only one (roflumilast, Fig. 4, 18) has received clinical approval for

    the treatment of COPD [66,67]. Cilomilast (Fig. 4, 19) has advanced

    to phase-III clinical trials [68], and numerous additional new drug

    entities in pre-clinical or early clinical development have recently

    been summarized in a comprehensive review [65].

    Due to these arguments, the necessity for detection assays

    capable of screening for PDE4-inhibitors such as resveratrol,

    rolipram, roflumilast, and cilomilast was noticed and first analyt-ical approaches were recently presented [69]. Based on published

    DMPK data and in vitro incubation reactions, target analytes were

    selected and mass spectrometrically characterized, including the

    intact drugs as well as major metabolites. While roflumilast and

    cilomilast eachyieldedmainlyone reasonably abundant metabolite

    invitro (roflumilast-N -oxide andhydroxylatedcilomilast), rolipram

    was converted into six intense metabolic products resulting from

    hydroxylation and dealkylation reactions. Employing an estab-

    lished sample preparation protocol for urine specimens consisting

    of enzymatichydrolysisfollowed by LLEand subsequentLC–MS/MS

    analysis, LODs of 1–5 ng/ml for the compounds of interest were

    accomplished, and proof-of-concept data were achieved with a

    patient’s urine sample collected after therapeutic use of roflumi-

    last. Due to the substantial increase in performance as observedwith rodents on a treadmill test (time-to-exhaustion) and the evi-

    dently elevated number of mitochondria in skeletal muscle of the

    test animals resulting from PDE4-inhibitor administrations, mis-

    use of these substances cannot be excluded and at least inclusion

    in monitoring programs seems advisable.

     2.1.6. Peptidic and non-peptidyl growth hormone secretagogues

    (GHS)

    The multifaceted(and arguablyperformance-enhancing)effects

    of human growth hormone (hGH), primarily mediated via insulin-

    like growth factor-1 (IGF-1) to the skeletal muscle, have triggered

    substantial interest and comprehensive research programs with

    medicinal and therapeutic intention in the past. One of many

    remarkable outcomes has been the discovery of orally activegrowth hormone secretagogues (GHS) based on various different

    pharmacophores including e.g. benzolactame, 4-spiropiperidine,

    capromorelin, and oxindole derivatives such as L-739943, MK-

    0677, CP-424391, andSM-130686, respectively,which are depicted

    in Fig. 4 as selected examples of an enormous variety of potential

    drug candidates [70,71]. Since GHS stimulate the hGH secretion

    via an alternative route from the growth hormone releasing hor-

    mone (GHRH, vide infra) and arguably provide synergistic effects

    [72], their clinical development has been pursued complementary

    to conventional therapies indicated in hGH/IGF-1 axis-related dis-

    orders and age-related decline. Sustainedincreases in plasma IGF-1

    were reported in GHSadministrationstudies; however, none of the

    leaddrug candidates hasyet receivedclinical approval,possiblydue

    to limited benefits compared to hGH replacement therapies and

    reported weight gain due to appetite-stimulating effects of GHS.

    Nevertheless, compounds such as MK-0677 have been available

    through Internet-based suppliers, and detection methods for the

    heterogeneous class of non-peptidyl GHS are desirable in routine

    doping controls, preferably using commonly available methodolo-

    gies such as mass spectrometry (Fig. 5a). In addition to these orally

    available GHS, a considerable variety of growth hormone releas-

    ing peptides (GHRPs) acting through the same pathways on hGH

    secretion has been developed since the seminal works of Bowers

    and co-workers demonstrated the capability of short peptides to

    regulate hGH release in the early 1980s [73–75]. One representa-

    tive (Pralmorelin, GHRP-2) received clinical approval 2004in Japan

    as a diagnostic tool for GH deficiency in adults [76]. Whilst the

    majority of GHRPs, a selection of which is summarized in Table 2,

    does currentlynot holdapproval by anyhealth authority for human

    therapeutic use, numerous black-market sources have been iden-

    tified as suppliers of such agents [77,78], which underlines the

    importance of doping control assays allowing the sensitive and

    comprehensive analysis of GHS.

    A generic approach toward this issue was presented in 2012 by

    Pinyot et al., who employed a competitive receptor-binding assay

    [79,80]. Since all GHS share the commonality to bind to the GHS

    receptor 1a, the membrane-bound receptor is preincubated with a

    radiolabeled ligand (125I-ghrelin), which is displaced by (urinary)GHS in a dose-dependentmanneras demonstratedwith 7 synthetic

    GHS in a proof-of-concept study. The responses or minimal positive

    concentrations for GHS in urine varied from 1.5E−10M to1.0E−06

    with MK-0677 being most sensitively detected. The analysis of the

    growth hormone releasing peptide GHRP-2 in post-administration

    study urine samples was further presented, suggesting a detection

    window of approximately 4.5h for this particular drug. Confirma-

    tion of the presence of a prohibited substance belongingto the class

    of GHS was recommended and conducted viadedicated LC–MS/MS

    assays.

    Such a mass spectrometry-based methodology focusing par-

    ticularly on the detection of GHRP-2 and its main metabolite

    (d-Ala-d(-naphthyl)-Ala-Ala-OH) in human urine was published

    in2010 byOkanoet al. [81]. Here, an isotope-labeledGHRP-2 inter-nal standard was used to ensure appropriate sample preparation

    andanalysisconditions before the specimenwas subjectedto solid-

    phase extraction (SPE) and subsequent LC–MS/MS analysis. The

    assay enabled LODs of 20–50 pg/ml and was applied to an excre-

    tion study with ten volunteers who received an intravenous bolus

    of 100g of GHRP-2dihydrochloride. While theintact GHRP-2wasfoundup to13 h,the aforementionedmetabolitewas detectedup to

    24h using the established approach. Similarly, a screening method

    was established in 2011 targeting a family of 8 GHRPs (GHRP-1,

    -2, -4, -5, -6, alexamorelin, hexarelin, and ipamorelin) plus the

    earlier mentioned GHRP-2 metabolite using an isotope-dilution

    LC–MS approach [82]. Here, two deuterated internal standards (d4-

    GHRP-4 and a d3-GHRP-2 metabolite) were added and SPE was

    conducted with a weak cation exchange resin prior to microborereversed-phase LC separation and full scan high resolution/high

    accuracymass spectrometry,which enabled LODs of 0.2–0.5ng/ml.

    The applicability of the developed method to authentic urine sam-

    ples was tested with an elimination study with 10mg of GHRP-2

    orally administered to one male volunteer. The analyses revealed

    the absence of intact GHRP-2 and the traceability of the GHRP-2

    metabolite up to 20 h post-administration. In a follow-upstudy, the

    instrumental setupwas modified to consist of a nanoUHPLC system

    interfaced to a quadrupole-orbitrap mass analyzer [83]. As a result,

    the LODs were lowered approximately 20-fold to allow for the

    detectionof 2–10pg/ml of eachsubstance.Since DMPKdata of most

    GHRPs are not available but of great importance in terms of doping

    controls, animal invivo studies and invitrosimulations of metabolic

    reactions were conducted to identify and characterize viable target

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    8/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    8 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

    Fig. 5. (a) Product ion mass spectrum of the protonated molecule [M+H]+ at m/z 528of MK-0677, recorded at a collision energy of 25eV;(b) product ion mass spectrum of 

    the protonated molecule [M+H]+ at m/z 655 of GHRP-3, recorded at a collision energy of20 eV; (c) full scanMS spectrum of the intact anti-myostatin antibody Stamulumab,

    recorded at a resolution of 35,000 (FWHM).

    compounds for sports drug testing approaches [84]. Focusing on

    currently non-approved GHRPs (except for GHRP-2), seven com-

    pounds were administered to rats via oral and intravenous routes.

    These compounds (GHRP-1, -2, -4, -5, -6, alexamorelin, hexarelin,

    and ipamorelin) yielded at least 3 urinary metabolites each after

    i.v. application, which were confirmed by human in vitro simula-

    tions andwillextend the initial testing options forGHRPs in routine

    doping controls. A typical product ion mass spectrum of an intact

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    9/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx 9

     Table 2

    Primary structures of selected peptidic drug candidates. Peptides with 10 or less

    amino acid residues arepresented in 3-letter code, larger peptides in 1-letter code.

    Compound Amino acid sequence Molecular mass

    monoisotopic

    [Da]

    GHRP-1 Ala-His-d-Nal-Ala-Trp-d-Phe-Lys-NH2

    954.5

    GHRP-2 d-Ala-d-Nal-Ala-Trp-d-Phe-

    Lys-NH2

    817.4

    GHRP-3 Aib-d-Trp-d-Pro-d-Ile-Arg-

    NH2

    654.4

    GHRP-4 d-Trp-Ala-Trp-d-Phe-NH2   607.3

    GHRP-5 Tyr-d-Trp-Ala-Trp-d-Phe-NH2   770.4

    GHRP-6 His-d-Trp-Ala-Trp-d-Phe-Lys-

    NH2

    872.4

    Alexamorelin Ala-His-d-Mrp-Ala-Trp-d-Phe-

    Lys-NH2

    957.5

    Hexarelin His-d-Mrp-Ala-Trp-d-Phe-Lys-

    NH2

    886.5

    Ipamorelin Aib-His-d-2-Nal-d-Phe-Lys-

    NH2

    711.4

    GHRH YADAIFTNSY RKVLGQLSAR  

    KLLQDIMSRQ QGESNQERGA

    RARL 

    5037.6

    Sermorelin YADAIFTNSY RKVLGQLSAR  

    KLLQDIMSR-NH2

    3355.8

    CJC-1288 YADAIFTNSY RKVLGQLSAR  

    KLLQDIMSR K*-NH2

    3634.9

    CJC-1293 Y  dA DAIFTNSY RKVLGQLSAR 

    KLLQDIMSR K*-NH2

    3634.9

    CJC-1295 Y  dA DAIFTQ SY RKV A GQLSAR 

    KLLQDIL SR K*-NH2

    3437.9

    IGF-1 GPETLCGAEL VDALQFVCGD

    RGFYFNKPTG YGSSSRRAPQ 

    TGIVDECCFR SCDLRRLEMY 

    CAPLKPAKSA

    7643.6

    des(1-3)-IGF-1 TLCGAELVDA L QFVCGDRGF

    YFNKPTGYGS SSRRAPQTGI

    VDECCFRSCD LRRLEMYCAP

    LKPAKSA

    7360.5

    R 3-IGF-1 GPR TLCGAEL VDALQFVCGD

    RGFYFNKPTG YGSSSRRAPQ 

    TGIVDECCFR SCDLRRLEMY 

    CAPLKPAKSA

    7670.6

    long-R 3-IGF-1 MFPAMPLSSL FVNGPR TLCG

    AELVDALQFV CGDRGFYFNK

    PTGYGSSSRR APQTGIVDEC

    CFRSCDLRRL EMYCAPLKPAKSA

    9105.4

    MGF YQPPSTNKNT KSQRRKGSTF

    EERK-NH2

    2865.5

    ‘full length’ MGF GPETLCGAEL VDALQFVCGD

    RGFYFNKPTG YGSSSRRAPQ 

    TGIVDECCFR SCDLRRLEMY 

    CAPLKPAKSA RSVRAQRHTD

    MPKTQKYQPP STNKNTKSQR 

    RKGSTFEEH

    12264.9

    Non-standard abbreviations:

    Aib= aminoisobutyric acid, Nal= naphthylalanine, Mrp 2-methyltryptophane

    *maleimido-propionic acid (MPA) tag for bioconjugation, accounting for a mass of 

    151 Da.

    GHRP is depicted in Fig. 5b representing GHRP-3. Human phar-

    macokinetic data for GHRP-6 were presented by Cabrales et al. in

    2012/2013 [85,86]. Employing an isotope-dilution mass spectro-

    metric approach, GHRP-6 was determined from plasma with an

    LOQ of 5 ng/ml. Samples were enriched with 13 C-labeled GHRP-6,

    plasma proteinswere depleted by acetone-facilitated precipitation,

    and the target analyte was quantifiedfrom the concentratedsuper-

    natant by nanoLC-Q/TOF MS using a monolithic nano LC column.

    Following an i.v. bolus dose of 100, 200, or 400g/kg of body-weight, plasma samples were collected up to 72h, and GHRP-6

    concentrations were found to fall below the LOQ (5ng/ml) after

    12 h post-administration.

     2.2. High(er) molecular mass substances

     2.2.1. Growth hormone releasing hormones (GHRHs)

    In addition to GHS, growth hormone releasing hormone (GHRH)

    and its modified synthetic analogs received an enormous stimulus

    for illicit and mostly Internet-based sales [77,87,88]. In contrast to

    GHS, GHRHs actvia receptorsat the anteriorpituitary, andclinically

    approved representatives of GHRHs are tesamorelin and sermore-

    lin.In addition, research concerning newpotent analogs of GHRHas

    therapeutic agents has beencontinued uninterruptedly for decades

    [89]. This has resulted in a series of potential drug candidates, one

    of the most frequently mentioned compound of which has been

    CJC-1295 (Table 2). Bearing sequence modifications at 5 positions

    as well as a 3-maleimido-propionic acid for in vivo bioconjuga-

    tion to albumin [90], its efficacy in rats with extended plasma

    half-life was reported. Hence, complementary to GHS test meth-

    ods, screening protocols for GHRHs such as CJC-1288, CJC-1293,

    and CJC-1295 have been desirable albeit, similar to most GHRPs,

    DMPK data in humans and information on the renal elimination

    of the intact drug and relevant metabolite(s) are largely missing. A

    methodology employing immunoaffinity purification followed by

    nanoLC–MS/MS analysis was presented in 2012 [91]. By means of 

    preconcentration of 5 ml of urine using SPEand subsequent extrac-

    tion of the target analytes GHRH (1–29, sermorelin) and CJC-1295(withoutthe maleimidopropionic acidmoiety), the highly sensitive

    and specific analysis of the compounds was accomplished allow-

    ing for detection limits of 5 and 1 pg/ml, respectively. It remains to

    be clarified, however, whether the intact peptides or correspond-

    ing metabolites will be most efficient as target analytes in routine

    doping controlswhen usingurineas primarydopingcontrolmatrix.

    Alternatively, blood/serum might be a viable option to test for the

    intact drugs.

     2.2.2. Mechano growth factors (MGFs)

    Peptide hormones categorized as mechano growth factors

    (MGFs) have been explicitly mentioned as prohibited substances

    in relevant WADA regulations since 2005. The use of the term MGF

    has been ambiguous in the literature, i.e. referringto IGF-1Eb (orEcin humans) mRNA, pro-IGF-1Eb, as well as the synthetic MGF pep-

    tide, and it is hence recommended to use the name “MGF” solely

    for synthetic mechano growth factor peptides [92]. While IGF-1Ec

    mRNA is derived by alternative splicing from the IGF-1 gene, the

    formation and in vivo existence of MGF in humans comprising 24

    amino acid residues (Table 2), as well as the effects attributed to

    MGF itself are subject of substantial controversy. Earlier reports

    described MGFs capability to stimulate muscle (stem) cell prolif-

    eration and thus to increase muscle strength and regeneration,

    suggesting that its abuse in sport is cannot be excluded, despite

    missing clinical approval [93]. However, comprehensive tests with

    synthetic MGF failed to reproduce the anabolic and regeneration-

    promoting effects of MGF and a dedicated MGF receptor has yet to

    be identified to support and corroborate the commonly acceptedMGF hypothesis [94].

    To date, no dedicated detection assay for doping control pur-

    poses has been reported but findings of MGF in black-market vials

    were reported in 2012 [95], stressing the potential abuse of such

    compounds in amateur and/or elite sport despite the above men-

    tioned questionable efficacy. The black-market product was shown

    to be composed of the appropriate 24 amino acid residues as listed

    in Table 2 but comprised a C-terminal amidation, which has not

    beenpostulated forthe potentially naturallyoccurringhumanMGF.

     2.2.3. Anti-myostatin antibody MYO-029

    Myostatin, also known as growth and differentiation factor-

    8 (GFD-8), is a highly conserved member of the transforming

    growth factor beta (TGF-ˇ) superfamily which acts as a negative

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    10/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    10 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

    regulator of skeletal muscle mass [96,97]. In myostatin knock-

    out mice (Mstn−/−), a substantial increase in muscle mass was

    observed resulting from both an elevated volume (hypertrophy)

    and number (hyperplasia) of skeletal muscle fibers [97–99]. Hence,

    selective blocking of the myostatin signaling pathway has been

    considered as a therapeuticmeans to counteract or cure severedis-

    eases such as muscular dystrophies but also other arenas such as

    improving livestock production were mentioned as desirable goals

    [98,100–102]. Approaches specifically targeting myostatin include

    injectable myostatin-binding proteins such as the GDF-8 pro-

    peptide [103,104] as well as recombinant antibodies [104–106].

    In a mouse model of muscular dystrophy, the inhibition of endoge-

    nous myostatin by specific antibodies considerably improved the

    dystrophic phenotype of the animals as their muscle mass, mus-

    cle size, muscle strength and body weight were significantly

    increased [105]. Consequently, antibody-based drug candidates

    for humans were developed with MYO-029, also referred to as

    stamulumab, being first in class in 2005 [107–109]. Despite dis-

    continuation from clinical development, a considerable misuse

    potential is eminent and test methods on immunological or mass

    spectrometric platforms are desirable. With the improving capa-

    bility of high resolution/high accuracy mass spectrometry also the

    direct analysis of the (purified) antibody could be a future option

    (Fig. 5c).

     2.2.4. RNA interference

    Considering the number of research articles, ribonucleic acid

    (RNA) interference (RNAi) has continued to be one of the

    most dynamic arenas of biotechnological research (along with

    proteomics and epigenetics) [110]. The enormous therapeutic

    potential of posttranscriptional gene silencing has been recog-

    nized in numerous fields of medicinal treatment, particularly

    where the temporary knock-down of negative regulators is desir-

    able. Here, a frequently referenced example is the inhibition of 

    myostatin, the prominent negative regulator of myogenesis, the

    elimination of which has resulted in significant increases of mus-

    cle mass and strength in animal models and, thus, suggesting

    a viable means to cure myopathies such as Duchenne muscu-lar dystrophy (DMD) [111–113]. Strategies employing antisense

    oligonucleotides or small interfering RNA (siRNA) were described

    as successful means but a main prerequisite of RNAi has been the

    stability of the drug candidate. Due to the rapid degradation of 

    RNA in general, modified ribonucleic acids were introduced such as

    2-fluoronucleotides, 2-O-methylated nucleotides, phosphorothi-

    oate nucleotides, or locked nucleic acids, effectively protecting the

    siRNAfrom hydrolysis and earlyeliminationfrom the system [114].

    Due to the availability of software allowing to predict viable RNAi

    motifs and the rapid and the option to purchase fully automated

    synthesized tailored siRNA, detection strategies for this new thera-

    peutic approach bearing the risk abuse in sport have been initiated

    [115,116].

    A major step toward demonstrating the fitness-for-purpose of developed anti-doping methods was provided in 2013, when ani-

    mal studies were conducted with model siRNA [117]. Following

    the intravenous administration of therapeutic amounts of siRNA,

    urine samples were collected and subjected to two analytical

    platforms consisting of biochemical and LC-HRMS methodologies.

    Sample preparation included the specific enrichment of urinary

    siRNA using solid-phase extraction spin columns, the extract of 

    which was applied to gel electrophoresis, LC-HRMS of intact

    target analytes (i.e. intact siRNA or truncated metabolic prod-

    ucts), or chemical hydrolysis and LC-HRMS(/MS) determination

    of synthetically mono- and oligonucleotides. Further to these, a

    combination of gel electrophoresis followed by mass spectromet-

    ric analysis in a bottom-up approach was demonstrated to provide

    the desired information whether chemically modified RNA was

    present in a urine sampleor not. Overall,screening forsiRNA by gel

    electrophoresis followed by confirmatory measurements employ-

    ing LC-HRMS was found to be suitable for doping control purposes

    offering detection limits of 25pmol/ml of urine and detection win-

    dowsof 24 h whensingle dose administration to laboratory rodents

    was conducted.

    3. Compounds affecting the oxygen transfer capacity 

    Since oxygen transfer capacity is one of the key parameters

    of athletic performance, its manipulation has been attempted

    by numerous means, and doping control laboratories and anti-

    doping authorities have been urged to expand their analytical

    scope beyond approved erythropoiesis-stimulating agents (ESAs)

    such as erythropoietin (EPO). These have been successfully cov-

    ered in the past by continuously improved and updated isoelectric

    focusing (IEF) and sodium dodecylsulfate polyacrylamide gel elec-

    trophoresis (SDS-PAGE) methods [118–121]; however, recent drug

    developments have necessitated further considerations as shown

    below.

     3.1. Hypoxia-inducible factor (HIF) stabilizers

    One of the main targets of pharmacological stimulation of ery-

    thropoiesis have been hypoxia-inducible factors (HIFs) since their

    role in ‘oxygen sensing’ and production of EPO was recognized and

    demonstrated in 1992 [122]. Prolylhydroxylase-catalyzed hydrox-

    ylation of HIFs and their subsequent ubiquitination followed by

    proteasomal degradation was shown to be reduced/eliminated by

    various categories of small molecules, acting as prolylhydroxylase

    inhibitors (PHIs), resulting in an increased expression of EPO and,

    consequently, an elevated erythropoiesis [123]. The main advan-

    tage of these compounds over approved ESAs such as EPO and its

    second- and third-generation successors is their oral availability

    and lack of concerns about immunogenicity [124]. A multitude of 

    drug candidates has been presented in the past as compiled in

    recent reviews [125,126], and a selection of disclosed substancesis summarized in Table 3 as well as Fig. 6.

    The plethora of emerging HIF stabilizing therapeutics and the

    concomitant potential for abuse as doping agents has necessitated

    proactive detection method developments in doping controls. First

    attempts were conducted in 2008 with the characterization of 

    FG-2216 (Fig. 6, 24) and its implementation into routine doping

    controls followed by investigations into structurally related com-

    pounds such as FG-4592 (Fig. 7a), in vitrometabolism elucidations,

    and the development of comprehensive analytical approaches

    [127–131]. Since most of the potential HIF stabilizers comprise

    structures that suggest physico-chemical properties suitable for

    chromatographic and mass spectrometric analyses, LC–MS/MS has

    been the method of choice in most of the published analyti-

    cal approaches. By means of targeted MRM-based measurements,detection limits for FG-2216 and related (model) compounds

    between 1 and 10ng/ml were accomplished [130], which is suffi-

    cientlysensitive consideringthe expectedtherapeuticamountsand

    resulting urinary concentrations of the drugs’ metabolites. More-

    over, in order to account for the yet largely unknown structures of 

    drug candidates and respective metabolic products, a mass spec-

    trometric peculiarity of isoquinoline-derived HIF stabilizers was

    exploited to allow for a specific initial testing approach. In sev-

    eral studies, the combined elimination of methylenamine (29Da)

    or carbon monoxide(28 Da)accompaniedby theaddition ofa water

    molecule (18 Da) was observed, resulting in a nominal loss of 11 or

    10Da, respectively. Hence, a neutral loss scan for this rather spe-

    cific mass difference can support and broaden the screening for

    current and future drug candidates with similar structural features

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    11/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx 11

     Table 3

    Structure characteristics of selected HIF stabilizers.

    No. (Fig. 6) HIF stabilizer/company Pharmacophore Elemental composition Molecular mass (Da)

    24 FG-2216 Isoquinoline-glycineamide C12 H9ClN2O4   280.0251

    25 FG-4592 Isoquinoline-glycineamide C19 H16 N2O5   352.1059

    26 Fibrogen Thienopyridine-glycineamide C10 H7ClN2O4S 285.9815

    Fibrogen Thiazolopyridine-glycineamide C9H6ClN3O4 S 286.9738

    Fibrogen isothiazolopyridine-glycineamide C24 H18 N4O4S 458.1049

    27 GSK360A Quinoline-glycineamide C17 H17 FN2O5   348.1121

    28 GSK Tetrahydropyrimidine-glycineamide C19 H27 N3O6   393.190029 GSK Dihydropyrimidine-glycineamide C21 H19 N3O5   393.1325

    GSK Dihydropyrazolopyrimidine-glycineamide C16 H14 N4O5   342.0964

    GSK Pyridopyrimidine-glycineamide C11 H8BrN3O5   340.9647

    30 Amgen Naphthyridine-glycineamide C12 H11 N3O5   277.0699

    31 Amgen Dihydropyridopyrimidine-glycineamide C17 H13 FN4O5   372.0870

    32 Bayer Pyrazol-picolinonitrile C15 H11 N5O 277.0964

    33 Bayer Pyrazol-nicotinic acid C14 H9BrN4O3   359.9858

    34  Janssen Benzoimidazol C13 H11 ClN4O2S 322.0291

    35 Merck Naphthyridine-glycineamide C18 H13 F3N4 O5   422.0838

    Merck Tetrahydropyrrolo-glycineamide C22 H17 F3N4 O6S 522.0821

    although this mass spectrometric scan mode is not particularly

    sensitive.

     3.2. Cobaltous chloride

    An arguablyoutdated andold-fashionedoptionto stimulateery-

    thropoiesis is the administration of cobaltous (II) chloride [132].

    Having been used since the early 1950s, it later served as refer-

    ence to assess the activity of one international unit of EPO [133].

    Technically,cobaltous chloridecan be categorized among the group

    of HIF stabilizers as its erythropoetic effect is, at least partially,

    attributed to an HIF stabilizationviadisplacementof iron inthe cat-

    alytically active site of relevant prolylhydroxylases. In addition or

    alternatively, its mechanism of action was hypothesized to include

    a reduction of ascorbate biosynthesis or a direct binding to HIF[125]. At any rate, its erythropoietic effect was therapeutically

    exploited prior to the EPO era (until its considerable health risks

    necessitated the search for alternative therapeutics) and its abusein sport for the very same reasons cannot be excluded [134]. In the

    contrary, particularly in animal sport, the abuse of cobaltous chlo-

    ride has been raised very recently [135] and analytical approaches

    such as LC–MS/MS following complexation of cobalt (Fig. 7b) [136]

    as well as commonly employed inductively coupled plasma (ICP)

    MS are viable options to quantify the naturally occurring trace ele-

    ment in doping control specimens [137–139]. However, threshold

    levels might have to be established similarly to guidelines of horse

    racing authorities, if the quantity of cobalt is considered relevant

    for human sports drug testing in the future. This is of particular

    importance in the light of dietary supplementation with cobaltous

    chloride, for which amounts of up to 600 and 1400g/day havebeen reported to be acceptably safe for humans [140–142].

     3.3. Erythropoietin fusion protein EPO-Fc 

    Despite the fact that EPO has been clinically approved, exten-

    sively used in therapeutic settings, and further developed for 25

    years, alternatives with either improved pharmacokinetics, pro-

    files of undesirable effects, and/or easier routes of administration

    have been explored in the past [123,143–146]. Among these, the

    fusion protein composed of EPO and the fragment crystallizable

    (Fc) part of immunoglobulin G (IgG), commonly referred to as EPO-

    Fc,has been pursued in pre-clinical and clinical studies [147]. Here,

    a longer half-life of the drug candidate as compared to EPO itself 

    was observed and, more importantly, the inhalative application of 

    the substance wasenabled as supportedby thepresence of airway-

    epithelial Fc receptors.

    Despite missing clinical approval, doping control test methods

    had to be assessed in terms of their capability to detect a potential

    abuse of the experimental drug by athletes. A first and compre-

    hensive study concerning the traceability of EPO-Fc in human

    serum was done in 2012 [148]. Both screening and confirmatoryapproaches were suggested allowing the detection of EPO-Fc at

    concentrations as low as 5pg/ml of serum employing ELISA and

    gel electrophoretic/immunological approaches. The ELISA-based

    methodology exploited the fact that the Fc-part of the drug can-

    didate can be captured by protein A-coated beads, which will not

    allow for retaining natural EPO. Subsequently, the analysis of the

    beads eluate with a commercial EPO ELISA kit is conducted, which

    would trigger a confirmatory analyses if a measurable signal indi-

    cating the presence of EPO is obtained [148]. The confirmation

    of an EPO-Fc finding is ideally included in routine doping control

    protocols such as IEF or SDS-PAGE, both followed by Western blot-

    ting. The IEF properties of EPO-Fc under routine doping control

    conditions were found to be unfavorable; however, the apparent

    molecular mass of 60kDa of EPO-Fc as determined by SDS-PAGEyielded a distinct image enabling the unequivocal differentiation

    of EPO-Fc from EPO and other related ESAs.

     3.4. Peginesatide

    In addition to EPO-based ESAs and HIF stabilizers, EPO mimetic

    peptides (EMPs) have received much attention ever since first

    medicinal reports were published in 1996 on the capability of 

    EMPs to bind to and stimulate the EPO receptor [149]. A first-in-

    class approved EMP-based drug referred to as peginesatide was

    launched in 2012, comprising a homodimeric EMP structure linked

    t o a 2×20 kDa polyethylene glycol (PEG) support (Fig.8). However,

    in early 2013, the manufacturer voluntarily recalled the therapeu-

    ticagent as safetyendpoint data of cardiovascularevents anddeathwere worse for peginesatide than for the comparator EPO prod-

    uct, and until now it has not been re-introduced to the market.

    Nevertheless, the availability of this ESA and, thus, its abuse can-

    not be excluded requiring appropriate test methods as established

    between 2011 and 2012 [150–153].

    In order to allow for comprehensive doping controls, viable

    matrices including serum/plasma, urine, and dried blood spots

    (DBS)were evaluated, demonstratingthat the drug can be detected

    if therapeutic amounts have been administered. When using MS-

    basedmethodologies, precipitationof highabundant proteins from

    serum (or plasma) was followed by enzymatic hydrolysis of the

    peptidic moiety using subtilizin, yielding a diagnostic pentapep-

    tide [GPIT(1-nal)] for LC–MS/MS analysis [151]. The assay allowed

    for detection limits of 1n g/ml, which was found appropriate

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    12/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    12 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

    Fig. 6. Structures of selectedHIF stabilizers including FG-2216 (24),FG-4592 (25), and GSK360A (27). Further details are given in Table 3.

    consideringtherapeuticplasmaconcentrations of up to 1000ng/ml.

    Due to the considerably high blood concentrations, DBS analysis

    was pursued offering a faster and cheaper sample collection strat-

    egy [152]. Here, essentially the same sample preparation strategy

    was applied including extraction of the DBS, subtilizin digestion,

    and subsequent LC–MS/MS measurement. However, as a result of 

    the limited sample volume (approximately 20l of DBS), detec-tion limits were determined at 10ng/ml, which nevertheless will

    serve the purpose of sports drug testing. As urine is the most fre-

    quently collected doping control specimen, animal in vivo study

    urine samples were used toprobefor the renal elimination of either

    the intact peginesatide or metabolite(s) that could be utilized as

    target analytes [150]. Since the intact drug was observed up to 4

    days post-administration of a single therapeutic dose of the ESA,

    the methodology as adapted and optimizedfor urine was validated,

    enabling for the analysis of 0.5 ng of peginesatide per ml of urine.

    Complementary, immunological (ELISA) and gel electrophoretic

    methods for the detection of peginesatide in serum were reported,

    allowing for detection limits of 0.5ng/ml [153]. The ELISA consisted

    of a sandwich-like test method employing an immobilized mono-

    clonal anti-PEG antibody and a monoclonal biotinylated antibody

    directed against the peptidic moiety of peginesatide. Using serum

    samples from an administration study with 50g/kg bodyweight,the drug was detectable up to 10 days post-administration using

    the ELISA-based initial test method. Confirmatory analyses without

    mass spectrometry were then suggested by means of gel elec-

    trophoretic determination of immunoprecipitated peginesatide.

    Here, a different monoclonal antibody targeting a different epitope

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    13/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx 13

    Fig. 7. (a) Product ion mass spectrum of the protonated molecule [M+H]+ at m/z 353 of FG-4592, recorded at a collision energy of 30eV; (b) product ion mass spectrum of 

    M+ at m/z 355 of thecobalt-diethyldithiocarbamate (DDC) complex as analyzed with ESI-MS/MSat a collision energy of 40eV.

    of thepeptide residue wasused to capture and extract theESA from

    500l of serum. Subsequently, conventional SDS-PAGE with dou-ble Western blotting was conducted, enabling the visualization of 

    0.5ng/ml of the target analyte.

    4. Other compounds

    As the questfor faster recovery,better athleticperformance,and

    optimized bodycomposition continues, new substances advertised

    with such properties have frequently been observed with Internet-

    based suppliers as well as in products confiscated at customs.

    4.1. TB-500

    An arguablynaturaland chemical-freesubstance for horse treat-

    ment is TB-500 (Fig. 9a). Following anecdotal evidence of its abuse

    in human sport, the substance was in fact seized from possessions

    of a professional athletes’ team entourage [56], and confiscated

    material was characterized by mass spectrometry [154], chemi-

    cal synthesis and NMR [155]. Advertised as a synthetic analog to

    thymosin beta 4 (T4), it was identified as an N -terminally acety-lated heptapeptide, representing the amino acid sequence 17–23

    (the major actin-binding site) of T4. This sequence was shown topossess (corneal) tissue-repair supportive and angiogenic proper-

    ties in animal models [156,157], hence a potentialbenefit to human

    and/or animal athletes was attributed. Due to the non-natural com-

    position of the product, its traceability in doping control sampleswas demonstrated for plasma and urine specimens [155]. Samples

    were subjected to SPE (in case of plasma, high abundant proteins

    were first precipitated), and extracts were analyzed by LC–MS/MS,

    allowing for detection limits of 500pg/ml. Even though no human

    clinical study data have been published for TB-500, extrapolation

    Fig. 8. Structure cartoon of peginesatide.

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    14/18

    Please cite this article in press as: M. Thevis, W. Schänzer, Analytical approaches for the detection of emerging therapeutics and non-

    approved drugs in human doping controls, J. Pharm. Biomed. Anal. (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.020

    ARTICLE IN PRESSG Model

    PBA-9583; No.of Pages18

    14 M. Thevis, W. Schänzer / Journal of Pharmaceutical andBiomedical Analysis xxx(2014) xxx–xxx

    Fig. 9. (a)Product ionmass spectrum of theprotonated molecule [M+H]+ at m/z 889 of TB-500, recorded at a collisionenergy of 45eV; (b)product ionmass spectrum of the

    doubly charged precursor ion [M+2H]2+ at m/z 908 of AOD-9604, measured at a collisionenergy of 30eV.

    fromrecommended dosing (10mg) and horse administration study

    results [158] suggest that such LODs are adequate also for human

    doping controls.

    4.2. AOD-9604

    The peptidic compound referred to as AOD-9604 consists

    of 16 amino acids (Fig. 9b), largely identical to the primary

    structure found within the C-terminal region of human growth

    hormone (hGH), with an average molecular mass of 1815D a.

    Due to reported lipolytic and anti-lipogenic properties [159], it

    has been under development for assisting anti-obesity treatments

    but has not reached full clinical approval yet [160,161]. Since2013, AOD-9604 has been considered as prohibited according

    to WADA’s anti-doping regulations [162]; however, similarly to

    other non-approved substances, its metabolism has not been fully

    investigated or publicized. A first concern as to doping control

    analyses was whether this substance could have any impact on

    the antibody-based detection assay for hGH, and studies were

    conducted demonstrating that the so-called growth hormone iso-

    form test is specific and not affected by AOD-9604 [163]. Due to

    the peptidic nature of AOD-9604, its implementation into rou-

    tine doping controls has been accomplished according analytical

    strategies employed e.g . for GHRPs (vide supra); it remains how-

    ever to be shown whether urine, serum, and/or DBS are the matrix

    of choice for this substance, if the intact drug or metabolic prod-

    ucts are viable target analytes, and if the presumably limited

    performance-affecting properties of AOD-9604 will be of suffi-

    cient temptation to athletes to conduct anti-doping rule violations

    [164]. Clearly, trafficking of AOD-9604 via illicit and black-market

    providershas been monitoredin the past [154], requiring attention

    of doping and customs controls.

    5. Conclusion

    Preventive and proactive anti-doping work, particularly con-

    cerning analytical strategies, is of paramount importance consider-

    ingthe enormous breadth of newemergingas well as discontinued

    drugs and drug candidates. A constantly increasing plethora of 

    substances with abuse potential has been recognized over the

    past years, necessitating comprehensive, sensitive, and specific

    analytical approaches, the development of which requires state-of-

    the-art instrumentation as well as insights into pharmacology and

    metabolicpathways of these compounds, which spanfrom low(est)

    molecular mass analytes such as cobalt to intact antibodies consti-

    tuting organic molecules of more than 150kDa. Hence, continuous

    research and improvements in sports drug testing are indicated to

    ensure an adequate support of the clean athlete.

     Acknowledgments

    The authors thank the Federal Ministry of the Interior of the

    Federal Republic of Germany and the Manfred-Donike Institute for

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.jpba.2014.05.020

  • 8/18/2019 The Vis 2014

    15/18

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.bbagen.2013.1011.1016http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0250http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0245http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0240http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0235http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085(14)00260-X/sbref0230http://refhub.elsevier.com/S0731-7085