A simple and rapid ESI-LC-MS/MS method for simultaneous screening of doping agents in urine samples

Similar documents
Simultaneous Determination of a Panel of 22 Steroids in Urine and Serum by SPE and LC-MS/MS

May 1, By John Heim,Doug Staples

The Application of QuEChERS in the Extraction of Anabolic Steroids in Whole Blood

Trace-Level Analysis of Metanephrines in Plasma by HILIC LC-MS/MS

Sample purification procedure for confirmation of 3 -hydroxy stanozolol by gas chromatography/high resolution mass spectrometry

WADA Technical Document TD2014EAAS. Endogenous Anabolic Androgenic Steroids Measurement and Reporting

LCMSMS Solutions For Steroid Analysis

High speed UHPLC-MS/MS determination of multiple steroids in human serum using the Nexera MX system for multiplex analysis

Extend the time window for the detection of low level anabolic androgenic steroids and their metabolites

Proudly serving laboratories worldwide since 1979 CALL for Refurbished & Certified Lab Equipment Varian 310

The instrument should have a mass range from 2 to 2000 m/z or better Interface

Longterm Detection of Anabolic Steroid Metabolites in Urine

Analysis of Benzenesulfonic Acid and P-Toluenesufonic Acid Esters in Genotox Monitoring using UPLC/UV-MS

WADA Technical Document TD2018MRPL

Renzo BAGNATI Mario Negri Institute of Pharmacological Research- IRCCS Milano - Italy

WADA Technical Document TD2016EAAS. Endogenous Anabolic Androgenic Steroids Measurement and Reporting

Extended Application Note

WADA Technical Document TD2017NA

Determination of Selected ASTM Perfluorinated Compounds (PFCs) Using the Shimadzu LCMS No. LCMS-083. No. SSI-LCMS-083

Analysis of Melamine and Cyanuric Acid in Food Matrices by LC-MS/MS

extraction of EG and DEG from the matrix. However, the addition of all diluent at once resulted in poor recoveries.

Methylprednisolone detection in urine following local and oral administrations

Technical Procedure for Drug Chemistry Gas Chromatograph/Mass Spectrometry (GC-MS)

Development of a Clinical Research Method for the Measurement of Testosterone. Dihydrotestosterone

SIMPLE, EFFICIENT AND SIMULTANEOUS DETERMINATION OF ANABOLIC STEROIDS IN HUMAN URINE

Integration of amino acid, acylcarnitine and steroids analysis in single FIA/LC-MS/MS platform

Now it is. easy to switch. CHS TM Steroid Profiling kit and software. Brochure not for distribution in the USA and Canada

Rapid and Reliable Detection of Dissolved Gases in Water

Toxicology Gas Chromatography-Mass Spectrometry (GC-MS)

GCMSD-Headspace Analysis SOP

WADA Technical Document TD2018EAAS. Endogenous Anabolic Androgenic Steroids Measurement and Reporting

SOP: Derivatized testosterone_01 Revision: 01 Date Effective: 05/17/15

Sensitive Analysis of Genotoxins by HPLC-ECD Using Boron-Doped Diamond Electrochemical Cell

OMCL Network of the Council of Europe QUALITY ASSURANCE DOCUMENT

User guide for operating the Agilent 6420A QqQ mass spectrometer by direct infusion/injection

TRACE LEVEL DETERMINATION OF PER- AND POLYFLUOROALKYL SUBSTANCES (PFASs) IN WATER USING THE AGILENT 6460 LC/MS/MS

Introduction to the Doping Problem

MSD 96-Well MULTI-ARRAY and MULTI-SPOT Human Granulocyte Colony Stimulating Factor (hg-csf) Ultrasensitive Assay

1. INTRODUCTION CH 3 CH3

Automated Determination of Dissolved Gases in Water Anne Jurek. Abstract: Discussion:

A VALIDATED STABILITY-INDICATING RP-HPLC ASSAY METHOD FOR BOLDENONE UNDECYLENATE AND ITS RELATED SUBSTANCES

RECENT ADVANCES IN DOPING ANALYSIS (2)

Introduction. Experimental

International Journal of Research in Pharmaceutical and Nano Sciences Journal homepage:

Evaluation of linear and step gradient performance and retention time precision of the Agilent 1200 Series Rapid Resolution LC

Introduction. Saurabh Dubey 1, * and Alka Beotra 2

Retention Time Locking: Concepts and Applications. Application

IJPAR Vol.6 Issue 1 Jan - Mar Journal Home page:

TROUBLESHOOTING. Dwell Volume Revisited. Gradient transfer does not have to be problematic.

CORESTA RECOMMENDED METHOD Nº 67

Analysis of Melamine and Cyanuric Acid in Food Matrices by LC-MS/MS

Client: Ultimate Fitness Events - USA 600 Golden Gate St Lake Orion, MI Phone: (248) Identification: 104 DOB: Sex:

Agilent Dimension Software for ELSD User Manual

WADA Technical Document TD2010EAAS. Endogenous Anabolic Androgenic Steroids Testing, Reporting and Interpretive Guidance

SMART 4 Sports Panel. Drug Free Sport 1 7/13/07

Setting a New Standard in Flash Chromatography Performance

SPECIFIC DETECTION OF ANDROST-4-ENE-3,17-DIONE

Sports drug testing an analyst s perspective

ASTM WK Standard Test Method for Dissolved Gases. Anne Jurek Applications Chemist

Gases&Technology. Measurement of Impurities in Helium Using the Dielectric Barrier Discharge Helium Ionization Detector. FEATURE.

M/s. Thermo fisher Scientific

Using the PreVent System in Time-Saver Mode with the AutoSystem XL Gas Chromatograph and the TurboMass Mass Spectrometer

Chemical Analysis for Methyltestosterone

FRANZ CELL TEST OF NANO GLUTATHIONE WITH HPLC ANALYSIS

Gas chromatographic determination of 1,4-dioxane at the

A NOVEL RP-HPLC METHOD FOR THE QUANTIFICATION OF ICATIBANT IN FORMULATIONS

Agilent 1290 Infinity LC System Applications requiring the Agilent Ultra-Low Dispersion Kit

(2 pts) Draw the line of best fit through the data and estimate the concentration of Fe in your sample solution.

Quantitative Assay Validation for Oxandrolone in Human Plasma Using LC MS-MS

Automated Determination of Dissolved Gases in Water By Headspace Calibration of Mixed Gases Anne Jurek

Fast method for Ginseng Analyses using Agilent Poroshell 120 Columns Scaled from a Traditional Method

Acclaim RSLC. (Rapid Separation Liquid Chromatography)

Low-Pressure Retention Time Locking with the 7890A GC. Application. Authors. Abstract. Introduction HPI

Updates on Anti-doping and TUE Management in Paralympic Sport

SUPPORTING INFORMATION

ANALYTICAL SCALE HPLC AGILENT SYSTEMS OPERATIONAL QUALIFICATION

Viapath Innovation Academy 5 December 2014

GC-MS(/MS) investigations on long-term metabolites of 17-methyl steroids

Laboratory Hardware. Custom Gas Chromatography Solutions WASSON - ECE INSTRUMENTATION. Engineered Solutions, Guaranteed Results.

Simplified Backflush Using Agilent 6890 GC Post Run Command Application Note

Agilent 7800 ICP-MS. Specifications and Typical Performance. Fast track metals analysis with Solution-Ready ICP-MS

International Journal of Generic Drugs ANALYTICAL METHOD PROCEDURES THIS SOP IS 'SWITCHED : OFF : ON'

Drugs in Sport. Fifth Edition. Edited by David R. Mottram. Routledge. Taylor & Francis Group LONDON AND NEW YORK

During today's webinar we're going to look at the following topics:

METHOD 3C - DETERMINATION OF CARBON DIOXIDE, METHANE, NITROGEN, AND OXYGEN FROM STATIONARY SOURCES

Automated Determination of Dissolved Gases in Water Anne Jurek

RECENT ADVANCES IN DOPING ANALYSIS (4)

Laboratory Hardware. Custom Gas Chromatography Solutions WASSON - ECE INSTRUMENTATION. Custom solutions for your analytical needs.

Validation of the MRM pesticides from the Working Document SANCO/12745/2013 of November 2017 rev. 9(1)

EVALUATION OF PROFICIENCY TESTING PROGRAM, SETTING PRIORITIES FOR THE FUTURE.

Experiment GC : Analysis of BTEX by GC-FID

STD-3-V1M4_1.7.1_AND_ /15/2015 page 1 of 6. TNI Standard. EL-V1M4 Sections and September 2015

DETERMINATION OF TETRAHYDROTHIOPHENE IN AMBIENT AIR BY GAS CHROMATOGRAPHY WITH A PFPD DETECTOR COUPLED TO A PRECONCENTRATION TECHNOLOGY

Automated Low Background Solid Phase Extraction of Perfluorinated Compounds in Water. Ruud Addink Fluid Management Systems Watertown MA

CHAPTER 3 Development and Validation of a RP-HPLC method for Dutasteride and its impurities in bulk drug

Australian Supplement Survey Summary

More Speed and Resolution for Current Methods

APPLICATION NOTE. GC Integrated Permeation Device

Following an Olympics shadowed by the Russian

Structure Characterization and Analysis of Drugs Relevant to Sports Drug Testing by GC-Q/TOF

Transcription:

Research Article A simple and rapid ESI-LC-MS/MS method for simultaneous screening of doping agents in urine samples I. Madhusudhana Reddy, Alka Beotra, S. Jain, S. Ahi ABSTRACT National Dope Testing Laboratory, Ministry of Youth Affairs and Sports, J. N. Stadium, New Delhi, India Received: 06.12.2008 Revised: 06.02.2009 Accepted: 06.02.2009 DOI: 10.4103/0253-7613.51347 Correspondence to: Dr. Alka Beotra E-mail: drabeotra@rediffmail.com Objective: The use of performance enhancing substances is banned in sports by the World Anti-Doping Agency (WADA). Though most prohibited substances can be detected by GC/MS, inclusion of corticosteroids and designer drugs has made it essential to detect these critical doping agents on LC/MS/MS due to their better separation and detection. Materials and Methods: A common extraction procedure for the isolation of acidic, basic and neutral drugs from urine samples was developed. A total of 28 doping drugs were analyzed on API 3200 Triple quadrupole mass spectrometer using C18 column in atmospheric pressure electrospray ionization. The mobile phase composition was a mixture of 1% formic acid and acetonitrile with gradient time period. Results: The method developed was very sensitive for detection of 28 doping agents. The linearity was performed for each drug and the total recovery percentage ranged from 57 to 114. Limit of detection is found to be 0.5 ng/ml for carboxy finasteride and 1-5 ng/ml for other drugs. The method was successfully used to detect positive urine samples of 3-OHstanozolol, methyl phenidate, mesocarb, clomiphene metabolite and carboxy finasteride. Conclusion: The method developed based on controlled ph extraction method and HPLC-mass spectrometry analysis allowed better identification and confirmation of glucocorticosteroids and a few other drugs in different categories. The validated method has been used successfully for testing of 1000 In-competition samples. The method helped in detection of chemically and pharmacologically different banned drugs in urine in a single short run at a minimum required performance limit set by WADA. KEY WORDS: Detection limit, glucocorticosteroids, LC/MS/MS, validation Introduction The use and abuse of performance-enhancing substances has been an issue in sports since ancient times. The availability of numerous synthetic steroids and peptide hormones has made testing an analytical challenge. [1] Acknowledging the facts of performance enhancing and deleterious side effects, the World Anti-doping Agency (WADA) banned these drugs in sports. [2] The techniques of dope testing have improved immensely from 1972 to 2008 using improved extraction methods [3] and sophisticated equipment. [4] Mass spectrometry was used for the first time in 1972 during the Munich Olympics. Since then various new chromatographic techniques viz. high-resolution mass spectrometry (Atlanta Olympics Games 1996), isotope ratio mass spectrometry (Special Olympic Winter Games, 1998) and liquid chromatography mass spectrometry (Athens Olympic Games, 2004) have come into dope testing. There are a total of 35 WADA accredited labs in the world. Each lab has their own set testing protocols utilizing various equipment viz. GC, GC/MS, HRMS, LC/MS/MS and IRMS. There are a number of methods available for individual groups of drugs viz. diuretics, [5] beta blockers, [6] stimulants and glucocorticosteroids [7] but during testing in our lab it was felt that apart from certain drugs like glucocorticosteroids for which it is mandatory to detect on LC/ MS/MS, there are a few other drugs in various other categories of drugs viz. anabolic steroids (6), diuretics (1), stimulants (4) which are required to be included in the LC/MS/MS methods because of difficulty in detection by conventional GC and GC/ MS. Therefore, the purpose of the present study was to explore the possibility of detecting by the glucocorticosteroids method a few other drugs which were difficult to detect at required lower concentration levels by GC and GC/MS methods. Materials and Methods Chemicals All reference standards were purchased from Sigma (St. Louis, MO, USA), Steraloids (Newport, USA) or National 80

Measurement Institute (Pymble, NSW, Australia). Acetonitrile and ethyl acetate were obtained from Qualigens (Worli, Mumbai, India), methanol from JT Baker (Phillipburg, USA), tertiary butyl methyl ether (TBME) from Acros Organics (New Jersey, USA), formic acid from Merck (Worli, Mumbai, India). De-ionized water was prepared on a MilliQ Laboratory Plant (Millipore, Bedford, USA). Organic solvents and reagents were all of analytical grade. LC and MSMS Apparatus The chromatographic system consisted of an Agilent 1100 series (Agilent Technologies, Waldbronn, Germany) equipped with G1311A high pressure gradient pumping system, G1329A Autosampler, G1379A degasser, G1316A column compartment and G1315B diode array detector. Mass spectrometric analyses were conducted using an API 3200 Triple quadrupole instrument (Applied-Biosystem-Sciex, Concord, Canada), equipped with a pneumatically assisted APCI (heated nebulizer) and ESI (electrospray) ionization source. The main analytical parameters of the mass spectrometer are summarized in Table 1. The whole system was controlled using Analyst 1.4 software (Applied- Biosystem-Sciex, Concord, Canada). Sample Preparation Procedure A common procedure for the isolation of acidic, basic and neutral drugs from urine sample prior to LC/MS/MS was developed. The sample extraction procedure was illustrated in Figure 1. Table 2 reports different categories of drugs extracted by this procedure. Method development Electrospray ionization (ESI) and LC conditions Mobile phase was introduced into the mass spectrometer via the ESI source operating in the positive ion/negative ion mode under multiple reaction monitoring conditions (MRM). Nitrogen was used as the nebulizing and curtain gas. Fragmentation was achieved with nitrogen. The dwell time for each transition was 60 msec and interchannel delay was 5 msec. For maximum sensitivity, the mass spectrometer parameters such as nebulizing gas, curtain gas and collision gas were optimized and collision energy is presented in Table 3. The temperature was 550 o C and resolution was set at the unit. Figure 1: Sample preparation ß ow steps Urine Sample 2 ml Sp.Gr.> 1.010 4 ml Sp.Gr.< 1.010 Add internal standard Add 1ml 0.2M-phosphate buffer ph 7.0, 50µl β-glucuronidase into the dry residue and mix well. Incubate at 60 C for 60 min. Cool and add 250µl 7% K 2 CO 3 solution ph 9-10 Add 4mL TBME Mix by horizontal shaking for 10 min Organic layer Aqueous layer Add 6N HCL to maintain ph 2-3 Add 4 ml ethyl acetate Separate Organic layer Dry & reconstitute 50:50 Mobile phase Inject LC-MS/MS 81

Figure 2: LCMSMS total ion chromatogram resulting from (A) analysis of urine sample spiked with mixture of doping agents; (B) blank urine sample with internal standard A B Table 1 Tandem mass spectrometer and HPLC working parameters Parameter Value Tandem mass spectrometer working parameters Scan type MRM Polarity Positive/Negative Ion source ESI Nebulizing gas 3 Curtain gas 10 Source temperature 550oC HPLC working parameters Column C-18 ODS Inertsil, 50 x 4.6 x 3.5 µm Flow 700 µl per minute Injection volume 10 µl Mobile phase 1% Formic acid, Acetonitrile Gradient 0-5 min B 15%, 5-6 min B 60%, 6-7 min B 100%, 7-7.10 min B 15%, 7.10-11.0 min B 15% LC conditions The chromatogram was on Inertsil ODS-3 (3.0 um, 50 mm x 4.6 mm i.d.) C 18 column (GL Sciences Inc., Tokyo, Japan) at 25 o C temperature. Figure 3 shows the chromatogram of the single MRM transition of a few doping agents. The mobile phase composition was a mixture of 1% formic acid and acetonitrile with gradient time period. The flow rate was 700 µl per minute. Results Internal standard 17-α methyl testosterone was used as the internal standard. The total recovery and elution time of the internal standard were perfectly fit for the chromatographic conditions of the method. Limit of detection (LOD) Limit of detection is defined as the measure of the background noise with that of the baseline peak. Three times the standard deviation of noise in the negative urine was used to estimate the limit of detection. LOD was measured by spiking seven aliquots of negative urine samples. The analysis of these was done for a period of few weeks for instrument performance. LOD varied much for steroids, diuretics and glucocorticosteroids due to differences in the ionization criteria of individual drugs. Table 4 shows the LOD of different drugs. Calibration and quality control samples Calibration standards were prepared by addition of ten 82

Table 2 Category of compounds analyzed Category Compound Diuretic Chlorothiazide Stimulants Ritalinic acid, Methyl Phenidate, Mesocarb Anabolic androgenic steroids Formebolone, Clomiphene metabolite, Epiternbolone, Gestrinone, Methy testosterone, THG, 3-OH-Stanozolol Glucocorticosteroids Triamcinolone, Prednisolone, Prednisone, Hydrocortisone, Fludrocortisone, Methyl prednisolone, Betamethasone, Dexamethasone, Flumethasone, Beclomethasone, Triamcinolone Acetonide, Desonide, Flunisolide, Fluocortolone, Fludrocortisone Acetate, Budesonide 5 α - reductase inhibitor Carboxy Þ nasteride Table 3 Analyte dependent mass spectrometry parameter for doping compounds Analyte ESI Precursor Product Collision Ionization Ions Ions Energy Ritalinic acid +ve 220 84 30 Chlorothiazide -ve 294 214,179-50 Methyl Phenidate +ve 234 84 30 Triamcinolone +ve 395 357,225 20 Formebolone +ve 347 173 30 3-OH-Stanozolol +ve 345 97,121, 30 107,111, 219 Mesocarb +ve 339 193,135 30 Prednisolone +ve 361 343,307 30 Prednisone +ve 359 147,341 25 Hydrocortisone +ve 363 121 31 Clomiphene metabolite +ve 422 100,72 30 Fludrocortisone +ve 381 181,105 35 Carboxy Þ nasteride +ve 403 335,175 30 Methyl prednisolone +ve 375 161,357 30 Betamethasone +ve 393 147,355 30 Dexamethasone +ve 393 147,355 30 Flumethasone +ve 411 253,121 26 Beclomethasone +ve 409 279,391 30 Triamcinolone Acetonide +ve 435 213,397 27 Desonide +ve 417 399,147 27 Flunisolide +ve 435 321,121 25 Fluocortolone +ve 377 171,303 22 Epitrenbolone +ve 271 199,227 30 Fludrocortisone Acetate +ve 423 239,181 45 Budesonide +ve 431 173,323 33 Gestrinone +ve 309 241 30 Methy testosterone +ve 303 109 30 THG +ve 313 241 30 microliters of working solution to drug free urine (DFU) to obtain required concentration levels. Quality control samples were prepared at concentrations 30 ng/ml (glucocorticosteroids), 10 ng/ml (steroids except 3-OH-stanozolol at 2 ng/ml) and 100 ng/ml (diuretics and stimulants) for minimum required performance limit (MRPL) set by WADA. The internal standard (17 α-methyl testosterone) solution (500 ng/ml) was prepared by diluting its stock solution with methanol. Working solutions were prepared from the stock solution by dilution using methanol. Calibration curve A five-point calibration curve was plotted for each drug with the internal standard covering the LOD and MRPL. The calibration curves were generated for three consecutive days and linearity was assessed [Table 4]. The calibration curve had a correlation coefficient of 0.9998. The five-point calibration curve gave acceptable results for the standard and was used for all quantitative calculations. Precision and accuracy Precision and accuracy was determined by analyzing six sets of quality control samples. The within batch and between batch precision and accuracy was done by randomized daily process and analyzed in position at the end of the batch. Table 4 summarizes the cumulative variance of all drugs. Specificity and matrix effect The negative urine sample is spiked with the internal standard to find the specificity and matrix effect of the method. Figure 2(b) shows the total ion chromatogram of blank urine sample with internal standard. Matrix interferences were observed due to the short run time but eliminated by increasing post run time of the method. The method found to be specific for the drugs summarized in Figure 2(a). Total recovery Total recovery completely relied on the sample preparation procedure. The total recovery of steroids, glucocorticosteroids, diuretics and narcotics was found to be good and acceptable with range of 57 to 114% as these compounds were extracted at different ph conditions. The total recovery was evaluated by comparing the mean peak areas of ten processed samples with the mean peak areas of unprocessed direct reference standard solutions of same concentrations. Figure 4 reports average recoveries for extraction of doping agents. Discussion The present method developed on HPLC-mass spectrometer could detect all the 28 doping agents in a single run with the limit of detection set by WADA. The total run time of the present method was short with better separation. In the last 3 to 4 years, a number of methods have been published on analysis of doping agents on LC/MS/MS. However, these methods are on specific group of drugs viz. diuretics, [5] beta-blockers, [6] glucocorticosteroids [7] and a few anabolic steroids. [8] The present method was developed for 14 glucocorticosteroids 83

Figure 3: Single MRM transition of few doping drugs Table 4 Compounds with limit of detection (LOD), retention time, linear range and CV% Analyte RT LOD Linear range CV (Min) (ng/ml) (ng/ml) % Ritalinic acid 2.86 NA NA NA Chlorothiazide 2.98 5 50-500 10.5 Methyl Phenidate 3.00 5 50-500 10.5 Triamcinolone 4.46 2 5-60 2.1 Formebolone 4.64 5 5-50 10.3 Mesocarb 4.68 NA NA NA 3-OH-Stanozolol 4.90 1 1-30 12.3 Prednisolone 5.09 2 15-120 10.2 Prednisone 5.10 2 15-120 6.3 Hydrocortisone 5.15 NA NA NA Clomiphene metabolite 5.15 NA NA NA Fludrocortisone 5.26 2 5-60 9.8 Carboxy Þ nasteride 5.31 0.5 5-60 2.1 Methyl prednisolone 5.65 3 15-120 10.7 Betamethasone 5.77 1 1-20 13.6 Dexamethasone 5.77 1 1-20 13.6 Flumethasone 5.81 1 15-120 0.5 Beclomethasone 5.99 5 15-120 8.5 Triamcinolone Acetonide 6.12 2 15-120 4.7 Desonide 6.15 2 15-120 3.7 Flunisolide 6.15 1 15-120 11.5 Fluocortolone 6.34 1 15-120 5.4 Epiternbolone 6.45 2 5-60 8.6 Fludrocortisone Acetate 6.51 2 15-120 5.4 Budesonide 7.19 1 15-120 6.4 Gestrinone 7.30 3 5-50 4.3 Methy testosterone 7.45 NA NA NA THG 8.35 1 2.5-20 7.1 Figure 4: Average recoveries for extraction of doping agents Beclomethasone Flumethasone Dexamethasone Carboxy finasteride Budesonide Fludrocortisone acetate Fluocortolone Flunisolide Desonide Triamcinolone acetonide Betamethasone Methylprednisolone Fludrocortisone Prednisone Prednisolone Triamcinolone Chlorothiazide 3-OH-Stanozolol THG Methyl testosterone Gestrinone Epitrenbolone Formebolone Mesocarb Methyl phenidate 0.0 20.0 40.0 60.0 80.0 100.0 120.0 140.0 % recovery 84

Figure 5: TIC and mass spectrum of 3-OH-Stanozolol from human urine excretion study sample and 6 anabolic steroids and then extended to a few estrogenic drugs, stimulants and diuretics, thereby detecting a total of 28 drugs in a single screen. It helped to detect problematic drugs from diuretics (chlorothiazide), stimulants (methyl phenidate, mesocarb) and steroids (3-OH-stanozolol) thereby making one versatile method for various drugs. The ionization and fragmentation pattern differs for each category of drugs; the results suggest that glucocorticosteroids are neutral compounds to ionize [9] whereas steroids and diuretics are polar [10] and easily fragmentable. Also observed was no polarity switching between chlorothiazide (detected at a retention time of 2.98 min in negative mode) and methyl phenidate (detected at a retention time of 3.00 min in positive mode) though they are co-eluting substances. Furthermore, analysis shows stimulants and narcotics are highly sensitive and ESI appeared to be the best sensitive ionization for methyl phenidate. The ionization pattern and separation were earlier discussed in a few publications which suggest buffering agents are significant in ionization. [11] In the present study formic acid and acetonitrile are used as eluting mobile phases that gave good separation and sensitivity. The method was successfully used to detect positive urine samples of 3-OH-stanozolol, methylphenidate, mesocarb, clomiphene metabolite and carboxy finasteride. The positive urine samples were collected for 48-72 hrs after single dose administration of 3-OH-stanozolol, methyl phenidate, mesocarb, clomiphene and finasteride to healthy volunteers. The drugs and metabolites could be detected up to 48 hrs later. However, methyl phenidate and ritalinic acid were tested in a positive sample purchased from Australian Doping Lab. The total recovery percentage ranged from 57 to 114 where the lower end of the recovery may be due to the matrix associated with the samples at different ph conditions. Further, the recovery percentage for different categories of drugs done by this method is at concentrations of 10 ng/ml for steroids except 3-OH-Stanozolol at 2 ng/ml, 250 ng/ml for stimulants, 100 ng/ ml for diuretics, 30 ng/ml for glucocorticosteroids and 100 ng/ ml for carboxy finasteride. The extraction recovery percentage in the range of 80-120% is considered to be acceptable by the international validation protocols. [12] However, the protocol for bioanalytical method validation explains that the recovery of an analyte need not be 100%, but the extent of recovery of an analyte and of the internal standard should be consistent, precise and reproducible. [13] The validated method was used for testing of 1000 In-competition samples successfully. However, during this testing adverse analytical findings were reported for 3-OH-Stanozolol in ten samples. In conclusion, the method developed based on controlled ph extraction method and HPLC-mass spectrometry analysis allowed better identification and confirmation of chemically and pharmacologically different banned drugs in urine in a single short run. The method could detect target analytes at a concentration of minimum required performance limit set by WADA. The assay provides a useful alternative approach to individual methods for screening and confirmation of a few diuretics, stimulants, narcotics, steroids and glucocorticosteroids. The approach has great potential in doping 85

and clinical testing and might simplify the analytical screening and confirmation cost effectively. Acknowledgments We thank Dr. Sangeeta Shukla, Professor, SOS of Zoology, Jiwaji University, Gwalior, who helped and provided constant support during this study. References 1. Larry D Bowers. Analytical advances in detection of performance enhancing compounds. Clin Chem 1997;43:7:1299-304. 2. Duntas L H, Parisis C. Doping: A challenge to the endocrinologist: A reappraisal in view of the Olympic games of 2004. Hormones 2003;2:35-42. 3. Reddy MI, Beotra A, Jain S, Kaur T, Lal R. PuriÞcation of urine samples to improve detection limit of anabolic agents. Indian J Pharmacol 2007;39:39-41. 4. Thevis M, Schanzer W. Mass spectrometry in doping control analysis. Curr Organ Chem 2005;9:825-48. 5. Fouracre C, Kazlauskas R. Diuretics screening and conþrmation using LC/ MS/MS. Recent advances in doping analysis (9). Sport und Buch Strauβ, Koln 2001. p. 321-6. 6. Thevis M, Schanzer W. Examples of doping control analysis by liquid chromatogrqaphy-tandem mass spectrometry: Ephedrines, β-receptor blocking agents, diuretics, sympathomimetics, and cross-linked heamoglobins. J Chr Sci 2005;43:22-31. 7. Fluri K, Rivier L. Method for conformation of synthetic corticosteroids in doping urine samples by liquid chromatography-electrospray ionization mass spectrometry. J Chromatography A 2001;926:87-95. 8. Thevis M, Bommerich U, Opfermann G, Schanzer W. Characterization of chemically modiþed steroids for doping control purposes by electrospray ionization tandem mass spectrometry. J Mass Spectrom 2005;40:494-502. 9. Steffenrud S, Maylin G. Thermospray liquid chromatography-mass spectrometry of corticosteroids. J Chromatography 1992;577:221-7. 10. Thieme D, Grosse J, Lang R, Mueller R K, Wahl A. Suitability of HPLC/MS-MS for the detection of diuretics: Recent advances in doping analysis (8). Sport und Buch Strauβ, Koln 2000. p. 47-55. 11. Guan F, Uboh C, Soma L, Hess A, Luo Y, Tsang DS. Sensitive liquid chromatography/tandem mass spectrometric method for the determination of beclomethasone dipropionate and its metabolites in equine plasma and urine. J Mass Spectrom 2003;38:823-38. 12. De Bievre. The Þtness for purpose of analytical methods. A laboratory guide to method validation and related topics. EURACHEM guidance document. October 2004. 13. Guidance for Industry-Bioanalytical method validation, U.S. Department of health and human services, FDA, CDER, CVM. May 2001. AUTHOR INSTITUTION MAP FOR THIS ISSUE Please note that not all the institutions may get mapped due to non-availability of requisite information in Google Map. For AIM of other issues, please check Archives/Back Issues page on the journal s website. 86